Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Comput Biol Med ; 171: 108165, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38402838

RESUMO

Virtual screening (VS) has been incorporated into the paradigm of modern drug discovery. This field is now undergoing a new wave of revolution driven by artificial intelligence and more specifically, machine learning (ML). In terms of those out-of-the-box datasets for model training or benchmarking, their data volume and applicability domain are limited. They are suffering from the biases constantly reported in the ML application. To address these issues, we present a novel benchmark named MUBDsyn. The utilization of synthetic decoys (i.e., presumed inactives) is the main feature of MUBDsyn, where deep reinforcement learning was leveraged for bias control during decoy generation. Then, we carried out extensive validations on this new benchmark. First, we confirmed that MUBDsyn was superior to the classical benchmarks in control of domain bias, artificial enrichment bias and analogue bias. Moreover, we found that the assessment of ML models based on MUBDsyn was less biased as revealed by the analysis of asymmetric validation embedding bias. In addition, MUBDsyn showed better setting of benchmarking challenge for deep learning models compared with NRLiSt-BDB. Overall, we have proven that MUBDsyn is the close-to-ideal benchmark for VS. The computational tool is publicly available for the easy extension of MUBDsyn.


Assuntos
Inteligência Artificial , Benchmarking , Descoberta de Drogas , Aprendizado de Máquina , Viés
2.
Chemistry ; 30(26): e202304279, 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38409580

RESUMO

Artificial intelligence (AI)/machine learning (ML) is emerging as pivotal in synthetic chemistry, offering revolutionary potential in retrosynthetic analysis, reaction conditions and reaction prediction. We have combined chemical descriptors, primarily based on Density Functional Theory (DFT) calculations, with various AI/ML tools such as Multi-Layer Perceptron (MLP) and Random Forest (RF), to predict the synthesis of 2-arylbenzothiazole in photoredox reactions. Significantly, our models underscore the critical role of the molecular structure and physicochemical characteristics of the base, especially the total atomic polarizabilities, in the rate-determining steps involving cyclohexyl and phenethyl moieties of the substrate. Moreover, we validated our findings in articles through experimental studies. It showcases the power of AI/ML and quantum chemistry in shaping the future of organic chemistry.

3.
Expert Opin Drug Discov ; 16(10): 1175-1192, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34011222

RESUMO

Introduction: Structure-based virtual screening (SBVS) is an essential strategy for hit identification. SBVS primarily uses molecular docking, which exploits the protein-ligand binding mode and associated affinity score for compound ranking. Previous studies have shown that computational representation of protein-ligand interfaces and the later establishment of machine learning models are efficacious in improving the accuracy of SBVS.Areas covered: The authors review the computational methods for representing protein-ligand interfaces, which include the traditional ones that use deliberately designed fingerprints and descriptors and the more recent methods that automatically extract features with deep learning. The effects of these methods on the performance of machine learning models are briefly discussed. Additionally, case studies that applied various computational representations to machine learning are cited with remarks.Expert opinion: It has become a trend to extract binding features automatically by deep learning, which uses a completely end-to-end representation. However, there is still plenty of scope for improvement . The interpretability of deep-learning models, the organization of data management, the quantity and quality of available data, and the optimization of hyperparameters could impact the accuracy of feature extraction. In addition, other important structural factors such as water molecules and protein flexibility should be considered.


Assuntos
Aprendizado de Máquina , Proteínas , Humanos , Ligantes , Simulação de Acoplamento Molecular , Ligação Proteica , Proteínas/metabolismo
4.
Chem Biol Drug Des ; 97(4): 944-961, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33386704

RESUMO

CC chemokine receptor 2 (CCR2) antagonists that disrupt CCR2/MCP-1 interaction are expected to treat a variety of inflammatory and autoimmune diseases. The lack of CCR2 crystal structure limits the application of structure-based drug design (SBDD) to this target. Although a few three-dimensional theoretical models have been reported, their accuracy remains to be improved in terms of templates and modeling approaches. In this study, we developed a unique ligand-steered strategy for CCR2 homology modeling. It starts with an initial model based on the X-ray structure of the closest homolog so far, that is, CXCR4. Then, it uses Elastic Network Normal Mode Analysis (EN-NMA) and flexible docking (FD) by AutoDock Vina software to generate ligand-induced fit models. It selects optimal model(s) as well as scoring function(s) via extensive evaluation of model performance based on a unique benchmarking set constructed by our in-house tool, that is, MUBD-DecoyMaker. The model of 81_04 presents the optimal enrichment when combined with the scoring function of PMF04, and the proposed binding mode between CCR2 and Teijin lead by this model complies with the reported mutagenesis data. To highlight the advantage of our strategy, we compared it with the only reported ligand-steered strategy for CCR2 homology modeling, that is, Discovery Studio/Ligand Minimization. Lastly, we performed prospective virtual screening based on 81_04 and CCR2 antagonist bioassay. The identification of two hit compounds, that is, E859-1281 and MolPort-007-767-945, validated the efficacy of our model and the ligand-steered strategy.


Assuntos
Ligantes , Simulação de Acoplamento Molecular , Receptores CCR2/metabolismo , Sítios de Ligação , Cálcio/metabolismo , Humanos , Mutagênese , Ligação Proteica , Receptores CCR2/antagonistas & inibidores , Receptores CCR2/genética
5.
J Chem Inf Model ; 60(3): 1202-1214, 2020 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-32050066

RESUMO

Farnesoid X receptor (FXR) agonists can reverse dysregulated bile acid metabolism, and thus, they are potential therapeutics to prevent and treat nonalcoholic fatty liver disease. The low success rate of FXR agonists' R&D and the side effects of clinical candidates such as obeticholic acid make it urgent to discover new chemotypes. Unfortunately, structure-based virtual screening (SBVS) that can speed up drug discovery has rarely been reported with success for FXR, which was likely hindered by the failure in addressing protein flexibility. To address this issue, we devised human FXR (hFXR)-specific ensemble learning models based on pose filters from 24 agonist-bound hFXR crystal structures and coupled them to traditional SBVS approaches of the FRED docking plus Chemgauss4 scoring function. It turned out that the hFXR-specific pose filter ensemble (PFE) was able to improve ligand enrichment significantly, which rendered 3RUT-based SBVS with its PFE the ideal approach for FXR agonist discovery. By screening of the Specs chemical library and in vitro FXR transactivation bioassay, we identified a new class of FXR agonists with compound XJ034 as the representative, which would have been missed if the PFE was not coupled. Following that, we performed in-depth biological studies which demonstrated that XJ034 resulted in a downtrend of intracellular triglyceride in vitro, significantly decreased the serum/liver TG in high fat diet-induced C57BL/6J obese mice, and more importantly, showed metabolic stabilities in both plasma and liver microsomes. To provide insight into further structure-based lead optimization, we solved the crystal structure of hFXR complexed with compound XJ034, uncovering a unique hydrogen bond between compound XJ034 and residue Y375. The current work highlights the power of our pose filter-based ensemble learning approach in terms of scaffold hopping and provides a promising lead compound for further development.


Assuntos
Fígado , Receptores Citoplasmáticos e Nucleares , Animais , Ligantes , Aprendizado de Máquina , Camundongos , Camundongos Endogâmicos C57BL
6.
Mol Inform ; 39(4): e1900151, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31828959

RESUMO

Ligand enrichment assessment based on benchmarking data sets has become a necessity for the rational selection of the best-suited approach for prospective data mining of drug-like molecules. Up to now, a variety of benchmarking data sets had been generated and frequently used. Among them, MUBD-HDACs from our prior research efforts was regarded as one of five state-of-the-art benchmarks in 2017 by Frontiers in Pharmacology. This benchmarking set was generated by one of our unique de-biasing algorithms. It also rendered quite a few other cases of successful applications in recent years, thus is expected to have more impact in modern drug discovery. To make our algorithm amenable to more users, we developed a Python GUI application called MUBD-DecoyMaker 2.0. Moreover, it has two new additional functional modules, i. e. "Detect 2D Bias" and "Quality Control". This new GUI version had been proved to be easy to use while generate benchmarking data sets of the same quality. MUBD-DecoyMaker 2.0 is freely available at https://github.com/jwxia2014/MUBD-DecoyMaker2.0, along with its manual and testcase.


