6
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      How accurately can one predict drug binding modes using AlphaFold models?

      research-article
      1 , 2 , 3 , 4 , 5 , 2 , 3 , 4 , 5 , 1 , 2 , 3 , 4 , 5 ,
      ,
      eLife
      eLife Sciences Publications, Ltd
      ligand pose prediction, GPCR, drug discovery, small molecule, homology modeling, molecular docking, None

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Computational prediction of protein structure has been pursued intensely for decades, motivated largely by the goal of using structural models for drug discovery. Recently developed machine-learning methods such as AlphaFold 2 (AF2) have dramatically improved protein structure prediction, with reported accuracy approaching that of experimentally determined structures. To what extent do these advances translate to an ability to predict more accurately how drugs and drug candidates bind to their target proteins? Here, we carefully examine the utility of AF2 protein structure models for predicting binding poses of drug-like molecules at the largest class of drug targets, the G-protein-coupled receptors. We find that AF2 models capture binding pocket structures much more accurately than traditional homology models, with errors nearly as small as differences between structures of the same protein determined experimentally with different ligands bound. Strikingly, however, the accuracy of ligand-binding poses predicted by computational docking to AF2 models is not significantly higher than when docking to traditional homology models and is much lower than when docking to structures determined experimentally without these ligands bound. These results have important implications for all those who might use predicted protein structures for drug discovery.

          Related collections

          Most cited references49

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Highly accurate protein structure prediction with AlphaFold

          Proteins are essential to life, and understanding their structure can facilitate a mechanistic understanding of their function. Through an enormous experimental effort 1 – 4 , the structures of around 100,000 unique proteins have been determined 5 , but this represents a small fraction of the billions of known protein sequences 6 , 7 . Structural coverage is bottlenecked by the months to years of painstaking effort required to determine a single protein structure. Accurate computational approaches are needed to address this gap and to enable large-scale structural bioinformatics. Predicting the three-dimensional structure that a protein will adopt based solely on its amino acid sequence—the structure prediction component of the ‘protein folding problem’ 8 —has been an important open research problem for more than 50 years 9 . Despite recent progress 10 – 14 , existing methods fall far short of atomic accuracy, especially when no homologous structure is available. Here we provide the first computational method that can regularly predict protein structures with atomic accuracy even in cases in which no similar structure is known. We validated an entirely redesigned version of our neural network-based model, AlphaFold, in the challenging 14th Critical Assessment of protein Structure Prediction (CASP14) 15 , demonstrating accuracy competitive with experimental structures in a majority of cases and greatly outperforming other methods. Underpinning the latest version of AlphaFold is a novel machine learning approach that incorporates physical and biological knowledge about protein structure, leveraging multi-sequence alignments, into the design of the deep learning algorithm. AlphaFold predicts protein structures with an accuracy competitive with experimental structures in the majority of cases using a novel deep learning architecture.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            UCSF Chimera--a visualization system for exploratory research and analysis.

            The design, implementation, and capabilities of an extensible visualization system, UCSF Chimera, are discussed. Chimera is segmented into a core that provides basic services and visualization, and extensions that provide most higher level functionality. This architecture ensures that the extension mechanism satisfies the demands of outside developers who wish to incorporate new features. Two unusual extensions are presented: Multiscale, which adds the ability to visualize large-scale molecular assemblies such as viral coats, and Collaboratory, which allows researchers to share a Chimera session interactively despite being at separate locales. Other extensions include Multalign Viewer, for showing multiple sequence alignments and associated structures; ViewDock, for screening docked ligand orientations; Movie, for replaying molecular dynamics trajectories; and Volume Viewer, for display and analysis of volumetric data. A discussion of the usage of Chimera in real-world situations is given, along with anticipated future directions. Chimera includes full user documentation, is free to academic and nonprofit users, and is available for Microsoft Windows, Linux, Apple Mac OS X, SGI IRIX, and HP Tru64 Unix from http://www.cgl.ucsf.edu/chimera/. Copyright 2004 Wiley Periodicals, Inc.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              The Protein Data Bank.

              The Protein Data Bank (PDB; http://www.rcsb.org/pdb/ ) is the single worldwide archive of structural data of biological macromolecules. This paper describes the goals of the PDB, the systems in place for data deposition and access, how to obtain further information, and near-term plans for the future development of the resource.
                Bookmark

                Author and article information

                Contributors
                Role: Reviewing Editor
                Role: Senior Editor
                Journal
                eLife
                Elife
                eLife
                eLife
                eLife Sciences Publications, Ltd
                2050-084X
                22 December 2023
                2023
                : 12
                : RP89386
                Affiliations
                [1 ] Biophysics Program, Stanford University ( https://ror.org/00f54p054) Stanford United States
                [2 ] Department of Computer Science, Stanford University ( https://ror.org/00f54p054) Stanford United States
                [3 ] Department of Molecular and Cellular Physiology, Stanford University School of Medicine ( https://ror.org/00f54p054) Stanford United States
                [4 ] Department of Structural Biology, Stanford University School of Medicine ( https://ror.org/00f54p054) Stanford United States
                [5 ] Institute for Computational and Mathematical Engineering, Stanford University ( https://ror.org/00f54p054) Stanford United States
                Arrakis Therapeutics ( https://ror.org/049d04r12) United States
                Boston University ( https://ror.org/05qwgg493) United States
                Arrakis Therapeutics United States
                Stanford University Stanford United States
                Stanford University Stanford United States
                Stanford University Stanford United States
                Author information
                https://orcid.org/0000-0003-1880-4536
                https://orcid.org/0000-0002-6418-2793
                Article
                89386
                10.7554/eLife.89386
                10746139
                38131311
                ca315d95-e3fb-4038-a3fd-3d01b3de24a1
                © 2023, Karelina et al

                This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.

                History
                : 30 May 2023
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/501100008982, National Science Foundation;
                Award ID: Graduate Research Fellowship Program
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100004191, Novo Nordisk;
                Award Recipient :
                The funders had no role in study design, data collection, and interpretation, or the decision to submit the work for publication.
                Categories
                Research Article
                Structural Biology and Molecular Biophysics
                Custom metadata
                AlphaFold 2 models capture binding pocket structures accurately but fare poorly when used to predict ligand-binding poses.
                prc

                Life sciences
                ligand pose prediction,gpcr,drug discovery,small molecule,homology modeling,molecular docking,none

                Comments

                Comment on this article