51
|
Kadsanit N, Worsawat P, Sakonsinsiri C, McElroy CR, Macquarrie D, Noppawan P, Hunt AJ. Sustainable methods for the carboxymethylation and methylation of ursolic acid with dimethyl carbonate under mild and acidic conditions. RSC Adv 2024; 14:16921-16934. [PMID: 38799212 PMCID: PMC11124730 DOI: 10.1039/d4ra02122c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024] Open
Abstract
Ursolic acid is a triterpene plant extract that exhibits significant potential as an anti-cancer, anti-tumour, and anti-inflammatory agent. Its direct use in the pharmaceutical industry is hampered by poor uptake of ursolic acid in the human body coupled with rapid metabolism causing a decrease in bioactivity. Modification of ursolic acid can overcome such issues, however, use of toxic reagents, unsustainable synthetic routes and poor reaction metrics have limited its potential. Herein, we demonstrate the first reported carboxymethylation and/or methylation of ursolic acid with dimethyl carbonate (DMC) as a green solvent and sustainable reagent under acidic conditions. The reaction of DMC with ursolic acid, in the presence of PTSA, ZnCl2, or H2SO4-SiO2 yielded the carboxymethylation product 3β-[[methoxy]carbonyl]oxyurs-12-en-28-oic acid, the methylation product 3β-methoxyurs-12-en-28-oic acid and the dehydration product urs-2,12-dien-28-oic acid. PTSA demonstrated high conversion and selectivity towards the previously unreported carboxymethylation of ursolic acid, while the application of formic acid in the system led to formylation of ursolic acid (3β-formylurs-12-en-28-oic acid) in quantitative yields via esterification, with DMC acting solely as a solvent. Meanwhile, the methylation product of ursolic acid, 3β-methoxyurs-12-en-28-oic acid, was successfully synthesised with FeCl3, demonstrating exceptional conversion and selectivity, >99% and 99%, respectively. Confirmed with the use of qualitative and quantitative green metrics, this result represents a significant improvement in conversion, selectivity, safety, and sustainability over previously reported methods of ursolic acid modification. It was demonstrated that these methods could be applied to other triterpenoids, including corosolic acid. The study also explored the potential pharmaceutical applications of ursolic acid, corosolic acid, and their derivatives, particularly in anti-inflammatory, anti-cancer, and anti-tumour treatments, using molecular ADMET and docking methods. The methods developed in this work have led to the synthesis of novel molecules, thus creating opportunities for the future investigation of biological activity and the modification of a wide range of triterpenoids applying acidic DMC systems to deliver novel active pharmaceutical intermediates.
Collapse
Affiliation(s)
- Nuttapong Kadsanit
- Materials Chemistry Research Center (MCRC), Department of Chemistry and Centre of Excellence for Innovation in Chemistry, Faculty of Science, Khon Kaen University Khon Kaen 40002 Thailand
| | - Pattamabhorn Worsawat
- Materials Chemistry Research Center (MCRC), Department of Chemistry and Centre of Excellence for Innovation in Chemistry, Faculty of Science, Khon Kaen University Khon Kaen 40002 Thailand
| | - Chadamas Sakonsinsiri
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University Khon Kaen 40002 Thailand
| | - Con R McElroy
- School of Chemistry, University of Lincoln Brayford Pool Campus Lincoln LN6 7TS UK
- Green Chemistry Centre of Excellence, Department of Chemistry, University of York Heslington York YO10 5DD UK
| | - Duncan Macquarrie
- Green Chemistry Centre of Excellence, Department of Chemistry, University of York Heslington York YO10 5DD UK
| | - Pakin Noppawan
- Department of Chemistry, Faculty of Science, Mahasarakham University Maha Sarakham 44150 Thailand
| | - Andrew J Hunt
- Materials Chemistry Research Center (MCRC), Department of Chemistry and Centre of Excellence for Innovation in Chemistry, Faculty of Science, Khon Kaen University Khon Kaen 40002 Thailand
| |
Collapse
|
52
|
Yoon JH, Lee D, Lee C, Cho E, Lee S, Cazenave-Gassiot A, Kim K, Chae S, Dennis EA, Suh PG. Paradigm shift required for translational research on the brain. Exp Mol Med 2024; 56:1043-1054. [PMID: 38689090 PMCID: PMC11148129 DOI: 10.1038/s12276-024-01218-x] [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: 10/13/2023] [Revised: 02/07/2024] [Accepted: 02/20/2024] [Indexed: 05/02/2024] Open
Abstract
Biomedical research on the brain has led to many discoveries and developments, such as understanding human consciousness and the mind and overcoming brain diseases. However, historical biomedical research on the brain has unique characteristics that differ from those of conventional biomedical research. For example, there are different scientific interpretations due to the high complexity of the brain and insufficient intercommunication between researchers of different disciplines owing to the limited conceptual and technical overlap of distinct backgrounds. Therefore, the development of biomedical research on the brain has been slower than that in other areas. Brain biomedical research has recently undergone a paradigm shift, and conducting patient-centered, large-scale brain biomedical research has become possible using emerging high-throughput analysis tools. Neuroimaging, multiomics, and artificial intelligence technology are the main drivers of this new approach, foreshadowing dramatic advances in translational research. In addition, emerging interdisciplinary cooperative studies provide insights into how unresolved questions in biomedicine can be addressed. This review presents the in-depth aspects of conventional biomedical research and discusses the future of biomedical research on the brain.
Collapse
Affiliation(s)
- Jong Hyuk Yoon
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea.
| | - Dongha Lee
- Cognitive Science Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Chany Lee
- Cognitive Science Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Eunji Cho
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Seulah Lee
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Amaury Cazenave-Gassiot
- Department of Biochemistry and Precision Medicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
| | - Kipom Kim
- Research Strategy Office, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Sehyun Chae
- Neurovascular Unit Research Group, Korean Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Edward A Dennis
- Department of Pharmacology and Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, 92093-0601, USA
| | - Pann-Ghill Suh
- Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| |
Collapse
|
53
|
Duo L, Chen Y, Liu Q, Ma Z, Farjudian A, Ho WY, Low SS, Ren J, Hirst JD, Xie H, Tang B. Discovery of novel SOS1 inhibitors using machine learning. RSC Med Chem 2024; 15:1392-1403. [PMID: 38665844 PMCID: PMC11042245 DOI: 10.1039/d4md00063c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/14/2024] [Indexed: 04/28/2024] Open
Abstract
Overactivation of the rat sarcoma virus (RAS) signaling is responsible for 30% of all human malignancies. Son of sevenless 1 (SOS1), a crucial node in the RAS signaling pathway, could modulate RAS activation, offering a promising therapeutic strategy for RAS-driven cancers. Applying machine learning (ML)-based virtual screening (VS) on small-molecule databases, we selected a random forest (RF) regressor for its robustness and performance. Screening was performed with the L-series and EGFR-related datasets, and was extended to the Chinese National Compound Library (CNCL) with more than 1.4 million compounds. In addition to a series of documented SOS1-related molecules, we uncovered nine compounds that have an unexplored chemical framework and displayed inhibitory activity, with the most potent achieving more than 50% inhibition rate in the KRAS G12C/SOS1 PPI assay and an IC50 value in the proximity of 20 μg mL-1. Compared with the manner that known inhibitory agents bind to the target, hit compounds represented by CL01545365 occupy a unique pocket in molecular docking. An in silico drug-likeness assessment suggested that the compound has moderately favorable drug-like properties and pharmacokinetic characteristics. Altogether, our findings strongly support that, characterized by the distinctive binding modes, the recognition of novel skeletons from the carboxylic acid series could be candidates for developing promising SOS1 inhibitors.
Collapse
Affiliation(s)
- Lihui Duo
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, Key Laboratory for Carbonaceous Waste Processing and Process Intensification Research of Zhejiang Province, Department of Chemical and Environmental Engineering, The University of Nottingham Ningbo China 199 Taikang East Road Ningbo 315100 P. R. China
| | - Yi Chen
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences 555 Zuchongzhi Road 201203 Shanghai China
- University of Chinese Academy of Sciences No.19A Yuquan Road Beijing 100049 China
| | - Qiupei Liu
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, Key Laboratory for Carbonaceous Waste Processing and Process Intensification Research of Zhejiang Province, Department of Chemical and Environmental Engineering, The University of Nottingham Ningbo China 199 Taikang East Road Ningbo 315100 P. R. China
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences 555 Zuchongzhi Road 201203 Shanghai China
| | - Zhangyi Ma
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, Key Laboratory for Carbonaceous Waste Processing and Process Intensification Research of Zhejiang Province, Department of Chemical and Environmental Engineering, The University of Nottingham Ningbo China 199 Taikang East Road Ningbo 315100 P. R. China
| | - Amin Farjudian
- School of Mathematics, Watson Building, University of Birmingham Edgbaston Birmingham B15 2TT UK
| | - Wan Yong Ho
- Faculty of Medicine and Health Sciences, University of Nottingham (Malaysia Campus) Semenyih 43500 Malaysia
| | - Sze Shin Low
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, Key Laboratory for Carbonaceous Waste Processing and Process Intensification Research of Zhejiang Province, Department of Chemical and Environmental Engineering, The University of Nottingham Ningbo China 199 Taikang East Road Ningbo 315100 P. R. China
| | - Jianfeng Ren
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, Key Laboratory for Carbonaceous Waste Processing and Process Intensification Research of Zhejiang Province, Department of Chemical and Environmental Engineering, The University of Nottingham Ningbo China 199 Taikang East Road Ningbo 315100 P. R. China
| | - Jonathan D Hirst
- School of Chemistry, University of Nottingham University Park Nottingham NG7 2RD UK
| | - Hua Xie
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences 555 Zuchongzhi Road 201203 Shanghai China
- University of Chinese Academy of Sciences No.19A Yuquan Road Beijing 100049 China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Zhongshan Tsuihang New District Zhongshan 528400 China
| | - Bencan Tang
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, Key Laboratory for Carbonaceous Waste Processing and Process Intensification Research of Zhejiang Province, Department of Chemical and Environmental Engineering, The University of Nottingham Ningbo China 199 Taikang East Road Ningbo 315100 P. R. China
| |
Collapse
|
54
|
Gu Z, Yan Y, Liu H, Wu D, Yao H, Lin K, Li X. Discovery of Covalent Lead Compounds Targeting 3CL Protease with a Lateral Interactions Spiking Neural Network. J Chem Inf Model 2024; 64:3047-3058. [PMID: 38520328 DOI: 10.1021/acs.jcim.3c01900] [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: 03/25/2024]
Abstract
Covalent drugs exhibit advantages in that noncovalent drugs cannot match, and covalent docking is an important method for screening covalent lead compounds. However, it is difficult for covalent docking to screen covalent compounds on a large scale because covalent docking requires determination of the covalent reaction type of the compound. Here, we propose to use deep learning of a lateral interactions spiking neural network to construct a covalent lead compound screening model to quickly screen covalent lead compounds. We used the 3CL protease (3CL Pro) of SARS-CoV-2 as the screen target and constructed two classification models based on LISNN to predict the covalent binding and inhibitory activity of compounds. The two classification models were trained on the covalent complex data set targeting cysteine (Cys) and the compound inhibitory activity data set targeting 3CL Pro, respected, with good prediction accuracy (ACC > 0.9). We then screened the screening compound library with 6 covalent binding screening models and 12 inhibitory activity screening models. We tested the inhibitory activity of the 32 compounds, and the best compound inhibited SARS-CoV-2 3CL Pro with an IC50 value of 369.5 nM. Further assay implied that dithiothreitol can affect the inhibitory activity of the compound to 3CL Pro, indicating that the compound may covalently bind 3CL Pro. The selectivity test showed that the compound had good target selectivity to 3CL Pro over cathepsin L. These correlation assays can prove the rationality of the covalent lead compound screening model. Finally, covalent docking was performed to demonstrate the binding conformation of the compound with 3CL Pro. The source code can be obtained from the GitHub repository (https://github.com/guzh970630/Screen_Covalent_Compound_by_LISNN).
Collapse
Affiliation(s)
- Zhihao Gu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yong Yan
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Hanwen Liu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Di Wu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Hequan Yao
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Kejiang Lin
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xuanyi Li
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
55
|
Corbaux P, Bayle A, Besle S, Vinceneux A, Vanacker H, Ouali K, Hanvic B, Baldini C, Cassier PA, Terret C, Verlingue L. Patients' selection and trial matching in early-phase oncology clinical trials. Crit Rev Oncol Hematol 2024; 196:104307. [PMID: 38401694 DOI: 10.1016/j.critrevonc.2024.104307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Early-phase clinical trials (EPCT) represent an important part of innovations in medical oncology and a valuable therapeutic option for patients with metastatic cancers, particularly in the era of precision medicine. Nevertheless, adult patients' participation in oncology clinical trials is low, ranging from 2% to 8% worldwide, with unequal access, and up to 40% risk of early discontinuation in EPCT, mostly due to cancer-related complications. DESIGN We review the tools and initiatives to increase patients' orientation and access to early phase cancer clinical trials, and to limit early discontinuation. RESULTS New approaches to optimize the early-phase clinical trial referring process in oncology include automatic trial matching, tools to facilitate the estimation of patients' prognostic and/or to better predict patients' eligibility to clinical trials. Classical and innovative approaches should be associated to double patient recruitment, improve clinical trial enrollment experience and reduce early discontinuation rates. CONCLUSIONS Whereas EPCT are essential for patients to access the latest medical innovations in oncology, offering the appropriate trial when it is relevant for patients should increase by organizational and technological innovations. The oncologic community will need to closely monitor their performance, portability and simplicity for implementation in daily clinical practice.
Collapse
Affiliation(s)
- P Corbaux
- Medical Oncology Department, Centre Léon Bérard, Lyon, France; Medical Oncology, Institut de Cancérologie et d'Hématologie Universitaire de Saint-Étienne (ICHUSE), Centre Hospitalier Universitaire de Saint-Etienne, France
| | - A Bayle
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif F-94805, France
| | - S Besle
- Centre de Recherche en Cancérologie de Lyon (CRCL), France
| | - A Vinceneux
- Medical Oncology Department, Centre Léon Bérard, Lyon, France
| | - H Vanacker
- Medical Oncology Department, Centre Léon Bérard, Lyon, France; Centre de Recherche en Cancérologie de Lyon (CRCL), France
| | - K Ouali
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif F-94805, France
| | - B Hanvic
- Medical Oncology Department, Centre Léon Bérard, Lyon, France
| | - C Baldini
- Drug Development Department (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif F-94805, France
| | - P A Cassier
- Medical Oncology Department, Centre Léon Bérard, Lyon, France; Centre de Recherche en Cancérologie de Lyon (CRCL), France
| | - C Terret
- Medical Oncology Department, Centre Léon Bérard, Lyon, France
| | - L Verlingue
- Medical Oncology Department, Centre Léon Bérard, Lyon, France; Centre de Recherche en Cancérologie de Lyon (CRCL), France.
| |
Collapse
|
56
|
Li X, Yang Q, Xu L, Dong W, Luo G, Wang W, Dong S, Wang K, Xuan P, Zhang X, Gao X. DrugMGR: a deep bioactive molecule binding method to identify compounds targeting proteins. Bioinformatics 2024; 40:btae176. [PMID: 38561176 PMCID: PMC11015954 DOI: 10.1093/bioinformatics/btae176] [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: 08/19/2023] [Revised: 03/04/2024] [Accepted: 03/28/2024] [Indexed: 04/04/2024] Open
Abstract
MOTIVATION Understanding the intermolecular interactions of ligand-target pairs is key to guiding the optimization of drug research on cancers, which can greatly mitigate overburden workloads for wet labs. Several improved computational methods have been introduced and exhibit promising performance for these identification tasks, but some pitfalls restrict their practical applications: (i) first, existing methods do not sufficiently consider how multigranular molecule representations influence interaction patterns between proteins and compounds; and (ii) second, existing methods seldom explicitly model the binding sites when an interaction occurs to enable better prediction and interpretation, which may lead to unexpected obstacles to biological researchers. RESULTS To address these issues, we here present DrugMGR, a deep multigranular drug representation model capable of predicting binding affinities and regions for each ligand-target pair. We conduct consistent experiments on three benchmark datasets using existing methods and introduce a new specific dataset to better validate the prediction of binding sites. For practical application, target-specific compound identification tasks are also carried out to validate the capability of real-world compound screen. Moreover, the visualization of some practical interaction scenarios provides interpretable insights from the results of the predictions. The proposed DrugMGR achieves excellent overall performance in these datasets, exhibiting its advantages and merits against state-of-the-art methods. Thus, the downstream task of DrugMGR can be fine-tuned for identifying the potential compounds that target proteins for clinical treatment. AVAILABILITY AND IMPLEMENTATION https://github.com/lixiaokun2020/DrugMGR.
Collapse
Affiliation(s)
- Xiaokun Li
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
- School of Computer Science and Technology, Heilongjiang University, Harbin 150080, China
- Postdoctoral Program of Heilongjiang Hengxun Technology Co., Ltd., Harbin 150090, China
| | - Qiang Yang
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150000, China
| | - Long Xu
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Weihe Dong
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
| | - Gongning Luo
- Computer, Electrical and Mathematical Sciences & Engineering Division, King Abdullah University of Science and Technology, KAUST, Thuwal 23955, Saudi Arabia
| | - Wei Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Suyu Dong
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
| | - Kuanquan Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Ping Xuan
- Department of Computer Science, School of Engineering, Shantou University, Shantou 515063, China
| | - Xianyu Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Xin Gao
- Computer, Electrical and Mathematical Sciences & Engineering Division, King Abdullah University of Science and Technology, KAUST, Thuwal 23955, Saudi Arabia
| |
Collapse
|
57
|
Zhang C, Xie L, Lu X, Mao R, Xu L, Xu X. Developing an Improved Cycle Architecture for AI-Based Generation of New Structures Aimed at Drug Discovery. Molecules 2024; 29:1499. [PMID: 38611779 PMCID: PMC11013495 DOI: 10.3390/molecules29071499] [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: 01/25/2024] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Drug discovery involves a crucial step of optimizing molecules with the desired structural groups. In the domain of computer-aided drug discovery, deep learning has emerged as a prominent technique in molecular modeling. Deep generative models, based on deep learning, play a crucial role in generating novel molecules when optimizing molecules. However, many existing molecular generative models have limitations as they solely process input information in a forward way. To overcome this limitation, we propose an improved generative model called BD-CycleGAN, which incorporates BiLSTM (bidirectional long short-term memory) and Mol-CycleGAN (molecular cycle generative adversarial network) to preserve the information of molecular input. To evaluate the proposed model, we assess its performance by analyzing the structural distribution and evaluation matrices of generated molecules in the process of structural transformation. The results demonstrate that the BD-CycleGAN model achieves a higher success rate and exhibits increased diversity in molecular generation. Furthermore, we demonstrate its application in molecular docking, where it successfully increases the docking score for the generated molecules. The proposed BD-CycleGAN architecture harnesses the power of deep learning to facilitate the generation of molecules with desired structural features, thus offering promising advancements in the field of drug discovery processes.
Collapse
Affiliation(s)
| | | | | | | | - Lei Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou 213001, China; (C.Z.); (L.X.); (X.L.); (R.M.)
| | - Xiaojun Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou 213001, China; (C.Z.); (L.X.); (X.L.); (R.M.)
| |
Collapse
|
58
|
Katsoulakis E, Wang Q, Wu H, Shahriyari L, Fletcher R, Liu J, Achenie L, Liu H, Jackson P, Xiao Y, Syeda-Mahmood T, Tuli R, Deng J. Digital twins for health: a scoping review. NPJ Digit Med 2024; 7:77. [PMID: 38519626 PMCID: PMC10960047 DOI: 10.1038/s41746-024-01073-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 03/07/2024] [Indexed: 03/25/2024] Open
Abstract
The use of digital twins (DTs) has proliferated across various fields and industries, with a recent surge in the healthcare sector. The concept of digital twin for health (DT4H) holds great promise to revolutionize the entire healthcare system, including management and delivery, disease treatment and prevention, and health well-being maintenance, ultimately improving human life. The rapid growth of big data and continuous advancement in data science (DS) and artificial intelligence (AI) have the potential to significantly expedite DT research and development by providing scientific expertise, essential data, and robust cybertechnology infrastructure. Although various DT initiatives have been underway in the industry, government, and military, DT4H is still in its early stages. This paper presents an overview of the current applications of DTs in healthcare, examines consortium research centers and their limitations, and surveys the current landscape of emerging research and development opportunities in healthcare. We envision the emergence of a collaborative global effort among stakeholders to enhance healthcare and improve the quality of life for millions of individuals worldwide through pioneering research and development in the realm of DT technology.
Collapse
Affiliation(s)
- Evangelia Katsoulakis
- VA Informatics and Computing Infrastructure, Salt Lake City, UT, 84148, USA
- Department of Radiation Oncology, University of South Florida, Tampa, FL, 33606, USA
| | - Qi Wang
- Department of Mathematics, University of South Carolina, Columbia, SC, 29208, USA
| | - Huanmei Wu
- Department of Health Services Administration and Policy, Temple University, Philadelphia, PA, 19122, USA
| | - Leili Shahriyari
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Richard Fletcher
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, 02139, USA
| | - Jinwei Liu
- Department of Computer and Information Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Luke Achenie
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24060, USA
| | - Hongfang Liu
- McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Pamela Jackson
- Precision Neurotherapeutics Innovation Program & Department of Neurosurgery, Mayo Clinic, Phoenix, AZ, 85003, USA
| | - Ying Xiao
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | - Richard Tuli
- Department of Radiation Oncology, University of South Florida, Tampa, FL, 33606, USA
| | - Jun Deng
- Department of Therapeutic Radiology, Yale University, New Haven, CT, 06510, USA.
| |
Collapse
|
59
|
Siddique F, Anwaar A, Bashir M, Nadeem S, Rawat R, Eyupoglu V, Afzal S, Bibi M, Bin Jardan YA, Bourhia M. Revisiting methotrexate and phototrexate Zinc15 library-based derivatives using deep learning in-silico drug design approach. Front Chem 2024; 12:1380266. [PMID: 38576849 PMCID: PMC10991842 DOI: 10.3389/fchem.2024.1380266] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/05/2024] [Indexed: 04/06/2024] Open
Abstract
Introduction: Cancer is the second most prevalent cause of mortality in the world, despite the availability of several medications for cancer treatment. Therefore, the cancer research community emphasized on computational techniques to speed up the discovery of novel anticancer drugs. Methods: In the current study, QSAR-based virtual screening was performed on the Zinc15 compound library (271 derivatives of methotrexate (MTX) and phototrexate (PTX)) to predict their inhibitory activity against dihydrofolate reductase (DHFR), a potential anticancer drug target. The deep learning-based ADMET parameters were employed to generate a 2D QSAR model using the multiple linear regression (MPL) methods with Leave-one-out cross-validated (LOO-CV) Q2 and correlation coefficient R2 values as high as 0.77 and 0.81, respectively. Results: From the QSAR model and virtual screening analysis, the top hits (09, 27, 41, 68, 74, 85, 99, 180) exhibited pIC50 ranging from 5.85 to 7.20 with a minimum binding score of -11.6 to -11.0 kcal/mol and were subjected to further investigation. The ADMET attributes using the message-passing neural network (MPNN) model demonstrated the potential of selected hits as an oral medication based on lipophilic profile Log P (0.19-2.69) and bioavailability (76.30% to 78.46%). The clinical toxicity score was 31.24% to 35.30%, with the least toxicity score (8.30%) observed with compound 180. The DFT calculations were carried out to determine the stability, physicochemical parameters and chemical reactivity of selected compounds. The docking results were further validated by 100 ns molecular dynamic simulation analysis. Conclusion: The promising lead compounds found endorsed compared to standard reference drugs MTX and PTX that are best for anticancer activity and can lead to novel therapies after experimental validations. Furthermore, it is suggested to unveil the inhibitory potential of identified hits via in-vitro and in-vivo approaches.
Collapse
Affiliation(s)
- Farhan Siddique
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Ahmar Anwaar
- Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Maryam Bashir
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
- Southern Punjab Institute of Health Sciences, Multan, Pakistan
| | - Sumaira Nadeem
- Department of Pharmacy, The Women University, Multan, Pakistan
| | - Ravi Rawat
- School of Health Sciences & Technology, UPES University, Dehradun, India
| | - Volkan Eyupoglu
- Department of Chemistry, Cankırı Karatekin University, Cankırı, Türkiye
| | - Samina Afzal
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Mehvish Bibi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Yousef A. Bin Jardan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed Bourhia
- Laboratory of Biotechnology and Natural Resources Valorization, Faculty of Sciences, Ibn Zohr University, Agadir, Morocco
| |
Collapse
|
60
|
Shoombuatong W, Homdee N, Schaduangrat N, Chumnanpuen P. Leveraging a meta-learning approach to advance the accuracy of Na v blocking peptides prediction. Sci Rep 2024; 14:4463. [PMID: 38396246 PMCID: PMC10891130 DOI: 10.1038/s41598-024-55160-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 02/21/2024] [Indexed: 02/25/2024] Open
Abstract
The voltage-gated sodium (Nav) channel is a crucial molecular component responsible for initiating and propagating action potentials. While the α subunit, forming the channel pore, plays a central role in this function, the complete physiological function of Nav channels relies on crucial interactions between the α subunit and auxiliary proteins, known as protein-protein interactions (PPI). Nav blocking peptides (NaBPs) have been recognized as a promising and alternative therapeutic agent for pain and itch. Although traditional experimental methods can precisely determine the effect and activity of NaBPs, they remain time-consuming and costly. Hence, machine learning (ML)-based methods that are capable of accurately contributing in silico prediction of NaBPs are highly desirable. In this study, we develop an innovative meta-learning-based NaBP prediction method (MetaNaBP). MetaNaBP generates new feature representations by employing a wide range of sequence-based feature descriptors that cover multiple perspectives, in combination with powerful ML algorithms. Then, these feature representations were optimized to identify informative features using a two-step feature selection method. Finally, the selected informative features were applied to develop the final meta-predictor. To the best of our knowledge, MetaNaBP is the first meta-predictor for NaBP prediction. Experimental results demonstrated that MetaNaBP achieved an accuracy of 0.948 and a Matthews correlation coefficient of 0.898 over the independent test dataset, which were 5.79% and 11.76% higher than the existing method. In addition, the discriminative power of our feature representations surpassed that of conventional feature descriptors over both the training and independent test datasets. We anticipate that MetaNaBP will be exploited for the large-scale prediction and analysis of NaBPs to narrow down the potential NaBPs.
Collapse
Affiliation(s)
- Watshara Shoombuatong
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand.
| | - Nutta Homdee
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Nalini Schaduangrat
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Pramote Chumnanpuen
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand
- Omics Center for Agriculture, Bioresources, Food, and Health, Kasetsart University (OmiKU), Bangkok, 10900, Thailand
| |
Collapse
|
61
|
Visan AI, Negut I. Integrating Artificial Intelligence for Drug Discovery in the Context of Revolutionizing Drug Delivery. Life (Basel) 2024; 14:233. [PMID: 38398742 PMCID: PMC10890405 DOI: 10.3390/life14020233] [Citation(s) in RCA: 46] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/03/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Drug development is expensive, time-consuming, and has a high failure rate. In recent years, artificial intelligence (AI) has emerged as a transformative tool in drug discovery, offering innovative solutions to complex challenges in the pharmaceutical industry. This manuscript covers the multifaceted role of AI in drug discovery, encompassing AI-assisted drug delivery design, the discovery of new drugs, and the development of novel AI techniques. We explore various AI methodologies, including machine learning and deep learning, and their applications in target identification, virtual screening, and drug design. This paper also discusses the historical development of AI in medicine, emphasizing its profound impact on healthcare. Furthermore, it addresses AI's role in the repositioning of existing drugs and the identification of drug combinations, underscoring its potential in revolutionizing drug delivery systems. The manuscript provides a comprehensive overview of the AI programs and platforms currently used in drug discovery, illustrating the technological advancements and future directions of this field. This study not only presents the current state of AI in drug discovery but also anticipates its future trajectory, highlighting the challenges and opportunities that lie ahead.
Collapse
Affiliation(s)
| | - Irina Negut
- National Institute for Lasers, Plasma and Radiation Physics, 409 Atomistilor Street, 077125 Magurele, Ilfov, Romania;
| |
Collapse
|
62
|
Cichońska A, Ravikumar B, Rahman R. AI for targeted polypharmacology: The next frontier in drug discovery. Curr Opin Struct Biol 2024; 84:102771. [PMID: 38215530 DOI: 10.1016/j.sbi.2023.102771] [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: 09/15/2023] [Revised: 11/30/2023] [Accepted: 12/20/2023] [Indexed: 01/14/2024]
Abstract
In drug discovery, targeted polypharmacology, i.e., targeting multiple molecular targets with a single drug, is redefining therapeutic design to address complex diseases. Pre-selected pharmacological profiles, as exemplified in kinase drugs, promise enhanced efficacy and reduced toxicity. Historically, many of such drugs were discovered serendipitously, limiting predictability and efficacy, but currently artificial intelligence (AI) offers a transformative solution. Machine learning and deep learning techniques enable modeling protein structures, generating novel compounds, and decoding their polypharmacological effects, opening an avenue for more systematic and predictive multi-target drug design. This review explores the use of AI in identifying synergistic co-targets and delineating them from anti-targets that lead to adverse effects, and then discusses advances in AI-enabled docking, generative chemistry, and proteochemometric modeling of proteome-wide compound interactions, in the context of polypharmacology. We also provide insights into challenges ahead.
