首页 > 最新文献

Journal of Chemical Information and Modeling 最新文献

英文 中文
AABBA Graph Kernel: Atom-Atom, Bond-Bond, and Bond-Atom Autocorrelations for Machine Learning. AABBA Graph Kernel:用于机器学习的原子-原子、邦德-邦德和邦德-原子自相关。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-12-09 Epub Date: 2024-11-24 DOI: 10.1021/acs.jcim.4c01583
Lucía Morán-González, Jørn Eirik Betten, Hannes Kneiding, David Balcells

Graphs are one of the most natural and powerful representations available for molecules; natural because they have an intuitive correspondence to skeletal formulas, the language used by chemists worldwide, and powerful, because they are highly expressive both globally (molecular topology) and locally (atom and bond properties). Graph kernels are used to transform molecular graphs into fixed-length vectors, which, based on their capacity of measuring similarity, can be used as fingerprints for machine learning (ML). To date, graph kernels have mostly focused on the atomic nodes of the graph. In this work, we developed a graph kernel based on atom-atom, bond-bond, and bond-atom (AABBA) autocorrelations. The resulting vector representations were tested on regression ML tasks on a data set of transition metal complexes; a benchmark motivated by the higher complexity of these compounds relative to organic molecules. In particular, we tested different flavors of the AABBA kernel in the prediction of the energy barriers and bond distances of the Vaska's complex data set (Friederich et al., Chem. Sci., 2020, 11, 4584). For a variety of ML models, including neural networks, gradient boosting machines, and Gaussian processes, we showed that AABBA outperforms the baseline including only atom-atom autocorrelations. Dimensionality reduction studies also showed that the bond-bond and bond-atom autocorrelations yield many of the most relevant features. We believe that the AABBA graph kernel can accelerate the exploration of large chemical spaces and inspire novel molecular representations in which both atomic and bond properties play an important role.

分子图是最自然、最强大的分子表征之一;自然是因为分子图与全球化学家使用的语言--骨骼公式有着直观的对应关系;强大是因为分子图在全局(分子拓扑)和局部(原子和化学键属性)两方面都具有很强的表现力。图核用于将分子图转化为固定长度的向量,基于其测量相似性的能力,这些向量可用作机器学习(ML)的指纹。迄今为止,图核主要集中在图的原子节点上。在这项工作中,我们开发了一种基于原子-原子、键-键和键-原子(AABBA)自相关性的图核。我们在过渡金属复合物数据集的回归 ML 任务中测试了由此产生的矢量表示法;与有机分子相比,这些化合物具有更高的复杂性,因此我们对这些数据集进行了基准测试。特别是,我们在预测瓦斯卡复合物数据集(Friederich 等人,《化学科学》,2020 年,11 期,4584 页)的能垒和键距时,测试了 AABBA 核的不同类型。对于包括神经网络、梯度提升机和高斯过程在内的各种 ML 模型,我们发现 AABBA 优于仅包含原子-原子自相关性的基线模型。降维研究还表明,键-键和键-原子自相关性产生了许多最相关的特征。我们相信,AABBA 图核可以加速对大型化学空间的探索,并激发新的分子表征,其中原子和化学键特性都发挥了重要作用。
{"title":"AABBA Graph Kernel: Atom-Atom, Bond-Bond, and Bond-Atom Autocorrelations for Machine Learning.","authors":"Lucía Morán-González, Jørn Eirik Betten, Hannes Kneiding, David Balcells","doi":"10.1021/acs.jcim.4c01583","DOIUrl":"10.1021/acs.jcim.4c01583","url":null,"abstract":"<p><p>Graphs are one of the most natural and powerful representations available for molecules; natural because they have an intuitive correspondence to skeletal formulas, the language used by chemists worldwide, and powerful, because they are highly expressive both globally (molecular topology) and locally (atom and bond properties). Graph kernels are used to transform molecular graphs into fixed-length vectors, which, based on their capacity of measuring similarity, can be used as fingerprints for machine learning (ML). To date, graph kernels have mostly focused on the atomic nodes of the graph. In this work, we developed a graph kernel based on atom-atom, bond-bond, and bond-atom (AABBA) autocorrelations. The resulting vector representations were tested on regression ML tasks on a data set of transition metal complexes; a benchmark motivated by the higher complexity of these compounds relative to organic molecules. In particular, we tested different flavors of the AABBA kernel in the prediction of the energy barriers and bond distances of the Vaska's complex data set (Friederich et al., <i>Chem. Sci.</i>, 2020, <b>11,</b> 4584). For a variety of ML models, including neural networks, gradient boosting machines, and Gaussian processes, we showed that AABBA outperforms the baseline including only atom-atom autocorrelations. Dimensionality reduction studies also showed that the bond-bond and bond-atom autocorrelations yield many of the most relevant features. We believe that the AABBA graph kernel can accelerate the exploration of large chemical spaces and inspire novel molecular representations in which both atomic and bond properties play an important role.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":" ","pages":"8756-8769"},"PeriodicalIF":5.6,"publicationDate":"2024-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11632777/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142708590","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Application of Machine Learning in Doping Detection. 机器学习在兴奋剂检测中的应用。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-12-09 Epub Date: 2024-11-22 DOI: 10.1021/acs.jcim.4c01234
Qingqing Yang, Wennuo Xu, Xiaodong Sun, Qin Chen, Bing Niu

Detecting doping agents in sports poses a significant challenge due to the continuous emergence of new prohibited substances and methods. Traditional detection methods primarily rely on targeted analysis, which is often labor-intensive and is susceptible to errors. In response, machine learning offers a transformative approach to enhancing doping screening and detection. With its powerful data analysis capabilities, machine learning enables the rapid identification of patterns and features in complex compound data, increasing both the efficiency and the accuracy of detection. Moreover, when integrated with nontargeted metabolomics, machine learning can predict unknown metabolites, aiding the discovery of long-lasting biomarkers of doping. It also excels in classifying novel compounds, thereby reducing false-negative rates. As instrumental analysis and machine learning technologies continue to advance, the development of rapid, scalable, and highly efficient doping detection methods becomes increasingly feasible, supporting the pursuit of fairness and integrity in sports competitions.

由于新的禁用物质和禁用方法不断涌现,检测体育运动中的兴奋剂成为一项重大挑战。传统的检测方法主要依赖于靶向分析,这通常需要大量人力,而且容易出错。对此,机器学习为加强兴奋剂筛查和检测提供了一种变革性方法。凭借强大的数据分析能力,机器学习能够快速识别复杂化合物数据中的模式和特征,从而提高检测的效率和准确性。此外,当与非靶向代谢组学相结合时,机器学习还能预测未知代谢物,帮助发现兴奋剂的长效生物标记物。它还能对新型化合物进行分类,从而降低假阴性率。随着仪器分析和机器学习技术的不断进步,开发快速、可扩展和高效的兴奋剂检测方法变得越来越可行,从而为追求体育竞赛的公平性和公正性提供支持。
{"title":"The Application of Machine Learning in Doping Detection.","authors":"Qingqing Yang, Wennuo Xu, Xiaodong Sun, Qin Chen, Bing Niu","doi":"10.1021/acs.jcim.4c01234","DOIUrl":"10.1021/acs.jcim.4c01234","url":null,"abstract":"<p><p>Detecting doping agents in sports poses a significant challenge due to the continuous emergence of new prohibited substances and methods. Traditional detection methods primarily rely on targeted analysis, which is often labor-intensive and is susceptible to errors. In response, machine learning offers a transformative approach to enhancing doping screening and detection. With its powerful data analysis capabilities, machine learning enables the rapid identification of patterns and features in complex compound data, increasing both the efficiency and the accuracy of detection. Moreover, when integrated with nontargeted metabolomics, machine learning can predict unknown metabolites, aiding the discovery of long-lasting biomarkers of doping. It also excels in classifying novel compounds, thereby reducing false-negative rates. As instrumental analysis and machine learning technologies continue to advance, the development of rapid, scalable, and highly efficient doping detection methods becomes increasingly feasible, supporting the pursuit of fairness and integrity in sports competitions.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":" ","pages":"8673-8683"},"PeriodicalIF":5.6,"publicationDate":"2024-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142685417","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CPIScore: A Deep Learning Approach for Rapid Scoring and Interpretation of Protein-Ligand Binding Interactions. CPIScore:用于快速评分和解释蛋白质配体结合相互作用的深度学习方法。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-12-09 Epub Date: 2024-11-19 DOI: 10.1021/acs.jcim.4c01175
Li Liang, Yunxin Duan, Chen Zeng, Boheng Wan, Huifeng Yao, Haichun Liu, Tao Lu, Yanmin Zhang, Yadong Chen, Jun Shen

Protein-ligand binding affinity prediction is a crucial and challenging task in the field of drug discovery. However, traditional simulation-based computational approaches are often prohibitively time-consuming, limiting their practical utility. In this study, we introduce a novel deep learning method, CPIScore, which leverages the capabilities of Transformer and Graph Convolutional Networks (GCN) to enhance the prediction of protein-ligand binding affinity. CPIScore utilizes the Transformer architecture to capture comprehensive global contexts of protein and ligand sequences, while the GCN component effectively extracts local features from small molecular graphs. Our results demonstrate that CPIScore surpasses both traditional machine learning and other deep learning models in accuracy, achieving a Pearson's r of 0.74 on our test set. Furthermore, CPIScore has been validated across multiple targets, proving its ability to discern inhibitors from a diverse compound library with high enrichment rates. Notably, when applied to a generated focused library of compounds, CPIScore successfully identified six potent small-molecule inhibitors of ATR, which were tested experimentally and four small molecules exhibited inhibitory activity below ten nanomoles. These results highlight CPIScore's potential to significantly streamline and enhance the efficiency of drug discovery processes.

蛋白质与配体的结合亲和力预测是药物发现领域中一项至关重要且极具挑战性的任务。然而,传统的基于模拟的计算方法往往耗时过长,限制了其实用性。在本研究中,我们介绍了一种新颖的深度学习方法 CPIScore,它充分利用了 Transformer 和图形卷积网络(GCN)的功能,以增强对蛋白质配体结合亲和力的预测。CPIScore 利用 Transformer 架构捕捉蛋白质和配体序列的全面全局上下文,而 GCN 组件则有效地从小型分子图中提取局部特征。我们的研究结果表明,CPIScore 在准确性上超越了传统机器学习和其他深度学习模型,在测试集上的皮尔森 r 达到了 0.74。此外,CPIScore 还在多个靶点上进行了验证,证明它有能力从具有高富集率的多样化化合物库中识别抑制剂。值得注意的是,当将 CPIScore 应用于生成的重点化合物库时,它成功地鉴定出了六种强效的 ATR 小分子抑制剂,经过实验测试,其中四种小分子的抑制活性低于 10 纳摩尔。这些结果凸显了 CPIScore 在显著简化和提高药物发现过程效率方面的潜力。
{"title":"CPIScore: A Deep Learning Approach for Rapid Scoring and Interpretation of Protein-Ligand Binding Interactions.","authors":"Li Liang, Yunxin Duan, Chen Zeng, Boheng Wan, Huifeng Yao, Haichun Liu, Tao Lu, Yanmin Zhang, Yadong Chen, Jun Shen","doi":"10.1021/acs.jcim.4c01175","DOIUrl":"10.1021/acs.jcim.4c01175","url":null,"abstract":"<p><p>Protein-ligand binding affinity prediction is a crucial and challenging task in the field of drug discovery. However, traditional simulation-based computational approaches are often prohibitively time-consuming, limiting their practical utility. In this study, we introduce a novel deep learning method, CPIScore, which leverages the capabilities of Transformer and Graph Convolutional Networks (GCN) to enhance the prediction of protein-ligand binding affinity. CPIScore utilizes the Transformer architecture to capture comprehensive global contexts of protein and ligand sequences, while the GCN component effectively extracts local features from small molecular graphs. Our results demonstrate that CPIScore surpasses both traditional machine learning and other deep learning models in accuracy, achieving a Pearson's <i>r</i> of 0.74 on our test set. Furthermore, CPIScore has been validated across multiple targets, proving its ability to discern inhibitors from a diverse compound library with high enrichment rates. Notably, when applied to a generated focused library of compounds, CPIScore successfully identified six potent small-molecule inhibitors of ATR, which were tested experimentally and four small molecules exhibited inhibitory activity below ten nanomoles. These results highlight CPIScore's potential to significantly streamline and enhance the efficiency of drug discovery processes.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":" ","pages":"8809-8823"},"PeriodicalIF":5.6,"publicationDate":"2024-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142674418","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Matini-Net: Versatile Material Informatics Research Framework for Feature Engineering and Deep Neural Network Design. Matini-Net:用于特征工程和深度神经网络设计的多功能材料信息学研究框架。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-12-09 Epub Date: 2024-11-21 DOI: 10.1021/acs.jcim.4c01676
Myeonghun Lee, Taehyun Park, Kyoungmin Min

In this study, we introduced Matini-Net, which is a versatile framework for feature engineering and automated architecture design for materials informatics research using deep neural networks. Matini-Net provides the flexibility to design feature-based, graph-based, and combinations of these models, accommodating both single- and multimodal model architectures. For validation, we performed a performance evaluation on the MatBench benchmarking dataset of five properties, targeting five types of regression architectures that can be designed using Matini-Net. When applied to each of the five material property datasets, the best model performance for the various architectures exhibited R2 > 0.84. This highlights the usefulness and flexibility of Matini-Net for accelerating materials discovery. Specifically, this framework was developed for researchers with limited experience in deep learning to easily apply it to research through automated feature engineering, hyperparameter tuning, and network construction. Moreover, Matini-Net improves the model interpretability by performing an importance analysis of the selected features. We believe that by employing Matini-Net, machine and deep learning can be applied more easily and effectively in various types of materials research.

在这项研究中,我们介绍了 Matini-Net,它是一个多功能框架,用于利用深度神经网络进行材料信息学研究的特征工程和自动架构设计。Matini-Net 可灵活设计基于特征的模型、基于图的模型以及这些模型的组合,同时适用于单模态和多模态模型架构。为了进行验证,我们在 MatBench 基准数据集上对五种属性进行了性能评估,目标是使用 Matini-Net 设计的五种回归架构。当应用于五个材料属性数据集时,各种架构的最佳模型性能表现为 R2 > 0.84。这凸显了 MatiniNet 在加速材料发现方面的实用性和灵活性。具体来说,该框架是为在深度学习方面经验有限的研究人员开发的,他们可以通过自动特征工程、超参数调整和网络构建,轻松地将其应用到研究中。此外,Matini-Net 还通过对所选特征进行重要性分析来提高模型的可解释性。我们相信,通过使用 Matini-Net,机器学习和深度学习可以更轻松、更有效地应用于各类材料研究。
{"title":"Matini-Net: Versatile Material Informatics Research Framework for Feature Engineering and Deep Neural Network Design.","authors":"Myeonghun Lee, Taehyun Park, Kyoungmin Min","doi":"10.1021/acs.jcim.4c01676","DOIUrl":"10.1021/acs.jcim.4c01676","url":null,"abstract":"<p><p>In this study, we introduced Matini-Net, which is a versatile framework for feature engineering and automated architecture design for materials informatics research using deep neural networks. Matini-Net provides the flexibility to design feature-based, graph-based, and combinations of these models, accommodating both single- and multimodal model architectures. For validation, we performed a performance evaluation on the MatBench benchmarking dataset of five properties, targeting five types of regression architectures that can be designed using Matini-Net. When applied to each of the five material property datasets, the best model performance for the various architectures exhibited <i>R</i><sup>2</sup> > 0.84. This highlights the usefulness and flexibility of Matini-Net for accelerating materials discovery. Specifically, this framework was developed for researchers with limited experience in deep learning to easily apply it to research through automated feature engineering, hyperparameter tuning, and network construction. Moreover, Matini-Net improves the model interpretability by performing an importance analysis of the selected features. We believe that by employing Matini-Net, machine and deep learning can be applied more easily and effectively in various types of materials research.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":" ","pages":"8770-8783"},"PeriodicalIF":5.6,"publicationDate":"2024-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142680026","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Hierarchical Graph Attention Network with Positive and Negative Attentions for Improved Interpretability: ISA-PN.
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-12-09 DOI: 10.1021/acs.jcim.4c01035
Jinyong Park, Minhi Han, Kiwoong Lee, Sungnam Park

With the advancement of deep learning (DL) methods in chemistry and materials science, the interpretability of DL models has become a critical issue in elucidating quantitative (molecular) structure-property relationships. Although attention mechanisms have been generally employed to explain the importance of molecular substructures that contribute to molecular properties, their interpretability remains limited. In this work, we introduce a versatile segmentation method and develop an interpretable subgraph attention (ISA) network with positive and negative streams (ISA-PN) to enhance the understanding of molecular structure-property relationships. The predictive performance of the ISA models was validated using data sets for aqueous solubility, lipophilicity, and melting temperature, with a particular focus on evaluating interpretability for the aqueous solubility data set. The ISA-PN model enables the quantification of the contributions of molecular substructures through positive and negative attention scores. Comparative analyses of the ISA, ISA-PN, and GC-Net (group contribution network) models demonstrate that the ISA-PN model significantly improves interpretability while maintaining similar accuracy levels. This study highlights the efficacy of the ISA-PN model in providing meaningful insights into the contributions of molecular substructures to molecular properties, thereby enhancing the interpretability of DL models in chemical applications.

{"title":"Hierarchical Graph Attention Network with Positive and Negative Attentions for Improved Interpretability: ISA-PN.","authors":"Jinyong Park, Minhi Han, Kiwoong Lee, Sungnam Park","doi":"10.1021/acs.jcim.4c01035","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c01035","url":null,"abstract":"<p><p>With the advancement of deep learning (DL) methods in chemistry and materials science, the interpretability of DL models has become a critical issue in elucidating quantitative (molecular) structure-property relationships. Although attention mechanisms have been generally employed to explain the importance of molecular substructures that contribute to molecular properties, their interpretability remains limited. In this work, we introduce a versatile segmentation method and develop an interpretable subgraph attention (ISA) network with positive and negative streams (ISA-PN) to enhance the understanding of molecular structure-property relationships. The predictive performance of the ISA models was validated using data sets for aqueous solubility, lipophilicity, and melting temperature, with a particular focus on evaluating interpretability for the aqueous solubility data set. The ISA-PN model enables the quantification of the contributions of molecular substructures through positive and negative attention scores. Comparative analyses of the ISA, ISA-PN, and GC-Net (group contribution network) models demonstrate that the ISA-PN model significantly improves interpretability while maintaining similar accuracy levels. This study highlights the efficacy of the ISA-PN model in providing meaningful insights into the contributions of molecular substructures to molecular properties, thereby enhancing the interpretability of DL models in chemical applications.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":" ","pages":""},"PeriodicalIF":5.6,"publicationDate":"2024-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142798647","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Mapping Protein Conformational Landscapes from Crystallographic Drug Fragment Screens. 从晶体学药物片段筛选中绘制蛋白质构象图谱
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-12-09 Epub Date: 2024-11-12 DOI: 10.1021/acs.jcim.4c01380
Ammaar A Saeed, Margaret A Klureza, Doeke R Hekstra

Proteins are dynamic macromolecules. Knowledge of a protein's thermally accessible conformations is critical to determining important transitions and designing therapeutics. Accessible conformations are highly constrained by a protein's structure such that concerted structural changes due to external perturbations likely track intrinsic conformational transitions. These transitions can be thought of as paths through a conformational landscape. Crystallographic drug fragment screens are high-throughput perturbation experiments, in which thousands of crystals of a drug target are soaked with small-molecule drug precursors (fragments) and examined for fragment binding, mapping potential drug binding sites on the target protein. Here, we describe an open-source Python package, COnformational LAndscape Visualization (COLAV), to infer conformational landscapes from such large-scale crystallographic perturbation studies. We apply COLAV to drug fragment screens of two medically important systems: protein tyrosine phosphatase 1B (PTP1B), which regulates insulin signaling, and the SARS CoV-2 Main Protease (MPro). With enough fragment-bound structures, we find that such drug screens enable detailed mapping of proteins' conformational landscapes.

蛋白质是动态大分子。了解蛋白质的热可获得构象对于确定重要转变和设计疗法至关重要。可获得的构象受到蛋白质结构的高度约束,因此外部扰动导致的协同结构变化很可能会跟踪内在构象转变。这些转变可被视为构象景观的路径。晶体学药物片段筛选是一种高通量扰动实验,在这种实验中,成千上万的药物靶点晶体被小分子药物前体(片段)浸泡,并检查片段结合情况,从而绘制出靶点蛋白质上潜在的药物结合位点。在这里,我们介绍了一个开源 Python 软件包 COnformational LAndscape Visualization (COLAV),它可以从这种大规模晶体学扰动研究中推断构象景观。我们将 COLAV 应用于两个重要医学系统的药物片段筛选:调节胰岛素信号的蛋白酪氨酸磷酸酶 1B (PTP1B) 和 SARS CoV-2 主要蛋白酶 (MPro)。我们发现,有了足够的片段结合结构,此类药物筛选就能详细绘制蛋白质的构象图谱。
{"title":"Mapping Protein Conformational Landscapes from Crystallographic Drug Fragment Screens.","authors":"Ammaar A Saeed, Margaret A Klureza, Doeke R Hekstra","doi":"10.1021/acs.jcim.4c01380","DOIUrl":"10.1021/acs.jcim.4c01380","url":null,"abstract":"<p><p>Proteins are dynamic macromolecules. Knowledge of a protein's thermally accessible conformations is critical to determining important transitions and designing therapeutics. Accessible conformations are highly constrained by a protein's structure such that concerted structural changes due to external perturbations likely track intrinsic conformational transitions. These transitions can be thought of as paths through a conformational landscape. Crystallographic drug fragment screens are high-throughput perturbation experiments, in which thousands of crystals of a drug target are soaked with small-molecule drug precursors (fragments) and examined for fragment binding, mapping potential drug binding sites on the target protein. Here, we describe an open-source Python package, COnformational LAndscape Visualization (COLAV), to infer conformational landscapes from such large-scale crystallographic perturbation studies. We apply COLAV to drug fragment screens of two medically important systems: protein tyrosine phosphatase 1B (PTP1B), which regulates insulin signaling, and the SARS CoV-2 Main Protease (MPro). With enough fragment-bound structures, we find that such drug screens enable detailed mapping of proteins' conformational landscapes.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":" ","pages":"8937-8951"},"PeriodicalIF":5.6,"publicationDate":"2024-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11633654/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142612435","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Input Pose is Key to Performance of Free Energy Perturbation: Benchmarking with Monoacylglycerol Lipase. 输入姿势是自由能扰动性能的关键:以单酰甘油脂肪酶为基准。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-12-09 Epub Date: 2024-11-19 DOI: 10.1021/acs.jcim.4c01223
Donya Ohadi, Kiran Kumar, Suchitra Ravula, Renee L DesJarlais, Mark J Seierstad, Amy Y Shih, Michael D Hack, Jamie M Schiffer

Free energy perturbation (FEP) methodologies have become commonplace methods for modeling potency in hit-to-lead and lead optimization stages of drug discovery. The conformational states of the initial poses of compounds for FEP+ calculations are often set up by alignment to a cocrystal structure ligand, but it is not clear if this method provides the best result for all proteins or all ligands. Not only are ligand conformational states potential variables in modeling compound potency in FEP but also the selection of crystallographic water molecules for inclusion in the FEP input structures can impact FEP models. Here, we report the results of FEP calculations using FEP+ from Schrödinger and starting from maximum common substructure alignment and docked poses generated with an array of docking methodologies. As a benchmark data set, we use monoacylglycerol lipase (MAGL), an important clinical drug target in cancer malignancy, neurological diseases, and metabolic disorders, and a set of 17 MAGL inhibitors. We found a large variation among FEP+ correlations to experimental IC50 values depending on the method used to generate the input pose and that the inclusion of ligand-based information in the docking process, with some methods, increases the correlation between FEP+ free energies and IC50 values. Upon analysis of the initial poses, we found that the differences in FEP+ correlations stemmed from rotation around a tertiary amide bond as well as translation of the compound toward the more hydrophobic side of the MAGL pocket. FEP+ estimation improved across all pose modeling methods when hydrogen bond constraint information was added. However, simple maximum common substructure alignment in the presence of all crystallographic water molecules outperformed all other methods in correlation between estimated and experimental IC50 values. Taken together, these findings suggest that pose selection and crystallographic water inclusion greatly impact how well FEP+ estimated IC50 values align with experimental IC50 values and that modelers should benchmark a few different pose generation methodologies and different water inclusion strategies for their hit-to-lead and lead optimization drug discovery projects.

自由能扰动(FEP)方法已成为在药物发现的 "命中先导 "和 "先导优化 "阶段建立药效模型的常用方法。用于 FEP+ 计算的化合物初始姿势的构象状态通常是通过与共晶体结构配体的配位来建立的,但目前还不清楚这种方法是否能为所有蛋白质或所有配体提供最佳结果。配体构象状态不仅是 FEP 中化合物效力建模的潜在变量,而且选择晶体学水分子纳入 FEP 输入结构也会影响 FEP 模型。在此,我们报告了使用薛定谔的 FEP+ 并从最大通用亚结构配位和一系列对接方法生成的对接姿势开始进行 FEP 计算的结果。作为基准数据集,我们使用了单酰基甘油脂肪酶(MAGL)和一组 17 种 MAGL 抑制剂,单酰基甘油脂肪酶是恶性肿瘤、神经系统疾病和代谢紊乱的重要临床药物靶点。我们发现 FEP+ 与实验 IC50 值之间的相关性存在很大差异,这取决于生成输入姿势所使用的方法,而且某些方法在对接过程中加入配体信息会提高 FEP+ 自由能与 IC50 值之间的相关性。对初始姿势进行分析后,我们发现 FEP+ 相关性的差异源于围绕三级酰胺键的旋转以及化合物向 MAGL 口袋疏水性更强的一侧的平移。添加氢键约束信息后,所有姿势建模方法的 FEP+ 估计值都有所提高。不过,在存在所有晶体学水分子的情况下,简单的最大共同子结构配准在估计 IC50 值与实验 IC50 值的相关性方面优于所有其他方法。综上所述,这些研究结果表明,姿势选择和晶体学水包含在很大程度上影响了 FEP+ 估算的 IC50 值与实验 IC50 值的一致性,因此建模人员应该为他们的 "命中先导 "和 "先导优化 "药物发现项目设定一些不同姿势生成方法和不同水包含策略的基准。
{"title":"Input Pose is Key to Performance of Free Energy Perturbation: Benchmarking with Monoacylglycerol Lipase.","authors":"Donya Ohadi, Kiran Kumar, Suchitra Ravula, Renee L DesJarlais, Mark J Seierstad, Amy Y Shih, Michael D Hack, Jamie M Schiffer","doi":"10.1021/acs.jcim.4c01223","DOIUrl":"10.1021/acs.jcim.4c01223","url":null,"abstract":"<p><p>Free energy perturbation (FEP) methodologies have become commonplace methods for modeling potency in hit-to-lead and lead optimization stages of drug discovery. The conformational states of the initial poses of compounds for FEP+ calculations are often set up by alignment to a cocrystal structure ligand, but it is not clear if this method provides the best result for all proteins or all ligands. Not only are ligand conformational states potential variables in modeling compound potency in FEP but also the selection of crystallographic water molecules for inclusion in the FEP input structures can impact FEP models. Here, we report the results of FEP calculations using FEP+ from Schrödinger and starting from maximum common substructure alignment and docked poses generated with an array of docking methodologies. As a benchmark data set, we use monoacylglycerol lipase (MAGL), an important clinical drug target in cancer malignancy, neurological diseases, and metabolic disorders, and a set of 17 MAGL inhibitors. We found a large variation among FEP+ correlations to experimental IC<sub>50</sub> values depending on the method used to generate the input pose and that the inclusion of ligand-based information in the docking process, with some methods, increases the correlation between FEP+ free energies and IC<sub>50</sub> values. Upon analysis of the initial poses, we found that the differences in FEP+ correlations stemmed from rotation around a tertiary amide bond as well as translation of the compound toward the more hydrophobic side of the MAGL pocket. FEP+ estimation improved across all pose modeling methods when hydrogen bond constraint information was added. However, simple maximum common substructure alignment in the presence of all crystallographic water molecules outperformed all other methods in correlation between estimated and experimental IC<sub>50</sub> values. Taken together, these findings suggest that pose selection and crystallographic water inclusion greatly impact how well FEP+ estimated IC<sub>50</sub> values align with experimental IC<sub>50</sub> values and that modelers should benchmark a few different pose generation methodologies and different water inclusion strategies for their hit-to-lead and lead optimization drug discovery projects.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":" ","pages":"8859-8869"},"PeriodicalIF":5.6,"publicationDate":"2024-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142666471","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
ReduMixDTI: Prediction of Drug-Target Interaction with Feature Redundancy Reduction and Interpretable Attention Mechanism. ReduMixDTI:通过减少特征冗余和可解释的注意机制预测药物与目标的相互作用。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-12-09 Epub Date: 2024-11-21 DOI: 10.1021/acs.jcim.4c01554
Mingqing Liu, Xuechun Meng, Yiyang Mao, Hongqi Li, Ji Liu

Identifying drug-target interactions (DTIs) is essential for drug discovery and development. Existing deep learning approaches to DTI prediction often employ powerful feature encoders to represent drugs and targets holistically, which usually cause significant redundancy and noise by neglecting the restricted binding regions. Furthermore, many previous DTI networks ignore or simplify the complex intermolecular interaction process involving diverse binding types, which significantly limits both predictive ability and interpretability. We propose ReduMixDTI, an end-to-end model that addresses feature redundancy and explicitly captures complex local interactions for DTI prediction. In this study, drug and target features are encoded by using graph neural networks and convolutional neural networks, respectively. These features are refined from channel and spatial perspectives to enhance the representations. The proposed attention mechanism explicitly models pairwise interactions between drug and target substructures, improving the model's understanding of binding processes. In extensive comparisons with seven state-of-the-art methods, ReduMixDTI demonstrates superior performance across three benchmark data sets and external test sets reflecting real-world scenarios. Additionally, we perform comprehensive ablation studies and visualize protein attention weights to enhance the interpretability. The results confirm that ReduMixDTI serves as a robust and interpretable model for reducing feature redundancy, contributing to advances in DTI prediction.

识别药物-靶点相互作用(DTI)对于药物发现和开发至关重要。现有的 DTI 预测深度学习方法通常采用功能强大的特征编码器来整体表示药物和靶点,但由于忽略了受限的结合区域,通常会产生大量冗余和噪声。此外,以前的许多 DTI 网络都忽略或简化了涉及不同结合类型的复杂分子间相互作用过程,这大大限制了预测能力和可解释性。我们提出的 ReduMixDTI 是一种端到端模型,可解决特征冗余问题,并明确捕捉复杂的局部相互作用,用于 DTI 预测。在这项研究中,药物和目标特征分别通过图神经网络和卷积神经网络进行编码。从通道和空间角度对这些特征进行细化,以增强表征。所提出的注意机制明确地模拟了药物和靶标亚结构之间的成对相互作用,从而提高了模型对结合过程的理解。在与七种最先进方法的广泛比较中,ReduMixDTI 在三个基准数据集和反映真实世界场景的外部测试集上表现出卓越的性能。此外,我们还进行了全面的消融研究,并将蛋白质关注权重可视化,以提高可解释性。结果证实,ReduMixDTI 是减少特征冗余的稳健且可解释的模型,有助于推动 DTI 预测的发展。
{"title":"ReduMixDTI: Prediction of Drug-Target Interaction with Feature Redundancy Reduction and Interpretable Attention Mechanism.","authors":"Mingqing Liu, Xuechun Meng, Yiyang Mao, Hongqi Li, Ji Liu","doi":"10.1021/acs.jcim.4c01554","DOIUrl":"10.1021/acs.jcim.4c01554","url":null,"abstract":"<p><p>Identifying drug-target interactions (DTIs) is essential for drug discovery and development. Existing deep learning approaches to DTI prediction often employ powerful feature encoders to represent drugs and targets holistically, which usually cause significant redundancy and noise by neglecting the restricted binding regions. Furthermore, many previous DTI networks ignore or simplify the complex intermolecular interaction process involving diverse binding types, which significantly limits both predictive ability and interpretability. We propose ReduMixDTI, an end-to-end model that addresses feature redundancy and explicitly captures complex local interactions for DTI prediction. In this study, drug and target features are encoded by using graph neural networks and convolutional neural networks, respectively. These features are refined from channel and spatial perspectives to enhance the representations. The proposed attention mechanism explicitly models pairwise interactions between drug and target substructures, improving the model's understanding of binding processes. In extensive comparisons with seven state-of-the-art methods, ReduMixDTI demonstrates superior performance across three benchmark data sets and external test sets reflecting real-world scenarios. Additionally, we perform comprehensive ablation studies and visualize protein attention weights to enhance the interpretability. The results confirm that ReduMixDTI serves as a robust and interpretable model for reducing feature redundancy, contributing to advances in DTI prediction.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":" ","pages":"8952-8962"},"PeriodicalIF":5.6,"publicationDate":"2024-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142685415","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Combining a Chemical Language Model and the Structure-Activity Relationship Matrix Formalism for Generative Design of Potent Compounds with Core Structure and Substituent Modifications. 结合化学语言模型和结构-活性关系矩阵形式,设计具有核心结构和取代基修饰的强效化合物。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-12-09 Epub Date: 2024-11-15 DOI: 10.1021/acs.jcim.4c01781
Hengwei Chen, Jürgen Bajorath

In medicinal chemistry, compound optimization relies on the generation of analogue series (AS) for exploring structure-activity relationships (SARs). Potency progression is a critical criterion for advancing AS. During optimization, a key question is which analogues to synthesize next. We introduce a new computational methodology for the extension of AS with potent compounds containing both core structure and substituent modifications at multiple sites, which has been reported for the first time. The approach combines a transformer chemical language model (CLM) with a SAR matrix (SARM) methodology that identifies and organizes structurally related AS. Therefore, the SARM approach was expanded to cover multisite AS. Consensus series extracted from SARMs representing a potency gradient served as input for CLM training to extend test AS with potent analogues. Different model variants were derived and investigated. Both general and fine-tuned models correctly predicted known potent analogues at high positions in probability-based compound rankings and chemically diversified AS through core structure modifications of the generated candidate compounds and substituent replacements at multiple sites.

在药物化学中,化合物的优化依赖于生成用于探索结构-活性关系(SARs)的类似物系列(AS)。药效进展是推进 AS 的关键标准。在优化过程中,一个关键问题是下一步合成哪些类似物。我们介绍了一种新的计算方法,用于扩展包含核心结构和多个位点取代基修饰的强效化合物的 AS。该方法将转换化学语言模型(CLM)与 SAR 矩阵(SARM)方法相结合,可识别和组织结构相关的 AS。因此,SARM 方法被扩展到涵盖多位点 AS。从代表药效梯度的 SARM 中提取的共识系列作为 CLM 训练的输入,以扩展测试 AS 的强效类似物。对不同的模型变体进行了推导和研究。通用模型和微调模型都能正确预测基于概率的化合物排名中处于高位的已知强效类似物,并通过对生成的候选化合物进行核心结构修改和在多个位点进行取代基替换,实现了化学多样化的 AS。
{"title":"Combining a Chemical Language Model and the Structure-Activity Relationship Matrix Formalism for Generative Design of Potent Compounds with Core Structure and Substituent Modifications.","authors":"Hengwei Chen, Jürgen Bajorath","doi":"10.1021/acs.jcim.4c01781","DOIUrl":"10.1021/acs.jcim.4c01781","url":null,"abstract":"<p><p>In medicinal chemistry, compound optimization relies on the generation of analogue series (AS) for exploring structure-activity relationships (SARs). Potency progression is a critical criterion for advancing AS. During optimization, a key question is which analogues to synthesize next. We introduce a new computational methodology for the extension of AS with potent compounds containing both core structure and substituent modifications at multiple sites, which has been reported for the first time. The approach combines a transformer chemical language model (CLM) with a SAR matrix (SARM) methodology that identifies and organizes structurally related AS. Therefore, the SARM approach was expanded to cover multisite AS. Consensus series extracted from SARMs representing a potency gradient served as input for CLM training to extend test AS with potent analogues. Different model variants were derived and investigated. Both general and fine-tuned models correctly predicted known potent analogues at high positions in probability-based compound rankings and chemically diversified AS through core structure modifications of the generated candidate compounds and substituent replacements at multiple sites.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":" ","pages":"8784-8795"},"PeriodicalIF":5.6,"publicationDate":"2024-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142638031","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Widespread Misinterpretation of pKa Terminology for Zwitterionic Compounds and Its Consequences. 对齐聚物 pKa 术语的广泛误读及其后果。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-12-09 Epub Date: 2024-11-19 DOI: 10.1021/acs.jcim.4c01420
Jonathan W Zheng, Ivo Leito, William H Green

The acid dissociation constant (pKa), which quantifies the propensity for a solute to donate a proton to its solvent, is crucial for drug design and synthesis, environmental fate studies, chemical manufacturing, and many other fields. Unfortunately, the terminology used for describing acid-base phenomena is sometimes inconsistent, causing large potential for misinterpretation. In this work, we examine a systematic confusion underlying the definition of "acidic" and "basic" pKa values for zwitterionic compounds. Due to this confusion, some pKa data are misrepresented in data repositories, including the widely used and highly trusted ChEMBL database. Such datasets are frequently used to supply training data for pKa prediction models, and hence, confusion and errors in the data make the model performance worse. Herein, we discuss the intricacies of this issue. We make suggestions for describing acid-base phenomena, training pKa prediction models, and stewarding pKa datasets, given the high potential for confusion and potentially high impact in downstream applications.

酸解离常数(pKa)量化了溶质向其溶剂捐赠质子的倾向,对于药物设计与合成、环境归宿研究、化学制造以及许多其他领域都至关重要。遗憾的是,用于描述酸碱现象的术语有时并不一致,导致误读的可能性很大。在这项工作中,我们研究了齐聚物的 "酸性 "和 "碱性 "pKa 值定义中存在的系统性混淆。由于这种混淆,一些 pKa 数据在数据存储库(包括广泛使用且高度可信的 ChEMBL 数据库)中被误用。这些数据集经常被用来为 pKa 预测模型提供训练数据,因此数据中的混淆和误差会使模型性能变差。在此,我们将讨论这一问题的复杂性。鉴于 pKa 数据集极易混淆并可能对下游应用产生重大影响,我们就如何描述酸碱现象、训练 pKa 预测模型以及管理 pKa 数据集提出了建议。
{"title":"Widespread Misinterpretation of p<i>K</i><sub>a</sub> Terminology for Zwitterionic Compounds and Its Consequences.","authors":"Jonathan W Zheng, Ivo Leito, William H Green","doi":"10.1021/acs.jcim.4c01420","DOIUrl":"10.1021/acs.jcim.4c01420","url":null,"abstract":"<p><p>The acid dissociation constant (p<i>K</i><sub>a</sub>), which quantifies the propensity for a solute to donate a proton to its solvent, is crucial for drug design and synthesis, environmental fate studies, chemical manufacturing, and many other fields. Unfortunately, the terminology used for describing acid-base phenomena is sometimes inconsistent, causing large potential for misinterpretation. In this work, we examine a systematic confusion underlying the definition of \"acidic\" and \"basic\" p<i>K</i><sub>a</sub> values for zwitterionic compounds. Due to this confusion, some p<i>K</i><sub>a</sub> data are misrepresented in data repositories, including the widely used and highly trusted ChEMBL database. Such datasets are frequently used to supply training data for p<i>K</i><sub>a</sub> prediction models, and hence, confusion and errors in the data make the model performance worse. Herein, we discuss the intricacies of this issue. We make suggestions for describing acid-base phenomena, training p<i>K</i><sub>a</sub> prediction models, and stewarding p<i>K</i><sub>a</sub> datasets, given the high potential for confusion and potentially high impact in downstream applications.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":" ","pages":"8838-8847"},"PeriodicalIF":5.6,"publicationDate":"2024-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142666199","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","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
Journal of Chemical Information and Modeling
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