Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Anal Chem ; 96(15): 5763-5770, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38564366

RESUMO

Library matching by comparing carbon-13 nuclear magnetic resonance (13C NMR) spectra with spectral data in the library is a crucial method for compound identification. In our previous paper, we introduced a deep contrastive learning system called CReSS, which used a library that contained more structures. However, CReSS has two limitations: there were no unknown structures in the library, and a redundant library reduces the structure-elucidation accuracy. Herein, we replaced the oversize traditional libraries with focused libraries containing a small number of molecules. A previously generative model, CMGNet, was used to generate focused libraries for CReSS. The combined model achieved a Top-10 accuracy of 54.03% when tested on 6,471 13C NMR spectra. In comparison, CReSS with a random reference structure library achieved an accuracy of only 9.17%. Furthermore, to expand the advantages of the focused libraries, we proposed SAmpRNN, which is a recurrent neural network (RNN). With the large focused library amplified by SAmpRNN, the structure-identification accuracy of the model increased in 70.0% of the 30 random example cases. In general, cross-modal retrieval between 13C NMR spectra and structures based on focused libraries (CFLS) achieved high accuracy and provided more accurate candidate structures than traditional libraries for compound identification.


Assuntos
Imageamento por Ressonância Magnética , Espectroscopia de Ressonância Magnética
2.
Phys Chem Chem Phys ; 26(13): 10323-10335, 2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38501198

RESUMO

Ribonucleic acid (RNA)-ligand interactions play a pivotal role in a wide spectrum of biological processes, ranging from protein biosynthesis to cellular reproduction. This recognition has prompted the broader acceptance of RNA as a viable candidate for drug targets. Delving into the atomic-scale understanding of RNA-ligand interactions holds paramount importance in unraveling intricate molecular mechanisms and further contributing to RNA-based drug discovery. Computational approaches, particularly molecular docking, offer an efficient way of predicting the interactions between RNA and small molecules. However, the accuracy and reliability of these predictions heavily depend on the performance of scoring functions (SFs). In contrast to the majority of SFs used in RNA-ligand docking, the end-point binding free energy calculation methods, such as molecular mechanics/generalized Born surface area (MM/GBSA) and molecular mechanics/Poisson Boltzmann surface area (MM/PBSA), stand as theoretically more rigorous approaches. Yet, the evaluation of their effectiveness in predicting both binding affinities and binding poses within RNA-ligand systems remains unexplored. This study first reported the performance of MM/PBSA and MM/GBSA with diverse solvation models, interior dielectric constants (εin) and force fields in the context of binding affinity prediction for 29 RNA-ligand complexes. MM/GBSA is based on short (5 ns) molecular dynamics (MD) simulations in an explicit solvent with the YIL force field; the GBGBn2 model with higher interior dielectric constant (εin = 12, 16 or 20) yields the best correlation (Rp = -0.513), which outperforms the best correlation (Rp = -0.317, rDock) offered by various docking programs. Then, the efficacy of MM/GBSA in identifying the near-native binding poses from the decoys was assessed based on 56 RNA-ligand complexes. However, it is evident that MM/GBSA has limitations in accurately predicting binding poses for RNA-ligand systems, particularly compared with notably proficient docking programs like rDock and PLANTS. The best top-1 success rate achieved by MM/GBSA rescoring is 39.3%, which falls below the best results given by docking programs (50%, PLNATS). This study represents the first evaluation of MM/PBSA and MM/GBSA for RNA-ligand systems and is expected to provide valuable insights into their successful application to RNA targets.


Assuntos
Simulação de Dinâmica Molecular , RNA , Simulação de Acoplamento Molecular , Ligantes , Reprodutibilidade dos Testes , Ligação Proteica , Termodinâmica , Sítios de Ligação
3.
J Chem Theory Comput ; 20(3): 1465-1478, 2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-38300792

RESUMO

Multisite λ-dynamics (MSLD) is a highly efficient binding free energy calculation method that samples multiple ligands in a single round by assigning different λ values to the alchemical part of each ligand. This method holds great promise for lead optimization (LO) in drug discovery. However, the complex data preparation and simulation process limits its widespread application in diverse protein-ligand systems. To address this challenge, we developed a comprehensive, open-source, and automated workflow for MSLD calculations based on the BLaDE dynamics engine. This workflow incorporates the Ligand Internal and Cartesian coordinate reconstruction-based alignment algorithm (LIC-align) and an optimized maximum common substructure (MCS) search algorithm to accurately generate MSLD multiple topologies with ideal perturbation patterns. Furthermore, our workflow is highly modularized, allowing straightforward integration and extension of various simulation techniques, and is highly accessible to nonexperts. This workflow was validated by calculating the relative binding free energies of large-scale congeneric ligands, many of which have large perturbing groups. The agreement between the calculations and experiments was excellent, with an average unsigned error of 1.08 ± 0.47 kcal/mol. More than 57.1% of the ligands had an error of less than 1.0 kcal/mol, and the perturbations of 6 targets were fully connected via the calculations, while those of 2 targets were connected via both calculations and experimental data. The Pearson correlation coefficient reached 0.88, indicating that the MSLD workflow provides accurate predictions that can guide lead optimization in drug discovery. We also examined the impact of single-site versus multisite perturbations, ligand grouping by perturbing group size, and the position of the anchor atom on the MSLD performance. By integrating our proposed LIC-align and optimized MCS search algorithm along with the coping strategies to handle challenging molecular substructures, our workflow can handle many realistic scenarios more reasonably than all previously published methods. Moreover, we observed that our MSLD workflow achieved similar accuracy to free energy perturbation (FEP) while improving computational efficiency by over 1 order of magnitude in speedup. These findings provide valuable insights and strategies for further MSLD development, making MSLD a competitive tool for lead optimization.


Assuntos
Simulação de Dinâmica Molecular , Proteínas , Termodinâmica , Ligantes , Fluxo de Trabalho , Proteínas/química , Ligação Proteica
4.
J Chem Inf Model ; 64(4): 1213-1228, 2024 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-38302422

RESUMO

Deep learning-based de novo molecular design has recently gained significant attention. While numerous DL-based generative models have been successfully developed for designing novel compounds, the majority of the generated molecules lack sufficiently novel scaffolds or high drug-like profiles. The aforementioned issues may not be fully captured by commonly used metrics for the assessment of molecular generative models, such as novelty, diversity, and quantitative estimation of the drug-likeness score. To address these limitations, we proposed a genetic algorithm-guided generative model called GARel (genetic algorithm-based receptor-ligand interaction generator), a novel framework for training a DL-based generative model to produce drug-like molecules with novel scaffolds. To efficiently train the GARel model, we utilized dense net to update the parameters based on molecules with novel scaffolds and drug-like features. To demonstrate the capability of the GARel model, we used it to design inhibitors for three targets: AA2AR, EGFR, and SARS-Cov2. The results indicate that GARel-generated molecules feature more diverse and novel scaffolds and possess more desirable physicochemical properties and favorable docking scores. Compared with other generative models, GARel makes significant progress in balancing novelty and drug-likeness, providing a promising direction for the further development of DL-based de novo design methodology with potential impacts on drug discovery.


Assuntos
Desenho de Fármacos , RNA Viral , Ligantes , Algoritmos , Descoberta de Drogas
5.
Chem Sci ; 15(4): 1449-1471, 2024 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-38274053

RESUMO

The expertise accumulated in deep neural network-based structure prediction has been widely transferred to the field of protein-ligand binding pose prediction, thus leading to the emergence of a variety of deep learning-guided docking models for predicting protein-ligand binding poses without relying on heavy sampling. However, their prediction accuracy and applicability are still far from satisfactory, partially due to the lack of protein-ligand binding complex data. To this end, we create a large-scale complex dataset containing ∼9 M protein-ligand docking complexes for pre-training, and propose CarsiDock, the first deep learning-guided docking approach that leverages pre-training of millions of predicted protein-ligand complexes. CarsiDock contains two main stages, i.e., a deep learning model for the prediction of protein-ligand atomic distance matrices, and a translation, rotation and torsion-guided geometry optimization procedure to reconstruct the matrices into a credible binding pose. The pre-training and multiple innovative architectural designs facilitate the dramatically improved docking accuracy of our approach over the baselines in terms of multiple docking scenarios, thereby contributing to its outstanding early recognition performance in several retrospective virtual screening campaigns. Further explorations demonstrate that CarsiDock can not only guarantee the topological reliability of the binding poses but also successfully reproduce the crucial interactions in crystalized structures, highlighting its superior applicability.

6.
Research (Wash D C) ; 7: 0292, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38213662

RESUMO

Deep learning (DL)-driven efficient synthesis planning may profoundly transform the paradigm for designing novel pharmaceuticals and materials. However, the progress of many DL-assisted synthesis planning (DASP) algorithms has suffered from the lack of reliable automated pathway evaluation tools. As a critical metric for evaluating chemical reactions, accurate prediction of reaction yields helps improve the practicality of DASP algorithms in the real-world scenarios. Currently, accurately predicting yields of interesting reactions still faces numerous challenges, mainly including the absence of high-quality generic reaction yield datasets and robust generic yield predictors. To compensate for the limitations of high-throughput yield datasets, we curated a generic reaction yield dataset containing 12 reaction categories and rich reaction condition information. Subsequently, by utilizing 2 pretraining tasks based on chemical reaction masked language modeling and contrastive learning, we proposed a powerful bidirectional encoder representations from transformers (BERT)-based reaction yield predictor named Egret. It achieved comparable or even superior performance to the best previous models on 4 benchmark datasets and established state-of-the-art performance on the newly curated dataset. We found that reaction-condition-based contrastive learning enhances the model's sensitivity to reaction conditions, and Egret is capable of capturing subtle differences between reactions involving identical reactants and products but different reaction conditions. Furthermore, we proposed a new scoring function that incorporated Egret into the evaluation of multistep synthesis routes. Test results showed that yield-incorporated scoring facilitated the prioritization of literature-supported high-yield reaction pathways for target molecules. In addition, through meta-learning strategy, we further improved the reliability of the model's prediction for reaction types with limited data and lower data quality. Our results suggest that Egret holds the potential to become an essential component of the next-generation DASP tools.

7.
Nat Protoc ; 19(4): 1105-1121, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38263521

RESUMO

Lead optimization is a crucial step in the drug discovery process, which aims to design potential drug candidates from biologically active hits. During lead optimization, active hits undergo modifications to improve their absorption, distribution, metabolism, excretion and toxicity (ADMET) profiles. Medicinal chemists face key questions regarding which compound(s) should be synthesized next and how to balance multiple ADMET properties. Reliable transformation rules from multiple experimental analyses are critical to improve this decision-making process. We developed OptADMET ( https://cadd.nscc-tj.cn/deploy/optadmet/ ), an integrated web-based platform that provides chemical transformation rules for 32 ADMET properties and leverages prior experimental data for lead optimization. The multiproperty transformation rule database contains a total of 41,779 validated transformation rules generated from the analysis of 177,191 reliable experimental datasets. Additionally, 146,450 rules were generated by analyzing 239,194 molecular data predictions. OptADMET provides the ADMET profiles of all optimized molecules from the queried molecule and enables the prediction of desirable substructure transformations and subsequent validation of drug candidates. OptADMET is based on matched molecular pairs analysis derived from synthetic chemistry, thus providing improved practicality over other methods. OptADMET is designed for use by both experimental and computational scientists.


Assuntos
Descoberta de Drogas , Internet , Bases de Dados Factuais
8.
J Chem Inf Model ; 63(24): 7617-7627, 2023 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-38079566

RESUMO

The application of Explainable Artificial Intelligence (XAI) in the field of chemistry has garnered growing interest for its potential to justify the prediction of black-box machine learning models and provide actionable insights. We first survey a range of XAI techniques adapted for chemical applications and categorize them based on the technical details of each methodology. We then present a few case studies to illustrate the practical utility of XAI, such as identifying carcinogenic molecules and guiding molecular optimizations, in order to provide chemists with concrete examples of ways to take full advantage of XAI-augmented machine learning for chemistry. Despite the initial success of XAI in chemistry, we still face the challenges of developing more reliable explanations, assuring robustness against adversarial actions, and customizing the explanation for different applications and needs of the diverse scientific community. Finally, we discuss the emerging role of large language models like GPT in generating natural language explanations and discusses the specific challenges associated with them. We advocate that addressing the aforementioned challenges and actively embracing new techniques may contribute to establishing machine learning as an indispensable technique for chemistry in this digital era.


Assuntos
Inteligência Artificial , Aprendizado de Máquina , Idioma
9.
Chem Sci ; 14(43): 12166-12181, 2023 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-37969589

RESUMO

Contemporary structure-based molecular generative methods have demonstrated their potential to model the geometric and energetic complementarity between ligands and receptors, thereby facilitating the design of molecules with favorable binding affinity and target specificity. Despite the introduction of deep generative models for molecular generation, the atom-wise generation paradigm that partially contradicts chemical intuition limits the validity and synthetic accessibility of the generated molecules. Additionally, the dependence of deep learning models on large-scale structural data has hindered their adaptability across different targets. To overcome these challenges, we present a novel search-based framework, 3D-MCTS, for structure-based de novo drug design. Distinct from prevailing atom-centric methods, 3D-MCTS employs a fragment-based molecular editing strategy. The fragments decomposed from small-molecule drugs are recombined under predefined retrosynthetic rules, offering improved drug-likeness and synthesizability, overcoming the inherent limitations of atom-based approaches. Leveraging multi-threaded parallel simulations combined with a real-time energy constraint-based pruning strategy, 3D-MCTS achieves remarkable efficiency. At a fixed computational cost, it outperforms other state-of-the-art (SOTA) methods by producing molecules with enhanced binding affinity. Furthermore, its fragment-based approach ensures the generation of more dependable binding conformations, exhibiting a success rate 43.6% higher than that of other SOTAs. This advantage becomes even more pronounced when handling targets that significantly deviate from the training dataset. 3D-MCTS is capable of achieving thirty times more hits with high binding affinity than traditional virtual screening methods, which demonstrates the superior ability of 3D-MCTS to explore chemical space. Moreover, the flexibility of our framework makes it easy to incorporate domain knowledge during the process, thereby enabling the generation of molecules with desirable pharmacophores and enhanced binding affinity. The adaptability of 3D-MCTS is further showcased in metalloprotein applications, highlighting its potential across various drug design scenarios.

10.
Research (Wash D C) ; 6: 0231, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37849643

RESUMO

Effective synthesis planning powered by deep learning (DL) can significantly accelerate the discovery of new drugs and materials. However, most DL-assisted synthesis planning methods offer either none or very limited capability to recommend suitable reaction conditions (RCs) for their reaction predictions. Currently, the prediction of RCs with a DL framework is hindered by several factors, including: (a) lack of a standardized dataset for benchmarking, (b) lack of a general prediction model with powerful representation, and (c) lack of interpretability. To address these issues, we first created 2 standardized RC datasets covering a broad range of reaction classes and then proposed a powerful and interpretable Transformer-based RC predictor named Parrot. Through careful design of the model architecture, pretraining method, and training strategy, Parrot improved the overall top-3 prediction accuracy on catalysis, solvents, and other reagents by as much as 13.44%, compared to the best previous model on a newly curated dataset. Additionally, the mean absolute error of the predicted temperatures was reduced by about 4 °C. Furthermore, Parrot manifests strong generalization capacity with superior cross-chemical-space prediction accuracy. Attention analysis indicates that Parrot effectively captures crucial chemical information and exhibits a high level of interpretability in the prediction of RCs. The proposed model Parrot exemplifies how modern neural network architecture when appropriately pretrained can be versatile in making reliable, generalizable, and interpretable recommendation for RCs even when the underlying training dataset may still be limited in diversity.

11.
J Chem Inf Model ; 63(20): 6169-6176, 2023 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-37820365

RESUMO

Target identification and bioactivity prediction are critical steps in the drug discovery process. Here we introduce CODD-Pred (COmprehensive Drug Design Predictor), an online web server with well-curated data sets from the GOSTAR database, which is designed with a dual purpose of predicting potential protein drug targets and computing bioactivity values of small molecules. We first designed a double molecular graph perception (DMGP) framework for target prediction based on a large library of 646 498 small molecules interacting with 640 human targets. The framework achieved a top-5 accuracy of over 80% for hitting at least one target on both external validation sets. Additionally, its performance on the external validation set comprising 200 molecules surpassed that of four existing target prediction servers. Second, we collected 56 targets closely related to the occurrence and development of cancer, metabolic diseases, and inflammatory immune diseases and developed a multi-model self-validation activity prediction (MSAP) framework that enables accurate bioactivity quantification predictions for small-molecule ligands of these 56 targets. CODD-Pred is a handy tool for rapid evaluation and optimization of small molecules with specific target activity. CODD-Pred is freely accessible at http://codd.iddd.group/.


Assuntos
Computadores , Proteínas , Humanos , Proteínas/química , Desenho de Fármacos , Descoberta de Drogas , Bases de Dados Factuais
12.
Brief Bioinform ; 24(5)2023 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-37738401

RESUMO

Cracking the entangling code of protein-ligand interaction (PLI) is of great importance to structure-based drug design and discovery. Different physical and biochemical representations can be used to describe PLI such as energy terms and interaction fingerprints, which can be analyzed by machine learning (ML) algorithms to create ML-based scoring functions (MLSFs). Here, we propose the ML-based PLI capturer (ML-PLIC), a web platform that automatically characterizes PLI and generates MLSFs to identify the potential binders of a specific protein target through virtual screening (VS). ML-PLIC comprises five modules, including Docking for ligand docking, Descriptors for PLI generation, Modeling for MLSF training, Screening for VS and Pipeline for the integration of the aforementioned functions. We validated the MLSFs constructed by ML-PLIC in three benchmark datasets (Directory of Useful Decoys-Enhanced, Active as Decoys and TocoDecoy), demonstrating accuracy outperforming traditional docking tools and competitive performance to the deep learning-based SF, and provided a case study of the Serine/threonine-protein kinase WEE1 in which MLSFs were developed by using the ML-based VS pipeline in ML-PLIC. Underpinning the latest version of ML-PLIC is a powerful platform that incorporates physical and biological knowledge about PLI, leveraging PLI characterization and MLSF generation into the design of structure-based VS pipeline. The ML-PLIC web platform is now freely available at http://cadd.zju.edu.cn/plic/.


Assuntos
Algoritmos , Benchmarking , Ligantes , Desenho de Fármacos , Aprendizado de Máquina
13.
Anal Methods ; 15(36): 4692-4699, 2023 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-37675461

RESUMO

By adjusting the reactants and reaction conditions, the particle size and surface state of fluorescent carbon dots (CDs) can be controlled, and CDs with different photoluminescence colors can be finally prepared. However, this multi-step procedure is relatively time-consuming and complex. Therefore, it is of great significance to explore a more convenient and efficient preparation route. In this paper, SA (P-aminobenzenesulfonic acid) and οPD (o-phenylenediamine) were used as precursors, and water and ethanol were used as reaction solvents. By adjusting the proportion of the precursor or reaction solvent, self-doping and co-doping of the precursor were realized, and CDs with various fluorescent colors were finally prepared. It was found that red-emission CDs (r-CDs) could be prepared with SA and οPD as precursors and water as the solvent. Through comparative study, it was found that r-CDs were affected by H+ in the formation process and photoluminescence process. The fluorescence stability of r-CDs indicated that they have good selectivity for some metal ions. The r-CDs prepared in this paper realized the specific recognition of Cu2+ and Ag+ through the "off-on" process, and the detection limits were 0.165 µm and 1.53 µm, respectively. And this test has the potential for practical qualitative testing.

14.
Anal Chem ; 95(37): 13733-13745, 2023 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-37688541

RESUMO

The interpretation of spectral data, including mass, nuclear magnetic resonance, infrared, and ultraviolet-visible spectra, is critical for obtaining molecular structural information. The development of advanced sensing technology has multiplied the amount of available spectral data. Chemical experts must use basic principles corresponding to the spectral information generated by molecular fragments and functional groups. This is a time-consuming process that requires a solid professional knowledge base. In recent years, the rapid development of computer science and its applications in cheminformatics and the emergence of computer-aided expert systems have greatly reduced the difficulty in analyzing large quantities of data. For expert systems, however, the problem-solving strategy must be known in advance or extracted by human experts and translated into algorithms. Gratifyingly, the development of artificial intelligence (AI) methods has shown great promise for solving such problems. Traditional algorithms, including the latest neural network algorithms, have shown great potential for both extracting useful information and processing massive quantities of data. This Perspective highlights recent innovations covering all of the emerging AI-based spectral interpretation techniques. In addition, the main limitations and current obstacles are presented, and the corresponding directions for further research are proposed. Moreover, this Perspective gives the authors' personal outlook on the development and future applications of spectral interpretation.

15.
Chem Sci ; 14(30): 8129-8146, 2023 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-37538816

RESUMO

Applying machine learning algorithms to protein-ligand scoring functions has aroused widespread attention in recent years due to the high predictive accuracy and affordable computational cost. Nevertheless, most machine learning-based scoring functions are only applicable to a specific task, e.g., binding affinity prediction, binding pose prediction or virtual screening, suggesting that the development of a scoring function with balanced performance in all critical tasks remains a grand challenge. To this end, we propose a novel parameterization strategy by introducing an adjustable binding affinity term that represents the correlation between the predicted outcomes and experimental data into the training of mixture density network. The resulting residue-atom distance likelihood potential not only retains the superior docking and screening power over all the other state-of-the-art approaches, but also achieves a remarkable improvement in scoring and ranking performance. We emphatically explore the impacts of several key elements on prediction accuracy as well as the task preference, and demonstrate that the performance of scoring/ranking and docking/screening tasks of a certain model could be well balanced through an appropriate manner. Overall, our study highlights the potential utility of our innovative parameterization strategy as well as the resulting scoring framework in future structure-based drug design.

16.
J Chem Theory Comput ; 19(16): 5633-5647, 2023 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-37480347

RESUMO

Nucleic acid (NA)-ligand interactions are of paramount importance in a variety of biological processes, including cellular reproduction and protein biosynthesis, and therefore, NAs have been broadly recognized as potential drug targets. Understanding NA-ligand interactions at the atomic scale is essential for investigating the molecular mechanism and further assisting in NA-targeted drug discovery. Molecular docking is one of the predominant computational approaches for predicting the interactions between NAs and small molecules. Despite the availability of versatile docking programs, their performance profiles for NA-ligand complexes have not been thoroughly characterized. In this study, we first compiled the largest structure-based NA-ligand binding data set to date, containing 800 noncovalent NA-ligand complexes with clearly identified ligands. Based on this extensive data set, eight frequently used docking programs, including six protein-ligand docking programs (LeDock, Surflex-Dock, UCSF Dock6, AutoDock, AutoDock Vina, and PLANTS) and two specific NA-ligand docking programs (rDock and RLDOCK), were systematically evaluated in terms of binding pose and binding affinity predictions. The results demonstrated that some protein-ligand docking programs, specifically PLANTS and LeDock, produced more promising or comparable results compared with the specialized NA-ligand docking programs. Among the programs evaluated, PLANTS, rDock, and LeDock showed the highest performance in binding pose prediction, and their top-1 and best root-mean-square deviation (rmsd) success rates were as follows: PLANTS (35.93 and 76.05%), rDock (27.25 and 72.16%), and LeDock (27.40 and 64.37%). Compared with the moderate level of binding pose prediction, few programs were successful in binding affinity prediction, and the best correlation (Rp = -0.461) was observed with PLANTS. Finally, further comparison with the latest NA-ligand docking program (NLDock) on four well-established data sets revealed that PLANTS and LeDock outperformed NLDock in terms of binding pose prediction on all data sets, demonstrating their significant potential for NA-ligand docking. To the best of our knowledge, this study is the most comprehensive evaluation of popular molecular docking programs for NA-ligand systems.


Assuntos
Descoberta de Drogas , Ácidos Nucleicos , Ligantes , Simulação de Acoplamento Molecular
17.
J Med Chem ; 66(13): 9174-9183, 2023 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-37317043

RESUMO

Machine-learning-based scoring functions (MLSFs) have gained attention for their potential to improve accuracy in binding affinity prediction and structure-based virtual screening (SBVS) compared to classical SFs. Developing accurate MLSFs for SBVS requires a large and unbiased dataset that includes structurally diverse actives and decoys. Unfortunately, most datasets suffer from hidden biases and data insufficiency. Here, we developed topology-based and conformation-based decoys database (ToCoDDB). The biological targets and active ligands in ToCoDDB were collected from scientific literature and established datasets. The decoys were generated and debiased by using conditional recurrent neural networks and molecular docking. ToCoDDB is presently the largest unbiased database with 2.4 million decoys encompassing 155 targets. The detailed information and performance benchmark for each target are provided, which are beneficial for training and evaluating MLSFs. Moreover, the online decoys generation function of ToCoDDB further expands its application range to any target. ToCoDDB is freely available at http://cadd.zju.edu.cn/tocodecoy/.


Assuntos
Benchmarking , Aprendizado de Máquina , Simulação de Acoplamento Molecular , Conformação Molecular , Bases de Dados Factuais , Ligantes , Ligação Proteica
18.
Brief Bioinform ; 24(4)2023 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-37193672

RESUMO

The rational design of chemical entities with desired properties for a specific target is a long-standing challenge in drug design. Generative neural networks have emerged as a powerful approach to sample novel molecules with specific properties, termed as inverse drug design. However, generating molecules with biological activity against certain targets and predefined drug properties still remains challenging. Here, we propose a conditional molecular generation net (CMGN), the backbone of which is a bidirectional and autoregressive transformer. CMGN applies large-scale pretraining for molecular understanding and navigates the chemical space for specified targets by fine-tuning with corresponding datasets. Additionally, fragments and properties were trained to recover molecules to learn the structure-properties relationships. Our model crisscrosses the chemical space for specific targets and properties that control fragment-growth processes. Case studies demonstrated the advantages and utility of our model in fragment-to-lead processes and multi-objective lead optimization. The results presented in this paper illustrate that CMGN has the potential to accelerate the drug discovery process.


Assuntos
Desenho de Fármacos , Descoberta de Drogas , Aprendizagem , Redes Neurais de Computação , Receptores Proteína Tirosina Quinases
19.
Nat Commun ; 14(1): 2585, 2023 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-37142585

RESUMO

Graph neural networks (GNNs) have been widely used in molecular property prediction, but explaining their black-box predictions is still a challenge. Most existing explanation methods for GNNs in chemistry focus on attributing model predictions to individual nodes, edges or fragments that are not necessarily derived from a chemically meaningful segmentation of molecules. To address this challenge, we propose a method named substructure mask explanation (SME). SME is based on well-established molecular segmentation methods and provides an interpretation that aligns with the understanding of chemists. We apply SME to elucidate how GNNs learn to predict aqueous solubility, genotoxicity, cardiotoxicity and blood-brain barrier permeation for small molecules. SME provides interpretation that is consistent with the understanding of chemists, alerts them to unreliable performance, and guides them in structural optimization for target properties. Hence, we believe that SME empowers chemists to confidently mine structure-activity relationship (SAR) from reliable GNNs through a transparent inspection on how GNNs pick up useful signals when learning from data.


Assuntos
Barreira Hematoencefálica , Cardiotoxicidade , Humanos , Dano ao DNA , Redes Neurais de Computação , Registros
20.
J Fluoresc ; 33(6): 2273-2280, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37017894

RESUMO

Most fluorescent probes based on carbon dots (CDs) fluorescence color or intensity change are still used for detection in solution, but in practical fluorescence detection applications, detection in the solid state is necessary. Therefore, a CDs-based fluorescence sensing device is designed in this paper, which can be used for water detection in liquid and solid states. Using oPD as a single precursor, yellow fluorescent CDs (y-CDs) were prepared by hydrothermal method, which can be used in the field of water detection and anti-counterfeiting by using its solvent-sensitive properties. First, y-CDs can be used to visually and intelligently detect the water content in ethanol. Secondly, it can be used to detect the Relative Humidity (RH) of the environment by combining it with cellulose to form a fluorescent film. Finally, y-CDs can also be used as a fluorescent material for fluorescence anti-counterfeiting.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...