Assuntos
Bases de Dados de Produtos Farmacêuticos , Conjuntos de Dados como Assunto/normas , Avaliação Pré-Clínica de Medicamentos , Preparações Farmacêuticas/química , Linguagens de Programação , Interface Usuário-Computador , Algoritmos , Descoberta de Drogas
7.
Bioorg Chem ; 86: 224-234, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30716620

RESUMO

Protein tyrosine phosphatase 1B (PTP1B) has recently been identified as a potential target of Norathyriol. Unfortunately, Norathyriol is not a potent PTP1B inhibitor, which somewhat hinders its further application. Based on the fact that no study on the relationship of chemical structure and PTP1B inhibitory activity of Norathyriol has been reported so far, we attempted to perform structural optimization so as to improve the potency for PTP1B. Via structure-based drug design (SBDD), a rational strategy based on the binding mode of Norathyriol to PTP1B, we designed 26 derivatives with substitutions at the four phenolic hydroxyl groups of Norathyriol. By chemical synthesis and in vitro bioassay, we identified seven PTP1B inhibitors that were more potent than Norathyriol, of which XWJ24 showed the highest potency (IC50: 0.6 µM). We also found out that XWJ24 was a competitive inhibitor and showed the 4.5-fold selectivity over its close homolog, TC-PTP. Through molecular docking of XWJ24 against PTP1B, we highlighted the essential role of its hydrogen bond with Asp181 for PTP1B inhibition and identified a potential halogen bond with Asp48 that was not observed for Norathyriol. The current data indicate that our SBDD strategy is effective to discover potent PTP1B-targeted Norathyriol derivatives, and XWJ24 is a promising lead compound for further development.


Assuntos
Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Proteína Tirosina Fosfatase não Receptora Tipo 1/antagonistas & inibidores , Xantenos/farmacologia , Bioensaio , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Simulação de Acoplamento Molecular , Estrutura Molecular , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Relação Estrutura-Atividade , Xantenos/síntese química , Xantenos/química
8.
J Chem Inf Model ; 58(5): 1104-1120, 2018 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-29698608

RESUMO

Chemokine receptors (CRs) have long been druggable targets for the treatment of inflammatory diseases and HIV-1 infection. As a powerful technique, virtual screening (VS) has been widely applied to identifying small molecule leads for modern drug targets including CRs. For rational selection of a wide variety of VS approaches, ligand enrichment assessment based on a benchmarking data set has become an indispensable practice. However, the lack of versatile benchmarking sets for the whole CRs family that are able to unbiasedly evaluate every single approach including both structure- and ligand-based VS somewhat hinders modern drug discovery efforts. To address this issue, we constructed Maximal Unbiased Benchmarking Data sets for human Chemokine Receptors (MUBD-hCRs) using our recently developed tools of MUBD-DecoyMaker. The MUBD-hCRs encompasses 13 subtypes out of 20 chemokine receptors, composed of 404 ligands and 15756 decoys so far and is readily expandable in the future. It had been thoroughly validated that MUBD-hCRs ligands are chemically diverse while its decoys are maximal unbiased in terms of "artificial enrichment", "analogue bias". In addition, we studied the performance of MUBD-hCRs, in particular CXCR4 and CCR5 data sets, in ligand enrichment assessments of both structure- and ligand-based VS approaches in comparison with other benchmarking data sets available in the public domain and demonstrated that MUBD-hCRs is very capable of designating the optimal VS approach. MUBD-hCRs is a unique and maximal unbiased benchmarking set that covers major CRs subtypes so far.


Assuntos
Descoberta de Drogas , Receptores de Quimiocinas/química , Receptores de Quimiocinas/metabolismo , Benchmarking , Bases de Dados de Proteínas , Humanos , Ligantes
9.
J Enzyme Inhib Med Chem ; 33(1): 525-535, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29464997

RESUMO

Histone deacetylase 3 (HDAC3) is a potential target for the treatment of human diseases such as cancers, diabetes, chronic inflammation and neurodegenerative diseases. Previously, we proposed a virtual screening (VS) pipeline named "Hypo1_FRED_SAHA-3" for the discovery of HDAC3 inhibitors (HDAC3Is) and had thoroughly validated it by theoretical calculations. In this study, we attempted to explore its practical utility in a large-scale VS campaign. To this end, we used the VS pipeline to hierarchically screen the Specs chemical library. In order to facilitate compound cherry-picking, we then developed a knowledge-based pose filter (PF) by using our in-house quantitative structure activity relationship- (QSAR-) modelling approach and coupled it with FRED and Autodock Vina. Afterward, we purchased and tested 11 diverse compounds for their HDAC3 inhibitory activity in vitro. The bioassay has identified compound 2 (Specs ID: AN-979/41971160) as a HDAC3I (IC50 = 6.1 µM), which proved the efficacy of our workflow. As a medicinal chemistry study, we performed a follow-up substructure search and identified two more hit compounds of the same chemical type, i.e. 2-1 (AQ-390/42122119, IC50 = 1.3 µM) and 2-2 (AN-329/43450111, IC50 = 12.5 µM). Based on the chemical structures and activities, we have demonstrated the essential role of the capping group in maintaining the activity for this class of HDAC3Is. In addition, we tested the hit compounds for their in vitro activities on other HDACs, including HDAC1, HDAC2, HDAC8, HDAC4 and HDAC6. We have identified these compounds are HDAC1/2/3 selective inhibitors, of which compound 2 show the best selectivity profile. Taken together, the present study is an experimental validation and an update to our earlier VS strategy. The identified hits could be used as starting structures for the development of highly potent and selective HDAC3Is.


Assuntos
Descoberta de Drogas , Bioensaio , Relação Dose-Resposta a Droga , Inibidores de Histona Desacetilases , Histona Desacetilases , Humanos , Modelos Moleculares , Estrutura Molecular , Relação Quantitativa Estrutura-Atividade
10.
Molecules ; 22(6)2017 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-28635653

RESUMO

Inhibition of apoptosis is a potential therapy to treat human diseases such as neurodegenerative disorders (e.g., Parkinson's disease), stroke, and sepsis. Due to the lack of druggable targets, it remains a major challenge to discover apoptosis inhibitors. The recent repositioning of a marketed drug (i.e., terazosin) as an anti-apoptotic agent uncovered a novel target (i.e., human phosphoglycerate kinase 1 (hPgk1)). In this study, we developed a virtual screening (VS) pipeline based on the X-ray structure of Pgk1/terazosin complex and applied it to a screening campaign for potential anti-apoptotic agents. The hierarchical filters in the pipeline (i.e., similarity search, a pharmacophore model, a shape-based model, and molecular docking) rendered 13 potential hits from Specs chemical library. By using PC12 cells (exposed to rotenone) as a cell model for bioassay, we first identified that AK-918/42829299, AN-465/41520984, and AT-051/43421517 were able to protect PC12 cells from rotenone-induced cell death. Molecular docking suggested these hit compounds were likely to bind to hPgk1 in a similar mode to terazosin. In summary, we not only present a versatile VS pipeline for potential apoptosis inhibitors discovery, but also provide three novel-scaffold hit compounds that are worthy of further development and biological study.


Assuntos
Apoptose/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Fosfoglicerato Quinase/antagonistas & inibidores , Fosfoglicerato Quinase/metabolismo , Prazosina/análogos & derivados , Inibidores de Proteínas Quinases/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Bases de Dados de Compostos Químicos , Humanos , Modelos Moleculares , Simulação de Acoplamento Molecular/métodos , Células PC12 , Fosfoglicerato Quinase/química , Prazosina/química , Prazosina/metabolismo , Prazosina/farmacologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/metabolismo , Ratos , Bibliotecas de Moléculas Pequenas
11.
J Chem Inf Model ; 57(6): 1414-1425, 2017 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-28511009

RESUMO

Structure-based virtual screening (SBVS) has become an indispensable technique for hit identification at the early stage of drug discovery. However, the accuracy of current scoring functions is not high enough to confer success to every target and thus remains to be improved. Previously, we had developed binary pose filters (PFs) using knowledge derived from the protein-ligand interface of a single X-ray structure of a specific target. This novel approach had been validated as an effective way to improve ligand enrichment. Continuing from it, in the present work we attempted to incorporate knowledge collected from diverse protein-ligand interfaces of multiple crystal structures of the same target to build PF ensembles (PFEs). Toward this end, we first constructed a comprehensive data set to meet the requirements of ensemble modeling and validation. This set contains 10 diverse targets, 118 well-prepared X-ray structures of protein-ligand complexes, and large benchmarking actives/decoys sets. Notably, we designed a unique workflow of two-layer classifiers based on the concept of ensemble learning and applied it to the construction of PFEs for all of the targets. Through extensive benchmarking studies, we demonstrated that (1) coupling PFE with Chemgauss4 significantly improves the early enrichment of Chemgauss4 itself and (2) PFEs show greater consistency in boosting early enrichment and larger overall enrichment than our prior PFs. In addition, we analyzed the pairwise topological similarities among cognate ligands used to construct PFEs and found that it is the higher chemical diversity of the cognate ligands that leads to the improved performance of PFEs. Taken together, the results so far prove that the incorporation of knowledge from diverse protein-ligand interfaces by ensemble modeling is able to enhance the screening competence of SBVS scoring functions.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Benchmarking , Ligantes , Simulação de Acoplamento Molecular , Conformação Proteica , Interface Usuário-Computador
12.
Int J Mol Sci ; 18(1)2017 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-28106794

RESUMO

Histone deacetylase 3 (HDAC3) has been recently identified as a potential target for the treatment of cancer and other diseases, such as chronic inflammation, neurodegenerative diseases, and diabetes. Virtual screening (VS) is currently a routine technique for hit identification, but its success depends on rational development of VS strategies. To facilitate this process, we applied our previously released benchmarking dataset, i.e., MUBD-HDAC3 to the evaluation of structure-based VS (SBVS) and ligand-based VS (LBVS) combinatorial approaches. We have identified FRED (Chemgauss4) docking against a structural model of HDAC3, i.e., SAHA-3 generated by a computationally inexpensive "flexible docking", as the best SBVS approach and a common feature pharmacophore model, i.e., Hypo1 generated by Catalyst/HipHop as the optimal model for LBVS. We then developed a pipeline that was composed of Hypo1, FRED (Chemgauss4), and SAHA-3 sequentially, and demonstrated that it was superior to other combinations in terms of ligand enrichment. In summary, we present the first highly-validated, rationally-designed VS strategy specific to HDAC3 inhibitor discovery. The constructed pipeline is publicly accessible for the scientific community to identify novel HDAC3 inhibitors in a time-efficient and cost-effective way.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Inibidores de Histona Desacetilases/análise , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Interface Usuário-Computador , Área Sob a Curva , Catálise , Inibidores de Histona Desacetilases/química , Ligantes , Simulação de Acoplamento Molecular , Curva ROC , Reprodutibilidade dos Testes , Relação Estrutura-Atividade
13.
J Chemom ; 31(12)2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29606793

RESUMO

Quionolone carboxylic acid derivatives as inhibitors of HIV-1 integrase were investigated as a potential class of drugs for the treatment of acquired immunodeficiency syndrome (AIDS). Hologram quantitative structure-activity relationships (HQSAR) and translocation comparative molecular field vector analysis (topomer CoMFA) were applied to a series of 48 quionolone carboxylic acid derivatives. The most effective HQSAR model was obtained using atoms and bonds as fragment distinctions: cross-validation q2 = 0.796, standard error of prediction SDCV = 0.36, the non-cross-validated r2 = 0.967, non-cross validated standard error SD = 0.17, the correlation coefficient of external validation Qext2 = 0.955, and the best hologram length HL = 180. topomer CoMFA models were built based on different fragment cutting models, with the most effective model of q2 = 0.775, SDCV = 0.37, r2 = 0.967, SD = 0.15, Qext2 = 0.915, and F = 163.255. These results show that the models generated form HQSAR and topomer CoMFA were able to effectively predict the inhibitory potency of this class of compounds. The molecular docking method was also used to study the interactions of these drugs by docking the ligands into the HIV-1 integrase active site, which revealed the likely bioactive conformations. This study showed that there are extensive interactions between the quionolone carboxylic acid derivatives and THR80, VAL82, GLY27, ASP29, and ARG8 residues in the active site of HIV-1 integrase. These results provide useful insights for the design of potent new inhibitors of HIV-1 integrase.

14.
Comb Chem High Throughput Screen ; 18(7): 685-92, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26138565

RESUMO

The human 5-hydroxytryptamine receptor subtype 1A (5-HT1A) is highly expressed in the raphe nuclei region and limbic structures; for that reason 5-HT1A has served as a promising target for treating human mood disorders and neurodegenerative diseases. We have developed binary quantitative structure-activity relationship (QSAR) models for 5- HT1A binding using data retrieved from the WOMBAT database and the k-Nearest Neighbor (kNN) machine learning method. A rigorous QSAR modeling and screening workflow had been followed, with extensive internal and external validation processes. The models' classification accuracies to discriminate 5-HT1A binders from the non-binders are as high as 96% for the external validation. These models were employed further to mine two major natural products screening libraries, i.e. TimTec Natural Product Library (NPL) and Natural Derivatives Library (NDL). In the end five screening hits were tested by radioligand binding assays with a success rate of 40%, and two Library compounds were confirmed to be binders at the µM concentration against the human 5-HT1A receptor. The combined application of rigorous QSAR modeling and model-based virtual screening presents a powerful means for profiling natural products compounds with important biomedical activities.


Assuntos
Produtos Biológicos , Descoberta de Drogas , Avaliação Pré-Clínica de Medicamentos , Modelos Biológicos , Receptor 5-HT1A de Serotonina/química , Bibliotecas de Moléculas Pequenas/farmacologia , Produtos Biológicos/química , Classificação , Relação Dose-Resposta a Droga , Humanos , Concentração Inibidora 50 , Relação Quantitativa Estrutura-Atividade , Receptor 5-HT1A de Serotonina/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/química
15.
J Chem Inf Model ; 55(2): 374-88, 2015 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-25633490

RESUMO

Histone deacetylases (HDACs) are an important class of drug targets for the treatment of cancers, neurodegenerative diseases, and other types of diseases. Virtual screening (VS) has become fairly effective approaches for drug discovery of novel and highly selective histone deacetylase inhibitors (HDACIs). To facilitate the process, we constructed maximal unbiased benchmarking data sets for HDACs (MUBD-HDACs) using our recently published methods that were originally developed for building unbiased benchmarking sets for ligand-based virtual screening (LBVS). The MUBD-HDACs cover all four classes including Class III (Sirtuins family) and 14 HDAC isoforms, composed of 631 inhibitors and 24609 unbiased decoys. Its ligand sets have been validated extensively as chemically diverse, while the decoy sets were shown to be property-matching with ligands and maximal unbiased in terms of "artificial enrichment" and "analogue bias". We also conducted comparative studies with DUD-E and DEKOIS 2.0 sets against HDAC2 and HDAC8 targets and demonstrate that our MUBD-HDACs are unique in that they can be applied unbiasedly to both LBVS and SBVS approaches. In addition, we defined a novel metric, i.e. NLBScore, to detect the "2D bias" and "LBVS favorable" effect within the benchmarking sets. In summary, MUBD-HDACs are the only comprehensive and maximal-unbiased benchmark data sets for HDACs (including Sirtuins) that are available so far. MUBD-HDACs are freely available at http://www.xswlab.org/ .


Assuntos
Histona Desacetilases/química , Sirtuínas/química , Algoritmos , Benchmarking , Bases de Dados de Compostos Químicos , Ensaios de Triagem em Larga Escala , Inibidores de Histona Desacetilases/química , Inibidores de Histona Desacetilases/farmacologia , Humanos , Ligantes , Modelos Químicos , Modelos Moleculares
16.
Methods ; 71: 146-57, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25481478

RESUMO

Retrospective small-scale virtual screening (VS) based on benchmarking data sets has been widely used to estimate ligand enrichments of VS approaches in the prospective (i.e. real-world) efforts. However, the intrinsic differences of benchmarking sets to the real screening chemical libraries can cause biased assessment. Herein, we summarize the history of benchmarking methods as well as data sets and highlight three main types of biases found in benchmarking sets, i.e. "analogue bias", "artificial enrichment" and "false negative". In addition, we introduce our recent algorithm to build maximum-unbiased benchmarking sets applicable to both ligand-based and structure-based VS approaches, and its implementations to three important human histone deacetylases (HDACs) isoforms, i.e. HDAC1, HDAC6 and HDAC8. The leave-one-out cross-validation (LOO CV) demonstrates that the benchmarking sets built by our algorithm are maximum-unbiased as measured by property matching, ROC curves and AUCs.


Assuntos
Benchmarking , Avaliação Pré-Clínica de Medicamentos/métodos , Algoritmos , Área Sob a Curva , Descoberta de Drogas/métodos , Ligantes , Curva ROC
17.
J Chem Inf Model ; 54(5): 1433-50, 2014 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-24749745

RESUMO

Benchmarking data sets have become common in recent years for the purpose of virtual screening, though the main focus had been placed on the structure-based virtual screening (SBVS) approaches. Due to the lack of crystal structures, there is great need for unbiased benchmarking sets to evaluate various ligand-based virtual screening (LBVS) methods for important drug targets such as G protein-coupled receptors (GPCRs). To date these ready-to-apply data sets for LBVS are fairly limited, and the direct usage of benchmarking sets designed for SBVS could bring the biases to the evaluation of LBVS. Herein, we propose an unbiased method to build benchmarking sets for LBVS and validate it on a multitude of GPCRs targets. To be more specific, our methods can (1) ensure chemical diversity of ligands, (2) maintain the physicochemical similarity between ligands and decoys, (3) make the decoys dissimilar in chemical topology to all ligands to avoid false negatives, and (4) maximize spatial random distribution of ligands and decoys. We evaluated the quality of our Unbiased Ligand Set (ULS) and Unbiased Decoy Set (UDS) using three common LBVS approaches, with Leave-One-Out (LOO) Cross-Validation (CV) and a metric of average AUC of the ROC curves. Our method has greatly reduced the "artificial enrichment" and "analogue bias" of a published GPCRs benchmarking set, i.e., GPCR Ligand Library (GLL)/GPCR Decoy Database (GDD). In addition, we addressed an important issue about the ratio of decoys per ligand and found that for a range of 30 to 100 it does not affect the quality of the benchmarking set, so we kept the original ratio of 39 from the GLL/GDD.


Assuntos
Benchmarking , Avaliação Pré-Clínica de Medicamentos/métodos , Receptores Acoplados a Proteínas G/metabolismo , Fenômenos Químicos , Bases de Dados de Produtos Farmacêuticos , Humanos , Ligantes , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Interface Usuário-Computador
18.
J Chem Inf Model ; 54(2): 634-47, 2014 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-24410373

RESUMO

The 5-hydroxytryptamine 1A (5-HT1A) serotonin receptor has been an attractive target for treating mood and anxiety disorders such as schizophrenia. We have developed binary classification quantitative structure-activity relationship (QSAR) models of 5-HT1A receptor binding activity using data retrieved from the PDSP Ki database. The prediction accuracy of these models was estimated by external 5-fold cross-validation as well as using an additional validation set comprising 66 structurally distinct compounds from the World of Molecular Bioactivity database. These validated models were then used to mine three major types of chemical screening libraries, i.e., drug-like libraries, GPCR targeted libraries, and diversity libraries, to identify novel computational hits. The five best hits from each class of libraries were chosen for further experimental testing in radioligand binding assays, and nine of the 15 hits were confirmed to be active experimentally with binding affinity better than 10 µM. The most active compound, Lysergol, from the diversity library showed very high binding affinity (Ki) of 2.3 nM against 5-HT1A receptor. The novel 5-HT1A actives identified with the QSAR-based virtual screening approach could be potentially developed as novel anxiolytics or potential antischizophrenic drugs.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Relação Quantitativa Estrutura-Atividade , Receptor 5-HT1A de Serotonina/metabolismo , Interface Usuário-Computador , Antipsicóticos/química , Antipsicóticos/metabolismo , Antipsicóticos/farmacologia , Ligantes , Ligação Proteica
19.
Curr Top Med Chem ; 13(11): 1353-62, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23675941

RESUMO

Serotonin (5-hydroxytryptamine, 5-HT) receptors are neuromodulator neurotransmitter receptors which when activated trigger a signal transduction cascade within cells resulting in cell-cell communication. 5-hydroxytryptamine receptor 2B (5-HT2B) is a subtype of the seven members of 5-hydroxytrytamine receptors family which is the largest member of the super family of 7-transmembrane G-protein coupled receptors (GPCRs). Not only do 5-HT receptors play physiological roles in the cardiovascular system, gastrointestinal and endocrine function as well as the central nervous system, but they also play a role in behavioral functions. In particular 5-HT2B receptor is widely spread with regards to its distribution throughout bodily tissues and is expressed at high levels in the lungs, peripheral tissues, liver, kidneys and prostate, just to name a few. Hence 5-HT2B participates in multiple biological functions including CNS regulation, regulation of gastrointestinal motality, cardiovascular regulation and 5-HT transport system regulation. While 5-HT2B is a viable drug target and has therapeutic indications for treating obesity, psychosis, Parkinson's disease etc. there is a growing concern regarding adverse drug reactions, specifically valvulopathy associated with 5-HT2B agonists. Due to the sequence homology experienced by 5-HT2 subtypes there is also a concern regarding the off-target effects of 5-HT2A and 5-HT2C agonists. The concepts of sensitivity and subtype selectivity are of paramount importance and now can be tackled with the aid of in silico studies, especially cheminformatics, to develop models to predict valvulopathy associated toxicity of drug candidates prior to clinical trials. This review has highlighted three in silico approaches thus far that have been successful in either predicting 5-HT2B toxicity of molecules or identifying important interactions between 5-HT2B and drug molecules that bring about valvulopathy related toxicities.


Assuntos
Avaliação Pré-Clínica de Medicamentos , Doenças das Valvas Cardíacas/prevenção & controle , Modelos Moleculares , Receptor 5-HT2B de Serotonina/química , Agonistas do Receptor 5-HT2 de Serotonina/efeitos adversos , Simulação por Computador , Desenho de Fármacos , Doenças das Valvas Cardíacas/induzido quimicamente , Humanos , Relação Quantitativa Estrutura-Atividade , Receptor 5-HT2A de Serotonina/química , Receptor 5-HT2B de Serotonina/metabolismo , Receptor 5-HT2C de Serotonina/química , Serotonina/metabolismo , Agonistas do Receptor 5-HT2 de Serotonina/química , Antagonistas do Receptor 5-HT2 de Serotonina/química , Transdução de Sinais
20.
Proteins ; 80(6): 1503-21, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22275072

RESUMO

Recent highly expected structural characterizations of agonist-bound and antagonist-bound beta-2 adrenoreceptor (ß2AR) by X-ray crystallography have been widely regarded as critical advances to enable more effective structure-based discovery of GPCRs ligands. It appears that this very important development may have undermined many previous efforts to develop 3D theoretical models of GPCRs. To address this question directly, we have compared several historical ß2AR models versus the inactive state and nanobody-stabilized active state of ß2AR crystal structures in terms of their structural similarity and effectiveness of use in virtual screening for ß2AR specific agonists and antagonists. Theoretical models, incluing both homology and de novo types, were collected from five different groups who have published extensively in the field of GPCRs modeling. All models were built before X-ray structures became available. In general, ß2AR theoretical models differ significantly from the crystal structure in terms of TMH definition and the global packing. Nevertheless, surprisingly, several models afforded hit rates resulting from virtual screening of large chemical library enriched by known ß2AR ligands that exceeded those using X-ray structures. The hit rates were particularly higher for agonists. Furthemore, the screening performance of models is associated with local structural quality, such as the RMSDs for binding pocket residues and the ability to capture accurately, most if not all critical protein/ligand interactions. These results suggest that carefully built models of GPCRs could capture critical chemical and structural features of the binding pocket, and thus may be even more useful for practical structure-based drug discovery than X-ray structures.


Assuntos
Biologia Computacional/métodos , Simulação de Dinâmica Molecular , Receptores Adrenérgicos beta 2/química , Receptores Acoplados a Proteínas G/química , Agonistas de Receptores Adrenérgicos beta 2/química , Agonistas de Receptores Adrenérgicos beta 2/metabolismo , Antagonistas de Receptores Adrenérgicos beta 2/química , Antagonistas de Receptores Adrenérgicos beta 2/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Análise por Conglomerados , Cristalografia por Raios X , Descoberta de Drogas , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Curva ROC , Receptores Adrenérgicos beta 2/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...