1
|
Zeng C, Zhuo C, Gao J, Liu H, Zhao Y. Advances and Challenges in Scoring Functions for RNA-Protein Complex Structure Prediction. Biomolecules 2024; 14:1245. [PMID: 39456178 PMCID: PMC11506084 DOI: 10.3390/biom14101245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
RNA-protein complexes play a crucial role in cellular functions, providing insights into cellular mechanisms and potential therapeutic targets. However, experimental determination of these complex structures is often time-consuming and resource-intensive, and it rarely yields high-resolution data. Many computational approaches have been developed to predict RNA-protein complex structures in recent years. Despite these advances, achieving accurate and high-resolution predictions remains a formidable challenge, primarily due to the limitations inherent in current RNA-protein scoring functions. These scoring functions are critical tools for evaluating and interpreting RNA-protein interactions. This review comprehensively explores the latest advancements in scoring functions for RNA-protein docking, delving into the fundamental principles underlying various approaches, including coarse-grained knowledge-based, all-atom knowledge-based, and machine-learning-based methods. We critically evaluate the strengths and limitations of existing scoring functions, providing a detailed performance assessment. Considering the significant progress demonstrated by machine learning techniques, we discuss emerging trends and propose future research directions to enhance the accuracy and efficiency of scoring functions in RNA-protein complex prediction. We aim to inspire the development of more sophisticated and reliable computational tools in this rapidly evolving field.
Collapse
Affiliation(s)
| | | | | | | | - Yunjie Zhao
- Institute of Biophysics and Department of Physics, Central China Normal University, Wuhan 430079, China; (C.Z.); (C.Z.); (J.G.); (H.L.)
| |
Collapse
|
2
|
Collins KW, Copeland MM, Brysbaert G, Wodak SJ, Bonvin AMJJ, Kundrotas PJ, Vakser IA, Lensink MF. CAPRI-Q: The CAPRI resource evaluating the quality of predicted structures of protein complexes. J Mol Biol 2024; 436:168540. [PMID: 39237205 PMCID: PMC11458157 DOI: 10.1016/j.jmb.2024.168540] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 09/07/2024]
Abstract
Protein interactions are essential for cellular processes. In recent years there has been significant progress in computational prediction of 3D structures of individual protein chains, with the best-performing algorithms reaching sub-Ångström accuracy. These techniques are now finding their way into the prediction of protein interactions, adding to the existing modeling approaches. The community-wide Critical Assessment of Predicted Interactions (CAPRI) has been a catalyst for the development of procedures for the structural modeling of protein assemblies by organizing blind prediction experiments. The predicted structures are assessed against unpublished experimentally determined structures using a set of metrics with proven robustness that have been established in the CAPRI community. In addition, several advanced benchmarking databases provide targets against which users can test docking and assembly modeling software. These include the Protein-Protein Docking Benchmark, the CAPRI Scoreset, and the Dockground database, all developed by members of the CAPRI community. Here we present CAPRI-Q, a stand-alone model quality assessment tool, which can be freely downloaded or used via a publicly available web server. This tool applies the CAPRI metrics to assess the quality of query structures against given target structures, along with other popular quality metrics such as DockQ, TM-score and l-DDT, and classifies the models according to the CAPRI model quality criteria. The tool can handle a variety of protein complex types including those involving peptides, nucleic acids, and oligosaccharides. The source code is freely available from https://gitlab.in2p3.fr/cmsb-public/CAPRI-Q and its web interface through the Dockground resource at https://dockground.compbio.ku.edu/assessment/.
Collapse
Affiliation(s)
- Keeley W Collins
- Computational Biology Program, The University of Kansas, Lawrence, KS 66045, USA
| | - Matthew M Copeland
- Computational Biology Program, The University of Kansas, Lawrence, KS 66045, USA
| | - Guillaume Brysbaert
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | | | - Alexandre M J J Bonvin
- Bijvoet Centre for Biomolecular Research, Faculty of Science - Chemistry, Utrecht University, The Netherlands
| | - Petras J Kundrotas
- Computational Biology Program, The University of Kansas, Lawrence, KS 66045, USA.
| | - Ilya A Vakser
- Computational Biology Program, The University of Kansas, Lawrence, KS 66045, USA; Department of Molecular Biology, The University of Kansas, Lawrence, KS 66045, USA.
| | - Marc F Lensink
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France.
| |
Collapse
|
3
|
Kiani YS, Jabeen I. Challenges of Protein-Protein Docking of the Membrane Proteins. Methods Mol Biol 2024; 2780:203-255. [PMID: 38987471 DOI: 10.1007/978-1-0716-3985-6_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Despite the recent advances in the determination of high-resolution membrane protein (MP) structures, the structural and functional characterization of MPs remains extremely challenging, mainly due to the hydrophobic nature, low abundance, poor expression, purification, and crystallization difficulties associated with MPs. Whereby the major challenges/hurdles for MP structure determination are associated with the expression, purification, and crystallization procedures. Although there have been significant advances in the experimental determination of MP structures, only a limited number of MP structures (approximately less than 1% of all) are available in the Protein Data Bank (PDB). Therefore, the structures of a large number of MPs still remain unresolved, which leads to the availability of widely unplumbed structural and functional information related to MPs. As a result, recent developments in the drug discovery realm and the significant biological contemplation have led to the development of several novel, low-cost, and time-efficient computational methods that overcome the limitations of experimental approaches, supplement experiments, and provide alternatives for the characterization of MPs. Whereby the fine tuning and optimizations of these computational approaches remains an ongoing endeavor.Computational methods offer a potential way for the elucidation of structural features and the augmentation of currently available MP information. However, the use of computational modeling can be extremely challenging for MPs mainly due to insufficient knowledge of (or gaps in) atomic structures of MPs. Despite the availability of numerous in silico methods for 3D structure determination the applicability of these methods to MPs remains relatively low since all methods are not well-suited or adequate for MPs. However, sophisticated methods for MP structure predictions are constantly being developed and updated to integrate the modifications required for MPs. Currently, different computational methods for (1) MP structure prediction, (2) stability analysis of MPs through molecular dynamics simulations, (3) modeling of MP complexes through docking, (4) prediction of interactions between MPs, and (5) MP interactions with its soluble partner are extensively used. Towards this end, MP docking is widely used. It is notable that the MP docking methods yet few in number might show greater potential in terms of filling the knowledge gap. In this chapter, MP docking methods and associated challenges have been reviewed to improve the applicability, accuracy, and the ability to model macromolecular complexes.
Collapse
Affiliation(s)
- Yusra Sajid Kiani
- School of Interdisciplinary Engineering and Sciences (SINES), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Ishrat Jabeen
- School of Interdisciplinary Engineering and Sciences (SINES), National University of Sciences and Technology (NUST), Islamabad, Pakistan.
| |
Collapse
|
4
|
Zeng C, Jian Y, Zhuo C, Li A, Zeng C, Zhao Y. Evaluation of DNA-protein complex structures using the deep learning method. Phys Chem Chem Phys 2023; 26:130-143. [PMID: 38063012 DOI: 10.1039/d3cp04980a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
Biological processes such as transcription, repair, and regulation require interactions between DNA and proteins. To unravel their functions, it is imperative to determine the high-resolution structures of DNA-protein complexes. However, experimental methods for this purpose are costly and technically demanding. Consequently, there is an urgent need for computational techniques to identify the structures of DNA-protein complexes. Despite technological advancements, accurately identifying DNA-protein complexes through computational methods still poses a challenge. Our team has developed a cutting-edge deep-learning approach called DDPScore that assesses DNA-protein complex structures. DDPScore utilizes a 4D convolutional neural network to overcome limited training data. This approach effectively captures local and global features while comprehensively considering the conformational changes arising from the flexibility during the DNA-protein docking process. DDPScore consistently outperformed the available methods in comprehensive DNA-protein complex docking evaluations, even for the flexible docking challenges. DDPScore has a wide range of applications in predicting and designing structures of DNA-protein complexes.
Collapse
Affiliation(s)
- Chengwei Zeng
- Institute of Biophysics and Department of Physics, Central China Normal University, Wuhan, 430079, China.
| | - Yiren Jian
- Department of Computer Science, Dartmouth College, Hanover, NH 03755, USA
| | - Chen Zhuo
- Institute of Biophysics and Department of Physics, Central China Normal University, Wuhan, 430079, China.
| | - Anbang Li
- Institute of Biophysics and Department of Physics, Central China Normal University, Wuhan, 430079, China.
| | - Chen Zeng
- Department of Physics, The George Washington University, Washington, DC 20052, USA
| | - Yunjie Zhao
- Institute of Biophysics and Department of Physics, Central China Normal University, Wuhan, 430079, China.
| |
Collapse
|
5
|
Georg Magalhães C, Ploeger Mansueli C, Manieri TM, Quintilio W, Garbuio A, de Jesus Marinho J, de Moraes JZ, Tsuruta LR, Moro AM. Impaired proliferation and migration of HUVEC and melanoma cells by human anti-FGF2 mAbs derived from a murine hybridoma by guided selection. Bioengineered 2023; 14:2252667. [PMID: 37661761 PMCID: PMC10478743 DOI: 10.1080/21655979.2023.2252667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 09/05/2023] Open
Abstract
Disadvantages of using murine monoclonal antibodies (mAb) in human therapy, such as immunogenicity response, led to the development of technologies to transform murine antibodies into human antibodies. The murine anti-FGF2 3F12E7 mAb was proposed as a promising agent to treat metastatic melanoma tumors; once it blocks the FGF2, responsible for playing a role in tumor growth, angiogenesis, and metastasis. Considering the therapeutic potential of anti-FGF2 3F12E7 mAb and its limited use in humans due to its origin, we used this antibody as the template for a guided selection humanization technique to obtain human anti-FGF2 mAbs. Three Fab libraries (murine, hybrid, and human) were constructed for humanization. The libraries were phage-displayed, and the panning was performed against recombinant human FGF2 (rFGF2). The selected human variable light and heavy chains were cloned into AbVec vectors for full-length IgG expression into HEK293-F cells. Surface plasmon resonance analyses showed binding to rFGF2 of seven mAbs out of 20 expressed. Assays performed with these mAbs resulted in two that showed proliferation reduction and cell migration attenuation of HUVEC and SK-Mel-28 melanoma cells. In-silico analyses predicted that these two human anti-FGF2 mAbs interact with FGF2 at a similar patch of residues than the chimeric anti-FGF2 antibody, comprehending a region within the heparin-binding domains of FGF2, essential for its function. These results are comparable to those achieved by the murine anti-FGF2 3F12E7 mAb and showed success in the humanization process and selection of two human mAbs with the potential to inhibit undesirable FGF2 roles.
Collapse
Affiliation(s)
| | | | | | - Wagner Quintilio
- Laboratory of Biopharmaceuticals, Butantan Institute, São Paulo, Brazil
| | - Angélica Garbuio
- Laboratory of Biopharmaceuticals, Butantan Institute, São Paulo, Brazil
| | | | - Jane Zveiter de Moraes
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Ana Maria Moro
- Laboratory of Biopharmaceuticals, Butantan Institute, São Paulo, Brazil
- CeRDI, Center for Research and Development in Immunobiologicals, Butantan Institute, São Paulo, Brazil
| |
Collapse
|
6
|
Pereira GP, Jiménez-García B, Pellarin R, Launay G, Wu S, Martin J, Souza PCT. Rational Prediction of PROTAC-Compatible Protein-Protein Interfaces by Molecular Docking. J Chem Inf Model 2023; 63:6823-6833. [PMID: 37877240 DOI: 10.1021/acs.jcim.3c01154] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
Proteolysis targeting chimeras (PROTACs) are heterobifunctional ligands that mediate the interaction between a protein target and an E3 ligase, resulting in a ternary complex, whose interaction with the ubiquitination machinery leads to target degradation. This technology is emerging as an exciting new avenue for therapeutic development, with several PROTACs currently undergoing clinical trials targeting cancer. Here, we describe a general and computationally efficient methodology combining restraint-based docking, energy-based rescoring, and a filter based on the minimal solvent-accessible surface distance to produce PROTAC-compatible PPIs suitable for when there is no a priori known PROTAC ligand. In a benchmark employing a manually curated data set of 13 ternary complex crystals, we achieved an accuracy of 92% when starting from bound structures and 77% when starting from unbound structures, respectively. Our method only requires that the ligand-bound structures of the monomeric forms of the E3 ligase and target proteins be given to run, making it general, accurate, and highly efficient, with the ability to impact early-stage PROTAC-based drug design campaigns where no structural information about the ternary complex structure is available.
Collapse
Affiliation(s)
- Gilberto P Pereira
- Molecular Microbiology and Structural Biochemistry, CNRS UMR 5086 and Université Claude Bernard Lyon 1, 7 Passage du Vercors, 69007 Lyon, France
- Laboratory of Biology and Modeling of the Cell, École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5239 and Inserm U1293, 46 Allée d'Italie, 69007 Lyon, France
| | | | - Riccardo Pellarin
- Molecular Microbiology and Structural Biochemistry, CNRS UMR 5086 and Université Claude Bernard Lyon 1, 7 Passage du Vercors, 69007 Lyon, France
- Laboratory of Biology and Modeling of the Cell, École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5239 and Inserm U1293, 46 Allée d'Italie, 69007 Lyon, France
| | - Guillaume Launay
- Molecular Microbiology and Structural Biochemistry, CNRS UMR 5086 and Université Claude Bernard Lyon 1, 7 Passage du Vercors, 69007 Lyon, France
- Laboratory of Biology and Modeling of the Cell, École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5239 and Inserm U1293, 46 Allée d'Italie, 69007 Lyon, France
| | - Sangwook Wu
- PharmCADD, Busan 48792, Republic of Korea
- Department of Physics, Pukyong National University, Busan 48513, Republic of Korea
| | - Juliette Martin
- Molecular Microbiology and Structural Biochemistry, CNRS UMR 5086 and Université Claude Bernard Lyon 1, 7 Passage du Vercors, 69007 Lyon, France
- Laboratory of Biology and Modeling of the Cell, École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5239 and Inserm U1293, 46 Allée d'Italie, 69007 Lyon, France
| | - Paulo C T Souza
- Molecular Microbiology and Structural Biochemistry, CNRS UMR 5086 and Université Claude Bernard Lyon 1, 7 Passage du Vercors, 69007 Lyon, France
- Laboratory of Biology and Modeling of the Cell, École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5239 and Inserm U1293, 46 Allée d'Italie, 69007 Lyon, France
| |
Collapse
|
7
|
Kellogg GE, Marabotti A, Spyrakis F, Mozzarelli A. HINT, a code for understanding the interaction between biomolecules: a tribute to Donald J. Abraham. Front Mol Biosci 2023; 10:1194962. [PMID: 37351551 PMCID: PMC10282649 DOI: 10.3389/fmolb.2023.1194962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/24/2023] [Indexed: 06/24/2023] Open
Abstract
A long-lasting goal of computational biochemists, medicinal chemists, and structural biologists has been the development of tools capable of deciphering the molecule-molecule interaction code that produces a rich variety of complex biomolecular assemblies comprised of the many different simple and biological molecules of life: water, small metabolites, cofactors, substrates, proteins, DNAs, and RNAs. Software applications that can mimic the interactions amongst all of these species, taking account of the laws of thermodynamics, would help gain information for understanding qualitatively and quantitatively key determinants contributing to the energetics of the bimolecular recognition process. This, in turn, would allow the design of novel compounds that might bind at the intermolecular interface by either preventing or reinforcing the recognition. HINT, hydropathic interaction, was a model and software code developed from a deceptively simple idea of Donald Abraham with the close collaboration with Glen Kellogg at Virginia Commonwealth University. HINT is based on a function that scores atom-atom interaction using LogP, the partition coefficient of any molecule between two phases; here, the solvents are water that mimics the cytoplasm milieu and octanol that mimics the protein internal hydropathic environment. This review summarizes the results of the extensive and successful collaboration between Abraham and Kellogg at VCU and the group at the University of Parma for testing HINT in a variety of different biomolecular interactions, from proteins with ligands to proteins with DNA.
Collapse
Affiliation(s)
- Glen E. Kellogg
- Department of Medicinal Chemistry and Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA, United States
| | - Anna Marabotti
- Department of Chemistry and Biology “A Zambelli”, University of Salerno, Fisciano (SA), Italy
| | - Francesca Spyrakis
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Andrea Mozzarelli
- Department of Food and Drug, University of Parma and Institute of Biophysics, Parma, Italy
| |
Collapse
|
8
|
Zeng C, Jian Y, Vosoughi S, Zeng C, Zhao Y. Evaluating native-like structures of RNA-protein complexes through the deep learning method. Nat Commun 2023; 14:1060. [PMID: 36828844 PMCID: PMC9958188 DOI: 10.1038/s41467-023-36720-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/14/2023] [Indexed: 02/26/2023] Open
Abstract
RNA-protein complexes underlie numerous cellular processes, including basic translation and gene regulation. The high-resolution structure determination of the RNA-protein complexes is essential for elucidating their functions. Therefore, computational methods capable of identifying the native-like RNA-protein structures are needed. To address this challenge, we thus develop DRPScore, a deep-learning-based approach for identifying native-like RNA-protein structures. DRPScore is tested on representative sets of RNA-protein complexes with various degrees of binding-induced conformation change ranging from fully rigid docking (bound-bound) to fully flexible docking (unbound-unbound). Out of the top 20 predictions, DRPScore selects native-like structures with a success rate of 91.67% on the testing set of bound RNA-protein complexes and 56.14% on the unbound complexes. DRPScore consistently outperforms existing methods with a roughly 10.53-15.79% improvement, even for the most difficult unbound cases. Furthermore, DRPScore significantly improves the accuracy of the native interface interaction predictions. DRPScore should be broadly useful for modeling and designing RNA-protein complexes.
Collapse
Affiliation(s)
- Chengwei Zeng
- Institute of Biophysics and Department of Physics, Central China Normal University, Wuhan, 430079, China
| | - Yiren Jian
- Department of Computer Science, Dartmouth College, Hanover, NH, 03755, USA
| | - Soroush Vosoughi
- Department of Computer Science, Dartmouth College, Hanover, NH, 03755, USA
| | - Chen Zeng
- Department of Physics, The George Washington University, Washington, DC, 20052, USA
| | - Yunjie Zhao
- Institute of Biophysics and Department of Physics, Central China Normal University, Wuhan, 430079, China.
| |
Collapse
|
9
|
Cohen T, Halfon M, Carter L, Sharkey B, Jain T, Sivasubramanian A, Schneidman-Duhovny D. Multi-state modeling of antibody-antigen complexes with SAXS profiles and deep-learning models. Methods Enzymol 2022; 678:237-262. [PMID: 36641210 DOI: 10.1016/bs.mie.2022.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Antibodies are an established class of human therapeutics. Epitope characterization is an important part of therapeutic antibody discovery. However, structural characterization of antibody-antigen complexes remains challenging. On the one hand, X-ray crystallography or cryo-electron microscopy provide atomic resolution characterization of the epitope, but the data collection process is typically long and the success rate is low. On the other hand, computational methods for modeling antibody-antigen structures from the individual components frequently suffer from a high false positive rate, rarely resulting in a unique solution. Recent deep learning models for structure prediction are also successful in predicting protein-protein complexes. However, they do not perform well for antibody-antigen complexes. Small Angle X-ray Scattering (SAXS) is a reliable technique for rapid structural characterization of protein samples in solution albeit at low resolution. Here, we present an integrative approach for modeling antigen-antibody complexes using the antibody sequence, antigen structure, and experimentally determined SAXS profiles of the antibody, antigen, and the complex. The method models antibody structures using a novel deep-learning approach, NanoNet. The structures of the antibodies and antigens are represented using multiple 3D conformations to account for compositional and conformational heterogeneity of the protein samples that are used to collect the SAXS data. The complexes are predicted by integrating the SAXS profiles with scoring functions for protein-protein interfaces that are based on statistical potentials and antibody-specific deep-learning models. We validated the method via application to four Fab:EGFR and one Fab:PCSK9 antibody:antigen complexes with experimentally available SAXS datasets. The integrative approach returns accurate predictions (interface RMSD<4Å) in the top five predictions for four out of five complexes (respective interface RMSD values of 1.95, 2.18, 2.66 and 3.87Å), providing support for the utility of such a computational pipeline for epitope characterization during therapeutic antibody discovery.
Collapse
Affiliation(s)
- Tomer Cohen
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Matan Halfon
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lester Carter
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Menlo Park, CA, United States
| | - Beth Sharkey
- High-Throughput Expression, Adimab LLC, Lebanon, NH, United States
| | - Tushar Jain
- Computational Biology, Adimab LLC, Palo Alto, CA, United States
| | | | - Dina Schneidman-Duhovny
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
10
|
Cohen T, Halfon M, Schneidman-Duhovny D. NanoNet: Rapid and accurate end-to-end nanobody modeling by deep learning. Front Immunol 2022; 13:958584. [PMID: 36032123 PMCID: PMC9411858 DOI: 10.3389/fimmu.2022.958584] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/15/2022] [Indexed: 11/20/2022] Open
Abstract
Antibodies are a rapidly growing class of therapeutics. Recently, single domain camelid VHH antibodies, and their recognition nanobody domain (Nb) appeared as a cost-effective highly stable alternative to full-length antibodies. There is a growing need for high-throughput epitope mapping based on accurate structural modeling of the variable domains that share a common fold and differ in the Complementarity Determining Regions (CDRs). We develop a deep learning end-to-end model, NanoNet, that given a sequence directly produces the 3D coordinates of the backbone and Cβ atoms of the entire VH domain. For the Nb test set, NanoNet achieves 3.16Å average RMSD for the most variable CDR3 loops and 2.65Å, 1.73Å for the CDR1, CDR2 loops, respectively. The accuracy for antibody VH domains is even higher: 2.38Å RMSD for CDR3 and 0.89Å, 0.96Å for the CDR1, CDR2 loops, respectively. NanoNet run times allow generation of ∼1M nanobody structures in less than 4 hours on a standard CPU computer enabling high-throughput structure modeling. NanoNet is available at GitHub: https://github.com/dina-lab3D/NanoNet.
Collapse
Affiliation(s)
- Tomer Cohen
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Dina Schneidman-Duhovny
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
11
|
da Rocha L, Baptista AM, Campos SRR. Approach to Study pH-Dependent Protein Association Using Constant-pH Molecular Dynamics: Application to the Dimerization of β-Lactoglobulin. J Chem Theory Comput 2022; 18:1982-2001. [PMID: 35171602 PMCID: PMC9775224 DOI: 10.1021/acs.jctc.1c01187] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Protein-protein association is often mediated by electrostatic interactions and modulated by pH. However, experimental and computational studies have often overlooked the effect of association on the protonation state of the protein. In this work, we present a methodological approach based on constant-pH molecular dynamics (MD), which aims to provide a detailed description of a pH-dependent protein-protein association, and apply it to the dimerization of β-lactoglobulin (BLG). A selection of analyses is performed using the data generated by constant-pH MD simulations of monomeric and dimeric forms of bovine BLG, in the pH range 3-8. First, we estimate free energies of dimerization using a computationally inexpensive approach based on the Wyman-Tanford linkage theory, calculated in a new way through the use of thermodynamically based splines. The individual free energy contribution of each titratable site is also calculated, allowing for identification of relevant residues. Second, the correlations between the proton occupancies of pairs of sites are calculated (using the Pearson coefficient), and extensive networks of correlated sites are observed at acidic pH values, sometimes involving distant pairs. In general, strongly correlated sites are also slow proton exchangers and contribute significantly to the pH-dependency of the dimerization free energy. Third, we use ionic density as a fingerprint of protein charge distribution and observe electrostatic complementarity between the monomer faces that form the dimer interface, more markedly at the isoionic point (where maximum dimerization occurs) than at other pH values, which might contribute to guide the association. Finally, the pH-dependent dimerization modes are inspected using PCA, among other analyses, and two states are identified: a relaxed state at pH 4-8 (with the typical alignment of the crystallographic structure) and a compact state at pH 3-4 (with a tighter association and rotated alignment). This work shows that an approach based on constant-pH MD simulations can produce rich detailed pictures of pH-dependent protein associations, as illustrated for BLG dimerization.
Collapse
|
12
|
Chen YC, Chen YH, Wright JD, Lim C. PPI-Hotspot DB: Database of Protein-Protein Interaction Hot Spots. J Chem Inf Model 2022; 62:1052-1060. [PMID: 35147037 DOI: 10.1021/acs.jcim.2c00025] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Single-point mutations of certain residues (so-called hot spots) impair/disrupt protein-protein interactions (PPIs), leading to pathogenesis and drug resistance. Conventionally, a PPI-hot spot is identified when its replacement decreased the binding free energy significantly, generally by ≥2 kcal/mol. The relatively few mutations with such a significant binding free energy drop limited the number of distinct PPI-hot spots. By defining PPI-hot spots based on mutations that have been manually curated in UniProtKB to significantly impair/disrupt PPIs in addition to binding free energy changes, we have greatly expanded the number of distinct PPI-hot spots by an order of magnitude. These experimentally determined PPI-hot spots along with available structures have been collected in a database called PPI-HotspotDB. We have applied the PPI-HotspotDB to create a nonredundant benchmark, PPI-Hotspot+PDBBM, for assessing methods to predict PPI-hot spots using the free structure as input. PPI-HotspotDB will benefit the design of mutagenesis experiments and development of PPI-hot spot prediction methods. The database and benchmark are freely available at https://ppihotspot.limlab.dnsalias.org.
Collapse
Affiliation(s)
- Yao Chi Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Yu-Hsien Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Jon D Wright
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Carmay Lim
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan.,Department of Chemistry, National Tsing Hua University, Hsinchu 300, Taiwan
| |
Collapse
|
13
|
Li H, Yan Y, Zhao X, Huang SY. Inclusion of Desolvation Energy into Protein–Protein Docking through Atomic Contact Potentials. J Chem Inf Model 2022; 62:740-750. [DOI: 10.1021/acs.jcim.1c01483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Hao Li
- School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, P. R. China
| | - Yumeng Yan
- School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, P. R. China
| | - Xuejun Zhao
- School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, P. R. China
| | - Sheng-You Huang
- School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, P. R. China
| |
Collapse
|
14
|
Ozden B, Kryshtafovych A, Karaca E. Assessment of the CASP14 assembly predictions. Proteins 2021; 89:1787-1799. [PMID: 34337786 PMCID: PMC9109697 DOI: 10.1002/prot.26199] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/08/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022]
Abstract
In CASP14, 39 research groups submitted more than 2500 3D models on 22 protein complexes. In general, the community performed well in predicting the fold of the assemblies (for 80% of the targets), although it faced significant challenges in reproducing the native contacts. This is especially the case for the complexes without whole-assembly templates. The leading predictor, BAKER-experimental, used a methodology combining classical techniques (template-based modeling, protein docking) with deep learning-based contact predictions and a fold-and-dock approach. The Venclovas team achieved the runner-up position with template-based modeling and docking. By analyzing the target interfaces, we showed that the complexes with depleted charged contacts or dominating hydrophobic interactions were the most challenging ones to predict. We also demonstrated that if AlphaFold2 predictions were at hand, the interface prediction challenge could be alleviated for most of the targets. All in all, it is evident that new approaches are needed for the accurate prediction of assemblies, which undoubtedly will expand on the significant improvements in the tertiary structure prediction field.
Collapse
Affiliation(s)
- Burcu Ozden
- Izmir Biomedicine and Genome Center, 35340, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340, Izmir, Turkey
| | - Andriy Kryshtafovych
- Protein Structure Prediction Center, Genome and Biomedical Sciences Facilities, University of California, Davis, California, USA
| | - Ezgi Karaca
- Izmir Biomedicine and Genome Center, 35340, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340, Izmir, Turkey
| |
Collapse
|
15
|
Abstract
The biological significance of proteins attracted the scientific community in exploring their characteristics. The studies shed light on the interaction patterns and functions of proteins in a living body. Due to their practical difficulties, reliable experimental techniques pave the way for introducing computational methods in the interaction prediction. Automated methods reduced the difficulties but could not yet replace experimental studies as the field is still evolving. Interaction prediction problem being critical needs highly accurate results, but none of the existing methods could offer reliable performance that can parallel with experimental results yet. This article aims to assess the existing computational docking algorithms, their challenges, and future scope. Blind docking techniques are quite helpful when no information other than the individual structures are available. As more and more complex structures are being added to different databases, information-driven approaches can be a good alternative. Artificial intelligence, ruling over the major fields, is expected to take over this domain very shortly.
Collapse
|
16
|
Prévost C, Sacquin-Mora S. Moving pictures: Reassessing docking experiments with a dynamic view of protein interfaces. Proteins 2021; 89:1315-1323. [PMID: 34038009 DOI: 10.1002/prot.26152] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/22/2021] [Accepted: 05/19/2021] [Indexed: 11/06/2022]
Abstract
The modeling of protein assemblies at the atomic level remains a central issue in structural biology, as protein interactions play a key role in numerous cellular processes. This problem is traditionally addressed using docking tools, where the quality of the models is based on their similarity to a single reference experimental structure. However, using a static reference does not take into account the dynamic quality of the protein interface. Here, we used all-atom classical Molecular Dynamics simulations to investigate the stability of the reference interface for three complexes that previously served as targets in the CAPRI competition. For each one of these targets, we also ran MD simulations for ten models that are distributed over the High, Medium and Acceptable accuracy categories. To assess the quality of these models from a dynamic perspective, we set up new criteria which take into account the stability of the reference experimental protein interface. We show that, when the protein interfaces are allowed to evolve along time, the original ranking based on the static CAPRI criteria no longer holds as over 50% of the docking models undergo a category change (which can be either toward a better or a lower accuracy group) when reassessing their quality using dynamic information.
Collapse
Affiliation(s)
- Chantal Prévost
- CNRS, Laboratoire de Biochimie Théorique, UPR9080, Université de Paris, Paris, France.,Institut de Biologie Physico-Chimique, Fondation Edmond de Rothschild, PSL Research University, Paris, France
| | - Sophie Sacquin-Mora
- CNRS, Laboratoire de Biochimie Théorique, UPR9080, Université de Paris, Paris, France.,Institut de Biologie Physico-Chimique, Fondation Edmond de Rothschild, PSL Research University, Paris, France
| |
Collapse
|
17
|
Zhang W, Meng Q, Tang J, Guo F. Exploring effectiveness of ab-initio protein-protein docking methods on a novel antibacterial protein complex dataset. Brief Bioinform 2021; 22:6265196. [PMID: 33959764 DOI: 10.1093/bib/bbab150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/12/2021] [Accepted: 03/27/2021] [Indexed: 12/27/2022] Open
Abstract
Diseases caused by bacterial infections become a critical problem in public heath. Antibiotic, the traditional treatment, gradually loses their effectiveness due to the resistance. Meanwhile, antibacterial proteins attract more attention because of broad spectrum and little harm to host cells. Therefore, exploring new effective antibacterial proteins is urgent and necessary. In this paper, we are committed to evaluating the effectiveness of ab-initio docking methods in antibacterial protein-protein docking. For this purpose, we constructed a three-dimensional (3D) structure dataset of antibacterial protein complex, called APCset, which contained $19$ protein complexes whose receptors or ligands are homologous to antibacterial peptides from Antimicrobial Peptide Database. Then we selected five representative ab-initio protein-protein docking tools including ZDOCK3.0.2, FRODOCK3.0, ATTRACT, PatchDock and Rosetta to identify these complexes' structure, whose performance differences were obtained by analyzing from five aspects, including top/best pose, first hit, success rate, average hit count and running time. Finally, according to different requirements, we assessed and recommended relatively efficient protein-protein docking tools. In terms of computational efficiency and performance, ZDOCK was more suitable as preferred computational tool, with average running time of $6.144$ minutes, average Fnat of best pose of $0.953$ and average rank of best pose of $4.158$. Meanwhile, ZDOCK still yielded better performance on Benchmark 5.0, which proved ZDOCK was effective in performing docking on large-scale dataset. Our survey can offer insights into the research on the treatment of bacterial infections by utilizing the appropriate docking methods.
Collapse
Affiliation(s)
- Wei Zhang
- School of Computer Science and Technology, College of Intelligence and Computing, Tianjin University, Tianjin, China
| | - Qiaozhen Meng
- School of Computer Science and Technology, College of Intelligence and Computing, Tianjin University, Tianjin, China
| | - Jijun Tang
- School of Computer Science and Technology, College of Intelligence and Computing, Tianjin University, Tianjin, China.,School of Computational Science and Engineering, University of South Carolina, Columbia, U.S.,Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, China
| | - Fei Guo
- School of Computer Science and Technology, College of Intelligence and Computing, Tianjin University, Tianjin, China
| |
Collapse
|
18
|
Egbert M, Porter KA, Ghani U, Kotelnikov S, Nguyen T, Ashizawa R, Kozakov D, Vajda S. Conservation of binding properties in protein models. Comput Struct Biotechnol J 2021; 19:2549-2566. [PMID: 34025942 PMCID: PMC8114079 DOI: 10.1016/j.csbj.2021.04.048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 04/19/2021] [Accepted: 04/22/2021] [Indexed: 01/09/2023] Open
Abstract
We study the models submitted to round 12 of the Critical Assessment of protein Structure Prediction (CASP) experiment to assess how well the binding properties are conserved when the X-ray structures of the target proteins are replaced by their models. To explore small molecule binding we generate distributions of molecular probes - which are fragment-sized organic molecules of varying size, shape, and polarity - around the protein, and count the number of interactions between each residue and the probes, resulting in a vector of interactions we call a binding fingerprint. The similarity between two fingerprints, one for the X-ray structure and the other for a model of the protein, is determined by calculating the correlation coefficient between the two vectors. The resulting correlation coefficients are shown to correlate with global measures of accuracy established in CASP, and the relationship yields an accuracy threshold that has to be reached for meaningful binding surface conservation. The clusters formed by the probe molecules reliably predict binding hot spots and ligand binding sites in both X-ray structures and reasonably accurate models of the target, but ensembles of models may be needed for assessing the availability of proper binding pockets. We explored ligand docking to the few targets that had bound ligands in the X-ray structure. More targets were available to assess the ability of the models to reproduce protein-protein interactions by docking both the X-ray structures and models to their interaction partners in complexes. It was shown that this application is more difficult than finding small ligand binding sites, and the success rates heavily depend on the local structure in the potential interface. In particular, predicted conformations of flexible loops are frequently incorrect in otherwise highly accurate models, and may prevent predicting correct protein-protein interactions.
Collapse
Affiliation(s)
- Megan Egbert
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
| | - Kathryn A. Porter
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
| | - Usman Ghani
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
| | - Sergei Kotelnikov
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794, United States
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, United States
| | - Thu Nguyen
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, United States
| | - Ryota Ashizawa
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794, United States
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, United States
| | - Dima Kozakov
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794, United States
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, United States
| | - Sandor Vajda
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States
- Department of Chemistry, Boston University, Boston, MA 02215, United States
| |
Collapse
|
19
|
McCafferty CL, Marcotte EM, Taylor DW. Simplified geometric representations of protein structures identify complementary interaction interfaces. Proteins 2021; 89:348-360. [PMID: 33140424 PMCID: PMC7855953 DOI: 10.1002/prot.26020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/22/2020] [Accepted: 10/25/2020] [Indexed: 12/12/2022]
Abstract
Protein-protein interactions are critical to protein function, but three-dimensional (3D) arrangements of interacting proteins have proven hard to predict, even given the identities and 3D structures of the interacting partners. Specifically, identifying the relevant pairwise interaction surfaces remains difficult, often relying on shape complementarity with molecular docking while accounting for molecular motions to optimize rigid 3D translations and rotations. However, such approaches can be computationally expensive, and faster, less accurate approximations may prove useful for large-scale prediction and assembly of 3D structures of multi-protein complexes. We asked if a reduced representation of protein geometry retains enough information about molecular properties to predict pairwise protein interaction interfaces that are tolerant of limited structural rearrangements. Here, we describe a reduced representation of 3D protein accessible surfaces on which molecular properties such as charge, hydrophobicity, and evolutionary rate can be easily mapped, implemented in the MorphProt package. Pairs of surfaces are compared to rapidly assess partner-specific potential surface complementarity. On two available benchmarks of 185 overall known protein complexes, we observe predictions comparable to other structure-based tools at correctly identifying protein interaction surfaces. Furthermore, we examined the effect of molecular motion through normal mode simulation on a benchmark receptor-ligand pair and observed no marked loss of predictive accuracy for distortions of up to 6 Å Cα-RMSD. Thus, a shape reduction of protein surfaces retains considerable information about surface complementarity, offers enhanced speed of comparison relative to more complex geometric representations, and exhibits tolerance to conformational changes.
Collapse
Affiliation(s)
- Caitlyn L. McCafferty
- Department of Molecular BiosciencesUniversity of Texas at AustinAustinTexasUSA
- Center for Systems and Synthetic BiologyUniversity of Texas at AustinAustinTexasUSA
- Institute for Cellular and Molecular BiologyUniversity of Texas at AustinAustinTexasUSA
| | - Edward M. Marcotte
- Department of Molecular BiosciencesUniversity of Texas at AustinAustinTexasUSA
- Center for Systems and Synthetic BiologyUniversity of Texas at AustinAustinTexasUSA
- Institute for Cellular and Molecular BiologyUniversity of Texas at AustinAustinTexasUSA
| | - David W. Taylor
- Department of Molecular BiosciencesUniversity of Texas at AustinAustinTexasUSA
- Center for Systems and Synthetic BiologyUniversity of Texas at AustinAustinTexasUSA
- Institute for Cellular and Molecular BiologyUniversity of Texas at AustinAustinTexasUSA
- LIVESTRONG Cancer InstitutesDell Medical SchoolAustinTexasUSA
| |
Collapse
|
20
|
Slater O, Miller B, Kontoyianni M. Decoding Protein-protein Interactions: An Overview. Curr Top Med Chem 2021; 20:855-882. [PMID: 32101126 DOI: 10.2174/1568026620666200226105312] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 11/27/2019] [Accepted: 11/27/2019] [Indexed: 12/24/2022]
Abstract
Drug discovery has focused on the paradigm "one drug, one target" for a long time. However, small molecules can act at multiple macromolecular targets, which serves as the basis for drug repurposing. In an effort to expand the target space, and given advances in X-ray crystallography, protein-protein interactions have become an emerging focus area of drug discovery enterprises. Proteins interact with other biomolecules and it is this intricate network of interactions that determines the behavior of the system and its biological processes. In this review, we briefly discuss networks in disease, followed by computational methods for protein-protein complex prediction. Computational methodologies and techniques employed towards objectives such as protein-protein docking, protein-protein interactions, and interface predictions are described extensively. Docking aims at producing a complex between proteins, while interface predictions identify a subset of residues on one protein that could interact with a partner, and protein-protein interaction sites address whether two proteins interact. In addition, approaches to predict hot spots and binding sites are presented along with a representative example of our internal project on the chemokine CXC receptor 3 B-isoform and predictive modeling with IP10 and PF4.
Collapse
Affiliation(s)
- Olivia Slater
- Department of Pharmaceutical Sciences, Southern Illinois University, Edwardsville, IL 62026, United States
| | - Bethany Miller
- Department of Pharmaceutical Sciences, Southern Illinois University, Edwardsville, IL 62026, United States
| | - Maria Kontoyianni
- Department of Pharmaceutical Sciences, Southern Illinois University, Edwardsville, IL 62026, United States
| |
Collapse
|
21
|
Bux K, Hofer TS, Moin ST. Exploring interfacial dynamics in homodimeric S-ribosylhomocysteine lyase (LuxS) from Vibrio cholerae through molecular dynamics simulations. RSC Adv 2021; 11:1700-1714. [PMID: 35424088 PMCID: PMC8693604 DOI: 10.1039/d0ra08809a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/22/2020] [Indexed: 11/21/2022] Open
Abstract
To the best of our knowledge, this is the first molecular dynamics simulation study on the dimeric form of the LuxS enzyme from Vibrio cholerae to evaluate its structural and dynamical properties including the dynamics of the interface formed by the two monomeric chains of the enzyme. The dynamics of the interfacial region were investigated in terms of inter-residual contacts and the associated interface area of the enzyme in its ligand-free and ligand–bound states which produced characteristics contrast in the interfacial dynamics. Moreover, the binding patterns of the two inhibitors (RHC and KRI) to the enzyme forming two different enzyme–ligand complexes were analyzed which pointed towards a varying inhibition potential of the inhibitors as also revealed by the free energies of ligand binding. It is shown that KRI is a more potent inhibitor than RHC – a substrate analogue, showing correlation with experimental data. Moreover, the role of a loop in chain B of the enzyme was found to facilitate the binding of RHC similar to that of the substrate, while KRI demonstrates a differing binding pattern. The computation of the free energy of binding for the two ligands was also carried out via thermodynamic integration which ultimately served to correlate the dynamical properties with the inhibition potential of two different ligands against the enzyme. Furthermore, this successful study provides a rational to suggest novel LuxS inhibitors which could become promising candidates to treat the diseases caused by a broad variety of bacterial species. To the best of our knowledge, this is the first molecular dynamics simulation study on the dimeric form of the LuxS enzyme from Vibrio cholerae to evaluate its structural and dynamical properties including the dynamics of the interface formed by the two monomeric chains of the enzyme.![]()
Collapse
Affiliation(s)
- Khair Bux
- H.E.J. Research Institute of Chemistry
- International Center for Chemical and Biological Sciences
- University of Karachi
- Karachi-75270
- Pakistan
| | - Thomas S. Hofer
- Theoretical Chemistry Division
- Institute of General, Inorganic and Theoretical Chemistry
- University of Innsbruck
- A-6020 Innsbruck
- Austria
| | - Syed Tarique Moin
- H.E.J. Research Institute of Chemistry
- International Center for Chemical and Biological Sciences
- University of Karachi
- Karachi-75270
- Pakistan
| |
Collapse
|
22
|
Pestana-Nobles R, Leyva-Rojas JA, Yosa J. Searching Hit Potential Antimicrobials in Natural Compounds Space against Biofilm Formation. Molecules 2020; 25:E5334. [PMID: 33207596 PMCID: PMC7696173 DOI: 10.3390/molecules25225334] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/10/2020] [Accepted: 10/20/2020] [Indexed: 01/06/2023] Open
Abstract
Biofilms are communities of microorganisms that can colonize biotic and abiotic surfaces and thus play a significant role in the persistence of bacterial infection and resistance to antimicrobial. About 65% and 80% of microbial and chronic infections are associated with biofilm formation, respectively. The increase in infections by multi-resistant bacteria instigates the need for the discovery of novel natural-based drugs that act as inhibitory molecules. The inhibition of diguanylate cyclases (DGCs), the enzyme implicated in the synthesis of the second messenger, cyclic diguanylate (c-di-GMP), involved in the biofilm formation, represents a potential approach for preventing the biofilm development. It has been extensively studied using PleD protein as a model of DGC for in silico studies as virtual screening and as a model for in vitro studies in biofilms formation. This study aimed to search for natural products capable of inhibiting the Caulobacter crescentus enzyme PleD. For this purpose, 224,205 molecules from the natural products ZINC15 database, have been evaluated through molecular docking and molecular dynamic simulation. Our results suggest trans-Aconitic acid (TAA) as a possible starting point for hit-to-lead methodologies to obtain new inhibitors of the PleD protein and hence blocking the biofilm formation.
Collapse
|
23
|
Drummond ML, Henry A, Li H, Williams CI. Improved Accuracy for Modeling PROTAC-Mediated Ternary Complex Formation and Targeted Protein Degradation via New In Silico Methodologies. J Chem Inf Model 2020; 60:5234-5254. [PMID: 32969649 DOI: 10.1021/acs.jcim.0c00897] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Extending upon our previous publication [Drummond, M.; J. Chem. Inf. Model. 2019, 59, 1634-1644], two additional computational methods are presented to model PROTAC-mediated ternary complex structures, which are then used to predict the efficacy of any accompanying protein degradation. Method 4B, an extension to one of our previous approaches, incorporates a clustering procedure uniquely suited for considering ternary complexes. Method 4B yields the highest proportion to date of crystal-like poses in modeled ternary complex ensembles, nearing 100% in two cases and always giving a hit rate of at least 10%. Techniques to further improve this performance for particularly troublesome cases are suggested and validated. This demonstrated ability to reliably reproduce known crystallographic ternary complex structures is further established through modeling of a newly released crystal structure. Moreover, for the far more common scenario where the structure of the ternary complex intermediate is unknown, the methods detailed in this work nonetheless consistently yield results that reliably follow experimental protein degradation trends, as established through seven retrospective case studies. These various case studies cover challenging yet common modeling situations, such as when the precise orientation of the PROTAC binding moiety in one (or both) of the protein pockets has not been experimentally established. Successful results are presented for one PROTAC targeting many proteins, for different PROTACs targeting the same protein, and even for degradation effected by an E3 ligase that has not been structurally characterized in a ternary complex. Overall, the computational modeling approaches detailed in this work should greatly facilitate PROTAC screening and design efforts, so that the many advantages of a PROTAC-based degradation approach can be effectively utilized both rapidly and at reduced cost.
Collapse
Affiliation(s)
| | - Andrew Henry
- Chemical Computing Group, Cambridge, CB4 0WS, United Kingdom
| | - Huifang Li
- Chemical Computing Group, Montreal, Quebec H3A 2R7, Canada
| | | |
Collapse
|
24
|
Torchala M, Gerguri T, Chaleil RAG, Gordon P, Russell F, Keshani M, Bates PA. Enhanced sampling of protein conformational states for dynamic cross-docking within the protein-protein docking server SwarmDock. Proteins 2020; 88:962-972. [PMID: 31697436 PMCID: PMC7496321 DOI: 10.1002/prot.25851] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/02/2019] [Accepted: 11/03/2019] [Indexed: 12/12/2022]
Abstract
The formation of specific protein-protein interactions is often a key to a protein's function. During complex formation, each protein component will undergo a change in the conformational state, for some these changes are relatively small and reside primarily at the sidechain level; however, others may display notable backbone adjustments. One of the classic problems in the protein-docking field is to be able to a priori predict the extent of such conformational changes. In this work, we investigated three protocols to find the most suitable input structure conformations for cross-docking, including a robust sampling approach in normal mode space. Counterintuitively, knowledge of the theoretically best combination of normal modes for unbound-bound transitions does not always lead to the best results. We used a novel spatial partitioning library, Aether Engine (see Supplementary Materials), to efficiently search the conformational states of 56 receptor/ligand pairs, including a recent CAPRI target, in a systematic manner and selected diverse conformations as input to our automated docking server, SwarmDock, a server that allows moderate conformational adjustments during the docking process. In essence, here we present a dynamic cross-docking protocol, which when benchmarked against the simpler approach of just docking the unbound components shows a 10% uplift in the quality of the top docking pose.
Collapse
Affiliation(s)
- Mieczyslaw Torchala
- Biomolecular Modelling LaboratoryThe Francis Crick InstituteLondonUK
- Hadean Supercomputing LtdLondonUK
| | - Tereza Gerguri
- Biomolecular Modelling LaboratoryThe Francis Crick InstituteLondonUK
| | | | | | | | | | - Paul A. Bates
- Biomolecular Modelling LaboratoryThe Francis Crick InstituteLondonUK
| |
Collapse
|
25
|
Khramushin A, Marcu O, Alam N, Shimony O, Padhorny D, Brini E, Dill KA, Vajda S, Kozakov D, Schueler-Furman O. Modeling beta-sheet peptide-protein interactions: Rosetta FlexPepDock in CAPRI rounds 38-45. Proteins 2020; 88:1037-1049. [PMID: 31891416 PMCID: PMC7539656 DOI: 10.1002/prot.25871] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/17/2019] [Accepted: 12/26/2019] [Indexed: 01/09/2023]
Abstract
Peptide-protein docking is challenging due to the considerable conformational freedom of the peptide. CAPRI rounds 38-45 included two peptide-protein interactions, both characterized by a peptide forming an additional beta strand of a beta sheet in the receptor. Using the Rosetta FlexPepDock peptide docking protocol we generated top-performing, high-accuracy models for targets 134 and 135, involving an interaction between a peptide derived from L-MAG with DLC8. In addition, we were able to generate the only medium-accuracy models for a particularly challenging target, T121. In contrast to the classical peptide-mediated interaction, in which receptor side chains contact both peptide backbone and side chains, beta-sheet complementation involves a major contribution to binding by hydrogen bonds between main chain atoms. To establish how binding affinity and specificity are established in this special class of peptide-protein interactions, we extracted PeptiDBeta, a benchmark of solved structures of different protein domains that are bound by peptides via beta-sheet complementation, and tested our protocol for global peptide-docking PIPER-FlexPepDock on this dataset. We find that the beta-strand part of the peptide is sufficient to generate approximate and even high resolution models of many interactions, but inclusion of adjacent motif residues often provides additional information necessary to achieve high resolution model quality.
Collapse
Affiliation(s)
- Alisa Khramushin
- Department of Microbiologyand Molecular Genetics, Institute
for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University,
Jerusalem, Israel
| | - Orly Marcu
- Department of Microbiologyand Molecular Genetics, Institute
for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University,
Jerusalem, Israel
| | - Nawsad Alam
- Department of Microbiologyand Molecular Genetics, Institute
for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University,
Jerusalem, Israel
| | - Orly Shimony
- Department of Microbiologyand Molecular Genetics, Institute
for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University,
Jerusalem, Israel
| | - Dzmitry Padhorny
- Department of Applied Mathematics and Statistics, Stony
Brook University, New York, New York
- Laufer Center for Physical and Quantitative Biology, Stony
Brook University, New York, New York
| | - Emiliano Brini
- Laufer Center for Physical and Quantitative Biology, Stony
Brook University, New York, New York
| | - Ken A. Dill
- Laufer Center for Physical and Quantitative Biology, Stony
Brook University, New York, New York
- Department of Physics and Astronomy, Stony Brook
University, New York, New York
- Department of Chemistry, Stony Brook University, New York,
New York
| | - Sandor Vajda
- Department of Biomedical Engineering, Boston University,
Boston, Massachusetts
- Department of Chemistry, Boston University, Boston,
Massachusetts
| | - Dima Kozakov
- Department of Applied Mathematics and Statistics, Stony
Brook University, New York, New York
- Laufer Center for Physical and Quantitative Biology, Stony
Brook University, New York, New York
| | - Ora Schueler-Furman
- Department of Microbiologyand Molecular Genetics, Institute
for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University,
Jerusalem, Israel
| |
Collapse
|
26
|
Townsend DJ, Mala B, Hughes E, Hussain R, Siligardi G, Fullwood NJ, Middleton DA. Circular Dichroism Spectroscopy Identifies the β-Adrenoceptor Agonist Salbutamol As a Direct Inhibitor of Tau Filament Formation in Vitro. ACS Chem Neurosci 2020; 11:2104-2116. [PMID: 32520518 DOI: 10.1021/acschemneuro.0c00154] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Potential drug treatments for Alzheimer's disease (AD) may be found by identifying compounds that block the assembly of the microtubule-associated protein tau into neurofibrillar tangles associated with neuron destabilization and cell death. Here, a small library of structurally diverse compounds was screened in vitro for the ability to inhibit tau aggregation, using high-throughput synchrotron radiation circular dichroism as a novel tool to monitor the structural changes in the protein as it assembles into filaments. The catecholamine epinephrine was found to be the most effective tau aggregation inhibitor of all 88 screened compounds. Subsequently, we tested chemically similar phenolamine drugs from the β-adrenergic receptor agonist class, using conventional circular dichroism spectroscopy, thioflavin T fluorescence, and transmission electron microscopy. Two compounds, salbutamol and dobutamine, used widely in the treatment of respiratory and cardiovascular disease, impede the aggregation of tau in vitro. Dobutamine reduces both the rate and yield of tau filament formation over 24 h; however, it has little effect on the structural transition of tau into β-sheet structures over 24 h. Salbutamol also reduces the yield and rate of filament formation and additionally inhibits tau's structural change into β-sheet-rich aggregates. Salbutamol has a good safety profile and a half-life that facilitates permeation through the blood-brain barrier and could represent an expediated approach to developing AD therapeutics. These results provide the motivation for the in vivo evaluation of pre-existing β-adrenergic receptor agonists as a potential therapy for AD through the reduction of tau deposition.
Collapse
Affiliation(s)
- David J Townsend
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB, United Kingdom
| | - Barbora Mala
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB, United Kingdom
| | - Eleri Hughes
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB, United Kingdom
| | - Rohanah Hussain
- Diamond House, Harwell Science & Innovation Campus, Diamond Light Source Ltd., Didcot OX11 ODE, United Kingdom
| | - Giuliano Siligardi
- Diamond House, Harwell Science & Innovation Campus, Diamond Light Source Ltd., Didcot OX11 ODE, United Kingdom
| | - Nigel J. Fullwood
- Biomedical and Life Sciences, Lancaster University, Lancaster LA1 4YG, United Kingdom
| | - David A. Middleton
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB, United Kingdom
| |
Collapse
|
27
|
Desta IT, Porter KA, Xia B, Kozakov D, Vajda S. Performance and Its Limits in Rigid Body Protein-Protein Docking. Structure 2020; 28:1071-1081.e3. [PMID: 32649857 DOI: 10.1016/j.str.2020.06.006] [Citation(s) in RCA: 354] [Impact Index Per Article: 88.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/19/2020] [Accepted: 06/19/2020] [Indexed: 12/13/2022]
Abstract
The development of fast Fourier transform (FFT) algorithms enabled the sampling of billions of complex conformations and thus revolutionized protein-protein docking. FFT-based methods are now widely available and have been used in hundreds of thousands of docking calculations. Although the methods perform "soft" docking, which allows for some overlap of component proteins, the rigid body assumption clearly introduces limitations on accuracy and reliability. In addition, the method can work only with energy expressions represented by sums of correlation functions. In this paper we use a well-established protein-protein docking benchmark set to evaluate the results of these limitations by focusing on the performance of the docking server ClusPro, which implements one of the best rigid body methods. Furthermore, we explore the theoretical limits of accuracy when using established energy terms for scoring, provide comparison with flexible docking algorithms, and review the historical performance of servers in the CAPRI docking experiment.
Collapse
Affiliation(s)
- Israel T Desta
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Kathryn A Porter
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Bing Xia
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Dima Kozakov
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Sandor Vajda
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| |
Collapse
|
28
|
Cao Y, Shen Y. Bayesian Active Learning for Optimization and Uncertainty Quantification in Protein Docking. J Chem Theory Comput 2020; 16:5334-5347. [PMID: 32558561 DOI: 10.1021/acs.jctc.0c00476] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ab initio protein docking represents a major challenge for optimizing a noisy and costly "black box"-like function in a high-dimensional space. Despite progress in this field, there is a lack of rigorous uncertainty quantification (UQ). To fill the gap, we introduce a novel algorithm, Bayesian active learning (BAL), for optimization and UQ of such black-box functions with applications to flexible protein docking. BAL directly models the posterior distribution of the global optimum (i.e., native structures) with active sampling and posterior estimation iteratively feeding each other. Furthermore, it uses complex normal modes to span a homogeneous, Euclidean conformation space suitable for high-dimensional optimization and constructs funnel-like energy models for quality estimation of encounter complexes. Over a protein-docking benchmark set and a CAPRI set including homology docking, we establish that BAL significantly improves against starting points from rigid docking and refinements by particle swarm optimization, providing a top-3 near-native prediction for one third targets. Quality assessment empowered with UQ leads to tight quality intervals with half range around 25% of the actual interface root-mean-square deviation and confidence level at 85%. BAL's estimated probability of a prediction being near-native achieves binary classification AUROC at 0.93 and area under the precision recall curve over 0.60 (compared to 0.50 and 0.14, respectively, by chance), which also improves ranking predictions. This study represents the first UQ solution for protein docking, with rigorous theoretical frameworks and comprehensive empirical assessments.
Collapse
Affiliation(s)
- Yue Cao
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, Texas 77843, United States
| | - Yang Shen
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, Texas 77843, United States.,TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering, Texas A&M University, College Station, Texas 77840, United States
| |
Collapse
|
29
|
He J, Tao H, Huang SY. Protein-ensemble-RNA docking by efficient consideration of protein flexibility through homology models. Bioinformatics 2020; 35:4994-5002. [PMID: 31086984 DOI: 10.1093/bioinformatics/btz388] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 04/28/2019] [Accepted: 05/03/2019] [Indexed: 12/18/2022] Open
Abstract
MOTIVATION Given the importance of protein-ribonucleic acid (RNA) interactions in many biological processes, a variety of docking algorithms have been developed to predict the complex structure from individual protein and RNA partners in the past decade. However, due to the impact of molecular flexibility, the performance of current methods has hit a bottleneck in realistic unbound docking. Pushing the limit, we have proposed a protein-ensemble-RNA docking strategy to explicitly consider the protein flexibility in protein-RNA docking through an ensemble of multiple protein structures, which is referred to as MPRDock. Instead of taking conformations from MD simulations or experimental structures, we obtained the multiple structures of a protein by building models from its homologous templates in the Protein Data Bank (PDB). RESULTS Our approach can not only avoid the reliability issue of structures from MD simulations but also circumvent the limited number of experimental structures for a target protein in the PDB. Tested on 68 unbound-bound and 18 unbound-unbound protein-RNA complexes, our MPRDock/DITScorePR considerably improved the docking performance and achieved a significantly higher success rate than single-protein rigid docking whether pseudo-unbound templates are included or not. Similar improvements were also observed when combining our ensemble docking strategy with other scoring functions. The present homology model-based ensemble docking approach will have a general application in molecular docking for other interactions. AVAILABILITY AND IMPLEMENTATION http://huanglab.phys.hust.edu.cn/mprdock/. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Jiahua He
- Institute of Biophysics, School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huanyu Tao
- Institute of Biophysics, School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Sheng-You Huang
- Institute of Biophysics, School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
30
|
Duan R, Qiu L, Xu X, Ma Z, Merideth BR, Shyu CR, Zou X. Performance of human and server prediction in CAPRI rounds 38-45. Proteins 2020; 88:1110-1120. [PMID: 32483825 DOI: 10.1002/prot.25956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/26/2020] [Accepted: 05/27/2020] [Indexed: 11/11/2022]
Abstract
CAPRI challenges offer a variety of blind tests for protein-protein interaction prediction. In CAPRI Rounds 38-45, we generated a set of putative binding modes for each target with an FFT-based docking algorithm, and then scored and ranked these binding modes with a proprietary scoring function, ITScorePP. We have also developed a novel web server, Rebipp. The algorithm utilizes information retrieval to identify relevant biological information to significantly reduce the search space for a particular protein. In parallel, we have also constructed a GPU-based docking server, MDockPP, for protein-protein complex structure prediction. Here, the performance of our protocol in CAPRI rounds 38-45 is reported, which include 16 docking and scoring targets. Among them, three targets contain multiple interfaces: Targets 124, 125, and 136 have 2, 4, and 3 interfaces, respectively. In the predictor experiments, we predicted correct binding modes for nine targets, including one high-accuracy interface, six medium-accuracy binding modes, and six acceptable-accuracy binding modes. For the docking server prediction experiments, we predicted correct binding modes for eight targets, including one high-accuracy, three medium-accuracy, and five acceptable-accuracy binding modes.
Collapse
Affiliation(s)
- Rui Duan
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Liming Qiu
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Xianjin Xu
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Zhiwei Ma
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA.,Department of Physics and Astronomy, University of Missouri, Columbia, Missouri, USA
| | - Benjamin Ryan Merideth
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA.,Institute for Data Science and Informatics, University of Missouri, Columbia, Missouri, USA
| | - Chi-Ren Shyu
- Institute for Data Science and Informatics, University of Missouri, Columbia, Missouri, USA.,Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, Missouri, USA
| | - Xiaoqin Zou
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA.,Department of Physics and Astronomy, University of Missouri, Columbia, Missouri, USA.,Institute for Data Science and Informatics, University of Missouri, Columbia, Missouri, USA.,Department of Biochemistry, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
31
|
Koukos P, Bonvin A. Integrative Modelling of Biomolecular Complexes. J Mol Biol 2020; 432:2861-2881. [DOI: 10.1016/j.jmb.2019.11.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 12/31/2022]
|
32
|
Yan Y, He J, Feng Y, Lin P, Tao H, Huang SY. Challenges and opportunities of automated protein-protein docking: HDOCK server vs human predictions in CAPRI Rounds 38-46. Proteins 2020; 88:1055-1069. [PMID: 31994779 DOI: 10.1002/prot.25874] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 01/02/2020] [Accepted: 01/22/2020] [Indexed: 12/12/2022]
Abstract
Protein-protein docking plays an important role in the computational prediction of the complex structure between two proteins. For years, a variety of docking algorithms have been developed, as witnessed by the critical assessment of prediction interactions (CAPRI) experiments. However, despite their successes, many docking algorithms often require a series of manual operations like modeling structures from sequences, incorporating biological information, and selecting final models. The difficulties in these manual steps have significantly limited the applications of protein-protein docking, as most of the users in the community are nonexperts in docking. Therefore, automated docking like a web server, which can give a comparable performance to human docking protocol, is pressingly needed. As such, we have participated in the blind CAPRI experiments for Rounds 38-45 and CASP13-CAPRI challenge for Round 46 with both our HDOCK automated docking web server and human docking protocol. It was shown that our HDOCK server achieved an "acceptable" or higher CAPRI-rated model in the top 10 submitted predictions for 65.5% and 59.1% of the targets in the docking experiments of CAPRI and CASP13-CAPRI, respectively, which are comparable to 66.7% and 54.5% for human docking protocol. Similar trends can also be observed in the scoring experiments. These results validated our HDOCK server as an efficient automated docking protocol for nonexpert users. Challenges and opportunities of automated docking are also discussed.
Collapse
Affiliation(s)
- Yumeng Yan
- School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Jiahua He
- School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yuyu Feng
- School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Peicong Lin
- School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Huanyu Tao
- School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Sheng-You Huang
- School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
33
|
Lensink MF, Nadzirin N, Velankar S, Wodak SJ. Modeling protein‐protein, protein‐peptide, and protein‐oligosaccharide complexes: CAPRI 7th edition. Proteins 2020; 88:916-938. [DOI: 10.1002/prot.25870] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/19/2019] [Accepted: 12/26/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Marc F. Lensink
- University of Lille, CNRS UMR8576 UGSF, Unité de Glycobiologie Structurale et Fonctionnelle F‐59000 Lille France
| | - Nurul Nadzirin
- European Molecular Biology LaboratoryEuropean Bioinformatics Institute (EMBL‐EBI), Wellcome Trust Genome Campus Cambridge UK
| | - Sameer Velankar
- European Molecular Biology LaboratoryEuropean Bioinformatics Institute (EMBL‐EBI), Wellcome Trust Genome Campus Cambridge UK
| | | |
Collapse
|
34
|
Koukos PI, Roel-Touris J, Ambrosetti F, Geng C, Schaarschmidt J, Trellet ME, Melquiond ASJ, Xue LC, Honorato RV, Moreira I, Kurkcuoglu Z, Vangone A, Bonvin AMJJ. An overview of data-driven HADDOCK strategies in CAPRI rounds 38-45. Proteins 2019; 88:1029-1036. [PMID: 31886559 DOI: 10.1002/prot.25869] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 12/17/2019] [Accepted: 12/26/2019] [Indexed: 01/18/2023]
Abstract
Our information-driven docking approach HADDOCK has demonstrated a sustained performance since the start of its participation to CAPRI. This is due, in part, to its ability to integrate data into the modeling process, and to the robustness of its scoring function. We participated in CAPRI both as server and manual predictors. In CAPRI rounds 38-45, we have used various strategies depending on the available information. These ranged from imposing restraints to a few residues identified from literature as being important for the interaction, to binding pockets identified from homologous complexes or template-based refinement/CA-CA restraint-guided docking from identified templates. When relevant, symmetry restraints were used to limit the conformational sampling. We also tested for a large decamer target a new implementation of the MARTINI coarse-grained force field in HADDOCK. Overall, we obtained acceptable or better predictions for 13 and 11 server and manual submissions, respectively, out of the 22 interfaces. Our server performance (acceptable or higher-quality models when considering the top 10) was better (59%) than the manual (50%) one, in which we typically experiment with various combinations of protocols and data sources. Again, our simple scoring function based on a linear combination of intermolecular van der Waals and electrostatic energies and an empirical desolvation term demonstrated a good performance in the scoring experiment with a 63% success rate across all 22 interfaces. An analysis of model quality indicates that, while we are consistently performing well in generating acceptable models, there is room for improvement for generating/identifying higher quality models.
Collapse
Affiliation(s)
- Panagiotis I Koukos
- Faculty of Science, Department of Chemistry, Bijvoet Center for Biomolecular Research, Computational Structural Biology Group, Utrecht University, Utrecht, The Netherlands
| | - Jorge Roel-Touris
- Faculty of Science, Department of Chemistry, Bijvoet Center for Biomolecular Research, Computational Structural Biology Group, Utrecht University, Utrecht, The Netherlands
| | - Francesco Ambrosetti
- Faculty of Science, Department of Chemistry, Bijvoet Center for Biomolecular Research, Computational Structural Biology Group, Utrecht University, Utrecht, The Netherlands.,Department of Physics, Sapienza University, Rome, Italy
| | - Cunliang Geng
- Faculty of Science, Department of Chemistry, Bijvoet Center for Biomolecular Research, Computational Structural Biology Group, Utrecht University, Utrecht, The Netherlands
| | - Jörg Schaarschmidt
- Faculty of Science, Department of Chemistry, Bijvoet Center for Biomolecular Research, Computational Structural Biology Group, Utrecht University, Utrecht, The Netherlands.,Multiscale Materials Modelling and Virtual Design, Institute of Nanotechnology, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Mikael E Trellet
- Faculty of Science, Department of Chemistry, Bijvoet Center for Biomolecular Research, Computational Structural Biology Group, Utrecht University, Utrecht, The Netherlands
| | - Adrien S J Melquiond
- Faculty of Science, Department of Chemistry, Bijvoet Center for Biomolecular Research, Computational Structural Biology Group, Utrecht University, Utrecht, The Netherlands
| | - Li C Xue
- Faculty of Science, Department of Chemistry, Bijvoet Center for Biomolecular Research, Computational Structural Biology Group, Utrecht University, Utrecht, The Netherlands
| | - Rodrigo V Honorato
- Faculty of Science, Department of Chemistry, Bijvoet Center for Biomolecular Research, Computational Structural Biology Group, Utrecht University, Utrecht, The Netherlands
| | - Irina Moreira
- Faculty of Science, Department of Chemistry, Bijvoet Center for Biomolecular Research, Computational Structural Biology Group, Utrecht University, Utrecht, The Netherlands.,CNC-Center for Neuroscience and Cell Biology, Rua Larga, FMUC, Polo I, 1° andar, Universidade de Coimbra, Coimbra, Portugal
| | - Zeynep Kurkcuoglu
- Faculty of Science, Department of Chemistry, Bijvoet Center for Biomolecular Research, Computational Structural Biology Group, Utrecht University, Utrecht, The Netherlands
| | - Anna Vangone
- Faculty of Science, Department of Chemistry, Bijvoet Center for Biomolecular Research, Computational Structural Biology Group, Utrecht University, Utrecht, The Netherlands
| | - Alexandre M J J Bonvin
- Faculty of Science, Department of Chemistry, Bijvoet Center for Biomolecular Research, Computational Structural Biology Group, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
35
|
Abstract
Background Protein-protein docking is a valuable computational approach for investigating protein-protein interactions. Shape complementarity is the most basic component of a scoring function and plays an important role in protein-protein docking. Despite significant progresses, shape representation remains an open question in the development of protein-protein docking algorithms, especially for grid-based docking approaches. Results We have proposed a new pairwise shape-based scoring function (LSC) for protein-protein docking which adopts an exponential form to take into account long-range interactions between protein atoms. The LSC scoring function was incorporated into our FFT-based docking program and evaluated for both bound and unbound docking on the protein docking benchmark 4.0. It was shown that our LSC achieved a significantly better performance than four other similar docking methods, ZDOCK 2.1, MolFit/G, GRAMM, and FTDock/G, in both success rate and number of hits. When considering the top 10 predictions, LSC obtained a success rate of 51.71% and 6.82% for bound and unbound docking, respectively, compared to 42.61% and 4.55% for the second-best program ZDOCK 2.1. LSC also yielded an average of 8.38 and 3.94 hits per complex in the top 1000 predictions for bound and unbound docking, respectively, followed by 6.38 and 2.96 hits for the second-best ZDOCK 2.1. Conclusions The present LSC method will not only provide an initial-stage docking approach for post-docking processes but also have a general implementation for accurate representation of other energy terms on grids in protein-protein docking. The software has been implemented in our HDOCK web server at http://hdock.phys.hust.edu.cn/.
Collapse
|
36
|
Agrawal P, Singh H, Srivastava HK, Singh S, Kishore G, Raghava GPS. Benchmarking of different molecular docking methods for protein-peptide docking. BMC Bioinformatics 2019; 19:426. [PMID: 30717654 PMCID: PMC7394329 DOI: 10.1186/s12859-018-2449-y] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 10/29/2018] [Indexed: 11/10/2022] Open
Abstract
Background Molecular docking studies on protein-peptide interactions are a challenging and time-consuming task because peptides are generally more flexible than proteins and tend to adopt numerous conformations. There are several benchmarking studies on protein-protein, protein-ligand and nucleic acid-ligand docking interactions. However, a series of docking methods is not rigorously validated for protein-peptide complexes in the literature. Considering the importance and wide application of peptide docking, we describe benchmarking of 6 docking methods on 133 protein-peptide complexes having peptide length between 9 to 15 residues. The performance of docking methods was evaluated using CAPRI parameters like FNAT, I-RMSD, L-RMSD. Result Firstly, we performed blind docking and evaluate the performance of the top docking pose of each method. It was observed that FRODOCK performed better than other methods with average L-RMSD of 12.46 Å. The performance of all methods improved significantly for their best docking pose and achieved highest average L-RMSD of 3.72 Å in case of FRODOCK. Similarly, we performed re-docking and evaluated the performance of the top and best docking pose of each method. We achieved the best performance in case of ZDOCK with average L-RMSD 8.60 Å and 2.88 Å for the top and best docking pose respectively. Methods were also evaluated on 40 protein-peptide complexes used in the previous benchmarking study, where peptide have length up to 5 residues. In case of best docking pose, we achieved the highest average L-RMSD of 4.45 Å and 2.09 Å for the blind docking using FRODOCK and re-docking using AutoDock Vina respectively. Conclusion The study shows that FRODOCK performed best in case of blind docking and ZDOCK in case of re-docking. There is a need to improve the ranking of docking pose generated by different methods, as the present ranking scheme is not satisfactory. To facilitate the scientific community for calculating CAPRI parameters between native and docked complexes, we developed a web-based service named PPDbench (http://webs.iiitd.edu.in/raghava/ppdbench/). Electronic supplementary material The online version of this article (10.1186/s12859-018-2449-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Piyush Agrawal
- Center for Computation Biology, Indraprastha Institute of Information Technology, Okhla Phase III, New Delhi, 110020, India.,CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, India
| | - Harinder Singh
- CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, India
| | | | - Sandeep Singh
- CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, India
| | - Gaurav Kishore
- CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, India
| | - Gajendra P S Raghava
- Center for Computation Biology, Indraprastha Institute of Information Technology, Okhla Phase III, New Delhi, 110020, India. .,CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, India.
| |
Collapse
|
37
|
Pfeiffenberger E, Bates PA. Refinement of protein-protein complexes in contact map space with metadynamics simulations. Proteins 2019; 87:12-22. [PMID: 30370948 PMCID: PMC6492248 DOI: 10.1002/prot.25612] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 09/21/2018] [Accepted: 09/26/2018] [Indexed: 12/18/2022]
Abstract
Accurate protein-protein complex prediction, to atomic detail, is a challenging problem. For flexible docking cases, current state-of-the-art docking methods are limited in their ability to exhaustively search the high dimensionality of the problem space. In this study, to obtain more accurate models, an investigation into the local optimization of initial docked solutions is presented with respect to a reference crystal structure. We show how physics-based refinement of protein-protein complexes in contact map space (CMS), within a metadynamics protocol, can be performed. The method uses 5 times replicated 10 ns simulations for sampling and ranks the generated conformational snapshots with ZRANK to identify an ensemble of n snapshots for final model building. Furthermore, we investigated whether the reconstructed free energy surface (FES), or a combination of both FES and ZRANK, referred to as CSα , can help to reduce snapshot ranking error.
Collapse
Affiliation(s)
- Erik Pfeiffenberger
- Biomolecular Modelling LaboratoryThe Francis Crick InstituteLondonUnited Kingdom
| | - Paul A. Bates
- Biomolecular Modelling LaboratoryThe Francis Crick InstituteLondonUnited Kingdom
| |
Collapse
|
38
|
Bourquard T, Musnier A, Puard V, Tahir S, Ayoub MA, Jullian Y, Boulo T, Gallay N, Watier H, Bruneau G, Reiter E, Crépieux P, Poupon A. MAbTope: A Method for Improved Epitope Mapping. THE JOURNAL OF IMMUNOLOGY 2018; 201:3096-3105. [PMID: 30322966 DOI: 10.4049/jimmunol.1701722] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 09/13/2018] [Indexed: 11/19/2022]
Abstract
Abs are very efficient drugs, ∼70 of them are already approved for medical use, over 500 are in clinical development, and many more are in preclinical development. One important step in the characterization and protection of a therapeutic Ab is the determination of its cognate epitope. The gold standard is the three-dimensional structure of the Ab/Ag complex by crystallography or nuclear magnetic resonance spectroscopy. However, it remains a tedious task, and its outcome is uncertain. We have developed MAbTope, a docking-based prediction method of the epitope associated with straightforward experimental validation procedures. We show that MAbTope predicts the correct epitope for each of 129 tested examples of Ab/Ag complexes of known structure. We further validated this method through the successful determination, and experimental validation (using human embryonic kidney cells 293), of the epitopes recognized by two therapeutic Abs targeting TNF-α: certolizumab and golimumab.
Collapse
Affiliation(s)
- Thomas Bourquard
- Unité de Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, Université François Rabelais-Tours, CNRS, 37380 Nouzilly, France.,Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX 77030
| | - Astrid Musnier
- Unité de Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, Université François Rabelais-Tours, CNRS, 37380 Nouzilly, France.,MAbSilico Société par Actions Simplifiée, Domaine de l'Orfrasière, 37380 Nouzilly, France
| | - Vincent Puard
- MAbSilico Société par Actions Simplifiée, Domaine de l'Orfrasière, 37380 Nouzilly, France
| | - Shifa Tahir
- Unité de Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, Université François Rabelais-Tours, CNRS, 37380 Nouzilly, France
| | - Mohammed Akli Ayoub
- Unité de Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, Université François Rabelais-Tours, CNRS, 37380 Nouzilly, France.,Biology Department, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Yann Jullian
- Calcul Scientifique et Modélisation Orléans Tours, l'Unité de Formation et de Recherche Sciences et Techniques, Université François-Rabelais, 37041 Tours, France; and
| | - Thomas Boulo
- Unité de Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, Université François Rabelais-Tours, CNRS, 37380 Nouzilly, France
| | - Nathalie Gallay
- Unité de Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, Université François Rabelais-Tours, CNRS, 37380 Nouzilly, France.,Centre Hospitalier Régional Universitaire de Tours, Université François-Rabelais de Tours, CNRS, UMR 7292, 37041 Tours, France
| | - Hervé Watier
- Centre Hospitalier Régional Universitaire de Tours, Université François-Rabelais de Tours, CNRS, UMR 7292, 37041 Tours, France
| | - Gilles Bruneau
- Unité de Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, Université François Rabelais-Tours, CNRS, 37380 Nouzilly, France
| | - Eric Reiter
- Unité de Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, Université François Rabelais-Tours, CNRS, 37380 Nouzilly, France
| | - Pascale Crépieux
- Unité de Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, Université François Rabelais-Tours, CNRS, 37380 Nouzilly, France
| | - Anne Poupon
- Unité de Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, Université François Rabelais-Tours, CNRS, 37380 Nouzilly, France;
| |
Collapse
|
39
|
Hogues H, Gaudreault F, Corbeil CR, Deprez C, Sulea T, Purisima EO. ProPOSE: Direct Exhaustive Protein-Protein Docking with Side Chain Flexibility. J Chem Theory Comput 2018; 14:4938-4947. [PMID: 30107730 DOI: 10.1021/acs.jctc.8b00225] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Despite decades of development, protein-protein docking remains a largely unsolved problem. The main difficulties are the immense space spanned by the translational and rotational degrees of freedom and the prediction of the conformational changes of proteins upon binding. FFT is generally the preferred method to exhaustively explore the translation-rotation space at a fine grid resolution, albeit with the trade-off of approximating force fields with correlation functions. This work presents a direct search alternative that samples the states in Cartesian space at the same resolution and computational cost as standard FFT methods. Operating in real space allows the use of standard force field functional forms used in typical non-FFT methods as well as the implementation of strategies for focused exploration of conformational flexibility. Currently, a few misplaced side chains can cause docking programs to fail. This work specifically addresses the problem of side chain rearrangements upon complex formation. Based on the observation that most side chains retain their unbound conformation upon binding, each rigidly docked pose is initially scored ignoring up to a limited number of side chain overlaps which are resolved in subsequent repacking and minimization steps. On test systems where side chains are altered and backbones held in their bound state, this implementation provides significantly better native pose recovery and higher quality (lower RMSD) predictions when compared with five of the most popular docking programs. The method is implemented in the software program ProPOSE (Protein Pose Optimization by Systematic Enumeration).
Collapse
Affiliation(s)
- Hervé Hogues
- Human Health Therapeutics , National Research Council Canada , 6100 Royalmount Avenue , Montreal , Quebec H4P 2R2 , Canada
| | - Francis Gaudreault
- Human Health Therapeutics , National Research Council Canada , 6100 Royalmount Avenue , Montreal , Quebec H4P 2R2 , Canada
| | - Christopher R Corbeil
- Human Health Therapeutics , National Research Council Canada , 6100 Royalmount Avenue , Montreal , Quebec H4P 2R2 , Canada
| | - Christophe Deprez
- Human Health Therapeutics , National Research Council Canada , 6100 Royalmount Avenue , Montreal , Quebec H4P 2R2 , Canada
| | - Traian Sulea
- Human Health Therapeutics , National Research Council Canada , 6100 Royalmount Avenue , Montreal , Quebec H4P 2R2 , Canada
| | - Enrico O Purisima
- Human Health Therapeutics , National Research Council Canada , 6100 Royalmount Avenue , Montreal , Quebec H4P 2R2 , Canada
| |
Collapse
|
40
|
Abstract
Protein-RNA interactions play an important role in many biological processes. Computational methods such as docking have been developed to complement existing biophysical and structural biology techniques. Computational prediction of protein-RNA complex structures includes two steps: generating candidate structures from the individual protein and RNA parts and scoring the generated poses to pick out the correct one. In this work, we considered three recently developed data sets of protein-RNA complexes to evaluate and improve the performance of the FFT-based rigid-body docking algorithm implemented in the ICM package. An electrostatic term describing interactions between negatively charged phosphate groups and positively charged protein residues was added to the energy function used during the docking step to take into account the greater role that electrostatic interactions play in protein-RNA complexes. Next, the docking results were used to optimize a scoring function including van der Waals, electrostatic, and solvation terms. This optimization yielded a much smaller weight for the solvation term indicating that solvation energy may be less important for the scoring of protein-RNA structures. Rescoring of the generated poses with the new scoring function led to much higher success rates, while pose clustering by contact fingerprints produced further improvements, achieving a success rate of 0.66 for the top 100 structures.
Collapse
Affiliation(s)
- Yelena A Arnautova
- Molsoft L.L.C., 11199 Sorrento Valley Road, S209 , San Diego , California 92121 , United States
| | - Ruben Abagyan
- Skaggs School of Pharmacy and Pharmaceutical Sciences , University of California San Diego , La Jolla , California 92093 , United States
| | - Maxim Totrov
- Molsoft L.L.C., 11199 Sorrento Valley Road, S209 , San Diego , California 92121 , United States
| |
Collapse
|
41
|
Development of a new benchmark for assessing the scoring functions applicable to protein–protein interactions. Future Med Chem 2018; 10:1555-1574. [DOI: 10.4155/fmc-2017-0261] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aim: Scoring functions are important component of protein–protein docking methods. They need to be evaluated on high-quality benchmarks to reveal their strengths and weaknesses. Evaluation results obtained on such benchmarks can provide valuable guidance for developing more advanced scoring functions. Methodology & results: In our comparative assessment of scoring functions for protein–protein interactions benchmark, the performance of a scoring function was characterized by ‘docking power’ and ‘scoring power’. A high-quality dataset of 273 protein–protein complexes was compiled and employed in both tests. Four scoring functions, including FASTCONTACT, ZRANK, dDFIRE and ATTRACT were tested as demonstration. ZRANK and ATTRACT exhibited encouraging performance in the docking power test. However, all four scoring functions failed badly in the scoring power test. Conclusion: Our comparative assessment of scoring functions for protein–protein interaction benchmark is created especially for assessing the scoring functions applicable to protein–protein interactions. It is different from other benchmarks for assessing protein–protein docking methods. Our benchmark is available to the public at www.pdbbind-cn.org/download/CASF-PPI/ .
Collapse
|
42
|
Rani A, Dhillon A, Sharma K, Goyal A. Insights into the structural characteristics and substrate binding analysis of chondroitin AC lyase (PsPL8A) from Pedobacter saltans. Int J Biol Macromol 2018; 109:980-991. [PMID: 29155196 DOI: 10.1016/j.ijbiomac.2017.11.087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 11/12/2017] [Accepted: 11/13/2017] [Indexed: 10/18/2022]
Abstract
The structure of chondroitin AC lyase (PsPL8A) of family 8 polysaccharide lyase was characterized. Modeled PsPL8A structure showed, it contains N-terminal (α/α)6 incomplete toroidal fold and a layered β sandwich structure at C-terminal. Ramchandran plot displayed 98.5% residues in favoured and 1.2% in generously allowed region. Secondary structure of PsPL8A by CD revealed 27.31% α helices 22.7% β sheets and 49.9% random coils. Protein melting study showed, PsPL8A completely unfolds at 60°C. SAXS analysis showed, PsPL8A is fully folded in solution form. The ab initio derived dummy model of PsPL8A superposed well with its modeled structure excluding some α-helices and loop region. Structural superposition and docking analysis showed, N153, W105, H203, Y208, Y212, R266 and E349 were involved in catalysis. Mutants N153A, H203A, Y212F, R266A and E349A created by SDM revealed no residual activity. Isothermal titration calorimetry analysis of Y212F and H203A with C4S polysaccharide, showed moderate binding by Y212F (Ka=9.56±3.81×105) and no binding with H203A, showing active contribution of Y212 in substrate binding. Residues Y212 and H203 or R266 might act as general base and general acid respectively. Residues N153 and E349 are likely contributing in charge neutralization and stabilizing enolate anion intermediate during β-elimination.
Collapse
Affiliation(s)
- Aruna Rani
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Arun Dhillon
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Kedar Sharma
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Arun Goyal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| |
Collapse
|
43
|
Feng T, Chen F, Kang Y, Sun H, Liu H, Li D, Zhu F, Hou T. HawkRank: a new scoring function for protein-protein docking based on weighted energy terms. J Cheminform 2017; 9:66. [PMID: 29282565 PMCID: PMC5745212 DOI: 10.1186/s13321-017-0254-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 12/14/2017] [Indexed: 01/09/2023] Open
Abstract
Deciphering the structural determinants of protein–protein interactions (PPIs) is essential to gain a deep understanding of many important biological functions in the living cells. Computational approaches for the structural modeling of PPIs, such as protein–protein docking, are quite needed to complement existing experimental techniques. The reliability of a protein–protein docking method is dependent on the ability of the scoring function to accurately distinguish the near-native binding structures from a huge number of decoys. In this study, we developed HawkRank, a novel scoring function designed for the sampling stage of protein–protein docking by summing the contributions from several energy terms, including van der Waals potentials, electrostatic potentials and desolvation potentials. First, based on the solvation free energies predicted by the Generalized Born model for ~ 800 proteins, a SASA (solvent accessible surface area)-based solvation model was developed, which can give the aqueous solvation free energies for proteins by summing the contributions of 21 atom types. Then, the van der Waals potentials and electrostatic potentials based on the Amber ff14SB force field were computed. Finally, the HawkRank scoring function was derived by determining the most optimal weights for five energy terms based on the training set. Here, MSR (modified success rate), a novel protein–protein scoring quality index, was used to assess the performance of HawkRank and three other popular protein–protein scoring functions, including ZRANK, FireDock and dDFIRE. The results show that HawkRank outperformed the other three scoring functions according to the total number of hits and MSR. HawkRank is available at http://cadd.zju.edu.cn/programs/hawkrank.
Collapse
Affiliation(s)
- Ting Feng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Fu Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Yu Kang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Huiyong Sun
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Hui Liu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Dan Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Tingjun Hou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China. .,State Key Lab of CAD&CG, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| |
Collapse
|
44
|
Alam N, Goldstein O, Xia B, Porter KA, Kozakov D, Schueler-Furman O. High-resolution global peptide-protein docking using fragments-based PIPER-FlexPepDock. PLoS Comput Biol 2017; 13:e1005905. [PMID: 29281622 PMCID: PMC5760072 DOI: 10.1371/journal.pcbi.1005905] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 01/09/2018] [Accepted: 11/29/2017] [Indexed: 11/24/2022] Open
Abstract
Peptide-protein interactions contribute a significant fraction of the protein-protein interactome. Accurate modeling of these interactions is challenging due to the vast conformational space associated with interactions of highly flexible peptides with large receptor surfaces. To address this challenge we developed a fragment based high-resolution peptide-protein docking protocol. By streamlining the Rosetta fragment picker for accurate peptide fragment ensemble generation, the PIPER docking algorithm for exhaustive fragment-receptor rigid-body docking and Rosetta FlexPepDock for flexible full-atom refinement of PIPER docked models, we successfully addressed the challenge of accurate and efficient global peptide-protein docking at high-resolution with remarkable accuracy, as validated on a small but representative set of peptide-protein complex structures well resolved by X-ray crystallography. Our approach opens up the way to high-resolution modeling of many more peptide-protein interactions and to the detailed study of peptide-protein association in general. PIPER-FlexPepDock is freely available to the academic community as a server at http://piperfpd.furmanlab.cs.huji.ac.il. Peptide-protein interactions are crucial components of various important biological processes in living cells. High-resolution structural information of such interactions provides insight about the underlying biophysical principles governing the interactions, and a starting point for their targeted manipulations. Accurate docking algorithms can help fill the gap between the vast number of these interactions and the small number of experimentally solved structures. However, the accuracies of the existing protocols have been limited, in particular for ab initio docking when no information about the peptide beyond its sequence is available. Here we introduce PIPER-FlexPepDock, a fragment-based global docking protocol for high-resolution modeling of peptide-protein interactions. Integration of accurate and efficient representation of the peptide using fragment ensembles, their fast and exhaustive rigid-body docking, and their subsequent accurate flexible refinement, enables peptide-protein docking of remarkable accuracy. The validation on a representative benchmark set of crystallographically solved high-resolution peptide-protein complexes demonstrates significantly improved performance over all existing docking protocols. This opens up the way to the modeling of many more peptide-protein interactions, and to a more detailed study of peptide-protein association in general.
Collapse
Affiliation(s)
- Nawsad Alam
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Oriel Goldstein
- School of Computer Sciences and Engineering, The Hebrew University, Jerusalem, Israel
| | - Bing Xia
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
| | - Kathryn A. Porter
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
| | - Dima Kozakov
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York, United States of America
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York, United States of America
- Institute for Advanced Computational Sciences, Stony Brook University, Stony Brook, New York, United States of America
- * E-mail: (OSF); (DK)
| | - Ora Schueler-Furman
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
- * E-mail: (OSF); (DK)
| |
Collapse
|
45
|
Roberts VA, Pique ME, Hsu S, Li S. Combining H/D Exchange Mass Spectrometry and Computational Docking To Derive the Structure of Protein-Protein Complexes. Biochemistry 2017; 56:6329-6342. [PMID: 29099587 DOI: 10.1021/acs.biochem.7b00643] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Protein-protein interactions are essential for biological function, but structures of protein-protein complexes are difficult to obtain experimentally. To derive the protein complex of the DNA-repair enzyme human uracil-DNA-glycosylase (hUNG) with its protein inhibitor (UGI), we combined rigid-body computational docking with hydrogen/deuterium exchange mass spectrometry (DXMS). Computational docking of the unbound protein structures provides a list of possible three-dimensional models of the complex; DXMS identifies solvent-protected protein residues. DXMS showed that unbound hUNG is compactly folded, but unbound UGI is loosely packed. An increased level of solvent protection of hUNG in the complex was localized to four regions on the same face. The decrease in the number of incorporated deuterons was quantitatively interpreted as the minimum number of main-chain hUNG amides buried in the protein-protein interface. The level of deuteration of complexed UGI decreased throughout the protein chain, indicating both tighter packing and direct solvent protection by hUNG. Three UGI regions showing the greatest decreases were best interpreted leniently, requiring just one main-chain amide from each in the interface. Applying the DXMS constraints as filters to a list of docked complexes gave the correct complex as the largest favorable energy cluster. Thus, identification of approximate protein interfaces was sufficient to distinguish the protein complex. Surprisingly, incorporating the DXMS data as added favorable potentials in the docking calculation was less effective in finding the correct complex. The filtering method has greater flexibility, with the capability to test each constraint and enforce simultaneous contact by multiple regions, but with the caveat that the list from the unbiased docking must include correct complexes.
Collapse
Affiliation(s)
- Victoria A Roberts
- San Diego Supercomputer Center, University of California, San Diego , La Jolla, California 92093, United States
| | - Michael E Pique
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute , La Jolla, California 92037, United States
| | - Simon Hsu
- School of Medicine, University of California, San Diego , La Jolla, California 92093, United States
| | - Sheng Li
- School of Medicine, University of California, San Diego , La Jolla, California 92093, United States
| |
Collapse
|
46
|
Tramontano A. The computational prediction of protein assemblies. Curr Opin Struct Biol 2017; 46:170-175. [PMID: 29102305 DOI: 10.1016/j.sbi.2017.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 10/04/2017] [Accepted: 10/05/2017] [Indexed: 10/18/2022]
Abstract
The function of proteins in the cell is almost always mediated by their interaction with different partners, including other proteins, nucleic acids or small organic molecules. The ability of identifying all of them is an essential step in our quest for understanding life at the molecular level. The inference of the protein complex composition and of its molecular details can also provide relevant clues for the development and the design of drugs. In this short review, I will discuss the computational aspects of the analysis and prediction of protein-protein assemblies and discuss some of the most recent developments as seen in the last Critical Assessment of Techniques for Protein Structure Prediction (CASP) experiment.
Collapse
Affiliation(s)
- Anna Tramontano
- Physics Department, Sapienza University of Rome, Piazzale Aldo Moro, 5 I-00185 Roma, Italy; Istituto Pasteur - Fondazione Cenci Bolognetti, Sapienza University of Rome, Piazzale Aldo Moro, 5 I-00185 Roma, Italy
| |
Collapse
|
47
|
Marcu O, Dodson EJ, Alam N, Sperber M, Kozakov D, Lensink MF, Schueler-Furman O. FlexPepDock lessons from CAPRI peptide-protein rounds and suggested new criteria for assessment of model quality and utility. Proteins 2017; 85:445-462. [PMID: 28002624 PMCID: PMC6618814 DOI: 10.1002/prot.25230] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 11/15/2016] [Accepted: 11/23/2016] [Indexed: 12/21/2022]
Abstract
CAPRI rounds 28 and 29 included, for the first time, peptide-receptor targets of three different systems, reflecting increased appreciation of the importance of peptide-protein interactions. The CAPRI rounds allowed us to objectively assess the performance of Rosetta FlexPepDock, one of the first protocols to explicitly include peptide flexibility in docking, accounting for peptide conformational changes upon binding. We discuss here successes and challenges in modeling these targets: we obtain top-performing, high-resolution models of the peptide motif for cases with known binding sites but there is a need for better modeling of flanking regions, as well as better selection criteria, in particular for unknown binding sites. These rounds have also provided us the opportunity to reassess the success criteria, to better reflect the quality of a peptide-protein complex model. Using all models submitted to CAPRI, we analyze the correlation between current classification criteria and the ability to retrieve critical interface features, such as hydrogen bonds and hotspots. We find that loosening the backbone (and ligand) RMSD threshold, together with a restriction on the side chain RMSD measure, allows us to improve the selection of high-accuracy models. We also suggest a new measure to assess interface hydrogen bond recovery, which is not assessed by the current CAPRI criteria. Finally, we find that surprisingly much can be learned from rather inaccurate models about binding hotspots, suggesting that the current status of peptide-protein docking methods, as reflected by the submitted CAPRI models, can already have a significant impact on our understanding of protein interactions. Proteins 2017; 85:445-462. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Orly Marcu
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, the Hebrew University of Jerusalem, Israel
| | - Emma-Joy Dodson
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, the Hebrew University of Jerusalem, Israel
| | - Nawsad Alam
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, the Hebrew University of Jerusalem, Israel
| | - Michal Sperber
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, the Hebrew University of Jerusalem, Israel
| | - Dima Kozakov
- Department of Applied Mathematics and Statistics, Stony Brooks University, Stony Brook, New York, 11794
| | - Marc F Lensink
- University of Lille, CNRS UMR8576 UGSF, Lille, 59000, France
| | - Ora Schueler-Furman
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, the Hebrew University of Jerusalem, Israel
| |
Collapse
|
48
|
de Vries SJ, Zacharias M. Fast and accurate grid representations for atom-based docking with partner flexibility. J Comput Chem 2017; 38:1538-1546. [DOI: 10.1002/jcc.24795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 01/18/2017] [Accepted: 01/19/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Sjoerd J. de Vries
- MTi, UMR-S 973, Physics Department T38; Technische Universität München; James-Franck-Strasse 1 85748 Garching Germany
| | - Martin Zacharias
- MTi, UMR-S 973, Physics Department T38; Technische Universität München; James-Franck-Strasse 1 85748 Garching Germany
| |
Collapse
|
49
|
Pfeiffenberger E, Chaleil RA, Moal IH, Bates PA. A machine learning approach for ranking clusters of docked protein-protein complexes by pairwise cluster comparison. Proteins 2017; 85:528-543. [PMID: 27935158 PMCID: PMC5396268 DOI: 10.1002/prot.25218] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 11/14/2016] [Accepted: 11/21/2016] [Indexed: 01/28/2023]
Abstract
Reliable identification of near-native poses of docked protein-protein complexes is still an unsolved problem. The intrinsic heterogeneity of protein-protein interactions is challenging for traditional biophysical or knowledge based potentials and the identification of many false positive binding sites is not unusual. Often, ranking protocols are based on initial clustering of docked poses followed by the application of an energy function to rank each cluster according to its lowest energy member. Here, we present an approach of cluster ranking based not only on one molecular descriptor (e.g., an energy function) but also employing a large number of descriptors that are integrated in a machine learning model, whereby, an extremely randomized tree classifier based on 109 molecular descriptors is trained. The protocol is based on first locally enriching clusters with additional poses, the clusters are then characterized using features describing the distribution of molecular descriptors within the cluster, which are combined into a pairwise cluster comparison model to discriminate near-native from incorrect clusters. The results show that our approach is able to identify clusters containing near-native protein-protein complexes. In addition, we present an analysis of the descriptors with respect to their power to discriminate near native from incorrect clusters and how data transformations and recursive feature elimination can improve the ranking performance. Proteins 2017; 85:528-543. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | | | - Iain H. Moal
- European Molecular Biology LaboratoryEuropean Bioinformatics Institute, Wellcome Trust Genome Campus, HinxtonCambridgeCB10 1SDUK
| | - Paul A. Bates
- Biomolecular Modelling LaboratoryThe Francis Crick InstituteLondonNW1 1ATUK
| |
Collapse
|
50
|
Weitzner BD, Jeliazkov JR, Lyskov S, Marze N, Kuroda D, Frick R, Adolf-Bryfogle J, Biswas N, Dunbrack RL, Gray JJ. Modeling and docking of antibody structures with Rosetta. Nat Protoc 2017; 12:401-416. [PMID: 28125104 DOI: 10.1038/nprot.2016.180] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We describe Rosetta-based computational protocols for predicting the 3D structure of an antibody from sequence (RosettaAntibody) and then docking the antibody to protein antigens (SnugDock). Antibody modeling leverages canonical loop conformations to graft large segments from experimentally determined structures, as well as offering (i) energetic calculations to minimize loops, (ii) docking methodology to refine the VL-VH relative orientation and (iii) de novo prediction of the elusive complementarity determining region (CDR) H3 loop. To alleviate model uncertainty, antibody-antigen docking resamples CDR loop conformations and can use multiple models to represent an ensemble of conformations for the antibody, the antigen or both. These protocols can be run fully automated via the ROSIE web server (http://rosie.rosettacommons.org/) or manually on a computer with user control of individual steps. For best results, the protocol requires roughly 1,000 CPU-hours for antibody modeling and 250 CPU-hours for antibody-antigen docking. Tasks can be completed in under a day by using public supercomputers.
Collapse
Affiliation(s)
- Brian D Weitzner
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jeliazko R Jeliazkov
- Program in Molecular Biophysics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sergey Lyskov
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Nicholas Marze
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Daisuke Kuroda
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Analytical and Physical Chemistry, Showa University School of Pharmacy, Tokyo, Japan
| | - Rahel Frick
- Centre for Immune Regulation, Department of Biosciences, University of Oslo, Oslo, Norway.,Centre for Immune Regulation, Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Jared Adolf-Bryfogle
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, USA.,Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Naireeta Biswas
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Roland L Dunbrack
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Jeffrey J Gray
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Program in Molecular Biophysics, Johns Hopkins University, Baltimore, Maryland, USA.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, USA.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|