Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
Proteomics ; 23(17): e2200323, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37365936

RESUMEN

Reliably scoring and ranking candidate models of protein complexes and assigning their oligomeric state from the structure of the crystal lattice represent outstanding challenges. A community-wide effort was launched to tackle these challenges. The latest resources on protein complexes and interfaces were exploited to derive a benchmark dataset consisting of 1677 homodimer protein crystal structures, including a balanced mix of physiological and non-physiological complexes. The non-physiological complexes in the benchmark were selected to bury a similar or larger interface area than their physiological counterparts, making it more difficult for scoring functions to differentiate between them. Next, 252 functions for scoring protein-protein interfaces previously developed by 13 groups were collected and evaluated for their ability to discriminate between physiological and non-physiological complexes. A simple consensus score generated using the best performing score of each of the 13 groups, and a cross-validated Random Forest (RF) classifier were created. Both approaches showed excellent performance, with an area under the Receiver Operating Characteristic (ROC) curve of 0.93 and 0.94, respectively, outperforming individual scores developed by different groups. Additionally, AlphaFold2 engines recalled the physiological dimers with significantly higher accuracy than the non-physiological set, lending support to the reliability of our benchmark dataset annotations. Optimizing the combined power of interface scoring functions and evaluating it on challenging benchmark datasets appears to be a promising strategy.


Asunto(s)
Proteínas , Reproducibilidad de los Resultados , Proteínas/metabolismo , Unión Proteica
2.
J Mol Biol ; 435(14): 168155, 2023 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-37356902

RESUMEN

Multiple sequence alignments (MSAs) are the workhorse of molecular evolution and structural biology research. From MSAs, the amino acids that are tolerated at each site during protein evolution can be inferred. However, little is known regarding the repertoire of tolerated amino acids in proteins when only a few or no sequence homologs are available, such as orphan and de novo designed proteins. Here we present EvoRator2, a deep-learning algorithm trained on over 15,000 protein structures that can predict which amino acids are tolerated at any given site, based exclusively on protein structural information mined from atomic coordinate files. We show that EvoRator2 obtained satisfying results for the prediction of position-weighted scoring matrices (PSSM). We further show that EvoRator2 obtained near state-of-the-art performance on proteins with high quality structures in predicting the effect of mutations in deep mutation scanning (DMS) experiments and that for certain DMS targets, EvoRator2 outperformed state-of-the-art methods. We also show that by combining EvoRator2's predictions with those obtained by a state-of-the-art deep-learning method that accounts for the information in the MSA, the prediction of the effect of mutation in DMS experiments was improved in terms of both accuracy and stability. EvoRator2 is designed to predict which amino-acid substitutions are tolerated in such proteins without many homologous sequences, including orphan or de novo designed proteins. We implemented our approach in the EvoRator web server (https://evorator.tau.ac.il).


Asunto(s)
Sustitución de Aminoácidos , Aprendizaje Profundo , Algoritmos , Aminoácidos/genética , Biología Computacional/métodos , Proteínas/química , Proteínas/genética , Conformación Proteica
3.
PLoS Comput Biol ; 19(2): e1010874, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36730443

RESUMEN

Design of peptide binders is an attractive strategy for targeting "undruggable" protein-protein interfaces. Current design protocols rely on the extraction of an initial sequence from one known protein interactor of the target protein, followed by in-silico or in-vitro mutagenesis-based optimization of its binding affinity. Wet lab protocols can explore only a minor portion of the vast sequence space and cannot efficiently screen for other desirable properties such as high specificity and low toxicity, while in-silico design requires intensive computational resources and often relies on simplified binding models. Yet, for a multivalent protein target, dozens to hundreds of natural protein partners already exist in the cellular environment. Here, we describe a peptide design protocol that harnesses this diversity via a machine learning generative model. After identifying putative natural binding fragments by literature and homology search, a compositional Restricted Boltzmann Machine is trained and sampled to yield hundreds of diverse candidate peptides. The latter are further filtered via flexible molecular docking and an in-vitro microchip-based binding assay. We validate and test our protocol on calcineurin, a calcium-dependent protein phosphatase involved in various cellular pathways in health and disease. In a single screening round, we identified multiple 16-length peptides with up to six mutations from their closest natural sequence that successfully interfere with the binding of calcineurin to its substrates. In summary, integrating protein interaction and sequence databases, generative modeling, molecular docking and interaction assays enables the discovery of novel protein-protein interaction modulators.


Asunto(s)
Calcineurina , Péptidos , Calcineurina/química , Calcineurina/genética , Calcineurina/metabolismo , Simulación del Acoplamiento Molecular , Péptidos/química , Unión Proteica
4.
Cell Rep ; 41(3): 111512, 2022 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-36223774

RESUMEN

The SARS-CoV-2 Omicron variant evades most neutralizing vaccine-induced antibodies and is associated with lower antibody titers upon breakthrough infections than previous variants. However, the mechanism remains unclear. Here, we find using a geometric deep-learning model that Omicron's extensively mutated receptor binding site (RBS) features reduced antigenicity compared with previous variants. Mice immunization experiments with different recombinant receptor binding domain (RBD) variants confirm that the serological response to Omicron is drastically attenuated and less potent. Analyses of serum cross-reactivity and competitive ELISA reveal a reduction in antibody response across both variable and conserved RBD epitopes. Computational modeling confirms that the RBS has a potential for further antigenicity reduction while retaining efficient receptor binding. Finally, we find a similar trend of antigenicity reduction over decades for hCoV229E, a common cold coronavirus. Thus, our study explains the reduced antibody titers associated with Omicron infection and reveals a possible trajectory of future viral evolution.


Asunto(s)
COVID-19 , Vacunas Virales , Ratones , Animales , Glicoproteína de la Espiga del Coronavirus , Pruebas de Neutralización , Anticuerpos Antivirales/química , SARS-CoV-2 , Anticuerpos Neutralizantes/química , Epítopos/química
5.
J Mol Biol ; 434(19): 167758, 2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-36116806

RESUMEN

Predicting the various binding sites of a protein from its structure sheds light on its function and paves the way towards design of interaction inhibitors. Here, we report ScanNet, a freely available web server for prediction of protein-protein, protein - disordered protein and protein - antibody binding sites from structure. ScanNet (Spatio-Chemical Arrangement of Neighbors Network) is an end-to-end, interpretable geometric deep learning model that learns spatio-chemical patterns directly from 3D structures. ScanNet consistently outperforms Machine Learning models based on handcrafted features and comparative modeling approaches. The web server is linked to both the PDB and AlphaFoldDB, and supports user-provided structure files. Predictions can be readily visualized on the website via the Molstar web app and locally via ChimeraX. ScanNet is available at http://bioinfo3d.cs.tau.ac.il/ScanNet/.


Asunto(s)
Aprendizaje Profundo , Uso de Internet , Unión Proteica , Proteínas , Programas Informáticos , Sitios de Unión , Proteínas/química
6.
Proteins ; 90(11): 1886-1895, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35598299

RESUMEN

Designing peptides for protein-protein interaction inhibition is of significant interest in computer-aided drug design. Such inhibitory peptides could mimic and compete with the binding of the partner protein to the inhibition target. Experimental peptide design is a laborious, time consuming, and expensive multi-step process. Therefore, in silico peptide design can be beneficial for achieving this task. We present a novel algorithm, Pep-Whisperer, which aims to design inhibitory peptides for protein-protein interaction. The desirable peptides would have a relatively high predicted binding affinity to the target protein in a given protein-protein complex. The algorithm outputs linear peptides which are based on an initial template. The template could either be a peptide which is retrieved from the interaction site, or a patch of nonconsecutive amino acids from the protein-protein interface which is completed to a linear peptide by short polyalanine linkers. In addition, the algorithm takes into consideration the conservation of the amino acids in the ligand-protein binding site by using evolutionary information for choosing the preferred amino acids in each position of the designed peptide. Our algorithm was able to design peptides with high predicted binding affinity to the target protein. The method is fully automated and available as a web server at http://bioinfo3d.cs.tau.ac.il/PepWhisperer/.


Asunto(s)
Péptidos , Proteínas , Aminoácidos/metabolismo , Diseño de Fármacos , Ligandos , Péptidos/química , Unión Proteica , Proteínas/química
7.
Nat Methods ; 19(6): 730-739, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35637310

RESUMEN

Predicting the functional sites of a protein from its structure, such as the binding sites of small molecules, other proteins or antibodies, sheds light on its function in vivo. Currently, two classes of methods prevail: machine learning models built on top of handcrafted features and comparative modeling. They are, respectively, limited by the expressivity of the handcrafted features and the availability of similar proteins. Here, we introduce ScanNet, an end-to-end, interpretable geometric deep learning model that learns features directly from 3D structures. ScanNet builds representations of atoms and amino acids based on the spatio-chemical arrangement of their neighbors. We train ScanNet for detecting protein-protein and protein-antibody binding sites, demonstrate its accuracy-including for unseen protein folds-and interpret the filters learned. Finally, we predict epitopes of the SARS-CoV-2 spike protein, validating known antigenic regions and predicting previously uncharacterized ones. Overall, ScanNet is a versatile, powerful and interpretable model suitable for functional site prediction tasks. A webserver for ScanNet is available from http://bioinfo3d.cs.tau.ac.il/ScanNet/ .


Asunto(s)
COVID-19 , Aprendizaje Profundo , Sitios de Unión , Humanos , Unión Proteica , Proteínas/química , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus
8.
bioRxiv ; 2022 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-35194608

RESUMEN

SARS-CoV-2 Omicron variant of concern (VOC) contains fifteen mutations on the receptor binding domain (RBD), evading most neutralizing antibodies from vaccinated sera. Emerging evidence suggests that Omicron breakthrough cases are associated with substantially lower antibody titers than other VOC cases. However, the mechanism remains unclear. Here, using a novel geometric deep-learning model, we discovered that the antigenic profile of Omicron RBD is distinct from the prior VOCs, featuring reduced antigenicity in its remodeled receptor binding sites (RBS). To substantiate our deep-learning prediction, we immunized mice with different recombinant RBD variants and found that the Omicron's extensive mutations can lead to a drastically attenuated serologic response with limited neutralizing activity in vivo , while the T cell response remains potent. Analyses of serum cross-reactivity and competitive ELISA with epitope-specific nanobodies revealed that the antibody response to Omicron was reduced across RBD epitopes, including both the variable RBS and epitopes without any known VOC mutations. Moreover, computational modeling confirmed that the RBS is highly versatile with a capacity to further decrease antigenicity while retaining efficient receptor binding. Longitudinal analysis showed that this evolutionary trend of decrease in antigenicity was also found in hCoV229E, a common cold coronavirus that has been circulating in humans for decades. Thus, our study provided unprecedented insights into the reduced antibody titers associated with Omicron infection, revealed a possible trajectory of future viral evolution and may inform the vaccine development against future outbreaks.

9.
Methods Mol Biol ; 2315: 111-117, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34302673

RESUMEN

Memdock is a tool for docking α-helical membrane proteins which takes into consideration the lipid bilayer environment. Given two α-helical membrane located protein molecules, the method outputs a list of potential complexes sorted by energy criteria. The program includes three steps: docking, refinement, and re-ranking of the results. All three docking steps have been customized to the membrane environment in order to improve performance and reduce program run-time. In this chapter, we describe the application of our web server, referred to as Memdock, for prediction of the docking complex for a pair of input membrane protein structures. Memdock is freely available for academic users without registration at http://bioinfo3d.cs.tau.ac.il/Memdock/index.html .


Asunto(s)
Biología Computacional/métodos , Proteínas de la Membrana/química , Simulación del Acoplamiento Molecular/métodos , Conformación Proteica en Hélice alfa/fisiología , Algoritmos , Internet , Modelos Moleculares , Programas Informáticos , Interfaz Usuario-Computador
10.
Methods Mol Biol ; 2112: 163-174, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32006285

RESUMEN

Macromolecular complexes play a key role in cellular function. Predicting the structure and dynamics of these complexes is one of the key challenges in structural biology. Docking applications have traditionally been used to predict pairwise interactions between proteins. However, few methods exist for modeling multi-protein assemblies. Here we present two methods, CombDock and DockStar, that can predict multi-protein assemblies starting from subunit structural models. CombDock can assemble subunits without any assumptions about the pairwise interactions between subunits, while DockStar relies on the interaction graph or, alternatively, a homology model or a cryo-electron microscopy (EM) density map of the entire complex. We demonstrate the two methods using RNA polymerase II with 12 subunits and TRiC/CCT chaperonin with 16 subunits.


Asunto(s)
Biología Computacional/métodos , Proteínas/química , Chaperonina con TCP-1/química , Chaperoninas/química , Microscopía por Crioelectrón/métodos , Simulación de Dinámica Molecular , Subunidades de Proteína/química , ARN Polimerasa II/química
11.
Bioinformatics ; 33(14): i30-i36, 2017 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-28881968

RESUMEN

MOTIVATION: A highly efficient template-based protein-protein docking algorithm, nicknamed SnapDock, is presented. It employs a Geometric Hashing-based structural alignment scheme to align the target proteins to the interfaces of non-redundant protein-protein interface libraries. Docking of a pair of proteins utilizing the 22 600 interface PIFACE library is performed in < 2 min on the average. A flexible version of the algorithm allowing hinge motion in one of the proteins is presented as well. RESULTS: To evaluate the performance of the algorithm a blind re-modelling of 3547 PDB complexes, which have been uploaded after the PIFACE publication has been performed with success ratio of about 35%. Interestingly, a similar experiment with the template free PatchDock docking algorithm yielded a success rate of about 23% with roughly 1/3 of the solutions different from those of SnapDock. Consequently, the combination of the two methods gave a 42% success ratio. AVAILABILITY AND IMPLEMENTATION: A web server of the application is under development. CONTACT: michaelestrin@gmail.com or wolfson@tau.ac.il.


Asunto(s)
Biología Computacional/métodos , Simulación del Acoplamiento Molecular , Mapeo de Interacción de Proteínas/métodos , Programas Informáticos , Algoritmos
12.
Methods Mol Biol ; 1561: 279-290, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28236244

RESUMEN

In this chapter we present two methods related to rational design of inhibitory peptides: PepCrawler: A tool to derive binding peptides from protein-protein complexes and the prediction of protein-peptide complexes. Given an initial protein-peptide complex, the method detects improved predicted peptide binding conformations which bind the protein with higher affinity. This program is a robotics motivated algorithm, representing the peptide as a robotic arm moving among obstacles and exploring its conformational space in an efficient way. PinaColada: A peptide design program for the discovery of novel peptide candidates that inhibit protein-protein interactions. PinaColada uses PepCrawler while introducing sequence mutations, in order to find novel inhibitory peptides for PPIs. It uses the ant colony optimization approach to explore the peptide's sequence space, while using PepCrawler in the refinement stage.


Asunto(s)
Algoritmos , Biología Computacional/métodos , Diseño de Fármacos , Fragmentos de Péptidos/farmacología , Mapas de Interacción de Proteínas/efectos de los fármacos , Proteínas/metabolismo , Humanos , Modelos Moleculares , Fragmentos de Péptidos/química , Unión Proteica , Conformación Proteica , Proteínas/química
13.
Bioinformatics ; 32(15): 2289-96, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27153578

RESUMEN

MOTIVATION: Design of protein-protein interaction (PPI) inhibitors is a major challenge in Structural Bioinformatics. Peptides, especially short ones (5-15 amino acid long), are natural candidates for inhibition of protein-protein complexes due to several attractive features such as high structural compatibility with the protein binding site (mimicking the surface of one of the proteins), small size and the ability to form strong hotspot binding connections with the protein surface. Efficient rational peptide design is still a major challenge in computer aided drug design, due to the huge space of possible sequences, which is exponential in the length of the peptide, and the high flexibility of peptide conformations. RESULTS: In this article we present PinaColada, a novel computational method for the design of peptide inhibitors for protein-protein interactions. We employ a version of the ant colony optimization heuristic, which is used to explore the exponential space ([Formula: see text]) of length n peptide sequences, in combination with our fast robotics motivated PepCrawler algorithm, which explores the conformational space for each candidate sequence. PinaColada is being run in parallel, on a DELL PowerEdge 2.8 GHZ computer with 20 cores and 256 GB memory, and takes up to 24 h to design a peptide of 5-15 amino acids length. AVAILABILITY AND IMPLEMENTATION: An online server available at: http://bioinfo3d.cs.tau.ac.il/PinaColada/. CONTACT: danielza@post.tau.ac.il; wolfson@tau.ac.il.


Asunto(s)
Algoritmos , Biología Computacional/métodos , Péptidos , Proteínas
14.
Bioinformatics ; 32(16): 2444-50, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27153621

RESUMEN

MOTIVATION: A wide range of fundamental biological processes are mediated by membrane proteins. Despite their large number and importance, less than 1% of all 3D protein structures deposited in the Protein Data Bank are of membrane proteins. This is mainly due to the challenges of crystallizing such proteins or performing NMR spectroscopy analyses. All the more so, there is only a small number of membrane protein-protein complexes with known structure. Therefore, developing computational tools for docking membrane proteins is crucial. Numerous methods for docking globular proteins exist, however few have been developed especially for membrane proteins and designed to address docking within the lipid bilayer environment. RESULTS: We present a novel algorithm, Memdock, for docking α-helical membrane proteins which takes into consideration the lipid bilayer environment for docking as well as for refining and ranking the docking candidates. We show that our algorithm improves both the docking accuracy and the candidates ranking compared to a standard protein-protein docking algorithm. AVAILABILITY AND IMPLEMENTATION: http://bioinfo3d.cs.tau.ac.il/Memdock/ CONTACTS: namih@tau.ac.il or wolfson@tau.ac.il SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Algoritmos , Proteínas de la Membrana , Modelos Moleculares , Bases de Datos de Proteínas
15.
Bioinformatics ; 31(17): 2801-7, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25913207

RESUMEN

MOTIVATION: Atomic resolution modeling of large multimolecular assemblies is a key task in Structural Cell Biology. Experimental techniques can provide atomic resolution structures of single proteins and small complexes, or low resolution data of large multimolecular complexes. RESULTS: We present a novel integrative computational modeling method, which integrates both low and high resolution experimental data. The algorithm accepts as input atomic resolution structures of the individual subunits obtained from X-ray, NMR or homology modeling, and interaction data between the subunits obtained from mass spectrometry. The optimal assembly of the individual subunits is formulated as an Integer Linear Programming task. The method was tested on several representative complexes, both in the bound and unbound cases. It placed correctly most of the subunits of multimolecular complexes of up to 16 subunits and significantly outperformed the CombDock and Haddock multimolecular docking methods. AVAILABILITY AND IMPLEMENTATION: http://bioinfo3d.cs.tau.ac.il/DockStar CONTACT: naamaamir@mail.tau.ac.il or wolfson@tau.ac.il SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Algoritmos , Biología Computacional/métodos , Modelos Moleculares , Complejos Multiproteicos/química , Proteínas/química , Homología Estructural de Proteína , Animales , Bovinos , Humanos , Simulación del Acoplamiento Molecular , Conformación Proteica , Proteínas de Saccharomyces cerevisiae
16.
Oncoscience ; 1(1): 39-48, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25593987

RESUMEN

LIM kinases (LIMKs) are important cell cytoskeleton regulators that play a prominent role in cancer manifestation and neuronal diseases. The LIMK family consists of two homologues, LIMK1 and LIMK2, which differ from one another in expression profile, intercellular localization, and function. The main substrate of LIMK is cofilin, a member of the actin-depolymerizing factor (ADF) protein family. When phosphorylated by LIMK, cofilin is inactive. LIMKs play a contributory role in several neurodevelopmental disorders and in cancer growth and metastasis. We recently reported the development and validation of a novel LIMK inhibitor, referred to here as T56-LIMKi, using a combination of computational methods and classical biochemistry techniques. Here we report that T56-LIMKi inhibits LIMK2 with high specificity, and shows little or no cross-reactivity with LIMK1. We found that T56-LIMKi decreases phosphorylated cofilin (p-cofilin) levels and thus inhibits growth of several cancerous cell lines, including those of pancreatic cancer, glioma and schwannoma. Because the most promising in-vitro effect of T56-LIMKi was observed in the pancreatic cancer cell line Panc-1, we tested the inhibitor on a nude mouse Panc-1 xenograft model. T56-LIMKi reduced tumor size and p-cofilin levels in the Panc-1 tumors, leading us to propose T56-LIMKi as a candidate drug for cancer therapy.

17.
Proteins ; 82(4): 620-32, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24155158

RESUMEN

We report the first assessment of blind predictions of water positions at protein-protein interfaces, performed as part of the critical assessment of predicted interactions (CAPRI) community-wide experiment. Groups submitting docking predictions for the complex of the DNase domain of colicin E2 and Im2 immunity protein (CAPRI Target 47), were invited to predict the positions of interfacial water molecules using the method of their choice. The predictions-20 groups submitted a total of 195 models-were assessed by measuring the recall fraction of water-mediated protein contacts. Of the 176 high- or medium-quality docking models-a very good docking performance per se-only 44% had a recall fraction above 0.3, and a mere 6% above 0.5. The actual water positions were in general predicted to an accuracy level no better than 1.5 Å, and even in good models about half of the contacts represented false positives. This notwithstanding, three hotspot interface water positions were quite well predicted, and so was one of the water positions that is believed to stabilize the loop that confers specificity in these complexes. Overall the best interface water predictions was achieved by groups that also produced high-quality docking models, indicating that accurate modelling of the protein portion is a determinant factor. The use of established molecular mechanics force fields, coupled to sampling and optimization procedures also seemed to confer an advantage. Insights gained from this analysis should help improve the prediction of protein-water interactions and their role in stabilizing protein complexes.


Asunto(s)
Colicinas/química , Mapeo de Interacción de Proteínas , Agua/química , Algoritmos , Biología Computacional , Modelos Moleculares , Simulación del Acoplamiento Molecular , Conformación Proteica
18.
Colloids Surf B Biointerfaces ; 112: 16-22, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23933103

RESUMEN

The ß-lactoglobulin (ß-LG) protein was discovered to be an efficient and selective dispersant for carbon nanotubes (CTNs) with certain diameters. A dispersion process of CTNs by the ß-LG was studied, focusing on the relationships between the surface curvature of the CNT and the ß-LG's efficiency in dispersing them, using cryogenic-transmission electron microscopy (cryo-TEM) and optical spectroscopy. Plausible binding sites of the ß-LG, responsible for the interaction of the protein with CNTs of various diameters (surface curvatures) were also investigated and were found to be in good agreement with corresponding docking calculations.


Asunto(s)
Lactoglobulinas/química , Proteínas de la Leche/química , Nanotubos de Carbono/química , Nanotubos de Carbono/ultraestructura , Animales , Sitios de Unión , Bovinos , Dicroismo Circular , Microscopía por Crioelectrón , Microscopía Electrónica de Transmisión , Modelos Moleculares , Tamaño de la Partícula , Estructura Cuaternaria de Proteína , Espectrofotometría , Proteína de Suero de Leche
19.
Nucleic Acids Res ; 41(21): 9956-66, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23980029

RESUMEN

Translin is a highly conserved RNA- and DNA-binding protein that plays essential roles in eukaryotic cells. Human translin functions as an octamer, but in the octameric crystallographic structure, the residues responsible for nucleic acid binding are not accessible. Moreover, electron microscopy data reveal very different octameric configurations. Consequently, the functional assembly and the mechanism of nucleic acid binding by the protein remain unclear. Here, we present an integrative study combining small-angle X-ray scattering (SAXS), site-directed mutagenesis, biochemical analysis and computational techniques to address these questions. Our data indicate a significant conformational heterogeneity for translin in solution, formed by a lesser-populated compact octameric state resembling the previously solved X-ray structure, and a highly populated open octameric state that had not been previously identified. On the other hand, our SAXS data and computational analyses of translin in complex with the RNA oligonucleotide (GU)12 show that the internal cavity found in the octameric assemblies can accommodate different nucleic acid conformations. According to this model, the nucleic acid binding residues become accessible for binding, which facilitates the entrance of the nucleic acids into the cavity. Our data thus provide a structural basis for the functions that translin performs in RNA metabolism and transport.


Asunto(s)
Proteínas de Unión al ADN/química , ARN/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Multimerización de Proteína , ARN/metabolismo
20.
EMBO J ; 32(4): 538-51, 2013 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-23361315

RESUMEN

The ubiquitylation signal promotes trafficking of endogenous and retroviral transmembrane proteins. The signal is decoded by a large set of ubiquitin (Ub) receptors that tether Ub-binding domains (UBDs) to the trafficking machinery. We developed a structure-based procedure to scan the protein data bank for hidden UBDs. The screen retrieved many of the known UBDs. Intriguingly, new potential UBDs were identified, including the ALIX-V domain. Pull-down, cross-linking and E3-independent ubiquitylation assays biochemically corroborated the in silico findings. Guided by the output model, we designed mutations at the postulated ALIX-V:Ub interface. Biophysical affinity measurements using microscale-thermophoresis of wild-type and mutant proteins revealed some of the interacting residues of the complex. ALIX-V binds mono-Ub with a K(d) of 119 µM. We show that ALIX-V oligomerizes with a Hill coefficient of 5.4 and IC(50) of 27.6 µM and that mono-Ub induces ALIX-V oligomerization. Moreover, we show that ALIX-V preferentially binds K63 di-Ub compared with mono-Ub and K48 di-Ub. Finally, an in vivo functionality assay demonstrates the significance of ALIX-V:Ub interaction in equine infectious anaemia virus budding. These results not only validate the new procedure, but also demonstrate that ALIX-V directly interacts with Ub in vivo and that this interaction can influence retroviral budding.


Asunto(s)
Virus de la Anemia Infecciosa Equina/metabolismo , Complejos Multienzimáticos , Mutación , Ubiquitina-Proteína Ligasas , Liberación del Virus/fisiología , Animales , Humanos , Virus de la Anemia Infecciosa Equina/genética , Ratones , Modelos Biológicos , Complejos Multienzimáticos/química , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Estructura Terciaria de Proteína , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA