Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Comput Chem ; 2024 Jun 20.
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.

2.
J Comput Chem ; 41(1): 69-73, 2020 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-31410856

RESUMEN

Evaluation of ligand-binding affinity using the atomic coordinates of a protein-ligand complex is a challenge from the computational point of view. The availability of crystallographic structures of complexes with binding affinity data opens the possibility to create machine-learning models targeted to a specific protein system. Here, we describe a new methodology that combines a mass-spring system approach with supervised machine-learning techniques to predict the binding affinity of protein-ligand complexes. The combination of these techniques allows exploring the scoring function space, generating a model targeted to a protein system of interest. The new model shows superior predictive performance when compared with classical scoring functions implemented in the programs Molegro Virtual Docker, AutoDock4, and AutoDock Vina. We implemented this methodology in a new program named Taba. Taba is implemented in Python and available to download under the GNU license at https://github.com/azevedolab/taba. © 2019 Wiley Periodicals, Inc.


Asunto(s)
Proteínas/química , Programas Informáticos , Ligandos , Aprendizaje Automático , Modelos Moleculares , Termodinámica
3.
Biochem Biophys Res Commun ; 494(1-2): 305-310, 2017 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-29017921

RESUMEN

Here we report the development of a machine-learning model to predict binding affinity based on the crystallographic structures of protein-ligand complexes. We used an ensemble of crystallographic structures (resolution better than 1.5 Å resolution) for which half-maximal inhibitory concentration (IC50) data is available. Polynomial scoring functions were built using as explanatory variables the energy terms present in the MolDock and PLANTS scoring functions. Prediction performance was tested and the supervised machine learning models showed improvement in the prediction power, when compared with PLANTS and MolDock scoring functions. In addition, the machine-learning model was applied to predict binding affinity of CDK2, which showed a better performance when compared with AutoDock4, AutoDock Vina, MolDock, and PLANTS scores.


Asunto(s)
Antineoplásicos/química , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Aprendizaje Automático Supervisado , Quinasa 2 Dependiente de la Ciclina/química , Bases de Datos de Proteínas , Conjuntos de Datos como Asunto , Diseño de Fármacos , Humanos , Concentración 50 Inhibidora , Ligandos , Simulación del Acoplamiento Molecular , Curva ROC , Termodinámica
4.
Front Chem ; 11: 1128859, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36778030

RESUMEN

SARS-CoV-2 is the virus responsible for the COVID-19 pandemic. For the virus to enter the host cell, its spike (S) protein binds to the ACE2 receptor, and the transmembrane protease serine 2 (TMPRSS2) cleaves the binding for the fusion. As part of the research on COVID-19 treatments, several Casiopeina-analogs presented here were looked at as TMPRSS2 inhibitors. Using the DFT and conceptual-DFT methods, it was found that the global reactivity indices of the optimized molecular structures of the inhibitors could be used to predict their pharmacological activity. In addition, molecular docking programs (AutoDock4, Molegro Virtual Docker, and GOLD) were used to find the best potential inhibitors by looking at how they interact with key amino acid residues (His296, Asp 345, and Ser441) in the catalytic triad. The results show that in many cases, at least one of the amino acids in the triad is involved in the interaction. In the best cases, Asp435 interacts with the terminal nitrogen atoms of the side chains in a similar way to inhibitors such as nafamostat, camostat, and gabexate. Since the copper compounds localize just above the catalytic triad, they could stop substrates from getting into it. The binding energies are in the range of other synthetic drugs already on the market. Because serine protease could be an excellent target to stop the virus from getting inside the cell, the analyzed complexes are an excellent place to start looking for new drugs to treat COVID-19.

5.
J Struct Biol ; 169(3): 379-88, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19932753

RESUMEN

In humans, purine nucleoside phosphorylase (HsPNP) is responsible for degradation of deoxyguanosine, and genetic deficiency of this enzyme leads to profound T-cell mediated immunosuppression. HsPNP is a target for inhibitor development aiming at T-cell immune response modulation. Here we report the crystal structure of HsPNP in complex with 7-deazaguanine (HsPNP:7DG) at 2.75 A. Molecular dynamics simulations were employed to assess the structural features of HsPNP in both free form and in complex with 7DG. Our results show that some regions, responsible for entrance and exit of substrate, present a conformational variability, which is dissected by dynamics simulation analysis. Enzymatic assays were also carried out and revealed that 7-deazaguanine presents a lower inhibitory activity against HsPNP (K(i)=200 microM). The present structure may be employed in both structure-based design of PNP inhibitors and in development of specific empirical scoring functions.


Asunto(s)
Guanina/análogos & derivados , Simulación de Dinámica Molecular , Purina-Nucleósido Fosforilasa/química , Purina-Nucleósido Fosforilasa/metabolismo , Difracción de Rayos X/métodos , Guanina/química , Guanina/metabolismo , Humanos , Estructura Molecular , Análisis de Componente Principal , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Espectrometría de Fluorescencia
6.
Curr Med Chem ; 27(28): 4741-4749, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31490743

RESUMEN

BACKGROUND: Cannabinoid receptor 1 has its crystallographic structure available in complex with agonists and inverse agonists, which paved the way to establish an understanding of the structural basis of interactions with ligands. Dipyrone is a prodrug with analgesic capabilities and is widely used in some countries. Recently some evidence of a dipyrone metabolite acting over the Cannabinoid Receptor 1has been shown. OBJECTIVE: Our goal here is to explore the dipyrone metabolite 4-aminoantipyrine as a Cannabinoid Receptor 1 agonist, reviewing dipyrone characteristics, and investigating the structural basis for its interaction with the Cannabinoid Receptor 1. METHOD: We reviewed here recent functional studies related to the dipyrone metabolite focusing on its action as a Cannabinoid Receptor 1 agonist. We also analyzed protein-ligand interactions for this complex obtained through docking simulations against the crystallographic structure of the Cannabinoid Receptor 1. RESULTS: Analysis of the crystallographic structure and docking simulations revealed that most of the interactions present in the docked pose were also present in the crystallographic structure of Cannabinoid Receptor 1 and agonist. CONCLUSION: Analysis of the complex of 4-aminoantipyrine and Cannabinoid Receptor 1 revealed the pivotal role played by residues Phe 170, Phe 174, Phe 177, Phe 189, Leu 193, Val 196, and Phe 379, besides the conserved hydrogen bond at Ser 383. The mechanistic analysis and the present computational study suggest that the dipyrone metabolite 4-aminoantipyrine interacts with the Cannabinoid Receptor 1.


Asunto(s)
Agonistas de Receptores de Cannabinoides/farmacología , Ampirona , Analgésicos , Cannabinoides , Dipirona
7.
Curr Med Chem ; 27(5): 745-759, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-30501592

RESUMEN

BACKGROUND: The enzyme trans-enoyl-[acyl carrier protein] reductase (InhA) is a central protein for the development of antitubercular drugs. This enzyme is the target for the pro-drug isoniazid, which is catalyzed by the enzyme catalase-peroxidase (KatG) to become active. OBJECTIVE: Our goal here is to review the studies on InhA, starting with general aspects and focusing on the recent structural studies, with emphasis on the crystallographic structures of complexes involving InhA and inhibitors. METHOD: We start with a literature review, and then we describe recent studies on InhA crystallographic structures. We use this structural information to depict protein-ligand interactions. We also analyze the structural basis for inhibition of InhA. Furthermore, we describe the application of computational methods to predict binding affinity based on the crystallographic position of the ligands. RESULTS: Analysis of the structures in complex with inhibitors revealed the critical residues responsible for the specificity against InhA. Most of the intermolecular interactions involve the hydrophobic residues with two exceptions, the residues Ser 94 and Tyr 158. Examination of the interactions has shown that many of the key residues for inhibitor binding were found in mutations of the InhA gene in the isoniazid-resistant Mycobacterium tuberculosis. Computational prediction of the binding affinity for InhA has indicated a moderate uphill relationship with experimental values. CONCLUSION: Analysis of the structures involving InhA inhibitors shows that small modifications on these molecules could modulate their inhibition, which may be used to design novel antitubercular drugs specific for multidrug-resistant strains.


Asunto(s)
Mycobacterium tuberculosis , Proteína Transportadora de Acilo , Antituberculosos , Proteínas Bacterianas , Isoniazida , Oxidorreductasas
8.
Methods Mol Biol ; 2053: 35-50, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31452097

RESUMEN

Protein-ligand docking simulations are of central interest for computer-aided drug design. Docking is also of pivotal importance to understand the structural basis for protein-ligand binding affinity. In the last decades, we have seen an explosion in the number of three-dimensional structures of protein-ligand complexes available at the Protein Data Bank. These structures gave further support for the development and validation of in silico approaches to address the binding of small molecules to proteins. As a result, we have now dozens of open source programs and web servers to carry out molecular docking simulations. The development of the docking programs and the success of such simulations called the attention of a broad spectrum of researchers not necessarily familiar with computer simulations. In this scenario, it is essential for those involved in experimental studies of protein-ligand interactions and biophysical techniques to have a glimpse of the basics of the protein-ligand docking simulations. Applications of protein-ligand docking simulations to drug development and discovery were able to identify hits, inhibitors, and even drugs. In the present chapter, we cover the fundamental ideas behind protein-ligand docking programs for non-specialists, which may benefit from such knowledge when studying molecular recognition mechanism.


Asunto(s)
Ligandos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Proteínas/química , Algoritmos , Sitios de Unión , Diseño de Fármacos , Conformación Molecular , Unión Proteica , Relación Estructura-Actividad , Flujo de Trabajo
9.
Methods Mol Biol ; 2053: 51-65, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31452098

RESUMEN

Since the early 1980s, we have witnessed considerable progress in the development and application of docking programs to assess protein-ligand interactions. Most of these applications had as a goal the identification of potential new binders to protein targets. Another remarkable progress is taking place in the determination of the structures of protein-ligand complexes, mostly using X-ray diffraction crystallography. Considering these developments, we have a favorable scenario for the creation of a computational tool that integrates into one workflow all steps involved in molecular docking simulations. We had these goals in mind when we developed the program SAnDReS. This program allows the integration of all computational features related to modern docking studies into one workflow. SAnDReS not only carries out docking simulations but also evaluates several docking protocols allowing the selection of the best approach for a given protein system. SAnDReS is a free and open-source (GNU General Public License) computational environment for running docking simulations. Here, we describe the combination of SAnDReS and AutoDock4 for protein-ligand docking simulations. AutoDock4 is a free program that has been applied to over a thousand receptor-ligand docking simulations. The dataset described in this chapter is available for downloading at https://github.com/azevedolab/sandres.


Asunto(s)
Biología Computacional/métodos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Programas Informáticos , Sitios de Unión , Bases de Datos Factuales , Diseño de Fármacos , Ligandos , Unión Proteica , Proteínas/química , Interfaz Usuario-Computador , Navegador Web
10.
Methods Mol Biol ; 2053: 109-124, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31452102

RESUMEN

X-ray diffraction crystallography is the primary technique to determine the three-dimensional structures of biomolecules. Although a robust method, X-ray crystallography is not able to access the dynamical behavior of macromolecules. To do so, we have to carry out molecular dynamics simulations taking as an initial system the three-dimensional structure obtained from experimental techniques or generated using homology modeling. In this chapter, we describe in detail a tutorial to carry out molecular dynamics simulations using the program NAMD2. We chose as a molecular system to simulate the structure of human cyclin-dependent kinase 2.


Asunto(s)
Simulación de Dinámica Molecular , Programas Informáticos , Adenosina Trifosfato/química , Algoritmos , Cristalografía por Rayos X , Quinasa 2 Dependiente de la Ciclina/química , Humanos , Conformación Proteica , Electricidad Estática , Interfaz Usuario-Computador
11.
Methods Mol Biol ; 2053: 149-167, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31452104

RESUMEN

Molegro Virtual Docker is a protein-ligand docking simulation program that allows us to carry out docking simulations in a fully integrated computational package. MVD has been successfully applied to hundreds of different proteins, with docking performance similar to other docking programs such as AutoDock4 and AutoDock Vina. The program MVD has four search algorithms and four native scoring functions. Considering that we may have water molecules or not in the docking simulations, we have a total of 32 docking protocols. The integration of the programs SAnDReS ( https://github.com/azevedolab/sandres ) and MVD opens the possibility to carry out a detailed statistical analysis of docking results, which adds to the native capabilities of the program MVD. In this chapter, we describe a tutorial to carry out docking simulations with MVD and how to perform a statistical analysis of the docking results with the program SAnDReS. To illustrate the integration of both programs, we describe the redocking simulation focused the cyclin-dependent kinase 2 in complex with a competitive inhibitor.


Asunto(s)
Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Programas Informáticos , Sitios de Unión , Quinasa 2 Dependiente de la Ciclina/química , Diseño de Fármacos , Humanos , Ligandos , Unión Proteica , Proteínas/química , Interfaz Usuario-Computador
12.
Methods Mol Biol ; 2053: 169-188, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31452105

RESUMEN

GEMDOCK is a protein-ligand docking software that makes use of an elegant biologically inspired computational methodology based on the differential evolution algorithm. As any docking program, GEMDOCK has two major features to predict the binding of a small-molecule ligand to the binding site of a protein target: the search algorithm and the scoring function to evaluate the generated poses. The GEMDOCK scoring function uses a piecewise potential energy function integrated into the differential evolutionary algorithm. GEMDOCK has been applied to a wide range of protein systems with docking accuracy similar to other docking programs such as Molegro Virtual Docker, AutoDock4, and AutoDock Vina. In this chapter, we explain how to carry out protein-ligand docking simulations with GEMDOCK. We focus this tutorial on the protein target cyclin-dependent kinase 2.


Asunto(s)
Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Proteínas/química , Programas Informáticos , Adenosina Trifosfato/química , Quinasa 2 Dependiente de la Ciclina/química , Bases de Datos de Proteínas , Diseño de Fármacos , Ligandos , Interfaz Usuario-Computador
13.
Methods Mol Biol ; 2053: 189-202, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31452106

RESUMEN

Protein-ligand docking simulation is central in drug design and development. Therefore, the development of web servers intended to docking simulations is of pivotal importance. SwissDock is a web server dedicated to carrying out protein-ligand docking simulation intuitively and elegantly. SwissDock is based on the protein-ligand docking program EADock DSS and has a simple and integrated interface. The SwissDock allows the user to upload structure files for a protein and a ligand, and returns the results by e-mail. To facilitate the upload of the protein and ligand files, we can prepare these input files using the program UCSF Chimera. In this chapter, we describe how to use UCSF Chimera and SwissDock to perform protein-ligand docking simulations. To illustrate the process, we describe the molecular docking of the competitive inhibitor roscovitine against the structure of human cyclin-dependent kinase 2.


Asunto(s)
Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Programas Informáticos , Algoritmos , Quinasa 2 Dependiente de la Ciclina/química , Diseño de Fármacos , Humanos , Ligandos , Proteínas/química , Interfaz Usuario-Computador , Navegador Web
14.
Methods Mol Biol ; 2053: 203-220, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31452107

RESUMEN

Molecular docking is the major computational technique employed in the early stages of computer-aided drug discovery. The availability of free software to carry out docking simulations of protein-ligand systems has allowed for an increasing number of studies using this technique. Among the available free docking programs, we discuss the use of ArgusLab ( http://www.arguslab.com/arguslab.com/ArgusLab.html ) for protein-ligand docking simulation. This easy-to-use computational tool makes use of a genetic algorithm as a search algorithm and a fast scoring function that allows users with minimal experience in the simulations of protein-ligand simulations to carry out docking simulations. In this chapter, we present a detailed tutorial to perform docking simulations using ArgusLab.


Asunto(s)
Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Programas Informáticos , Algoritmos , Sitios de Unión , Diseño de Fármacos , Ligandos , Unión Proteica , Proteínas/química , Interfaz Usuario-Computador , Navegador Web
15.
Methods Mol Biol ; 2053: 231-249, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31452109

RESUMEN

Homology modeling is a computational approach to generate three-dimensional structures of protein targets when experimental data about similar proteins are available. Although experimental methods such as X-ray crystallography and nuclear magnetic resonance spectroscopy successfully solved the structures of nearly 150,000 macromolecules, there is still a gap in our structural knowledge. We can fulfill this gap with computational methodologies. Our goal in this chapter is to explain how to perform homology modeling of protein targets for drug development. We choose as a homology modeling tool the program MODELLER. To illustrate its use, we describe how to model the structure of human cyclin-dependent kinase 3 using MODELLER. We explain the modeling procedure of CDK3 apoenzyme and the structure of this enzyme in complex with roscovitine.


Asunto(s)
Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Proteínas/química , Programas Informáticos , Secuencia de Aminoácidos , Diseño de Fármacos , Humanos , Conformación Proteica , Homología Estructural de Proteína , Interfaz Usuario-Computador , Navegador Web
16.
Methods Mol Biol ; 2053: 251-273, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31452110

RESUMEN

Recent progress in the development of scientific libraries with machine-learning techniques paved the way for the implementation of integrated computational tools to predict ligand-binding affinity. The prediction of binding affinity uses the atomic coordinates of protein-ligand complexes. These new computational tools made application of a broad spectrum of machine-learning techniques to study protein-ligand interactions possible. The essential aspect of these machine-learning approaches is to train a new computational model by using technologies such as supervised machine-learning techniques, convolutional neural network, and random forest to mention the most commonly applied methods. In this chapter, we focus on supervised machine-learning techniques and their applications in the development of protein-targeted scoring functions for the prediction of binding affinity. We discuss the development of the program SAnDReS and its application to the creation of machine-learning models to predict inhibition of cyclin-dependent kinase and HIV-1 protease. Moreover, we describe the scoring function space, and how to use it to explain the development of targeted scoring functions.


Asunto(s)
Aprendizaje Automático , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Proteínas/química , Programas Informáticos , Algoritmos , Quinasa 2 Dependiente de la Ciclina/química , Bases de Datos de Proteínas , Proteasa del VIH/química , Humanos , Ligandos , Modelos Estadísticos , Aprendizaje Automático Supervisado , Navegador Web
17.
Methods Mol Biol ; 2053: 275-281, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31452111

RESUMEN

In the analysis of protein-ligand interactions, two abstractions have been widely employed to build a systematic approach to analyze these complexes: protein and chemical spaces. The pioneering idea of the protein space dates back to 1970, and the chemical space is newer, later 1990s. With the progress of computational methodologies to create machine-learning models to predict the ligand-binding affinity, clearly there is a need for novel approaches to the problem of protein-ligand interactions. New abstractions are required to guide the conceptual analysis of the molecular recognition problem. Using a systems approach, we proposed to address protein-ligand scoring functions using the modern idea of the scoring function space. In this chapter, we describe the fundamental concept behind the scoring function space and how it has been applied to develop the new generation of targeted-scoring functions.


Asunto(s)
Aprendizaje Automático , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Proteínas/química , Programas Informáticos , Algoritmos , Navegador Web
18.
Curr Med Chem ; 26(10): 1908-1919, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-29667549

RESUMEN

BACKGROUND: Cannabinoid Receptor 1 (CB1) is a membrane protein prevalent in the central nervous system, whose crystallographic structure has recently been solved. Studies will be needed to investigate CB1 complexes with its ligands and its role in the development of new drugs. OBJECTIVE: Our goal here is to review the studies on CB1, starting with general aspects and focusing on the recent structural studies, with emphasis on the inverse agonists bound structures. METHODS: We start with a literature review, and then we describe recent studies on CB 1 crystallographic structure and docking simulations. We use this structural information to depict protein-ligand interactions. We also describe the molecular docking method to obtain complex structures of CB 1 with inverse agonists. RESULTS: Analysis of the crystallographic structure and docking results revealed the residues responsible for the specificity of the inverse agonists for CB 1. Most of the intermolecular interactions involve hydrophobic residues, with the participation of the residues Phe 170 and Leu 359 in all complex structures investigated in the present study. For the complexes with otenabant and taranabant, we observed intermolecular hydrogen bonds involving residues His 178 (otenabant) and Thr 197 and Ser 383 (taranabant). CONCLUSION: Analysis of the structures involving inverse agonists and CB 1 revealed the pivotal role played by residues Phe 170 and Leu 359 in their interactions and the strong intermolecular hydrogen bonds highlighting the importance of the exploration of intermolecular interactions in the development of novel inverse agonists.


Asunto(s)
Agonistas de Receptores de Cannabinoides/metabolismo , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/metabolismo , Agonistas de Receptores de Cannabinoides/química , Cristalografía por Rayos X , Agonismo Inverso de Drogas , Humanos , Leucina/química , Ligandos , Simulación del Acoplamiento Molecular , Fenilalanina/química , Unión Proteica , Receptor Cannabinoide CB1/química
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA