101
|
Gaillard T. Evaluation of AutoDock and AutoDock Vina on the CASF-2013 Benchmark. J Chem Inf Model 2018; 58:1697-1706. [DOI: 10.1021/acs.jcim.8b00312] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Thomas Gaillard
- Laboratoire de Biochimie (CNRS UMR7654), Department of Biology, Ecole Polytechnique, 91128 Palaiseau, France
| |
Collapse
|
102
|
Development of a new benchmark for assessing the scoring functions applicable to protein–protein interactions. Future Med Chem 2018; 10:1555-1574. [DOI: 10.4155/fmc-2017-0261] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aim: Scoring functions are important component of protein–protein docking methods. They need to be evaluated on high-quality benchmarks to reveal their strengths and weaknesses. Evaluation results obtained on such benchmarks can provide valuable guidance for developing more advanced scoring functions. Methodology & results: In our comparative assessment of scoring functions for protein–protein interactions benchmark, the performance of a scoring function was characterized by ‘docking power’ and ‘scoring power’. A high-quality dataset of 273 protein–protein complexes was compiled and employed in both tests. Four scoring functions, including FASTCONTACT, ZRANK, dDFIRE and ATTRACT were tested as demonstration. ZRANK and ATTRACT exhibited encouraging performance in the docking power test. However, all four scoring functions failed badly in the scoring power test. Conclusion: Our comparative assessment of scoring functions for protein–protein interaction benchmark is created especially for assessing the scoring functions applicable to protein–protein interactions. It is different from other benchmarks for assessing protein–protein docking methods. Our benchmark is available to the public at www.pdbbind-cn.org/download/CASF-PPI/ .
Collapse
|
103
|
Kalinowsky L, Weber J, Balasupramaniam S, Baumann K, Proschak E. A Diverse Benchmark Based on 3D Matched Molecular Pairs for Validating Scoring Functions. ACS OMEGA 2018; 3:5704-5714. [PMID: 31458770 PMCID: PMC6641919 DOI: 10.1021/acsomega.7b01194] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 01/22/2018] [Indexed: 06/10/2023]
Abstract
The prediction of protein-ligand interactions and their corresponding binding free energy is a challenging task in structure-based drug design and related applications. Docking and scoring is broadly used to propose the binding mode and underlying interactions as well as to provide a measure for ligand affinity or differentiate between active and inactive ligands. Various studies have revealed that most docking software packages reliably predict the binding mode, although scoring remains a challenge. Here, a diverse benchmark data set of 99 matched molecular pairs (3D-MMPs) with experimentally determined X-ray structures and corresponding binding affinities is introduced. This data set was used to study the predictive power of 13 commonly used scoring functions to demonstrate the applicability of the 3D-MMP data set as a valuable tool for benchmarking scoring functions.
Collapse
Affiliation(s)
- Lena Kalinowsky
- Institute
of Pharmaceutical Chemistry, Goethe University
Frankfurt, Max-von-Laue
Str. 9, Frankfurt am Main D-60438, Germany
| | - Julia Weber
- Institute
of Pharmaceutical Chemistry, Goethe University
Frankfurt, Max-von-Laue
Str. 9, Frankfurt am Main D-60438, Germany
| | - Shantheya Balasupramaniam
- Institute
of Medicinal and Pharmaceutical Chemistry, University of Technology of Braunschweig, Beethovenstr. 55, Braunschweig D-38106, Germany
| | - Knut Baumann
- Institute
of Medicinal and Pharmaceutical Chemistry, University of Technology of Braunschweig, Beethovenstr. 55, Braunschweig D-38106, Germany
| | - Ewgenij Proschak
- Institute
of Pharmaceutical Chemistry, Goethe University
Frankfurt, Max-von-Laue
Str. 9, Frankfurt am Main D-60438, Germany
| |
Collapse
|
104
|
|
105
|
Carlson HA. Lessons Learned over Four Benchmark Exercises from the Community Structure-Activity Resource. J Chem Inf Model 2018; 56:951-4. [PMID: 27345761 DOI: 10.1021/acs.jcim.6b00182] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- Heather A Carlson
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan , 428 Church Street, Ann Arbor, Michigan 48109-1065, United States
| |
Collapse
|
106
|
Fu DY, Meiler J. Predictive Power of Different Types of Experimental Restraints in Small Molecule Docking: A Review. J Chem Inf Model 2018; 58:225-233. [PMID: 29286651 DOI: 10.1021/acs.jcim.7b00418] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Incorporating experimental restraints is a powerful method of increasing accuracy in computational protein small molecule docking simulations. Different algorithms integrate distinct forms of biochemical data during the docking and/or scoring stages. These so-called hybrid methods make use of receptor-based information such as nuclear magnetic resonance (NMR) restraints or small molecule-based information such as structure-activity relationships (SARs). A third class of methods directly interrogates contacts between the protein receptor and the small molecule. This work reviews the current state of using such restraints in docking simulations, evaluates their feasibility across broad systems, and identifies potential areas of algorithm development.
Collapse
Affiliation(s)
- Darwin Y Fu
- Department of Chemistry Vanderbilt University Nashville, Tennessee 37235, United States
| | - Jens Meiler
- Department of Chemistry Vanderbilt University Nashville, Tennessee 37235, United States
| |
Collapse
|
107
|
Ehmki ESR, Rarey M. Exploring Structure-Activity Relationships with Three-Dimensional Matched Molecular Pairs-A Review. ChemMedChem 2018; 13:482-489. [PMID: 29211343 DOI: 10.1002/cmdc.201700628] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 11/27/2017] [Indexed: 11/10/2022]
Abstract
A matched molecular pair (MMP) consists of two small molecules that differ by a few atoms only. The minor structural difference between the molecules allows a detailed analysis of changes in properties. Three-dimensional (3D) MMPs extend the concept of chemical similarity by spatial similarity. Conformations must be generated, and superimpositions have to be calculated. The additional complexity and uncertainty as well as the smaller amount of available experimental data substantially complicates the derivation of models. Nonetheless, there are some benefits that make the transition worthwhile. The 3D concept gives detailed insight into mechanisms behind several methods classically used by the 2D MMP approach. It can help to analyze disrupted series of structure-activity relationships or extend the 2D MMP concept with scaffold hopping. One of the most powerful features is the high confidence structure-activity relationship transfer between series of analogues. Several research groups have approached the problem from different directions. The models vary especially in the 3D similarity measure used and complexity of the applied descriptor selected or designed. Nonetheless, all approaches have increased the amount of information available by incorporating 3D structural information.
Collapse
Affiliation(s)
- Emanuel S R Ehmki
- Center for Bioinformatics, Universität Hamburg, Bundesstraße 43, 20146, Hamburg, Germany
| | - Matthias Rarey
- Center for Bioinformatics, Universität Hamburg, Bundesstraße 43, 20146, Hamburg, Germany
| |
Collapse
|
108
|
Jiménez J, Škalič M, Martínez-Rosell G, De Fabritiis G. KDEEP: Protein–Ligand Absolute Binding Affinity Prediction via 3D-Convolutional Neural Networks. J Chem Inf Model 2018; 58:287-296. [DOI: 10.1021/acs.jcim.7b00650] [Citation(s) in RCA: 389] [Impact Index Per Article: 64.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- José Jiménez
- Computational
Biophysics Laboratory, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona, Carrer del Dr. Aiguader
88, Barcelona 08003, Spain
| | - Miha Škalič
- Computational
Biophysics Laboratory, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona, Carrer del Dr. Aiguader
88, Barcelona 08003, Spain
| | - Gerard Martínez-Rosell
- Computational
Biophysics Laboratory, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona, Carrer del Dr. Aiguader
88, Barcelona 08003, Spain
| | - Gianni De Fabritiis
- Computational
Biophysics Laboratory, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona, Carrer del Dr. Aiguader
88, Barcelona 08003, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Passeig Lluis Companys 23, 08010 Barcelona, Spain
| |
Collapse
|
109
|
Padhorny D, Hall DR, Mirzaei H, Mamonov AB, Moghadasi M, Alekseenko A, Beglov D, Kozakov D. Protein-ligand docking using FFT based sampling: D3R case study. J Comput Aided Mol Des 2018; 32:225-230. [PMID: 29101520 PMCID: PMC5767528 DOI: 10.1007/s10822-017-0069-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 09/16/2017] [Indexed: 12/15/2022]
Abstract
Fast Fourier transform (FFT) based approaches have been successful in application to modeling of relatively rigid protein-protein complexes. Recently, we have been able to adapt the FFT methodology to treatment of flexible protein-peptide interactions. Here, we report our latest attempt to expand the capabilities of the FFT approach to treatment of flexible protein-ligand interactions in application to the D3R PL-2016-1 challenge. Based on the D3R assessment, our FFT approach in conjunction with Monte Carlo minimization off-grid refinement was among the top performing methods in the challenge. The potential advantage of our method is its ability to globally sample the protein-ligand interaction landscape, which will be explored in further applications.
Collapse
Affiliation(s)
- Dzmitry Padhorny
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, 11794, USA
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, 11794, USA
| | | | - Hanieh Mirzaei
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Artem B Mamonov
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Mohammad Moghadasi
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Andrey Alekseenko
- Moscow Institute of Physics and Technology (State University), Institutskii per. 9, Dolgoprudny, Moscow Oblast, Russia, 141700
- Institute of Computer Aided Design of the Russian Academy of Sciences, 19/18, 2-nd Brestskaya St, Moscow, Russia, 123056
| | - Dmitri Beglov
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA.
| | - Dima Kozakov
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, 11794, USA.
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, 11794, USA.
| |
Collapse
|
110
|
Morency LP, Gaudreault F, Najmanovich R. Applications of the NRGsuite and the Molecular Docking Software FlexAID in Computational Drug Discovery and Design. Methods Mol Biol 2018; 1762:367-388. [PMID: 29594781 DOI: 10.1007/978-1-4939-7756-7_18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Docking simulations help us understand molecular interactions. Here we present a hands-on tutorial to utilize FlexAID (Flexible Artificial Intelligence Docking), an open source molecular docking software between ligands such as small molecules or peptides and macromolecules such as proteins and nucleic acids. The tutorial uses the NRGsuite PyMOL plugin graphical user interface to set up and visualize docking simulations in real time as well as detect and refine target cavities. The ease of use of FlexAID and the NRGsuite combined with its superior performance relative to widely used docking software provides nonexperts with an important tool to understand molecular interactions with direct applications in structure-based drug design and virtual high-throughput screening.
Collapse
Affiliation(s)
- Louis-Philippe Morency
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | | | - Rafael Najmanovich
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
111
|
Ashtawy HM, Mahapatra NR. Task-Specific Scoring Functions for Predicting Ligand Binding Poses and Affinity and for Screening Enrichment. J Chem Inf Model 2017; 58:119-133. [DOI: 10.1021/acs.jcim.7b00309] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Hossam M. Ashtawy
- Department of Electrical and Computer
Engineering, Michigan State University, East Lansing, Michigan 48824-1226, United States
| | - Nihar R. Mahapatra
- Department of Electrical and Computer
Engineering, Michigan State University, East Lansing, Michigan 48824-1226, United States
| |
Collapse
|
112
|
Ashtawy HM, Mahapatra NR. Descriptor Data Bank (DDB): A Cloud Platform for Multiperspective Modeling of Protein–Ligand Interactions. J Chem Inf Model 2017; 58:134-147. [DOI: 10.1021/acs.jcim.7b00310] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Hossam M. Ashtawy
- Department of Electrical
and Computer Engineering, Michigan State University, East Lansing, Michigan 48824-1226, United States
| | - Nihar R. Mahapatra
- Department of Electrical
and Computer Engineering, Michigan State University, East Lansing, Michigan 48824-1226, United States
| |
Collapse
|
113
|
Workflows and performances in the ranking prediction of 2016 D3R Grand Challenge 2: lessons learned from a collaborative effort. J Comput Aided Mol Des 2017; 32:129-142. [PMID: 28986733 DOI: 10.1007/s10822-017-0072-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 09/23/2017] [Indexed: 12/21/2022]
Abstract
The 2016 D3R Grand Challenge 2 includes both pose and affinity or ranking predictions. This article is focused exclusively on affinity predictions submitted to the D3R challenge from a collaborative effort of the modeling and informatics group. Our submissions include ranking of 102 ligands covering 4 different chemotypes against the FXR ligand binding domain structure, and the relative binding affinity predictions of the two designated free energy subsets of 15 and 18 compounds. Using all the complex structures prepared in the same way allowed us to cover many types of workflows and compare their performances effectively. We evaluated typical workflows used in our daily structure-based design modeling support, which include docking scores, force field-based scores, QM/MM, MMGBSA, MD-MMGBSA, and MacroModel interaction energy estimations. The best performing methods for the two free energy subsets are discussed. Our results suggest that affinity ranking still remains very challenging; that the knowledge of more structural information does not necessarily yield more accurate predictions; and that visual inspection and human intervention are considerably important for ranking. Knowledge of the mode of action and protein flexibility along with visualization tools that depict polar and hydrophobic maps are very useful for visual inspection. QM/MM-based workflows were found to be powerful in affinity ranking and are encouraged to be applied more often. The standardized input and output enable systematic analysis and support methodology development and improvement for high level blinded predictions.
Collapse
|
114
|
Meyder A, Nittinger E, Lange G, Klein R, Rarey M. Estimating Electron Density Support for Individual Atoms and Molecular Fragments in X-ray Structures. J Chem Inf Model 2017; 57:2437-2447. [PMID: 28981269 DOI: 10.1021/acs.jcim.7b00391] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Macromolecular structures resolved by X-ray crystallography are essential for life science research. While some methods exist to automatically quantify the quality of the electron density fit, none of them is without flaws. Especially the question of how well individual parts like atoms, small fragments, or molecules are supported by electron density is difficult to quantify. While taking experimental uncertainties correctly into account, they do not offer an answer on how reliable an individual atom position is. A rapid quantification of this atomic position reliability would be highly valuable in structure-based molecular design. To overcome this limitation, we introduce the electron density score EDIA for individual atoms and molecular fragments. EDIA assesses rapidly, automatically, and intuitively the fit of individual as well as multiple atoms (EDIAm) into electron density accompanied by an integrated error analysis. The computation is based on the standard 2fo - fc electron density map in combination with the model of the molecular structure. For evaluating partial structures, EDIAm shows significant advantages compared to the real-space R correlation coefficient (RSCC) and the real-space difference density Z score (RSZD) from the molecular modeler's point of view. Thus, EDIA abolishes the time-consuming step of visually inspecting the electron density during structure selection and curation. It supports daily modeling tasks of medicinal and computational chemists and enables a fully automated assembly of large-scale, high-quality structure data sets. Furthermore, EDIA scores can be applied for model validation and method development in computer-aided molecular design. In contrast to measuring the deviation from the structure model by root-mean-squared deviation, EDIA scores allow comparison to the underlying experimental data taking its uncertainty into account.
Collapse
Affiliation(s)
- Agnes Meyder
- ZBH-Center for Bioinformatics, Universität Hamburg , Hamburg 20146, Germany
| | - Eva Nittinger
- ZBH-Center for Bioinformatics, Universität Hamburg , Hamburg 20146, Germany
| | | | | | - Matthias Rarey
- ZBH-Center for Bioinformatics, Universität Hamburg , Hamburg 20146, Germany
| |
Collapse
|
115
|
Convex-PL: a novel knowledge-based potential for protein-ligand interactions deduced from structural databases using convex optimization. J Comput Aided Mol Des 2017; 31:943-958. [DOI: 10.1007/s10822-017-0068-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 09/08/2017] [Indexed: 12/16/2022]
|
116
|
Ajani H, Pecina A, Eyrilmez SM, Fanfrlík J, Haldar S, Řezáč J, Hobza P, Lepšík M. Superior Performance of the SQM/COSMO Scoring Functions in Native Pose Recognition of Diverse Protein-Ligand Complexes in Cognate Docking. ACS OMEGA 2017; 2:4022-4029. [PMID: 30023710 PMCID: PMC6044937 DOI: 10.1021/acsomega.7b00503] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/18/2017] [Indexed: 06/08/2023]
Abstract
General and reliable description of structures and energetics in protein-ligand (PL) binding using the docking/scoring methodology has until now been elusive. We address this urgent deficiency of scoring functions (SFs) by the systematic development of corrected semiempirical quantum mechanical (SQM) methods, which correctly describe all types of noncovalent interactions and are fast enough to treat systems of thousands of atoms. Two most accurate SQM methods, PM6-D3H4X and SCC-DFTB3-D3H4X, are coupled with the conductor-like screening model (COSMO) implicit solvation model in so-called "SQM/COSMO" SFs and have shown unique recognition of native ligand poses in cognate docking in four challenging PL systems, including metalloprotein. Here, we apply the two SQM/COSMO SFs to 17 diverse PL complexes and compare their performance with four widely used classical SFs (Glide XP, AutoDock4, AutoDock Vina, and UCSF Dock). We observe superior performance of the SQM/COSMO SFs and identify challenging systems. This method, due to its generality, comparability across the chemical space, and lack of need for any system-specific parameters, gives promise of becoming, after comprehensive large-scale testing in the near future, a useful computational tool in structure-based drug design and serving as a reference method for the development of other SFs.
Collapse
Affiliation(s)
- Haresh Ajani
- Department
of Computational Chemistry, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, v.v.i., Flemingovo nam. 2, 16610 Praha 6, Czech Republic
- Department
of Physical Chemistry, Palacký University, tř. 17. listopadu 1192/12, 77146 Olomouc, Czech Republic
| | - Adam Pecina
- Department
of Computational Chemistry, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, v.v.i., Flemingovo nam. 2, 16610 Praha 6, Czech Republic
| | - Saltuk M. Eyrilmez
- Department
of Computational Chemistry, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, v.v.i., Flemingovo nam. 2, 16610 Praha 6, Czech Republic
- Department
of Physical Chemistry, Palacký University, tř. 17. listopadu 1192/12, 77146 Olomouc, Czech Republic
| | - Jindřich Fanfrlík
- Department
of Computational Chemistry, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, v.v.i., Flemingovo nam. 2, 16610 Praha 6, Czech Republic
| | - Susanta Haldar
- Department
of Computational Chemistry, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, v.v.i., Flemingovo nam. 2, 16610 Praha 6, Czech Republic
| | - Jan Řezáč
- Department
of Computational Chemistry, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, v.v.i., Flemingovo nam. 2, 16610 Praha 6, Czech Republic
| | - Pavel Hobza
- Department
of Computational Chemistry, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, v.v.i., Flemingovo nam. 2, 16610 Praha 6, Czech Republic
- Department
of Physical Chemistry, Regional Centre of Advanced Technologies and
Materials, Palacký University, 77146 Olomouc, Czech Republic
| | - Martin Lepšík
- Department
of Computational Chemistry, Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, v.v.i., Flemingovo nam. 2, 16610 Praha 6, Czech Republic
| |
Collapse
|
117
|
Liu J, Su M, Liu Z, Li J, Li Y, Wang R. Enhance the performance of current scoring functions with the aid of 3D protein-ligand interaction fingerprints. BMC Bioinformatics 2017; 18:343. [PMID: 28720122 PMCID: PMC5516336 DOI: 10.1186/s12859-017-1750-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 07/05/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In structure-based drug design, binding affinity prediction remains as a challenging goal for current scoring functions. Development of target-biased scoring functions provides a new possibility for tackling this problem, but this approach is also associated with certain technical difficulties. We previously reported the Knowledge-Guided Scoring (KGS) method as an alternative approach (BMC Bioinformatics, 2010, 11, 193-208). The key idea is to compute the binding affinity of a given protein-ligand complex based on the known binding data of an appropriate reference complex, so the error in binding affinity prediction can be reduced effectively. RESULTS In this study, we have developed an upgraded version, i.e. KGS2, by employing 3D protein-ligand interaction fingerprints in reference selection. KGS2 was evaluated in combination with four scoring functions (X-Score, ChemPLP, ASP, and GoldScore) on five drug targets (HIV-1 protease, carbonic anhydrase 2, beta-secretase 1, beta-trypsin, and checkpoint kinase 1). In the in situ scoring test, considerable improvements were observed in most cases after application of KGS2. Besides, the performance of KGS2 was always better than KGS in all cases. In the more challenging molecular docking test, application of KGS2 also led to improved structure-activity relationship in some cases. CONCLUSIONS KGS2 can be applied as a convenient "add-on" to current scoring functions without the need to re-engineer them, and its application is not limited to certain target proteins as customized scoring functions. As an interpolation method, its accuracy in principle can be improved further with the increasing knowledge of protein-ligand complex structures and binding affinity data. We expect that KGS2 will become a practical tool for enhancing the performance of current scoring functions in binding affinity prediction. The KGS2 software is available upon contacting the authors.
Collapse
Affiliation(s)
- Jie Liu
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Collaborative Innovation Center of Chemistry for Life Sciences, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai, 200032, China
| | - Minyi Su
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Collaborative Innovation Center of Chemistry for Life Sciences, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai, 200032, China
| | - Zhihai Liu
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Collaborative Innovation Center of Chemistry for Life Sciences, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai, 200032, China
| | - Jie Li
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Collaborative Innovation Center of Chemistry for Life Sciences, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai, 200032, China
| | - Yan Li
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Collaborative Innovation Center of Chemistry for Life Sciences, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai, 200032, China.
| | - Renxiao Wang
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Collaborative Innovation Center of Chemistry for Life Sciences, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai, 200032, China. .,State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, People's Republic of China.
| |
Collapse
|
118
|
GalaxyDock BP2 score: a hybrid scoring function for accurate protein–ligand docking. J Comput Aided Mol Des 2017. [DOI: 10.1007/s10822-017-0030-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
119
|
Chéron N, Shakhnovich EI. Effect of sampling on BACE-1 ligands binding free energy predictions via MM-PBSA calculations. J Comput Chem 2017; 38:1941-1951. [PMID: 28568844 DOI: 10.1002/jcc.24839] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 05/02/2017] [Accepted: 05/04/2017] [Indexed: 01/04/2023]
Abstract
The BACE-1 enzyme is a prime target to find a cure to Alzheimer's disease. In this article, we used the MM-PBSA approach to compute the binding free energies of 46 reported ligands to this enzyme. After showing that the most probable protonation state of the catalytic dyad is mono-protonated (on ASP32), we performed a thorough analysis of the parameters influencing the sampling of the conformational space (in total, more than 35 μs of simulations were performed). We show that ten simulations of 2 ns gives better results than one of 50 ns. We also investigated the influence of the protein force field, the water model, the periodic boundary conditions artifacts (box size), as well as the ionic strength. Amber03 with TIP3P, a minimal distance of 1.0 nm between the protein and the box edges and a ionic strength of I = 0.2 M provides the optimal correlation with experiments. Overall, when using these parameters, a Pearson correlation coefficient of R = 0.84 (R2 = 0.71) is obtained for the 46 ligands, spanning eight orders of magnitude of Kd (from 0.017 nm to 2000 μM, i.e., from -14.7 to -3.7 kcal/mol), with a ligand size from 22 to 136 atoms (from 138 to 937 g/mol). After a two-parameter fit of the binding affinities for 12 of the ligands, an error of RMSD = 1.7 kcal/mol was obtained for the remaining ligands. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Nicolas Chéron
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, 02138.,Département de Chimie, UMR 8640 PASTEUR, Ecole Normale Supérieure, PSL Research University, UPMC Univ. Paris 06, CNRS, 24 rue Lhomond, Paris, 75005, France
| | - Eugene I Shakhnovich
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, 02138
| |
Collapse
|
120
|
Yu Z, Li P, Merz KM. Using Ligand-Induced Protein Chemical Shift Perturbations To Determine Protein–Ligand Structures. Biochemistry 2017; 56:2349-2362. [DOI: 10.1021/acs.biochem.7b00170] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Zhuoqin Yu
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824-1322, United States
| | - Pengfei Li
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824-1322, United States
| | - Kenneth M. Merz
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824-1322, United States
| |
Collapse
|
121
|
Debroise T, Shakhnovich EI, Chéron N. A Hybrid Knowledge-Based and Empirical Scoring Function for Protein–Ligand Interaction: SMoG2016. J Chem Inf Model 2017; 57:584-593. [DOI: 10.1021/acs.jcim.6b00610] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Théau Debroise
- Department
of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Eugene I. Shakhnovich
- Department
of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Nicolas Chéron
- Department
of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
- Ecole Normale Supérieure, PSL Research University, UPMC Univ. Paris 06, CNRS, Département de Chimie,
UMR 8640 PASTEUR, 24 rue
Lhomond, 75005 Paris, France
| |
Collapse
|
122
|
Liu Z, Su M, Han L, Liu J, Yang Q, Li Y, Wang R. Forging the Basis for Developing Protein-Ligand Interaction Scoring Functions. Acc Chem Res 2017; 50:302-309. [PMID: 28182403 DOI: 10.1021/acs.accounts.6b00491] [Citation(s) in RCA: 217] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
In structure-based drug design, scoring functions are widely used for fast evaluation of protein-ligand interactions. They are often applied in combination with molecular docking and de novo design methods. Since the early 1990s, a whole spectrum of protein-ligand interaction scoring functions have been developed. Regardless of their technical difference, scoring functions all need data sets combining protein-ligand complex structures and binding affinity data for parametrization and validation. However, data sets of this kind used to be rather limited in terms of size and quality. On the other hand, standard metrics for evaluating scoring function used to be ambiguous. Scoring functions are often tested in molecular docking or even virtual screening trials, which do not directly reflect the genuine quality of scoring functions. Collectively, these underlying obstacles have impeded the invention of more advanced scoring functions. In this Account, we describe our long-lasting efforts to overcome these obstacles, which involve two related projects. On the first project, we have created the PDBbind database. It is the first database that systematically annotates the protein-ligand complexes in the Protein Data Bank (PDB) with experimental binding data. This database has been updated annually since its first public release in 2004. The latest release (version 2016) provides binding data for 16 179 biomolecular complexes in PDB. Data sets provided by PDBbind have been applied to many computational and statistical studies on protein-ligand interaction and various subjects. In particular, it has become a major data resource for scoring function development. On the second project, we have established the Comparative Assessment of Scoring Functions (CASF) benchmark for scoring function evaluation. Our key idea is to decouple the "scoring" process from the "sampling" process, so scoring functions can be tested in a relatively pure context to reflect their quality. In our latest work on this track, i.e. CASF-2013, the performance of a scoring function was quantified in four aspects, including "scoring power", "ranking power", "docking power", and "screening power". All four performance tests were conducted on a test set containing 195 high-quality protein-ligand complexes selected from PDBbind. A panel of 20 standard scoring functions were tested as demonstration. Importantly, CASF is designed to be an open-access benchmark, with which scoring functions developed by different researchers can be compared on the same grounds. Indeed, it has become a popular choice for scoring function validation in recent years. Despite the considerable progress that has been made so far, the performance of today's scoring functions still does not meet people's expectations in many aspects. There is a constant demand for more advanced scoring functions. Our efforts have helped to overcome some obstacles underlying scoring function development so that the researchers in this field can move forward faster. We will continue to improve the PDBbind database and the CASF benchmark in the future to keep them as useful community resources.
Collapse
Affiliation(s)
- Zhihai Liu
- State
Key Laboratory of Bioorganic and Natural Products Chemistry, Collaborative
Innovation Center of Chemistry for Life Sciences, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, People’s Republic of China
| | - Minyi Su
- State
Key Laboratory of Bioorganic and Natural Products Chemistry, Collaborative
Innovation Center of Chemistry for Life Sciences, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, People’s Republic of China
| | - Li Han
- State
Key Laboratory of Bioorganic and Natural Products Chemistry, Collaborative
Innovation Center of Chemistry for Life Sciences, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, People’s Republic of China
| | - Jie Liu
- State
Key Laboratory of Bioorganic and Natural Products Chemistry, Collaborative
Innovation Center of Chemistry for Life Sciences, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, People’s Republic of China
| | - Qifan Yang
- State
Key Laboratory of Bioorganic and Natural Products Chemistry, Collaborative
Innovation Center of Chemistry for Life Sciences, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, People’s Republic of China
| | - Yan Li
- State
Key Laboratory of Bioorganic and Natural Products Chemistry, Collaborative
Innovation Center of Chemistry for Life Sciences, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, People’s Republic of China
| | - Renxiao Wang
- State
Key Laboratory of Bioorganic and Natural Products Chemistry, Collaborative
Innovation Center of Chemistry for Life Sciences, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, People’s Republic of China
- State
Key Laboratory of Quality Research in Chinese Medicine, Macau Institute
for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, People’s Republic of China
| |
Collapse
|
123
|
Friedrich NO, Meyder A, de Bruyn Kops C, Sommer K, Flachsenberg F, Rarey M, Kirchmair J. High-Quality Dataset of Protein-Bound Ligand Conformations and Its Application to Benchmarking Conformer Ensemble Generators. J Chem Inf Model 2017; 57:529-539. [PMID: 28206754 DOI: 10.1021/acs.jcim.6b00613] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We developed a cheminformatics pipeline for the fully automated selection and extraction of high-quality protein-bound ligand conformations from X-ray structural data. The pipeline evaluates the validity and accuracy of the 3D structures of small molecules according to multiple criteria, including their fit to the electron density and their physicochemical and structural properties. Using this approach, we compiled two high-quality datasets from the Protein Data Bank (PDB): a comprehensive dataset and a diversified subset of 4626 and 2912 structures, respectively. The datasets were applied to benchmarking seven freely available conformer ensemble generators: Balloon (two different algorithms), the RDKit standard conformer ensemble generator, the Experimental-Torsion basic Knowledge Distance Geometry (ETKDG) algorithm, Confab, Frog2 and Multiconf-DOCK. Substantial differences in the performance of the individual algorithms were observed, with RDKit and ETKDG generally achieving a favorable balance of accuracy, ensemble size and runtime. The Platinum datasets are available for download from http://www.zbh.uni-hamburg.de/platinum_dataset .
Collapse
Affiliation(s)
- Nils-Ole Friedrich
- University of Hamburg , ZBH - Center for Bioinformatics, Bundesstraße 43, Hamburg 20146, Germany
| | - Agnes Meyder
- University of Hamburg , ZBH - Center for Bioinformatics, Bundesstraße 43, Hamburg 20146, Germany
| | - Christina de Bruyn Kops
- University of Hamburg , ZBH - Center for Bioinformatics, Bundesstraße 43, Hamburg 20146, Germany
| | - Kai Sommer
- University of Hamburg , ZBH - Center for Bioinformatics, Bundesstraße 43, Hamburg 20146, Germany
| | - Florian Flachsenberg
- University of Hamburg , ZBH - Center for Bioinformatics, Bundesstraße 43, Hamburg 20146, Germany
| | - Matthias Rarey
- University of Hamburg , ZBH - Center for Bioinformatics, Bundesstraße 43, Hamburg 20146, Germany
| | - Johannes Kirchmair
- University of Hamburg , ZBH - Center for Bioinformatics, Bundesstraße 43, Hamburg 20146, Germany
| |
Collapse
|
124
|
Wang C, Zhang Y. Improving scoring-docking-screening powers of protein-ligand scoring functions using random forest. J Comput Chem 2017; 38:169-177. [PMID: 27859414 PMCID: PMC5140681 DOI: 10.1002/jcc.24667] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 09/06/2016] [Accepted: 10/26/2016] [Indexed: 12/16/2022]
Abstract
The development of new protein-ligand scoring functions using machine learning algorithms, such as random forest, has been of significant interest. By efficiently utilizing expanded feature sets and a large set of experimental data, random forest based scoring functions (RFbScore) can achieve better correlations to experimental protein-ligand binding data with known crystal structures; however, more extensive tests indicate that such enhancement in scoring power comes with significant under-performance in docking and screening power tests compared to traditional scoring functions. In this work, to improve scoring-docking-screening powers of protein-ligand docking functions simultaneously, we have introduced a Δvina RF parameterization and feature selection framework based on random forest. Our developed scoring function Δvina RF20 , which employs 20 descriptors in addition to the AutoDock Vina score, can achieve superior performance in all power tests of both CASF-2013 and CASF-2007 benchmarks compared to classical scoring functions. The Δvina RF20 scoring function and its code are freely available on the web at: https://www.nyu.edu/projects/yzhang/DeltaVina. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Cheng Wang
- Department of Chemistry, New York University, New York, New York 10003
| | - Yingkai Zhang
- Department of Chemistry, New York University, New York, New York 10003
- NYU-ECNU Center for Computational Chemistry at NYU Shanghai, Shanghai 200062, China
| |
Collapse
|
125
|
A Thoroughly Validated Virtual Screening Strategy for Discovery of Novel HDAC3 Inhibitors. Int J Mol Sci 2017; 18:ijms18010137. [PMID: 28106794 PMCID: PMC5297770 DOI: 10.3390/ijms18010137] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 01/09/2017] [Accepted: 01/09/2017] [Indexed: 12/15/2022] Open
Abstract
Histone deacetylase 3 (HDAC3) has been recently identified as a potential target for the treatment of cancer and other diseases, such as chronic inflammation, neurodegenerative diseases, and diabetes. Virtual screening (VS) is currently a routine technique for hit identification, but its success depends on rational development of VS strategies. To facilitate this process, we applied our previously released benchmarking dataset, i.e., MUBD-HDAC3 to the evaluation of structure-based VS (SBVS) and ligand-based VS (LBVS) combinatorial approaches. We have identified FRED (Chemgauss4) docking against a structural model of HDAC3, i.e., SAHA-3 generated by a computationally inexpensive “flexible docking”, as the best SBVS approach and a common feature pharmacophore model, i.e., Hypo1 generated by Catalyst/HipHop as the optimal model for LBVS. We then developed a pipeline that was composed of Hypo1, FRED (Chemgauss4), and SAHA-3 sequentially, and demonstrated that it was superior to other combinations in terms of ligand enrichment. In summary, we present the first highly-validated, rationally-designed VS strategy specific to HDAC3 inhibitor discovery. The constructed pipeline is publicly accessible for the scientific community to identify novel HDAC3 inhibitors in a time-efficient and cost-effective way.
Collapse
|
126
|
Exploring the stability of ligand binding modes to proteins by molecular dynamics simulations. J Comput Aided Mol Des 2017; 31:201-211. [PMID: 28074360 DOI: 10.1007/s10822-016-0005-2] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 12/22/2016] [Indexed: 02/01/2023]
Abstract
The binding mode prediction is of great importance to structure-based drug design. The discrimination of various binding poses of ligand generated by docking is a great challenge not only to docking score functions but also to the relatively expensive free energy calculation methods. Here we systematically analyzed the stability of various ligand poses under molecular dynamics (MD) simulation. First, a data set of 120 complexes was built based on the typical physicochemical properties of drug-like ligands. Three potential binding poses (one correct pose and two decoys) were selected for each ligand from self-docking in addition to the experimental pose. Then, five independent MD simulations for each pose were performed with different initial velocities for the statistical analysis. Finally, the stabilities of ligand poses under MD were evaluated and compared with the native one from crystal structure. We found that about 94% of the native poses were maintained stable during the simulations, which suggests that MD simulations are accurate enough to judge most experimental binding poses as stable properly. Interestingly, incorrect decoy poses were maintained much less and 38-44% of decoys could be excluded just by performing equilibrium MD simulations, though 56-62% of decoys were stable. The computationally-heavy binding free energy calculation can be performed only for these survived poses.
Collapse
|
127
|
Prediction of N-Methyl-D-Aspartate Receptor GluN1-Ligand Binding Affinity by a Novel SVM-Pose/SVM-Score Combinatorial Ensemble Docking Scheme. Sci Rep 2017; 7:40053. [PMID: 28059133 PMCID: PMC5216401 DOI: 10.1038/srep40053] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/30/2016] [Indexed: 01/24/2023] Open
Abstract
The glycine-binding site of the N-methyl-D-aspartate receptor (NMDAR) subunit GluN1 is a potential pharmacological target for neurodegenerative disorders. A novel combinatorial ensemble docking scheme using ligand and protein conformation ensembles and customized support vector machine (SVM)-based models to select the docked pose and to predict the docking score was generated for predicting the NMDAR GluN1-ligand binding affinity. The predicted root mean square deviation (RMSD) values in pose by SVM-Pose models were found to be in good agreement with the observed values (n = 30, r2 = 0.928–0.988, = 0.894–0.954, RMSE = 0.002–0.412, s = 0.001–0.214), and the predicted pKi values by SVM-Score were found to be in good agreement with the observed values for the training samples (n = 24, r2 = 0.967, = 0.899, RMSE = 0.295, s = 0.170) and test samples (n = 13, q2 = 0.894, RMSE = 0.437, s = 0.202). When subjected to various statistical validations, the developed SVM-Pose and SVM-Score models consistently met the most stringent criteria. A mock test asserted the predictivity of this novel docking scheme. Collectively, this accurate novel combinatorial ensemble docking scheme can be used to predict the NMDAR GluN1-ligand binding affinity for facilitating drug discovery.
Collapse
|
128
|
Yan Z, Wang J. Scoring Functions of Protein-Ligand Interactions. Oncology 2017. [DOI: 10.4018/978-1-5225-0549-5.ch036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Scoring function of protein-ligand interactions is used to recognize the “native” binding pose of a ligand on the protein and to predict the binding affinity, so that the active small molecules can be discriminated from the non-active ones. Scoring function is widely used in computationally molecular docking and structure-based drug discovery. The development and improvement of scoring functions have broad implications in pharmaceutical industry and academic research. During the past three decades, much progress have been made in methodology and accuracy for scoring functions, and many successful cases have be witnessed in virtual database screening. In this chapter, the authors introduced the basic types of scoring functions and their derivations, the commonly-used evaluation methods and benchmarks, as well as the underlying challenges and current solutions. Finally, the authors discussed the promising directions to improve and develop scoring functions for future molecular docking-based drug discovery.
Collapse
|
129
|
Shin WH, Christoffer CW, Wang J, Kihara D. PL-PatchSurfer2: Improved Local Surface Matching-Based Virtual Screening Method That Is Tolerant to Target and Ligand Structure Variation. J Chem Inf Model 2016; 56:1676-91. [PMID: 27500657 PMCID: PMC5037053 DOI: 10.1021/acs.jcim.6b00163] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Virtual screening has become an indispensable procedure in drug discovery. Virtual screening methods can be classified into two categories: ligand-based and structure-based. While the former have advantages, including being quick to compute, in general they are relatively weak at discovering novel active compounds because they use known actives as references. On the other hand, structure-based methods have higher potential to find novel compounds because they directly predict the binding affinity of a ligand in a target binding pocket, albeit with substantially lower speed than ligand-based methods. Here we report a novel structure-based virtual screening method, PL-PatchSurfer2. In PL-PatchSurfer2, protein and ligand surfaces are represented by a set of overlapping local patches, each of which is represented by three-dimensional Zernike descriptors (3DZDs). By means of 3DZDs, the shapes and physicochemical complementarities of local surface regions of a pocket surface and a ligand molecule can be concisely and effectively computed. Compared with the previous version of the program, the performance of PL-PatchSurfer2 is substantially improved by the addition of two more features, atom-based hydrophobicity and hydrogen-bond acceptors and donors. Benchmark studies showed that PL-PatchSurfer2 performed better than or comparable to popular existing methods. Particularly, PL-PatchSurfer2 significantly outperformed existing methods when apo-form or template-based protein models were used for queries. The computational time of PL-PatchSurfer2 is about 20 times shorter than those of conventional structure-based methods. The PL-PatchSurfer2 program is available at http://www.kiharalab.org/plps2/ .
Collapse
Affiliation(s)
- Woong-Hee Shin
- Department of Biological Science, Purdue University, 249 S. Martin Jischke Street, West Lafayette, IN, USA
| | - Charles W. Christoffer
- Department of Computer Science, Purdue University, 305 N. University Street, West Lafayette, IN, USA
| | - Jibo Wang
- Discovery Chemistry Research and Technologies, Eli Lilly and Company, 893 S. Delaware Street, Indianapolis, IN, USA
| | - Daisuke Kihara
- Department of Biological Science, Purdue University, 249 S. Martin Jischke Street, West Lafayette, IN, USA
- Department of Computer Science, Purdue University, 305 N. University Street, West Lafayette, IN, USA
| |
Collapse
|
130
|
Ligand binding cooperativity: Bioisosteric replacement of CO with SO2 among thrombin inhibitors. Bioorg Med Chem Lett 2016; 26:3850-4. [DOI: 10.1016/j.bmcl.2016.07.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 07/07/2016] [Accepted: 07/08/2016] [Indexed: 01/22/2023]
|
131
|
Fang Y, Ding Y, Feinstein WP, Koppelman DM, Moreno J, Jarrell M, Ramanujam J, Brylinski M. GeauxDock: Accelerating Structure-Based Virtual Screening with Heterogeneous Computing. PLoS One 2016; 11:e0158898. [PMID: 27420300 PMCID: PMC4946785 DOI: 10.1371/journal.pone.0158898] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 06/23/2016] [Indexed: 12/19/2022] Open
Abstract
Computational modeling of drug binding to proteins is an integral component of direct drug design. Particularly, structure-based virtual screening is often used to perform large-scale modeling of putative associations between small organic molecules and their pharmacologically relevant protein targets. Because of a large number of drug candidates to be evaluated, an accurate and fast docking engine is a critical element of virtual screening. Consequently, highly optimized docking codes are of paramount importance for the effectiveness of virtual screening methods. In this communication, we describe the implementation, tuning and performance characteristics of GeauxDock, a recently developed molecular docking program. GeauxDock is built upon the Monte Carlo algorithm and features a novel scoring function combining physics-based energy terms with statistical and knowledge-based potentials. Developed specifically for heterogeneous computing platforms, the current version of GeauxDock can be deployed on modern, multi-core Central Processing Units (CPUs) as well as massively parallel accelerators, Intel Xeon Phi and NVIDIA Graphics Processing Unit (GPU). First, we carried out a thorough performance tuning of the high-level framework and the docking kernel to produce a fast serial code, which was then ported to shared-memory multi-core CPUs yielding a near-ideal scaling. Further, using Xeon Phi gives 1.9× performance improvement over a dual 10-core Xeon CPU, whereas the best GPU accelerator, GeForce GTX 980, achieves a speedup as high as 3.5×. On that account, GeauxDock can take advantage of modern heterogeneous architectures to considerably accelerate structure-based virtual screening applications. GeauxDock is open-sourced and publicly available at www.brylinski.org/geauxdock and https://figshare.com/articles/geauxdock_tar_gz/3205249.
Collapse
Affiliation(s)
- Ye Fang
- School of Electrical Engineering and Computer Science, Louisiana State University, Baton Rouge, Louisiana, United States of America
- Center for Computation & Technology, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Yun Ding
- Department of Physics and Astronomy, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Wei P. Feinstein
- High-Performance Computing, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - David M. Koppelman
- School of Electrical Engineering and Computer Science, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Juana Moreno
- Department of Physics and Astronomy, Louisiana State University, Baton Rouge, Louisiana, United States of America
- Center for Computation & Technology, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Mark Jarrell
- Department of Physics and Astronomy, Louisiana State University, Baton Rouge, Louisiana, United States of America
- Center for Computation & Technology, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - J. Ramanujam
- School of Electrical Engineering and Computer Science, Louisiana State University, Baton Rouge, Louisiana, United States of America
- Center for Computation & Technology, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Michal Brylinski
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States of America
- Center for Computation & Technology, Louisiana State University, Baton Rouge, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
132
|
Pason LP, Sotriffer CA. Empirical Scoring Functions for Affinity Prediction of Protein-ligand Complexes. Mol Inform 2016; 35:541-548. [PMID: 27870243 DOI: 10.1002/minf.201600048] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 06/01/2016] [Indexed: 12/31/2022]
Abstract
The ability to rapidly assess the quality of a protein-ligand complex in terms of its affinity is of fundamental importance for various methods of computer-aided drug design. While simple filtering or matching critieria may be sufficient in fast docking methods or at early stages of virtual screening, estimates of the actual free energy of binding are needed whenever refined docking solutions, ligand rankings or support for the optimization of hit compounds are required. If rigorous free energy calculations based on molecular simulations are impractical, such affinity estimates are provided by scoring functions. The class of empirical scoring functions aims to provide them via a regression-based approach. Using experimental structures and affinity data of protein-ligand complexes and descriptors suitable to capture the essential features of the interaction, these functions are trained with classical linear regression techniques or machine-learning methods. The latter have led to considerable improvements in terms of prediction accuracy for large generic data sets. Nevertheless, many limitations are not yet resolved and pose significant challenges for future developments.
Collapse
Affiliation(s)
- Lukas P Pason
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, D-97074, Würzburg, Germany
| | - Christoph A Sotriffer
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, D-97074, Würzburg, Germany
| |
Collapse
|
133
|
Bjerrum EJ. Machine learning optimization of cross docking accuracy. Comput Biol Chem 2016; 62:133-44. [DOI: 10.1016/j.compbiolchem.2016.04.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 04/08/2016] [Accepted: 04/09/2016] [Indexed: 12/13/2022]
|
134
|
Pires DEV, Ascher DB. CSM-lig: a web server for assessing and comparing protein-small molecule affinities. Nucleic Acids Res 2016; 44:W557-61. [PMID: 27151202 PMCID: PMC4987933 DOI: 10.1093/nar/gkw390] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 04/28/2016] [Indexed: 12/21/2022] Open
Abstract
Determining the affinity of a ligand for a given protein is a crucial component of drug development and understanding their biological effects. Predicting binding affinities is a challenging and difficult task, and despite being regarded as poorly predictive, scoring functions play an important role in the analysis of molecular docking results. Here, we present CSM-Lig (http://structure.bioc.cam.ac.uk/csm_lig), a web server tailored to predict the binding affinity of a protein-small molecule complex, encompassing both protein and small-molecule complementarity in terms of shape and chemistry via graph-based structural signatures. CSM-Lig was trained and evaluated on different releases of the PDBbind databases, achieving a correlation of up to 0.86 on 10-fold cross validation and 0.80 in blind tests, performing as well as or better than other widely used methods. The web server allows users to rapidly and automatically predict binding affinities of collections of structures and assess the interactions made. We believe CSM-lig would be an invaluable tool for helping assess docking poses, the effects of multiple mutations, including insertions, deletions and alternative splicing events, in protein-small molecule affinity, unraveling important aspects that drive protein–compound recognition.
Collapse
Affiliation(s)
- Douglas E V Pires
- Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, 30190-002, Brazil
| | - David B Ascher
- Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, 30190-002, Brazil Department of Biochemistry, University of Cambridge, Cambridge, CB2 1GA, UK Department of Biochemistry, University of Melbourne, Victoria 3010, Australia
| |
Collapse
|
135
|
Liu X, Liu J, Zhu T, Zhang L, He X, Zhang JZH. PBSA_E: A PBSA-Based Free Energy Estimator for Protein–Ligand Binding Affinity. J Chem Inf Model 2016; 56:854-61. [DOI: 10.1021/acs.jcim.6b00001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Xiao Liu
- Department
of Physics, State Key Laboratory of Precision Spectroscopy, College
of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Jinfeng Liu
- Department
of Physics, State Key Laboratory of Precision Spectroscopy, College
of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Tong Zhu
- Department
of Physics, State Key Laboratory of Precision Spectroscopy, College
of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
- NYU−ECNU
Center for Computational Chemistry at NYU Shanghai, Shanghai 200062, China
| | - Lujia Zhang
- State
Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Xiao He
- Department
of Physics, State Key Laboratory of Precision Spectroscopy, College
of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
- NYU−ECNU
Center for Computational Chemistry at NYU Shanghai, Shanghai 200062, China
| | - John Z. H. Zhang
- Department
of Physics, State Key Laboratory of Precision Spectroscopy, College
of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
- NYU−ECNU
Center for Computational Chemistry at NYU Shanghai, Shanghai 200062, China
- Department
of Chemistry, New York University, New York, New York 10003, United States
- Collaborative
Innovation Center of Extreme Optics, Shanxi University, Taiyuan, Shanxi 030006, China
| |
Collapse
|
136
|
Wang Z, Sun H, Yao X, Li D, Xu L, Li Y, Tian S, Hou T. Comprehensive evaluation of ten docking programs on a diverse set of protein-ligand complexes: the prediction accuracy of sampling power and scoring power. Phys Chem Chem Phys 2016; 18:12964-75. [PMID: 27108770 DOI: 10.1039/c6cp01555g] [Citation(s) in RCA: 563] [Impact Index Per Article: 70.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
As one of the most popular computational approaches in modern structure-based drug design, molecular docking can be used not only to identify the correct conformation of a ligand within the target binding pocket but also to estimate the strength of the interaction between a target and a ligand. Nowadays, as a variety of docking programs are available for the scientific community, a comprehensive understanding of the advantages and limitations of each docking program is fundamentally important to conduct more reasonable docking studies and docking-based virtual screening. In the present study, based on an extensive dataset of 2002 protein-ligand complexes from the PDBbind database (version 2014), the performance of ten docking programs, including five commercial programs (LigandFit, Glide, GOLD, MOE Dock, and Surflex-Dock) and five academic programs (AutoDock, AutoDock Vina, LeDock, rDock, and UCSF DOCK), was systematically evaluated by examining the accuracies of binding pose prediction (sampling power) and binding affinity estimation (scoring power). Our results showed that GOLD and LeDock had the best sampling power (GOLD: 59.8% accuracy for the top scored poses; LeDock: 80.8% accuracy for the best poses) and AutoDock Vina had the best scoring power (rp/rs of 0.564/0.580 and 0.569/0.584 for the top scored poses and best poses), suggesting that the commercial programs did not show the expected better performance than the academic ones. Overall, the ligand binding poses could be identified in most cases by the evaluated docking programs but the ranks of the binding affinities for the entire dataset could not be well predicted by most docking programs. However, for some types of protein families, relatively high linear correlations between docking scores and experimental binding affinities could be achieved. To our knowledge, this study has been the most extensive evaluation of popular molecular docking programs in the last five years. It is expected that our work can offer useful information for the successful application of these docking tools to different requirements and targets.
Collapse
Affiliation(s)
- Zhe Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | | | | | | | | | | | | | | |
Collapse
|
137
|
Duan L, Liu X, Zhang JZ. Interaction Entropy: A New Paradigm for Highly Efficient and Reliable Computation of Protein–Ligand Binding Free Energy. J Am Chem Soc 2016; 138:5722-8. [DOI: 10.1021/jacs.6b02682] [Citation(s) in RCA: 205] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Lili Duan
- Department
of Physics, College of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
- School
of Physics and Electronics, Shandong Normal University, Jinan 250014, China
| | - Xiao Liu
- Department
of Physics, College of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - John Z.H. Zhang
- Department
of Physics, College of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
- NYU-ECNU Center for Computational Chemistry at NYU Shanghai, Shanghai 200062, China
- Department
of Chemistry, New York University, New York, New York 10003, United States
- Collaborative
Innovation Center of Extreme Optics, Shanxi University, Taiyuan, Shanxi 030006, PRC
| |
Collapse
|
138
|
Yan Z, Wang J. Incorporating specificity into optimization: evaluation of SPA using CSAR 2014 and CASF 2013 benchmarks. J Comput Aided Mol Des 2016; 30:219-27. [DOI: 10.1007/s10822-016-9897-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/28/2016] [Indexed: 01/04/2023]
|
139
|
Tanchuk VY, Tanin VO, Vovk AI, Poda G. A New, Improved Hybrid Scoring Function for Molecular Docking and Scoring Based on AutoDock and AutoDock Vina. Chem Biol Drug Des 2015; 87:618-25. [DOI: 10.1111/cbdd.12697] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 10/07/2015] [Accepted: 11/20/2015] [Indexed: 01/13/2023]
Affiliation(s)
- Vsevolod Yu Tanchuk
- Department of Bioorganic Mechanisms; Institute of Bioorganic Chemistry and Petrochemistry; National Academy of Sciences of Ukraine; 1 Murmanska Street-94 Kyiv 02660 Ukraine
| | - Volodymyr O. Tanin
- Department of Bioorganic Mechanisms; Institute of Bioorganic Chemistry and Petrochemistry; National Academy of Sciences of Ukraine; 1 Murmanska Street-94 Kyiv 02660 Ukraine
| | - Andriy I. Vovk
- Department of Bioorganic Mechanisms; Institute of Bioorganic Chemistry and Petrochemistry; National Academy of Sciences of Ukraine; 1 Murmanska Street-94 Kyiv 02660 Ukraine
| | - Gennady Poda
- Drug Discovery Program; Ontario Institute for Cancer Research (OICR); 661 University Avenue, Suite 510 Toronto ON Canada M5G 0A3
- Leslie Dan Faculty of Pharmacy; University of Toronto; 144 College Street Toronto ON Canada M5S 3M2
| |
Collapse
|
140
|
Lizunov AY, Gonchar AL, Zaitseva NI, Zosimov VV. Accounting for Intraligand Interactions in Flexible Ligand Docking with a PMF-Based Scoring Function. J Chem Inf Model 2015; 55:2121-37. [DOI: 10.1021/acs.jcim.5b00158] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- A. Y. Lizunov
- Department
of Mathematics, Moscow Institute of Physics and Technology, Moscow 117303, Russia
- Faculty
of Fundamental Medicine, Moscow State University, Moscow 119991, Russia
| | - A. L. Gonchar
- Faculty
of Fundamental Medicine, Moscow State University, Moscow 119991, Russia
| | - N. I. Zaitseva
- The
Faculty of Pharmacy, First Moscow State Medical University, Moscow 119991, Russia
| | - V. V. Zosimov
- Department
of Mathematics, Moscow Institute of Physics and Technology, Moscow 117303, Russia
| |
Collapse
|
141
|
Kumar A, Zhang KYJ. Application of Shape Similarity in Pose Selection and Virtual Screening in CSARdock2014 Exercise. J Chem Inf Model 2015; 56:965-73. [PMID: 26247231 DOI: 10.1021/acs.jcim.5b00279] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
To evaluate the applicability of shape similarity in docking-based pose selection and virtual screening, we participated in the CSARdock2014 benchmark exercise for identifying the correct docking pose of inhibitors targeting factor XA, spleen tyrosine kinase, and tRNA methyltransferase. This exercise provides a valuable opportunity for researchers to test their docking programs, methods, and protocols in a blind testing environment. In the CSARdock2014 benchmark exercise, we have implemented an approach that uses ligand 3D shape similarity to facilitate docking-based pose selection and virtual screening. We showed here that ligand 3D shape similarity between bound poses could be used to identify the native-like pose from an ensemble of docking-generated poses. Our method correctly identified the native pose as the top-ranking pose for 73% of test cases in a blind testing environment. Moreover, the pose selection results also revealed an excellent correlation between ligand 3D shape similarity scores and RMSD to X-ray crystal structure ligand. In the virtual screening exercise, the average RMSD for our pose prediction was found to be 1.02 Å, and it was one of the top performances achieved in CSARdock2014 benchmark exercise. Furthermore, the inclusion of shape similarity improved virtual screening performance of docking-based scoring and ranking. The coefficient of determination (r(2)) between experimental activities and docking scores for 276 spleen tyrosine kinase inhibitors was found to be 0.365 but reached 0.614 when the ligand 3D shape similarity was included.
Collapse
Affiliation(s)
- Ashutosh Kumar
- Structural Bioinformatics Team, Center for Life Science Technologies, RIKEN , 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Kam Y J Zhang
- Structural Bioinformatics Team, Center for Life Science Technologies, RIKEN , 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| |
Collapse
|
142
|
Yan Z, Wang J. Optimizing the affinity and specificity of ligand binding with the inclusion of solvation effect. Proteins 2015; 83:1632-42. [PMID: 26111900 DOI: 10.1002/prot.24848] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 06/03/2015] [Accepted: 06/21/2015] [Indexed: 01/08/2023]
Abstract
Solvation effect is an important factor for protein-ligand binding in aqueous water. Previous scoring function of protein-ligand interactions rarely incorporates the solvation model into the quantification of protein-ligand interactions, mainly due to the immense computational cost, especially in the structure-based virtual screening, and nontransferable application of independently optimized atomic solvation parameters. In order to overcome these barriers, we effectively combine knowledge-based atom-pair potentials and the atomic solvation energy of charge-independent implicit solvent model in the optimization of binding affinity and specificity. The resulting scoring functions with optimized atomic solvation parameters is named as specificity and affinity with solvation effect (SPA-SE). The performance of SPA-SE is evaluated and compared to 20 other scoring functions, as well as SPA. The comparative results show that SPA-SE outperforms all other scoring functions in binding affinity prediction and "native" pose identification. Our optimization validates that solvation effect is an important regulator to the stability and specificity of protein-ligand binding. The development strategy of SPA-SE sets an example for other scoring function to account for the solvation effect in biomolecular recognitions.
Collapse
Affiliation(s)
- Zhiqiang Yan
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun, Jilin, 130022, China
| | - Jin Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun, Jilin, 130022, China.,Department of Chemistry & Physics, State University of New York at Stony Brook, Stony Brook, New York, 11794-3400, USA
| |
Collapse
|
143
|
Liu Z, Li J, Liu J, Liu Y, Nie W, Han L, Li Y, Wang R. Cross-Mapping of Protein - Ligand Binding Data Between ChEMBL and PDBbind. Mol Inform 2015; 34:568-76. [DOI: 10.1002/minf.201500010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 04/17/2015] [Indexed: 02/06/2023]
|
144
|
Li H, Leung KS, Wong MH, Ballester PJ. Low-Quality Structural and Interaction Data Improves Binding Affinity Prediction via Random Forest. Molecules 2015; 20:10947-62. [PMID: 26076113 PMCID: PMC6272292 DOI: 10.3390/molecules200610947] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 06/04/2015] [Accepted: 06/09/2015] [Indexed: 12/17/2022] Open
Abstract
Docking scoring functions can be used to predict the strength of protein-ligand binding. It is widely believed that training a scoring function with low-quality data is detrimental for its predictive performance. Nevertheless, there is a surprising lack of systematic validation experiments in support of this hypothesis. In this study, we investigated to which extent training a scoring function with data containing low-quality structural and binding data is detrimental for predictive performance. We actually found that low-quality data is not only non-detrimental, but beneficial for the predictive performance of machine-learning scoring functions, though the improvement is less important than that coming from high-quality data. Furthermore, we observed that classical scoring functions are not able to effectively exploit data beyond an early threshold, regardless of its quality. This demonstrates that exploiting a larger data volume is more important for the performance of machine-learning scoring functions than restricting to a smaller set of higher data quality.
Collapse
Affiliation(s)
- Hongjian Li
- Department of Computer Science and Engineering, Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong.
| | - Kwong-Sak Leung
- Department of Computer Science and Engineering, Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong.
| | - Man-Hon Wong
- Department of Computer Science and Engineering, Chinese University of Hong Kong, Sha Tin, New Territories 999077, Hong Kong.
| | - Pedro J Ballester
- Cancer Research Center of Marseille, INSERM U1068, F-13009 Marseille, France.
| |
Collapse
|
145
|
Wei K, Wang GQ, Bai X, Niu YF, Chen HP, Wen CN, Li ZH, Dong ZJ, Zuo ZL, Xiong WY, Liu JK. Structure-Based Optimization and Biological Evaluation of Pancreatic Lipase Inhibitors as Novel Potential Antiobesity Agents. NATURAL PRODUCTS AND BIOPROSPECTING 2015; 5:129-157. [PMID: 26085282 PMCID: PMC4488150 DOI: 10.1007/s13659-015-0062-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 05/20/2015] [Indexed: 05/06/2023]
Abstract
The unusual fused β-lactone vibralactone was isolated from cultures of the basidiomycete Boreostereum vibrans and has been shown to significantly inhibit pancreatic lipase. In this study, a structure-based lead optimization of vibralactone resulted in three series of 104 analogs, among which compound C1 exhibited the most potent inhibition of pancreatic lipase, with an IC50 value of 14 nM. This activity is more than 3000-fold higher than that of vibralactone. The effect of compound C1 on obesity was investigated using high-fat diet (HFD)-induced C57BL/6 J obese mice. Treatment with compound C1 at a dose of 100 mg/kg significantly decreased HFD-induced obesity, primarily through the improvement of metabolic parameters, such as triglyceride levels.
Collapse
Affiliation(s)
- Kun Wei
- />State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 China
| | - Gang-Qiang Wang
- />State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 China
- />School of Nuclear Technology and Chemistry & Biology, Hubei University of Science and Technology, Xianning, 437100 China
| | - Xue Bai
- />State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 China
| | - Yan-Fen Niu
- />State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 China
| | - He-Ping Chen
- />State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 China
| | - Chun-Nan Wen
- />State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 China
| | - Zheng-Hui Li
- />State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 China
| | - Ze-Jun Dong
- />State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 China
| | - Zhi-Li Zuo
- />State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 China
| | - Wen-Yong Xiong
- />State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 China
| | - Ji-Kai Liu
- />State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201 China
| |
Collapse
|
146
|
Said AM, Hangauer DG. Binding cooperativity between a ligand carbonyl group and a hydrophobic side chain can be enhanced by additional H-bonds in a distance dependent manner: A case study with thrombin inhibitors. Eur J Med Chem 2015; 96:405-24. [DOI: 10.1016/j.ejmech.2015.03.059] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 03/21/2015] [Accepted: 03/25/2015] [Indexed: 01/07/2023]
|
147
|
Affiliation(s)
- Jie Liu
- State
Key Laboratory of Bioorganic and Natural Products Chemistry, Collaborative
Innovation Center of Chemistry for Life Sciences, Shanghai Institute
of Organic Chemistry, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Renxiao Wang
- State
Key Laboratory of Bioorganic and Natural Products Chemistry, Collaborative
Innovation Center of Chemistry for Life Sciences, Shanghai Institute
of Organic Chemistry, Chinese Academy of Sciences, Shanghai, People’s Republic of China
- State
Key Laboratory of Quality Research in Chinese Medicine, Macau Institute
for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, People’s Republic of China
| |
Collapse
|
148
|
The Use of Random Forest to Predict Binding Affinity in Docking. BIOINFORMATICS AND BIOMEDICAL ENGINEERING 2015. [DOI: 10.1007/978-3-319-16480-9_24] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
149
|
Ahmed A, Smith RD, Clark JJ, Dunbar JB, Carlson HA. Recent improvements to Binding MOAD: a resource for protein-ligand binding affinities and structures. Nucleic Acids Res 2014; 43:D465-9. [PMID: 25378330 PMCID: PMC4383918 DOI: 10.1093/nar/gku1088] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
For over 10 years, Binding MOAD (Mother of All Databases; http://www.BindingMOAD.org) has been one of the largest resources for high-quality protein-ligand complexes and associated binding affinity data. Binding MOAD has grown at the rate of 1994 complexes per year, on average. Currently, it contains 23,269 complexes and 8156 binding affinities. Our annual updates curate the data using a semi-automated literature search of the references cited within the PDB file, and we have recently upgraded our website and added new features and functionalities to better serve Binding MOAD users. In order to eliminate the legacy application server of the old platform and to accommodate new changes, the website has been completely rewritten in the LAMP (Linux, Apache, MySQL and PHP) environment. The improved user interface incorporates current third-party plugins for better visualization of protein and ligand molecules, and it provides features like sorting, filtering and filtered downloads. In addition to the field-based searching, Binding MOAD now can be searched by structural queries based on the ligand. In order to remove redundancy, Binding MOAD records are clustered in different families based on 90% sequence identity. The new Binding MOAD, with the upgraded platform, features and functionalities, is now equipped to better serve its users.
Collapse
Affiliation(s)
- Aqeel Ahmed
- Department of Medicinal Chemistry, University of Michigan, 428 Church St, Ann Arbor, MI 48109-1065, USA
| | - Richard D Smith
- Department of Medicinal Chemistry, University of Michigan, 428 Church St, Ann Arbor, MI 48109-1065, USA
| | - Jordan J Clark
- Department of Medicinal Chemistry, University of Michigan, 428 Church St, Ann Arbor, MI 48109-1065, USA
| | - James B Dunbar
- Department of Medicinal Chemistry, University of Michigan, 428 Church St, Ann Arbor, MI 48109-1065, USA
| | - Heather A Carlson
- Department of Medicinal Chemistry, University of Michigan, 428 Church St, Ann Arbor, MI 48109-1065, USA
| |
Collapse
|
150
|
Liu Z, Li Y, Han L, Li J, Liu J, Zhao Z, Nie W, Liu Y, Wang R. PDB-wide collection of binding data: current status of the PDBbind database. Bioinformatics 2014; 31:405-12. [DOI: 10.1093/bioinformatics/btu626] [Citation(s) in RCA: 264] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|