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.
J Comput Chem ; 45(27): 2333-2346, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-38900052

RESUMEN

Classical scoring functions may exhibit low accuracy in determining ligand binding affinity for proteins. The availability of both protein-ligand structures and affinity data make it possible to develop machine-learning models focused on specific protein systems with superior predictive performance. Here, we report a new methodology named SAnDReS that combines AutoDock Vina 1.2 with 54 regression methods available in Scikit-Learn to calculate binding affinity based on protein-ligand structures. This approach allows exploration of the scoring function space. SAnDReS generates machine-learning models based on crystal, docked, and AlphaFold-generated structures. As a proof of concept, we examine the performance of SAnDReS-generated models in three case studies. For all three cases, our models outperformed classical scoring functions. Also, SAnDReS-generated models showed predictive performance close to or better than other machine-learning models such as KDEEP, CSM-lig, and ΔVinaRF20. SAnDReS 2.0 is available to download at https://github.com/azevedolab/sandres.


Asunto(s)
Aprendizaje Automático , Proteínas , Proteínas/química , Proteínas/metabolismo , Ligandos , Programas Informáticos , Simulación del Acoplamiento Molecular
2.
Curr Med Chem ; 29(14): 2438-2455, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34365938

RESUMEN

BACKGROUND: CDK2 participates in the control of eukaryotic cell-cycle progression. Due to the great interest in CDK2 for drug development and the relative easiness in crystallizing this enzyme, we have over 400 structural studies focused on this protein target. This structural data is the basis for the development of computational models to estimate CDK2-ligand binding affinity. OBJECTIVE: This work focuses on the recent developments in the application of supervised machine learning modeling to develop scoring functions to predict the binding affinity of CDK2. METHOD: We employed the structures available at the protein data bank and the ligand information accessed from the BindingDB, Binding MOAD, and PDBbind to evaluate the predictive performance of machine learning techniques combined with physical modeling used to calculate binding affinity. We compared this hybrid methodology with classical scoring functions available in docking programs. RESULTS: Our comparative analysis of previously published models indicated that a model created using a combination of a mass-spring system and cross-validated Elastic Net to predict the binding affinity of CDK2-inhibitor complexes outperformed classical scoring functions available in AutoDock4 and AutoDock Vina. CONCLUSION: All studies reviewed here suggest that targeted machine learning models are superior to classical scoring functions to calculate binding affinities. Specifically for CDK2, we see that the combination of physical modeling with supervised machine learning techniques exhibits improved predictive performance to calculate the protein-ligand binding affinity. These results find theoretical support in the application of the concept of scoring function space.


Asunto(s)
Antineoplásicos , Neoplasias , Antineoplásicos/farmacología , Quinasa 2 Dependiente de la Ciclina/metabolismo , Bases de Datos de Proteínas , Descubrimiento de Drogas , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Unión Proteica , Proteínas/metabolismo
3.
Curr Med Chem ; 28(34): 7006-7022, 2021 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-33568025

RESUMEN

BACKGROUND: One of the main challenges in the early stages of drug discovery is the computational assessment of protein-ligand binding affinity. Machine learning techniques can contribute to predicting this type of interaction. We may apply these techniques following two approaches. Firstly, using the experimental structures for which affinity data is available. Secondly, using protein-ligand docking simulations. OBJECTIVE: In this review, we describe recently published machine learning models based on crystal structures, for which binding affinity and thermodynamic data are available. METHOD: We used experimental structures available at the protein data bank and binding affinity and thermodynamic data was accessed through BindingDB, Binding MOAD, and PDBbind databases. We reviewed machine learning models to predict binding created using open source programs, such as SAnDReS and Taba. RESULTS: Analysis of machine learning models trained against datasets, composed of crystal structure complexes indicated the high predictive performance of these models when compared with classical scoring functions. CONCLUSION: The rapid increase in the number of crystal structures of protein-ligand complexes created a favorable scenario for developing machine learning models to predict binding affinity. These models rely on experimental data from two sources, the structural and the affinity data. The combination of experimental data generates computational models that outperform the classical scoring functions.


Asunto(s)
Aprendizaje Automático , Proteínas , Bases de Datos de Proteínas , Ligandos , Simulación del Acoplamiento Molecular , Unión Proteica , Proteínas/metabolismo
4.
Methods Mol Biol ; 2053: 67-77, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31452099

RESUMEN

Computational analysis of protein-ligand interactions is of pivotal importance for drug design. Assessment of ligand binding energy allows us to have a glimpse of the potential of a small organic molecule as a ligand to the binding site of a protein target. Considering scoring functions available in docking programs such as AutoDock4, AutoDock Vina, and Molegro Virtual Docker, we could say that they all rely on equations that sum each type of protein-ligand interactions to model the binding affinity. Most of the scoring functions consider electrostatic interactions involving the protein and the ligand. In this chapter, we present the main physics concepts necessary to understand electrostatics interactions relevant to molecular recognition of a ligand by the binding pocket of a protein target. Moreover, we analyze the electrostatic potential energy for an ensemble of structures to highlight the main features related to the importance of this interaction for binding affinity.


Asunto(s)
Ligandos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Proteínas/química , Electricidad Estática , Algoritmos , Sitios de Unión , Diseño de Fármacos , Modelos Moleculares , Unión Proteica
5.
Methods Mol Biol ; 2053: 79-91, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31452100

RESUMEN

Van der Waals forces are determinants of the formation of protein-ligand complexes. Physical models based on the Lennard-Jones potential can estimate van der Waals interactions with considerable accuracy and with a computational complexity that allows its application to molecular docking simulations and virtual screening of large databases of small organic molecules. Several empirical scoring functions used to evaluate protein-ligand interactions approximate van der Waals interactions with the Lennard-Jones potential. In this chapter, we present the main concepts necessary to understand van der Waals interactions relevant to molecular recognition of a ligand by the binding pocket of a protein target. We describe the Lennard-Jones potential and its application to calculate potential energy for an ensemble of structures to highlight the main features related to the importance of this interaction for binding affinity.


Asunto(s)
Diseño de Fármacos , Modelos Teóricos , Complejos Multiproteicos/química , Proteínas/química , Algoritmos , Ligandos
6.
Methods Mol Biol ; 2053: 93-107, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31452101

RESUMEN

Fast and reliable evaluation of the hydrogen bond potential energy has a significant impact in the drug design and development since it allows the assessment of large databases of organic molecules in virtual screening projects focused on a protein of interest. Semi-empirical force fields implemented in molecular docking programs make it possible the evaluation of protein-ligand binding affinity where the hydrogen bond potential is a common term used in the calculation. In this chapter, we describe the concepts behind the programs used to predict hydrogen bond potential energy employing semi-empirical force fields as the ones available in the programs AMBER, AutoDock4, TreeDock, and ReplicOpter. We described here the 12-10 potential and applied it to evaluate the binding affinity for an ensemble of crystallographic structures for which experimental data about binding affinity are available.


Asunto(s)
Enlace de Hidrógeno , Ligandos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Proteínas/química , Algoritmos , Diseño de Fármacos , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA