Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Bioinformatics ; 38(4): 1110-1117, 2022 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-34849593

RESUMO

MOTIVATION: In recent years, cyclic peptide drugs have been receiving increasing attention because they can target proteins that are difficult to be tackled by conventional small-molecule drugs or antibody drugs. Plasma protein binding rate (%PPB) is a significant pharmacokinetic property of a compound in drug discovery and design. However, due to structural differences, previous computational prediction methods developed for small-molecule compounds cannot be successfully applied to cyclic peptides, and methods for predicting the PPB rate of cyclic peptides with high accuracy are not yet available. RESULTS: Cyclic peptides are larger than small molecules, and their local structures have a considerable impact on PPB; thus, molecular descriptors expressing residue-level local features of cyclic peptides, instead of those expressing the entire molecule, as well as the circularity of the cyclic peptides should be considered. Therefore, we developed a prediction method named CycPeptPPB using deep learning that considers both factors. First, the macrocycle ring of cyclic peptides was decomposed residue by residue. The residue-based descriptors were arranged according to the sequence information of the cyclic peptide. Furthermore, the circular data augmentation method was used, and the circular convolution method CyclicConv was devised to express the cyclic structure. CycPeptPPB exhibited excellent performance, with mean absolute error (MAE) of 4.79% and correlation coefficient (R) of 0.92 for the public drug dataset, compared to the prediction performance of the existing PPB rate prediction software (MAE=15.08%, R=0.63). AVAILABILITY AND IMPLEMENTATION: The data underlying this article are available in the online supplementary material. The source code of CycPeptPPB is available at https://github.com/akiyamalab/cycpeptppb. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Aprendizado Profundo , Peptídeos Cíclicos , Proteínas Sanguíneas , Ligação Proteica , Software
2.
J Chem Inf Model ; 63(7): 2240-2250, 2023 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-36930969

RESUMO

Recently, cyclic peptides have been considered breakthrough drugs because they can interact with "undruggable" targets such as intracellular protein-protein interactions. Membrane permeability is an essential indicator of oral bioavailability and intracellular targeting, and the development of membrane-permeable peptides is a bottleneck in cyclic peptide drug discovery. Although many experimental data on membrane permeability of cyclic peptides have been reported, a comprehensive database is not yet available. A comprehensive membrane permeability database is essential for developing computational methods for cyclic peptide drug design. In this study, we constructed CycPeptMPDB, the first web-accessible database of cyclic peptide membrane permeability. We collected information on a total of 7334 cyclic peptides, including the structure and experimentally measured membrane permeability, from 45 published papers and 2 patents from pharmaceutical companies. To unambiguously represent cyclic peptides larger than small molecules, we used the hierarchical editing language for macromolecules notation to generate a uniform sequence representation of peptides. In addition to data storage, CycPeptMPDB provides several supporting functions such as online data visualization, data analysis, and downloading. CycPeptMPDB is expected to be a valuable platform to support membrane permeability research on cyclic peptides. CycPeptMPDB can be freely accessed at http://cycpeptmpdb.com.


Assuntos
Peptídeos Cíclicos , Peptídeos , Peptídeos Cíclicos/química , Peptídeos/química , Desenho de Fármacos , Descoberta de Drogas/métodos , Permeabilidade , Permeabilidade da Membrana Celular
3.
Int J Mol Sci ; 24(17)2023 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-37686057

RESUMO

More than 930,000 protein-protein interactions (PPIs) have been identified in recent years, but their physicochemical properties differ from conventional drug targets, complicating the use of conventional small molecules as modalities. Cyclic peptides are a promising modality for targeting PPIs, but it is difficult to predict the structure of a target protein-cyclic peptide complex or to design a cyclic peptide sequence that binds to the target protein using computational methods. Recently, AlphaFold with a cyclic offset has enabled predicting the structure of cyclic peptides, thereby enabling de novo cyclic peptide designs. We developed a cyclic peptide complex offset to enable the structural prediction of target proteins and cyclic peptide complexes and found AlphaFold2 with a cyclic peptide complex offset can predict structures with high accuracy. We also applied the cyclic peptide complex offset to the binder hallucination protocol of AfDesign, a de novo protein design method using AlphaFold, and we could design a high predicted local-distance difference test and lower separated binding energy per unit interface area than the native MDM2/p53 structure. Furthermore, the method was applied to 12 other protein-peptide complexes and one protein-protein complex. Our approach shows that it is possible to design putative cyclic peptide sequences targeting PPI.


Assuntos
Sistemas de Liberação de Medicamentos , Peptídeos Cíclicos , Excipientes , Aparelhos Ortopédicos
4.
Molecules ; 28(15)2023 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-37570623

RESUMO

Protein-protein interactions (PPIs) are associated with various diseases; hence, they are important targets in drug discovery. However, the physicochemical empirical properties of PPI-targeted drugs are distinct from those of conventional small molecule oral pharmaceuticals, which adhere to the "rule of five (RO5)". Therefore, developing PPI-targeted drugs using conventional methods, such as molecular generation models, is challenging. In this study, we propose a molecular generation model based on deep reinforcement learning that is specialized for the production of PPI inhibitors. By introducing a scoring function that can represent the properties of PPI inhibitors, we successfully generated potential PPI inhibitor compounds. These newly constructed virtual compounds possess the desired properties for PPI inhibitors, and they show similarity to commercially available PPI libraries. The virtual compounds are freely available as a virtual library.


Assuntos
Descoberta de Drogas , Mapeamento de Interação de Proteínas , Descoberta de Drogas/métodos
5.
J Chem Inf Model ; 62(18): 4549-4560, 2022 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-36053061

RESUMO

Cyclic peptides have attracted attention as a promising pharmaceutical modality due to their potential to selectively inhibit previously undruggable targets, such as intracellular protein-protein interactions. Poor membrane permeability is the biggest bottleneck hindering successful drug discovery based on cyclic peptides. Therefore, the development of computational methods that can predict membrane permeability and support elucidation of the membrane permeation mechanism of drug candidate peptides is much sought after. In this study, we developed a protocol to simulate the behavior in membrane permeation steps and estimate the membrane permeability of large cyclic peptides with more than or equal to 10 residues. This protocol requires the use of a more realistic membrane model than a single-lipid phospholipid bilayer. To select a membrane model, we first analyzed the effect of cholesterol concentration in the model membrane on the potential of mean force and hydrogen bonding networks along the direction perpendicular to the membrane surface as predicted by molecular dynamics simulations using cyclosporine A. These results suggest that a membrane model with 40 or 50 mol % cholesterol was suitable for predicting the permeation process. Subsequently, two types of membrane models containing 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine and 40 and 50 mol % cholesterol were used. To validate the efficiency of our protocol, the membrane permeability of 18 ten-residue peptides was predicted. Correlation coefficients of R > 0.8 between the experimental and calculated permeability values were obtained with both model membranes. The results of this study demonstrate that the lipid membrane is not just a medium but also among the main factors determining the membrane permeability of molecules. The computational protocol proposed in this study and the findings obtained on the effect of membrane model composition will contribute to building a schematic view of the membrane permeation process. Furthermore, the results of this study will eventually aid the elucidation of design rules for peptide drugs with high membrane permeability.


Assuntos
Simulação de Dinâmica Molecular , Peptídeos Cíclicos , Colesterol/química , Ciclosporina , Bicamadas Lipídicas/química , Peptídeos/química , Peptídeos Cíclicos/farmacologia , Permeabilidade , Preparações Farmacêuticas , Fosfatidilcolinas/química , Fosfolipídeos
6.
J Chem Inf Model ; 61(7): 3681-3695, 2021 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-34236179

RESUMO

Membrane permeability is a significant obstacle facing the development of cyclic peptide drugs. However, membrane permeation mechanisms are poorly understood. To investigate common features of permeable (and nonpermeable) designs, it is necessary to reproduce the membrane permeation process of cyclic peptides through the lipid bilayer. We simulated the membrane permeation process of 100 six-residue cyclic peptides across the lipid bilayer based on steered molecular dynamics (MD) and replica-exchange umbrella sampling simulations and predicted membrane permeability using the inhomogeneous solubility-diffusion model and a modified version of it. Furthermore, we confirmed the effectiveness of this protocol by predicting the membrane permeability of 56 eight-residue cyclic peptides with diverse chemical structures, including some confidential designs from a pharmaceutical company. As a result, a reasonable correlation between experimentally assessed and calculated membrane permeability of cyclic peptides was observed for the peptide libraries, except for strongly hydrophobic peptides. Our analysis of the MD trajectory demonstrated that most peptides were stabilized in the boundary region between bulk water and membrane and that for most peptides, the process of crossing the center of the membrane is the main obstacle to membrane permeation. The height of this barrier is well correlated with the electrostatic interaction between the peptide and the surrounding media. The structural and energetic features of the representative peptide at each vertical position within the membrane were also analyzed, revealing that peptides permeate the membrane by changing their orientation and conformation according to the surrounding environment.


Assuntos
Bicamadas Lipídicas , Simulação de Dinâmica Molecular , Conformação Molecular , Peptídeos Cíclicos , Permeabilidade
7.
Int J Mol Sci ; 22(20)2021 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-34681589

RESUMO

Drug-likeness quantification is useful for screening drug candidates. Quantitative estimates of drug-likeness (QED) are commonly used to assess quantitative drug efficacy but are not suitable for screening compounds targeting protein-protein interactions (PPIs), which have recently gained attention. Therefore, we developed a quantitative estimate index for compounds targeting PPIs (QEPPI), specifically for early-stage screening of PPI-targeting compounds. QEPPI is an extension of the QED method for PPI-targeting drugs that models physicochemical properties based on the information available for drugs/compounds, specifically those reported to act on PPIs. FDA-approved drugs and compounds in iPPI-DB, which comprise PPI inhibitors and stabilizers, were evaluated using QEPPI. The results showed that QEPPI is more suitable than QED for early screening of PPI-targeting compounds. QEPPI was also considered an extended concept of the "Rule-of-Four" (RO4), a PPI inhibitor index. We evaluated the discriminatory performance of QEPPI and RO4 for datasets of PPI-target compounds and FDA-approved drugs using F-score and other indices. The F-scores of RO4 and QEPPI were 0.451 and 0.501, respectively. QEPPI showed better performance and enabled quantification of drug-likeness for early-stage PPI drug discovery. Hence, it can be used as an initial filter to efficiently screen PPI-targeting compounds.


Assuntos
Descoberta de Drogas/métodos , Mapas de Interação de Proteínas , Área Sob a Curva , Modelos Moleculares , Preparações Farmacêuticas/química , Preparações Farmacêuticas/metabolismo , Curva ROC
8.
Int J Mol Sci ; 22(10)2021 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-34069916

RESUMO

Periodontitis is an inflammation of tooth-supporting tissues, which is caused by bacteria in the subgingival plaque (biofilm) and the host immune response. Traditionally, subgingival pathogens have been investigated using methods such as culturing, DNA probes, or PCR. The development of next-generation sequencing made it possible to investigate the whole microbiome in the subgingival plaque. Previous studies have implicated dysbiosis of the subgingival microbiome in the etiology of periodontitis. However, details are still lacking. In this study, we conducted a metagenomic analysis of subgingival plaque samples from a group of Japanese individuals with and without periodontitis. In the taxonomic composition analysis, genus Bacteroides and Mycobacterium demonstrated significantly different compositions between healthy sites and sites with periodontal pockets. The results from the relative abundance of functional gene categories, carbohydrate metabolism, glycan biosynthesis and metabolism, amino acid metabolism, replication and repair showed significant differences between healthy sites and sites with periodontal pockets. These results provide important insights into the shift in the taxonomic and functional gene category abundance caused by dysbiosis, which occurs during the progression of periodontal disease.


Assuntos
Placa Dentária/microbiologia , Gengiva/microbiologia , Periodontite/microbiologia , Adulto , Idoso , Bactérias/genética , Placa Dentária/genética , Disbiose/genética , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Japão/epidemiologia , Masculino , Metagenoma , Microbiota/genética , Pessoa de Meia-Idade , Bolsa Periodontal/genética , Bolsa Periodontal/microbiologia , Periodontite/genética , RNA Ribossômico 16S/genética
9.
Mol Divers ; 23(1): 11-18, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29971617

RESUMO

Druglikeness is a useful concept for screening drug candidate compounds. We developed QEX, which is a new druglikeness index specific to individual targets. QEX is an improvement of the quantitative estimate of druglikeness (QED) method, which is a popular quantitative evaluation method of druglikeness proposed by Bickerton et al. QEX models the physicochemical properties of compounds that act on each target protein based on the concept of QED modeling physicochemical properties from information on US Food and Drug Administration-approved drugs. The result of the evaluation of PubChem assay data revealed that QEX showed better performance than the original QED did (the area under the curve value of the receiver operating characteristic curve improved by 0.069-0.236). We also present the c-Src inhibitor filtering results of the QEX constructed using Src family kinase inhibitors as a case study. QEX distinguished the inhibitors and non-inhibitors better than QED did. QEX works efficiently even when datasets of inactive compounds are unavailable. If both active and inactive compounds are present, QEX can be used as an initial filter to enhance the screening ability of conventional ligand-based virtual screenings.


Assuntos
Descoberta de Drogas , Inibidores de Proteínas Quinases , Quinases da Família src/antagonistas & inibidores , Modelos Moleculares
10.
BMC Bioinformatics ; 19(Suppl 4): 62, 2018 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-29745830

RESUMO

BACKGROUND: Protein-protein interactions (PPIs) play several roles in living cells, and computational PPI prediction is a major focus of many researchers. The three-dimensional (3D) structure and binding surface are important for the design of PPI inhibitors. Therefore, rigid body protein-protein docking calculations for two protein structures are expected to allow elucidation of PPIs different from known complexes in terms of 3D structures because known PPI information is not explicitly required. We have developed rapid PPI prediction software based on protein-protein docking, called MEGADOCK. In order to fully utilize the benefits of computational PPI predictions, it is necessary to construct a comprehensive database to gather prediction results and their predicted 3D complex structures and to make them easily accessible. Although several databases exist that provide predicted PPIs, the previous databases do not contain a sufficient number of entries for the purpose of discovering novel PPIs. RESULTS: In this study, we constructed an integrated database of MEGADOCK PPI predictions, named MEGADOCK-Web. MEGADOCK-Web provides more than 10 times the number of PPI predictions than previous databases and enables users to conduct PPI predictions that cannot be found in conventional PPI prediction databases. In MEGADOCK-Web, there are 7528 protein chains and 28,331,628 predicted PPIs from all possible combinations of those proteins. Each protein structure is annotated with PDB ID, chain ID, UniProt AC, related KEGG pathway IDs, and known PPI pairs. Additionally, MEGADOCK-Web provides four powerful functions: 1) searching precalculated PPI predictions, 2) providing annotations for each predicted protein pair with an experimentally known PPI, 3) visualizing candidates that may interact with the query protein on biochemical pathways, and 4) visualizing predicted complex structures through a 3D molecular viewer. CONCLUSION: MEGADOCK-Web provides a huge amount of comprehensive PPI predictions based on docking calculations with biochemical pathways and enables users to easily and quickly assess PPI feasibilities by archiving PPI predictions. MEGADOCK-Web also promotes the discovery of new PPIs and protein functions and is freely available for use at http://www.bi.cs.titech.ac.jp/megadock-web/ .


Assuntos
Bases de Dados de Proteínas , Internet , Mapeamento de Interação de Proteínas/métodos , Proteínas/química , Proteínas/metabolismo , Software
11.
BMC Bioinformatics ; 19(Suppl 19): 527, 2018 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-30598072

RESUMO

BACKGROUND: Cyclic peptide-based drug discovery is attracting increasing interest owing to its potential to avoid target protein depletion. In drug discovery, it is important to maintain the biostability of a drug within the proper range. Plasma protein binding (PPB) is the most important index of biostability, and developing a computational method to predict PPB of drug candidate compounds contributes to the acceleration of drug discovery research. PPB prediction of small molecule drug compounds using machine learning has been conducted thus far; however, no study has investigated cyclic peptides because experimental information of cyclic peptides is scarce. RESULTS: First, we adopted sparse modeling and small molecule information to construct a PPB prediction model for cyclic peptides. As cyclic peptide data are limited, applying multidimensional nonlinear models involves concerns regarding overfitting. However, models constructed by sparse modeling can avoid overfitting, offering high generalization performance and interpretability. More than 1000 PPB data of small molecules are available, and we used them to construct a prediction models with two enumeration methods: enumerating lasso solutions (ELS) and forward beam search (FBS). The accuracies of the prediction models constructed by ELS and FBS were equal to or better than those of conventional non-linear models (MAE = 0.167-0.174) on cross-validation of a small molecule compound dataset. Moreover, we showed that the prediction accuracies for cyclic peptides were close to those for small molecule compounds (MAE = 0.194-0.288). Such high accuracy could not be obtained by a simple method of learning from cyclic peptide data directly by lasso regression (MAE = 0.286-0.671) or ridge regression (MAE = 0.244-0.354). CONCLUSION: In this study, we proposed a machine learning techniques that uses low-dimensional sparse modeling to predict the PPB value of cyclic peptides computationally. The low-dimensional sparse model not only exhibits excellent generalization performance but also improves interpretation of the prediction model. This can provide common an noteworthy knowledge for future cyclic peptide drug discovery studies.


Assuntos
Proteínas Sanguíneas/metabolismo , Simulação por Computador , Aprendizado de Máquina , Modelos Teóricos , Peptídeos Cíclicos/metabolismo , Preparações Farmacêuticas/metabolismo , Bibliotecas de Moléculas Pequenas/metabolismo , Humanos , Ligação Proteica
12.
Bioinformatics ; 33(23): 3836-3843, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-28369284

RESUMO

MOTIVATION: Recently, the number of available protein tertiary structures and compounds has increased. However, structure-based virtual screening is computationally expensive owing to docking simulations. Thus, methods that filter out obviously unnecessary compounds prior to computationally expensive docking simulations have been proposed. However, the calculation speed of these methods is not fast enough to evaluate ≥ 10 million compounds. RESULTS: In this article, we propose a novel, docking-based pre-screening protocol named Spresso (Speedy PRE-Screening method with Segmented cOmpounds). Partial structures (fragments) are common among many compounds; therefore, the number of fragment variations needed for evaluation is smaller than that of compounds. Our method increases calculation speeds by ∼200-fold compared to conventional methods. AVAILABILITY AND IMPLEMENTATION: Spresso is written in C ++ and Python, and is available as an open-source code (http://www.bi.cs.titech.ac.jp/spresso/) under the GPLv3 license. CONTACT: akiyama@c.titech.ac.jp. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Simulação de Acoplamento Molecular/métodos , Proteínas/química , Software , Estrutura Terciária de Proteína , Proteínas/ultraestrutura
13.
Bioinformatics ; 30(22): 3281-3, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25100686

RESUMO

SUMMARY: The application of protein-protein docking in large-scale interactome analysis is a major challenge in structural bioinformatics and requires huge computing resources. In this work, we present MEGADOCK 4.0, an FFT-based docking software that makes extensive use of recent heterogeneous supercomputers and shows powerful, scalable performance of >97% strong scaling. AVAILABILITY AND IMPLEMENTATION: MEGADOCK 4.0 is written in C++ with OpenMPI and NVIDIA CUDA 5.0 (or later) and is freely available to all academic and non-profit users at: http://www.bi.cs.titech.ac.jp/megadock. CONTACT: akiyama@cs.titech.ac.jp SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Simulação de Acoplamento Molecular/métodos , Mapeamento de Interação de Proteínas/métodos , Software , Computadores
14.
Comput Struct Biotechnol J ; 23: 1214-1225, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38545599

