Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Molecules ; 29(12)2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38930976

RESUMEN

Accurately predicting drug-target interactions is a critical yet challenging task in drug discovery. Traditionally, pocket detection and drug-target affinity prediction have been treated as separate aspects of drug-target interaction, with few methods combining these tasks within a unified deep learning system to accelerate drug development. In this study, we propose EMPDTA, an end-to-end framework that integrates protein pocket prediction and drug-target affinity prediction to provide a comprehensive understanding of drug-target interactions. The EMPDTA framework consists of three main modules: pocket online detection, multimodal representation learning for affinity prediction, and multi-task joint training. The performance and potential of the proposed framework have been validated across diverse benchmark datasets, achieving robust results in both tasks. Furthermore, the visualization results of the predicted pockets demonstrate accurate pocket detection, confirming the effectiveness of our framework.


Asunto(s)
Descubrimiento de Drogas , Descubrimiento de Drogas/métodos , Proteínas/química , Proteínas/metabolismo , Aprendizaje Profundo , Unión Proteica , Sitios de Unión , Humanos , Algoritmos
2.
Comput Struct Biotechnol J ; 23: 1320-1338, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38585646

RESUMEN

Many research groups and institutions have created a variety of databases curating experimental and predicted data related to protein-ligand binding. The landscape of available databases is dynamic, with new databases emerging and established databases becoming defunct. Here, we review the current state of databases that contain binding pockets and protein-ligand binding interactions. We have compiled a list of such databases, fifty-three of which are currently available for use. We discuss variation in how binding pockets are defined and summarize pocket-finding methods. We organize the fifty-three databases into subgroups based on goals and contents, and describe standard use cases. We also illustrate that pockets within the same protein are characterized differently across different databases. Finally, we assess critical issues of sustainability, accessibility and redundancy.

3.
Cell Syst ; 14(8): 692-705.e6, 2023 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-37516103

RESUMEN

Protein-ligand interactions are essential for cellular activities and drug discovery processes. Appropriately and effectively representing protein features is of vital importance for developing computational approaches, especially data-driven methods, for predicting protein-ligand interactions. However, existing approaches may not fully investigate the features of the ligand-occupying regions in the protein pockets. Here, we design a structure-based protein representation method, named PocketAnchor, for capturing the local environmental and spatial features of protein pockets to facilitate protein-ligand interaction-related learning tasks. We define "anchors" as probe points reaching into the cavities and those located near the surface of proteins, and we design a specific message passing strategy for gathering local information from the atoms and surface neighboring these anchors. Comprehensive evaluation of our method demonstrated its successful applications in pocket detection and binding affinity prediction, which indicated that our anchor-based approach can provide effective protein feature representations for improving the prediction of protein-ligand interactions.


Asunto(s)
Algoritmos , Proteínas , Sitios de Unión , Ligandos , Proteínas/metabolismo
4.
Int J Mol Sci ; 23(9)2022 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-35563148

RESUMEN

The prediction of how a ligand binds to its target is an essential step for Structure-Based Drug Design (SBDD) methods. Molecular docking is a standard tool to predict the binding mode of a ligand to its macromolecular receptor and to quantify their mutual complementarity, with multiple applications in drug design. However, docking programs do not always find correct solutions, either because they are not sampled or due to inaccuracies in the scoring functions. Quantifying the docking performance in real scenarios is essential to understanding their limitations, managing expectations and guiding future developments. Here, we present a fully automated pipeline for pose prediction validated by participating in the Continuous Evaluation of Ligand Pose Prediction (CELPP) Challenge. Acknowledging the intrinsic limitations of the docking method, we devised a strategy to automatically mine and exploit pre-existing data, defining-whenever possible-empirical restraints to guide the docking process. We prove that the pipeline is able to generate predictions for most of the proposed targets as well as obtain poses with low RMSD values when compared to the crystal structure. All things considered, our pipeline highlights some major challenges in the automatic prediction of protein-ligand complexes, which will be addressed in future versions of the pipeline.


Asunto(s)
Diseño de Fármacos , Sitios de Unión , Cristalografía por Rayos X , Ligandos , Simulación del Acoplamiento Molecular , Unión Proteica , Conformación Proteica
5.
Methods Mol Biol ; 2165: 1-11, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32621216

RESUMEN

Structural data of biomolecules, such as those of proteins and nucleic acids, provide much information for estimation of their functions. For structure-unknown proteins, structure information is obtainable by modeling their structures based on sequence similarity of proteins. Moreover, information related to ligands or ligand-binding sites is necessary to elucidate protein functions because the binding of ligands can engender not only the activation and inactivation of the proteins but also the modification of protein functions. This chapter presents methods using our profile-profile alignment server FORTE and the PoSSuM ligand-binding site database for prediction of the structure and potential ligand-binding sites of structure-unknown and function-unknown proteins, aimed at protein function prediction.


Asunto(s)
Simulación del Acoplamiento Molecular/métodos , Conformación Proteica , Alineación de Secuencia/métodos , Programas Informáticos , Sitios de Unión , Humanos , Ligandos , Unión Proteica
6.
BMC Bioinformatics ; 18(1): 493, 2017 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-29145826

RESUMEN

BACKGROUND: Protein cavities play a key role in biomolecular recognition and function, particularly in protein-ligand interactions, as usual in drug discovery and design. Grid-based cavity detection methods aim at finding cavities as aggregates of grid nodes outside the molecule, under the condition that such cavities are bracketed by nodes on the molecule surface along a set of directions (not necessarily aligned with coordinate axes). Therefore, these methods are sensitive to scanning directions, a problem that we call cavity ground-and-walls ambiguity, i.e., they depend on the position and orientation of the protein in the discretized domain. Also, it is hard to distinguish grid nodes belonging to protein cavities amongst all those outside the protein, a problem that we call cavity ceiling ambiguity. RESULTS: We solve those two ambiguity problems using two implicit isosurfaces of the protein, the protein surface itself (called inner isosurface) that excludes all its interior nodes from any cavity, and the outer isosurface that excludes most of its exterior nodes from any cavity. Summing up, the cavities are formed from nodes located between these two isosurfaces. It is worth noting that these two surfaces do not need to be evaluated (i.e., sampled), triangulated, and rendered on the screen to find the cavities in between; their defining analytic functions are enough to determine which grid nodes are in the empty space between them. CONCLUSION: This article introduces a novel geometric algorithm to detect cavities on the protein surface that takes advantage of the real analytic functions describing two Gaussian surfaces of a given protein.


Asunto(s)
Algoritmos , Proteínas/química , Ligandos , Distribución Normal , Proteínas/metabolismo , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA