Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Brief Bioinform ; 25(2)2024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-38305457

RESUMEN

The structural modeling of peptides can be a useful aid in the discovery of new drugs and a deeper understanding of the molecular mechanisms of life. Here we present a novel multiscale protocol for the structure prediction of linear and cyclic peptides. The protocol combines two main stages: coarse-grained simulations using the CABS-flex standalone package and an all-atom reconstruction-optimization process using the Modeller program. We evaluated the protocol on a set of linear peptides and two sets of cyclic peptides, with cyclization through the backbone and disulfide bonds. A comparison with other state-of-the-art tools (APPTEST, PEP-FOLD, ESMFold and AlphaFold implementation in ColabFold) shows that for most cases, AlphaFold offers the highest resolution. However, CABS-flex is competitive, particularly when it comes to short linear peptides. As demonstrated, the protocol performance can be further improved by combination with the residue-residue contact prediction method or more efficient scoring. The protocol is included in the CABS-flex standalone package along with online documentation to aid users in predicting the structure of peptides and mini-proteins.


Asunto(s)
Péptidos Cíclicos , Proteínas , Proteínas/química , Péptidos/química , Conformación Proteica
2.
Nucleic Acids Res ; 50(W1): W474-W482, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35524560

RESUMEN

Correct identification and effective visualization of interactions in biomolecular structures facilitate understanding of their functions and molecular design. In response to the practical needs of structure-based analysis, we have created a Mapiya web server. The Mapiya integrates four main functionalities: (i) generation of contact maps - intramolecular and intermolecular-for proteins, nucleic acids, and their complexes; (ii) characterization of the interactions physicochemical nature, (iii) interactive visualization of biomolecular conformations with automatic zoom on selected contacts using Molstar and (iv) additional sequence- and structure-based analyses performed with third-party software and in-house algorithms combined into an easy-to-use interface. Thus, Mapiya offers a highly customized analysis of the molecular interactions' in various biological systems. The web server is available at: http://mapiya.lcbio.pl/.


Asunto(s)
Proteínas , Programas Informáticos , Proteínas/química , Algoritmos , Computadores , Conformación Proteica , Internet
3.
Int J Mol Sci ; 22(14)2021 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-34298961

RESUMEN

Most of the protein-protein docking methods treat proteins as almost rigid objects. Only the side-chains flexibility is usually taken into account. The few approaches enabling docking with a flexible backbone typically work in two steps, in which the search for protein-protein orientations and structure flexibility are simulated separately. In this work, we propose a new straightforward approach for docking sampling. It consists of a single simulation step during which a protein undergoes large-scale backbone rearrangements, rotations, and translations. Simultaneously, the other protein exhibits small backbone fluctuations. Such extensive sampling was possible using the CABS coarse-grained protein model and Replica Exchange Monte Carlo dynamics at a reasonable computational cost. In our proof-of-concept simulations of 62 protein-protein complexes, we obtained acceptable quality models for a significant number of cases.


Asunto(s)
Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Pliegue de Proteína , Proteínas/química , Método de Montecarlo
4.
Protein Sci ; 29(1): 211-222, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31682301

RESUMEN

Molecular docking of peptides to proteins can be a useful tool in the exploration of the possible peptide binding sites and poses. CABS-dock is a method for protein-peptide docking that features significant conformational flexibility of both the peptide and the protein molecules during the peptide search for a binding site. The CABS-dock has been made available as a web server and a standalone package. The web server is an easy to use tool with a simple web interface. The standalone package is a command-line program dedicated to professional users. It offers a number of advanced features, analysis tools and support for large-sized systems. In this article, we outline the current status of the CABS-dock method, its recent developments, applications, and challenges ahead.


Asunto(s)
Biología Computacional/métodos , Péptidos/química , Proteínas/química , Sitios de Unión , Modelos Moleculares , Simulación del Acoplamiento Molecular , Péptidos/metabolismo , Unión Proteica , Conformación Proteica , Proteínas/metabolismo , Programas Informáticos , Interfaz Usuario-Computador , Navegador Web
5.
Nucleic Acids Res ; 47(W1): W300-W307, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31049593

RESUMEN

Protein aggregation is a hallmark of a growing number of human disorders and constitutes a major bottleneck in the manufacturing of therapeutic proteins. Therefore, there is a strong need of in-silico methods that can anticipate the aggregative properties of protein variants linked to disease and assist the engineering of soluble protein-based drugs. A few years ago, we developed a method for structure-based prediction of aggregation properties that takes into account the dynamic fluctuations of proteins. The method has been made available as the Aggrescan3D (A3D) web server and applied in numerous studies of protein structure-aggregation relationship. Here, we present a major update of the A3D web server to version 2.0. The new features include: extension of dynamic calculations to significantly larger and multimeric proteins, simultaneous prediction of changes in protein solubility and stability upon mutation, rapid screening for functional protein variants with improved solubility, a REST-ful service to incorporate A3D calculations in automatic pipelines, and a new, enhanced web server interface. A3D 2.0 is freely available at: http://biocomp.chem.uw.edu.pl/A3D2/.


Asunto(s)
Algoritmos , Agregado de Proteínas , Proteínas/química , Programas Informáticos , Humanos , Difusión de la Información , Internet , Agregación Patológica de Proteínas/metabolismo , Multimerización de Proteína , Estabilidad Proteica , Solubilidad
6.
Bioinformatics ; 35(20): 4170-4172, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30865258

RESUMEN

SUMMARY: CABS-dock standalone is a multiplatform Python package for protein-peptide docking with backbone flexibility. The main feature of the CABS-dock method is its ability to simulate significant backbone flexibility of the entire protein-peptide system in a reasonable computational time. In the default mode, the package runs a simulation of fully flexible peptide searching for a binding site on the surface of a flexible protein receptor. The flexibility level of the molecules may be defined by the user. Furthermore, the CABS-dock standalone application provides users with full control over the docking simulation from the initial setup to the analysis of results. The standalone version is an upgrade of the original web server implementation-it introduces a number of customizable options, provides support for large-sized systems and offers a framework for deeper analysis of docking results. AVAILABILITY AND IMPLEMENTATION: CABS-dock standalone is distributed under the MIT licence, which is free for academic and non-profit users. It is implemented in Python and Fortran. The CABS-dock standalone source code, wiki with documentation and examples of use and installation instructions for Linux, macOS and Windows are available in the CABS-dock standalone repository at https://bitbucket.org/lcbio/cabsdock.


Asunto(s)
Programas Informáticos , Sitios de Unión , Simulación del Acoplamiento Molecular , Péptidos , Unión Proteica , Proteínas
7.
Bioinformatics ; 35(19): 3834-3835, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30825368

RESUMEN

SUMMARY: Aggrescan3D (A3D) standalone is a multiplatform Python package for structure-based prediction of protein aggregation properties and rational design of protein solubility. A3D allows the re-design of protein solubility by combining structural aggregation propensity and stability predictions, as demonstrated by a recent experimental study. It also enables predicting the impact of protein conformational fluctuations on the aggregation properties. The standalone A3D version is an upgrade of the original web server implementation-it introduces a number of customizable options, automated analysis of multiple mutations and offers a flexible computational framework for merging it with other computational tools. AVAILABILITY AND IMPLEMENTATION: A3D standalone is distributed under the MIT license, which is free for academic and non-profit users. It is implemented in Python. The A3D standalone source code, wiki with documentation and examples of use, and installation instructions for Linux, macOS and Windows are available in the A3D standalone repository at https://bitbucket.org/lcbio/aggrescan3d.


Asunto(s)
Programas Informáticos , Agregado de Proteínas , Conformación Proteica , Proteínas , Solubilidad
8.
Bioinformatics ; 35(4): 694-695, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30101282

RESUMEN

SUMMARY: CABS-flex standalone is a Python package for fast simulations of protein structure flexibility. The package combines simulations of protein dynamics using CABS coarse-grained protein model with the reconstruction of selected models to all-atom representation and analysis of modeling results. CABS-flex standalone is designed to allow for command-line access to the CABS computations and complete control over simulation process. CABS-flex standalone is equipped with features such as: modeling of multimeric and large-size protein systems, contact map visualizations, analysis of similarities to the reference structure and configurable modeling protocol. For instance, the user may modify the simulation parameters, distance restraints, structural clustering scheme or all-atom reconstruction parameters. With these features CABS-flex standalone can be easily incorporated into other methodologies of structural biology. AVAILABILITY AND IMPLEMENTATION: CABS-flex standalone is distributed under the MIT license, which is free for academic and non-profit users. It is implemented in Python. CABS-flex source code, wiki with examples of use and installation instructions for Linux, macOS and Windows are available from the CABS-flex standalone repository at https://bitbucket.org/lcbio/cabsflex.


Asunto(s)
Conformación Proteica , Proteínas/química , Programas Informáticos
9.
Brief Bioinform ; 20(6): 2299-2305, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30247502

RESUMEN

CABS-dock is a computational method for protein-peptide molecular docking that does not require predefinition of the binding site. The peptide is treated as fully flexible, while the protein backbone undergoes small fluctuations and, optionally, large-scale rearrangements. Here, we present a specific CABS-dock protocol that enhances the docking procedure using fragmentary information about protein-peptide contacts. The contact information is used to narrow down the search for the binding peptide pose to the proximity of the binding site. We used information on a single-chosen and randomly chosen native protein-peptide contact to validate the protocol on the peptiDB benchmark. The contact information significantly improved CABS-dock performance. The protocol has been made available as a new feature of the CABS-dock web server (at http://biocomp.chem.uw.edu.pl/CABSdock/). SHORT ABSTRACT: CABS-dock is a tool for flexible docking of peptides to proteins. In this article, we present a protocol for CABS-dock docking driven by information about protein-peptide contact(s). Using information on individual protein-peptide contacts allows to improve the accuracy of CABS-dock docking.


Asunto(s)
Simulación del Acoplamiento Molecular , Péptidos/metabolismo , Proteínas/metabolismo , Unión Proteica
10.
Nucleic Acids Res ; 46(W1): W338-W343, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29762700

RESUMEN

Classical simulations of protein flexibility remain computationally expensive, especially for large proteins. A few years ago, we developed a fast method for predicting protein structure fluctuations that uses a single protein model as the input. The method has been made available as the CABS-flex web server and applied in numerous studies of protein structure-function relationships. Here, we present a major update of the CABS-flex web server to version 2.0. The new features include: extension of the method to significantly larger and multimeric proteins, customizable distance restraints and simulation parameters, contact maps and a new, enhanced web server interface. CABS-flex 2.0 is freely available at http://biocomp.chem.uw.edu.pl/CABSflex2.


Asunto(s)
Internet , Conformación Proteica , Proteínas/química , Programas Informáticos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Proteínas/genética
11.
Drug Discov Today ; 23(8): 1530-1537, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29733895

RESUMEN

Peptides have recently attracted much attention as promising drug candidates. Rational design of peptide-derived therapeutics usually requires structural characterization of the underlying protein-peptide interaction. Given that experimental characterization can be difficult, reliable computational tools are needed. In recent years, a variety of approaches have been developed for 'protein-peptide docking', that is, predicting the structure of the protein-peptide complex, starting from the protein structure and the peptide sequence, including variable degrees of information about the peptide binding site and/or conformation. In this review, we provide an overview of protein-peptide docking methods and outline their capabilities, limitations, and applications in structure-based drug design. Key challenges are also briefly discussed, such as modeling of large-scale conformational changes upon binding, scoring of predicted models, and optimal inclusion of varied types of experimental data and theoretical predictions into an integrative modeling process.


Asunto(s)
Diseño de Fármacos , Simulación del Acoplamiento Molecular , Péptidos/farmacología , Animales , Sitios de Unión , Humanos , Péptidos/síntesis química , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad
12.
Molecules ; 23(2)2018 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-29463040

RESUMEN

Despite considerable advances over the past years in understanding the mechanisms of action and the role of the σ1 receptor, several questions regarding this receptor remain unanswered. This receptor has been identified as a useful target for the treatment of a diverse range of diseases, from various central nervous system disorders to cancer. The recently solved issue of the crystal structure of the σ1 receptor has made elucidating the structure-activity relationship feasible. The interaction of seven representative opioid ligands with the crystal structure of the σ1 receptor (PDB ID: 5HK1) was simulated for the first time using molecular dynamics (MD). Analysis of the MD trajectories has provided the receptor-ligand interaction fingerprints, combining information on the crucial receptor residues and frequency of the residue-ligand contacts. The contact frequencies and the contact maps suggest that for all studied ligands, the hydrophilic (hydrogen bonding) interactions with Glu172 are an important factor for the ligands' affinities toward the σ1 receptor. However, the hydrophobic interactions with Tyr120, Val162, Leu105, and Ile124 also significantly contribute to the ligand-receptor interplay and, in particular, differentiate the action of the agonistic morphine from the antagonistic haloperidol.


Asunto(s)
Analgésicos Opioides/química , Morfina/química , Receptores sigma/química , Analgésicos Opioides/uso terapéutico , Sitios de Unión , Cristalografía por Rayos X , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas/efectos de los fármacos , Ligandos , Simulación de Dinámica Molecular , Morfina/uso terapéutico , Unión Proteica , Relación Estructura-Actividad
13.
Biomed Eng Online ; 16(Suppl 1): 71, 2017 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-28830442

RESUMEN

BACKGROUND: Many protein-protein interactions are mediated by a short linear motif. Usually, amino acid sequences of those motifs are known or can be predicted. It is much harder to experimentally characterize or predict their structure in the bound form. In this work, we test a possibility of using flexible docking of a short linear motif to predict the interaction interface of the EphB4-EphrinB2 complex (a system extensively studied for its significance in tumor progression). METHODS: In the modeling, we only use knowledge about the motif sequence and experimental structures of EphB4-EphrinB2 complex partners. The proposed protocol enables efficient modeling of significant conformational changes in the short linear motif fragment during molecular docking simulation. For the docking simulations, we use the CABS-dock method for docking fully flexible peptides to flexible protein receptors (available as a server at http://biocomp.chem.uw.edu.pl/CABSdock/ ). Based on the docking result, the protein-protein complex is reconstructed and refined. RESULTS: Using this novel protocol, we obtained an accurate EphB4-EphrinB2 interaction model. CONCLUSIONS: The results show that the CABS-dock method may be useful as the primary docking tool in specific protein-protein docking cases similar to EphB4-EphrinB2 complex-that is, where a short linear motif fragment can be identified.


Asunto(s)
Efrina-B2/química , Efrina-B2/metabolismo , Simulación del Acoplamiento Molecular , Receptor EphB4/química , Receptor EphB4/metabolismo , Secuencias de Aminoácidos , Unión Proteica
14.
Biomed Eng Online ; 16(Suppl 1): 73, 2017 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-28830545

RESUMEN

BACKGROUND: The characterization of protein-peptide interactions is a challenge for computational molecular docking. Protein-peptide docking tools face at least two major difficulties: (1) efficient sampling of large-scale conformational changes induced by binding and (2) selection of the best models from a large set of predicted structures. In this paper, we merge an efficient sampling technique with external information about side-chain contacts to sample and select the best possible models. METHODS: In this paper we test a new protocol that uses information about side-chain contacts in CABS-dock protein-peptide docking. As shown in our recent studies, CABS-dock enables efficient modeling of large-scale conformational changes without knowledge about the binding site. However, the resulting set of binding sites and poses is in many cases highly diverse and difficult to score. RESULTS: As we demonstrate here, information about a single side-chain contact can significantly improve the prediction accuracy. Importantly, the imposed constraints for side-chain contacts are quite soft. Therefore, the developed protocol does not require precise contact information and ensures large-scale peptide flexibility in the broad contact area. CONCLUSIONS: The demonstrated protocol provides the extension of the CABS-dock method that can be practically used in the structure prediction of protein-peptide complexes guided by the knowledge of the binding interface.


Asunto(s)
Simulación del Acoplamiento Molecular , Péptidos/química , Péptidos/metabolismo , Proteínas/química , Proteínas/metabolismo , Sitios de Unión , Unión Proteica , Conformación Proteica
15.
Methods Mol Biol ; 1561: 69-94, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28236234

RESUMEN

Protein-peptide molecular docking is a difficult modeling problem. It is even more challenging when significant conformational changes that may occur during the binding process need to be predicted. In this chapter, we demonstrate the capabilities and features of the CABS-dock server for flexible protein-peptide docking. CABS-dock allows highly efficient modeling of full peptide flexibility and significant flexibility of a protein receptor. During CABS-dock docking, the peptide folding and binding process is explicitly simulated and no information about the peptide binding site or its structure is used. This chapter presents a successful CABS-dock use for docking a potentially therapeutic peptide to a protein target. Moreover, simulation contact maps, a new CABS-dock feature, are described and applied to the docking test case. Finally, a tutorial for running CABS-dock from the command line or command line scripts is provided. The CABS-dock web server is available from http://biocomp.chem.uw.edu.pl/CABSdock/ .


Asunto(s)
Bases de Datos de Proteínas , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Proteínas/metabolismo , Programas Informáticos , Sitios de Unión , Humanos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Unión Proteica , Conformación Proteica , Proteínas/química , Navegador Web
16.
Sci Rep ; 6: 37532, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27905468

RESUMEN

Protein-peptide interactions are often associated with large-scale conformational changes that are difficult to study either by classical molecular modeling or by experiment. Recently, we have developed the CABS-dock method for flexible protein-peptide docking that enables large-scale rearrangements of the protein chain. In this study, we use CABS-dock to investigate the binding of the p53-MDM2 complex, an element of the cell cycle regulation system crucial for anti-cancer drug design. Experimental data suggest that p53-MDM2 binding is affected by significant rearrangements of a lid region - the N-terminal highly flexible MDM2 fragment; however, the details are not clear. The large size of the highly flexible MDM2 fragments makes p53-MDM2 intractable for exhaustive binding dynamics studies using atomistic models. We performed extensive dynamics simulations using the CABS-dock method, including large-scale structural rearrangements of MDM2 flexible regions. Without a priori knowledge of the p53 peptide structure or its binding site, we obtained near-native models of the p53-MDM2 complex. The simulation results match well the experimental data and provide new insights into the possible role of the lid fragment in p53 binding. The presented case study demonstrates that CABS-dock methodology opens up new opportunities for protein-peptide docking with large-scale changes of the protein receptor structure.


Asunto(s)
Simulación del Acoplamiento Molecular/métodos , Proteínas Proto-Oncogénicas c-mdm2/química , Proteína p53 Supresora de Tumor/química , Sitios de Unión , Humanos , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Termodinámica , Proteína p53 Supresora de Tumor/metabolismo
17.
Methods ; 93: 72-83, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26165956

RESUMEN

Protein-peptide interactions play essential functional roles in living organisms and their structural characterization is a hot subject of current experimental and theoretical research. Computational modeling of the structure of protein-peptide interactions is usually divided into two stages: prediction of the binding site at a protein receptor surface, and then docking (and modeling) the peptide structure into the known binding site. This paper presents a comprehensive CABS-dock method for the simultaneous search of binding sites and flexible protein-peptide docking, available as a user's friendly web server. We present example CABS-dock results obtained in the default CABS-dock mode and using its advanced options that enable the user to increase the range of flexibility for chosen receptor fragments or to exclude user-selected binding modes from docking search. Furthermore, we demonstrate a strategy to improve CABS-dock performance by assessing the quality of models with classical molecular dynamics. Finally, we discuss the promising extensions and applications of the CABS-dock method and provide a tutorial appendix for the convenient analysis and visualization of CABS-dock results. The CABS-dock web server is freely available at http://biocomp.chem.uw.edu.pl/CABSdock/.


Asunto(s)
Modelos Moleculares , Simulación del Acoplamiento Molecular/métodos , Péptidos/metabolismo , Proteínas/metabolismo , Navegador Web , Sitios de Unión/fisiología , Péptidos/química , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas/química
18.
Molecules ; 20(6): 10763-80, 2015 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-26111167

RESUMEN

Most of the current docking procedures are focused on fine conformational adjustments of assembled complexes and fail to reproduce large-scale protein motion. In this paper, we test a new modeling approach developed to address this problem. CABS-dock is a versatile and efficient tool for modeling the structure, dynamics and interactions of protein complexes. The docking protocol employs a coarse-grained representation of proteins, a simplified model of interactions and advanced protocols for conformational sampling. CABS-dock is one of the very few tools that allow unrestrained docking with large conformational freedom of the receptor. In an example application we modeled the process of complex assembly between two proteins: Troponin C (TnC) and the N-terminal helix of Troponin I (TnI N-helix), which occurs in vivo during muscle contraction. Docking simulations illustrated how the TnC molecule undergoes significant conformational transition on complex formation, a phenomenon that can be modeled only when protein flexibility is properly accounted for. This way our procedure opens up a new possibility for studying mechanisms of protein complex assembly, which may be a supporting tool for rational drug design.


Asunto(s)
Contracción Muscular , Péptidos/química , Conformación Proteica , Troponina C/química , Troponina I/química , Secuencia de Aminoácidos , Proteínas Portadoras/metabolismo , Humanos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Péptidos/metabolismo , Unión Proteica , Estructura Secundaria de Proteína , Troponina C/metabolismo , Troponina I/metabolismo
19.
Nucleic Acids Res ; 43(W1): W419-24, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25943545

RESUMEN

Protein-peptide interactions play a key role in cell functions. Their structural characterization, though challenging, is important for the discovery of new drugs. The CABS-dock web server provides an interface for modeling protein-peptide interactions using a highly efficient protocol for the flexible docking of peptides to proteins. While other docking algorithms require pre-defined localization of the binding site, CABS-dock does not require such knowledge. Given a protein receptor structure and a peptide sequence (and starting from random conformations and positions of the peptide), CABS-dock performs simulation search for the binding site allowing for full flexibility of the peptide and small fluctuations of the receptor backbone. This protocol was extensively tested over the largest dataset of non-redundant protein-peptide interactions available to date (including bound and unbound docking cases). For over 80% of bound and unbound dataset cases, we obtained models with high or medium accuracy (sufficient for practical applications). Additionally, as optional features, CABS-dock can exclude user-selected binding modes from docking search or to increase the level of flexibility for chosen receptor fragments. CABS-dock is freely available as a web server at http://biocomp.chem.uw.edu.pl/CABSdock.


Asunto(s)
Simulación del Acoplamiento Molecular/métodos , Péptidos/química , Conformación Proteica , Programas Informáticos , Sitios de Unión , Internet , Péptidos/metabolismo , Proteínas/química , Proteínas/metabolismo
20.
J Chem Theory Comput ; 10(6): 2224-31, 2014 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-26580746

RESUMEN

A complex of the phosphorylated kinase-inducible domain (pKID) with its interacting domain (KIX) is a model system for studies of mechanisms by which intrinsically unfolded proteins perform their functions. These mechanisms are not fully understood. Using an efficient coarse-grained model, ab initio simulations were performed of the coupled folding and binding of the pKID to the KIX. The simulations start from an unbound, randomly positioned and disordered pKID structure. During the simulations the pKID chain and its position remain completely unrestricted, while the KIX backbone is limited to near-native fluctuations. Ab initio simulations of such large-scale conformational transitions, unaffected by any knowledge about the bound pKID structure, remain inaccessible to classical simulations. Our simulations recover an ensemble of transient encounter complexes in good agreement with experimental results. We find that a key folding and binding step is linked to the formation of weak native interactions between a preformed nativelike fragment of a pKID helix and KIX surface. Once that nucleus forms, the pKID chain may condense from a largely disordered encounter ensemble to a natively bound and ordered conformation. The observed mechanism is reminiscent of a nucleation-condensation model, a common scenario for folding of globular proteins.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...