Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Bioinformatics ; 33(20): 3299-3301, 2017 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-28430871

RESUMO

SUMMARY: We present an approach for the efficient docking of peptide motifs to their free receptor structures. Using a motif based search, we can retrieve structural fragments from the Protein Data Bank (PDB) that are very similar to the peptide's final, bound conformation. We use a Fast Fourier Transform (FFT) based docking method to quickly perform global rigid body docking of these fragments to the receptor. According to CAPRI peptide docking criteria, an acceptable conformation can often be found among the top-ranking predictions. AVAILABILITY AND IMPLEMENTATION: The method is available as part of the protein-protein docking server ClusPro at https://peptidock.cluspro.org/nousername.php. CONTACT: midas@laufercenter.org or oraf@ekmd.huji.ac.il. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Biologia Computacional/métodos , Simulação de Acoplamento Molecular/métodos , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Software , Algoritmos , Ciclinas/química , Ciclinas/metabolismo , Bases de Dados de Proteínas , Análise de Fourier , Peptídeos/química , Peptídeos/metabolismo
2.
Proteins ; 85(3): 435-444, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27936493

RESUMO

The heavily used protein-protein docking server ClusPro performs three computational steps as follows: (1) rigid body docking, (2) RMSD based clustering of the 1000 lowest energy structures, and (3) the removal of steric clashes by energy minimization. In response to challenges encountered in recent CAPRI targets, we added three new options to ClusPro. These are (1) accounting for small angle X-ray scattering data in docking; (2) considering pairwise interaction data as restraints; and (3) enabling discrimination between biological and crystallographic dimers. In addition, we have developed an extremely fast docking algorithm based on 5D rotational manifold FFT, and an algorithm for docking flexible peptides that include known sequence motifs. We feel that these developments will further improve the utility of ClusPro. However, CAPRI emphasized several shortcomings of the current server, including the problem of selecting the right energy parameters among the five options provided, and the problem of selecting the best models among the 10 generated for each parameter set. In addition, results convinced us that further development is needed for docking homology models. Finally, we discuss the difficulties we have encountered when attempting to develop a refinement algorithm that would be computationally efficient enough for inclusion in a heavily used server. Proteins 2017; 85:435-444. © 2016 Wiley Periodicals, Inc.


Assuntos
Algoritmos , Biologia Computacional/métodos , Simulação de Acoplamento Molecular/métodos , Proteínas/química , Software , Água/química , Benchmarking , Sítios de Ligação , Análise por Conglomerados , Cristalografia por Raios X , Bases de Dados de Proteínas , Internet , Ligação Proteica , Conformação Proteica , Mapeamento de Interação de Proteínas , Multimerização Proteica , Projetos de Pesquisa , Homologia Estrutural de Proteína , Termodinâmica
3.
Proteins ; 85(1): 10-16, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27172383

RESUMO

Protein docking procedures carry out the task of predicting the structure of a protein-protein complex starting from the known structures of the individual protein components. More often than not, however, the structure of one or both components is not known, but can be derived by homology modeling on the basis of known structures of related proteins deposited in the Protein Data Bank (PDB). Thus, the problem is to develop methods that optimally integrate homology modeling and docking with the goal of predicting the structure of a complex directly from the amino acid sequences of its component proteins. One possibility is to use the best available homology modeling and docking methods. However, the models built for the individual subunits often differ to a significant degree from the bound conformation in the complex, often much more so than the differences observed between free and bound structures of the same protein, and therefore additional conformational adjustments, both at the backbone and side chain levels need to be modeled to achieve an accurate docking prediction. In particular, even homology models of overall good accuracy frequently include localized errors that unfavorably impact docking results. The predicted reliability of the different regions in the model can also serve as a useful input for the docking calculations. Here we present a benchmark dataset that should help to explore and solve combined modeling and docking problems. This dataset comprises a subset of the experimentally solved 'target' complexes from the widely used Docking Benchmark from the Weng Lab (excluding antibody-antigen complexes). This subset is extended to include the structures from the PDB related to those of the individual components of each complex, and hence represent potential templates for investigating and benchmarking integrated homology modeling and docking approaches. Template sets can be dynamically customized by specifying ranges in sequence similarity and in PDB release dates, or using other filtering options, such as excluding sets of specific structures from the template list. Multiple sequence alignments, as well as structural alignments of the templates to their corresponding subunits in the target are also provided. The resource is accessible online or can be downloaded at http://cluspro.org/benchmark, and is updated on a weekly basis in synchrony with new PDB releases. Proteins 2016; 85:10-16. © 2016 Wiley Periodicals, Inc.


Assuntos
Benchmarking , Caspase 9/química , Simulação de Acoplamento Molecular , Software , Homologia Estrutural de Proteína , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/química , Sequência de Aminoácidos , Sítios de Ligação , Caspase 9/metabolismo , Bases de Dados de Proteínas , Humanos , Internet , Ligantes , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Alinhamento de Sequência , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo
4.
J Comput Chem ; 37(11): 961-70, 2016 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-26837000

RESUMO

The fast Fourier transform (FFT) sampling algorithm has been used with success in application to protein-protein docking and for protein mapping, the latter docking a variety of small organic molecules for the identification of binding hot spots on the target protein. Here we explore the local rather than global usage of the FFT sampling approach in docking applications. If the global FFT based search yields a near-native cluster of docked structures for a protein complex, then focused resampling of the cluster generally leads to a substantial increase in the number of conformations close to the native structure. In protein mapping, focused resampling of the selected hot spot regions generally reveals further hot spots that, while not as strong as the primary hot spots, also contribute to ligand binding. The detection of additional ligand binding regions is shown by the improved overlap between hot spots and bound ligands.


Assuntos
Análise de Fourier , Simulação de Acoplamento Molecular , Proteínas/química , Algoritmos , Ligantes , Conformação Proteica
5.
PLoS Comput Biol ; 10(10): e1003872, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25275445

RESUMO

Many protein-protein interactions (PPIs) are compelling targets for drug discovery, and in a number of cases can be disrupted by small molecules. The main goal of this study is to examine the mechanism of binding site formation in the interface region of proteins that are PPI targets by comparing ligand-free and ligand-bound structures. To avoid any potential bias, we focus on ensembles of ligand-free protein conformations obtained by nuclear magnetic resonance (NMR) techniques and deposited in the Protein Data Bank, rather than on ensembles specifically generated for this study. The measures used for structure comparison are based on detecting binding hot spots, i.e., protein regions that are major contributors to the binding free energy. The main tool of the analysis is computational solvent mapping, which explores the surface of proteins by docking a large number of small "probe" molecules. Although we consider conformational ensembles obtained by NMR techniques, the analysis is independent of the method used for generating the structures. Finding the energetically most important regions, mapping can identify binding site residues using ligand-free models based on NMR data. In addition, the method selects conformations that are similar to some peptide-bound or ligand-bound structure in terms of the properties of the binding site. This agrees with the conformational selection model of molecular recognition, which assumes such pre-existing conformations. The analysis also shows the maximum level of similarity between unbound and bound states that is achieved without any influence from a ligand. Further shift toward the bound structure assumes protein-peptide or protein-ligand interactions, either selecting higher energy conformations that are not part of the NMR ensemble, or leading to induced fit. Thus, forming the sites in protein-protein interfaces that bind peptides and can be targeted by small ligands always includes conformational selection, although other recognition mechanisms may also be involved.


Assuntos
Sítios de Ligação , Ligação Proteica , Conformação Proteica , Proteínas/química , Proteínas/metabolismo , Biologia Computacional , Bases de Dados de Proteínas , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular
6.
Nucleic Acids Res ; 40(Web Server issue): W271-5, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22589414

RESUMO

Binding hot spots, protein sites with high-binding affinity, can be identified using X-ray crystallography or NMR by screening libraries of small organic molecules that tend to cluster at such regions. FTMAP, a direct computational analog of the experimental screening approaches, globally samples the surface of a target protein using small organic molecules as probes, finds favorable positions, clusters the conformations and ranks the clusters on the basis of the average energy. The regions that bind several probe clusters predict the binding hot spots, in good agreement with experimental results. Small molecules discovered by fragment-based approaches to drug design also bind at the hot spot regions. To identify such molecules and their most likely bound positions, we extend the functionality of FTMAP (http://ftmap.bu.edu/param) to accept any small molecule as an additional probe. In its updated form, FTMAP identifies the hot spots based on a standard set of probes, and for each additional probe shows representative structures of nearby low energy clusters. This approach helps to predict bound poses of the user-selected molecules, detects if a compound is not likely to bind in the hot spot region, and provides input for the design of larger ligands.


Assuntos
Proteínas/química , Software , Algoritmos , Sítios de Ligação , Internet , Ligantes , Sondas Moleculares/química , Ligação Proteica , Trombina/química
7.
Nucleic Acids Res ; 40(16): 7644-52, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22705795

RESUMO

Formaldehyde has long been recognized as a hazardous environmental agent highly reactive with DNA. Recently, it has been realized that due to the activity of histone demethylation enzymes within the cell nucleus, formaldehyde is produced endogenously, in direct vicinity of genomic DNA. Should it lead to extensive DNA damage? We address this question with the aid of a computational mapping method, analogous to X-ray and nuclear magnetic resonance techniques for observing weakly specific interactions of small organic compounds with a macromolecule in order to establish important functional sites. We concentrate on the leading reaction of formaldehyde with free bases: hydroxymethylation of cytosine amino groups. Our results show that in B-DNA, cytosine amino groups are totally inaccessible for the formaldehyde attack. Then, we explore the effect of recently discovered transient flipping of Watson-Crick (WC) pairs into Hoogsteen (HG) pairs (HG breathing). Our results show that the HG base pair formation dramatically affects the accessibility for formaldehyde of cytosine amino nitrogens within WC base pairs adjacent to HG base pairs. The extensive literature on DNA interaction with formaldehyde is analyzed in light of the new findings. The obtained data emphasize the significance of DNA HG breathing.


Assuntos
DNA de Forma B/química , Formaldeído/química , Algoritmos , Pareamento de Bases , Sítios de Ligação , Biologia Computacional , Citosina/química , Modelos Moleculares , Nitrogênio/química
8.
Proteins ; 81(12): 2159-66, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23996272

RESUMO

The protein docking server ClusPro has been participating in critical assessment of prediction of interactions (CAPRI) since its introduction in 2004. This article evaluates the performance of ClusPro 2.0 for targets 46-58 in Rounds 22-27 of CAPRI. The analysis leads to a number of important observations. First, ClusPro reliably yields acceptable or medium accuracy models for targets of moderate difficulty that have also been successfully predicted by other groups, and fails only for targets that have few acceptable models submitted. Second, the quality of automated docking by ClusPro is very close to that of the best human predictor groups, including our own submissions. This is very important, because servers have to submit results within 48 h and the predictions should be reproducible, whereas human predictors have several weeks and can use any type of information. Third, while we refined the ClusPro results for manual submission by running computationally costly Monte Carlo minimization simulations, we observed significant improvement in accuracy only for two of the six complexes correctly predicted by ClusPro. Fourth, new developments, not seen in previous rounds of CAPRI, are that the top ranked model provided by ClusPro was acceptable or better quality for all these six targets, and that the top ranked model was also the highest quality for five of the six, confirming that ranking models based on cluster size can reliably identify the best near-native conformations.


Assuntos
Simulação de Acoplamento Molecular , Mapeamento de Interação de Proteínas , Proteínas/química , Software , Biologia Computacional , Simulação por Computador , Bases de Dados de Proteínas , Humanos , Internet , Modelos Moleculares , Método de Monte Carlo , Conformação Proteica , Proteômica
9.
Proteins ; 81(12): 2096-105, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24123488

RESUMO

Peptide-mediated interactions, in which a short linear motif binds to a globular domain, play major roles in cellular regulation. An accurate structural model of this type of interaction is an excellent starting point for the characterization of the binding specificity of a given peptide-binding domain. A number of different protocols have recently been proposed for the accurate modeling of peptide-protein complex structures, given the structure of the protein receptor and the binding site on its surface. When no information about the peptide binding site(s) is a priori available, there is a need for new approaches to locate peptide-binding sites on the protein surface. While several approaches have been proposed for the general identification of ligand binding sites, peptides show very specific binding characteristics, and therefore, there is a need for robust and accurate approaches that are optimized for the prediction of peptide-binding sites. Here, we present PeptiMap, a protocol for the accurate mapping of peptide binding sites on protein structures. Our method is based on experimental evidence that peptide-binding sites also bind small organic molecules of various shapes and polarity. Using an adaptation of ab initio ligand binding site prediction based on fragment mapping (FTmap), we optimize a protocol that specifically takes into account peptide binding site characteristics. In a high-quality curated set of peptide-protein complex structures PeptiMap identifies for most the accurate site of peptide binding among the top ranked predictions. We anticipate that this protocol will significantly increase the number of accurate structural models of peptide-mediated interactions.


Assuntos
Biologia Computacional , Proteínas de Membrana/química , Peptídeos/química , Mapas de Interação de Proteínas , Sítios de Ligação , Bases de Dados de Proteínas , Ligantes , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Software
10.
Bioinformatics ; 28(20): 2608-14, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23053206

RESUMO

MOTIVATION: An effective docking algorithm for antibody-protein antigen complex prediction is an important first step toward design of biologics and vaccines. We have recently developed a new class of knowledge-based interaction potentials called Decoys as the Reference State (DARS) and incorporated DARS into the docking program PIPER based on the fast Fourier transform correlation approach. Although PIPER was the best performer in the latest rounds of the CAPRI protein docking experiment, it is much less accurate for docking antibody-protein antigen pairs than other types of complexes, in spite of incorporating sequence-based information on the location of the paratope. Analysis of antibody-protein antigen complexes has revealed an inherent asymmetry within these interfaces. Specifically, phenylalanine, tryptophan and tyrosine residues highly populate the paratope of the antibody but not the epitope of the antigen. RESULTS: Since this asymmetry cannot be adequately modeled using a symmetric pairwise potential, we have removed the usual assumption of symmetry. Interaction statistics were extracted from antibody-protein complexes under the assumption that a particular atom on the antibody is different from the same atom on the antigen protein. The use of the new potential significantly improves the performance of docking for antibody-protein antigen complexes, even without any sequence information on the location of the paratope. We note that the asymmetric potential captures the effects of the multi-body interactions inherent to the complex environment in the antibody-protein antigen interface. AVAILABILITY: The method is implemented in the ClusPro protein docking server, available at http://cluspro.bu.edu.


Assuntos
Complexo Antígeno-Anticorpo/química , Simulação de Acoplamento Molecular/métodos , Proteínas/imunologia , Algoritmos , Complexo Antígeno-Anticorpo/metabolismo , Interpretação Estatística de Dados , Análise de Fourier , Bases de Conhecimento , Ligação Proteica , Proteínas/química , Proteínas/metabolismo
11.
Nat Biotechnol ; 41(5): 686-697, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36624149

RESUMO

Cytosine base editors (CBEs) enable programmable genomic C·G-to-T·A transition mutations and typically comprise a modified CRISPR-Cas enzyme, a naturally occurring cytidine deaminase, and an inhibitor of uracil repair. Previous studies have shown that CBEs utilizing naturally occurring cytidine deaminases may cause unguided, genome-wide cytosine deamination. While improved CBEs that decrease stochastic genome-wide off-targets have subsequently been reported, these editors can suffer from suboptimal on-target performance. Here, we report the generation and characterization of CBEs that use engineered variants of TadA (CBE-T) that enable high on-target C·G to T·A across a sequence-diverse set of genomic loci, demonstrate robust activity in primary cells and cause no detectable elevation in genome-wide mutation. Additionally, we report cytosine and adenine base editors (CABEs) catalyzing both A-to-I and C-to-U editing (CABE-Ts). Together with ABEs, CBE-Ts and CABE-Ts enable the programmable installation of all transition mutations using laboratory-evolved TadA variants with improved properties relative to previously reported CBEs.


Assuntos
Citosina , Edição de Genes , Mutação/genética , Citidina Desaminase/genética , Genoma , Sistemas CRISPR-Cas/genética
12.
Sci Rep ; 11(1): 15856, 2021 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-34349202

RESUMO

Novel modalities such as PROTAC and RNAi have the ability to inadvertently alter the abundance of endogenous proteins. Currently available in vitro secondary pharmacology assays, which evaluate off-target binding or activity of small molecules, do not fully assess the off-target effects of PROTAC and are not applicable to RNAi. To address this gap, we developed a proteomics-based platform to comprehensively evaluate the abundance of off-target proteins. First, we selected off-target proteins using genetics and pharmacology evidence. This process yielded 2813 proteins, which we refer to as the "selected off-target proteome" (SOTP). An iterative algorithm was then used to identify four human cell lines out of 932. The 4 cell lines collectively expressed ~ 80% of the SOTP based on transcriptome data. Second, we used mass spectrometry to quantify the intracellular and extracellular proteins from the selected cell lines. Among over 10,000 quantifiable proteins identified, 1828 were part of the predefined SOTP. The SOTP was designed to be easily modified or expanded, owing to the rational selection process developed and the label free LC-MS/MS approach chosen. This versatility inherent to our platform is essential to design fit-for-purpose studies that can address the dynamic questions faced in investigative toxicology.


Assuntos
Biomarcadores Tumorais/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias/metabolismo , Neoplasias/patologia , Proteoma/análise , Proteoma/metabolismo , Proliferação de Células , Humanos , Células Tumorais Cultivadas
13.
Protein Eng Des Sel ; 32(7): 347-354, 2019 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-31504835

RESUMO

Antibodies often undergo substantial engineering en route to the generation of a therapeutic candidate with good developability properties. Characterization of antibody libraries has shown that retaining native-like sequence improves the overall quality of the library. Motivated by recent advances in deep learning, we developed a bi-directional long short-term memory (LSTM) network model to make use of the large amount of available antibody sequence information, and use this model to quantify the nativeness of antibody sequences. The model scores sequences for their similarity to naturally occurring antibodies, which can be used as a consideration during design and engineering of libraries. We demonstrate the performance of this approach by training a model on human antibody sequences and show that our method outperforms other approaches at distinguishing human antibodies from those of other species. We show the applicability of this method for the evaluation of synthesized antibody libraries and humanization of mouse antibodies.


Assuntos
Anticorpos/química , Biologia Computacional , Animais , Anticorpos/imunologia , Humanos
14.
Methods Mol Biol ; 1561: 11-20, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28236230

RESUMO

Peptide-mediated interactions are of primordial importance to the cell, and the structure of such interaction provides an important starting point for their further characterization. In many cases, the structure of the peptide-protein complex has not been solved by experiment, and modeling tools need to be applied to generate structural models of the interaction. PeptiMap is a protocol that identifies the peptide-binding site when only the structure of the receptor is known, but no information about where the peptide binds is available. This is achieved by mapping the surface for solvents to identify ligand-binding sites, similar in approach to ANCHORMAP in which amino acids are mapped. Peptimap is a free open access web-based server. It can be accessed at http://peptimap.cluspro.org .


Assuntos
Bases de Dados de Proteínas , Fragmentos de Peptídeos/metabolismo , Proteínas/metabolismo , Software , Sítios de Ligação , Humanos , Fragmentos de Peptídeos/química , Proteínas/química , Navegador
15.
Nat Protoc ; 10(5): 733-55, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25855957

RESUMO

FTMap is a computational mapping server that identifies binding hot spots of macromolecules-i.e., regions of the surface with major contributions to the ligand-binding free energy. To use FTMap, users submit a protein, DNA or RNA structure in PDB (Protein Data Bank) format. FTMap samples billions of positions of small organic molecules used as probes, and it scores the probe poses using a detailed energy expression. Regions that bind clusters of multiple probe types identify the binding hot spots in good agreement with experimental data. FTMap serves as the basis for other servers, namely FTSite, which is used to predict ligand-binding sites, FTFlex, which is used to account for side chain flexibility, FTMap/param, used to parameterize additional probes and FTDyn, for mapping ensembles of protein structures. Applications include determining the druggability of proteins, identifying ligand moieties that are most important for binding, finding the most bound-like conformation in ensembles of unliganded protein structures and providing input for fragment-based drug design. FTMap is more accurate than classical mapping methods such as GRID and MCSS, and it is much faster than the more-recent approaches to protein mapping based on mixed molecular dynamics. By using 16 probe molecules, the FTMap server finds the hot spots of an average-size protein in <1 h. As FTFlex performs mapping for all low-energy conformers of side chains in the binding site, its completion time is proportionately longer.


Assuntos
Biologia Computacional/métodos , Proteínas/química , Proteínas/metabolismo , Sítios de Ligação , Bases de Dados de Proteínas , Internet , Ligantes , Sondas Moleculares , Conformação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA