首页 > 最新文献

Journal of Chemical Information and Modeling 最新文献

英文 中文
UNIQUE: A Framework for Uncertainty Quantification Benchmarking. UNIQUE:不确定性量化基准框架。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-25 Epub Date: 2024-11-14 DOI: 10.1021/acs.jcim.4c01578
Jessica Lanini, Minh Tam Davide Huynh, Gaetano Scebba, Nadine Schneider, Raquel Rodríguez-Pérez

Machine learning (ML) models have become key in decision-making for many disciplines, including drug discovery and medicinal chemistry. ML models are generally evaluated prior to their usage in high-stakes decisions, such as compound synthesis or experimental testing. However, no ML model is robust or predictive in all real-world scenarios. Therefore, uncertainty quantification (UQ) in ML predictions has gained importance in recent years. Many investigations have focused on developing methodologies that provide accurate uncertainty estimates for ML-based predictions. Unfortunately, there is no UQ strategy that consistently provides robust estimates about model's applicability on new samples. Depending on the dataset, prediction task, and algorithm, accurate uncertainty estimations might be unfeasible to obtain. Moreover, the optimum UQ metric also varies across applications, and previous investigations have shown a lack of consistency across benchmarks. Herein, the UNIQUE (UNcertaInty QUantification bEnchmarking) framework is introduced to facilitate a comparison of UQ strategies in ML-based predictions. This Python library unifies the benchmarking of multiple UQ metrics, including the calculation of nonstandard UQ metrics (combining information from the dataset and model), and provides a comprehensive evaluation. In this framework, UQ metrics are evaluated for different application scenarios, e.g., eliminating the predictions with the lowest confidence or obtaining a reliable uncertainty estimate for an acquisition function. Taken together, this library will help to standardize UQ investigations and evaluate new methodologies.

机器学习(ML)模型已成为许多学科决策的关键,包括药物发现和药物化学。在化合物合成或实验测试等重大决策中使用 ML 模型之前,通常会对其进行评估。然而,没有一个 ML 模型在现实世界的所有情况下都是稳健的或具有预测性的。因此,近年来 ML 预测的不确定性量化(UQ)变得越来越重要。许多研究都侧重于开发能为基于 ML 的预测提供准确不确定性估计的方法。遗憾的是,目前还没有一种不确定性量化策略能始终如一地对模型在新样本上的适用性提供可靠的估计。根据数据集、预测任务和算法的不同,准确的不确定性估计可能难以获得。此外,最佳 UQ 指标也因应用而异,以往的研究表明不同基准之间缺乏一致性。在此,我们引入了 UNIQUE(UNcertaInty QUantification bEnchmarking)框架,以方便比较基于 ML 的预测中的 UQ 策略。这个 Python 库统一了多个 UQ 指标的基准测试,包括非标准 UQ 指标的计算(结合数据集和模型的信息),并提供了全面的评估。在这一框架中,UQ 指标针对不同的应用场景进行评估,例如,剔除置信度最低的预测,或为获取函数获得可靠的不确定性估计。总之,该库将有助于标准化 UQ 调查和评估新方法。
{"title":"UNIQUE: A Framework for Uncertainty Quantification Benchmarking.","authors":"Jessica Lanini, Minh Tam Davide Huynh, Gaetano Scebba, Nadine Schneider, Raquel Rodríguez-Pérez","doi":"10.1021/acs.jcim.4c01578","DOIUrl":"10.1021/acs.jcim.4c01578","url":null,"abstract":"<p><p>Machine learning (ML) models have become key in decision-making for many disciplines, including drug discovery and medicinal chemistry. ML models are generally evaluated prior to their usage in high-stakes decisions, such as compound synthesis or experimental testing. However, no ML model is robust or predictive in all real-world scenarios. Therefore, uncertainty quantification (UQ) in ML predictions has gained importance in recent years. Many investigations have focused on developing methodologies that provide accurate uncertainty estimates for ML-based predictions. Unfortunately, there is no UQ strategy that consistently provides robust estimates about model's applicability on new samples. Depending on the dataset, prediction task, and algorithm, accurate uncertainty estimations might be unfeasible to obtain. Moreover, the optimum UQ metric also varies across applications, and previous investigations have shown a lack of consistency across benchmarks. Herein, the UNIQUE (UNcertaInty QUantification bEnchmarking) framework is introduced to facilitate a comparison of UQ strategies in ML-based predictions. This Python library unifies the benchmarking of multiple UQ metrics, including the calculation of nonstandard UQ metrics (combining information from the dataset and model), and provides a comprehensive evaluation. In this framework, UQ metrics are evaluated for different application scenarios, e.g., eliminating the predictions with the lowest confidence or obtaining a reliable uncertainty estimate for an acquisition function. Taken together, this library will help to standardize UQ investigations and evaluate new methodologies.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":" ","pages":"8379-8386"},"PeriodicalIF":5.6,"publicationDate":"2024-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11600502/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142612420","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
Challenge for Deep Learning: Protein Structure Prediction of Ligand-Induced Conformational Changes at Allosteric and Orthosteric Sites. 深度学习的挑战:配体诱导异位和正位构象变化的蛋白质结构预测。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-25 Epub Date: 2024-11-01 DOI: 10.1021/acs.jcim.4c01475
Gustav Olanders, Giulia Testa, Alessandro Tibo, Eva Nittinger, Christian Tyrchan

In the realm of biomedical research, understanding the intricate structure of proteins is crucial, as these structures determine how proteins function within our bodies and interact with potential drugs. Traditionally, methods like X-ray crystallography and cryo-electron microscopy have been used to unravel these structures, but they are often challenging, time-consuming and costly. Recently, a breakthrough in computational biology has emerged with the development of deep learning algorithms capable of predicting protein structures based on their amino acid sequences (Jumper, J., et al. Nature 2021, 596, 583. Lane, T. J. Nature Methods 2023, 20, 170. Kryshtafovych, A., et al. Proteins: Structure, Function and Bioinformatics 2021, 89, 1607). This study focuses on predicting the dynamic changes that proteins undergo upon ligand binding, specifically when they bind to allosteric sites, i.e. a pocket different from the active site. Allosteric modulators are particularly important for drug discovery, as they open new avenues for designing drugs that can target proteins more effectively and with fewer side effects (Nussinov, R.; Tsai, C. J. Cell 2013, 153, 293). To study this, we curated a data set of 578 X-ray structures comprised of proteins displaying orthosteric and allosteric binding as well as a general framework to evaluate deep learning-based structure prediction methods. Our findings demonstrate the potential and current limitations of deep learning methods, such as AlphaFold2 (Jumper, J., et al. Nature 2021, 596, 583), NeuralPLexer (Qiao, Z., et al. Nat Mach Intell 2024, 6, 195), and RoseTTAFold All-Atom (Krishna, R., et al. Science 2024, 384, eadl2528) to predict not just static protein structures but also the dynamic conformational changes. Herein we show that predicting the allosteric induce-fit conformation still poses a challenge to deep learning methods as they more accurately predict the orthosteric bound conformation compared to the allosteric induce fit conformation. For AlphaFold2, we observed that conformational diversity, and sampling between the apo and holo state could be increased by modifying the MSA depth, but this did not enhance the ability to generate conformations close to the allosteric induced-fit conformation. To further support advancements in protein structure prediction field, the curated data set and evaluation framework are made publicly available.

在生物医学研究领域,了解蛋白质的复杂结构至关重要,因为这些结构决定了蛋白质在人体内的功能以及与潜在药物的相互作用。传统上,人们使用 X 射线晶体学和冷冻电镜等方法来揭示这些结构,但这些方法往往具有挑战性、耗时且成本高昂。最近,随着能够根据氨基酸序列预测蛋白质结构的深度学习算法的发展,计算生物学出现了突破性进展(Jumper, J., et al. Nature 2021, 596, 583.Lane, T. J. Nature Methods 2023, 20, 170.Kryshtafovych, A., et al. Proteins: Structure, Function and Bioinformatics 2021, 89, 1607)。这项研究的重点是预测蛋白质与配体结合后发生的动态变化,特别是当配体与异构位点(即不同于活性位点的口袋)结合时。异位调节剂对药物发现尤为重要,因为它们为设计能更有效地靶向蛋白质且副作用更小的药物开辟了新途径(Nussinov, R.; Tsai, C. J. Cell 2013, 153, 293)。为了研究这一点,我们整理了一个包含 578 个 X 射线结构的数据集,这些数据集由显示正交和异位结合的蛋白质组成,同时还整理了一个通用框架,用于评估基于深度学习的结构预测方法。我们的研究结果表明了深度学习方法的潜力和目前的局限性,如 AlphaFold2(Jumper, J., et al. Nature 2021, 596, 583)、NeuralPLexer(Qiao, Z., et al. Nat Mach Intell 2024, 6, 195)和 RoseTTAFold All-Atom(Krishna, R., et al. Science 2024, 384, eadl2528),它们不仅能预测静态蛋白质结构,还能预测动态构象变化。在这里,我们发现预测异构诱导拟合构象仍然是深度学习方法面临的挑战,因为与异构诱导拟合构象相比,深度学习方法能更准确地预测正交结合构象。对于 AlphaFold2,我们观察到构象多样性以及 apo 和 holo 状态之间的采样可以通过修改 MSA 深度来增加,但这并没有提高生成接近于异生诱导拟合构象的能力。为了进一步支持蛋白质结构预测领域的进步,我们公开了经整理的数据集和评估框架。
{"title":"Challenge for Deep Learning: Protein Structure Prediction of Ligand-Induced Conformational Changes at Allosteric and Orthosteric Sites.","authors":"Gustav Olanders, Giulia Testa, Alessandro Tibo, Eva Nittinger, Christian Tyrchan","doi":"10.1021/acs.jcim.4c01475","DOIUrl":"10.1021/acs.jcim.4c01475","url":null,"abstract":"<p><p>In the realm of biomedical research, understanding the intricate structure of proteins is crucial, as these structures determine how proteins function within our bodies and interact with potential drugs. Traditionally, methods like X-ray crystallography and cryo-electron microscopy have been used to unravel these structures, but they are often challenging, time-consuming and costly. Recently, a breakthrough in computational biology has emerged with the development of deep learning algorithms capable of predicting protein structures based on their amino acid sequences (Jumper, J., et al. <i>Nature</i> <b>2021</b>, <i>596</i>, 583. Lane, T. J. <i>Nature Methods</i> <b>2023</b>, <i>20</i>, 170. Kryshtafovych, A., et al. <i>Proteins: Structure, Function and Bioinformatics</i> <b>2021</b>, <i>89</i>, 1607). This study focuses on predicting the dynamic changes that proteins undergo upon ligand binding, specifically when they bind to allosteric sites, i.e. a pocket different from the active site. Allosteric modulators are particularly important for drug discovery, as they open new avenues for designing drugs that can target proteins more effectively and with fewer side effects (Nussinov, R.; Tsai, C. J. <i>Cell</i> <b>2013</b>, <i>153</i>, 293). To study this, we curated a data set of 578 X-ray structures comprised of proteins displaying orthosteric and allosteric binding as well as a general framework to evaluate deep learning-based structure prediction methods. Our findings demonstrate the potential and current limitations of deep learning methods, such as AlphaFold2 (Jumper, J., et al. <i>Nature</i> <b>2021</b>, <i>596</i>, 583), NeuralPLexer (Qiao, Z., et al. <i>Nat Mach Intell</i> <b>2024</b>, <i>6</i>, 195), and RoseTTAFold All-Atom (Krishna, R., et al. <i>Science</i> <b>2024</b>, <i>384</i>, eadl2528) to predict not just static protein structures but also the dynamic conformational changes. Herein we show that predicting the allosteric induce-fit conformation still poses a challenge to deep learning methods as they more accurately predict the orthosteric bound conformation compared to the allosteric induce fit conformation. For AlphaFold2, we observed that conformational diversity, and sampling between the apo and holo state could be increased by modifying the MSA depth, but this did not enhance the ability to generate conformations close to the allosteric induced-fit conformation. To further support advancements in protein structure prediction field, the curated data set and evaluation framework are made publicly available.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":" ","pages":"8481-8494"},"PeriodicalIF":5.6,"publicationDate":"2024-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142556619","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
Konnektor: A Framework for Using Graph Theory to Plan Networks for Free Energy Calculations. Konnektor:使用图论规划自由能计算网络的框架。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-25 Epub Date: 2024-11-05 DOI: 10.1021/acs.jcim.4c01710
Benjamin Ries, Richard J Gowers, Hannah M Baumann, David W H Swenson, Michael M Henry, James R B Eastwood, Irfan Alibay, David Mobley

Alchemical free energy campaigns can be planned using graph theory by building networks that contain nodes representing molecules that are connected by possible transformations as edges. We introduce Konnektor, an open-source Python package, for systematically planning, modifying, and analyzing free energy calculation networks. Konnektor is designed to aid in the drug discovery process by enabling users to easily setup free energy campaigns using complex graph manipulation methods. The package contains functions for network operations including concatenation of networks, deletion of transformations, and clustering of molecules along with a framework for combining these tools with existing network generation algorithms to enable the development of more complex methods for network generation. A comparison of the various network layout features offered is carried out using toy data sets. Additionally, Konnektor contains visualization and analysis tools, making the investigation of network features much simpler. Besides the content of the package, the paper also offers application examples, demonstrating how Konnektor can be used and how the different networks perform from a graph theory perspective. Konnektor is freely available via GitHub at https://github.com/OpenFreeEnergy/konnektor under the permissive MIT License.

炼金术自由能活动可以通过图论进行规划,方法是构建包含代表分子的节点的网络,这些节点通过可能的转化作为边连接起来。我们介绍的 Konnektor 是一个开源 Python 软件包,用于系统地规划、修改和分析自由能计算网络。Konnektor 旨在帮助用户使用复杂的图操作方法轻松建立自由能计算网络,从而为药物发现过程提供帮助。该软件包包含网络操作功能,包括网络连接、删除转换和分子聚类,以及将这些工具与现有网络生成算法相结合的框架,从而开发出更复杂的网络生成方法。使用玩具数据集对所提供的各种网络布局功能进行了比较。此外,Konnektor 还包含可视化和分析工具,使网络特征研究变得更加简单。除了软件包的内容,论文还提供了应用示例,演示如何使用 Konnektor 以及从图论角度看不同网络的性能。Konnektor 可通过 GitHub https://github.com/OpenFreeEnergy/konnektor 免费获取,采用 MIT 许可。
{"title":"Konnektor: A Framework for Using Graph Theory to Plan Networks for Free Energy Calculations.","authors":"Benjamin Ries, Richard J Gowers, Hannah M Baumann, David W H Swenson, Michael M Henry, James R B Eastwood, Irfan Alibay, David Mobley","doi":"10.1021/acs.jcim.4c01710","DOIUrl":"10.1021/acs.jcim.4c01710","url":null,"abstract":"<p><p>Alchemical free energy campaigns can be planned using graph theory by building networks that contain nodes representing molecules that are connected by possible transformations as edges. We introduce Konnektor, an open-source Python package, for systematically planning, modifying, and analyzing free energy calculation networks. Konnektor is designed to aid in the drug discovery process by enabling users to easily setup free energy campaigns using complex graph manipulation methods. The package contains functions for network operations including concatenation of networks, deletion of transformations, and clustering of molecules along with a framework for combining these tools with existing network generation algorithms to enable the development of more complex methods for network generation. A comparison of the various network layout features offered is carried out using toy data sets. Additionally, Konnektor contains visualization and analysis tools, making the investigation of network features much simpler. Besides the content of the package, the paper also offers application examples, demonstrating how Konnektor can be used and how the different networks perform from a graph theory perspective. Konnektor is freely available via GitHub at https://github.com/OpenFreeEnergy/konnektor under the permissive MIT License.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":" ","pages":"8396-8403"},"PeriodicalIF":5.6,"publicationDate":"2024-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142580824","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
AI Promoted Virtual Screening, Structure-Based Hit Optimization, and Synthesis of Novel COVID-19 S-RBD Domain Inhibitors. 人工智能促进了新型 COVID-19 S-RBD 域抑制剂的虚拟筛选、基于结构的命中优化和合成。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-25 Epub Date: 2024-11-13 DOI: 10.1021/acs.jcim.4c01110
Ioannis Gkekas, Sotirios Katsamakas, Stelios Mylonas, Theano Fotopoulou, George Ε Magoulas, Alia Cristina Tenchiu, Marios Dimitriou, Apostolos Axenopoulos, Nafsika Rossopoulou, Simona Kostova, Erich E Wanker, Theodora Katsila, Demetris Papahatjis, Vassilis G Gorgoulis, Maria Koufaki, Ioannis Karakasiliotis, Theodora Calogeropoulou, Petros Daras, Spyros Petrakis

Coronavirus disease 2019 (COVID-19) is caused by a new, highly pathogenic severe-acute-respiratory syndrome coronavirus 2 (SARS-CoV-2) that infects human cells through its transmembrane spike (S) glycoprotein. The receptor-binding domain (RBD) of the S protein interacts with the angiotensin-converting enzyme II (ACE2) receptor of the host cells. Therefore, pharmacological targeting of this interaction might prevent infection or spread of the virus. Here, we performed a virtual screening to identify small molecules that block S-ACE2 interaction. Large compound libraries were filtered for drug-like properties, promiscuity and protein-protein interaction-targeting ability based on their ADME-Tox descriptors and also to exclude pan-assay interfering compounds. A properly designed AI-based virtual screening pipeline was applied to the remaining compounds, comprising approximately 10% of the starting data sets, searching for molecules that could bind to the RBD of the S protein. All molecules were sorted according to their screening score, grouped based on their structure and postfiltered for possible interaction patterns with the ACE2 receptor, yielding 31 hits. These hit molecules were further tested for their inhibitory effect on Spike RBD/ACE2 (19-615) interaction. Six compounds inhibited the S-ACE2 interaction in a dose-dependent manner while two of them also prevented infection of human cells from a pseudotyped virus whose entry is mediated by the S protein of SARS-CoV-2. Of the two compounds, the benzimidazole derivative CKP-22 protected Vero E6 cells from infection with SARS-CoV-2, as well. Subsequent, hit-to-lead optimization of CKP-22 was effected through the synthesis of 29 new derivatives of which compound CKP-25 suppressed the Spike RBD/ACE2 (19-615) interaction, reduced the cytopathic effect of SARS-CoV-2 in Vero E6 cells (IC50 = 3.5 μM) and reduced the viral load in cell culture supernatants. Early in vitro ADME-Tox studies showed that CKP-25 does not possess biodegradation or liver metabolism issues, while isozyme-specific CYP450 experiments revealed that CKP-25 was a weak inhibitor of the CYP450 system. Moreover, CKP-25 does not elicit mutagenic effect on Escherichia coli WP2 uvrA strain. Thus, CKP-25 is considered a lead compound against COVID-19 infection.

冠状病毒病 2019(COVID-19)是由一种新型高致病性严重急性呼吸系统综合征冠状病毒 2(SARS-CoV-2)引起的,它通过跨膜尖峰(S)糖蛋白感染人体细胞。S 蛋白的受体结合域(RBD)与宿主细胞的血管紧张素转换酶 II(ACE2)受体相互作用。因此,针对这种相互作用的药理作用可能会阻止病毒的感染或传播。在此,我们进行了一次虚拟筛选,以确定能阻断 S-ACE2 相互作用的小分子。我们根据ADME-Tox描述符对大型化合物库进行了筛选,以确定其药物样特性、杂合性和蛋白-蛋白相互作用靶向能力,同时排除泛检测干扰化合物。对剩余的化合物(约占起始数据集的 10%)采用了适当设计的基于人工智能的虚拟筛选流水线,搜索能与 S 蛋白的 RBD 结合的分子。根据筛选得分对所有分子进行排序,根据其结构进行分组,并对其与 ACE2 受体可能的相互作用模式进行后过滤,最终得出 31 个命中分子。我们进一步测试了这些命中分子对 Spike RBD/ACE2 (19-615) 相互作用的抑制作用。六种化合物以剂量依赖的方式抑制了 S-ACE2 相互作用,其中两种化合物还阻止了由 SARS-CoV-2 的 S 蛋白介导的伪型病毒对人体细胞的感染。在这两种化合物中,苯并咪唑衍生物 CKP-22 也能保护 Vero E6 细胞免受 SARS-CoV-2 感染。随后,通过合成 29 种新的衍生物对 CKP-22 进行了 "一击即中 "的优化,其中化合物 CKP-25 抑制了 Spike RBD/ACE2 (19-615) 的相互作用,降低了 SARS-CoV-2 在 Vero E6 细胞中的细胞病理效应(IC50 = 3.5 μM),并减少了细胞培养上清液中的病毒载量。早期的体外 ADME-Tox 研究表明,CKP-25 不存在生物降解或肝脏代谢问题,而 CYP450 同工酶特异性实验表明,CKP-25 是 CYP450 系统的弱抑制剂。此外,CKP-25 对大肠杆菌 WP2 uvrA 菌株没有诱变作用。因此,CKP-25 被认为是抗 COVID-19 感染的先导化合物。
{"title":"AI Promoted Virtual Screening, Structure-Based Hit Optimization, and Synthesis of Novel COVID-19 S-RBD Domain Inhibitors.","authors":"Ioannis Gkekas, Sotirios Katsamakas, Stelios Mylonas, Theano Fotopoulou, George Ε Magoulas, Alia Cristina Tenchiu, Marios Dimitriou, Apostolos Axenopoulos, Nafsika Rossopoulou, Simona Kostova, Erich E Wanker, Theodora Katsila, Demetris Papahatjis, Vassilis G Gorgoulis, Maria Koufaki, Ioannis Karakasiliotis, Theodora Calogeropoulou, Petros Daras, Spyros Petrakis","doi":"10.1021/acs.jcim.4c01110","DOIUrl":"10.1021/acs.jcim.4c01110","url":null,"abstract":"<p><p>Coronavirus disease 2019 (COVID-19) is caused by a new, highly pathogenic severe-acute-respiratory syndrome coronavirus 2 (SARS-CoV-2) that infects human cells through its transmembrane spike (S) glycoprotein. The receptor-binding domain (RBD) of the S protein interacts with the angiotensin-converting enzyme II (ACE2) receptor of the host cells. Therefore, pharmacological targeting of this interaction might prevent infection or spread of the virus. Here, we performed a virtual screening to identify small molecules that block S-ACE2 interaction. Large compound libraries were filtered for drug-like properties, promiscuity and protein-protein interaction-targeting ability based on their ADME-Tox descriptors and also to exclude pan-assay interfering compounds. A properly designed AI-based virtual screening pipeline was applied to the remaining compounds, comprising approximately 10% of the starting data sets, searching for molecules that could bind to the RBD of the S protein. All molecules were sorted according to their screening score, grouped based on their structure and postfiltered for possible interaction patterns with the ACE2 receptor, yielding 31 hits. These hit molecules were further tested for their inhibitory effect on Spike RBD/ACE2 (19-615) interaction. Six compounds inhibited the S-ACE2 interaction in a dose-dependent manner while two of them also prevented infection of human cells from a pseudotyped virus whose entry is mediated by the S protein of SARS-CoV-2. Of the two compounds, the benzimidazole derivative <b>CKP-22</b> protected Vero E6 cells from infection with SARS-CoV-2, as well. Subsequent, hit-to-lead optimization of <b>CKP-22</b> was effected through the synthesis of 29 new derivatives of which compound <b>CKP-25</b> suppressed the Spike RBD/ACE2 (19-615) interaction, reduced the cytopathic effect of SARS-CoV-2 in Vero E6 cells (IC<sub>50</sub> = 3.5 μM) and reduced the viral load in cell culture supernatants. Early in vitro ADME-Tox studies showed that <b>CKP-25</b> does not possess biodegradation or liver metabolism issues, while isozyme-specific CYP450 experiments revealed that <b>CKP-25</b> was a weak inhibitor of the CYP450 system. Moreover, <b>CKP-25</b> does not elicit mutagenic effect on <i>Escherichia coli</i> WP2 uvrA strain. Thus, <b>CKP-25</b> is considered a lead compound against COVID-19 infection.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":" ","pages":"8562-8585"},"PeriodicalIF":5.6,"publicationDate":"2024-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11600510/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142612427","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
Latin American Natural Product Database (LANaPDB): An Update. 拉丁美洲天然产品数据库(LANaPDB):更新。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-25 Epub Date: 2024-11-06 DOI: 10.1021/acs.jcim.4c01560
Alejandro Gómez-García, Daniel A Acuña Jiménez, William J Zamora, Haruna L Barazorda-Ccahuana, Miguel Á Chávez-Fumagalli, Marilia Valli, Adriano D Andricopulo, Vanderlan da S Bolzani, Dionisio A Olmedo, Pablo N Solís, Marvin J Núñez, Johny R Rodríguez Pérez, Hoover A Valencia Sánchez, Héctor F Cortés Hernández, Oscar M Mosquera Martinez, José L Medina-Franco

Natural product (NP) databases are crucial tools in computer-aided drug design (CADD). Over the past decade, there has been a worldwide effort to assemble information regarding natural products (NPs) isolated and characterized in certain geographical regions. In 2023, it was published LANaPDB, and to our knowledge, this is the first attempt to gather and standardize all the NP databases of Latin America. Herein, we present and analyze in detail the contents of an updated version of LANaPDB, which includes 619 newly added compounds from Colombia, Costa Rica, and Mexico. The present version of LANaPDB has a total of 13 578 compounds, coming from ten databases of seven Latin American countries. A chemoinformatic characterization of LANaPDB was carried out, which includes the structural classification of the compounds, calculation of six physicochemical properties of pharmaceutical interest, and visualization of the chemical space by employing and comparing two different fingerprints (MACCS keys (166-bit) and Morgan2 (2048-bit)). Furthermore, additional analyses were made, and valuable information not included in the first version of LANaPDB was added, which includes structural diversity, molecular complexity, synthetic feasibility, commercial availability, and reported and predicted biological activity. In addition, the LANaPDB compounds were cross-referenced to two of the largest public chemical compound databases annotated with biological activity: ChEMBL and PubChem.

天然产物(NP)数据库是计算机辅助药物设计(CADD)的重要工具。过去十年来,全世界都在努力收集在某些地理区域分离和鉴定的天然产物(NPs)的相关信息。2023 年,LANaPDB 出版,据我们所知,这是收集拉丁美洲所有 NP 数据库并使之标准化的首次尝试。在此,我们详细介绍并分析了 LANaPDB 更新版的内容,其中包括来自哥伦比亚、哥斯达黎加和墨西哥的 619 种新化合物。目前版本的 LANaPDB 共有 13 578 个化合物,来自 7 个拉美国家的 10 个数据库。对 LANaPDB 进行了化学信息学鉴定,包括化合物的结构分类、六种药物理化性质的计算,以及通过使用和比较两种不同的指纹(MACCS 密钥(166 位)和 Morgan2(2048 位))实现化学空间的可视化。此外,还进行了其他分析,并添加了第一版 LANaPDB 未包含的有价值信息,包括结构多样性、分子复杂性、合成可行性、商业可用性以及报告和预测的生物活性。此外,LANaPDB 化合物还与两个最大的注释了生物活性的公共化合物数据库进行了交叉引用:ChEMBL 和 PubChem。
{"title":"Latin American Natural Product Database (LANaPDB): An Update.","authors":"Alejandro Gómez-García, Daniel A Acuña Jiménez, William J Zamora, Haruna L Barazorda-Ccahuana, Miguel Á Chávez-Fumagalli, Marilia Valli, Adriano D Andricopulo, Vanderlan da S Bolzani, Dionisio A Olmedo, Pablo N Solís, Marvin J Núñez, Johny R Rodríguez Pérez, Hoover A Valencia Sánchez, Héctor F Cortés Hernández, Oscar M Mosquera Martinez, José L Medina-Franco","doi":"10.1021/acs.jcim.4c01560","DOIUrl":"10.1021/acs.jcim.4c01560","url":null,"abstract":"<p><p>Natural product (NP) databases are crucial tools in computer-aided drug design (CADD). Over the past decade, there has been a worldwide effort to assemble information regarding natural products (NPs) isolated and characterized in certain geographical regions. In 2023, it was published LANaPDB, and to our knowledge, this is the first attempt to gather and standardize all the NP databases of Latin America. Herein, we present and analyze in detail the contents of an updated version of LANaPDB, which includes 619 newly added compounds from Colombia, Costa Rica, and Mexico. The present version of LANaPDB has a total of 13 578 compounds, coming from ten databases of seven Latin American countries. A chemoinformatic characterization of LANaPDB was carried out, which includes the structural classification of the compounds, calculation of six physicochemical properties of pharmaceutical interest, and visualization of the chemical space by employing and comparing two different fingerprints (MACCS keys (166-bit) and Morgan2 (2048-bit)). Furthermore, additional analyses were made, and valuable information not included in the first version of LANaPDB was added, which includes structural diversity, molecular complexity, synthetic feasibility, commercial availability, and reported and predicted biological activity. In addition, the LANaPDB compounds were cross-referenced to two of the largest public chemical compound databases annotated with biological activity: ChEMBL and PubChem.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":" ","pages":"8495-8509"},"PeriodicalIF":5.6,"publicationDate":"2024-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11600509/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142580826","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
Investigating Ligand-Mediated Conformational Dynamics of Pre-miR21: A Machine-Learning-Aided Enhanced Sampling Study. 调查配体介导的 Pre-miR21 构象动力学:机器学习辅助增强采样研究。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-25 Epub Date: 2024-11-11 DOI: 10.1021/acs.jcim.4c01166
Simone Aureli, Francesco Bellina, Valerio Rizzi, Francesco Luigi Gervasio

MicroRNAs (miRs) are short, noncoding RNA strands that regulate the activity of mRNAs by affecting the repression of protein translation, and their dysregulation has been implicated in several pathologies. miR21 in particular has been implicated in tumorigenesis and anticancer drug resistance, making it a critical target for drug design. miR21 biogenesis involves precise biochemical pathways, including the cleavage of its precursor, pre-miR21, by the enzyme Dicer. The present work investigates the conformational dynamics of pre-miR21, focusing on the role of adenine29 in switching between Dicer-binding-prone and inactive states. We also investigated the effect of L50, a cyclic peptide binder of pre-miR21 and a weak inhibitor of its processing. Using time series data and our novel collective variable-based enhanced sampling technique, OneOPES, we simulated these conformational changes and assessed the effect of L50 on the conformational plasticity of pre-miR21. Our results provide insight into peptide-induced conformational changes and pave the way for the development of a computational platform for the screening of inhibitors of pre-miR21 processing that considers RNA flexibility, a stepping stone for effective structure-based drug design, with potentially broad applications in drug discovery.

微RNA(miRs)是一种短的非编码RNA链,通过抑制蛋白质翻译来调节mRNA的活性。本研究调查了 pre-miR21 的构象动态,重点研究了腺嘌呤 29 在 Dicer 结合态和非活性态之间切换的作用。我们还研究了 L50 的影响,L50 是 pre-miR21 的环肽粘合剂,也是其加工过程的弱抑制剂。我们利用时间序列数据和基于集体变量的新型增强采样技术 OneOPES 模拟了这些构象变化,并评估了 L50 对前 miR21 构象可塑性的影响。我们的研究结果深入揭示了多肽诱导的构象变化,并为开发一种考虑到 RNA 灵活性的计算平台铺平了道路,该平台可用于筛选前 miR21 处理过程的抑制剂,是有效的基于结构的药物设计的垫脚石,在药物发现领域具有潜在的广泛应用前景。
{"title":"Investigating Ligand-Mediated Conformational Dynamics of Pre-miR21: A Machine-Learning-Aided Enhanced Sampling Study.","authors":"Simone Aureli, Francesco Bellina, Valerio Rizzi, Francesco Luigi Gervasio","doi":"10.1021/acs.jcim.4c01166","DOIUrl":"10.1021/acs.jcim.4c01166","url":null,"abstract":"<p><p>MicroRNAs (miRs) are short, noncoding RNA strands that regulate the activity of mRNAs by affecting the repression of protein translation, and their dysregulation has been implicated in several pathologies. miR21 in particular has been implicated in tumorigenesis and anticancer drug resistance, making it a critical target for drug design. miR21 biogenesis involves precise biochemical pathways, including the cleavage of its precursor, pre-miR21, by the enzyme Dicer. The present work investigates the conformational dynamics of pre-miR21, focusing on the role of adenine29 in switching between Dicer-binding-prone and inactive states. We also investigated the effect of L50, a cyclic peptide binder of pre-miR21 and a weak inhibitor of its processing. Using time series data and our novel collective variable-based enhanced sampling technique, OneOPES, we simulated these conformational changes and assessed the effect of L50 on the conformational plasticity of pre-miR21. Our results provide insight into peptide-induced conformational changes and pave the way for the development of a computational platform for the screening of inhibitors of pre-miR21 processing that considers RNA flexibility, a stepping stone for effective structure-based drug design, with potentially broad applications in drug discovery.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":" ","pages":"8595-8603"},"PeriodicalIF":5.6,"publicationDate":"2024-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11600507/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142612431","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
Exploring the Impact of Physiological C-Terminal Truncation on α-Synuclein Conformations to Unveil Mechanisms Regulating Pathological Aggregation. 探索生理 C 端截短对α-突触核蛋白构象的影响,揭示调节病理聚集的机制。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-25 Epub Date: 2024-11-06 DOI: 10.1021/acs.jcim.4c01839
Fengjuan Huang, Jiajia Yan, Huan Xu, Ying Wang, Xiaohan Zhang, Yu Zou, Jiangfang Lian, Feng Ding, Yunxiang Sun

Emerging evidence suggests that physiological C-terminal truncation of α-synuclein (αS) plays a critical role in regulating liquid-liquid phase separation and promoting amyloid aggregation, processes implicated in neurodegenerative diseases such as Parkinson's disease (PD). However, the molecular mechanisms through which C-terminal truncation influences αS conformation and modulates its aggregation remain poorly understood. In this study, we investigated the impact of C-terminal truncation on αS conformational dynamics by comparing full-length αS1-140 with truncated αS1-103 monomers using atomistic discrete molecular dynamics simulations. Our findings revealed that both αS1-140 and αS1-103 primarily adopted helical conformations around residues 7-32, while residues 36-95, located in the second half of the N-terminal and NAC domains, predominantly formed a dynamic β-sheet core. The C-terminus of αS1-140 was largely unstructured and dynamically wrapped around the β-sheet core. While residues 1-95 exhibited similar secondary structure propensities in both αS1-140 and αS1-103, the dynamic capping by the C-terminus in αS1-140 slightly enhanced β-sheet formation around residues 36-95. In contrast, key aggregation-driving regions (residues 2-9, 36-42, 45-57, and 68-78) were dynamically shielded by the C-terminus in αS1-140, reducing their exposure and potentially preventing interpeptide interactions that drive aggregation. C-terminal truncation, on the other hand, increased the exposed surface area of these aggregation-prone regions, thereby enhancing interpeptide interactions, phase separation, and amyloid aggregation. Overall, our simulations provide valuable insights into the conformational effects of C-terminal truncation on αS and its role in promoting pathological aggregation.

新的证据表明,α-突触核蛋白(αS)的生理性 C 端截断在调节液-液相分离和促进淀粉样蛋白聚集方面起着关键作用,而这些过程与帕金森病(PD)等神经退行性疾病有关。然而,人们对 C 端截短影响 αS 构象并调节其聚集的分子机制仍然知之甚少。在这项研究中,我们通过原子离散分子动力学模拟比较了全长αS1-140和截短的αS1-103单体,研究了C端截短对αS构象动力学的影响。我们的研究结果表明,αS1-140 和 αS1-103主要围绕残基 7-32 采用螺旋构象,而位于 N 端和 NAC 结构域后半部的残基 36-95 则主要形成动态的 β 片状核心。αS1-140 的 C 端在很大程度上是非结构化的,并动态地包裹在 β 片状核心周围。虽然残基 1-95 在 αS1-140 和 αS1-103 中表现出相似的二级结构倾向性,但 αS1-140 中 C 端的动态封顶略微增强了残基 36-95 周围的 β 片层形成。与此相反,αS1-140 中的 C 端动态屏蔽了关键的聚集驱动区(残基 2-9、36-42、45-57 和 68-78),减少了它们的暴露,并有可能防止肽间相互作用导致聚集。另一方面,C端截短增加了这些易聚集区域的暴露表面积,从而增强了肽间相互作用、相分离和淀粉样蛋白聚集。总之,我们的模拟为了解 C 端截短对αS 的构象效应及其在促进病理聚集中的作用提供了宝贵的见解。
{"title":"Exploring the Impact of Physiological C-Terminal Truncation on α-Synuclein Conformations to Unveil Mechanisms Regulating Pathological Aggregation.","authors":"Fengjuan Huang, Jiajia Yan, Huan Xu, Ying Wang, Xiaohan Zhang, Yu Zou, Jiangfang Lian, Feng Ding, Yunxiang Sun","doi":"10.1021/acs.jcim.4c01839","DOIUrl":"10.1021/acs.jcim.4c01839","url":null,"abstract":"<p><p>Emerging evidence suggests that physiological C-terminal truncation of α-synuclein (αS) plays a critical role in regulating liquid-liquid phase separation and promoting amyloid aggregation, processes implicated in neurodegenerative diseases such as Parkinson's disease (PD). However, the molecular mechanisms through which C-terminal truncation influences αS conformation and modulates its aggregation remain poorly understood. In this study, we investigated the impact of C-terminal truncation on αS conformational dynamics by comparing full-length αS<sub>1-140</sub> with truncated αS<sub>1-103</sub> monomers using atomistic discrete molecular dynamics simulations. Our findings revealed that both αS<sub>1-140</sub> and αS<sub>1-103</sub> primarily adopted helical conformations around residues 7-32, while residues 36-95, located in the second half of the N-terminal and NAC domains, predominantly formed a dynamic β-sheet core. The C-terminus of αS<sub>1-140</sub> was largely unstructured and dynamically wrapped around the β-sheet core. While residues 1-95 exhibited similar secondary structure propensities in both αS<sub>1-140</sub> and αS<sub>1-103</sub>, the dynamic capping by the C-terminus in αS<sub>1-140</sub> slightly enhanced β-sheet formation around residues 36-95. In contrast, key aggregation-driving regions (residues 2-9, 36-42, 45-57, and 68-78) were dynamically shielded by the C-terminus in αS<sub>1-140</sub>, reducing their exposure and potentially preventing interpeptide interactions that drive aggregation. C-terminal truncation, on the other hand, increased the exposed surface area of these aggregation-prone regions, thereby enhancing interpeptide interactions, phase separation, and amyloid aggregation. Overall, our simulations provide valuable insights into the conformational effects of C-terminal truncation on αS and its role in promoting pathological aggregation.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":" ","pages":"8616-8627"},"PeriodicalIF":5.6,"publicationDate":"2024-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11588551/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142580822","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
AABBA Graph Kernel: Atom-Atom, Bond-Bond, and Bond-Atom Autocorrelations for Machine Learning
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-24 DOI: 10.1021/acs.jcim.4c0158310.1021/acs.jcim.4c01583
Lucía Morán-González, Jørn Eirik Betten, Hannes Kneiding and 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.

{"title":"AABBA Graph Kernel: Atom-Atom, Bond-Bond, and Bond-Atom Autocorrelations for Machine Learning","authors":"Lucía Morán-González,&nbsp;Jørn Eirik Betten,&nbsp;Hannes Kneiding and David Balcells*,&nbsp;","doi":"10.1021/acs.jcim.4c0158310.1021/acs.jcim.4c01583","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c01583https://doi.org/10.1021/acs.jcim.4c01583","url":null,"abstract":"<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":"64 23","pages":"8756–8769 8756–8769"},"PeriodicalIF":5.6,"publicationDate":"2024-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/epdf/10.1021/acs.jcim.4c01583","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142850554","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
Effects of All-Atom and Coarse-Grained Molecular Mechanics Force Fields on Amyloid Peptide Assembly: The Case of a Tau K18 Monomer
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-23 DOI: 10.1021/acs.jcim.4c0144810.1021/acs.jcim.4c01448
Xibing He, Viet Hoang Man, Jie Gao and Junmei Wang*, 

To propose new mechanism-based therapeutics for Alzheimer’s disease (AD), it is crucial to study the kinetics and oligomerization/aggregation mechanisms of the hallmark tau proteins, which have various isoforms and are intrinsically disordered. In this study, multiple all-atom (AA) and coarse-grained (CG) force fields (FFs) have been benchmarked on molecular dynamics (MD) simulations of K18 tau (M243–E372), which is a truncated form (130 residues) of full-length tau (441 residues). FF19SB is first excluded because the dynamics are too slow, and the conformations are too stable. All other benchmarked AAFFs (Charmm36m, FF14SB, Gromos54A7, and OPLS-AA) and CGFFs (Martini3 and Sirah2.0) exhibit a trend of shrinking K18 tau into compact structures with the radius of gyration (ROG) around 2.0 nm, which is much smaller than the experimental value of 3.8 nm, within 200 ns of AA-MD or 2000 ns of CG-MD. Gromos54A7, OPLS-AA, and Martini3 shrink much faster than the other FFs. To perform meaningful postanalysis of various properties, we propose a strategy of selecting snapshots with 2.5 < ROG < 4.5 nm, instead of using all sampled snapshots. The calculated chemical shifts of all C, CA, and CB atoms have very good and close root-mean-square error (RMSE) values, while Charmm36m and Sirah2.0 exhibit better chemical shifts of N than other FFs. Comparing the calculated distributions of the distance between the CA atoms of CYS291 and CYS322 with the results of the FRET experiment demonstrates that Charmm36m is a perfect match with the experiment while other FFs exhibit limitations. In summary, Charmm36m is recommended as the best AAFF, and Sirah2.0 is recommended as an excellent CGFF for simulating tau K18.

{"title":"Effects of All-Atom and Coarse-Grained Molecular Mechanics Force Fields on Amyloid Peptide Assembly: The Case of a Tau K18 Monomer","authors":"Xibing He,&nbsp;Viet Hoang Man,&nbsp;Jie Gao and Junmei Wang*,&nbsp;","doi":"10.1021/acs.jcim.4c0144810.1021/acs.jcim.4c01448","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c01448https://doi.org/10.1021/acs.jcim.4c01448","url":null,"abstract":"<p >To propose new mechanism-based therapeutics for Alzheimer’s disease (AD), it is crucial to study the kinetics and oligomerization/aggregation mechanisms of the hallmark tau proteins, which have various isoforms and are intrinsically disordered. In this study, multiple all-atom (AA) and coarse-grained (CG) force fields (FFs) have been benchmarked on molecular dynamics (MD) simulations of K18 tau (M243–E372), which is a truncated form (130 residues) of full-length tau (441 residues). FF19SB is first excluded because the dynamics are too slow, and the conformations are too stable. All other benchmarked AAFFs (Charmm36m, FF14SB, Gromos54A7, and OPLS-AA) and CGFFs (Martini3 and Sirah2.0) exhibit a trend of shrinking K18 tau into compact structures with the radius of gyration (ROG) around 2.0 nm, which is much smaller than the experimental value of 3.8 nm, within 200 ns of AA-MD or 2000 ns of CG-MD. Gromos54A7, OPLS-AA, and Martini3 shrink much faster than the other FFs. To perform meaningful postanalysis of various properties, we propose a strategy of selecting snapshots with 2.5 &lt; ROG &lt; 4.5 nm, instead of using all sampled snapshots. The calculated chemical shifts of all C, CA, and CB atoms have very good and close root-mean-square error (RMSE) values, while Charmm36m and Sirah2.0 exhibit better chemical shifts of N than other FFs. Comparing the calculated distributions of the distance between the CA atoms of CYS291 and CYS322 with the results of the FRET experiment demonstrates that Charmm36m is a perfect match with the experiment while other FFs exhibit limitations. In summary, Charmm36m is recommended as the best AAFF, and Sirah2.0 is recommended as an excellent CGFF for simulating tau K18.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":"64 23","pages":"8880–8891 8880–8891"},"PeriodicalIF":5.6,"publicationDate":"2024-11-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142842735","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
Pairing a Global Optimization Algorithm with EXAFS to Characterize Lanthanide Structure in Solution 将全局优化算法与 EXAFS 配对,以确定溶液中的镧系元素结构特征
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-22 DOI: 10.1021/acs.jcim.4c0176910.1021/acs.jcim.4c01769
Thomas J. Summers, Difan Zhang, Josiane A. Sobrinho, Ana de Bettencourt-Dias, Roger Rousseau, Vassiliki-Alexandra Glezakou* and David C. Cantu*, 

Ensemble-average sampling of structures from ab initio molecular dynamics (AIMD) simulations can be used to predict theoretical extended X-ray absorption fine structure (EXAFS) signals that closely match experimental spectra. However, AIMD simulations are time-consuming and resource-intensive, particularly for solvated lanthanide ions, which often form multiple nonrigid geometries with high coordination numbers. To accelerate the characterization of lanthanide structures in solution, we employed the Northwest Potential Energy Surface Search Engine (NWPEsSe), an adaptive-learning global optimization algorithm, to efficiently screen first-shell structures. As case studies, we examine two systems: Eu(NO3)3 dissolved in acetonitrile with a terpyridine ligand (terpyNO2), and Nd(NO3)3 dissolved in acetonitrile. The theoretical spectra for structures identified by NWPEsSe were compared to both experimental and AIMD-derived EXAFS spectra. The NWPEsSe algorithm successfully identified the proper solvation structure for both Eu(NO3)3(terpyNO2) and Nd(NO3)(acetonitrile)3, with the calculated EXAFS signals closely matching the experimental spectra for the Eu-ligand complex and showing good similarity for the Nd salt; the better agreement with the ligand-containing structure is attributed to a less dynamic coordination environment due to the rigid ligand. The key advantage of the global optimization algorithm lies in its ability to sample the coordination environment across the potential energy surface and reduce the time required to identify structures from generally a month to within a week. Additionally, this approach is versatile and can be adapted to characterize main-group metal complexes.

{"title":"Pairing a Global Optimization Algorithm with EXAFS to Characterize Lanthanide Structure in Solution","authors":"Thomas J. Summers,&nbsp;Difan Zhang,&nbsp;Josiane A. Sobrinho,&nbsp;Ana de Bettencourt-Dias,&nbsp;Roger Rousseau,&nbsp;Vassiliki-Alexandra Glezakou* and David C. Cantu*,&nbsp;","doi":"10.1021/acs.jcim.4c0176910.1021/acs.jcim.4c01769","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c01769https://doi.org/10.1021/acs.jcim.4c01769","url":null,"abstract":"<p ><i>Ensemble</i>-average sampling of structures from <i>ab initio</i> molecular dynamics (AIMD) simulations can be used to predict theoretical extended X-ray absorption fine structure (EXAFS) signals that closely match experimental spectra. However, AIMD simulations are time-consuming and resource-intensive, particularly for solvated lanthanide ions, which often form multiple nonrigid geometries with high coordination numbers. To accelerate the characterization of lanthanide structures in solution, we employed the Northwest Potential Energy Surface Search Engine (NWPEsSe), an adaptive-learning global optimization algorithm, to efficiently screen first-shell structures. As case studies, we examine two systems: Eu(NO<sub>3</sub>)<sub>3</sub> dissolved in acetonitrile with a terpyridine ligand (terpyNO<sub>2</sub>), and Nd(NO<sub>3</sub>)<sub>3</sub> dissolved in acetonitrile. The theoretical spectra for structures identified by NWPEsSe were compared to both experimental and AIMD-derived EXAFS spectra. The NWPEsSe algorithm successfully identified the proper solvation structure for both Eu(NO<sub>3</sub>)<sub>3</sub>(terpyNO<sub>2</sub>) and Nd(NO<sub>3</sub>)(acetonitrile)<sub>3</sub>, with the calculated EXAFS signals closely matching the experimental spectra for the Eu-ligand complex and showing good similarity for the Nd salt; the better agreement with the ligand-containing structure is attributed to a less dynamic coordination environment due to the rigid ligand. The key advantage of the global optimization algorithm lies in its ability to sample the coordination environment across the potential energy surface and reduce the time required to identify structures from generally a month to within a week. Additionally, this approach is versatile and can be adapted to characterize main-group metal complexes.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":"64 23","pages":"8926–8936 8926–8936"},"PeriodicalIF":5.6,"publicationDate":"2024-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142842721","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