RESUMO

Rapid advancements in protein sequencing technology have resulted in gaps between proteins with identified sequences and those with mapped structures. Although sequence-based predictions offer insights, they can be incomplete due to the absence of structural details. Conversely, structure-based methods face challenges with respect to newly sequenced proteins. The AlphaFold Multimer has remarkable accuracy in predicting the structure of protein complexes. However, it cannot distinguish whether the input protein sequences can interact. Nonetheless, by analyzing the information in the models predicted by the AlphaFold Multimer, we propose a highly accurate method for predicting protein interactions. This study focuses on the use of deep neural networks, specifically to analyze protein complex structures predicted by the AlphaFold Multimer. By transforming atomic coordinates and utilizing sophisticated image-processing techniques, vital 3D structural details were extracted from protein complexes. Recognizing the significance of evaluating residue distances in protein interactions, this study leveraged image recognition approaches by integrating Densely Connected Convolutional Networks (DenseNet) and Deep Residual Network (ResNet) within 3D convolutional networks for protein 3D structure analysis. When benchmarked against leading protein-protein interaction prediction methods, such as SpeedPPI, D-script, DeepTrio, and PEPPI, our proposed method, named SpatialPPI, exhibited notable efficacy, emphasizing the promising role of 3D spatial processing in advancing the realm of structural biology. The SpatialPPI code is available at: https://github.com/ohuelab/SpatialPPI.

15.
Commun Chem ; 7(1): 74, 2024 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-38580841

RESUMO

Graph Neural Networks (GNNs) excel in compound property and activity prediction, but the choice of molecular graph representations significantly influences model learning and interpretation. While atom-level molecular graphs resemble natural topology, they overlook key substructures or functional groups and their interpretation partially aligns with chemical intuition. Recent research suggests alternative representations using reduced molecular graphs to integrate higher-level chemical information and leverages both representations for model. However, there is a lack of studies about applicability and impact of different molecular graphs on model learning and interpretation. Here, we introduce MMGX (Multiple Molecular Graph eXplainable discovery), investigating the effects of multiple molecular graphs, including Atom, Pharmacophore, JunctionTree, and FunctionalGroup, on model learning and interpretation with various perspectives. Our findings indicate that multiple graphs relatively improve model performance, but in varying degrees depending on datasets. Interpretation from multiple graphs in different views provides more comprehensive features and potential substructures consistent with background knowledge. These results help to understand model decisions and offer valuable insights for subsequent tasks. The concept of multiple molecular graph representations and diverse interpretation perspectives has broad applicability across tasks, architectures, and explanation techniques, enhancing model learning and interpretation for relevant applications in drug discovery.

16.
BMC Res Notes ; 16(1): 229, 2023 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-37737185

RESUMO

MOTIVATION: Since the advent of ColabFold, numerous software packages have been provided with Google Colaboratory-compatible ipynb files, allowing users to effortlessly test and reproduce results without the need for local installation or configuration. MEGADOCK, a protein-protein docking tool, is particularly well-suited for Google Colaboratory due to its lightweight computations and GPU acceleration capabilities. To increase accessibility and promote widespread use, it is crucial to provide a computing environment compatible with Google Colaboratory. RESULTS: In this study, we report the development of a Google Colaboratory environment for running our protein-protein docking software, MEGADOCK. We provide a comprehensive ipynb file, including the compilation of MEGADOCK with the FFTW library installation on Colaboratory, the introduction of related tools using PyPI/apt, and the execution and visualization of docking structures. This streamlined environment enables users to visualize docking structures with just one click. The code is available under a CC-BY NC 4.0 license from https://github.com/ohuelab/MEGADOCK-on-Colab .


Assuntos
Corrida , Ferramenta de Busca , Biblioteca Gênica , Software
17.
Commun Chem ; 6(1): 249, 2023 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-37973971

RESUMO

The structural diversity of chemical libraries, which are systematic collections of compounds that have potential to bind to biomolecules, can be represented by chemical latent space. A chemical latent space is a projection of a compound structure into a mathematical space based on several molecular features, and it can express structural diversity within a compound library in order to explore a broader chemical space and generate novel compound structures for drug candidates. In this study, we developed a deep-learning method, called NP-VAE (Natural Product-oriented Variational Autoencoder), based on variational autoencoder for managing hard-to-analyze datasets from DrugBank and large molecular structures such as natural compounds with chirality, an essential factor in the 3D complexity of compounds. NP-VAE was successful in constructing the chemical latent space from large-sized compounds that were unable to be handled in existing methods, achieving higher reconstruction accuracy, and demonstrating stable performance as a generative model across various indices. Furthermore, by exploring the acquired latent space, we succeeded in comprehensively analyzing a compound library containing natural compounds and generating novel compound structures with optimized functions.

18.
Biomedicines ; 10(7)2022 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-35884931

RESUMO

New protein-protein interactions (PPIs) are identified, but PPIs have different physicochemical properties compared with conventional targets, making it difficult to use small molecules. Peptides offer a new modality to target PPIs, but designing appropriate peptide sequences by computation is challenging. Recently, AlphaFold and RoseTTAFold have made it possible to predict protein structures from amino acid sequences with ultra-high accuracy, enabling de novo protein design. We designed peptides likely to have PPI as the target protein using the "binder hallucination" protocol of AfDesign, a de novo protein design method using AlphaFold. However, the solubility of the peptides tended to be low. Therefore, we designed a solubility loss function using solubility indices for amino acids and developed a solubility-aware AfDesign binder hallucination protocol. The peptide solubility in sequences designed using the new protocol increased with the weight of the solubility loss function; moreover, they captured the characteristics of the solubility indices. Moreover, the new protocol sequences tended to have higher affinity than random or single residue substitution sequences when evaluated by docking binding affinity. Our approach shows that it is possible to design peptide sequences that can bind to the interface of PPI while controlling solubility.

19.
ACS Omega ; 7(34): 30265-30274, 2022 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-36061673

RESUMO

Virtual screening is a commonly used process to search for feasible drug candidates from a huge number of compounds during the early stages of drug design. As the compound database continues to expand to billions of entries or more, there remains an urgent need to accelerate the process of docking calculations. Reuse of calculation results is a possible way to accelerate the process. In this study, we first propose yet another virtual screening-oriented docking strategy by combining three factors, namely, compound decomposition, simplified fragment grid storing k-best scores, and flexibility consideration with pregenerated conformers. Candidate compounds contain many common fragments (chemical substructures). Thus, the calculation results of these common fragments can be reused among them. As a proof-of-concept of the aforementioned strategies, we also conducted the development of REstretto, a tool that implements the three factors to enable the reuse of calculation results. We demonstrated that the speed and accuracy of REstretto were comparable to those of AutoDock Vina, a well-known free docking tool. The implementation of REstretto has much room for further performance improvement, and therefore, the results show the feasibility of the strategy. The code is available under an MIT license at https://github.com/akiyamalab/restretto.

20.
Genome Inform ; 25(1): 25-39, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22230937

RESUMO

Elucidating protein-RNA interactions (PRIs) is important for understanding many cellular systems. We developed a PRI prediction method by using a rigid-body protein-RNA docking calculation with tertiary structure data. We evaluated this method by using 78 protein-RNA complex structures from the Protein Data Bank. We predicted the interactions for pairs in 78×78 combinations. Of these, 78 original complexes were defined as positive pairs, and the other 6,006 complexes were defined as negative pairs; then an F-measure value of 0.465 was obtained with our prediction system.


Assuntos
Simulação de Acoplamento Molecular , Proteínas de Ligação a RNA/metabolismo , RNA/metabolismo , Software , Sequência de Aminoácidos , Sequência de Bases , Dados de Sequência Molecular , Ligação Proteica , RNA/química , Proteínas de Ligação a RNA/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA