Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Bioorg Med Chem ; 100: 117588, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38295487

RESUMEN

Microsatellite instability (MSI) is a hypermutable condition caused by DNA mismatch repair system defects, contributing to the development of various cancer types. Recent research has identified Werner syndrome ATP-dependent helicase (WRN) as a promising synthetic lethal target for MSI cancers. Herein, we report the first discovery of thiophen-2-ylmethylene bis-dimedone derivatives as novel WRN inhibitors for MSI cancer therapy. Initial computational analysis and biological evaluation identified a new scaffold for a WRN inhibitor. Subsequent SAR study led to the discovery of a highly potent WRN inhibitor. Furthermore, we demonstrated that the optimal compound induced DNA damage and apoptotic cell death in MSI cancer cells by inhibiting WRN. This study provides a new pharmacophore for WRN inhibitors, emphasizing their therapeutic potential for MSI cancers.


Asunto(s)
Inestabilidad de Microsatélites , Neoplasias , Tiofenos , Humanos , Ciclohexanonas , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Helicasa del Síndrome de Werner/antagonistas & inhibidores , Helicasa del Síndrome de Werner/metabolismo , Tiofenos/química , Tiofenos/farmacología
2.
Semin Cancer Biol ; 68: 84-91, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-31698087

RESUMEN

A pre-eminent subtype of lung carcinoma, Non-small cell lung cancer accounts for paramount causes of cancer-associated mortality worldwide. Undeterred by the endeavour in the treatment strategies, the overall cure and survival rates for NSCLC remain substandard, particularly in metastatic diseases. Moreover, the emergence of resistance to classic anticancer drugs further deteriorates the situation. These demanding circumstances culminate the need of extended and revamped research for the establishment of upcoming generation cancer therapeutics. Drug repositioning introduces an affordable and efficient strategy to discover novel drug action, especially when integrated with recent systems biology driven stratagem. This review illustrates the trendsetting approaches in repurposing along with their numerous success stories with an emphasize on the NSCLC therapeutics. Indeed, these novel hits, in combination with conventional anticancer agents, will ideally make their way the clinics and strengthen the therapeutic arsenal to combat drug resistance in the near future.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Descubrimiento de Drogas , Reposicionamiento de Medicamentos/métodos , Neoplasias Pulmonares/tratamiento farmacológico , Polifarmacología/métodos , Animales , Humanos
3.
Proteins ; 88(8): 948-961, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31697428

RESUMEN

We report the performance of the protein docking prediction pipeline of our group and the results for Critical Assessment of Prediction of Interactions (CAPRI) rounds 38-46. The pipeline integrates programs developed in our group as well as other existing scoring functions. The core of the pipeline is the LZerD protein-protein docking algorithm. If templates of the target complex are not found in PDB, the first step of our docking prediction pipeline is to run LZerD for a query protein pair. Meanwhile, in the case of human group prediction, we survey the literature to find information that can guide the modeling, such as protein-protein interface information. In addition to any literature information and binding residue prediction, generated docking decoys were selected by a rank aggregation of statistical scoring functions. The top 10 decoys were relaxed by a short molecular dynamics simulation before submission to remove atom clashes and improve side-chain conformations. In these CAPRI rounds, our group, particularly the LZerD server, showed robust performance. On the other hand, there are failed cases where some other groups were successful. To understand weaknesses of our pipeline, we analyzed sources of errors for failed targets. Since we noted that structure refinement is a step that needs improvement, we newly performed a comparative study of several refinement approaches. Finally, we show several examples that illustrate successful and unsuccessful cases by our group.


Asunto(s)
Simulación del Acoplamiento Molecular , Péptidos/química , Proteínas/química , Programas Informáticos , Algoritmos , Secuencia de Aminoácidos , Sitios de Unión , Humanos , Ligandos , Péptidos/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Proteínas/metabolismo , Proyectos de Investigación , Homología Estructural de Proteína
4.
Int J Mol Sci ; 21(22)2020 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-33182567

RESUMEN

Accurate prediction of the binding affinity of a protein-ligand complex is essential for efficient and successful rational drug design. Therefore, many binding affinity prediction methods have been developed. In recent years, since deep learning technology has become powerful, it is also implemented to predict affinity. In this work, a new neural network model that predicts the binding affinity of a protein-ligand complex structure is developed. Our model predicts the binding affinity of a complex using the ensemble of multiple independently trained networks that consist of multiple channels of 3-D convolutional neural network layers. Our model was trained using the 3772 protein-ligand complexes from the refined set of the PDBbind-2016 database and tested using the core set of 285 complexes. The benchmark results show that the Pearson correlation coefficient between the predicted binding affinities by our model and the experimental data is 0.827, which is higher than the state-of-the-art binding affinity prediction scoring functions. Additionally, our method ranks the relative binding affinities of possible multiple binders of a protein quite accurately, comparable to the other scoring functions. Last, we measured which structural information is critical for predicting binding affinity and found that the complementarity between the protein and ligand is most important.


Asunto(s)
Redes Neurales de la Computación , Unión Proteica , Proteínas/química , Proteínas/metabolismo , Diseño Asistido por Computadora , Bases de Datos de Proteínas , Aprendizaje Profundo , Diseño de Fármacos , Descubrimiento de Drogas , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Interfaz Usuario-Computador
5.
Proteins ; 87(12): 1200-1221, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31612567

RESUMEN

We present the results for CAPRI Round 46, the third joint CASP-CAPRI protein assembly prediction challenge. The Round comprised a total of 20 targets including 14 homo-oligomers and 6 heterocomplexes. Eight of the homo-oligomer targets and one heterodimer comprised proteins that could be readily modeled using templates from the Protein Data Bank, often available for the full assembly. The remaining 11 targets comprised 5 homodimers, 3 heterodimers, and two higher-order assemblies. These were more difficult to model, as their prediction mainly involved "ab-initio" docking of subunit models derived from distantly related templates. A total of ~30 CAPRI groups, including 9 automatic servers, submitted on average ~2000 models per target. About 17 groups participated in the CAPRI scoring rounds, offered for most targets, submitting ~170 models per target. The prediction performance, measured by the fraction of models of acceptable quality or higher submitted across all predictors groups, was very good to excellent for the nine easy targets. Poorer performance was achieved by predictors for the 11 difficult targets, with medium and high quality models submitted for only 3 of these targets. A similar performance "gap" was displayed by scorer groups, highlighting yet again the unmet challenge of modeling the conformational changes of the protein components that occur upon binding or that must be accounted for in template-based modeling. Our analysis also indicates that residues in binding interfaces were less well predicted in this set of targets than in previous Rounds, providing useful insights for directions of future improvements.


Asunto(s)
Biología Computacional , Conformación Proteica , Proteínas/ultraestructura , Programas Informáticos , Algoritmos , Sitios de Unión/genética , Bases de Datos de Proteínas , Modelos Moleculares , Unión Proteica/genética , Mapeo de Interacción de Proteínas , Proteínas/química , Proteínas/genética , Homología Estructural de Proteína
6.
PLoS Comput Biol ; 14(1): e1005937, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29329283

RESUMEN

Protein-protein interactions are the cornerstone of numerous biological processes. Although an increasing number of protein complex structures have been determined using experimental methods, relatively fewer studies have been performed to determine the assembly order of complexes. In addition to the insights into the molecular mechanisms of biological function provided by the structure of a complex, knowing the assembly order is important for understanding the process of complex formation. Assembly order is also practically useful for constructing subcomplexes as a step toward solving the entire complex experimentally, designing artificial protein complexes, and developing drugs that interrupt a critical step in the complex assembly. There are several experimental methods for determining the assembly order of complexes; however, these techniques are resource-intensive. Here, we present a computational method that predicts the assembly order of protein complexes by building the complex structure. The method, named Path-LzerD, uses a multimeric protein docking algorithm that assembles a protein complex structure from individual subunit structures and predicts assembly order by observing the simulated assembly process of the complex. Benchmarked on a dataset of complexes with experimental evidence of assembly order, Path-LZerD was successful in predicting the assembly pathway for the majority of the cases. Moreover, when compared with a simple approach that infers the assembly path from the buried surface area of subunits in the native complex, Path-LZerD has the strong advantage that it can be used for cases where the complex structure is not known. The path prediction accuracy decreased when starting from unbound monomers, particularly for larger complexes of five or more subunits, for which only a part of the assembly path was correctly identified. As the first method of its kind, Path-LZerD opens a new area of computational protein structure modeling and will be an indispensable approach for studying protein complexes.


Asunto(s)
Biología Computacional/métodos , Mapeo de Interacción de Proteínas/métodos , Proteínas/química , Algoritmos , Toxina del Cólera/química , Bases de Datos de Proteínas , Helicobacter pylori/metabolismo , Humanos , Modelos Estadísticos , Simulación del Acoplamiento Molecular , Unión Proteica , Dominios Proteicos , Programas Informáticos , Termodinámica
7.
J Comput Aided Mol Des ; 33(12): 1083-1094, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31506789

RESUMEN

Computational prediction of protein-ligand interactions is a useful approach that aids the drug discovery process. Two major tasks of computational approaches are to predict the docking pose of a compound in a known binding pocket and to rank compounds in a library according to their predicted binding affinities. There are many computational tools developed in the past decades both in academia and industry. To objectively assess the performance of existing tools, the community has held a blind assessment of computational predictions, the Drug Design Data Resource Grand Challenge. This round, Grand Challenge 4 (GC4), focused on two targets, protein beta-secretase 1 (BACE-1) and cathepsin S (CatS). We participated in GC4 in both BACE-1 and CatS challenges using our molecular surface-based virtual screening method, PL-PatchSurfer2.0. A unique feature of PL-PatchSurfer2.0 is that it uses the three-dimensional Zernike descriptor, a mathematical moment-based shape descriptor, to quantify local shape complementarity between a ligand and a receptor, which properly incorporates molecular flexibility and provides stable affinity assessment for a bound ligand-receptor complex. Since PL-PatchSurfer2.0 does not explicitly build a bound pose of a ligand, we used an external docking program, such as AutoDock Vina, to provide an ensemble of poses, which were then evaluated by PL-PatchSurfer2.0. Here, we provide an overview of our method and report the performance in GC4.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/química , Ácido Aspártico Endopeptidasas/química , Péptidos y Proteínas de Señalización Intracelular/química , Simulación del Acoplamiento Molecular , Proteínas Nucleares/química , Unión Proteica/genética , Secretasas de la Proteína Precursora del Amiloide/genética , Ácido Aspártico Endopeptidasas/genética , Sitios de Unión/genética , Diseño Asistido por Computadora , Cristalografía por Rayos X , Diseño de Fármacos , Descubrimiento de Drogas , Péptidos y Proteínas de Señalización Intracelular/genética , Ligandos , Proteínas Nucleares/genética , Conformación Proteica , Proteínas/química , Proteínas/genética , Relación Estructura-Actividad , Termodinámica
8.
Pattern Recognit ; 93: 534-545, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32042209

RESUMEN

Direct comparison of three-dimensional (3D) objects is computationally expensive due to the need for translation, rotation, and scaling of the objects to evaluate their similarity. In applications of 3D object comparison, often identifying specific local regions of objects is of particular interest. We have recently developed a set of 2D moment invariants based on discrete orthogonal Krawtchouk polynomials for comparison of local image patches. In this work, we extend them to 3D and construct 3D Krawtchouk descriptors (3DKDs) that are invariant under translation, rotation, and scaling. The new descriptors have the ability to extract local features of a 3D surface from any region-of-interest. This property enables comparison of two arbitrary local surface regions from different 3D objects. We present the new formulation of 3DKDs and apply it to the local shape comparison of protein surfaces in order to predict ligand molecules that bind to query proteins.

9.
Proteins ; 86 Suppl 1: 311-320, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28845596

RESUMEN

We report our group's performance for protein-protein complex structure prediction and scoring in Round 37 of the Critical Assessment of PRediction of Interactions (CAPRI), an objective assessment of protein-protein complex modeling. We demonstrated noticeable improvement in both prediction and scoring compared to previous rounds of CAPRI, with our human predictor group near the top of the rankings and our server scorer group at the top. This is the first time in CAPRI that a server has been the top scorer group. To predict protein-protein complex structures, we used both multi-chain template-based modeling (TBM) and our protein-protein docking program, LZerD. LZerD represents protein surfaces using 3D Zernike descriptors (3DZD), which are based on a mathematical series expansion of a 3D function. Because 3DZD are a soft representation of the protein surface, LZerD is tolerant to small conformational changes, making it well suited to docking unbound and TBM structures. The key to our improved performance in CAPRI Round 37 was to combine multi-chain TBM and docking. As opposed to our previous strategy of performing docking for all target complexes, we used TBM when multi-chain templates were available and docking otherwise. We also describe the combination of multiple scoring functions used by our server scorer group, which achieved the top rank for the scorer phase.


Asunto(s)
Biología Computacional/métodos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Conformación Proteica , Mapeo de Interacción de Proteínas/métodos , Proteínas/química , Sitios de Unión , Bases de Datos de Proteínas , Humanos , Unión Proteica , Proteínas/metabolismo , Análisis de Secuencia de Proteína , Homología Estructural de Proteína
10.
Methods ; 131: 22-32, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28802714

RESUMEN

A core concept behind modern drug discovery is finding a small molecule that modulates a function of a target protein. This concept has been successfully applied since the mid-1970s. However, the efficiency of drug discovery is decreasing because the druggable target space in the human proteome is limited. Recently, protein-protein interaction (PPI) has been identified asan emerging target space for drug discovery. PPI plays a pivotal role in biological pathways including diseases. Current human interactome research suggests that the number of PPIs is between 130,000 and 650,000, and only a small number of them have been targeted as drug targets. For traditional drug targets, in silico structure-based methods have been successful in many cases. However, their performance suffers on PPI interfaces because PPI interfaces are different in five major aspects: From a geometric standpoint, they have relatively large interface regions, flat geometry, and the interface surface shape tends to fluctuate upon binding. Also, their interactions are dominated by hydrophobic atoms, which is different from traditional binding-pocket-targeted drugs. Finally, PPI targets usually lack natural molecules that bind to the target PPI interface. Here, we first summarize characteristics of PPI interfaces and their known binders. Then, we will review existing in silico structure-based approaches for discovering small molecules that bind to PPI interfaces.


Asunto(s)
Diseño de Fármacos , Descubrimiento de Drogas/métodos , Simulación del Acoplamiento Molecular , Proteínas/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Biología Computacional , Descubrimiento de Drogas/tendencias , Humanos , Terapia Molecular Dirigida/métodos , Unión Proteica/efectos de los fármacos , Mapeo de Interacción de Proteínas/métodos , Mapas de Interacción de Proteínas/efectos de los fármacos , Proteínas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad
11.
J Proteome Res ; 16(2): 470-480, 2017 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-28152599

RESUMEN

Protein-ligand interaction plays a critical role in regulating the biochemical functions of proteins. Discovering protein targets for ligands is vital to new drug development. Here, we present a strategy that combines experimental and computational approaches to identify ligand-binding proteins in a proteomic scale. For the experimental part, we coupled pulse proteolysis with filter-assisted sample preparation (FASP) and quantitative mass spectrometry. Under denaturing conditions, ligand binding affected protein stability, which resulted in altered protein abundance after pulse proteolysis. For the computational part, we used the software Patch-Surfer2.0. We applied the integrated approach to identify nicotinamide adenine dinucleotide (NAD)-binding proteins in the Escherichia coli proteome, which has over 4200 proteins. Pulse proteolysis and Patch-Surfer2.0 identified 78 and 36 potential NAD-binding proteins, respectively, including 12 proteins that were consistently detected by the two approaches. Interestingly, the 12 proteins included 8 that are not previously known as NAD binders. Further validation of these eight proteins showed that their binding affinities to NAD computed by AutoDock Vina are higher than their cognate ligands and also that their protein ratios in the pulse proteolysis are consistent with known NAD-binding proteins. These results strongly suggest that these eight proteins are indeed newly identified NAD binders.


Asunto(s)
Biología Computacional/métodos , Proteínas de Escherichia coli/química , Escherichia coli/química , NAD/química , Proteoma/química , Mezclas Complejas/química , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Expresión Génica , Ligandos , Simulación del Acoplamiento Molecular , NAD/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Proteolisis , Proteoma/genética , Proteoma/metabolismo , Programas Informáticos , Termolisina/química
12.
Proteins ; 85(3): 513-527, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27654025

RESUMEN

We report the performance of protein-protein docking predictions by our group for recent rounds of the Critical Assessment of Prediction of Interactions (CAPRI), a community-wide assessment of state-of-the-art docking methods. Our prediction procedure uses a protein-protein docking program named LZerD developed in our group. LZerD represents a protein surface with 3D Zernike descriptors (3DZD), which are based on a mathematical series expansion of a 3D function. The appropriate soft representation of protein surface with 3DZD makes the method more tolerant to conformational change of proteins upon docking, which adds an advantage for unbound docking. Docking was guided by interface residue prediction performed with BindML and cons-PPISP as well as literature information when available. The generated docking models were ranked by a combination of scoring functions, including PRESCO, which evaluates the native-likeness of residues' spatial environments in structure models. First, we discuss the overall performance of our group in the CAPRI prediction rounds and investigate the reasons for unsuccessful cases. Then, we examine the performance of several knowledge-based scoring functions and their combinations for ranking docking models. It was found that the quality of a pool of docking models generated by LZerD, that is whether or not the pool includes near-native models, can be predicted by the correlation of multiple scores. Although the current analysis used docking models generated by LZerD, findings on scoring functions are expected to be universally applicable to other docking methods. Proteins 2017; 85:513-527. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Algoritmos , Biología Computacional/métodos , Simulación del Acoplamiento Molecular/métodos , Proteínas/química , Programas Informáticos , Agua/química , Secuencias de Aminoácidos , Benchmarking , Sitios de Unión , Humanos , Unión Proteica , Conformación Proteica , Mapeo de Interacción de Proteínas , Multimerización de Proteína , Proyectos de Investigación , Homología Estructural de Proteína , Termodinámica
13.
J Comput Aided Mol Des ; 31(7): 653-666, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28623486

RESUMEN

Protein-ligand docking is a useful tool for providing atomic-level understanding of protein functions in nature and design principles for artificial ligands or proteins with desired properties. The ability to identify the true binding pose of a ligand to a target protein among numerous possible candidate poses is an essential requirement for successful protein-ligand docking. Many previously developed docking scoring functions were trained to reproduce experimental binding affinities and were also used for scoring binding poses. However, in this study, we developed a new docking scoring function, called GalaxyDock BP2 Score, by directly training the scoring power of binding poses. This function is a hybrid of physics-based, empirical, and knowledge-based score terms that are balanced to strengthen the advantages of each component. The performance of the new scoring function exhibits significant improvement over existing scoring functions in decoy pose discrimination tests. In addition, when the score is used with the GalaxyDock2 protein-ligand docking program, it outperformed other state-of-the-art docking programs in docking tests on the Astex diverse set, the Cross2009 benchmark set, and the Astex non-native set. GalaxyDock BP2 Score and GalaxyDock2 with this score are freely available at http://galaxy.seoklab.org/softwares/galaxydock.html .


Asunto(s)
Simulación del Acoplamiento Molecular , Proteínas/química , Sitios de Unión , Bases de Datos de Proteínas , Ligandos , Unión Proteica , Conformación Proteica , Proyectos de Investigación , Programas Informáticos
14.
Methods ; 93: 41-50, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26427548

RESUMEN

Protein function prediction is an active area of research in computational biology. Function prediction can help biologists make hypotheses for characterization of genes and help interpret biological assays, and thus is a productive area for collaboration between experimental and computational biologists. Among various function prediction methods, predicting binding ligand molecules for a target protein is an important class because ligand binding events for a protein are usually closely intertwined with the proteins' biological function, and also because predicted binding ligands can often be directly tested by biochemical assays. Binding ligand prediction methods can be classified into two types: those which are based on protein-protein (or pocket-pocket) comparison, and those that compare a target pocket directly to ligands. Recently, our group proposed two computational binding ligand prediction methods, Patch-Surfer, which is a pocket-pocket comparison method, and PL-PatchSurfer, which compares a pocket to ligand molecules. The two programs apply surface patch-based descriptions to calculate similarity or complementarity between molecules. A surface patch is characterized by physicochemical properties such as shape, hydrophobicity, and electrostatic potentials. These properties on the surface are represented using three-dimensional Zernike descriptors (3DZD), which are based on a series expansion of a 3 dimensional function. Utilizing 3DZD for describing the physicochemical properties has two main advantages: (1) rotational invariance and (2) fast comparison. Here, we introduce Patch-Surfer and PL-PatchSurfer with an emphasis on PL-PatchSurfer, which is more recently developed. Illustrative examples of PL-PatchSurfer performance on binding ligand prediction as well as virtual drug screening are also provided.


Asunto(s)
Biología Computacional/métodos , Bases de Datos de Proteínas , Proteínas/química , Proteínas/metabolismo , Sitios de Unión/fisiología , Predicción , Ligandos , Unión Proteica/fisiología , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
15.
J Chem Inf Model ; 56(6): 988-95, 2016 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-26583962

RESUMEN

We analyze the results of the GalaxyDock protein-ligand docking program in the two recent experiments of Community Structure-Activity Resource (CSAR), CSAR 2013 and 2014. GalaxyDock performs global optimization of a modified AutoDock3 energy function by employing the conformational space annealing method. The energy function of GalaxyDock is quite sensitive to atomic clashes. Such energy functions can be effective for sampling physically correct conformations but may not be effective for scoring when conformations are not fully optimized. In phase 1 of CSAR 2013, we successfully selected all four true binders of digoxigenin along with three false positives. However, the energy values were rather high due to insufficient optimization of the conformations docked to homology models. A posteriori relaxation of the model complex structures by GalaxyRefine improved the docking energy values and differentiated the true binders from the false positives better. In the scoring test of CSAR 2013 phase 2, we selected the best poses for each of the two targets. The results of CSAR 2013 phase 3 suggested that an improved method for generating initial conformations for GalaxyDock is necessary for targets involving bulky ligands. Finally, combining existing binding information with GalaxyDock energy-based optimization may be needed for more accurate binding affinity prediction.


Asunto(s)
Simulación del Acoplamiento Molecular , Benchmarking , Ligandos , Conformación Proteica , Proteínas/química , Proteínas/metabolismo , Relación Estructura-Actividad
16.
J Chem Inf Model ; 56(6): 1088-99, 2016 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-26691286

RESUMEN

The Community Structure-Activity Resource (CSAR) benchmark exercise provides a unique opportunity for researchers to objectively evaluate the performance of protein-ligand docking methods. Patch-Surfer and PL-PatchSurfer, molecular surface-based methods for predicting binding ligands of proteins developed in our group, were tested on both CSAR 2013 and 2014 benchmark exercises in combination with an empirical scoring function-based method, AutoDock, while we only participated in CSAR 2013 using Patch-Surfer. The prediction results for Phase 1 task in CSAR 2013 showed that Patch-Surfer was able to rank all the four designed binding proteins within top ranks, outperforming AutoDock Vina. In Phase 2 of 2013, PL-PatchSurfer correctly selected the correct ligand pose for two target proteins. PL-PatchSurfer performed reasonably well in ranking ligands according to their binding affinity and in selecting near-native ligand poses in 2013 Phase 3 and 2014 Phase 1, respectively, although AutoDock Vina showed better performance. Lastly, in the 2014 Phase 2 exercise, the PL-PatchSurfer scores computed for ligands to target protein pairs correlated well with their pIC50 values, which was better or comparable to results by other participants. Overall, our methods showed fairly good performance in CSAR 2013 and 2014. Unique characteristics of the methods are discussed in comparison with AutoDock.


Asunto(s)
Biología Computacional , Diseño de Fármacos , Simulación del Acoplamiento Molecular , Proteínas/metabolismo , Benchmarking , Bases de Datos Farmacéuticas , Ligandos , Unión Proteica , Proteínas/química , Relación Estructura-Actividad
17.
J Chem Inf Model ; 56(9): 1676-91, 2016 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-27500657

RESUMEN

Virtual screening has become an indispensable procedure in drug discovery. Virtual screening methods can be classified into two categories: ligand-based and structure-based. While the former have advantages, including being quick to compute, in general they are relatively weak at discovering novel active compounds because they use known actives as references. On the other hand, structure-based methods have higher potential to find novel compounds because they directly predict the binding affinity of a ligand in a target binding pocket, albeit with substantially lower speed than ligand-based methods. Here we report a novel structure-based virtual screening method, PL-PatchSurfer2. In PL-PatchSurfer2, protein and ligand surfaces are represented by a set of overlapping local patches, each of which is represented by three-dimensional Zernike descriptors (3DZDs). By means of 3DZDs, the shapes and physicochemical complementarities of local surface regions of a pocket surface and a ligand molecule can be concisely and effectively computed. Compared with the previous version of the program, the performance of PL-PatchSurfer2 is substantially improved by the addition of two more features, atom-based hydrophobicity and hydrogen-bond acceptors and donors. Benchmark studies showed that PL-PatchSurfer2 performed better than or comparable to popular existing methods. Particularly, PL-PatchSurfer2 significantly outperformed existing methods when apo-form or template-based protein models were used for queries. The computational time of PL-PatchSurfer2 is about 20 times shorter than those of conventional structure-based methods. The PL-PatchSurfer2 program is available at http://www.kiharalab.org/plps2/ .


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Sitios de Unión , Ligandos , Simulación del Acoplamiento Molecular , Conformación Proteica , Interfaz Usuario-Computador
18.
Nucleic Acids Res ; 42(Web Server issue): W210-4, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24753427

RESUMEN

Knowledge of ligand-binding sites of proteins provides invaluable information for functional studies, drug design and protein design. Recent progress in ligand-binding-site prediction methods has demonstrated that using information from similar proteins of known structures can improve predictions. The GalaxySite web server, freely accessible at http://galaxy.seoklab.org/site, combines such information with molecular docking for more precise binding-site prediction for non-metal ligands. According to the recent critical assessments of structure prediction methods held in 2010 and 2012, this server was found to be superior or comparable to other state-of-the-art programs in the category of ligand-binding-site prediction. A strong merit of the GalaxySite program is that it provides additional predictions on binding ligands and their binding poses in terms of the optimized 3D coordinates of the protein-ligand complexes, whereas other methods predict only identities of binding-site residues or copy binding geometry from similar proteins. The additional information on the specific binding geometry would be very useful for applications in functional studies and computer-aided drug discovery.


Asunto(s)
Simulación del Acoplamiento Molecular/métodos , Proteínas/química , Proteínas/metabolismo , Programas Informáticos , Sitios de Unión , Internet , Ligandos , Conformación Proteica
19.
Molecules ; 20(7): 12841-62, 2015 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-26193243

RESUMEN

Virtual screening has been widely used in the drug discovery process. Ligand-based virtual screening (LBVS) methods compare a library of compounds with a known active ligand. Two notable advantages of LBVS methods are that they do not require structural information of a target receptor and that they are faster than structure-based methods. LBVS methods can be classified based on the complexity of ligand structure information utilized: one-dimensional (1D), two-dimensional (2D), and three-dimensional (3D). Unlike 1D and 2D methods, 3D methods can have enhanced performance since they treat the conformational flexibility of compounds. In this paper, a number of 3D methods will be reviewed. In addition, four representative 3D methods were benchmarked to understand their performance in virtual screening. Specifically, we tested overall performance in key aspects including the ability to find dissimilar active compounds, and computational speed.


Asunto(s)
Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Humanos , Procesamiento de Imagen Asistido por Computador , Imagenología Tridimensional/métodos , Ligandos , Modelos Químicos , Modelos Moleculares
20.
bioRxiv ; 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38464318

RESUMEN

Structure-based virtual screening (SBVS) is a widely used method in silico drug discovery. It necessitates a receptor structure or binding site to predict the binding pose and fitness of a ligand. Therefore, the performance of the SBVS is affected by the protein conformation. The most frequently used method in SBVS is the protein-ligand docking program, which utilizes atomic distance-based scoring functions. Hence, they are highly prone to sensitivity towards variation in receptor structure, and it is reported that the conformational change significantly drops the performance of the docking program. To address the problem, we have introduced a novel program of SBVS, named PL-PatchSurfer. This program makes use of molecular surface patches and the Zernike descriptor. The surfaces of the pocket and ligand are segmented into several patches by the program. These patches are then mapped with physico-chemical properties such as shape and electrostatic potential before being converted into the Zernike descriptor, which is rotationally invariant. A complementarity between the protein and the ligand is assessed by comparing the descriptors and geometric distribution of the patches in the molecules. A benchmarking study showed that PL-PatchSurfer2 was able to screen active molecules regardless of the receptor structure change with fast speed. However, the program could not achieve high performance for the targets that the hydrogen bonding feature is important such as nuclear hormone receptors. In this paper, we present the newer version of PL-PatchSurfer, PL-PatchSurfer3, which incorporates two new features: a change in the definition of hydrogen bond complementarity and consideration of visibility that contains curvature information of a patch. Our evaluation demonstrates that the new program outperforms its predecessor and other SBVS methods while retaining its characteristic tolerance to receptor structure changes. Interested individuals can access the program at kiharalab.org/plps3.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA