1
|
Liu J, Zhu J, Hao H, Bi J, Hou H, Zhang G. Transcriptomic and Molecular Docking Analysis Reveal Virulence Gene Regulation-Mediated Antibacterial Effects of Benzyl Isothiocyanate Against Staphylococcus aureus. Appl Biochem Biotechnol 2024; 196:8239-8253. [PMID: 38709426 DOI: 10.1007/s12010-024-04938-y] [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] [Accepted: 04/16/2024] [Indexed: 05/07/2024]
Abstract
Staphylococcus aureus (S. aureus) is a common pathogen that can cause many serious infections. Thus, efficient and practical techniques to fight S. aureus are required. In this study, transcriptomics was used to evaluate changes in S. aureus following treatment with benzyl isothiocyanate (BITC) to determine its antibacterial action. The results revealed that the BITC at subinhibitory concentrations (1/8th MIC) treated group had 94 differentially expressed genes compared to the control group, with 52 downregulated genes. Moreover, STRING analyses were used to reveal the protein interactions encoded by 36 genes. Then, we verified three significant virulence genes by qRT-PCR, including capsular polysaccharide synthesis enzyme (cp8F), capsular polysaccharide biosynthesis protein (cp5D), and thermonuclease (nuc). Furthermore, molecular docking analysis was performed to investigate the action site of BITC with the encoded proteins of cp8F, cp5D, and nuc. The results showed that the docking fraction of BITC with selected proteins ranged from - 6.00 to - 6.60 kcal/mol, predicting the stability of these complexes. BITC forms hydrophobic, hydrogen-bonded, π-π conjugated interactions with amino acids TRP (130), GLY (10), ILE (406), LYS (368), TYR (192), and ARG (114) of these proteins. These findings will aid future research into the antibacterial effects of BITC against S. aureus.
Collapse
Affiliation(s)
- Jianan Liu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, 116034, China
| | - Junya Zhu
- Jinkui Food Science and Technology (Dalian) Co., Ltd, Dalian, 116000, China
| | - Hongshun Hao
- Department of Inorganic Nonmetallic Materials Engineering, Dalian Polytechnic University, Dalian, 116034, China
| | - Jingran Bi
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, 116034, China
| | - Hongman Hou
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, 116034, China
| | - Gongliang Zhang
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, 116034, China.
| |
Collapse
|
2
|
Rossetti G, Mandelli D. How exascale computing can shape drug design: A perspective from multiscale QM/MM molecular dynamics simulations and machine learning-aided enhanced sampling algorithms. Curr Opin Struct Biol 2024; 86:102814. [PMID: 38631106 DOI: 10.1016/j.sbi.2024.102814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 03/11/2024] [Accepted: 03/25/2024] [Indexed: 04/19/2024]
Abstract
Molecular simulations are an essential asset in the first steps of drug design campaigns. However, the requirement of high-throughput limits applications mainly to qualitative approaches with low computational cost, but also low accuracy. Unlocking the potential of more rigorous quantum mechanical/molecular mechanics (QM/MM) models combined with molecular dynamics-based free energy techniques could have a tremendous impact. Indeed, these two relatively old techniques are emerging as promising methods in the field. This has been favored by the exponential growth of computer power and the proliferation of powerful data-driven methods. Here, we briefly review recent advances and applications, and give our perspective on the impact that QM/MM and free-energy methods combined with machine learning-aided algorithms can have on drug design.
Collapse
Affiliation(s)
- Giulia Rossetti
- Computational Biomedicine, Institute of Advanced Simulations IAS-5/Institute for Neuroscience and Medicine INM-9, Forschungszentrum Jülich GmbH, Jülich 52428, Germany; Department of Neurology, University Hospital Aachen (UKA), RWTH Aachen University, Aachen, Germany; Jülich Supercomputing Centre (JSC), Forschungszentrum Jülich GmbH, Jülich 52428, Germany. https://twitter.com/G_Rossetti_
| | - Davide Mandelli
- Computational Biomedicine, Institute of Advanced Simulations IAS-5/Institute for Neuroscience and Medicine INM-9, Forschungszentrum Jülich GmbH, Jülich 52428, Germany.
| |
Collapse
|
3
|
Tu G, Fu T, Zheng G, Xu B, Gou R, Luo D, Wang P, Xue W. Computational Chemistry in Structure-Based Solute Carrier Transporter Drug Design: Recent Advances and Future Perspectives. J Chem Inf Model 2024; 64:1433-1455. [PMID: 38294194 DOI: 10.1021/acs.jcim.3c01736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Solute carrier transporters (SLCs) are a class of important transmembrane proteins that are involved in the transportation of diverse solute ions and small molecules into cells. There are approximately 450 SLCs within the human body, and more than a quarter of them are emerging as attractive therapeutic targets for multiple complex diseases, e.g., depression, cancer, and diabetes. However, only 44 unique transporters (∼9.8% of the SLC superfamily) with 3D structures and specific binding sites have been reported. To design innovative and effective drugs targeting diverse SLCs, there are a number of obstacles that need to be overcome. However, computational chemistry, including physics-based molecular modeling and machine learning- and deep learning-based artificial intelligence (AI), provides an alternative and complementary way to the classical drug discovery approach. Here, we present a comprehensive overview on recent advances and existing challenges of the computational techniques in structure-based drug design of SLCs from three main aspects: (i) characterizing multiple conformations of the proteins during the functional process of transportation, (ii) identifying druggability sites especially the cryptic allosteric ones on the transporters for substrates and drugs binding, and (iii) discovering diverse small molecules or synthetic protein binders targeting the binding sites. This work is expected to provide guidelines for a deep understanding of the structure and function of the SLC superfamily to facilitate rational design of novel modulators of the transporters with the aid of state-of-the-art computational chemistry technologies including artificial intelligence.
Collapse
Affiliation(s)
- Gao Tu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Tingting Fu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | | | - Binbin Xu
- Chengdu Sintanovo Biotechnology Co., Ltd., Chengdu 610200, China
| | - Rongpei Gou
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Ding Luo
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Panpan Wang
- College of Chemistry and Pharmaceutical Engineering, Huanghuai University, Zhumadian 463000, China
| | - Weiwei Xue
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| |
Collapse
|
4
|
Cheng C, Beroza P. Shape-Aware Synthon Search (SASS) for Virtual Screening of Synthon-Based Chemical Spaces. J Chem Inf Model 2024; 64:1251-1260. [PMID: 38335044 DOI: 10.1021/acs.jcim.3c01865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
Virtual screening of large-scale chemical libraries has become increasingly useful for identifying high-quality candidates for drug discovery. While it is possible to exhaustively screen chemical spaces that number on the order of billions, indirect combinatorial approaches are needed to efficiently navigate larger, synthon-based virtual spaces. We describe Shape-Aware Synthon Search (SASS), a synthon-based virtual screening method that carries out shape similarity searches in the synthon space instead of the enumerated product space. SASS can replicate results from exhaustive searches in ultralarge, combinatorial spaces with high recall on a variety of query molecules while only scoring a small subspace of possible enumerated products, thereby significantly accelerating large-scale, shape-based virtual screening.
Collapse
Affiliation(s)
- Chen Cheng
- Discovery Chemistry, Genentech, South San Francisco, California 94080, United States
| | - Paul Beroza
- Discovery Chemistry, Genentech, South San Francisco, California 94080, United States
| |
Collapse
|
5
|
Das S, Merz KM. Molecular Gas-Phase Conformational Ensembles. J Chem Inf Model 2024; 64:749-760. [PMID: 38206321 DOI: 10.1021/acs.jcim.3c01309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Accurately determining the global minima of a molecular structure is important in diverse scientific fields, including drug design, materials science, and chemical synthesis. Conformational search engines serve as valuable tools for exploring the extensive conformational space of molecules and for identifying energetically favorable conformations. In this study, we present a comparison of Auto3D, CREST, Balloon, and ETKDG (from RDKit), which are freely available conformational search engines, to evaluate their effectiveness in locating global minima. These engines employ distinct methodologies, including machine learning (ML) potential-based, semiempirical, and force field-based approaches. To validate these methods, we propose the use of collisional cross-section (CCS) values obtained from ion mobility-mass spectrometry studies. We hypothesize that experimental gas-phase CCS values can provide experimental evidence that we likely have the global minimum for a given molecule. To facilitate this effort, we used our gas-phase conformation library (GPCL) which currently consists of the full ensembles of 20 small molecules and can be used by the community to validate any conformational search engine. Further members of the GPCL can be readily created for any molecule of interest using our standard workflow used to compute CCS values, expanding the ability of the GPCL in validation exercises. These innovative validation techniques enhance our understanding of the conformational landscape and provide valuable insights into the performance of conformational generation engines. Our findings shed light on the strengths and limitations of each search engine, enabling informed decisions for their utilization in various scientific fields, where accurate molecular structure determination is crucial for understanding biological activity and designing targeted interventions. By facilitating the identification of reliable conformations, this study significantly contributes to enhancing the efficiency and accuracy of molecular structure determination, with particular focus on metabolite structure elucidation. The findings of this research also provide valuable insights for developing effective workflows for predicting the structures of unknown compounds with high precision.
Collapse
Affiliation(s)
- Susanta Das
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, Michigan 48824, United States
| | - Kenneth M Merz
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, Michigan 48824, United States
| |
Collapse
|
6
|
Popov KI, Wellnitz J, Maxfield T, Tropsha A. HIt Discovery using docking ENriched by GEnerative Modeling (HIDDEN GEM): A novel computational workflow for accelerated virtual screening of ultra-large chemical libraries. Mol Inform 2024; 43:e202300207. [PMID: 37802967 PMCID: PMC11156482 DOI: 10.1002/minf.202300207] [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: 08/16/2023] [Revised: 10/03/2023] [Accepted: 10/06/2023] [Indexed: 10/08/2023]
Abstract
Recent rapid expansion of make-on-demand, purchasable, chemical libraries comprising dozens of billions or even trillions of molecules has challenged the efficient application of traditional structure-based virtual screening methods that rely on molecular docking. We present a novel computational methodology termed HIDDEN GEM (HIt Discovery using Docking ENriched by GEnerative Modeling) that greatly accelerates virtual screening. This workflow uniquely integrates machine learning, generative chemistry, massive chemical similarity searching and molecular docking of small, selected libraries in the beginning and the end of the workflow. For each target, HIDDEN GEM nominates a small number of top-scoring virtual hits prioritized from ultra-large chemical libraries. We have benchmarked HIDDEN GEM by conducting virtual screening campaigns for 16 diverse protein targets using Enamine REAL Space library comprising 37 billion molecules. We show that HIDDEN GEM yields the highest enrichment factors as compared to state of the art accelerated virtual screening methods, while requiring the least computational resources. HIDDEN GEM can be executed with any docking software and employed by users with limited computational resources.
Collapse
Affiliation(s)
- Konstantin I. Popov
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
- These authors contributed equally: Konstantin I. Popov, James Wellnitz, Travis Maxfield
| | - James Wellnitz
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
- These authors contributed equally: Konstantin I. Popov, James Wellnitz, Travis Maxfield
| | - Travis Maxfield
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
- These authors contributed equally: Konstantin I. Popov, James Wellnitz, Travis Maxfield
| | - Alexander Tropsha
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
7
|
Wang R, Feng H, Wei GW. ChatGPT in Drug Discovery: A Case Study on Anticocaine Addiction Drug Development with Chatbots. J Chem Inf Model 2023; 63:7189-7209. [PMID: 37956228 PMCID: PMC11021135 DOI: 10.1021/acs.jcim.3c01429] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The birth of ChatGPT, a cutting-edge language model-based chatbot developed by OpenAI, ushered in a new era in AI. However, due to potential pitfalls, its role in rigorous scientific research is not clear yet. This paper vividly showcases its innovative application within the field of drug discovery. Focused specifically on developing anticocaine addiction drugs, the study employs GPT-4 as a virtual guide, offering strategic and methodological insights to researchers working on generative models for drug candidates. The primary objective is to generate optimal drug-like molecules with desired properties. By leveraging the capabilities of ChatGPT, the study introduces a novel approach to the drug discovery process. This symbiotic partnership between AI and researchers transforms how drug development is approached. Chatbots become facilitators, steering researchers toward innovative methodologies and productive paths for creating effective drug candidates. This research sheds light on the collaborative synergy between human expertise and AI assistance, wherein ChatGPT's cognitive abilities enhance the design and development of pharmaceutical solutions. This paper not only explores the integration of advanced AI in drug discovery but also reimagines the landscape by advocating for AI-powered chatbots as trailblazers in revolutionizing therapeutic innovation.
Collapse
Affiliation(s)
- Rui Wang
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Hongsong Feng
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Guo-Wei Wei
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
8
|
Yu L, He X, Fang X, Liu L, Liu J. Deep Learning with Geometry-Enhanced Molecular Representation for Augmentation of Large-Scale Docking-Based Virtual Screening. J Chem Inf Model 2023; 63:6501-6514. [PMID: 37882338 DOI: 10.1021/acs.jcim.3c01371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023]
Abstract
Structure-based virtual screening has been a crucial tool in drug discovery for decades. However, as the chemical space expands, the existing structure-based virtual screening techniques based on molecular docking and scoring struggle to handle billion-entry ultralarge libraries due to the high computational cost. To address this challenge, people have resorted to machine learning techniques to enhance structure-based virtual screening for efficiently exploring the vast chemical space. In those cases, compounds are usually treated as sequential strings or two-dimensional topology graphs, limiting their ability to incorporate three-dimensional structural information for downstream tasks. We herein propose a novel deep learning protocol, GEM-Screen, which utilizes the geometry-enhanced molecular representation of the compounds docking to a specific target and is trained on docking scores of a small fraction of a library through an active learning strategy to approximate the docking outcome for yet nontraining entries. This protocol is applied to virtual screening campaigns against the AmpC and D4 targets, demonstrating that GEM-Screen enriches more than 90% of the hit scaffolds for AmpC in the top 4% of model predictions and more than 80% of the hit scaffolds for D4 in the same top-ranking size of library. GEM-Screen can be used in conjunction with traditional docking programs for docking of only the top-ranked compounds to avoid the exhaustive docking of the whole library, thus allowing for discovering top-scoring compounds from billion-entry libraries in a rapid yet accurate fashion.
Collapse
Affiliation(s)
- Lan Yu
- School of Science, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao He
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Shanghai Frontiers Science Center of Molecule Intelligent Syntheses, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
- New York University-East China Normal University Center for Computational Chemistry, New York University Shanghai, Shanghai 200062, China
| | - Xiaomin Fang
- Baidu International Technology (Shenzhen) Co., Ltd., Shenzhen 518063, China
| | - Lihang Liu
- Baidu International Technology (Shenzhen) Co., Ltd., Shenzhen 518063, China
| | - Jinfeng Liu
- School of Science, China Pharmaceutical University, Nanjing 210009, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
9
|
Wang R, Feng H, Wei GW. ChatGPT in Drug Discovery: A Case Study on Anti-Cocaine Addiction Drug Development with Chatbots. ARXIV 2023:arXiv:2308.06920v2. [PMID: 37645039 PMCID: PMC10462169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
The birth of ChatGPT, a cutting-edge language model-based chatbot developed by OpenAI, ushered in a new era in AI. However, due to potential pitfalls, its role in rigorous scientific research is not clear yet. This paper vividly showcases its innovative application within the field of drug discovery. Focused specifically on developing anti-cocaine addiction drugs, the study employs GPT-4 as a virtual guide, offering strategic and methodological insights to researchers working on generative models for drug candidates. The primary objective is to generate optimal drug-like molecules with desired properties. By leveraging the capabilities of ChatGPT, the study introduces a novel approach to the drug discovery process. This symbiotic partnership between AI and researchers transforms how drug development is approached. Chatbots become facilitators, steering researchers towards innovative methodologies and productive paths for creating effective drug candidates. This research sheds light on the collaborative synergy between human expertise and AI assistance, wherein ChatGPT's cognitive abilities enhance the design and development of potential pharmaceutical solutions. This paper not only explores the integration of advanced AI in drug discovery but also reimagines the landscape by advocating for AI-powered chatbots as trailblazers in revolutionizing therapeutic innovation.
Collapse
Affiliation(s)
- Rui Wang
- Department of Mathematics,Michigan State University, MI 48824, USA
| | - Hongsong Feng
- Department of Mathematics,Michigan State University, MI 48824, USA
| | - Guo-Wei Wei
- Department of Mathematics,Michigan State University, MI 48824, USA
- Department of Electrical and Computer Engineering, Michigan State University, MI 48824, USA
- Department of Biochemistry and Molecular Biology, Michigan State University, MI 48824, USA
| |
Collapse
|
10
|
Han R, Yoon H, Kim G, Lee H, Lee Y. Revolutionizing Medicinal Chemistry: The Application of Artificial Intelligence (AI) in Early Drug Discovery. Pharmaceuticals (Basel) 2023; 16:1259. [PMID: 37765069 PMCID: PMC10537003 DOI: 10.3390/ph16091259] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/24/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Artificial intelligence (AI) has permeated various sectors, including the pharmaceutical industry and research, where it has been utilized to efficiently identify new chemical entities with desirable properties. The application of AI algorithms to drug discovery presents both remarkable opportunities and challenges. This review article focuses on the transformative role of AI in medicinal chemistry. We delve into the applications of machine learning and deep learning techniques in drug screening and design, discussing their potential to expedite the early drug discovery process. In particular, we provide a comprehensive overview of the use of AI algorithms in predicting protein structures, drug-target interactions, and molecular properties such as drug toxicity. While AI has accelerated the drug discovery process, data quality issues and technological constraints remain challenges. Nonetheless, new relationships and methods have been unveiled, demonstrating AI's expanding potential in predicting and understanding drug interactions and properties. For its full potential to be realized, interdisciplinary collaboration is essential. This review underscores AI's growing influence on the future trajectory of medicinal chemistry and stresses the importance of ongoing synergies between computational and domain experts.
Collapse
Affiliation(s)
| | | | | | | | - Yoonji Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
11
|
Wang Z, Zheng L, Wang S, Lin M, Wang Z, Kong AWK, Mu Y, Wei Y, Li W. A fully differentiable ligand pose optimization framework guided by deep learning and a traditional scoring function. Brief Bioinform 2023; 24:6887112. [PMID: 36502369 DOI: 10.1093/bib/bbac520] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/17/2022] [Accepted: 10/31/2022] [Indexed: 12/14/2022] Open
Abstract
The recently reported machine learning- or deep learning-based scoring functions (SFs) have shown exciting performance in predicting protein-ligand binding affinities with fruitful application prospects. However, the differentiation between highly similar ligand conformations, including the native binding pose (the global energy minimum state), remains challenging that could greatly enhance the docking. In this work, we propose a fully differentiable, end-to-end framework for ligand pose optimization based on a hybrid SF called DeepRMSD+Vina combined with a multi-layer perceptron (DeepRMSD) and the traditional AutoDock Vina SF. The DeepRMSD+Vina, which combines (1) the root mean square deviation (RMSD) of the docking pose with respect to the native pose and (2) the AutoDock Vina score, is fully differentiable; thus is capable of optimizing the ligand binding pose to the energy-lowest conformation. Evaluated by the CASF-2016 docking power dataset, the DeepRMSD+Vina reaches a success rate of 94.4%, which outperforms most reported SFs to date. We evaluated the ligand conformation optimization framework in practical molecular docking scenarios (redocking and cross-docking tasks), revealing the high potentialities of this framework in drug design and discovery. Structural analysis shows that this framework has the ability to identify key physical interactions in protein-ligand binding, such as hydrogen-bonding. Our work provides a paradigm for optimizing ligand conformations based on deep learning algorithms. The DeepRMSD+Vina model and the optimization framework are available at GitHub repository https://github.com/zchwang/DeepRMSD-Vina_Optimization.
Collapse
Affiliation(s)
- Zechen Wang
- School of Physics, Shandong University, Jinan, Shandong 250100, China
| | - Liangzhen Zheng
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China.,Shanghai Zelixir Biotech Company Ltd., Shanghai 200030, China
| | - Sheng Wang
- Shanghai Zelixir Biotech Company Ltd., Shanghai 200030, China
| | - Mingzhi Lin
- Shanghai Zelixir Biotech Company Ltd., Shanghai 200030, China
| | - Zhihao Wang
- School of Physics, Shandong University, Jinan, Shandong 250100, China
| | - Adams Wai-Kin Kong
- Rolls-Royce Corporate Lab, Nanyang Technological University, Singapore 637551, Singapore
| | - Yuguang Mu
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Yanjie Wei
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Weifeng Li
- School of Physics, Shandong University, Jinan, Shandong 250100, China
| |
Collapse
|
12
|
Liu Z, Zubatiuk T, Roitberg A, Isayev O. Auto3D: Automatic Generation of the Low-Energy 3D Structures with ANI Neural Network Potentials. J Chem Inf Model 2022; 62:5373-5382. [PMID: 36112860 DOI: 10.1021/acs.jcim.2c00817] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Computational programs accelerate the chemical discovery processes but often need proper three-dimensional molecular information as part of the input. Getting optimal molecular structures is challenging because it requires enumerating and optimizing a huge space of stereoisomers and conformers. We developed the Python-based Auto3D package for generating the low-energy 3D structures using SMILES as the input. Auto3D is based on state-of-the-art algorithms and can automatize the isomer enumeration and duplicate filtering process, 3D building process, geometry optimization, and ranking process. Tested on 50 molecules with multiple unspecified stereocenters, Auto3D is guaranteed to find the stereoconfiguration that yields the lowest-energy conformer. With Auto3D, we provide an extension of the ANI model. The new model, dubbed ANI-2xt, is trained on a tautomer-rich data set. ANI-2xt is benchmarked with DFT methods on geometry optimization and electronic and Gibbs free energy calculations. Compared with ANI-2x, ANI-2xt provides a 42% error reduction for tautomeric reaction energy calculations when using the gold-standard coupled-cluster calculation as the reference. ANI-2xt can accurately predict the energies and is several orders of magnitude faster than DFT methods.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Chemistry, Mellon College of Science, Carnegie Mellon University, Pittsburgh, Pennsylvania15213, United States
| | - Tetiana Zubatiuk
- Department of Chemistry, Mellon College of Science, Carnegie Mellon University, Pittsburgh, Pennsylvania15213, United States
| | - Adrian Roitberg
- Department of Chemistry, University of Florida, Gainesville, Florida32611, United States
| | - Olexandr Isayev
- Department of Chemistry, Mellon College of Science, Carnegie Mellon University, Pittsburgh, Pennsylvania15213, United States
| |
Collapse
|
13
|
Mehta S, Goel M, Priyakumar UD. MO-MEMES: A method for accelerating virtual screening using multi-objective Bayesian optimization. Front Med (Lausanne) 2022; 9:916481. [PMID: 36213671 PMCID: PMC9537730 DOI: 10.3389/fmed.2022.916481] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
The pursuit of potential inhibitors for novel targets has become a very important problem especially over the last 2 years with the world in the midst of the COVID-19 pandemic. This entails performing high throughput screening exercises on drug libraries to identify potential “hits”. These hits are identified using analysis of their physical properties like binding affinity to the target receptor, octanol-water partition coefficient (LogP) and more. However, drug libraries can be extremely large and it is infeasible to calculate and analyze the physical properties for each of those molecules within acceptable time and moreover, each molecule must possess a multitude of properties apart from just the binding affinity. To address this problem, in this study, we propose an extension to the Machine learning framework for Enhanced MolEcular Screening (MEMES) framework for multi-objective Bayesian optimization. This approach is capable of identifying over 90% of the most desirable molecules with respect to all required properties while explicitly calculating the values of each of those properties on only 6% of the entire drug library. This framework would provide an immense boost in identifying potential hits that possess all properties required for a drug molecules.
Collapse
|
14
|
Identification of Potential Cytochrome P450 3A5 Inhibitors: An Extensive Virtual Screening through Molecular Docking, Negative Image-Based Screening, Machine Learning and Molecular Dynamics Simulation Studies. Int J Mol Sci 2022; 23:ijms23169374. [PMID: 36012627 PMCID: PMC9409045 DOI: 10.3390/ijms23169374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
Cytochrome P450 3A5 (CYP3A5) is one of the crucial CYP family members and has already proven to be an important drug target for cardiovascular diseases. In the current study, the PubChem database was screened through molecular docking and high-affinity molecules were adopted for further assessment. A negative image-based (NIB) model was used for a similarity search by considering the complementary shape and electrostatics of the target and small molecules. Further, the molecules were segregated into active and inactive groups through six machine learning (ML) matrices. The active molecules found in each ML model were used for in silico pharmacokinetics and toxicity assessments. A total of five molecules followed the acceptable pharmacokinetics and toxicity profiles. Several potential binding interactions between the proposed molecules and CYP3A5 were observed. The dynamic behavior of the selected molecules in the CYP3A5 was explored through a molecular dynamics (MD) simulation study. Several parameters obtained from the MD simulation trajectory explained the stability of the protein–ligand complexes in dynamic states. The high binding affinity of each molecule was revealed by the binding free energy calculation through the MM-GBSA methods. Therefore, it can be concluded that the proposed molecules might be potential CYP3A5 molecules for therapeutic application in cardiovascular diseases subjected to in vitro/in vivo validations.
Collapse
|
15
|
Gorgulla C, Jayaraj A, Fackeldey K, Arthanari H. Emerging frontiers in virtual drug discovery: From quantum mechanical methods to deep learning approaches. Curr Opin Chem Biol 2022; 69:102156. [PMID: 35576813 PMCID: PMC9990419 DOI: 10.1016/j.cbpa.2022.102156] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 03/16/2022] [Accepted: 04/07/2022] [Indexed: 11/19/2022]
Abstract
Virtual screening-based approaches to discover initial hit and lead compounds have the potential to reduce both the cost and time of early drug discovery stages, as well as to find inhibitors for even challenging target sites such as protein-protein interfaces. Here in this review, we provide an overview of the progress that has been made in virtual screening methodology and technology on multiple fronts in recent years. The advent of ultra-large virtual screens, in which hundreds of millions to billions of compounds are screened, has proven to be a powerful approach to discover highly potent hit compounds. However, these developments are just the tip of the iceberg, with new technologies and methods emerging to propel the field forward. Examples include novel machine-learning approaches, which can reduce the computational costs of virtual screening dramatically, while progress in quantum-mechanical approaches can increase the accuracy of predictions of various small molecule properties.
Collapse
Affiliation(s)
- Christoph Gorgulla
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA; Department of Physics, Faculty of Arts and Sciences, Harvard University, Cambridge, MA, USA; Department of Cancer Biology, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | | | - Konstantin Fackeldey
- Institute of Mathematics, Technical University Berlin, Berlin, Germany; Zuse Institute Berlin, Berlin, Germany
| | - Haribabu Arthanari
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA; Department of Cancer Biology, Dana-Farber Cancer Institute (DFCI), Boston, MA, USA.
| |
Collapse
|
16
|
Liu J, Zhou Z, Kong S, Ma Z. Application of random forest based on semi-automatic parameter adjustment for optimization of anti-breast cancer drugs. Front Oncol 2022; 12:956705. [PMID: 35936743 PMCID: PMC9353770 DOI: 10.3389/fonc.2022.956705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/28/2022] [Indexed: 11/19/2022] Open
Abstract
The optimization of drug properties in the process of cancer drug development is very important to save research and development time and cost. In order to make the anti-breast cancer drug candidates with good biological activity, this paper collected 1974 compounds, firstly, the top 20 molecular descriptors that have the most influence on biological activity were screened by using XGBoost-based data feature selection; secondly, on this basis, take pIC50 values as feature data and use a variety of machine learning algorithms to compare, soas to select a most suitable algorithm to predict the IC50 and pIC50 values. It is preliminarily found that the effects of Random Forest, XGBoost and Gradient-enhanced algorithms are good and have little difference, and the Support vector machine is the worst. Then, using the Semi-automatic parameter adjustment method to adjust the parameters of Random Forest, XGBoost and Gradient-enhanced algorithms to find the optimal parameters. It is found that the Random Forest algorithm has high accuracy and excellent anti over fitting, and the algorithm is stable. Its prediction accuracy is 0.745. Finally, the accuracy of the results is verified by training the model with the preliminarily selected data, which provides an innovative solution for the optimization of the properties of anti- breast cancer drugs, and can provide better support for the early research and development of anti-breast cancer drugs.
Collapse
Affiliation(s)
- Jiajia Liu
- College of Science, North China University of Science and Technology, Tangshan, China
- Hebei Engineering Research Center for the Intelligentization of Iron Ore Optimization and Ironmaking Raw Materials Preparation Processes, North China University of Science and Technology, Tangshan, China
- The Key Laboratory of Engineering Computing in Tangshan City, North China University of Science and Technology, Tangshan, China
| | - Zhihui Zhou
- College of Science, North China University of Science and Technology, Tangshan, China
- The Key Laboratory of Engineering Computing in Tangshan City, North China University of Science and Technology, Tangshan, China
- Hebei Key Laboratory of Data Science and Application, North China University of Science and Technology, Tangshan, China
| | - Shanshan Kong
- College of Science, North China University of Science and Technology, Tangshan, China
- Hebei Engineering Research Center for the Intelligentization of Iron Ore Optimization and Ironmaking Raw Materials Preparation Processes, North China University of Science and Technology, Tangshan, China
- The Key Laboratory of Engineering Computing in Tangshan City, North China University of Science and Technology, Tangshan, China
- Hebei Key Laboratory of Data Science and Application, North China University of Science and Technology, Tangshan, China
- Tangshan Intelligent Industry and Image Processing Technology Innovation Center, North China University of Science and Technology, Tangshan, China
- *Correspondence: Shanshan Kong,
| | - Zezhong Ma
- College of Science, North China University of Science and Technology, Tangshan, China
- Hebei Engineering Research Center for the Intelligentization of Iron Ore Optimization and Ironmaking Raw Materials Preparation Processes, North China University of Science and Technology, Tangshan, China
- Tangshan Intelligent Industry and Image Processing Technology Innovation Center, North China University of Science and Technology, Tangshan, China
| |
Collapse
|
17
|
Spataro V, Buetti-Dinh A. POH1/Rpn11/PSMD14: a journey from basic research in fission yeast to a prognostic marker and a druggable target in cancer cells. Br J Cancer 2022; 127:788-799. [PMID: 35501388 PMCID: PMC9428165 DOI: 10.1038/s41416-022-01829-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/06/2022] [Accepted: 04/11/2022] [Indexed: 12/11/2022] Open
Abstract
POH1/Rpn11/PSMD14 is a highly conserved protein in eukaryotes from unicellular organisms to human and has a crucial role in cellular homoeostasis. It is a subunit of the regulatory particle of the proteasome, where it acts as an intrinsic deubiquitinase removing polyubiquitin chains from substrate proteins. This function is not only coupled to the translocation of substrates into the core of the proteasome and their subsequent degradation but also, in some instances, to the stabilisation of ubiquitinated proteins through their deubiquitination. POH1 was initially discovered as a functional homologue of the fission yeast gene pad1+, which confers drug resistance when overexpressed. In translational studies, expression of POH1 has been found to be increased in several tumour types relative to normal adjacent tissue and to correlate with tumour progression, higher tumour grade, decreased sensitivity to cytotoxic drugs and poor prognosis. Proteasome inhibitors targeting the core particle of the proteasome are highly active in the treatment of myeloma, and recently developed POH1 inhibitors, such as capzimin and thiolutin, have shown promising anticancer activity in cell lines of solid tumours and leukaemia. Here we give an overview of POH1 function in the cell, of its potential role in oncogenesis and of recent progress in developing POH1-targeting drugs.
Collapse
Affiliation(s)
- Vito Spataro
- Service of Medical Oncology, Oncology Institute of Southern Switzerland (IOSI), Ospedale San Giovanni, Via Gallino, 6500, Bellinzona, Switzerland.
| | - Antoine Buetti-Dinh
- Institute of Microbiology, Department of Environmental Constructions and Design, University of Applied Sciences and Arts of Southern Switzerland (SUPSI), via Mirasole 22a, 6500, Bellinzona, Switzerland.,Swiss Institute of Bioinformatics, Quartier Sorge, Batiment Genopode, 1015, Lausanne, Switzerland
| |
Collapse
|
18
|
Zheng L, Meng J, Jiang K, Lan H, Wang Z, Lin M, Li W, Guo H, Wei Y, Mu Y. Improving protein-ligand docking and screening accuracies by incorporating a scoring function correction term. Brief Bioinform 2022; 23:6548372. [PMID: 35289359 PMCID: PMC9116214 DOI: 10.1093/bib/bbac051] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/30/2022] [Accepted: 01/31/2022] [Indexed: 12/13/2022] Open
Abstract
Scoring functions are important components in molecular docking for structure-based drug discovery. Traditional scoring functions, generally empirical- or force field-based, are robust and have proven to be useful for identifying hits and lead optimizations. Although multiple highly accurate deep learning- or machine learning-based scoring functions have been developed, their direct applications for docking and screening are limited. We describe a novel strategy to develop a reliable protein–ligand scoring function by augmenting the traditional scoring function Vina score using a correction term (OnionNet-SFCT). The correction term is developed based on an AdaBoost random forest model, utilizing multiple layers of contacts formed between protein residues and ligand atoms. In addition to the Vina score, the model considerably enhances the AutoDock Vina prediction abilities for docking and screening tasks based on different benchmarks (such as cross-docking dataset, CASF-2016, DUD-E and DUD-AD). Furthermore, our model could be combined with multiple docking applications to increase pose selection accuracies and screening abilities, indicating its wide usage for structure-based drug discoveries. Furthermore, in a reverse practice, the combined scoring strategy successfully identified multiple known receptors of a plant hormone. To summarize, the results show that the combination of data-driven model (OnionNet-SFCT) and empirical scoring function (Vina score) is a good scoring strategy that could be useful for structure-based drug discoveries and potentially target fishing in future.
Collapse
Affiliation(s)
- Liangzhen Zheng
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China.,Shanghai Zelixir Biotech Company Ltd., Shanghai 200030, China
| | - Jintao Meng
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China.,National Supercomputer Center in Shenzhen, Shenzhen, 518000, China
| | - Kai Jiang
- Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology (SUSTech), Shenzhen, Guangdong 518055, China
| | - Haidong Lan
- Tencent AI Lab, Shenzhen, Guangdong 518000, China
| | - Zechen Wang
- School of Physics, Shandong University, Jinan, Shandong 250101, China
| | - Mingzhi Lin
- Shanghai Zelixir Biotech Company Ltd., Shanghai 200030, China
| | - Weifeng Li
- School of Physics, Shandong University, Jinan, Shandong 250101, China
| | - Hongwei Guo
- Institute of Plant and Food Science, Department of Biology, School of Life Sciences, Southern University of Science and Technology (SUSTech), Shenzhen, Guangdong 518055, China
| | - Yanjie Wei
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Yuguang Mu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive 637551, Singapore
| |
Collapse
|
19
|
Basciu A, Callea L, Motta S, Bonvin AM, Bonati L, Vargiu AV. No dance, no partner! A tale of receptor flexibility in docking and virtual screening. VIRTUAL SCREENING AND DRUG DOCKING 2022. [DOI: 10.1016/bs.armc.2022.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
20
|
Using filters in virtual screening: A comprehensive guide to minimize errors and maximize efficiency. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2022. [DOI: 10.1016/bs.armc.2022.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|