Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
Front Chem ; 11: 1128859, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36778030

RESUMO

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.

4.
Curr Med Chem ; 27(5): 745-759, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-30501592

RESUMO

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.


Assuntos
Mycobacterium tuberculosis , Proteína de Transporte de Acila , Antituberculosos , Proteínas de Bactérias , Isoniazida , Oxirredutases
5.
Curr Med Chem ; 27(28): 4741-4749, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31490743

RESUMO

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.


Assuntos
Agonistas de Receptores de Canabinoides/farmacologia , Ampirona , Analgésicos , Canabinoides , Dipirona
6.
J Comput Chem ; 41(1): 69-73, 2020 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-31410856

RESUMO

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.


Assuntos
Proteínas/química , Software , Ligantes , Aprendizado de Máquina , Modelos Moleculares , Termodinâmica
7.
Methods Mol Biol ; 2053: 35-50, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31452097

RESUMO

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.


Assuntos
Ligantes , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Proteínas/química , Algoritmos , Sítios de Ligação , Desenho de Fármacos , Conformação Molecular , Ligação Proteica , Relação Estrutura-Atividade , Fluxo de Trabalho
8.
Methods Mol Biol ; 2053: 51-65, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31452098

RESUMO

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.


Assuntos
Biologia Computacional/métodos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Software , Sítios de Ligação , Bases de Dados Factuais , Desenho de Fármacos , Ligantes , Ligação Proteica , Proteínas/química , Interface Usuário-Computador , Navegador
9.
Methods Mol Biol ; 2053: 67-77, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31452099

RESUMO

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.


Assuntos
Ligantes , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Proteínas/química , Eletricidade Estática , Algoritmos , Sítios de Ligação , Desenho de Fármacos , Modelos Moleculares , Ligação Proteica
10.
Methods Mol Biol ; 2053: 79-91, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31452100

RESUMO

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.


Assuntos
Desenho de Fármacos , Modelos Teóricos , Complexos Multiproteicos/química , Proteínas/química , Algoritmos , Ligantes
11.
Methods Mol Biol ; 2053: 93-107, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31452101

RESUMO

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.


Assuntos
Ligação de Hidrogênio , Ligantes , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Proteínas/química , Algoritmos , Desenho de Fármacos , Ligação Proteica
12.
Methods Mol Biol ; 2053: 109-124, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31452102

RESUMO

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.


Assuntos
Simulação de Dinâmica Molecular , Software , Trifosfato de Adenosina/química , Algoritmos , Cristalografia por Raios X , Quinase 2 Dependente de Ciclina/química , Humanos , Conformação Proteica , Eletricidade Estática , Interface Usuário-Computador
13.
Methods Mol Biol ; 2053: 125-148, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31452103

RESUMO

AutoDock is one of the most popular receptor-ligand docking simulation programs. It was first released in the early 1990s and is in continuous development and adapted to specific protein targets. AutoDock has been applied to a wide range of biological systems. It has been used not only for protein-ligand docking simulation but also for the prediction of binding affinity with good correlation with experimental binding affinity for several protein systems. The latest version makes use of a semi-empirical force field to evaluate protein-ligand binding affinity and for selecting the lowest energy pose in docking simulation. AutoDock4.2.6 has an arsenal of four search algorithms to carry out docking simulation including simulated annealing, genetic algorithm, and Lamarckian algorithm. In this chapter, we describe a tutorial about how to perform docking with AutoDock4. We focus our simulations on the protein target cyclin-dependent kinase 2.


Assuntos
Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Software , Trifosfato de Adenosina/química , Quinase 2 Dependente de Ciclina/química , Desenho de Fármacos , Ligação de Hidrogênio , Ligantes , Conformação Molecular , Ligação Proteica , Proteínas/química , Interface Usuário-Computador
14.
Methods Mol Biol ; 2053: 149-167, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31452104

RESUMO

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.


Assuntos
Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Software , Sítios de Ligação , Quinase 2 Dependente de Ciclina/química , Desenho de Fármacos , Humanos , Ligantes , Ligação Proteica , Proteínas/química , Interface Usuário-Computador
15.
Methods Mol Biol ; 2053: 169-188, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31452105

RESUMO

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.


Assuntos
Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Proteínas/química , Software , Trifosfato de Adenosina/química , Quinase 2 Dependente de Ciclina/química , Bases de Dados de Proteínas , Desenho de Fármacos , Ligantes , Interface Usuário-Computador
16.
Methods Mol Biol ; 2053: 189-202, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31452106

RESUMO

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.


Assuntos
Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Software , Algoritmos , Quinase 2 Dependente de Ciclina/química , Desenho de Fármacos , Humanos , Ligantes , Proteínas/química , Interface Usuário-Computador , Navegador
17.
Methods Mol Biol ; 2053: 203-220, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31452107

RESUMO

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.


Assuntos
Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Software , Algoritmos , Sítios de Ligação , Desenho de Fármacos , Ligantes , Ligação Proteica , Proteínas/química , Interface Usuário-Computador , Navegador
18.
Methods Mol Biol ; 2053: 231-249, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31452109

RESUMO

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.


Assuntos
Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Proteínas/química , Software , Sequência de Aminoácidos , Desenho de Fármacos , Humanos , Conformação Proteica , Homologia Estrutural de Proteína , Interface Usuário-Computador , Navegador
19.
Methods Mol Biol ; 2053: 251-273, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31452110

RESUMO

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.


Assuntos
Aprendizado de Máquina , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Proteínas/química , Software , Algoritmos , Quinase 2 Dependente de Ciclina/química , Bases de Dados de Proteínas , Protease de HIV/química , Humanos , Ligantes , Modelos Estatísticos , Aprendizado de Máquina Supervisionado , Navegador
20.
Methods Mol Biol ; 2053: 275-281, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31452111

RESUMO

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.


Assuntos
Aprendizado de Máquina , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Proteínas/química , Software , Algoritmos , Navegador
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...