1
|
Sichrovsky M, Lacabanne D, Ruprecht JJ, Rana JJ, Stanik K, Dionysopoulou M, Sowton AP, King MS, Jones SA, Cooper L, Hardwick SW, Paris G, Chirgadze DY, Ding S, Fearnley IM, Palmer SM, Pardon E, Steyaert J, Leone V, Forrest LR, Tavoulari S, Kunji ERS. Molecular basis of pyruvate transport and inhibition of the human mitochondrial pyruvate carrier. SCIENCE ADVANCES 2025; 11:eadw1489. [PMID: 40249800 PMCID: PMC12007569 DOI: 10.1126/sciadv.adw1489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 03/14/2025] [Indexed: 04/20/2025]
Abstract
The mitochondrial pyruvate carrier transports pyruvate, produced by glycolysis from sugar molecules, into the mitochondrial matrix, as a crucial transport step in eukaryotic energy metabolism. The carrier is a drug target for the treatment of cancers, diabetes mellitus, neurodegeneration, and metabolic dysfunction-associated steatotic liver disease. We have solved the structure of the human MPC1L/MPC2 heterodimer in the inward- and outward-open states by cryo-electron microscopy, revealing its alternating access rocker-switch mechanism. The carrier has a central binding site for pyruvate, which contains an essential lysine and histidine residue, important for its ΔpH-dependent transport mechanism. We have also determined the binding poses of three chemically distinct inhibitor classes, which exploit the same binding site in the outward-open state by mimicking pyruvate interactions and by using aromatic stacking interactions.
Collapse
Affiliation(s)
- Maximilian Sichrovsky
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Denis Lacabanne
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Jonathan J. Ruprecht
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Jessica J. Rana
- Computational Structural Biology Section, National Institutes of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Klaudia Stanik
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Mariangela Dionysopoulou
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Alice P. Sowton
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Martin S. King
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Scott A. Jones
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Lee Cooper
- Department of Biochemistry, University of Cambridge, Sanger Building, Tennis Court Road, Cambridge CB2 1GA, UK
| | - Steven W. Hardwick
- Department of Biochemistry, University of Cambridge, Sanger Building, Tennis Court Road, Cambridge CB2 1GA, UK
| | - Giulia Paris
- Department of Biochemistry, University of Cambridge, Sanger Building, Tennis Court Road, Cambridge CB2 1GA, UK
| | - Dimitri Y. Chirgadze
- Department of Biochemistry, University of Cambridge, Sanger Building, Tennis Court Road, Cambridge CB2 1GA, UK
| | - Shujing Ding
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Ian M. Fearnley
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Shane M. Palmer
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Els Pardon
- VIB-VUB Center for Structural Biology, VIB, Pleinlaan 2, B-1050 Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
| | - Jan Steyaert
- VIB-VUB Center for Structural Biology, VIB, Pleinlaan 2, B-1050 Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium
| | - Vanessa Leone
- Computational Structural Biology Section, National Institutes of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
- Department of Biophysics and Data Science Institute, Medical College of Wisconsin, Milwaukee, WI 53226-3548, USA
| | - Lucy R. Forrest
- Computational Structural Biology Section, National Institutes of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Sotiria Tavoulari
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Edmund R. S. Kunji
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| |
Collapse
|
2
|
Shirali A, Stebliankin V, Karki U, Shi J, Chapagain P, Narasimhan G. A comprehensive survey of scoring functions for protein docking models. BMC Bioinformatics 2025; 26:25. [PMID: 39844036 PMCID: PMC11755896 DOI: 10.1186/s12859-024-05991-4] [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: 07/04/2024] [Accepted: 11/18/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND While protein-protein docking is fundamental to our understanding of how proteins interact, scoring protein-protein complex conformations is a critical component of successful docking programs. Without accurate and efficient scoring functions to differentiate between native and non-native binding complexes, the accuracy of current docking tools cannot be guaranteed. Although many innovative scoring functions have been proposed, a good scoring function for docking remains elusive. Deep learning models offer alternatives to using explicit empirical or mathematical functions for scoring protein-protein complexes. RESULTS In this study, we perform a comprehensive survey of the state-of-the-art scoring functions by considering the most popular and highly performant approaches, both classical and deep learning-based, for scoring protein-protein complexes. The methods were also compared based on their runtime as it directly impacts their use in large-scale docking applications. CONCLUSIONS We evaluate the strengths and weaknesses of classical and deep learning-based approaches across seven public and popular datasets to aid researchers in understanding the progress made in this field.
Collapse
Affiliation(s)
- Azam Shirali
- Bioinformatics Research Group (BioRG), Knight Foundation School of Computing and Information Sciences, Florida International University, 11200 SW 8th 10 St, Miami, 33199, USA
| | - Vitalii Stebliankin
- Bioinformatics Research Group (BioRG), Knight Foundation School of Computing and Information Sciences, Florida International University, 11200 SW 8th 10 St, Miami, 33199, USA
| | - Ukesh Karki
- Department of Physics, Florida International University, 11200 SW 8th 10 St, Miami, 33199, USA
| | - Jimeng Shi
- Bioinformatics Research Group (BioRG), Knight Foundation School of Computing and Information Sciences, Florida International University, 11200 SW 8th 10 St, Miami, 33199, USA
| | - Prem Chapagain
- Department of Physics, Florida International University, 11200 SW 8th 10 St, Miami, 33199, USA
- Biomolecular Sciences Institute, Florida International University, 11200 SW 8th St, Miami, 33199, USA
| | - Giri Narasimhan
- Bioinformatics Research Group (BioRG), Knight Foundation School of Computing and Information Sciences, Florida International University, 11200 SW 8th 10 St, Miami, 33199, USA.
- Biomolecular Sciences Institute, Florida International University, 11200 SW 8th St, Miami, 33199, USA.
| |
Collapse
|
3
|
Liang F, Sun M, Xie L, Zhao X, Liu D, Zhao K, Zhang G. Recent advances and challenges in protein complex model accuracy estimation. Comput Struct Biotechnol J 2024; 23:1824-1832. [PMID: 38707538 PMCID: PMC11066466 DOI: 10.1016/j.csbj.2024.04.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 05/07/2024] Open
Abstract
Estimation of model accuracy plays a crucial role in protein structure prediction, aiming to evaluate the quality of predicted protein structure models accurately and objectively. This process is not only key to screening candidate models that are close to the real structure, but also provides guidance for further optimization of protein structures. With the significant advancements made by AlphaFold2 in monomer structure, the problem of single-domain protein structure prediction has been widely solved. Correspondingly, the importance of assessing the quality of single-domain protein models decreased, and the research focus has shifted to estimation of model accuracy of protein complexes. In this review, our goal is to provide a comprehensive overview of the reference and statistical metrics, as well as representative methods, and the current challenges within four distinct facets (Topology Global Score, Interface Total Score, Interface Residue-Wise Score, and Tertiary Residue-Wise Score) in the field of complex EMA.
Collapse
Affiliation(s)
| | | | - Lei Xie
- College of Information Engineering, Zhejiang University of Technology, Hangzhou 310023, China
| | - Xuanfeng Zhao
- College of Information Engineering, Zhejiang University of Technology, Hangzhou 310023, China
| | - Dong Liu
- College of Information Engineering, Zhejiang University of Technology, Hangzhou 310023, China
| | - Kailong Zhao
- College of Information Engineering, Zhejiang University of Technology, Hangzhou 310023, China
| | - Guijun Zhang
- College of Information Engineering, Zhejiang University of Technology, Hangzhou 310023, China
| |
Collapse
|
4
|
Mirabello C, Wallner B. DockQ v2: improved automatic quality measure for protein multimers, nucleic acids, and small molecules. Bioinformatics 2024; 40:btae586. [PMID: 39348158 PMCID: PMC11467047 DOI: 10.1093/bioinformatics/btae586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/01/2024] Open
Abstract
MOTIVATION It is important to assess the quality of modeled biomolecules to benchmark and assess the performance of different prediction methods. DockQ has emerged as the standard tool for assessing the quality of protein interfaces in model structures against given references. However, as predictions of large multimers with multiple chains become more common, DockQ needs to be updated with more functionality for robustness and speed. Moreover, as the field progresses and more methods are released to predict interactions between proteins and other types of molecules, such as nucleic acids and small molecules, it becomes necessary to have a tool that can assess all types of interactions. RESULTS Here, we present a complete reimplementation of DockQ in pure Python. The updated version of DockQ is more portable, faster and introduces novel functionalities, such as automatic DockQ calculations for multiple interfaces and automatic chain mapping with multi-threading. These enhancements are designed to facilitate comparative analyses of protein complexes, particularly large multi-chain complexes. Furthermore, DockQ is now also able to score interfaces between proteins, nucleic acids, and small molecules. AVAILABILITY AND IMPLEMENTATION DockQ v2 is available online at: https://wallnerlab.org/DockQ.
Collapse
Affiliation(s)
- Claudio Mirabello
- Division of Bioinformatics, Department of Physics, Chemistry and Biology, Linköping University, SE-581 83 Linköping, Sweden
- National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Linköping University, SE-581 83 Linköping, Sweden
| | - Björn Wallner
- Division of Bioinformatics, Department of Physics, Chemistry and Biology, Linköping University, SE-581 83 Linköping, Sweden
| |
Collapse
|
5
|
Wang J, Wang X, Chu Y, Li C, Li X, Meng X, Fang Y, No KT, Mao J, Zeng X. Exploring the Conformational Ensembles of Protein-Protein Complex with Transformer-Based Generative Model. J Chem Theory Comput 2024; 20:4469-4480. [PMID: 38816696 DOI: 10.1021/acs.jctc.4c00255] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
Protein-protein interactions are the basis of many protein functions, and understanding the contact and conformational changes of protein-protein interactions is crucial for linking the protein structure to biological function. Although difficult to detect experimentally, molecular dynamics (MD) simulations are widely used to study the conformational ensembles and dynamics of protein-protein complexes, but there are significant limitations in sampling efficiency and computational costs. In this study, a generative neural network was trained on protein-protein complex conformations obtained from molecular simulations to directly generate novel conformations with physical realism. We demonstrated the use of a deep learning model based on the transformer architecture to explore the conformational ensembles of protein-protein complexes through MD simulations. The results showed that the learned latent space can be used to generate unsampled conformations of protein-protein complexes for obtaining new conformations complementing pre-existing ones, which can be used as an exploratory tool for the analysis and enhancement of molecular simulations of protein-protein complexes.
Collapse
Affiliation(s)
- Jianmin Wang
- The Interdisciplinary Graduate Program in Integrative Biotechnology, Yonsei University, Incheon 21983, Korea
| | - Xun Wang
- School of Computer Science and Technology, China University of Petroleum, Qingdao, Shandong 266580, P. R. China
- High Performance Computer Research Center, University of Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Yanyi Chu
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Chunyan Li
- School of Informatics, Yunnan Normal University, Kunming, Yunnan 650500, P. R. China
| | - Xue Li
- School of Computer Science and Technology, China University of Petroleum, Qingdao, Shandong 266580, P. R. China
| | - Xiangyu Meng
- School of Computer Science and Technology, China University of Petroleum, Qingdao, Shandong 266580, P. R. China
| | - Yitian Fang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
| | - Kyoung Tai No
- The Interdisciplinary Graduate Program in Integrative Biotechnology, Yonsei University, Incheon 21983, Korea
| | - Jiashun Mao
- School of Medical Information and Engineering, Southwest Medical University, Luzhou, Sichuan 646000, P. R. China
| | - Xiangxiang Zeng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, Hunan 410082, P. R. China
| |
Collapse
|
6
|
Chen X, Liu J, Park N, Cheng J. A Survey of Deep Learning Methods for Estimating the Accuracy of Protein Quaternary Structure Models. Biomolecules 2024; 14:574. [PMID: 38785981 PMCID: PMC11117562 DOI: 10.3390/biom14050574] [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: 03/01/2024] [Revised: 04/07/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
The quality prediction of quaternary structure models of a protein complex, in the absence of its true structure, is known as the Estimation of Model Accuracy (EMA). EMA is useful for ranking predicted protein complex structures and using them appropriately in biomedical research, such as protein-protein interaction studies, protein design, and drug discovery. With the advent of more accurate protein complex (multimer) prediction tools, such as AlphaFold2-Multimer and ESMFold, the estimation of the accuracy of protein complex structures has attracted increasing attention. Many deep learning methods have been developed to tackle this problem; however, there is a noticeable absence of a comprehensive overview of these methods to facilitate future development. Addressing this gap, we present a review of deep learning EMA methods for protein complex structures developed in the past several years, analyzing their methodologies, data and feature construction. We also provide a prospective summary of some potential new developments for further improving the accuracy of the EMA methods.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65211, USA
| | - Jian Liu
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65211, USA
- NextGen Precision Health Institute, University of Missouri, Columbia, MO 65211, USA
| | - Nolan Park
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65211, USA
| | - Jianlin Cheng
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65211, USA
- NextGen Precision Health Institute, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
7
|
Thathapudi NC, Callai-Silva N, Malhotra K, Basu S, Aghajanzadeh-Kiyaseh M, Zamani-Roudbaraki M, Groleau M, Lombard-Vadnais F, Lesage S, Griffith M. Modified host defence peptide GF19 slows TNT-mediated spread of corneal herpes simplex virus serotype I infection. Sci Rep 2024; 14:4096. [PMID: 38374240 PMCID: PMC10876564 DOI: 10.1038/s41598-024-53662-4] [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: 12/11/2023] [Accepted: 02/03/2024] [Indexed: 02/21/2024] Open
Abstract
Corneal HSV-1 infections are a leading cause of infectious blindness globally by triggering tissue damage due to the intense inflammation. HSV-1 infections are treated mainly with antiviral drugs that clear the infections but are inefficient as prophylactics. The body produces innate cationic host defence peptides (cHDP), such as the cathelicidin LL37. Various epithelia, including the corneal epithelium, express LL37. cHDPs can cause disintegration of pathogen membranes, stimulate chemokine production, and attract immune cells. Here, we selected GF17, a peptide containing the LL37 fragment with bioactivity but with minimal cytotoxicity, and added two cell-penetrating amino acids to enhance its activity. The resulting GF19 was relatively cell-friendly, inducing only partial activation of antigen presenting immune cells in vitro. We showed that HSV-1 spreads by tunneling nanotubes in cultured human corneal epithelial cells. GF19 given before infection was able to block infection, most likely by blocking viral entry. When cells were sequentially exposed to viruses and GF19, the infection was attenuated but not arrested, supporting the contention that the GF19 mode of action was to block viral entry. Encapsulation into silica nanoparticles allowed a more sustained release of GF19, enhancing its activity. GF19 is most likely suitable as a prevention rather than a virucidal treatment.
Collapse
Affiliation(s)
- Neethi C Thathapudi
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC, H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Institute of Biomedical Engineering, Université de Montréal, Montreal, QC, H3T 1J4, Canada
| | - Natalia Callai-Silva
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC, H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Institute of Biomedical Engineering, Université de Montréal, Montreal, QC, H3T 1J4, Canada
| | - Kamal Malhotra
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC, H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, University of Ottawa, Ottawa, K1Y 4W7, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, K1H 8M5, Canada
| | - Sankar Basu
- Department of Microbiology, Asutosh College, (Affiliated With University of Calcutta), Kolkata, 700026, India
| | - Mozhgan Aghajanzadeh-Kiyaseh
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC, H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Institute of Biomedical Engineering, Université de Montréal, Montreal, QC, H3T 1J4, Canada
| | - Mostafa Zamani-Roudbaraki
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC, H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Institute of Biomedical Engineering, Université de Montréal, Montreal, QC, H3T 1J4, Canada
| | - Marc Groleau
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC, H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | | | - Sylvie Lesage
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC, H1T 2M4, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - May Griffith
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC, H1T 2M4, Canada.
- Department of Ophthalmology, Université de Montréal, Montreal, QC, H3C 3J7, Canada.
- Institute of Biomedical Engineering, Université de Montréal, Montreal, QC, H3T 1J4, Canada.
| |
Collapse
|
8
|
Zhang L, Wang S, Hou J, Si D, Zhu J, Cao R. ComplexQA: a deep graph learning approach for protein complex structure assessment. Brief Bioinform 2023; 24:bbad287. [PMID: 37930021 DOI: 10.1093/bib/bbad287] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/09/2023] [Accepted: 07/24/2023] [Indexed: 11/07/2023] Open
Abstract
MOTIVATION In recent years, the end-to-end deep learning method for single-chain protein structure prediction has achieved high accuracy. For example, the state-of-the-art method AlphaFold, developed by Google, has largely increased the accuracy of protein structure predictions to near experimental accuracy in some of the cases. At the same time, there are few methods that can evaluate the quality of protein complexes at the residue level. In particular, evaluating the quality of residues at the interface of protein complexes can lead to a wide range of applications, such as protein function analysis and drug design. In this paper, we introduce a new deep graph neural network-based method ComplexQA, to evaluate the local quality of interfaces for protein complexes by utilizing the residue-level structural information in 3D space and the sequence-level constraints. RESULTS We benchmark our method to other state-of-the-art quality assessment approaches on the HAF2 and DBM55-AF2 datasets (high-quality structural models predicted by AlphaFold-Multimer), and the BM5 docking dataset. The experimental results show that our proposed method achieves better or similar performance compared with other state-of-the-art methods, especially on difficult targets which only contain a few acceptable models. Our method is able to suggest a score for each interfac e residue, which demonstrates a powerful assessment tool for the ever-increasing number of protein complexes. AVAILABILITY https://github.com/Cao-Labs/ComplexQA.git. Contact: caora@plu.edu.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Computer Science and Technology, AnHui University, Hefei, 230601, Anhui, China
| | - Sheng Wang
- Department of Computer Science and Technology, AnHui University, Hefei, 230601, Anhui, China
| | - Jie Hou
- Department of Computer Science, Saint Louis University, Saint. Louis, 63103, MO, USA
| | - Dong Si
- Division of Computing and Software Systems, University of Washington Bothell, Bothell, 98011, WA, USA
| | - Junyong Zhu
- Department of Computer Science and Technology, AnHui University, Hefei, 230601, Anhui, China
| | - Renzhi Cao
- Department of Humanities, Pacific Lutheran University, Tacoma, 98447, WA, USA
| |
Collapse
|
9
|
Biswas G, Mukherjee D, Dutta N, Ghosh P, Basu S. EnCPdock: a web-interface for direct conjoint comparative analyses of complementarity and binding energetics in inter-protein associations. J Mol Model 2023; 29:239. [PMID: 37423912 DOI: 10.1007/s00894-023-05626-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/20/2023] [Indexed: 07/11/2023]
Abstract
CONTEXT Protein-protein interaction (PPI) is a key component linked to virtually all cellular processes. Be it an enzyme catalysis ('classic type functions' of proteins) or a signal transduction ('non-classic'), proteins generally function involving stable or quasi-stable multi-protein associations. The physical basis for such associations is inherent in the combined effect of shape and electrostatic complementarities (Sc, EC) of the interacting protein partners at their interface, which provides indirect probabilistic estimates of the stability and affinity of the interaction. While Sc is a necessary criterion for inter-protein associations, EC can be favorable as well as disfavored (e.g., in transient interactions). Estimating equilibrium thermodynamic parameters (∆Gbinding, Kd) by experimental means is costly and time consuming, thereby opening windows for computational structural interventions. Attempts to empirically probe ∆Gbinding from coarse-grain structural descriptors (primarily, surface area based terms) have lately been overtaken by physics-based, knowledge-based and their hybrid approaches (MM/PBSA, FoldX, etc.) that directly compute ∆Gbinding without involving intermediate structural descriptors. METHODS Here, we present EnCPdock ( https://www.scinetmol.in/EnCPdock/ ), a user-friendly web-interface for the direct conjoint comparative analyses of complementarity and binding energetics in proteins. EnCPdock returns an AI-predicted ∆Gbinding computed by combining complementarity (Sc, EC) and other high-level structural descriptors (input feature vectors), and renders a prediction accuracy comparable to the state-of-the-art. EnCPdock further locates a PPI complex in terms of its {Sc, EC} values (taken as an ordered pair) in the two-dimensional complementarity plot (CP). In addition, it also generates mobile molecular graphics of the interfacial atomic contact network for further analyses. EnCPdock also furnishes individual feature trends along with the relative probability estimates (Prfmax) of the obtained feature-scores with respect to the events of their highest observed frequencies. Together, these functionalities are of real practical use for structural tinkering and intervention as might be relevant in the design of targeted protein-interfaces. Combining all its features and applications, EnCPdock presents a unique online tool that should be beneficial to structural biologists and researchers across related fraternities.
Collapse
Affiliation(s)
- Gargi Biswas
- Department of Chemistry and Structural Biology, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Debasish Mukherjee
- Institute of Molecular Biology gGmbH (IMB), Ackermannweg 4, 55128, Mainz, Germany
| | - Nalok Dutta
- Dept of Biochemical Engineering, Faculty of Engineering Science, University College London, London, WC1E 6BT, UK
| | - Prithwi Ghosh
- Department of Botany, Narajole Raj College, Vidyasagar University, Midnapore, 721211, India
| | - Sankar Basu
- Department of Microbiology, Asutosh College (affiliated with University of Calcutta), 92, Shyama Prasad Mukherjee Rd, Bhowanipore, 700026, Kolkata, India.
| |
Collapse
|
10
|
Chen X, Morehead A, Liu J, Cheng J. A gated graph transformer for protein complex structure quality assessment and its performance in CASP15. Bioinformatics 2023; 39:i308-i317. [PMID: 37387159 PMCID: PMC10311325 DOI: 10.1093/bioinformatics/btad203] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2023] Open
Abstract
MOTIVATION Proteins interact to form complexes to carry out essential biological functions. Computational methods such as AlphaFold-multimer have been developed to predict the quaternary structures of protein complexes. An important yet largely unsolved challenge in protein complex structure prediction is to accurately estimate the quality of predicted protein complex structures without any knowledge of the corresponding native structures. Such estimations can then be used to select high-quality predicted complex structures to facilitate biomedical research such as protein function analysis and drug discovery. RESULTS In this work, we introduce a new gated neighborhood-modulating graph transformer to predict the quality of 3D protein complex structures. It incorporates node and edge gates within a graph transformer framework to control information flow during graph message passing. We trained, evaluated and tested the method (called DProQA) on newly-curated protein complex datasets before the 15th Critical Assessment of Techniques for Protein Structure Prediction (CASP15) and then blindly tested it in the 2022 CASP15 experiment. The method was ranked 3rd among the single-model quality assessment methods in CASP15 in terms of the ranking loss of TM-score on 36 complex targets. The rigorous internal and external experiments demonstrate that DProQA is effective in ranking protein complex structures. AVAILABILITY AND IMPLEMENTATION The source code, data, and pre-trained models are available at https://github.com/jianlin-cheng/DProQA.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65201, United States
| | - Alex Morehead
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65201, United States
| | - Jian Liu
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65201, United States
| | - Jianlin Cheng
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO 65201, United States
| |
Collapse
|
11
|
McFee M, Kim PM. GDockScore: a graph-based protein-protein docking scoring function. BIOINFORMATICS ADVANCES 2023; 3:vbad072. [PMID: 37359726 PMCID: PMC10290236 DOI: 10.1093/bioadv/vbad072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/30/2023] [Accepted: 06/10/2023] [Indexed: 06/28/2023]
Abstract
Summary Protein complexes play vital roles in a variety of biological processes, such as mediating biochemical reactions, the immune response and cell signalling, with 3D structure specifying function. Computational docking methods provide a means to determine the interface between two complexed polypeptide chains without using time-consuming experimental techniques. The docking process requires the optimal solution to be selected with a scoring function. Here, we propose a novel graph-based deep learning model that utilizes mathematical graph representations of proteins to learn a scoring function (GDockScore). GDockScore was pre-trained on docking outputs generated with the Protein Data Bank biounits and the RosettaDock protocol, and then fine-tuned on HADDOCK decoys generated on the ZDOCK Protein Docking Benchmark. GDockScore performs similarly to the Rosetta scoring function on docking decoys generated using the RosettaDock protocol. Furthermore, state-of-the-art is achieved on the CAPRI score set, a challenging dataset for developing docking scoring functions. Availability and implementation The model implementation is available at https://gitlab.com/mcfeemat/gdockscore. Supplementary information Supplementary data are available at Bioinformatics Advances online.
Collapse
Affiliation(s)
- Matthew McFee
- Department of Molecular Genetics, The University of Toronto, Toronto, ON M5S 1A8, Canada
- Donnelly Centre for Cellular and Biomolecular Research, The University of Toronto, Toronto, ON M5S 3E1, Canada
| | | |
Collapse
|
12
|
Malhotra K, Buznyk O, Islam MM, Edin E, Basu S, Groleau M, Dégué DS, Fagerholm P, Fois A, Lesage S, Jangamreddy JR, Šimoliūnas E, Liszka A, Patra HK, Griffith M. Phosphorylcholine and KR12-Containing Corneal Implants in HSV-1-Infected Rabbit Corneas. Pharmaceutics 2023; 15:1658. [PMID: 37376106 DOI: 10.3390/pharmaceutics15061658] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/29/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Severe HSV-1 infection can cause blindness due to tissue damage from severe inflammation. Due to the high risk of graft failure in HSV-1-infected individuals, cornea transplantation to restore vision is often contraindicated. We tested the capacity for cell-free biosynthetic implants made from recombinant human collagen type III and 2-methacryloyloxyethyl phosphorylcholine (RHCIII-MPC) to suppress inflammation and promote tissue regeneration in the damaged corneas. To block viral reactivation, we incorporated silica dioxide nanoparticles releasing KR12, the small bioactive core fragment of LL37, an innate cationic host defense peptide produced by corneal cells. KR12 is more reactive and smaller than LL37, so more KR12 molecules can be incorporated into nanoparticles for delivery. Unlike LL37, which was cytotoxic, KR12 was cell-friendly and showed little cytotoxicity at doses that blocked HSV-1 activity in vitro, instead enabling rapid wound closure in cultures of human epithelial cells. Composite implants released KR12 for up to 3 weeks in vitro. The implant was also tested in vivo on HSV-1-infected rabbit corneas where it was grafted by anterior lamellar keratoplasty. Adding KR12 to RHCIII-MPC did not reduce HSV-1 viral loads or the inflammation resulting in neovascularization. Nevertheless, the composite implants reduced viral spread sufficiently to allow stable corneal epithelium, stroma, and nerve regeneration over a 6-month observation period.
Collapse
Affiliation(s)
- Kamal Malhotra
- Department of Ophthalmology, Université de Montréal, Montreal, QC H3C 3J7, Canada
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC H1T 2M4, Canada
| | - Oleksiy Buznyk
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden
- Filatov Institute of Eye Diseases and Tissue Therapy of the NAMS of Ukraine, 65061 Odessa, Ukraine
| | - Mohammad Mirazul Islam
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden
| | - Elle Edin
- Department of Ophthalmology, Université de Montréal, Montreal, QC H3C 3J7, Canada
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC H1T 2M4, Canada
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden
- Institute of Biomedical Engineering, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Sankar Basu
- Department of Microbiology, Asutosh College, Affiliated with University of Calcutta, Kolkata 700026, India
| | - Marc Groleau
- Department of Ophthalmology, Université de Montréal, Montreal, QC H3C 3J7, Canada
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC H1T 2M4, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Delali Shana Dégué
- Department of Ophthalmology, Université de Montréal, Montreal, QC H3C 3J7, Canada
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC H1T 2M4, Canada
- Institute of Biomedical Engineering, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Per Fagerholm
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden
| | - Adrien Fois
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC H1T 2M4, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Sylvie Lesage
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC H1T 2M4, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | | | - Egidijus Šimoliūnas
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, 01513 Vilnius, Lithuania
| | - Aneta Liszka
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden
| | - Hirak K Patra
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden
- Department of Surgical Biotechnology, UCL Division of Surgery and Interventional Science, University College London, London WC1E 6BT, UK
| | - May Griffith
- Department of Ophthalmology, Université de Montréal, Montreal, QC H3C 3J7, Canada
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC H1T 2M4, Canada
- Department of Clinical and Experimental Medicine, Linköping University, 58183 Linköping, Sweden
- Institute of Biomedical Engineering, Université de Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
13
|
Jung Y, Geng C, Bonvin AMJJ, Xue LC, Honavar VG. MetaScore: A Novel Machine-Learning-Based Approach to Improve Traditional Scoring Functions for Scoring Protein-Protein Docking Conformations. Biomolecules 2023; 13:121. [PMID: 36671507 PMCID: PMC9855734 DOI: 10.3390/biom13010121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 01/11/2023] Open
Abstract
Protein-protein interactions play a ubiquitous role in biological function. Knowledge of the three-dimensional (3D) structures of the complexes they form is essential for understanding the structural basis of those interactions and how they orchestrate key cellular processes. Computational docking has become an indispensable alternative to the expensive and time-consuming experimental approaches for determining the 3D structures of protein complexes. Despite recent progress, identifying near-native models from a large set of conformations sampled by docking-the so-called scoring problem-still has considerable room for improvement. We present MetaScore, a new machine-learning-based approach to improve the scoring of docked conformations. MetaScore utilizes a random forest (RF) classifier trained to distinguish near-native from non-native conformations using their protein-protein interfacial features. The features include physicochemical properties, energy terms, interaction-propensity-based features, geometric properties, interface topology features, evolutionary conservation, and also scores produced by traditional scoring functions (SFs). MetaScore scores docked conformations by simply averaging the score produced by the RF classifier with that produced by any traditional SF. We demonstrate that (i) MetaScore consistently outperforms each of the nine traditional SFs included in this work in terms of success rate and hit rate evaluated over conformations ranked among the top 10; (ii) an ensemble method, MetaScore-Ensemble, that combines 10 variants of MetaScore obtained by combining the RF score with each of the traditional SFs outperforms each of the MetaScore variants. We conclude that the performance of traditional SFs can be improved upon by using machine learning to judiciously leverage protein-protein interfacial features and by using ensemble methods to combine multiple scoring functions.
Collapse
Affiliation(s)
- Yong Jung
- Bioinformatics & Genomics Graduate Program, Pennsylvania State University, University Park, PA 16802, USA
- Artificial Intelligence Research Laboratory, Pennsylvania State University, University Park, PA 16802, USA
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
| | - Cunliang Geng
- Bijvoet Centre for Biomolecular Research, Faculty of Science—Chemistry, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Alexandre M. J. J. Bonvin
- Bijvoet Centre for Biomolecular Research, Faculty of Science—Chemistry, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Li C. Xue
- Bijvoet Centre for Biomolecular Research, Faculty of Science—Chemistry, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
- Center for Molecular and Biomolecular Informatics, Radboudumc, Greet Grooteplein 26-28, 6525 GA Nijmegen, The Netherlands
| | - Vasant G. Honavar
- Bioinformatics & Genomics Graduate Program, Pennsylvania State University, University Park, PA 16802, USA
- Artificial Intelligence Research Laboratory, Pennsylvania State University, University Park, PA 16802, USA
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
- Clinical and Translational Sciences Institute, Pennsylvania State University, University Park, PA 16802, USA
- College of Information Sciences & Technology, Pennsylvania State University, University Park, PA 16802, USA
- Institute for Computational and Data Sciences, Pennsylvania State University, University Park, PA 16802, USA
- Center for Big Data Analytics and Discovery Informatics, Pennsylvania State University, University Park, PA 16823, USA
| |
Collapse
|
14
|
Johansson-Åkhe I, Wallner B. Improving peptide-protein docking with AlphaFold-Multimer using forced sampling. FRONTIERS IN BIOINFORMATICS 2022; 2:959160. [PMID: 36304330 PMCID: PMC9580857 DOI: 10.3389/fbinf.2022.959160] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/16/2022] [Indexed: 12/02/2022] Open
Abstract
Protein interactions are key in vital biological processes. In many cases, particularly in regulation, this interaction is between a protein and a shorter peptide fragment. Such peptides are often part of larger disordered regions in other proteins. The flexible nature of peptides enables the rapid yet specific regulation of important functions in cells, such as their life cycle. Consequently, knowledge of the molecular details of peptide-protein interactions is crucial for understanding and altering their function, and many specialized computational methods have been developed to study them. The recent release of AlphaFold and AlphaFold-Multimer has led to a leap in accuracy for the computational modeling of proteins. In this study, the ability of AlphaFold to predict which peptides and proteins interact, as well as its accuracy in modeling the resulting interaction complexes, are benchmarked against established methods. We find that AlphaFold-Multimer predicts the structure of peptide-protein complexes with acceptable or better quality (DockQ ≥0.23) for 66 of the 112 complexes investigated-25 of which were high quality (DockQ ≥0.8). This is a massive improvement on previous methods with 23 or 47 acceptable models and only four or eight high quality models, when using energy-based docking or interaction templates, respectively. In addition, AlphaFold-Multimer can be used to predict whether a peptide and a protein will interact. At 1% false positives, AlphaFold-Multimer found 26% of the possible interactions with a precision of 85%, the best among the methods benchmarked. However, the most interesting result is the possibility of improving AlphaFold by randomly perturbing the neural network weights to force the network to sample more of the conformational space. This increases the number of acceptable models from 66 to 75 and improves the median DockQ from 0.47 to 0.55 (17%) for first ranked models. The best possible DockQ improves from 0.58 to 0.72 (24%), indicating that selecting the best possible model is still a challenge. This scheme of generating more structures with AlphaFold should be generally useful for many applications involving multiple states, flexible regions, and disorder.
Collapse
Affiliation(s)
| | - Björn Wallner
- Division of Bioinformatics, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| |
Collapse
|
15
|
Guo L, He J, Lin P, Huang SY, Wang J. TRScore: a three-dimensional RepVGG-based scoring method for ranking protein docking models. Bioinformatics 2022; 38:2444-2451. [PMID: 35199137 DOI: 10.1093/bioinformatics/btac120] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 01/19/2022] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
MOTIVATION Protein-protein interactions (PPI) play important roles in cellular activities. Due to the technical difficulty and high cost of experimental methods, there are considerable interests towards the development of computational approaches, such as protein docking, to decipher PPI patterns. One of the important and difficult aspects in protein docking is recognizing near-native conformations from a set of decoys, but unfortunately traditional scoring functions still suffer from limited accuracy. Therefore, new scoring methods are pressingly needed in methodological and/or practical implications. RESULTS We present a new deep learning-based scoring method for ranking protein-protein docking models based on a three-dimensional (3D) RepVGG network, named TRScore. To recognize near-native conformations from a set of decoys, TRScore voxelizes the protein-protein interface into a 3D grid labeled by the number of atoms in different physicochemical classes. Benefiting from the deep convolutional RepVGG architecture, TRScore can effectively capture the subtle differences between energetically favorable near-native models and unfavorable non-native decoys without needing extra information. TRScore was extensively evaluated on diverse test sets including protein-protein docking benchmark 5.0 update set, DockGround decoy set, as well as realistic CAPRI decoy set, and overall obtained a significant improvement over existing methods in cross validation and independent evaluations. AVAILABILITY Codes available at: https://github.com/BioinformaticsCSU/TRScore.
Collapse
Affiliation(s)
- Linyuan Guo
- School of Computer Science, Central South University, Changsha, Hunan 410083, China
| | - Jiahua He
- School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Peicong Lin
- School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Sheng-You Huang
- School of Physics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Jianxin Wang
- School of Computer Science, Central South University, Changsha, Hunan 410083, China
| |
Collapse
|
16
|
Capturing a Crucial ‘Disorder-to-Order Transition’ at the Heart of the Coronavirus Molecular Pathology—Triggered by Highly Persistent, Interchangeable Salt-Bridges. Vaccines (Basel) 2022; 10:vaccines10020301. [PMID: 35214759 PMCID: PMC8875383 DOI: 10.3390/vaccines10020301] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/27/2022] [Accepted: 02/05/2022] [Indexed: 02/05/2023] Open
Abstract
The COVID-19 origin debate has greatly been influenced by genome comparison studies of late, revealing the emergence of the Furin-like cleavage site at the S1/S2 junction of the SARS-CoV-2 Spike (FLCSSpike) containing its 681PRRAR685 motif, absent in other related respiratory viruses. Being the rate-limiting (i.e., the slowest) step, the host Furin cleavage is instrumental in the abrupt increase in transmissibility in COVID-19, compared to earlier onsets of respiratory viral diseases. In such a context, the current paper entraps a ‘disorder-to-order transition’ of the FLCSSpike (concomitant to an entropy arrest) upon binding to Furin. The interaction clearly seems to be optimized for a more efficient proteolytic cleavage in SARS-CoV-2. The study further shows the formation of dynamically interchangeable and persistent networks of salt-bridges at the Spike–Furin interface in SARS-CoV-2 involving the three arginines (R682, R683, R685) of the FLCSSpike with several anionic residues (E230, E236, D259, D264, D306) coming from Furin, strategically distributed around its catalytic triad. Multiplicity and structural degeneracy of plausible salt-bridge network archetypes seem to be the other key characteristic features of the Spike–Furin binding in SARS-CoV-2, allowing the system to breathe—a trademark of protein disorder transitions. Interestingly, with respect to the homologous interaction in SARS-CoV (2002/2003) taken as a baseline, the Spike–Furin binding events, generally, in the coronavirus lineage, seems to have preference for ionic bond formation, even with a lesser number of cationic residues at their potentially polybasic FLCSSpike patches. The interaction energies are suggestive of characteristic metastabilities attributed to Spike–Furin interactions, generally to the coronavirus lineage, which appears to be favorable for proteolytic cleavages targeted at flexible protein loops. The current findings not only offer novel mechanistic insights into the coronavirus molecular pathology and evolution, but also add substantially to the existing theories of proteolytic cleavages.
Collapse
|
17
|
Depetris RS, Lu D, Polonskaya Z, Zhang Z, Luna X, Tankard A, Kolahi P, Drummond M, Williams C, Ebert MCCJC, Patel JP, Poyurovsky MV. Functional antibody characterization via direct structural analysis and information-driven protein-protein docking. Proteins 2021; 90:919-935. [PMID: 34773424 PMCID: PMC9544432 DOI: 10.1002/prot.26280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 08/28/2021] [Accepted: 11/07/2021] [Indexed: 12/02/2022]
Abstract
Detailed description of the mechanism of action of the therapeutic antibodies is essential for the functional characterization and future optimization of potential clinical agents. We recently developed KD035, a fully human antibody targeting vascular endothelial growth factor receptor 2 (VEGFR2). KD035 blocked VEGF‐A, and VEGF‐C‐mediated VEGFR2 activation, as demonstrated by the in vitro binding and competition assays and functional cellular assays. Here, we report a computational model of the complex between the variable fragment of KD035 (KD035(Fv)) and the domains 2 and 3 of the extracellular portion of VEGFR2 (VEGFR2(D2‐3)). Our modeling was guided by a priori experimental information including the X‐ray structures of KD035 and related antibodies, binding assays, target domain mapping and comparison of KD035 affinity for VEGFR2 from different species. The accuracy of the model was assessed by molecular dynamics simulations, and subsequently validated by mutagenesis and binding analysis. Importantly, the steps followed during the generation of this model can set a precedent for future in silico efforts aimed at the accurate description of the antibody–antigen and more broadly protein–protein complexes.
Collapse
Affiliation(s)
| | - Dan Lu
- Kadmon Corporation, LLC, New York, New York, USA
| | | | - Zhikai Zhang
- Kadmon Corporation, LLC, New York, New York, USA
| | - Xenia Luna
- Kadmon Corporation, LLC, New York, New York, USA
| | | | - Pegah Kolahi
- Kadmon Corporation, LLC, New York, New York, USA
| | | | | | | | | | | |
Collapse
|
18
|
Johansson-Åkhe I, Mirabello C, Wallner B. InterPepRank: Assessment of Docked Peptide Conformations by a Deep Graph Network. FRONTIERS IN BIOINFORMATICS 2021; 1:763102. [PMID: 36303778 PMCID: PMC9581042 DOI: 10.3389/fbinf.2021.763102] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 10/05/2021] [Indexed: 11/13/2022] Open
Abstract
Peptide-protein interactions between a smaller or disordered peptide stretch and a folded receptor make up a large part of all protein-protein interactions. A common approach for modeling such interactions is to exhaustively sample the conformational space by fast-Fourier-transform docking, and then refine a top percentage of decoys. Commonly, methods capable of ranking the decoys for selection fast enough for larger scale studies rely on first-principle energy terms such as electrostatics, Van der Waals forces, or on pre-calculated statistical potentials. We present InterPepRank for peptide-protein complex scoring and ranking. InterPepRank is a machine learning-based method which encodes the structure of the complex as a graph; with physical pairwise interactions as edges and evolutionary and sequence features as nodes. The graph network is trained to predict the LRMSD of decoys by using edge-conditioned graph convolutions on a large set of peptide-protein complex decoys. InterPepRank is tested on a massive independent test set with no targets sharing CATH annotation nor 30% sequence identity with any target in training or validation data. On this set, InterPepRank has a median AUC of 0.86 for finding coarse peptide-protein complexes with LRMSD < 4Å. This is an improvement compared to other state-of-the-art ranking methods that have a median AUC between 0.65 and 0.79. When included as a selection-method for selecting decoys for refinement in a previously established peptide docking pipeline, InterPepRank improves the number of medium and high quality models produced by 80% and 40%, respectively. The InterPepRank program as well as all scripts for reproducing and retraining it are available from: http://wallnerlab.org/InterPepRank.
Collapse
|
19
|
MAP4K4 expression in cardiomyocytes: multiple isoforms, multiple phosphorylations and interactions with striatins. Biochem J 2021; 478:2121-2143. [PMID: 34032269 PMCID: PMC8203206 DOI: 10.1042/bcj20210003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/21/2021] [Accepted: 05/25/2021] [Indexed: 12/02/2022]
Abstract
The Ser/Thr kinase MAP4K4, like other GCKIV kinases, has N-terminal kinase and C-terminal citron homology (CNH) domains. MAP4K4 can activate c-Jun N-terminal kinases (JNKs), and studies in the heart suggest it links oxidative stress to JNKs and heart failure. In other systems, MAP4K4 is regulated in striatin-interacting phosphatase and kinase (STRIPAK) complexes, in which one of three striatins tethers PP2A adjacent to a kinase to keep it dephosphorylated and inactive. Our aim was to understand how MAP4K4 is regulated in cardiomyocytes. The rat MAP4K4 gene was not properly defined. We identified the first coding exon of the rat gene using 5′-RACE, we cloned the full-length sequence and confirmed alternative-splicing of MAP4K4 in rat cardiomyocytes. We identified an additional α-helix C-terminal to the kinase domain important for kinase activity. In further studies, FLAG-MAP4K4 was expressed in HEK293 cells or cardiomyocytes. The Ser/Thr protein phosphatase inhibitor calyculin A (CalA) induced MAP4K4 hyperphosphorylation, with phosphorylation of the activation loop and extensive phosphorylation of the linker between the kinase and CNH domains. This required kinase activity. MAP4K4 associated with myosin in untreated cardiomyocytes, and this was lost with CalA-treatment. FLAG-MAP4K4 associated with all three striatins in cardiomyocytes, indicative of regulation within STRIPAK complexes and consistent with activation by CalA. Computational analysis suggested the interaction was direct and mediated via coiled-coil domains. Surprisingly, FLAG-MAP4K4 inhibited JNK activation by H2O2 in cardiomyocytes and increased myofibrillar organisation. Our data identify MAP4K4 as a STRIPAK-regulated kinase in cardiomyocytes, and suggest it regulates the cytoskeleton rather than activates JNKs.
Collapse
|
20
|
Basu S, Chakravarty D, Bhattacharyya D, Saha P, Patra HK. Plausible blockers of Spike RBD in SARS-CoV2-molecular design and underlying interaction dynamics from high-level structural descriptors. J Mol Model 2021; 27:191. [PMID: 34057647 PMCID: PMC8165686 DOI: 10.1007/s00894-021-04779-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 04/26/2021] [Indexed: 12/24/2022]
Abstract
Abstract COVID-19 is characterized by an unprecedented abrupt increase in the viral transmission rate (SARS-CoV-2) relative to its pandemic evolutionary ancestor, SARS-CoV (2003). The complex molecular cascade of events related to the viral pathogenicity is triggered by the Spike protein upon interacting with the ACE2 receptor on human lung cells through its receptor binding domain (RBDSpike). One potential therapeutic strategy to combat COVID-19 could thus be limiting the infection by blocking this key interaction. In this current study, we adopt a protein design approach to predict and propose non-virulent structural mimics of the RBDSpike which can potentially serve as its competitive inhibitors in binding to ACE2. The RBDSpike is an independently foldable protein domain, resilient to conformational changes upon mutations and therefore an attractive target for strategic re-design. Interestingly, in spite of displaying an optimal shape fit between their interacting surfaces (attributed to a consequently high mutual affinity), the RBDSpike–ACE2 interaction appears to have a quasi-stable character due to a poor electrostatic match at their interface. Structural analyses of homologous protein complexes reveal that the ACE2 binding site of RBDSpike has an unusually high degree of solvent-exposed hydrophobic residues, attributed to key evolutionary changes, making it inherently “reaction-prone.” The designed mimics aimed to block the viral entry by occupying the available binding sites on ACE2, are tested to have signatures of stable high-affinity binding with ACE2 (cross-validated by appropriate free energy estimates), overriding the native quasi-stable feature. The results show the apt of directly adapting natural examples in rational protein design, wherein, homology-based threading coupled with strategic “hydrophobic ↔ polar” mutations serve as a potential breakthrough. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1007/s00894-021-04779-0.
Collapse
Affiliation(s)
- Sankar Basu
- Department of Microbiology, Asutosh College (affiliated to University of Calcutta), Kolkata, 700026, West Bengal, India.
| | - Devlina Chakravarty
- Department of Chemistry, University of Rutgers-Camden, Camden, 08102, NJ, USA
| | - Dhananjay Bhattacharyya
- Computational Science Division, Saha Institute of Nuclear Physics, Kolkata, 700064, West Bengal, India
| | - Pampa Saha
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Hirak K Patra
- Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London, London, NW3 2PF, UK
| |
Collapse
|
21
|
Sotudian S, Desta IT, Hashemi N, Zarbafian S, Kozakov D, Vakili P, Vajda S, Paschalidis IC. Improved cluster ranking in protein-protein docking using a regression approach. Comput Struct Biotechnol J 2021; 19:2269-2278. [PMID: 33995918 PMCID: PMC8102165 DOI: 10.1016/j.csbj.2021.04.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 11/21/2022] Open
Abstract
We develop a Regression-based Ranking by Pairwise Cluster Comparisons (RRPCC) method to rank clusters of similar protein complex conformations generated by an underlying docking program. The method leverages robust regression to predict the relative quality difference between any pair or clusters and combines these pairwise assessments to form a ranked list of clusters, from higher to lower quality. We apply RRPCC to clusters produced by the automated docking server ClusPro and, depending on the training/validation strategy, we show improvement by 24-100% in ranking acceptable or better quality clusters first, and by 15-100% in ranking medium or better quality clusters first. We compare the RRPCC-ClusPro combination to a number of alternatives, and show that very different machine learning approaches to scoring docked structures yield similar success rates. Finally, we discuss the current limitations on sampling and scoring, looking ahead to further improvements. Interestingly, some features important for improved scoring are internal energy terms that occur only due to the local energy minimization applied in the refinement stage following rigid body docking.
Collapse
Affiliation(s)
| | | | - Nasser Hashemi
- Division of Systems Engineering, Boston University, Boston, USA
| | | | - Dima Kozakov
- Laufer Center for Physical and Quantitative Biology, Institute for Advanced Computational Sciences, Stony Brook University, Stony Brook, USA
| | - Pirooz Vakili
- Division of Systems Engineering, Boston University, Boston, USA
| | - Sandor Vajda
- Department of Biomedical Engineering, Boston University
- Department of Chemistry, Boston University
| | - Ioannis Ch. Paschalidis
- Division of Systems Engineering, Boston University, Boston, USA
- Department of Biomedical Engineering, Boston University
- Department of Electrical & Computer Engineering, and Faculty for Computing & Data Sciences, Boston University
| |
Collapse
|
22
|
Alnomasy SF. Virus-receptor interactions of SARS-CoV-2 spikereceptor-binding domain and human neuropilin-1 b1 domain. Saudi J Biol Sci 2021; 28:3926-3928. [PMID: 33850424 PMCID: PMC8032480 DOI: 10.1016/j.sjbs.2021.03.074] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/28/2021] [Accepted: 03/30/2021] [Indexed: 12/13/2022] Open
Abstract
In late 2019, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in Wahan, China and it causes disease which is known as COVID-19. This infection spreads everywhere in the world, and it leads to an enormous number of death among individuals. The mystery issue about SARS-CoV-2 that appears not to have functions of a hemagglutinin and neuraminidase like other coronaviruses. Angiotensin-converting enzyme 2 (ACE2) is the main surface receptor for entering SARS-CoV-2 into the host cell. This entry process is mediated by binding the SARS-CoV-2 spike receptor-binding domain (RBD) to ACE2. Recently, researchers discover a new receptor responsible for the SARS-CoV-2 entry which is neuropilin-1 (NRP1). So, this work provides afford a knowledge of how the initial interaction between SARS-CoV-2 spike RBD and NRP1 b1 domain may occur. Understanding this interaction would be very necessary for drug design against SARS-CoV-2.
Collapse
Affiliation(s)
- Sultan F Alnomasy
- Department of Medical Laboratories Sciences, College of Applied Medical Sciences in Al-Quwayiyah, Shaqra University, Al-Quwayiyah, Riyadh, Saudi Arabia
| |
Collapse
|
23
|
Johansson-Åkhe I, Mirabello C, Wallner B. InterPep2: global peptide-protein docking using interaction surface templates. Bioinformatics 2020; 36:2458-2465. [PMID: 31917413 PMCID: PMC7178396 DOI: 10.1093/bioinformatics/btaa005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 12/16/2019] [Accepted: 01/03/2020] [Indexed: 12/23/2022] Open
Abstract
Motivation Interactions between proteins and peptides or peptide-like intrinsically disordered regions are involved in many important biological processes, such as gene expression and cell life-cycle regulation. Experimentally determining the structure of such interactions is time-consuming and difficult because of the inherent flexibility of the peptide ligand. Although several prediction-methods exist, most are limited in performance or availability. Results InterPep2 is a freely available method for predicting the structure of peptide–protein interactions. Improved performance is obtained by using templates from both peptide–protein and regular protein–protein interactions, and by a random forest trained to predict the DockQ-score for a given template using sequence and structural features. When tested on 252 bound peptide–protein complexes from structures deposited after the complexes used in the construction of the training and templates sets of InterPep2, InterPep2-Refined correctly positioned 67 peptides within 4.0 Å LRMSD among top10, similar to another state-of-the-art template-based method which positioned 54 peptides correctly. However, InterPep2 displays a superior ability to evaluate the quality of its own predictions. On a previously established set of 27 non-redundant unbound-to-bound peptide–protein complexes, InterPep2 performs on-par with leading methods. The extended InterPep2-Refined protocol managed to correctly model 15 of these complexes within 4.0 Å LRMSD among top10, without using templates from homologs. In addition, combining the template-based predictions from InterPep2 with ab initio predictions from PIPER-FlexPepDock resulted in 22% more near-native predictions compared to the best single method (22 versus 18). Availability and implementation The program is available from: http://wallnerlab.org/InterPep2. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Isak Johansson-Åkhe
- Division of Bioinformatics, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Claudio Mirabello
- Division of Bioinformatics, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Björn Wallner
- Division of Bioinformatics, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| |
Collapse
|
24
|
Tanemura KA, Pei J, Merz KM. Refinement of pairwise potentials via logistic regression to score protein-protein interactions. Proteins 2020; 88:1559-1568. [PMID: 32729132 DOI: 10.1002/prot.25973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/17/2020] [Accepted: 06/14/2020] [Indexed: 12/20/2022]
Abstract
Protein-protein interactions (PPIs) are ubiquitous and functionally of great importance in biological systems. Hence, the accurate prediction of PPIs by protein-protein docking and scoring tools is highly desirable in order to characterize their structure and biological function. Ab initio docking protocols are divided into the sampling of docking poses to produce at least one near-native structure, and then to evaluate the vast candidate structures by scoring. Concurrent development in both sampling and scoring is crucial for the deployment of protein-protein docking software. In the present work, we apply a machine learning model on pairwise potentials to refine the task of protein quaternary structure native structure detection among decoys. A decoy set was featurized using the Knowledge and Empirical Combined Scoring Algorithm 2 (KECSA2) pairwise potential. The highly unbalanced decoy set was then balanced using a comparison concept between native and decoy structures. The resultant comparison descriptors were used to train a logistic regression (LR) classifier. The LR model yielded the optimal performance for native detection among decoys compared with conventional scoring functions, while exhibiting lesser performance for the detection of low root mean square deviation decoy structures. Its deployment on an independent benchmark set confirms that the scoring function performs competitively relative to other scoring functions. The scripts used are available at https://github.com/TanemuraKiyoto/PPI-native-detection-via-LR.
Collapse
Affiliation(s)
- Kiyoto A Tanemura
- Department of Chemistry, Michigan State University, East Lansing, Michigan, USA
| | - Jun Pei
- Department of Chemistry, Michigan State University, East Lansing, Michigan, USA
| | - Kenneth M Merz
- Department of Chemistry, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
25
|
Kaku Y, Kuwata T, Gorny MK, Matsushita S. Prediction of Contact Residues in Anti-HIV Neutralizing Antibody by Deep Learning. Jpn J Infect Dis 2020; 73:235-241. [PMID: 32009060 DOI: 10.7883/yoken.jjid.2019.496] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The monoclonal antibody 1C10 targets the V3 loop of HIV-1 and neutralizes a broad range of clade B viruses. However, the mode of interaction between 1C10 and the V3 loop remains unclear because crystallization of 1C10 and the V3 peptide was unsuccessful due to the flexible regions present in both 1C10 and the V3 peptide. In this study, we predicted the 1C10 amino acid residues that make contact with the V3 loop using a deep learning (DL)-based method. Inputs from ROSIE for docking simulation and FastContact, Naccess, and PDBtools, to approximate interactions were processed by Chainer for DL, and outputs were obtained as probabilities of contact residues. Using this DL algorithm, D95, D97, P100a, and D100b of CDRH3; D53, and D56 of CDRH2; and D61 of FR3 were highly ranked as contact residues of 1C10. Substitution of these residues with alanine significantly decreased the affinity of 1C10 to the V3 peptide. Moreover, the higher the rank of the residue, the more the binding activity diminished. This study demonstrates that the prediction of contact residues using a DL-based approach is a precise and useful tool for the analysis of antibody-antigen interactions.
Collapse
Affiliation(s)
- Yu Kaku
- Clinical Retrovirology, Joint Research Center for Human Retrovirus Infection, Kumamoto University
| | - Takeo Kuwata
- Clinical Retrovirology, Joint Research Center for Human Retrovirus Infection, Kumamoto University
| | | | - Shuzo Matsushita
- Clinical Retrovirology, Joint Research Center for Human Retrovirus Infection, Kumamoto University
| |
Collapse
|
26
|
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
|
27
|
James N, Shanthi V, Ramanathan K. Density Functional Theory and Molecular Simulation Studies for Prioritizing Anaplastic Lymphoma Kinase Inhibitors. Appl Biochem Biotechnol 2019; 190:1127-1146. [PMID: 31712989 DOI: 10.1007/s12010-019-03156-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/23/2019] [Indexed: 12/12/2022]
Abstract
Targeting anaplastic lymphoma kinase (ALK) is one of the important treatment strategies for the treatment of non-small cell lung cancer (NSCLC). In the present perspective, multidimensional approaches were used for the identification of ALK inhibitors. Initially, an e-pharmacophore model was generated using the PHASE algorithm and was used as a 3D query to screen 468,200 molecules of ASINEX database. Prior to the screening process, the model was evaluated for its significance and the ability to differentiate actives from inactives, using enrichment analysis. Subsequently, the hierarchical docking protocol and binding free energy calculations were instigated using GLIDE algorithm and Prime module, respectively. Further, the pharmacokinetic/pharmacodynamics (PK/PD) properties and toxicities of the hit compounds were envisaged respectively using QikProp program, Osiris explorer, and Protox-II algorithm. These approaches retrieved two hits namely BAS 00137817 and BAS 00680055 with acceptable absorption, distribution, metabolism, excretion and toxicity (ADMET) properties and higher affinity towards ALK protein. Additionally, density functional theory calculations and molecular dynamics simulations were performed to validate the inhibitory activity of the lead compounds. It is noteworthy to mention that all the hits constitute of particular scaffolds which play a major role in the downregulation of some ALK-positive lung cancer pathways. We speculate that the outcomes of this research are of substantial prominence in the rational designing of novel and efficacious ALK inhibitors.
Collapse
Affiliation(s)
- Nivya James
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - V Shanthi
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - K Ramanathan
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
28
|
Turki T, Wei Z, Wang JTL. A transfer learning approach via procrustes analysis and mean shift for cancer drug sensitivity prediction. J Bioinform Comput Biol 2019; 16:1840014. [PMID: 29945499 DOI: 10.1142/s0219720018400140] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Transfer learning (TL) algorithms aim to improve the prediction performance in a target task (e.g. the prediction of cisplatin sensitivity in triple-negative breast cancer patients) via transferring knowledge from auxiliary data of a related task (e.g. the prediction of docetaxel sensitivity in breast cancer patients), where the distribution and even the feature space of the data pertaining to the tasks can be different. In real-world applications, we sometimes have a limited training set in a target task while we have auxiliary data from a related task. To obtain a better prediction performance in the target task, supervised learning requires a sufficiently large training set in the target task to perform well in predicting future test examples of the target task. In this paper, we propose a TL approach for cancer drug sensitivity prediction, where our approach combines three techniques. First, we shift the representation of a subset of examples from auxiliary data of a related task to a representation closer to a target training set of a target task. Second, we align the shifted representation of the selected examples of the auxiliary data to the target training set to obtain examples with representation aligned to the target training set. Third, we train machine learning algorithms using both the target training set and the aligned examples. We evaluate the performance of our approach against baseline approaches using the Area Under the receiver operating characteristic (ROC) Curve (AUC) on real clinical trial datasets pertaining to multiple myeloma, nonsmall cell lung cancer, triple-negative breast cancer, and breast cancer. Experimental results show that our approach is better than the baseline approaches in terms of performance and statistical significance.
Collapse
Affiliation(s)
- Turki Turki
- * Department of Computer Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Zhi Wei
- † Department of Computer Science, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Jason T L Wang
- † Department of Computer Science, New Jersey Institute of Technology, Newark, NJ 07102, USA
| |
Collapse
|
29
|
Characterization of phthalate reductase from Ralstonia eutropha CH34 and in silico study of phthalate dioxygenase and phthalate reductase interaction. J Mol Graph Model 2019; 90:161-170. [DOI: 10.1016/j.jmgm.2019.05.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 05/03/2019] [Accepted: 05/03/2019] [Indexed: 12/17/2022]
|
30
|
Ece A. Towards more effective acetylcholinesterase inhibitors: a comprehensive modelling study based on human acetylcholinesterase protein–drug complex. J Biomol Struct Dyn 2019; 38:565-572. [DOI: 10.1080/07391102.2019.1583606] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Abdulilah Ece
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Biruni University, Istanbul, Turkey
| |
Collapse
|
31
|
Inner-View of Nanomaterial Incited Protein Conformational Changes: Insights into Designable Interaction. RESEARCH 2018; 2018:9712832. [PMID: 31549040 PMCID: PMC6750102 DOI: 10.1155/2018/9712832] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 08/16/2018] [Indexed: 12/19/2022]
Abstract
Nanoparticle bioreactivity critically depends upon interaction between proteins and nanomaterials (NM). The formation of the "protein corona" (PC) is the effect of such nanoprotein interactions. PC has a wide usage in pharmaceuticals, drug delivery, medicine, and industrial biotechnology. Therefore, a detailed in-vitro, in-vivo, and in-silico understanding of nanoprotein interaction is fundamental and has a genuine contemporary appeal. NM surfaces can modify the protein conformation during interaction, or NMs themselves can lead to self-aggregations. Both phenomena can change the whole downstream bioreactivity of the concerned nanosystem. The main aim of this review is to understand the mechanistic view of NM-protein interaction and recapitulate the underlying physical chemistry behind the formation of such complicated macromolecular assemblies, to provide a critical overview of the different models describing NM induced structural and functional modification of proteins. The review also attempts to point out the current limitation in understanding the field and highlights the future scopes, involving a plausible proposition of how artificial intelligence could be aided to explore such systems for the prediction and directed design of the desired NM-protein interactions.
Collapse
|
32
|
Shape complementarity at protein interfaces via global docking optimisation. J Mol Graph Model 2018; 84:69-73. [DOI: 10.1016/j.jmgm.2018.06.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 06/11/2018] [Accepted: 06/12/2018] [Indexed: 11/24/2022]
|
33
|
Basu S, Biswas P. Salt-bridge dynamics in intrinsically disordered proteins: A trade-off between electrostatic interactions and structural flexibility. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2018; 1866:624-641. [DOI: 10.1016/j.bbapap.2018.03.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 02/13/2018] [Accepted: 03/07/2018] [Indexed: 12/29/2022]
|
34
|
Borges NM, Sartori GR, Ribeiro JFR, Rocha JR, Martins JBL, Montanari CA, Gargano R. Similarity search combined with docking and molecular dynamics for novel hAChE inhibitor scaffolds. J Mol Model 2018; 24:41. [PMID: 29332299 DOI: 10.1007/s00894-017-3548-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 11/27/2017] [Indexed: 12/31/2022]
Abstract
The main purpose of this study was to address the performance of virtual screening methods based on ligands and the protein structure of acetylcholinesterase (AChE) in order to retrieve novel human AChE (hAChE) inhibitors. In addition, a protocol was developed to identify novel hit compounds and propose new promising AChE inhibitors from the ZINC database with 10 million commercially available compounds. In this sense, 3D similarity searches using rapid overlay of chemical structures and similarity analysis through comparison of electrostatic overlay of docked hits were used to retrieve AChE inhibitors from collected databases. Molecular dynamics simulation of 100 ns was carried out to study the best docked compounds from similarity searches. Some key residues were identified as crucial for the dual binding mode of inhibitor with the interaction site. All results indicated the relevant use of EON and docking strategy for identifying novel hit compounds as promising potential anticholinesterase candidates, and seven new structures were selected as potential hAChE inhibitors. Graphical abstract Compound N01 in the 4M0E hAChE crystallography structure from docking results. Yellow dashed lines Hydrogen bonds, blue dashed lines π-stacking interactions, green dashed lines cation-π interactions.
Collapse
Affiliation(s)
| | | | - Jean F R Ribeiro
- Institute of Chemistry of São Carlos, University of São Paulo, São Carlos, SP, Brazil
| | - Josmar R Rocha
- Institute of Chemistry of São Carlos, University of São Paulo, São Carlos, SP, Brazil
| | - João B L Martins
- Institute of Chemistry, University of Brasilia, Brasilia, DF, Brazil
| | - Carlos A Montanari
- Institute of Chemistry of São Carlos, University of São Paulo, São Carlos, SP, Brazil
| | - Ricardo Gargano
- Institute of Physics, University of Brasilia, Brasilia, DF, Brazil
| |
Collapse
|
35
|
Basu S. CP dock: the complementarity plot for docking of proteins: implementing multi-dielectric continuum electrostatics. J Mol Model 2017; 24:8. [PMID: 29218430 DOI: 10.1007/s00894-017-3546-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Accepted: 11/24/2017] [Indexed: 01/18/2023]
Abstract
The complementarity plot (CP) is an established validation tool for protein structures, applicable to both globular proteins (folding) as well as protein-protein complexes (binding). It computes the shape and electrostatic complementarities (Sm, Em) for amino acid side-chains buried within the protein interior or interface and plots them in a two-dimensional plot having knowledge-based probabilistic quality estimates for the residues as well as for the whole structure. The current report essentially presents an upgraded version of the plot with the implementation of the advanced multi-dielectric functionality (as in Delphi version 6.2 or higher) in the computation of electrostatic complementarity to make the validation tool physico-chemically more realistic. The two methods (single- and multi-dielectric) agree decently in their resultant Em values, and hence, provisions for both methods have been kept in the software suite. So to speak, the global electrostatic balance within a well-folded protein and/or a well-packed interface seems only marginally perturbed by the choice of different internal dielectric values. However, both from theoretical as well as practical grounds, the more advanced multi-dielectric version of the plot is certainly recommended for potentially producing more reliable results. The report also presents a new methodology and a variant plot, namely CPdock, based on the same principles of complementarity specifically designed to be used in the docking of proteins. The efficacy of the method to discriminate between good and bad docked protein complexes has been tested on a recent state-of-the-art docking benchmark. The results unambiguously indicate that CPdock can indeed be effective in the initial screening phase of a docking scoring pipeline before going into more sophisticated and computationally expensive scoring functions. CPdock has been made available at https://github.com/nemo8130/CPdock . Graphical Abstract An example showing the efficacy of CPdock to be used in the initial screening phase of a protein-protein docking scoring pipeline.
Collapse
Affiliation(s)
- Sankar Basu
- Department of Chemistry, University of Delhi, New Delhi, India.
| |
Collapse
|
36
|
Salt-bridge networks within globular and disordered proteins: characterizing trends for designable interactions. J Mol Model 2017. [PMID: 28626846 DOI: 10.1007/s00894-017-3376-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
There has been considerable debate about the contribution of salt bridges to the stabilization of protein folds, in spite of their participation in crucial protein functions. Salt bridges appear to contribute to the activity-stability trade-off within proteins by bringing high-entropy charged amino acids into close contacts during the course of their functions. The current study analyzes the modes of association of salt bridges (in terms of networks) within globular proteins and at protein-protein interfaces. While the most common and trivial type of salt bridge is the isolated salt bridge, bifurcated salt bridge appears to be a distinct salt-bridge motif having a special topology and geometry. Bifurcated salt bridges are found ubiquitously in proteins and interprotein complexes. Interesting and attractive examples presenting different modes of interaction are highlighted. Bifurcated salt bridges appear to function as molecular clips that are used to stitch together large surface contours at interacting protein interfaces. The present work also emphasizes the key role of salt-bridge-mediated interactions in the partial folding of proteins containing long stretches of disordered regions. Salt-bridge-mediated interactions seem to be pivotal to the promotion of "disorder-to-order" transitions in small disordered protein fragments and their stabilization upon binding. The results obtained in this work should help to guide efforts to elucidate the modus operandi of these partially disordered proteins, and to conceptualize how these proteins manage to maintain the required amount of disorder even in their bound forms. This work could also potentially facilitate explorations of geometrically specific designable salt bridges through the characterization of composite salt-bridge networks. Graphical abstract ᅟ.
Collapse
|
37
|
Basu S, Söderquist F, Wallner B. Proteus: a random forest classifier to predict disorder-to-order transitioning binding regions in intrinsically disordered proteins. J Comput Aided Mol Des 2017; 31:453-466. [PMID: 28365882 PMCID: PMC5429364 DOI: 10.1007/s10822-017-0020-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 03/24/2017] [Indexed: 12/03/2022]
Abstract
The focus of the computational structural biology community has taken a dramatic shift over the past one-and-a-half decades from the classical protein structure prediction problem to the possible understanding of intrinsically disordered proteins (IDP) or proteins containing regions of disorder (IDPR). The current interest lies in the unraveling of a disorder-to-order transitioning code embedded in the amino acid sequences of IDPs/IDPRs. Disordered proteins are characterized by an enormous amount of structural plasticity which makes them promiscuous in binding to different partners, multi-functional in cellular activity and atypical in folding energy landscapes resembling partially folded molten globules. Also, their involvement in several deadly human diseases (e.g. cancer, cardiovascular and neurodegenerative diseases) makes them attractive drug targets, and important for a biochemical understanding of the disease(s). The study of the structural ensemble of IDPs is rather difficult, in particular for transient interactions. When bound to a structured partner, an IDPR adapts an ordered conformation in the complex. The residues that undergo this disorder-to-order transition are called protean residues, generally found in short contiguous stretches and the first step in understanding the modus operandi of an IDP/IDPR would be to predict these residues. There are a few available methods which predict these protean segments from their amino acid sequences; however, their performance reported in the literature leaves clear room for improvement. With this background, the current study presents ‘Proteus’, a random forest classifier that predicts the likelihood of a residue undergoing a disorder-to-order transition upon binding to a potential partner protein. The prediction is based on features that can be calculated using the amino acid sequence alone. Proteus compares favorably with existing methods predicting twice as many true positives as the second best method (55 vs. 27%) with a much higher precision on an independent data set. The current study also sheds some light on a possible ‘disorder-to-order’ transitioning consensus, untangled, yet embedded in the amino acid sequence of IDPs. Some guidelines have also been suggested for proceeding with a real-life structural modeling involving an IDPR using Proteus.
Collapse
Affiliation(s)
- Sankar Basu
- Bioinformatics Division, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden.,Department of Biochemistry, University of Calcutta, Kolkata, 700019, India
| | - Fredrik Söderquist
- Bioinformatics Division, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Björn Wallner
- Bioinformatics Division, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden. .,Swedish e-Science Research Center, Linköping University, Linköping, Sweden.
| |
Collapse
|
38
|
Peterson LX, Kim H, Esquivel-Rodriguez J, Roy A, Han X, Shin WH, Zhang J, Terashi G, Lee M, Kihara D. Human and server docking prediction for CAPRI round 30-35 using LZerD with combined scoring functions. Proteins 2017; 85:513-527. [PMID: 27654025 PMCID: PMC5313330 DOI: 10.1002/prot.25165] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 09/09/2016] [Accepted: 09/15/2016] [Indexed: 12/12/2022]
Abstract
We report the performance of protein-protein docking predictions by our group for recent rounds of the Critical Assessment of Prediction of Interactions (CAPRI), a community-wide assessment of state-of-the-art docking methods. Our prediction procedure uses a protein-protein docking program named LZerD developed in our group. LZerD represents a protein surface with 3D Zernike descriptors (3DZD), which are based on a mathematical series expansion of a 3D function. The appropriate soft representation of protein surface with 3DZD makes the method more tolerant to conformational change of proteins upon docking, which adds an advantage for unbound docking. Docking was guided by interface residue prediction performed with BindML and cons-PPISP as well as literature information when available. The generated docking models were ranked by a combination of scoring functions, including PRESCO, which evaluates the native-likeness of residues' spatial environments in structure models. First, we discuss the overall performance of our group in the CAPRI prediction rounds and investigate the reasons for unsuccessful cases. Then, we examine the performance of several knowledge-based scoring functions and their combinations for ranking docking models. It was found that the quality of a pool of docking models generated by LZerD, that is whether or not the pool includes near-native models, can be predicted by the correlation of multiple scores. Although the current analysis used docking models generated by LZerD, findings on scoring functions are expected to be universally applicable to other docking methods. Proteins 2017; 85:513-527. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lenna X. Peterson
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Hyungrae Kim
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | | | - Amitava Roy
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA
- Bioinformatics and Computational Biosciences Branch, Rocky Mountain Laboratories, NIAID, National Institutes of Health, Hamilton, Montana 59840, USA
| | - Xusi Han
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Woong-Hee Shin
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Jian Zhang
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Genki Terashi
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
- School of Pharmacy, Kitasato University, Minato-Ku, Tokyo, 108-8641, Japan
| | - Matt Lee
- Lilly Biotechnology Center San Diego, 10300 Campus Point Drive, San Diego, CA, 92121, USA
| | - Daisuke Kihara
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
- Department of Computer Science, Purdue University, West Lafayette, IN, 47907, USA
| |
Collapse
|
39
|
Basu S, Wallner B. DockQ: A Quality Measure for Protein-Protein Docking Models. PLoS One 2016; 11:e0161879. [PMID: 27560519 PMCID: PMC4999177 DOI: 10.1371/journal.pone.0161879] [Citation(s) in RCA: 226] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 08/12/2016] [Indexed: 01/26/2023] Open
Abstract
The state-of-the-art to assess the structural quality of docking models is currently based on three related yet independent quality measures: Fnat, LRMS, and iRMS as proposed and standardized by CAPRI. These quality measures quantify different aspects of the quality of a particular docking model and need to be viewed together to reveal the true quality, e.g. a model with relatively poor LRMS (>10Å) might still qualify as 'acceptable' with a descent Fnat (>0.50) and iRMS (<3.0Å). This is also the reason why the so called CAPRI criteria for assessing the quality of docking models is defined by applying various ad-hoc cutoffs on these measures to classify a docking model into the four classes: Incorrect, Acceptable, Medium, or High quality. This classification has been useful in CAPRI, but since models are grouped in only four bins it is also rather limiting, making it difficult to rank models, correlate with scoring functions or use it as target function in machine learning algorithms. Here, we present DockQ, a continuous protein-protein docking model quality measure derived by combining Fnat, LRMS, and iRMS to a single score in the range [0, 1] that can be used to assess the quality of protein docking models. By using DockQ on CAPRI models it is possible to almost completely reproduce the original CAPRI classification into Incorrect, Acceptable, Medium and High quality. An average PPV of 94% at 90% Recall demonstrating that there is no need to apply predefined ad-hoc cutoffs to classify docking models. Since DockQ recapitulates the CAPRI classification almost perfectly, it can be viewed as a higher resolution version of the CAPRI classification, making it possible to estimate model quality in a more quantitative way using Z-scores or sum of top ranked models, which has been so valuable for the CASP community. The possibility to directly correlate a quality measure to a scoring function has been crucial for the development of scoring functions for protein structure prediction, and DockQ should be useful in a similar development in the protein docking field. DockQ is available at http://github.com/bjornwallner/DockQ/.
Collapse
Affiliation(s)
- Sankar Basu
- Bioinformatics Division, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Björn Wallner
- Bioinformatics Division, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
- Swedish e-Science Research Center, Linköping University, Linköping, Sweden
| |
Collapse
|