Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Struct Dyn ; 11(3): 034701, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38774441

RESUMEN

Studying protein dynamics and conformational heterogeneity is crucial for understanding biomolecular systems and treating disease. Despite the deposition of over 215 000 macromolecular structures in the Protein Data Bank and the advent of AI-based structure prediction tools such as AlphaFold2, RoseTTAFold, and ESMFold, static representations are typically produced, which fail to fully capture macromolecular motion. Here, we discuss the importance of integrating experimental structures with computational clustering to explore the conformational landscapes that manifest protein function. We describe the method developed by the Protein Data Bank in Europe - Knowledge Base to identify distinct conformational states, demonstrate the resource's primary use cases, through examples, and discuss the need for further efforts to annotate protein conformations with functional information. Such initiatives will be crucial in unlocking the potential of protein dynamics data, expediting drug discovery research, and deepening our understanding of macromolecular mechanisms.

2.
Curr Protoc ; 4(5): e1047, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38720559

RESUMEN

Recent advancements in protein structure determination and especially in protein structure prediction techniques have led to the availability of vast amounts of macromolecular structures. However, the accessibility and integration of these structures into scientific workflows are hindered by the lack of standardization among publicly available data resources. To address this issue, we introduced the 3D-Beacons Network, a unified platform that aims to establish a standardized framework for accessing and displaying protein structure data. In this article, we highlight the importance of standardized approaches for accessing protein structure data and showcase the capabilities of 3D-Beacons. We describe four protocols for finding and accessing macromolecular structures from various specialist data resources via 3D-Beacons. First, we describe three scenarios for programmatically accessing and retrieving data using the 3D-Beacons API. Next, we show how to perform sequence-based searches to find structures from model providers. Then, we demonstrate how to search for structures and fetch them directly into a workflow using JalView. Finally, we outline the process of facilitating access to data from providers interested in contributing their structures to the 3D-Beacons Network. © 2024 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Programmatic access to the 3D-Beacons API Basic Protocol 2: Sequence-based search using the 3D-Beacons API Basic Protocol 3: Accessing macromolecules from 3D-Beacons with JalView Basic Protocol 4: Enhancing data accessibility through 3D-Beacons.


Asunto(s)
Conformación Proteica , Proteínas , Proteínas/química , Bases de Datos de Proteínas , Programas Informáticos
3.
Nucleic Acids Res ; 52(D1): D10-D17, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-38015445

RESUMEN

The European Molecular Biology Laboratory's European Bioinformatics Institute (EMBL-EBI) is one of the world's leading sources of public biomolecular data. Based at the Wellcome Genome Campus in Hinxton, UK, EMBL-EBI is one of six sites of the European Molecular Biology Laboratory (EMBL), Europe's only intergovernmental life sciences organisation. This overview summarises the latest developments in the services provided by EMBL-EBI data resources to scientific communities globally. These developments aim to ensure EMBL-EBI resources meet the current and future needs of these scientific communities, accelerating the impact of open biological data for all.


Asunto(s)
Academias e Institutos , Biología Computacional , Biología Computacional/organización & administración , Biología Computacional/tendencias , Academias e Institutos/organización & administración , Academias e Institutos/tendencias , Bases de Datos de Ácidos Nucleicos , Europa (Continente)
4.
Nucleic Acids Res ; 52(D1): D368-D375, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-37933859

RESUMEN

The AlphaFold Database Protein Structure Database (AlphaFold DB, https://alphafold.ebi.ac.uk) has significantly impacted structural biology by amassing over 214 million predicted protein structures, expanding from the initial 300k structures released in 2021. Enabled by the groundbreaking AlphaFold2 artificial intelligence (AI) system, the predictions archived in AlphaFold DB have been integrated into primary data resources such as PDB, UniProt, Ensembl, InterPro and MobiDB. Our manuscript details subsequent enhancements in data archiving, covering successive releases encompassing model organisms, global health proteomes, Swiss-Prot integration, and a host of curated protein datasets. We detail the data access mechanisms of AlphaFold DB, from direct file access via FTP to advanced queries using Google Cloud Public Datasets and the programmatic access endpoints of the database. We also discuss the improvements and services added since its initial release, including enhancements to the Predicted Aligned Error viewer, customisation options for the 3D viewer, and improvements in the search engine of AlphaFold DB.


The AlphaFold Protein Structure Database (AlphaFold DB) is a massive digital library of predicted protein structures, with over 214 million entries, marking a 500-times expansion in size since its initial release in 2021. The structures are predicted using Google DeepMind's AlphaFold 2 artificial intelligence (AI) system. Our new report highlights the latest updates we have made to this database. We have added more data on specific organisms and proteins related to global health and expanded to cover almost the complete UniProt database, a primary data resource of protein sequences. We also made it easier for our users to access the data by directly downloading files or using advanced cloud-based tools. Finally, we have also improved how users view and search through these protein structures, making the user experience smoother and more informative. In short, AlphaFold DB has been growing rapidly and has become more user-friendly and robust to support the broader scientific community.


Asunto(s)
Inteligencia Artificial , Estructura Secundaria de Proteína , Proteoma , Secuencia de Aminoácidos , Bases de Datos de Proteínas , Motor de Búsqueda , Proteínas/química
5.
J Cheminform ; 15(1): 117, 2023 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-38042830

RESUMEN

While the Protein Data Bank (PDB) contains a wealth of structural information on ligands bound to macromolecules, their analysis can be challenging due to the large amount and diversity of data. Here, we present PDBe CCDUtils, a versatile toolkit for processing and analysing small molecules from the PDB in PDBx/mmCIF format. PDBe CCDUtils provides streamlined access to all the metadata for small molecules in the PDB and offers a set of convenient methods to compute various properties using RDKit, such as 2D depictions, 3D conformers, physicochemical properties, scaffolds, common fragments, and cross-references to small molecule databases using UniChem. The toolkit also provides methods for identifying all the covalently attached chemical components in a macromolecular structure and calculating similarity among small molecules. By providing a broad range of functionality, PDBe CCDUtils caters to the needs of researchers in cheminformatics, structural biology, bioinformatics and computational chemistry.

6.
Sci Data ; 10(1): 853, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-38040737

RESUMEN

Macromolecular complexes are essential functional units in nearly all cellular processes, and their atomic-level understanding is critical for elucidating and modulating molecular mechanisms. The Protein Data Bank (PDB) serves as the global repository for experimentally determined structures of macromolecules. Structural data in the PDB offer valuable insights into the dynamics, conformation, and functional states of biological assemblies. However, the current annotation practices lack standardised naming conventions for assemblies in the PDB, complicating the identification of instances representing the same assembly. In this study, we introduce a method leveraging resources external to PDB, such as the Complex Portal, UniProt and Gene Ontology, to describe assemblies and contextualise them within their biological settings accurately. Employing the proposed approach, we assigned standard names to over 90% of unique assemblies in the PDB and provided persistent identifiers for each assembly. This standardisation of assembly data enhances the PDB, facilitating a deeper understanding of macromolecular complexes. Furthermore, the data standardisation improves the PDB's FAIR attributes, fostering more effective basic and translational research and scientific education.


Asunto(s)
Investigación Biomédica Traslacional , Conformación Molecular , Bases de Datos de Proteínas , Sustancias Macromoleculares , Conformación Proteica
7.
Bioinformatics ; 39(12)2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-38085238

RESUMEN

SUMMARY: PDBImages is an innovative, open-source Node.js package that harnesses the power of the popular macromolecule structure visualization software Mol*. Designed for use by the scientific community, PDBImages provides a means to generate high-quality images for PDB and AlphaFold DB models. Its unique ability to render and save images directly to files in a browserless mode sets it apart, offering users a streamlined, automated process for macromolecular structure visualization. Here, we detail the implementation of PDBImages, enumerating its diverse image types, and elaborating on its user-friendly setup. This powerful tool opens a new gateway for researchers to visualize, analyse, and share their work, fostering a deeper understanding of bioinformatics. AVAILABILITY AND IMPLEMENTATION: PDBImages is available as an npm package from https://www.npmjs.com/package/pdb-images. The source code is available from https://github.com/PDBeurope/pdb-images.


Asunto(s)
Biología Computacional , Programas Informáticos , Estructura Molecular , Biología Computacional/métodos
8.
Proteins ; 2023 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-37850517

RESUMEN

The rapid evolution of protein structure prediction tools has significantly broadened access to protein structural data. Although predicted structure models have the potential to accelerate and impact fundamental and translational research significantly, it is essential to note that they are not validated and cannot be considered the ground truth. Thus, challenges persist, particularly in capturing protein dynamics, predicting multi-chain structures, interpreting protein function, and assessing model quality. Interdisciplinary collaborations are crucial to overcoming these obstacles. Databases like the AlphaFold Protein Structure Database, the ESM Metagenomic Atlas, and initiatives like the 3D-Beacons Network provide FAIR access to these data, enabling their interpretation and application across a broader scientific community. Whilst substantial advancements have been made in protein structure prediction, further progress is required to address the remaining challenges. Developing training materials, nurturing collaborations, and ensuring open data sharing will be paramount in this pursuit. The continued evolution of these tools and methodologies will deepen our understanding of protein function and accelerate disease pathogenesis and drug development discoveries.

9.
Nature ; 622(7983): 637-645, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37704730

RESUMEN

Proteins are key to all cellular processes and their structure is important in understanding their function and evolution. Sequence-based predictions of protein structures have increased in accuracy1, and over 214 million predicted structures are available in the AlphaFold database2. However, studying protein structures at this scale requires highly efficient methods. Here, we developed a structural-alignment-based clustering algorithm-Foldseek cluster-that can cluster hundreds of millions of structures. Using this method, we have clustered all of the structures in the AlphaFold database, identifying 2.30 million non-singleton structural clusters, of which 31% lack annotations representing probable previously undescribed structures. Clusters without annotation tend to have few representatives covering only 4% of all proteins in the AlphaFold database. Evolutionary analysis suggests that most clusters are ancient in origin but 4% seem to be species specific, representing lower-quality predictions or examples of de novo gene birth. We also show how structural comparisons can be used to predict domain families and their relationships, identifying examples of remote structural similarity. On the basis of these analyses, we identify several examples of human immune-related proteins with putative remote homology in prokaryotic species, illustrating the value of this resource for studying protein function and evolution across the tree of life.


Asunto(s)
Algoritmos , Análisis por Conglomerados , Proteínas , Homología Estructural de Proteína , Humanos , Bases de Datos de Proteínas , Proteínas/química , Proteínas/clasificación , Proteínas/metabolismo , Alineación de Secuencia , Anotación de Secuencia Molecular , Células Procariotas/química , Filogenia , Especificidad de la Especie , Evolución Molecular
10.
Sci Data ; 10(1): 204, 2023 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-37045837

RESUMEN

More than 61,000 proteins have up-to-date correspondence between their amino acid sequence (UniProtKB) and their 3D structures (PDB), enabled by the Structure Integration with Function, Taxonomy and Sequences (SIFTS) resource. SIFTS incorporates residue-level annotations from many other biological resources. SIFTS data is available in various formats like XML, CSV and TSV format or also accessible via the PDBe REST API but always maintained separately from the structure data (PDBx/mmCIF file) in the PDB archive. Here, we extended the wwPDB PDBx/mmCIF data dictionary with additional categories to accommodate SIFTS data and added the UniProtKB, Pfam, SCOP2, and CATH residue-level annotations directly into the PDBx/mmCIF files from the PDB archive. With the integrated UniProtKB annotations, these files now provide consistent numbering of residues in different PDB entries allowing easy comparison of structure models. The extended dictionary yields a more consistent, standardised metadata description without altering the core PDB information. This development enables up-to-date cross-reference information at the residue level resulting in better data interoperability, supporting improved data analysis and visualisation.

11.
J Mol Biol ; 435(14): 168021, 2023 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-36828268

RESUMEN

ModelCIF (github.com/ihmwg/ModelCIF) is a data information framework developed for and by computational structural biologists to enable delivery of Findable, Accessible, Interoperable, and Reusable (FAIR) data to users worldwide. ModelCIF describes the specific set of attributes and metadata associated with macromolecular structures modeled by solely computational methods and provides an extensible data representation for deposition, archiving, and public dissemination of predicted three-dimensional (3D) models of macromolecules. It is an extension of the Protein Data Bank Exchange / macromolecular Crystallographic Information Framework (PDBx/mmCIF), which is the global data standard for representing experimentally-determined 3D structures of macromolecules and associated metadata. The PDBx/mmCIF framework and its extensions (e.g., ModelCIF) are managed by the Worldwide Protein Data Bank partnership (wwPDB, wwpdb.org) in collaboration with relevant community stakeholders such as the wwPDB ModelCIF Working Group (wwpdb.org/task/modelcif). This semantically rich and extensible data framework for representing computed structure models (CSMs) accelerates the pace of scientific discovery. Herein, we describe the architecture, contents, and governance of ModelCIF, and tools and processes for maintaining and extending the data standard. Community tools and software libraries that support ModelCIF are also described.


Asunto(s)
Bases de Datos de Proteínas , Sustancias Macromoleculares/química , Conformación Proteica , Programas Informáticos
12.
Curr Opin Struct Biol ; 79: 102543, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36807079

RESUMEN

The function of proteins can often be inferred from their three-dimensional structures. Experimental structural biologists spent decades studying these structures, but the accelerated pace of protein sequencing continuously increases the gaps between sequences and structures. The early 2020s saw the advent of a new generation of deep learning-based protein structure prediction tools that offer the potential to predict structures based on any number of protein sequences. In this review, we give an overview of the impact of this new generation of structure prediction tools, with examples of the impacted field in the life sciences. We discuss the novel opportunities and new scientific and technical challenges these tools present to the broader scientific community. Finally, we highlight some potential directions for the future of computational protein structure prediction.


Asunto(s)
Aprendizaje Profundo , Biología Computacional/métodos , Proteínas/química , Secuencia de Aminoácidos
13.
Proteomics ; 23(17): e2200128, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-36382391

RESUMEN

Arguably, 2020 was the year of high-accuracy protein structure predictions, with AlphaFold 2.0 achieving previously unseen accuracy in the Critical Assessment of Protein Structure Prediction (CASP). In 2021, DeepMind and EMBL-EBI developed the AlphaFold Protein Structure Database to make an unprecedented number of reliable protein structure predictions easily accessible to the broad scientific community. We provide a brief overview and describe the latest developments in the AlphaFold database. We highlight how the fields of data services, bioinformatics, structural biology, and drug discovery are directly affected by the influx of protein structure data. We also show examples of cutting-edge research that took advantage of the AlphaFold database. It is apparent that connections between various fields through protein structures are now possible, but the amount of data poses new challenges. Finally, we give an outlook regarding the future direction of the database, both in terms of data sets and new functionalities.


Asunto(s)
Disciplinas de las Ciencias Biológicas , Proteínas , Conformación Proteica , Bases de Datos de Proteínas , Proteínas/química , Biología Computacional
14.
Gigascience ; 112022 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-36448847

RESUMEN

While scientists can often infer the biological function of proteins from their 3-dimensional quaternary structures, the gap between the number of known protein sequences and their experimentally determined structures keeps increasing. A potential solution to this problem is presented by ever more sophisticated computational protein modeling approaches. While often powerful on their own, most methods have strengths and weaknesses. Therefore, it benefits researchers to examine models from various model providers and perform comparative analysis to identify what models can best address their specific use cases. To make data from a large array of model providers more easily accessible to the broader scientific community, we established 3D-Beacons, a collaborative initiative to create a federated network with unified data access mechanisms. The 3D-Beacons Network allows researchers to collate coordinate files and metadata for experimentally determined and theoretical protein models from state-of-the-art and specialist model providers and also from the Protein Data Bank.


Asunto(s)
Metadatos , Registros , Secuencia de Aminoácidos , Bases de Datos de Proteínas , Simulación por Computador
15.
Protein Sci ; 31(10): e4439, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36173162

RESUMEN

The archiving and dissemination of protein and nucleic acid structures as well as their structural, functional and biophysical annotations is an essential task that enables the broader scientific community to conduct impactful research in multiple fields of the life sciences. The Protein Data Bank in Europe (PDBe; pdbe.org) team develops and maintains several databases and web services to address this fundamental need. From data archiving as a member of the Worldwide PDB consortium (wwPDB; wwpdb.org), to the PDBe Knowledge Base (PDBe-KB; pdbekb.org), we provide data, data-access mechanisms, and visualizations that facilitate basic and applied research and education across the life sciences. Here, we provide an overview of the structural data and annotations that we integrate and make freely available. We describe the web services and data visualization tools we offer, and provide information on how to effectively use or even further develop them. Finally, we discuss the direction of our data services, and how we aim to tackle new challenges that arise from the recent, unprecedented advances in the field of structure determination and protein structure modeling.


Asunto(s)
Ácidos Nucleicos , Proteínas , Bases de Datos de Proteínas , Europa (Continente) , Conformación Proteica , Proteínas/química
16.
Int J Mol Sci ; 23(16)2022 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-36012140

RESUMEN

The number of unique transmembrane (TM) protein structures doubled in the last four years, which can be attributed to the revolution of cryo-electron microscopy. In addition, AlphaFold2 (AF2) also provided a large number of predicted structures with high quality. However, if a specific protein family is the subject of a study, collecting the structures of the family members is highly challenging in spite of existing general and protein domain-specific databases. Here, we demonstrate this and assess the applicability and usability of automatic collection and presentation of protein structures via the ABC protein superfamily. Our pipeline identifies and classifies transmembrane ABC protein structures using the PFAM search and also aims to determine their conformational states based on special geometric measures, conftors. Since the AlphaFold database contains structure predictions only for single polypeptide chains, we performed AF2-Multimer predictions for human ABC half transporters functioning as dimers. Our AF2 predictions warn of possibly ambiguous interpretation of some biochemical data regarding interaction partners and call for further experiments and experimental structure determination. We made our predicted ABC protein structures available through a web application, and we joined the 3D-Beacons Network to reach the broader scientific community through platforms such as PDBe-KB.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Furilfuramida , Transportadoras de Casetes de Unión a ATP/metabolismo , Inteligencia Artificial , Biología , Microscopía por Crioelectrón , Humanos , Modelos Moleculares , Conformación Proteica
17.
Nucleic Acids Res ; 50(D1): D439-D444, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34791371

RESUMEN

The AlphaFold Protein Structure Database (AlphaFold DB, https://alphafold.ebi.ac.uk) is an openly accessible, extensive database of high-accuracy protein-structure predictions. Powered by AlphaFold v2.0 of DeepMind, it has enabled an unprecedented expansion of the structural coverage of the known protein-sequence space. AlphaFold DB provides programmatic access to and interactive visualization of predicted atomic coordinates, per-residue and pairwise model-confidence estimates and predicted aligned errors. The initial release of AlphaFold DB contains over 360,000 predicted structures across 21 model-organism proteomes, which will soon be expanded to cover most of the (over 100 million) representative sequences from the UniRef90 data set.


Asunto(s)
Bases de Datos de Proteínas , Pliegue de Proteína , Proteínas/química , Programas Informáticos , Secuencia de Aminoácidos , Animales , Bacterias/genética , Bacterias/metabolismo , Conjuntos de Datos como Asunto , Dictyostelium/genética , Dictyostelium/metabolismo , Hongos/genética , Hongos/metabolismo , Humanos , Internet , Modelos Moleculares , Plantas/genética , Plantas/metabolismo , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Proteínas/genética , Proteínas/metabolismo , Trypanosoma cruzi/genética , Trypanosoma cruzi/metabolismo
18.
BMC Bioinformatics ; 22(1): 383, 2021 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-34301175

RESUMEN

BACKGROUND: Biomacromolecular structural data outgrew the legacy Protein Data Bank (PDB) format which the scientific community relied on for decades, yet the use of its successor PDBx/Macromolecular Crystallographic Information File format (PDBx/mmCIF) is still not widespread. Perhaps one of the reasons is the availability of easy to use tools that only support the legacy format, but also the inherent difficulties of processing mmCIF files correctly, given the number of edge cases that make efficient parsing problematic. Nevertheless, to fully exploit macromolecular structure data and their associated annotations such as multiscale structures from integrative/hybrid methods or large macromolecular complexes determined using traditional methods, it is necessary to fully adopt the new format as soon as possible. RESULTS: To this end, we developed PDBeCIF, an open-source Python project for manipulating mmCIF and CIF files. It is part of the official list of mmCIF parsers recorded by the wwPDB and is heavily employed in the processes of the Protein Data Bank in Europe. The package is freely available both from the PyPI repository ( http://pypi.org/project/pdbecif ) and from GitHub ( https://github.com/pdbeurope/pdbecif ) along with rich documentation and many ready-to-use examples. CONCLUSIONS: PDBeCIF is an efficient and lightweight Python 2.6+/3+ package with no external dependencies. It can be readily integrated with 3rd party libraries as well as adopted for broad scientific analyses.


Asunto(s)
Programas Informáticos , Bases de Datos de Proteínas , Europa (Continente) , Sustancias Macromoleculares , Estructura Molecular
19.
Bioinformatics ; 37(21): 3950-3952, 2021 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-34081107

RESUMEN

SUMMARY: The PDBe aggregated API is an open-access and open-source RESTful API that provides programmatic access to a wealth of macromolecular structural data and their functional and biophysical annotations through 80+ API endpoints. The API is powered by the PDBe graph database (https://pdbe.org/graph-schema), an open-access integrative knowledge graph that can be used as a discovery tool to answer complex biological questions. AVAILABILITY AND IMPLEMENTATION: The PDBe aggregated API provides up-to-date access to the PDBe graph database, which has weekly releases with the latest data from the Protein Data Bank, integrated with updated annotations from UniProt, Pfam, CATH, SCOP and the PDBe-KB partner resources. The complete list of all the available API endpoints and their descriptions are available at https://pdbe.org/graph-api. The source code of the Python 3.6+ API application is publicly available at https://gitlab.ebi.ac.uk/pdbe-kb/services/pdbe-graph-api. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Reconocimiento de Normas Patrones Automatizadas , Programas Informáticos , Estructura Molecular , Bases de Datos de Proteínas , Conformación Proteica
20.
Brief Bioinform ; 22(2): 742-768, 2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33348379

RESUMEN

SARS-CoV-2 is the causative agent of COVID-19, the ongoing global pandemic. It has posed a worldwide challenge to human health as no effective treatment is currently available to combat the disease. Its severity has led to unprecedented collaborative initiatives for therapeutic solutions against COVID-19. Studies resorting to structure-based drug design for COVID-19 are plethoric and show good promise. Structural biology provides key insights into 3D structures, critical residues/mutations in SARS-CoV-2 proteins, implicated in infectivity, molecular recognition and susceptibility to a broad range of host species. The detailed understanding of viral proteins and their complexes with host receptors and candidate epitope/lead compounds is the key to developing a structure-guided therapeutic design. Since the discovery of SARS-CoV-2, several structures of its proteins have been determined experimentally at an unprecedented speed and deposited in the Protein Data Bank. Further, specialized structural bioinformatics tools and resources have been developed for theoretical models, data on protein dynamics from computer simulations, impact of variants/mutations and molecular therapeutics. Here, we provide an overview of ongoing efforts on developing structural bioinformatics tools and resources for COVID-19 research. We also discuss the impact of these resources and structure-based studies, to understand various aspects of SARS-CoV-2 infection and therapeutic development. These include (i) understanding differences between SARS-CoV-2 and SARS-CoV, leading to increased infectivity of SARS-CoV-2, (ii) deciphering key residues in the SARS-CoV-2 involved in receptor-antibody recognition, (iii) analysis of variants in host proteins that affect host susceptibility to infection and (iv) analyses facilitating structure-based drug and vaccine design against SARS-CoV-2.


Asunto(s)
Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Biología Computacional , SARS-CoV-2/aislamiento & purificación , COVID-19/virología , Humanos , Conformación Proteica , Proteínas Virales/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...