1
|
Zhou H, Skolnick J. Utility of the Morgan Fingerprint in Structure-Based Virtual Ligand Screening. J Phys Chem B 2024; 128:5363-5370. [PMID: 38783525 PMCID: PMC11163432 DOI: 10.1021/acs.jpcb.4c01875] [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: 03/21/2024] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
In modern drug discovery, virtual ligand screening (VLS) is frequently applied to identify possible hits before experimental testing and refinement due to its cost-effective nature for large compound libraries. For decades, efforts have been devoted to developing VLS methods with high accuracy. These include the state-of-the-art FINDSITE suite of approaches FINDSITEcomb2.0, FRAGSITE, and FRAGSITE2 and the meta version FRAGSITEcomb that were developed in our lab. These methods combine ligand homology modeling (LHM), traditional ligand similarity methods, and more recently machine learning approaches to rank ligands and have proven to be superior to most recent deep learning and large language model-based approaches. Here, we describe further improvements to our previous best methods by combining the Morgan fingerprint (MF) with the originally used PubChem fingerprint and FP2 fingerprint. We then benchmarked FINDSITEcomb2.0M, FRAGSITEM, FRAGSITE2M, and the composite meta-approach FRAGSITEcombM. On the 102 target DUD-E set, the 1% enrichment factor (EF1%) and area under the precision-recall curve (AUPR) of FRAGSITEcomb increased from 42.0/0.59 to 47.6/0.72. This 0.72 AUPR is significantly better than that of the state-of-the-art deep learning-based method DenseFS's AUPR of 0.443. An independent test on the 81 targets DEKOIS2.0 set shows that EF1%/AUPR increases from 18.3/0.520 to 23.1/0.683. An ablation investigation shows that the MF contributes to most of the improvement of all four approaches. Thus, the MF is a useful addition to structure-based VLS.
Collapse
Affiliation(s)
- Hongyi Zhou
- Center for the Study of Systems
Biology, School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Jeffrey Skolnick
- Center for the Study of Systems
Biology, School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
2
|
Zhou H, Skolnick J. FRAGSITE2: A structure and fragment-based approach for virtual ligand screening. Protein Sci 2024; 33:e4869. [PMID: 38100293 PMCID: PMC10751727 DOI: 10.1002/pro.4869] [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: 08/17/2023] [Revised: 12/06/2023] [Accepted: 12/09/2023] [Indexed: 12/17/2023]
Abstract
Protein function annotation and drug discovery often involve finding small molecule binders. In the early stages of drug discovery, virtual ligand screening (VLS) is frequently applied to identify possible hits before experimental testing. While our recent ligand homology modeling (LHM)-machine learning VLS method FRAGSITE outperformed approaches that combined traditional docking to generate protein-ligand poses and deep learning scoring functions to rank ligands, a more robust approach that could identify a more diverse set of binding ligands is needed. Here, we describe FRAGSITE2 that shows significant improvement on protein targets lacking known small molecule binders and no confident LHM identified template ligands when benchmarked on two commonly used VLS datasets: For both the DUD-E set and DEKOIS2.0 set and ligands having a Tanimoto coefficient (TC) < 0.7 to the template ligands, the 1% enrichment factor (EF1% ) of FRAGSITE2 is significantly better than those for FINDSITEcomb2.0 , an earlier LHM algorithm. For the DUD-E set, FRAGSITE2 also shows better ROC enrichment factor and AUPR (area under the precision-recall curve) than the deep learning DenseFS scoring function. Comparison with the RF-score-VS on the 76 target subset of DEKOIS2.0 and a TC < 0.99 to training DUD-E ligands, FRAGSITE2 has double the EF1% . Its boosted tree regression method provides for more robust performance than a deep learning multiple layer perceptron method. When compared with the pretrained language model for protein target features, FRAGSITE2 also shows much better performance. Thus, FRAGSITE2 is a promising approach that can discover novel hits for protein targets. FRAGSITE2's web service is freely available to academic users at http://sites.gatech.edu/cssb/FRAGSITE2.
Collapse
Affiliation(s)
- Hongyi Zhou
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Jeffrey Skolnick
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of TechnologyAtlantaGeorgiaUSA
| |
Collapse
|
3
|
Skolnick J, Zhou H. Implications of the Essential Role of Small Molecule Ligand Binding Pockets in Protein-Protein Interactions. J Phys Chem B 2022; 126:6853-6867. [PMID: 36044742 PMCID: PMC9484464 DOI: 10.1021/acs.jpcb.2c04525] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/18/2022] [Indexed: 11/28/2022]
Abstract
Protein-protein interactions (PPIs) and protein-metabolite interactions play a key role in many biochemical processes, yet they are often viewed as being independent. However, the fact that small molecule drugs have been successful in inhibiting PPIs suggests a deeper relationship between protein pockets that bind small molecules and PPIs. We demonstrate that 2/3 of PPI interfaces, including antibody-epitope interfaces, contain at least one significant small molecule ligand binding pocket. In a representative library of 50 distinct protein-protein interactions involving hundreds of mutations, >75% of hot spot residues overlap with small molecule ligand binding pockets. Hence, ligand binding pockets play an essential role in PPIs. In representative cases, evolutionary unrelated monomers that are involved in different multimeric interactions yet share the same pocket are predicted to bind the same metabolites/drugs; these results are confirmed by examples in the PDB. Thus, the binding of a metabolite can shift the equilibrium between monomers and multimers. This implicit coupling of PPI equilibria, termed "metabolic entanglement", was successfully employed to suggest novel functional relationships among protein multimers that do not directly interact. Thus, the current work provides an approach to unify metabolomics and protein interactomics.
Collapse
Affiliation(s)
- Jeffrey Skolnick
- Center for the Study of Systems
Biology, School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Drive, NW, Atlanta, Georgia 30332, United States
| | - Hongyi Zhou
- Center for the Study of Systems
Biology, School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Drive, NW, Atlanta, Georgia 30332, United States
| |
Collapse
|
4
|
Vigil-Vásquez C, Schüller A. De Novo Prediction of Drug Targets and Candidates by Chemical Similarity-Guided Network-Based Inference. Int J Mol Sci 2022; 23:ijms23179666. [PMID: 36077062 PMCID: PMC9455815 DOI: 10.3390/ijms23179666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/12/2022] [Accepted: 08/21/2022] [Indexed: 12/01/2022] Open
Abstract
Identifying drug–target interactions is a crucial step in discovering novel drugs and for drug repositioning. Network-based methods have shown great potential thanks to the straightforward integration of information from different sources and the possibility of extracting novel information from the graph topology. However, despite recent advances, there is still an urgent need for efficient and robust prediction methods. Here, we present SimSpread, a novel method that combines network-based inference with chemical similarity. This method employs a tripartite drug–drug–target network constructed from protein–ligand interaction annotations and drug–drug chemical similarity on which a resource-spreading algorithm predicts potential biological targets for both known or failed drugs and novel compounds. We describe small molecules as vectors of similarity indices to other compounds, thereby providing a flexible means to explore diverse molecular representations. We show that our proposed method achieves high prediction performance through multiple cross-validation and time-split validation procedures over a series of datasets. In addition, we demonstrate that our method performed a balanced exploration of both chemical ligand space (scaffold hopping) and biological target space (target hopping). Our results suggest robust and balanced performance, and our method may be useful for predicting drug targets, virtual screening, and drug repositioning.
Collapse
Affiliation(s)
- Carlos Vigil-Vásquez
- Department of Molecular Genetics and Microbiology, School of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Andreas Schüller
- Department of Molecular Genetics and Microbiology, School of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Correspondence:
| |
Collapse
|
5
|
Zhang W, Huang J. EViS: An Enhanced Virtual Screening Approach Based on Pocket-Ligand Similarity. J Chem Inf Model 2022; 62:498-510. [PMID: 35084171 DOI: 10.1021/acs.jcim.1c00944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Virtual screening (VS) is a popular technology in drug discovery to identify a new scaffold of actives for a specific drug target, which can be classified into ligand-based and structure-based approaches. As the number of protein-ligand complex structures available in public databases increases, it would be possible to develop a template searching-based VS approach that utilizes such information. In this work, we proposed an enhanced VS approach, which is termed EViS, to integrate ligand docking, protein pocket template searching, and ligand template shape similarity calculation. A novel and simple PL-score to characterize local pocket-ligand template similarity was used to evaluate the screening compounds. Benchmark tests were performed on three datasets including DUDE, LIT-PCBA, and DEKOIS. EViS achieved the average enrichment factors (EFs) of 27.8 and 23.4 at a 1% cutoff for experimental and predicted structures on the widely used DUDE dataset, respectively. Detailed data analysis shows that EViS benefits from obtaining favorable ligand poses from docking and using such ligand geometric information to perform three-dimensional (3D) ligand similarity calculations, and the PL-score is efficient to screen compounds based on template searching in the protein-ligand structure database.
Collapse
Affiliation(s)
- Wenyi Zhang
- Westlake AI Therapeutics Lab, Westlake Laboratory of Life Sciences and Biomedicine, 18 Shilongshan Road, Hangzhou, Zhejiang 310024, China.,Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou, Zhejiang 310024, China.,Institute of Biology, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou, Zhejiang 310024, China
| | - Jing Huang
- Westlake AI Therapeutics Lab, Westlake Laboratory of Life Sciences and Biomedicine, 18 Shilongshan Road, Hangzhou, Zhejiang 310024, China.,Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, 18 Shilongshan Road, Hangzhou, Zhejiang 310024, China.,Institute of Biology, Westlake Institute for Advanced Study, 18 Shilongshan Road, Hangzhou, Zhejiang 310024, China
| |
Collapse
|
6
|
Zhang B, Li H, Yu K, Jin Z. Molecular docking-based computational platform for high-throughput virtual screening. CCF TRANSACTIONS ON HIGH PERFORMANCE COMPUTING 2022; 4:63-74. [PMID: 35039800 PMCID: PMC8754542 DOI: 10.1007/s42514-021-00086-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 11/12/2021] [Indexed: 05/03/2023]
Abstract
Structure-based virtual screening is a key, routine computational method in computer-aided drug design. Such screening can be used to identify potentially highly active compounds, to speed up the progress of novel drug design. Molecular docking-based virtual screening can help find active compounds from large ligand databases by identifying the binding affinities between receptors and ligands. In this study, we analyzed the challenges of virtual screening, with the aim of identifying highly active compounds faster and more easily than is generally possible. We discuss the accuracy and speed of molecular docking software and the strategy of high-throughput molecular docking calculation, and we focus on current challenges and our solutions to these challenges of ultra-large-scale virtual screening. The development of Web services helps lower the barrier to drug virtual screening. We introduced some related web sites for docking and virtual screening, focusing on the development of pre- and post-processing interactive visualization and large-scale computing.
Collapse
Affiliation(s)
- Baohua Zhang
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Hui Li
- Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, 201203 China
- Shanghai Institute for Advanced Immunochemical Studies, and School of Life Science and Technology, Shanghai Tech University, Shanghai, 200031 China
| | - Kunqian Yu
- Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, 201203 China
| | - Zhong Jin
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100190 China
| |
Collapse
|
7
|
Clyde A. Ultrahigh Throughput Protein-Ligand Docking with Deep Learning. Methods Mol Biol 2022; 2390:301-319. [PMID: 34731475 DOI: 10.1007/978-1-0716-1787-8_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Ultrahigh-throughput virtual screening (uHTVS) is an emerging field linking together classical docking techniques with high-throughput AI methods. We outline mechanistic docking models' goals and successes. We present different AI accelerated workflows for uHTVS, mainly through surrogate docking models. We showcase a novel feature representation technique, molecular depictions (images), as a surrogate model for docking. Along with a discussion on analyzing screens using regression enrichment surfaces at the tens of billion scale, we outline a future for uHTVS screening pipelines with deep learning.
Collapse
Affiliation(s)
- Austin Clyde
- Department of Computer Science, University of Chicago, Chicago, IL, USA.
- Data Science and Learning Division, Argonne National Laboratory, Lemont, IL, USA.
| |
Collapse
|
8
|
Zhou H, Cao H, Skolnick J. FRAGSITE: A Fragment-Based Approach for Virtual Ligand Screening. J Chem Inf Model 2021; 61:2074-2089. [PMID: 33724022 DOI: 10.1021/acs.jcim.0c01160] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
To reduce time and cost, virtual ligand screening (VLS) often precedes experimental ligand screening in modern drug discovery. Traditionally, high-resolution structure-based docking approaches rely on experimental structures, while ligand-based approaches need known binders to the target protein and only explore their nearby chemical space. In contrast, our structure-based FINDSITEcomb2.0 approach takes advantage of predicted, low-resolution structures and information from ligands that bind distantly related proteins whose binding sites are similar to the target protein. Using a boosted tree regression machine learning framework, we significantly improved FINDSITEcomb2.0 by integrating ligand fragment scores as encoded by molecular fingerprints with the global ligand similarity scores of FINDSITEcomb2.0. The new approach, FRAGSITE, exploits our observation that ligand fragments, e.g., rings, tend to interact with stereochemically conserved protein subpockets that also occur in evolutionarily unrelated proteins. FRAGSITE was benchmarked on the 102 protein DUD-E set, where any template protein whose sequence identify >30% to the target was excluded. Within the top 100 ranked molecules, FRAGSITE improves VLS precision and recall by 14.3 and 18.5%, respectively, relative to FINDSITEcomb2.0. Moreover, the mean top 1% enrichment factor increases from 25.2 to 30.2. On average, both outperform state-of-the-art deep learning-based methods such as AtomNet. On the more challenging unbiased set LIT-PCBA, FRAGSITE also shows better performance than ligand similarity-based and docking approaches such as two-dimensional ECFP4 and Surflex-Dock v.3066. On a subset of 23 targets from DEKOIS 2.0, FRAGSITE shows much better performance than the boosted tree regression-based, vScreenML scoring function. Experimental testing of FRAGSITE's predictions shows that it has more hits and covers a more diverse region of chemical space than FINDSITEcomb2.0. For the two proteins that were experimentally tested, DHFR, a well-studied protein that catalyzes the conversion of dihydrofolate to tetrahydrofolate, and the kinase ACVR1, FRAGSITE identified new small-molecule nanomolar binders. Interestingly, one new binder of DHFR is a kinase inhibitor predicted to bind in a new subpocket. For ACVR1, FRAGSITE identified new molecules that have diverse scaffolds and estimated nanomolar to micromolar affinities. Thus, FRAGSITE shows significant improvement over prior state-of-the-art ligand virtual screening approaches. A web server is freely available for academic users at http:/sites.gatech.edu/cssb/FRAGSITE.
Collapse
Affiliation(s)
- Hongyi Zhou
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Drive, NW, Atlanta, Georgia 30332-2000, United States
| | - Hongnan Cao
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Drive, NW, Atlanta, Georgia 30332-2000, United States
| | - Jeffrey Skolnick
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Drive, NW, Atlanta, Georgia 30332-2000, United States
| |
Collapse
|
9
|
Identification of Potential HCV Inhibitors Based on the Interaction of Epigallocatechin-3-Gallate with Viral Envelope Proteins. Molecules 2021; 26:molecules26051257. [PMID: 33652639 PMCID: PMC7956288 DOI: 10.3390/molecules26051257] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/14/2021] [Accepted: 02/20/2021] [Indexed: 12/25/2022] Open
Abstract
Hepatitis C is affecting millions of people around the globe annually, which leads to death in very high numbers. After many years of research, hepatitis C virus (HCV) remains a serious threat to the human population and needs proper management. The in silico approach in the drug discovery process is an efficient method in identifying inhibitors for various diseases. In our study, the interaction between Epigallocatechin-3-gallate, a component of green tea, and envelope glycoprotein E2 of HCV is evaluated. Epigallocatechin-3-gallate is the most promising polyphenol approved through cell culture analysis that can inhibit the entry of HCV. Therefore, various in silico techniques have been employed to find out other potential inhibitors that can behave as EGCG. Thus, the homology modelling of E2 protein was performed. The potential lead molecules were predicted using ligand-based as well as structure-based virtual screening methods. The compounds obtained were then screened through PyRx. The drugs obtained were ranked based on their binding affinities. Furthermore, the docking of the topmost drugs was performed by AutoDock Vina, while its 2D interactions were plotted in LigPlot+. The lead compound mms02387687 (2-[[5-[(4-ethylphenoxy) methyl]-4-prop-2-enyl-1,2,4-triazol-3-yl] sulfanyl]-N-[3(trifluoromethyl) phenyl] acetamide) was ranked on top, and we believe it can serve as a drug against HCV in the future, owing to experimental validation.
Collapse
|
10
|
Zhou H, Cao H, Matyunina L, Shelby M, Cassels L, McDonald JF, Skolnick J. MEDICASCY: A Machine Learning Approach for Predicting Small-Molecule Drug Side Effects, Indications, Efficacy, and Modes of Action. Mol Pharm 2020; 17:1558-1574. [PMID: 32237745 PMCID: PMC7319183 DOI: 10.1021/acs.molpharmaceut.9b01248] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
To improve the drug discovery yield, a method which is implemented at the beginning of drug discovery that accurately predicts drug side effects, indications, efficacy, and mode of action based solely on the input of the drug's chemical structure is needed. In contrast, extant predictive methods do not comprehensively address these aspects of drug discovery and rely on features derived from extensive, often unavailable experimental information for novel molecules. To address these issues, we developed MEDICASCY, a multilabel-based boosted random forest machine learning method that only requires the small molecule's chemical structure for the drug side effect, indication, efficacy, and probable mode of action target predictions; however, it has comparable or even significantly better performance than existing approaches requiring far more information. In retrospective benchmarking on high confidence predictions, MEDICASCY shows about 78% precision and recall for predicting at least one severe side effect and 72% precision drug efficacy. Experimental validation of MEDICASCY's efficacy predictions on novel molecules shows close to 80% precision for the inhibition of growth in ovarian, breast, and prostate cancer cell lines. Thus, MEDICASCY should improve the success rate for new drug approval. A web service for academic users is available at http://pwp.gatech.edu/cssb/MEDICASCY.
Collapse
Affiliation(s)
- Hongyi Zhou
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Drive, N.W., Atlanta, GA 30332
| | - Hongnan Cao
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Drive, N.W., Atlanta, GA 30332
| | - Lilya Matyunina
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, 30332-0230, USA
| | - Madelyn Shelby
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, 30332-0230, USA
| | - Lauren Cassels
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, 30332-0230, USA
| | - John F. McDonald
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, 30332-0230, USA
| | - Jeffrey Skolnick
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Drive, N.W., Atlanta, GA 30332
| |
Collapse
|
11
|
Sachdev K, Gupta MK. A comprehensive review of computational techniques for the prediction of drug side effects. Drug Dev Res 2020; 81:650-670. [DOI: 10.1002/ddr.21669] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/18/2020] [Accepted: 03/30/2020] [Indexed: 12/28/2022]
Affiliation(s)
- Kanica Sachdev
- School of Computer Science and EngineeringShri Mata Vaishno Devi University Katra Jammu and Kashmir India
| | - Manoj K. Gupta
- School of Computer Science and EngineeringShri Mata Vaishno Devi University Katra Jammu and Kashmir India
| |
Collapse
|
12
|
He H, Liu B, Luo H, Zhang T, Jiang J. Big data and artificial intelligence discover novel drugs targeting proteins without 3D structure and overcome the undruggable targets. Stroke Vasc Neurol 2020; 5:381-387. [PMID: 33376199 PMCID: PMC7804061 DOI: 10.1136/svn-2019-000323] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 02/29/2020] [Accepted: 03/03/2020] [Indexed: 12/27/2022] Open
Abstract
The discovery of targeted drugs heavily relies on three-dimensional (3D) structures of target proteins. When the 3D structure of a protein target is unknown, it is very difficult to design its corresponding targeted drugs. Although the 3D structures of some proteins (the so-called undruggable targets) are known, their targeted drugs are still absent. As increasing crystal/cryogenic
electron microscopy structures are deposited in Protein Data Bank, it is much more possible to discover the targeted drugs. Moreover, it is also highly probable to turn previous undruggable targets into druggable ones when we identify their hidden allosteric sites. In this review, we focus on the currently available advanced methods for the discovery of novel compounds targeting proteins without 3D structure and how to turn undruggable targets into druggable ones.
Collapse
Affiliation(s)
- Huiqin He
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Benquan Liu
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Hongyi Luo
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Tingting Zhang
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Jingwei Jiang
- Institute of Pharmacologic Science, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
13
|
Alekseenko A, Kotelnikov S, Ignatov M, Egbert M, Kholodov Y, Vajda S, Kozakov D. ClusPro LigTBM: Automated Template-based Small Molecule Docking. J Mol Biol 2019; 432:3404-3410. [PMID: 31863748 DOI: 10.1016/j.jmb.2019.12.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/03/2019] [Accepted: 12/04/2019] [Indexed: 12/31/2022]
Abstract
The template-based approach has been essential for achieving high-quality models in the recent rounds of blind protein-protein docking competition CAPRI (Critical Assessment of Predicted Interactions). However, few such automated methods exist for protein-small molecule docking. In this paper, we present an algorithm for template-based docking of small molecules. It searches for known complexes with ligands that have partial coverage of the target ligand, performs conformational sampling and template-guided energy refinement to produce a variety of possible poses, and then scores the refined poses. The algorithm is available as the automated ClusPro LigTBM server. It allows the user to specify the target protein as a PDB file and the ligand as a SMILES string. The server then searches for templates and uses them for docking, presenting the user with top-scoring poses and their confidence scores. The method is tested on the Astex Diverse benchmark, as well as on the targets from the last round of the D3R (Drug Design Data Resource) Grand Challenge. The server is publicly available as part of the ClusPro docking server suite at https://ligtbm.cluspro.org/.
Collapse
Affiliation(s)
- Andrey Alekseenko
- Department of Applied Mathematics and Statistics, Stony Brook University, 11794 Stony Brook, NY, USA; Laufer Center for Physical and Quantitative Biology, Stony Brook University, 11794 Stony Brook, NY, USA
| | - Sergei Kotelnikov
- Department of Applied Mathematics and Statistics, Stony Brook University, 11794 Stony Brook, NY, USA; Laufer Center for Physical and Quantitative Biology, Stony Brook University, 11794 Stony Brook, NY, USA; Innopolis University, 420500, Innopolis, Russia
| | - Mikhail Ignatov
- Department of Applied Mathematics and Statistics, Stony Brook University, 11794 Stony Brook, NY, USA; Laufer Center for Physical and Quantitative Biology, Stony Brook University, 11794 Stony Brook, NY, USA; Institute for Advanced Computational Sciences, Stony Brook University, 11794, Stony Brook, NY, USA
| | - Megan Egbert
- Department of Biomedical Engineering, Boston University, 02215, Boston, MA, USA
| | | | - Sandor Vajda
- Department of Biomedical Engineering, Boston University, 02215, Boston, MA, USA; Department of Chemistry, Boston University, 02215, Boston, MA, USA
| | - Dima Kozakov
- Department of Applied Mathematics and Statistics, Stony Brook University, 11794 Stony Brook, NY, USA; Laufer Center for Physical and Quantitative Biology, Stony Brook University, 11794 Stony Brook, NY, USA; Institute for Advanced Computational Sciences, Stony Brook University, 11794, Stony Brook, NY, USA.
| |
Collapse
|
14
|
Computational methods and tools for binding site recognition between proteins and small molecules: from classical geometrical approaches to modern machine learning strategies. J Comput Aided Mol Des 2019; 33:887-903. [PMID: 31628659 DOI: 10.1007/s10822-019-00235-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 10/11/2019] [Indexed: 10/25/2022]
Abstract
In the current "genomic era" the number of identified genes is growing exponentially. However, the biological function of a large number of the corresponding proteins is still unknown. Recognition of small molecule ligands (e.g., substrates, inhibitors, allosteric regulators, etc.) is pivotal for protein functions in the vast majority of the cases and knowledge of the region where these processes take place is essential for protein function prediction and drug design. In this regard, computational methods represent essential tools to tackle this problem. A significant number of software tools have been developed in the last few years which exploit either protein sequence information, structure information or both. This review describes the most recent developments in protein function recognition and binding site prediction, in terms of both freely-available and commercial solutions and tools, detailing the main characteristics of the considered tools and providing a comparative analysis of their performance.
Collapse
|
15
|
Durán-Pérez SA, López-Moreno HS, Jiménez-Edeza M, Parra-Unda JR, Rangel-López E, Rendón-Maldonado JG. Upregulation of Cathepsin B-like Protease Activity During Apoptosis inGiardia duodenalis. CURR PROTEOMICS 2019. [DOI: 10.2174/1570164616666190204112452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:In eukaryotic cells, apoptosis signaling pathways are controlled mainly by aspartic acid cysteine proteases (caspases). However, certain unicellular microorganisms, such as Giardia duodenalis, lack these proteins. Thus, other cysteine proteases may play an important role in the parasite apoptosis signaling pathway.Objective:To understand the effect of cathepsin B-like inhibition on the cell viability of Giardia duodenalis and its cell death process.Methods:Bioinformatics analysis was performed to identify apoptotic proteases. Analysis showed that cathepsin B-like protease genes from G. duodenalis were the best candidate. A homology modeling technique was used to explore in silico the inhibitory effect of E-64 against cathepsin B-like proteases from G. duodenalis genome and to examine the effect of curcumin on cathepsin B-like activity regulation. In addition, the effect of E-64 on parasite survival and DNA fragmentation was tested.Results:Eight cathepsin B-like protease coding genes were identified in silico. Interestingly, while these sequences lacked the cathepsin B characteristic occluding loop, they maintained the catalytic active- site responsible for cathepsin B activity, which was evidenced by the increase in the degradation of the Z-RR-AMC substrate, suggesting the upregulation of the activity of these proteins. Additionally, inhibition of E-64 against G. duodenalis trophozoites caused a decrease in DNA fragmentation compared to control cells and had a positive effect on parasite survival after exposure to curcumin.Conclusion:Overall, these results suggested that Giardia duodenalis might have a cell death mechanism in which cathepsin B-like proteases play an important role.
Collapse
Affiliation(s)
- Sergio Alonso Durán-Pérez
- Doctorate in Biotechnology, Faculty of Biological Chemistry Sciences, Autonomous University of Sinaloa, Calzada de las Americas Norte 2771, Bureaucrat, 80030 Culiacan, Sinaloa, Mexico
| | - Héctor Samuel López-Moreno
- Doctorate in Biotechnology, Faculty of Biological Chemistry Sciences, Autonomous University of Sinaloa, Calzada de las Americas Norte 2771, Bureaucrat, 80030 Culiacan, Sinaloa, Mexico
| | - Maribel Jiménez-Edeza
- Doctorate in Biomedical Sciences, Faculty of Biological Chemistry Sciences, Autonomous University of Sinaloa, Calzada de las Americas Norte 2771, Bureaucrat, 80030 Culiacan, Sinaloa, Mexico
| | - Jesús Ricardo Parra-Unda
- Doctorate in Biotechnology, Faculty of Biological Chemistry Sciences, Autonomous University of Sinaloa, Calzada de las Americas Norte 2771, Bureaucrat, 80030 Culiacan, Sinaloa, Mexico
| | - Edgar Rangel-López
- Laboratory of Amino Acids Exciters, National Institute of Neurology and Neurosurgery Manuel Velasco Suarez, Insurgentes Sur 3877, 14269, Mexico City, Mexico
| | - José Guadalupe Rendón-Maldonado
- Doctorate in Biotechnology, Faculty of Biological Chemistry Sciences, Autonomous University of Sinaloa, Calzada de las Americas Norte 2771, Bureaucrat, 80030 Culiacan, Sinaloa, Mexico
| |
Collapse
|
16
|
Litfin T, Yang Y, Zhou Y. SPOT-Peptide: Template-Based Prediction of Peptide-Binding Proteins and Peptide-Binding Sites. J Chem Inf Model 2019; 59:924-930. [DOI: 10.1021/acs.jcim.8b00777] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Thomas Litfin
- School of Information and Communication Technology, Griffith University, Southport, QLD 4222, Australia
| | - Yuedong Yang
- School of Data and Computer Science, Sun-Yat Sen University, Guangzhou, Guangdong 510006, China
| | - Yaoqi Zhou
- School of Information and Communication Technology, Griffith University, Southport, QLD 4222, Australia
- Institute for Glycomics, Griffith University, Southport, QLD 4222, Australia
| |
Collapse
|
17
|
Zhou H, Cao H, Skolnick J. FINDSITE comb2.0: A New Approach for Virtual Ligand Screening of Proteins and Virtual Target Screening of Biomolecules. J Chem Inf Model 2018; 58:2343-2354. [PMID: 30278128 PMCID: PMC6437778 DOI: 10.1021/acs.jcim.8b00309] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Computational approaches for predicting protein-ligand interactions can facilitate drug lead discovery and drug target determination. We have previously developed a threading/structural-based approach, FINDSITEcomb, for the virtual ligand screening of proteins that has been extensively experimentally validated. Even when low resolution predicted protein structures are employed, FINDSITEcomb has the advantage of being faster and more accurate than traditional high-resolution structure-based docking methods. It also overcomes the limitations of traditional QSAR methods that require a known set of seed ligands that bind to the given protein target. Here, we further improve FINDSITEcomb by enhancing its template ligand selection from the PDB/DrugBank/ChEMBL libraries of known protein-ligand interactions by (1) parsing the template proteins and their corresponding binding ligands in the DrugBank and ChEMBL libraries into domains so that the ligands with falsely matched domains to the targets will not be selected as template ligands; (2) applying various thresholds to filter out falsely matched template structures in the structure comparison process and thus their corresponding ligands for template ligand selection. With a sequence identity cutoff of 30% of target to templates and modeled target structures, FINDSITEcomb2.0 is shown to significantly improve upon FINDSITEcomb on the DUD-E benchmark set by increasing the 1% enrichment factor from 16.7 to 22.1, with a p-value of 4.3 × 10-3 by the Student t-test. With an 80% sequence identity cutoff of target to templates for the DUD-E set and modeled target structures, FINDSITEcomb2.0, having a 1% ROC enrichment factor of 52.39, also outperforms state-of-the-art methods that employ machine learning such as a deep convolutional neural network, CNN, with an enrichment of 29.65. Thus, FINDSITEcomb2.0 represents a significant improvement in the state-of-the-art. The FINDSITEcomb2.0 web service is freely available for academic users at http://pwp.gatech.edu/cssb/FINDSITE-COMB-2 .
Collapse
Affiliation(s)
- Hongyi Zhou
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Drive, NW, Atlanta, GA 30332-2000
| | - Hongnan Cao
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Drive, NW, Atlanta, GA 30332-2000
| | - Jeffrey Skolnick
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology, 950 Atlantic Drive, NW, Atlanta, GA 30332-2000
| |
Collapse
|
18
|
Srinivasan B, Tonddast-Navaei S, Roy A, Zhou H, Skolnick J. Chemical space of Escherichia coli dihydrofolate reductase inhibitors: New approaches for discovering novel drugs for old bugs. Med Res Rev 2018; 39:684-705. [PMID: 30192413 DOI: 10.1002/med.21538] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/16/2018] [Accepted: 08/09/2018] [Indexed: 12/15/2022]
Abstract
Escherichia coli Dihydrofolate reductase is an important enzyme that is essential for the survival of the Gram-negative microorganism. Inhibitors designed against this enzyme have demonstrated application as antibiotics. However, either because of poor bioavailability of the small-molecules resulting from their inability to cross the double membrane in Gram-negative bacteria or because the microorganism develops resistance to the antibiotics by mutating the DHFR target, discovery of new antibiotics against the enzyme is mandatory to overcome drug-resistance. This review summarizes the field of DHFR inhibition with special focus on recent efforts to effectively interface computational and experimental efforts to discover novel classes of inhibitors that target allosteric and active-sites in drug-resistant variants of EcDHFR.
Collapse
Affiliation(s)
- Bharath Srinivasan
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, Atlanta, Georgia
| | - Sam Tonddast-Navaei
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, Atlanta, Georgia
| | - Ambrish Roy
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, Atlanta, Georgia
| | - Hongyi Zhou
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, Atlanta, Georgia
| | - Jeffrey Skolnick
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
19
|
Snell TW, Johnston RK, Matthews AB, Zhou H, Gao M, Skolnick J. Repurposed FDA-approved drugs targeting genes influencing aging can extend lifespan and healthspan in rotifers. Biogerontology 2018; 19:145-157. [PMID: 29340835 PMCID: PMC5834582 DOI: 10.1007/s10522-018-9745-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 01/09/2018] [Indexed: 12/29/2022]
Abstract
Pharmaceutical interventions can slow aging in animals, and have advantages because their dose can be tightly regulated and the timing of the intervention can be closely controlled. They also may complement environmental interventions like caloric restriction by acting additively. A fertile source for therapies slowing aging is FDA approved drugs whose safety has been investigated. Because drugs bind to several protein targets, they cause multiple effects, many of which have not been characterized. It is possible that some of the side effects of drugs prescribed for one therapy may have benefits in retarding aging. We used computationally guided drug screening for prioritizing drug targets to produce a short list of candidate compounds for in vivo testing. We applied the virtual ligand screening approach FINDSITEcomb for screening potential anti-aging protein targets against FDA approved drugs listed in DrugBank. A short list of 31 promising compounds was screened using a multi-tiered approach with rotifers as an animal model of aging. Primary and secondary survival screens and cohort life table experiments identified four drugs capable of extending rotifer lifespan by 8-42%. Exposures to 1 µM erythromycin, 5 µM carglumic acid, 3 µM capecitabine, and 1 µM ivermectin, extended rotifer lifespan without significant effect on reproduction. Some drugs also extended healthspan, as estimated by mitochondria activity and mobility (swimming speed). Our most promising result is that rotifer lifespan was extended by 7-8.9% even when treatment was started in middle age.
Collapse
Affiliation(s)
- Terry W Snell
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332-0230, USA.
| | - Rachel K Johnston
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332-0230, USA
| | - Amelia B Matthews
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332-0230, USA
| | - Hongyi Zhou
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332-0230, USA
| | - Mu Gao
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332-0230, USA
| | - Jeffrey Skolnick
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332-0230, USA
| |
Collapse
|
20
|
Ranking Enzyme Structures in the PDB by Bound Ligand Similarity to Biological Substrates. Structure 2018; 26:565-571.e3. [PMID: 29551288 PMCID: PMC5890617 DOI: 10.1016/j.str.2018.02.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 01/26/2018] [Accepted: 02/09/2018] [Indexed: 11/22/2022]
Abstract
There are numerous applications that use the structures of protein-ligand complexes from the PDB, such as 3D pharmacophore identification, virtual screening, and fragment-based drug design. The structures underlying these applications are potentially much more informative if they contain biologically relevant bound ligands, with high similarity to the cognate ligands. We present a study of ligand-enzyme complexes that compares the similarity of bound and cognate ligands, enabling the best matches to be identified. We calculate the molecular similarity scores using a method called PARITY (proportion of atoms residing in identical topology), which can conveniently be combined to give a similarity score for all cognate reactants or products in the reaction. Thus, we generate a rank-ordered list of related PDB structures, according to the biological similarity of the ligands bound in the structures. We present PARITY, matching atoms in identical topology to gauge ligand similarity Bound-cognate ligand similarity is a useful metric for ranking PDB structures Only 26% of enzyme structures in the PDB have bound-cognate ligand similarity ≥0.7 We provide rank-ordered lists of PDBs with the most biologically relevant ligands
Collapse
|
21
|
Sam E, Athri P. Web-based drug repurposing tools: a survey. Brief Bioinform 2017; 20:299-316. [DOI: 10.1093/bib/bbx125] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Indexed: 12/15/2022] Open
Affiliation(s)
- Elizabeth Sam
- Department of Computer Science & Engineering Amrita, University Bengaluru, India
| | - Prashanth Athri
- Department of Computer Science & Engineering Amrita, University Bengaluru, India
| |
Collapse
|
22
|
Srinivasan B, Tonddast-Navaei S, Skolnick J. Pocket detection and interaction-weighted ligand-similarity search yields novel high-affinity binders for Myocilin-OLF, a protein implicated in glaucoma. Bioorg Med Chem Lett 2017; 27:4133-4139. [PMID: 28739043 DOI: 10.1016/j.bmcl.2017.07.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/10/2017] [Accepted: 07/11/2017] [Indexed: 10/19/2022]
Abstract
Traditional structure and ligand based virtual screening approaches rely on the availability of structural and ligand binding information. To overcome this limitation, hybrid approaches were developed that relied on extraction of ligand binding information from proteins sharing similar folds and hence, evolutionarily relationship. However, they cannot target a chosen pocket in a protein. To address this, a pocket centric virtual ligand screening approach is required. Here, we employ a new, iterative implementation of a pocket and ligand-similarity based approach to virtual ligand screening to predict small molecule binders for the olfactomedin domain of human myocilin implicated in glaucoma. Small-molecule binders of the protein might prevent the aggregation of the protein, commonly seen during glaucoma. First round experimental assessment of the predictions using differential scanning fluorimetry with myoc-OLF yielded 7 hits with a success rate of 12.7%; the best hit had an apparent dissociation constant of 99nM. By matching to the key functional groups of the best ligand that were likely involved in binding, the affinity of the best hit was improved by almost 10,000 fold from the high nanomolar to the low picomolar range. Thus, this study provides preliminary validation of the methodology on a medically important glaucoma associated protein.
Collapse
Affiliation(s)
- Bharath Srinivasan
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, 950, Atlantic Drive, Atlanta, GA 30332, United States
| | - Sam Tonddast-Navaei
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, 950, Atlantic Drive, Atlanta, GA 30332, United States
| | - Jeffrey Skolnick
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, 950, Atlantic Drive, Atlanta, GA 30332, United States.
| |
Collapse
|
23
|
Shah MA, Ullah R, March MD, Shah MS, Ismat F, Habib M, Iqbal M, Onesti S, Rahman M. Overexpression and characterization of the 100K protein of Fowl adenovirus-4 as an antiviral target. Virus Res 2017; 238:218-225. [PMID: 28666898 DOI: 10.1016/j.virusres.2017.06.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/26/2017] [Accepted: 06/27/2017] [Indexed: 10/19/2022]
Abstract
100K is an important scaffolding protein of adenoviruses including fowl adenovirus serotype 4 (FAdV-4) that causes inclusion body hepatitis-hydropericardium syndrome (IBH-HPS) in poultry. 100K carries out the trimerization of the major capsid hexon protein of the virus for the generation of new virions inside the target host cells. Despite its critical role for FAdV-4, no structural study, in particular, has been conducted so far. Here, the overexpression of soluble 100K protein was successfully carried out in E. coli using various expression constructs and purification yield of 3mg per litre culture volume was obtained. Gel filtration chromatography suggested that 100K protein exists in trimeric form. Circular dichroism and Fourier transform infrared spectroscopy clearly reveal that 100K protein folds with a high content of α-helices. The 3-dimentional homology model of the 100K protein, refined with molecular dynamics tools also depicts higher α-helical content within the protein model. Moreover, overexpressed recombinant 100K protein could be used to differentiate vaccinated and FAdV-4 infected chickens on the basis of higher serum anti 100K antibody titres. Our work provides preliminary structural and functional results to study biological role of the 100K protein and for further investigations to develop 100K inhibitors to control IBH-HPS in poultry.
Collapse
Affiliation(s)
- Majid Ali Shah
- Drug Discovery and Structural Biology Group, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan; Structural Biology Laboratory, Elettra-Sincrotrone Trieste S.C.p.A., Basovizza 34149, Trieste, Italy; Pakistan Institute of Engineering and Applied Sciences, P.O. Nilore, Islamabad, Pakistan
| | - Raheem Ullah
- Drug Discovery and Structural Biology Group, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan; Structural Biology Laboratory, Elettra-Sincrotrone Trieste S.C.p.A., Basovizza 34149, Trieste, Italy; Pakistan Institute of Engineering and Applied Sciences, P.O. Nilore, Islamabad, Pakistan
| | - Matteo De March
- Structural Biology Laboratory, Elettra-Sincrotrone Trieste S.C.p.A., Basovizza 34149, Trieste, Italy
| | - Muhammad Salahuddin Shah
- Drug Discovery and Structural Biology Group, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan; Vaccine Development Group, Animal Sciences Division, NIAB, Faisalabad, Pakistan
| | - Fouzia Ismat
- Drug Discovery and Structural Biology Group, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan; Pakistan Institute of Engineering and Applied Sciences, P.O. Nilore, Islamabad, Pakistan
| | - Mudasser Habib
- Vaccine Development Group, Animal Sciences Division, NIAB, Faisalabad, Pakistan
| | - Mazhar Iqbal
- Drug Discovery and Structural Biology Group, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan; Pakistan Institute of Engineering and Applied Sciences, P.O. Nilore, Islamabad, Pakistan
| | - Silvia Onesti
- Structural Biology Laboratory, Elettra-Sincrotrone Trieste S.C.p.A., Basovizza 34149, Trieste, Italy
| | - Moazur Rahman
- Drug Discovery and Structural Biology Group, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan; Pakistan Institute of Engineering and Applied Sciences, P.O. Nilore, Islamabad, Pakistan.
| |
Collapse
|
24
|
Litfin T, Zhou Y, Yang Y. SPOT-ligand 2: improving structure-based virtual screening by binding-homology search on an expanded structural template library. Bioinformatics 2017; 33:1238-1240. [PMID: 28057679 DOI: 10.1093/bioinformatics/btw829] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 12/27/2016] [Indexed: 11/12/2022] Open
Abstract
Motivation The high cost of drug discovery motivates the development of accurate virtual screening tools. Binding-homology, which takes advantage of known protein-ligand binding pairs, has emerged as a powerful discrimination technique. In order to exploit all available binding data, modelled structures of ligand-binding sequences may be used to create an expanded structural binding template library. Results SPOT-Ligand 2 has demonstrated significantly improved screening performance over its previous version by expanding the template library 15 times over the previous one. It also performed better than or similar to other binding-homology approaches on the DUD and DUD-E benchmarks. Availability and Implementation The server is available online at http://sparks-lab.org . Contacts yaoqi.zhou@griffith.edu.au or yuedong.yang@griffith.edu.au. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
| | - Yaoqi Zhou
- School of Information and Communication Technology.,Institute for Glycomics, Griffith University, Southport, Queensland 4215, Australia
| | - Yuedong Yang
- School of Information and Communication Technology.,Institute for Glycomics, Griffith University, Southport, Queensland 4215, Australia
| |
Collapse
|
25
|
Hamzeh-Mivehroud M, Sokouti B, Dastmalchi S. Molecular Docking at a Glance. Oncology 2017. [DOI: 10.4018/978-1-5225-0549-5.ch030] [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
The current chapter introduces different aspects of molecular docking technique in order to give an overview to the readers about the topics which will be dealt with throughout this volume. Like many other fields of science, molecular docking studies has experienced a lagging period of slow and steady increase in terms of acquiring attention of scientific community as well as its frequency of application, followed by a pronounced era of exponential expansion in theory, methodology, areas of application and performance due to developments in related technologies such as computational resources and theoretical as well as experimental biophysical methods. In the following sections the evolution of molecular docking will be reviewed and its different components including methods, search algorithms, scoring functions, validation of the methods, and area of applications plus few case studies will be touched briefly.
Collapse
Affiliation(s)
| | | | - Siavoush Dastmalchi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Iran & School of Pharmacy, Tabriz University of Medical Sciences, Iran
| |
Collapse
|
26
|
Snell TW, Johnston RK, Srinivasan B, Zhou H, Gao M, Skolnick J. Repurposing FDA-approved drugs for anti-aging therapies. Biogerontology 2016; 17:907-920. [PMID: 27484416 PMCID: PMC5065615 DOI: 10.1007/s10522-016-9660-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 07/25/2016] [Indexed: 12/31/2022]
Abstract
There is great interest in drugs that are capable of modulating multiple aging pathways, thereby delaying the onset and progression of aging. Effective strategies for drug development include the repurposing of existing drugs already approved by the FDA for human therapy. FDA approved drugs have known mechanisms of action and have been thoroughly screened for safety. Although there has been extensive scientific activity in repurposing drugs for disease therapy, there has been little testing of these drugs for their effects on aging. The pool of FDA approved drugs therefore represents a large reservoir of drug candidates with substantial potential for anti-aging therapy. In this paper we employ FINDSITEcomb, a powerful ligand homology modeling program, to identify binding partners for proteins produced by temperature sensing genes that have been implicated in aging. This list of drugs with potential to modulate aging rates was then tested experimentally for lifespan and healthspan extension using a small invertebrate model. Three protein targets of the rotifer Brachionus manjavacas corresponding to products of the transient receptor potential gene 7, ribosomal protein S6 polypeptide 2 gene, or forkhead box C gene, were screened against a compound library consisting of DrugBank drugs including 1347 FDA approved, non-nutraceutical molecules. Twenty nine drugs ranked in the top 1 % for binding to each target were subsequently included in our experimental analysis. Continuous exposure of rotifers to 1 µM naproxen significantly extended rotifer mean lifespan by 14 %. We used three endpoints to estimate rotifer health: swimming speed (mobility proxy), reproduction (overall vitality), and mitochondria activity (cellular senescence proxy). The natural decline in swimming speed with aging was more gradual when rotifers were exposed to three drugs, so that on day 6, mean swimming speed of females was 1.19 mm/s for naproxen (P = 0.038), 1.20 for fludarabine (P = 0.040), 1.35 for hydralazine (P = 0.038), as compared to 0.88 mm/s in the control. The average reproduction of control females in the second half of their reproductive lifespan was 1.08 per day. In contrast, females treated with 1 µM naproxen produced 1.4 offspring per day (P = 0.027) and females treated with 10 µM fludarabine or 1 µM hydralazine produced 1.72 (P = <0.001) and 1.66 (P = 0.001) offspring per day, respectively. Mitochondrial activity naturally declines with rotifer aging, but B. manjavacas treated with 1 µM hydralazine or 10 µM fludarabine retained 49 % (P = 0.038) and 89 % (P = 0.002) greater mitochondria activity, respectively, than untreated controls. Our results demonstrate that coupling computation to experimentation can quickly identify new drug candidates with anti-aging potential. Screening drugs for anti-aging effects using a rotifer bioassay is a powerful first step in identifying compounds worthy of follow-up in vertebrate models. Even if lifespan extension is not observed, certain drugs could improve healthspan, slowing age-dependent losses in mobility and vitality.
Collapse
Affiliation(s)
- Terry W Snell
- School of Biology, Georgia Institute of Technology, Atlanta, GA, 30332-0230, USA.
| | - Rachel K Johnston
- School of Biology, Georgia Institute of Technology, Atlanta, GA, 30332-0230, USA
| | - Bharath Srinivasan
- School of Biology, Georgia Institute of Technology, Atlanta, GA, 30332-0230, USA
| | - Hongyi Zhou
- School of Biology, Georgia Institute of Technology, Atlanta, GA, 30332-0230, USA
| | - Mu Gao
- School of Biology, Georgia Institute of Technology, Atlanta, GA, 30332-0230, USA
| | - Jeffrey Skolnick
- School of Biology, Georgia Institute of Technology, Atlanta, GA, 30332-0230, USA
| |
Collapse
|
27
|
Skolnick J, Zhou H. Why Is There a Glass Ceiling for Threading Based Protein Structure Prediction Methods? J Phys Chem B 2016; 121:3546-3554. [PMID: 27748116 DOI: 10.1021/acs.jpcb.6b09517] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Despite their different implementations, comparison of the best threading approaches to the prediction of evolutionary distant protein structures reveals that they tend to succeed or fail on the same protein targets. This is true despite the fact that the structural template library has good templates for all cases. Thus, a key question is why are certain protein structures threadable while others are not. Comparison with threading results on a set of artificial sequences selected for stability further argues that the failure of threading is due to the nature of the protein structures themselves. Using a new contact map based alignment algorithm, we demonstrate that certain folds are highly degenerate in that they can have very similar coarse grained fractions of native contacts aligned and yet differ significantly from the native structure. For threadable proteins, this is not the case. Thus, contemporary threading approaches appear to have reached a plateau, and new approaches to structure prediction are required.
Collapse
Affiliation(s)
- Jeffrey Skolnick
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology , 950 Atlantic Drive Northwest, Atlanta, Georgia 30318, United States
| | - Hongyi Zhou
- Center for the Study of Systems Biology, School of Biological Sciences, Georgia Institute of Technology , 950 Atlantic Drive Northwest, Atlanta, Georgia 30318, United States
| |
Collapse
|
28
|
Srinivasan B, Zhou H, Mitra S, Skolnick J. Novel small molecule binders of human N-glycanase 1, a key player in the endoplasmic reticulum associated degradation pathway. Bioorg Med Chem 2016; 24:4750-4758. [PMID: 27567076 DOI: 10.1016/j.bmc.2016.08.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/08/2016] [Accepted: 08/12/2016] [Indexed: 12/30/2022]
Abstract
Peptide:N-glycanase (NGLY1) is an enzyme responsible for cleaving oligosaccharide moieties from misfolded glycoproteins to enable their proper degradation. Deletion and truncation mutations in this gene are responsible for an inherited disorder of the endoplasmic reticulum-associated degradation pathway. However, the literature is unclear whether the disorder is a result of mutations leading to loss-of-function, loss of substrate specificity, loss of protein stability or a combination of these factors. In this communication, without burdening ourselves with the mechanistic underpinning of disease causation because of mutations on the NGLY1 protein, we demonstrate the successful application of virtual ligand screening (VLS) combined with experimental high-throughput validation to the discovery of novel small-molecules that show binding to the transglutaminase domain of NGLY1. Attempts at recombinant expression and purification of six different constructs led to successful expression of five, with three constructs purified to homogeneity. Most mutant variants failed to purify possibly because of misfolding and the resultant exposure of surface hydrophobicity that led to protein aggregation. For the purified constructs, our threading/structure-based VLS algorithm, FINDSITE(comb), was employed to predict ligands that may bind to the protein. Then, the predictions were assessed by high-throughput differential scanning fluorimetry. This led to the identification of nine different ligands that bind to the protein of interest and provide clues to the nature of pharmacophore that facilitates binding. This is the first study that has identified novel ligands that bind to the NGLY1 protein as a possible starting point in the discovery of ligands with potential therapeutic applications in the treatment of the disorder caused by NGLY1 mutants.
Collapse
Affiliation(s)
- Bharath Srinivasan
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, 950, Atlantic Drive, Atlanta, GA 30332, United States.
| | - Hongyi Zhou
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, 950, Atlantic Drive, Atlanta, GA 30332, United States
| | - Sreyoshi Mitra
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, 950, Atlantic Drive, Atlanta, GA 30332, United States
| | - Jeffrey Skolnick
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, 950, Atlantic Drive, Atlanta, GA 30332, United States.
| |
Collapse
|
29
|
The Aryl Hydrocarbon Receptor Relays Metabolic Signals to Promote Cellular Regeneration. Stem Cells Int 2016; 2016:4389802. [PMID: 27563312 PMCID: PMC4987465 DOI: 10.1155/2016/4389802] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 06/16/2016] [Accepted: 07/05/2016] [Indexed: 02/04/2023] Open
Abstract
While sensing the cell environment, the aryl hydrocarbon receptor (AHR) interacts with different pathways involved in cellular homeostasis. This review summarizes evidence suggesting that cellular regeneration in the context of aging and diseases can be modulated by AHR signaling on stem cells. New insights connect orphaned observations into AHR interactions with critical signaling pathways such as WNT to propose a role of this ligand-activated transcription factor in the modulation of cellular regeneration by altering pathways that nurture cellular expansion such as changes in the metabolic efficiency rather than by directly altering cell cycling, proliferation, or cell death. Targeting the AHR to promote regeneration might prove to be a useful strategy to avoid unbalanced disruptions of homeostasis that may promote disease and also provide biological rationale for potential regenerative medicine approaches.
Collapse
|
30
|
Tan Y, Hu Y, Liu X, Yin Z, Chen XW, Liu M. Improving drug safety: From adverse drug reaction knowledge discovery to clinical implementation. Methods 2016; 110:14-25. [PMID: 27485605 DOI: 10.1016/j.ymeth.2016.07.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 07/13/2016] [Accepted: 07/30/2016] [Indexed: 12/16/2022] Open
Abstract
Adverse drug reactions (ADRs) are a major public health concern, causing over 100,000 fatalities in the United States every year with an annual cost of $136 billion. Early detection and accurate prediction of ADRs is thus vital for drug development and patient safety. Multiple scientific disciplines, namely pharmacology, pharmacovigilance, and pharmacoinformatics, have been addressing the ADR problem from different perspectives. With the same goal of improving drug safety, this article summarizes and links the research efforts in the multiple disciplines into a single framework from comprehensive understanding of the interactions between drugs and biological system and the identification of genetic and phenotypic predispositions of patients susceptible to higher ADR risks and finally to the current state of implementation of medication-related decision support systems. We start by describing available computational resources for building drug-target interaction networks with biological annotations, which provides a fundamental knowledge for ADR prediction. Databases are classified by functions to help users in selection. Post-marketing surveillance is then introduced where data-driven approach can not only enhance the prediction accuracy of ADRs but also enables the discovery of genetic and phenotypic risk factors of ADRs. Understanding genetic risk factors for ADR requires well organized patient genetics information and analysis by pharmacogenomic approaches. Finally, current state of clinical decision support systems is presented and described how clinicians can be assisted with the integrated knowledgebase to minimize the risk of ADR. This review ends with a discussion of existing challenges in each of disciplines with potential solutions and future directions.
Collapse
Affiliation(s)
- Yuxiang Tan
- Big Data Decision Institute, The First Affiliated Hospital, International Immunology Center, The Biomedical Translational Research Institute, Jinan University, Guangzhou, Guangdong, China
| | - Yong Hu
- Big Data Decision Institute, The First Affiliated Hospital, International Immunology Center, The Biomedical Translational Research Institute, Jinan University, Guangzhou, Guangdong, China
| | - Xiaoxiao Liu
- Big Data Decision Institute, The First Affiliated Hospital, International Immunology Center, The Biomedical Translational Research Institute, Jinan University, Guangzhou, Guangdong, China
| | - Zhinan Yin
- Big Data Decision Institute, The First Affiliated Hospital, International Immunology Center, The Biomedical Translational Research Institute, Jinan University, Guangzhou, Guangdong, China
| | - Xue-Wen Chen
- Department of Computer Science, Wayne State University, Detroit, USA
| | - Mei Liu
- Department of Internal Medicine, Division of Medical Informatics, University of Kansas Medical Center, Kansas City, USA.
| |
Collapse
|
31
|
Dhakshinamoorthy S, Dinh NT, Skolnick J, Styczynski MP. Metabolomics identifies the intersection of phosphoethanolamine with menaquinone-triggered apoptosis in an in vitro model of leukemia. MOLECULAR BIOSYSTEMS 2016; 11:2406-16. [PMID: 26175011 DOI: 10.1039/c5mb00237k] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Altered metabolism is increasingly acknowledged as an important aspect of cancer, and thus serves as a potentially fertile area for the identification of therapeutic targets or leads. Our recent work using transcriptional data to predict metabolite levels in cancer cells led to preliminary evidence of the antiproliferative role of menaquinone (vitamin K2) in the Jurkat cell line model of acute lymphoblastic leukemia. However, nothing is known about the direct metabolic impacts of menaquinone in cancer, which could provide insights into its mechanism of action. Here, we used metabolomics to investigate the process by which menaquinone exerts antiproliferative activity on Jurkat cells. We first validated the dose-dependent, semi-selective, pro-apoptotic activity of menaquinone treatment on Jurkat cells relative to non-cancerous lymphoblasts. We then used mass spectrometry-based metabolomics to identify systems-scale changes in metabolic dynamics that are distinct from changes induced in non-cancerous cells or by other chemotherapeutics. One of the most significantly affected metabolites was phosphoethanolamine, which exhibited a two-fold increase in menaquinone-treated Jurkat cells compared to vehicle-treated cells at 24 h, growing to a five-fold increase at 72 h. Phosphoethanolamine elevation was observed prior to the induction of apoptosis, and was not observed in menaquinone-treated lymphoblasts or chemotherapeutic-treated Jurkat cells. We also validated the link between menaquinone and phosphoethanolamine in an ovarian cancer cell line, suggesting potentially broad applicability of their relationship. This metabolomics-based work is the first detailed characterization of the metabolic impacts of menaquinone treatment and the first identified link between phosphoethanolamine and menaquinone-induced apoptosis.
Collapse
|
32
|
Hodos RA, Kidd BA, Khader S, Readhead BP, Dudley JT. In silico methods for drug repurposing and pharmacology. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2016; 8:186-210. [PMID: 27080087 PMCID: PMC4845762 DOI: 10.1002/wsbm.1337] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/08/2016] [Accepted: 02/11/2016] [Indexed: 12/18/2022]
Abstract
Data in the biological, chemical, and clinical domains are accumulating at ever-increasing rates and have the potential to accelerate and inform drug development in new ways. Challenges and opportunities now lie in developing analytic tools to transform these often complex and heterogeneous data into testable hypotheses and actionable insights. This is the aim of computational pharmacology, which uses in silico techniques to better understand and predict how drugs affect biological systems, which can in turn improve clinical use, avoid unwanted side effects, and guide selection and development of better treatments. One exciting application of computational pharmacology is drug repurposing-finding new uses for existing drugs. Already yielding many promising candidates, this strategy has the potential to improve the efficiency of the drug development process and reach patient populations with previously unmet needs such as those with rare diseases. While current techniques in computational pharmacology and drug repurposing often focus on just a single data modality such as gene expression or drug-target interactions, we argue that methods such as matrix factorization that can integrate data within and across diverse data types have the potential to improve predictive performance and provide a fuller picture of a drug's pharmacological action. WIREs Syst Biol Med 2016, 8:186-210. doi: 10.1002/wsbm.1337 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Rachel A Hodos
- New York University and Icahn School of Medicine at Mt. Sinai, New York, NY
| | - Brian A Kidd
- Icahn School of Medicine at Mt. Sinai, New York, NY
| | | | | | | |
Collapse
|
33
|
Yang Y, Zhan J, Zhou Y. SPOT‐Ligand: Fast and effective structure‐based virtual screening by binding homology search according to ligand and receptor similarity. J Comput Chem 2016; 37:1734-9. [DOI: 10.1002/jcc.24380] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 01/12/2016] [Accepted: 03/05/2016] [Indexed: 12/11/2022]
Affiliation(s)
- Yuedong Yang
- Institute for Glycomics and School of Information and Communication TechnologyGriffith UniversityParklands DrSouthport QLD4222 Australia
| | - Jian Zhan
- Institute for Glycomics and School of Information and Communication TechnologyGriffith UniversityParklands DrSouthport QLD4222 Australia
| | - Yaoqi Zhou
- Institute for Glycomics and School of Information and Communication TechnologyGriffith UniversityParklands DrSouthport QLD4222 Australia
| |
Collapse
|
34
|
ENTPRISE: An Algorithm for Predicting Human Disease-Associated Amino Acid Substitutions from Sequence Entropy and Predicted Protein Structures. PLoS One 2016; 11:e0150965. [PMID: 26982818 PMCID: PMC4794227 DOI: 10.1371/journal.pone.0150965] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 02/21/2016] [Indexed: 01/02/2023] Open
Abstract
The advance of next-generation sequencing technologies has made exome sequencing rapid and relatively inexpensive. A major application of exome sequencing is the identification of genetic variations likely to cause Mendelian diseases. This requires processing large amounts of sequence information and therefore computational approaches that can accurately and efficiently identify the subset of disease-associated variations are needed. The accuracy and high false positive rates of existing computational tools leave much room for improvement. Here, we develop a boosted tree regression machine-learning approach to predict human disease-associated amino acid variations by utilizing a comprehensive combination of protein sequence and structure features. On comparing our method, ENTPRISE, to the state-of-the-art methods SIFT, PolyPhen-2, MUTATIONASSESSOR, MUTATIONTASTER, FATHMM, ENTPRISE exhibits significant improvement. In particular, on a testing dataset consisting of only proteins with balanced disease-associated and neutral variations defined as having the ratio of neutral/disease-associated variations between 0.3 and 3, the Mathews Correlation Coefficient by ENTPRISE is 0.493 as compared to 0.432 by PPH2-HumVar, 0.406 by SIFT, 0.403 by MUTATIONASSESSOR, 0.402 by PPH2-HumDiv, 0.305 by MUTATIONTASTER, and 0.181 by FATHMM. ENTPRISE is then applied to nucleic acid binding proteins in the human proteome. Disease-associated predictions are shown to be highly correlated with the number of protein-protein interactions. Both these predictions and the ENTPRISE server are freely available for academic users as a web service at http://cssb.biology.gatech.edu/entprise/.
Collapse
|
35
|
Abstract
Native proteins perform an amazing variety of biochemical functions, including enzymatic catalysis, and can engage in protein-protein and protein-DNA interactions that are essential for life. A key question is how special are these functional properties of proteins. Are they extremely rare, or are they an intrinsic feature? Comparison to the properties of compact conformations of artificially generated compact protein structures selected for thermodynamic stability but not any type of function, the artificial (ART) protein library, demonstrates that a remarkable number of the properties of native-like proteins are recapitulated. These include the complete set of small molecule ligand-binding pockets and most protein-protein interfaces. ART structures are predicted to be capable of weakly binding metabolites and cover a significant fraction of metabolic pathways, with the most enriched pathways including ancient ones such as glycolysis. Native-like active sites are also found in ART proteins. A small fraction of ART proteins are predicted to have strong protein-protein and protein-DNA interactions. Overall, it appears that biochemical function is an intrinsic feature of proteins which nature has significantly optimized during evolution. These studies raise questions as to the relative roles of specificity and promiscuity in the biochemical function and control of cells that need investigation.
Collapse
Affiliation(s)
- Jeffrey Skolnick
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Mu Gao
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Hongyi Zhou
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
36
|
Ismaya WT, Yunita, Damayanti S, Wijaya C, Tjandrawinata RR, Retnoningrum DS, Rachmawati H. In Silico Study to Develop a Lectin-Like Protein from Mushroom Agaricus bisporus for Pharmaceutical Application. Sci Pharm 2016; 84:203-17. [PMID: 27110510 PMCID: PMC4839548 DOI: 10.3797/scipharm.isp.2015.11] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Accepted: 12/15/2015] [Indexed: 01/19/2023] Open
Abstract
A lectin-like protein of unknown function designated as LSMT was recently discovered in the edible mushroom Agaricus bisporus. The protein shares high structural similarity to HA-33 from Clostridium botulinum (HA33) and Ricin-B-like lectin from the mushroom Clitocybe nebularis (CNL), which have been developed as drug carrier and anti-cancer, respectively. These homologous proteins display the ability to penetrate the intestinal epithelial cell monolayer, and are beneficial for oral administration. As the characteristics of LSMT are unknown, a structural study in silico was performed to assess its potential pharmaceutical application. The study suggested potential binding to target ligands such as HA-33 and CNL although the nature, specificity, capacity, mode, and strength may differ. Further molecular docking experiments suggest that interactions between the LSMT and tested ligands may take place. This finding indicates the possible use of the LSMT protein, initiating new research on its use for pharmaceutical purposes.
Collapse
Affiliation(s)
- Wangsa Tirta Ismaya
- Dexa Laboratories of Biomolecular Sciences, Industri Selatan V, Blok PP No. 7, Kawasan Industri, Jababeka II, Cikarang 17550, Indonesia
| | - Yunita
- Research group of Pharmaceutics, School of Pharmacy, Institut Teknologi Bandung, Jl. Ganesa 10, Bandung 40132, Indonesia
| | - Sophi Damayanti
- Research group of Pharmacochemistry, School of Pharmacy, Institut Teknologi Bandung, Jl. Ganesa 10, Bandung 40132, Indonesia
| | - Caroline Wijaya
- Dexa Laboratories of Biomolecular Sciences, Industri Selatan V, Blok PP No. 7, Kawasan Industri, Jababeka II, Cikarang 17550, Indonesia
| | - Raymond R Tjandrawinata
- Dexa Laboratories of Biomolecular Sciences, Industri Selatan V, Blok PP No. 7, Kawasan Industri, Jababeka II, Cikarang 17550, Indonesia
| | - Debbie Sofie Retnoningrum
- Research group of Biotechnology, School of Pharmacy, Institut Teknologi Bandung, Jl. Ganesa 10, Bandung 40132, Indonesia
| | - Heni Rachmawati
- Research group of Pharmaceutics, School of Pharmacy, Institut Teknologi Bandung, Jl. Ganesa 10, Bandung 40132, Indonesia
| |
Collapse
|
37
|
Abstract
Enzymes are one of the most important groups of drug targets, and identifying possible ligand-enzyme interactions is of major importance in many drug discovery processes. Novel computational methods have been developed that can apply the information from the increasing number of resolved and available ligand-enzyme complexes to model new unknown interactions and therefore contribute to answer open questions in the field of drug discovery like the identification of unknown protein functions, off-target binding, ligand 3D homology modeling and induced-fit simulations.
Collapse
|
38
|
Roy A, Srinivasan B, Skolnick J. PoLi: A Virtual Screening Pipeline Based on Template Pocket and Ligand Similarity. J Chem Inf Model 2015. [PMID: 26225536 DOI: 10.1021/acs.jcim.5b00232] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Often in pharmaceutical research the goal is to identify small molecules that can interact with and appropriately modify the biological behavior of a new protein target. Unfortunately, most proteins lack both known structures and small molecule binders, prerequisites of many virtual screening, VS, approaches. For such proteins, ligand homology modeling, LHM, that copies ligands from homologous and perhaps evolutionarily distant template proteins, has been shown to be a powerful VS approach to identify possible binding ligands. However, if we want to target a specific pocket for which there is no homologous holo template protein structure, then LHM will not work. To address this issue, in a new pocket-based approach, PoLi, we generalize LHM by exploiting the fact that the number of distinct small molecule ligand-binding pockets in proteins is small. PoLi identifies similar ligand-binding pockets in a holo template protein library, selectively copies relevant parts of template ligands, and uses them for VS. In practice, PoLi is a hybrid structure and ligand-based VS algorithm that integrates 2D fingerprint-based and 3D shape-based similarity metrics for improved virtual screening performance. On standard DUD and DUD-E benchmark databases, using modeled receptor structures, PoLi achieves an average enrichment factor of 13.4 and 9.6, respectively, in the top 1% of the screened library. In contrast, traditional docking-based VS using AutoDock Vina and homology-based VS using FINDSITE(filt) have an average enrichment of 1.6 (3.0) and 9.0 (7.9) on the DUD (DUD-E) sets, respectively. Experimental validation of PoLi predictions on dihydrofolate reductase, DHFR, using differential scanning fluorimetry, DSF, identifies multiple ligands with diverse molecular scaffolds, thus demonstrating the advantage of PoLi over current state-of-the-art VS methods.
Collapse
Affiliation(s)
- Ambrish Roy
- Center for the Study of Systems Biology, Georgia Institute of Technology , 250 14th Street NW, Atlanta, Georgia 30318, United States
| | - Bharath Srinivasan
- Center for the Study of Systems Biology, Georgia Institute of Technology , 250 14th Street NW, Atlanta, Georgia 30318, United States
| | - Jeffrey Skolnick
- Center for the Study of Systems Biology, Georgia Institute of Technology , 250 14th Street NW, Atlanta, Georgia 30318, United States
| |
Collapse
|
39
|
Comprehensive prediction of drug-protein interactions and side effects for the human proteome. Sci Rep 2015; 5:11090. [PMID: 26057345 PMCID: PMC4603786 DOI: 10.1038/srep11090] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 05/14/2015] [Indexed: 01/23/2023] Open
Abstract
Identifying unexpected drug-protein interactions is crucial for drug repurposing. We develop a comprehensive proteome scale approach that predicts human protein targets and side effects of drugs. For drug-protein interaction prediction, FINDSITE(comb), whose average precision is ~30% and recall ~27%, is employed. For side effect prediction, a new method is developed with a precision of ~57% and a recall of ~24%. Our predictions show that drugs are quite promiscuous, with the average (median) number of human targets per drug of 329 (38), while a given protein interacts with 57 drugs. The result implies that drug side effects are inevitable and existing drugs may be useful for repurposing, with only ~1,000 human proteins likely causing serious side effects. A killing index derived from serious side effects has a strong correlation with FDA approved drugs being withdrawn. Therefore, it provides a pre-filter for new drug development. The methodology is free to the academic community on the DR. PRODIS (DRugome, PROteome, and DISeasome) webserver at http://cssb.biology.gatech.edu/dr.prodis/. DR. PRODIS provides protein targets of drugs, drugs for a given protein target, associated diseases and side effects of drugs, as well as an interface for the virtual target screening of new compounds.
Collapse
|
40
|
Insights into Disease-Associated Mutations in the Human Proteome through Protein Structural Analysis. Structure 2015; 23:1362-9. [PMID: 26027735 DOI: 10.1016/j.str.2015.03.028] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 03/03/2015] [Accepted: 03/06/2015] [Indexed: 01/12/2023]
Abstract
Most known disease-associated mutations are missense mutations involving changes of amino acids of proteins encoded by their genes. Given the plethora of genetic studies, sequenced exomes, and new protein structures determined each year, it is appropriate to revisit the role that structure plays in providing insights into the molecular basis of disease-associated mutations. In that regard, a large-scale structural analysis of 6,025 disease-associated mutations as well as 4,536 neutral variations for comparison was performed. While buried amino acids are common among the disease-associated mutations, as reported previously, more are statistically significantly enriched at observed or predicted functional sites. Interesting findings are that ligand-binding sites adjacent to protein-protein interfaces and residues involved in enzymatic function are especially vulnerable to disease-associated mutations. Finally, a compositional analysis of disease-associated mutations in comparison with variants identified in the 1000 Genomes Project provides a structural rationalization of the most disease-associated residue types.
Collapse
|
41
|
Currin A, Swainston N, Day PJ, Kell DB. Synthetic biology for the directed evolution of protein biocatalysts: navigating sequence space intelligently. Chem Soc Rev 2015; 44:1172-239. [PMID: 25503938 PMCID: PMC4349129 DOI: 10.1039/c4cs00351a] [Citation(s) in RCA: 256] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Indexed: 12/21/2022]
Abstract
The amino acid sequence of a protein affects both its structure and its function. Thus, the ability to modify the sequence, and hence the structure and activity, of individual proteins in a systematic way, opens up many opportunities, both scientifically and (as we focus on here) for exploitation in biocatalysis. Modern methods of synthetic biology, whereby increasingly large sequences of DNA can be synthesised de novo, allow an unprecedented ability to engineer proteins with novel functions. However, the number of possible proteins is far too large to test individually, so we need means for navigating the 'search space' of possible protein sequences efficiently and reliably in order to find desirable activities and other properties. Enzymologists distinguish binding (Kd) and catalytic (kcat) steps. In a similar way, judicious strategies have blended design (for binding, specificity and active site modelling) with the more empirical methods of classical directed evolution (DE) for improving kcat (where natural evolution rarely seeks the highest values), especially with regard to residues distant from the active site and where the functional linkages underpinning enzyme dynamics are both unknown and hard to predict. Epistasis (where the 'best' amino acid at one site depends on that or those at others) is a notable feature of directed evolution. The aim of this review is to highlight some of the approaches that are being developed to allow us to use directed evolution to improve enzyme properties, often dramatically. We note that directed evolution differs in a number of ways from natural evolution, including in particular the available mechanisms and the likely selection pressures. Thus, we stress the opportunities afforded by techniques that enable one to map sequence to (structure and) activity in silico, as an effective means of modelling and exploring protein landscapes. Because known landscapes may be assessed and reasoned about as a whole, simultaneously, this offers opportunities for protein improvement not readily available to natural evolution on rapid timescales. Intelligent landscape navigation, informed by sequence-activity relationships and coupled to the emerging methods of synthetic biology, offers scope for the development of novel biocatalysts that are both highly active and robust.
Collapse
Affiliation(s)
- Andrew Currin
- Manchester Institute of Biotechnology , The University of Manchester , 131, Princess St , Manchester M1 7DN , UK . ; http://dbkgroup.org/; @dbkell ; Tel: +44 (0)161 306 4492
- School of Chemistry , The University of Manchester , Manchester M13 9PL , UK
- Centre for Synthetic Biology of Fine and Speciality Chemicals (SYNBIOCHEM) , The University of Manchester , 131, Princess St , Manchester M1 7DN , UK
| | - Neil Swainston
- Manchester Institute of Biotechnology , The University of Manchester , 131, Princess St , Manchester M1 7DN , UK . ; http://dbkgroup.org/; @dbkell ; Tel: +44 (0)161 306 4492
- Centre for Synthetic Biology of Fine and Speciality Chemicals (SYNBIOCHEM) , The University of Manchester , 131, Princess St , Manchester M1 7DN , UK
- School of Computer Science , The University of Manchester , Manchester M13 9PL , UK
| | - Philip J. Day
- Manchester Institute of Biotechnology , The University of Manchester , 131, Princess St , Manchester M1 7DN , UK . ; http://dbkgroup.org/; @dbkell ; Tel: +44 (0)161 306 4492
- Centre for Synthetic Biology of Fine and Speciality Chemicals (SYNBIOCHEM) , The University of Manchester , 131, Princess St , Manchester M1 7DN , UK
- Faculty of Medical and Human Sciences , The University of Manchester , Manchester M13 9PT , UK
| | - Douglas B. Kell
- Manchester Institute of Biotechnology , The University of Manchester , 131, Princess St , Manchester M1 7DN , UK . ; http://dbkgroup.org/; @dbkell ; Tel: +44 (0)161 306 4492
- School of Chemistry , The University of Manchester , Manchester M13 9PL , UK
- Centre for Synthetic Biology of Fine and Speciality Chemicals (SYNBIOCHEM) , The University of Manchester , 131, Princess St , Manchester M1 7DN , UK
| |
Collapse
|
42
|
Petrey D, Chen TS, Deng L, Garzon JI, Hwang H, Lasso G, Lee H, Silkov A, Honig B. Template-based prediction of protein function. Curr Opin Struct Biol 2015; 32:33-8. [PMID: 25678152 DOI: 10.1016/j.sbi.2015.01.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 01/13/2015] [Accepted: 01/19/2015] [Indexed: 12/11/2022]
Abstract
We discuss recent approaches for structure-based protein function annotation. We focus on template-based methods where the function of a query protein is deduced from that of a template for which both the structure and function are known. We describe the different ways of identifying a template. These are typically based on sequence analysis but new methods based on purely structural similarity are also being developed that allow function annotation based on structural relationships that cannot be recognized by sequence. The growing number of available structures of known function, improved homology modeling techniques and new developments in the use of structure allow template-based methods to be applied on a proteome-wide scale and in many different biological contexts. This progress significantly expands the range of applicability of structural information in function annotation to a level that previously was only achievable by sequence comparison.
Collapse
Affiliation(s)
- Donald Petrey
- Howard Hughes Medical Institute, Department of Biochemistry and Molecular Biophysics, Department of Systems Biology, Center for Computational Biology and Bioinformatics, 1130 St. Nicholas Avenue, Room 815, New York, NY 10032, United States.
| | - T Scott Chen
- Howard Hughes Medical Institute, Department of Biochemistry and Molecular Biophysics, Department of Systems Biology, Center for Computational Biology and Bioinformatics, 1130 St. Nicholas Avenue, Room 815, New York, NY 10032, United States
| | - Lei Deng
- Howard Hughes Medical Institute, Department of Biochemistry and Molecular Biophysics, Department of Systems Biology, Center for Computational Biology and Bioinformatics, 1130 St. Nicholas Avenue, Room 815, New York, NY 10032, United States
| | - Jose Ignacio Garzon
- Howard Hughes Medical Institute, Department of Biochemistry and Molecular Biophysics, Department of Systems Biology, Center for Computational Biology and Bioinformatics, 1130 St. Nicholas Avenue, Room 815, New York, NY 10032, United States
| | - Howook Hwang
- Howard Hughes Medical Institute, Department of Biochemistry and Molecular Biophysics, Department of Systems Biology, Center for Computational Biology and Bioinformatics, 1130 St. Nicholas Avenue, Room 815, New York, NY 10032, United States
| | - Gorka Lasso
- Howard Hughes Medical Institute, Department of Biochemistry and Molecular Biophysics, Department of Systems Biology, Center for Computational Biology and Bioinformatics, 1130 St. Nicholas Avenue, Room 815, New York, NY 10032, United States
| | - Hunjoong Lee
- Howard Hughes Medical Institute, Department of Biochemistry and Molecular Biophysics, Department of Systems Biology, Center for Computational Biology and Bioinformatics, 1130 St. Nicholas Avenue, Room 815, New York, NY 10032, United States
| | - Antonina Silkov
- Howard Hughes Medical Institute, Department of Biochemistry and Molecular Biophysics, Department of Systems Biology, Center for Computational Biology and Bioinformatics, 1130 St. Nicholas Avenue, Room 815, New York, NY 10032, United States
| | - Barry Honig
- Howard Hughes Medical Institute, Department of Biochemistry and Molecular Biophysics, Department of Systems Biology, Center for Computational Biology and Bioinformatics, 1130 St. Nicholas Avenue, Room 815, New York, NY 10032, United States
| |
Collapse
|
43
|
Skolnick J, Gao M, Roy A, Srinivasan B, Zhou H. Implications of the small number of distinct ligand binding pockets in proteins for drug discovery, evolution and biochemical function. Bioorg Med Chem Lett 2015; 25:1163-70. [PMID: 25690787 DOI: 10.1016/j.bmcl.2015.01.059] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 01/23/2015] [Accepted: 01/24/2015] [Indexed: 01/05/2023]
Abstract
Coincidence of the properties of ligand binding pockets in native proteins with those in proteins generated by computer simulations without selection for function shows that pockets are a generic protein feature and the number of distinct pockets is small. Similar pockets occur in unrelated protein structures, an observation successfully employed in pocket-based virtual ligand screening. The small number of pockets suggests that off-target interactions among diverse proteins are inherent; kinases, proteases and phosphatases show this prototypical behavior. The ability to repurpose FDA approved drugs is general, and minor side effects cannot be avoided. Finally, the implications to drug discovery are explored.
Collapse
Affiliation(s)
- Jeffrey Skolnick
- Center for the Study of Systems Biology, Georgia Institute of Technology, 250 14th St NW, Atlanta, GA 30318, USA.
| | - Mu Gao
- Center for the Study of Systems Biology, Georgia Institute of Technology, 250 14th St NW, Atlanta, GA 30318, USA
| | - Ambrish Roy
- Center for the Study of Systems Biology, Georgia Institute of Technology, 250 14th St NW, Atlanta, GA 30318, USA
| | - Bharath Srinivasan
- Center for the Study of Systems Biology, Georgia Institute of Technology, 250 14th St NW, Atlanta, GA 30318, USA
| | - Hongyi Zhou
- Center for the Study of Systems Biology, Georgia Institute of Technology, 250 14th St NW, Atlanta, GA 30318, USA
| |
Collapse
|
44
|
Chen YC. Beware of docking! Trends Pharmacol Sci 2015; 36:78-95. [DOI: 10.1016/j.tips.2014.12.001] [Citation(s) in RCA: 344] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 11/23/2014] [Accepted: 12/02/2014] [Indexed: 12/16/2022]
|
45
|
|
46
|
Grinter SZ, Zou X. Challenges, applications, and recent advances of protein-ligand docking in structure-based drug design. Molecules 2014; 19:10150-76. [PMID: 25019558 PMCID: PMC6270832 DOI: 10.3390/molecules190710150] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 06/13/2014] [Accepted: 07/02/2014] [Indexed: 11/16/2022] Open
Abstract
The docking methods used in structure-based virtual database screening offer the ability to quickly and cheaply estimate the affinity and binding mode of a ligand for the protein receptor of interest, such as a drug target. These methods can be used to enrich a database of compounds, so that more compounds that are subsequently experimentally tested are found to be pharmaceutically interesting. In addition, like all virtual screening methods used for drug design, structure-based virtual screening can focus on curated libraries of synthesizable compounds, helping to reduce the expense of subsequent experimental verification. In this review, we introduce the protein-ligand docking methods used for structure-based drug design and other biological applications. We discuss the fundamental challenges facing these methods and some of the current methodological topics of interest. We also discuss the main approaches for applying protein-ligand docking methods. We end with a discussion of the challenging aspects of evaluating or benchmarking the accuracy of docking methods for their improvement, and discuss future directions.
Collapse
Affiliation(s)
- Sam Z Grinter
- Informatics Institute, University of Missouri, Columbia, MO 65211, USA.
| | - Xiaoqin Zou
- Informatics Institute, University of Missouri, Columbia, MO 65211, USA.
| |
Collapse
|
47
|
Xia J, Jin H, Liu Z, Zhang L, Wang XS. An unbiased method to build benchmarking sets for ligand-based virtual screening and its application to GPCRs. J Chem Inf Model 2014; 54:1433-50. [PMID: 24749745 PMCID: PMC4038372 DOI: 10.1021/ci500062f] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
![]()
Benchmarking data
sets have become common in recent years for the
purpose of virtual screening, though the main focus had been placed
on the structure-based virtual screening (SBVS) approaches. Due to
the lack of crystal structures, there is great need for unbiased benchmarking
sets to evaluate various ligand-based virtual screening (LBVS) methods
for important drug targets such as G protein-coupled receptors (GPCRs).
To date these ready-to-apply data sets for LBVS are fairly limited,
and the direct usage of benchmarking sets designed for SBVS could
bring the biases to the evaluation of LBVS. Herein, we propose an
unbiased method to build benchmarking sets for LBVS and validate it
on a multitude of GPCRs targets. To be more specific, our methods
can (1) ensure chemical diversity of ligands, (2) maintain the physicochemical
similarity between ligands and decoys, (3) make the decoys dissimilar
in chemical topology to all ligands to avoid false negatives, and
(4) maximize spatial random distribution of ligands and decoys. We
evaluated the quality of our Unbiased Ligand Set (ULS) and Unbiased
Decoy Set (UDS) using three common LBVS approaches, with Leave-One-Out
(LOO) Cross-Validation (CV) and a metric of average AUC of the ROC
curves. Our method has greatly reduced the “artificial enrichment”
and “analogue bias” of a published GPCRs benchmarking
set, i.e., GPCR Ligand Library (GLL)/GPCR Decoy Database (GDD). In
addition, we addressed an important issue about the ratio of decoys
per ligand and found that for a range of 30 to 100 it does not affect
the quality of the benchmarking set, so we kept the original ratio
of 39 from the GLL/GDD.
Collapse
Affiliation(s)
- Jie Xia
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University , Beijing 100191, China
| | | | | | | | | |
Collapse
|
48
|
Srinivasan B, Zhou H, Kubanek J, Skolnick J. Experimental validation of FINDSITE(comb) virtual ligand screening results for eight proteins yields novel nanomolar and micromolar binders. J Cheminform 2014; 6:16. [PMID: 24936211 PMCID: PMC4038399 DOI: 10.1186/1758-2946-6-16] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 04/15/2014] [Indexed: 01/09/2023] Open
Abstract
Background Identification of ligand-protein binding interactions is a critical step in drug discovery. Experimental screening of large chemical libraries, in spite of their specific role and importance in drug discovery, suffer from the disadvantages of being random, time-consuming and expensive. To accelerate the process, traditional structure- or ligand-based VLS approaches are combined with experimental high-throughput screening, HTS. Often a single protein or, at most, a protein family is considered. Large scale VLS benchmarking across diverse protein families is rarely done, and the reported success rate is very low. Here, we demonstrate the experimental HTS validation of a novel VLS approach, FINDSITEcomb, across a diverse set of medically-relevant proteins. Results For eight different proteins belonging to different fold-classes and from diverse organisms, the top 1% of FINDSITEcomb’s VLS predictions were tested, and depending on the protein target, 4%-47% of the predicted ligands were shown to bind with μM or better affinities. In total, 47 small molecule binders were identified. Low nanomolar (nM) binders for dihydrofolate reductase and protein tyrosine phosphatases (PTPs) and micromolar binders for the other proteins were identified. Six novel molecules had cytotoxic activity (<10 μg/ml) against the HCT-116 colon carcinoma cell line and one novel molecule had potent antibacterial activity. Conclusions We show that FINDSITEcomb is a promising new VLS approach that can assist drug discovery.
Collapse
Affiliation(s)
- Bharath Srinivasan
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, 250, 14th Street, N.W., Atlanta, GA 30318, USA
| | - Hongyi Zhou
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, 250, 14th Street, N.W., Atlanta, GA 30318, USA
| | - Julia Kubanek
- School of Biology, Atlanta, GA 30332, USA ; School of Chemistry and Biochemistry, Aquatic Chemical Ecology Center, Institute of Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Jeffrey Skolnick
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, 250, 14th Street, N.W., Atlanta, GA 30318, USA
| |
Collapse
|
49
|
On the use of knowledge-based potentials for the evaluation of models of protein-protein, protein-DNA, and protein-RNA interactions. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2014; 94:77-120. [PMID: 24629186 DOI: 10.1016/b978-0-12-800168-4.00004-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Proteins are the bricks and mortar of cells, playing structural and functional roles. In order to perform their function, they interact with each other as well as with other biomolecules such as DNA or RNA. Therefore, to fathom the function of a protein, we require knowing its partners and the atomic details of its interactions (i.e., the structure of the complex). However, the amount of protein interactions with an experimentally determined three-dimensional structure is scarce. Therefore, computational techniques such as homology modeling are foremost to fill this gap. Protein interactions can be modeled using as templates the interactions of homologous proteins, if the structure of the complex is known, or using docking methods. In both approaches, the estimation of the quality of models is essential. There are several ways to address this problem. In this review, we focus on the use of knowledge-based potentials for the analysis of protein interactions. We describe the procedure to derive statistical potentials and split them into different energetic terms that can be used for different purposes. We extensively discuss the fields where knowledge-based potentials have been successfully applied to (1) model protein-protein, protein-DNA, and protein-RNA interactions and (2) predict binding sites (in the protein and in the DNA). Moreover, we provide ready-to-use resources for docking and benchmarking protein interactions.
Collapse
|
50
|
Khar KR, Goldschmidt L, Karanicolas J. Fast docking on graphics processing units via Ray-Casting. PLoS One 2013; 8:e70661. [PMID: 23976948 PMCID: PMC3745428 DOI: 10.1371/journal.pone.0070661] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 06/20/2013] [Indexed: 11/22/2022] Open
Abstract
Docking Approach using Ray Casting (DARC) is structure-based computational method for carrying out virtual screening by docking small-molecules into protein surface pockets. In a complementary study we find that DARC can be used to identify known inhibitors from large sets of decoy compounds, and can identify new compounds that are active in biochemical assays. Here, we describe our adaptation of DARC for use on Graphics Processing Units (GPUs), leading to a speedup of approximately 27-fold in typical-use cases over the corresponding calculations carried out using a CPU alone. This dramatic speedup of DARC will enable screening larger compound libraries, screening with more conformations of each compound, and including multiple receptor conformations when screening. We anticipate that all three of these enhanced approaches, which now become tractable, will lead to improved screening results.
Collapse
Affiliation(s)
- Karen R. Khar
- Center for Bioinformatics, University of Kansas, Lawrence, Kansas, United States of America
| | - Lukasz Goldschmidt
- UCLA-DOE Institute for Genomics and Proteomics, University of California, Los Angeles, California, United States of America
| | - John Karanicolas
- Center for Bioinformatics, University of Kansas, Lawrence, Kansas, United States of America
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, United States of America
- * E-mail:
| |
Collapse
|