Collapse
|
63
|
Akermi S, Smaoui S, Chaari M, Elhadef K, Gentile R, Hait M, Roymahapatra G, Mellouli L. Combined in vitro/in silico approaches, molecular dynamics simulations and safety assessment of the multifunctional properties of thymol and carvacrol: A comparative insight. Chem Biodivers 2024; 21:e202301575. [PMID: 38116885 DOI: 10.1002/cbdv.202301575] [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: 10/07/2023] [Revised: 12/11/2023] [Accepted: 12/18/2023] [Indexed: 12/21/2023]
Abstract
Bioactive compounds derived from medicinal plants have acquired immense attentiveness in drug discovery and development. The present study investigated in vitro and predicted in silico the antibacterial, antifungal, and antiviral properties of thymol and carvacrol, and assessed their safety. The performed microbiological assays against Pseudomonas aeruginosa, Escherichia coli, Salmonella enterica Typhimurium revealed that the minimal inhibitory concentration values ranged from (0.078 to 0.312 mg/mL) and the minimal fungicidal concentration against Candida albicans was 0.625 mg/mL. Molecular docking simulations, stipulated that these compounds could inhibit bacterial replication and transcription functions by targeting DNA and RNA polymerases receptors with docking scores varying between (-5.1 to -6.9 kcal/mol). Studied hydroxylated monoterpenes could hinder C. albicans growth by impeding lanosterol 14α-demethylase enzyme and showed a (ΔG=-6.2 and -6.3 kcal/mol). Computational studies revealed that thymol and carvacrol could target the SARS-Cov-2 spike protein of the Omicron variant RBD domain. Molecular dynamics simulations disclosed that these compounds have a stable dynamic behavior over 100 ns as compared to remdesivir. Chemo-computational toxicity prediction using Protox II webserver indicated that thymol and carvacrol could be safely and effectively used as drug candidates to tackle bacterial, fungal, and viral infections as compared to chemical medication.
Collapse
Affiliation(s)
- Sarra Akermi
- Laboratory of Microbial and Enzymatic Biotechnologies and Biomolecules. Center of Biotechnology of Sfax (CBS), University of Sfax, Road of Sidi Mansour Km 6, P.O. Box 1177, Sfax, 3018, Sfax-, Tunisia
| | - Slim Smaoui
- Laboratory of Microbial and Enzymatic Biotechnologies and Biomolecules. Center of Biotechnology of Sfax (CBS), University of Sfax, Road of Sidi Mansour Km 6, P.O. Box 1177, Sfax, 3018, Sfax-, Tunisia
| | - Moufida Chaari
- Laboratory of Microbial and Enzymatic Biotechnologies and Biomolecules. Center of Biotechnology of Sfax (CBS), University of Sfax, Road of Sidi Mansour Km 6, P.O. Box 1177, Sfax, 3018, Sfax-, Tunisia
| | - Khaoula Elhadef
- Laboratory of Microbial and Enzymatic Biotechnologies and Biomolecules. Center of Biotechnology of Sfax (CBS), University of Sfax, Road of Sidi Mansour Km 6, P.O. Box 1177, Sfax, 3018, Sfax-, Tunisia
| | - Rocco Gentile
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Milan Hait
- Department of Chemistry, Dr. C. V. Raman University, Kota, 495113, Bilaspur, India
| | | | - Lotfi Mellouli
- Laboratory of Microbial and Enzymatic Biotechnologies and Biomolecules. Center of Biotechnology of Sfax (CBS), University of Sfax, Road of Sidi Mansour Km 6, P.O. Box 1177, Sfax, 3018, Sfax-, Tunisia
| |
Collapse
|
64
|
Dakilah I, Harb A, Abu-Gharbieh E, El-Huneidi W, Taneera J, Hamoudi R, Semreen MH, Bustanji Y. Potential of CDC25 phosphatases in cancer research and treatment: key to precision medicine. Front Pharmacol 2024; 15:1324001. [PMID: 38313315 PMCID: PMC10834672 DOI: 10.3389/fphar.2024.1324001] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/04/2024] [Indexed: 02/06/2024] Open
Abstract
The global burden of cancer continues to rise, underscoring the urgency of developing more effective and precisely targeted therapies. This comprehensive review explores the confluence of precision medicine and CDC25 phosphatases in the context of cancer research. Precision medicine, alternatively referred to as customized medicine, aims to customize medical interventions by taking into account the genetic, genomic, and epigenetic characteristics of individual patients. The identification of particular genetic and molecular drivers driving cancer helps both diagnostic accuracy and treatment selection. Precision medicine utilizes sophisticated technology such as genome sequencing and bioinformatics to elucidate genetic differences that underlie the proliferation of cancer cells, hence facilitating the development of customized therapeutic interventions. CDC25 phosphatases, which play a crucial role in governing the progression of the cell cycle, have garnered significant attention as potential targets for cancer treatment. The dysregulation of CDC25 is a characteristic feature observed in various types of malignancies, hence classifying them as proto-oncogenes. The proteins in question, which operate as phosphatases, play a role in the activation of Cyclin-dependent kinases (CDKs), so promoting the advancement of the cell cycle. CDC25 inhibitors demonstrate potential as therapeutic drugs for cancer treatment by specifically blocking the activity of CDKs and modulating the cell cycle in malignant cells. In brief, precision medicine presents a potentially fruitful option for augmenting cancer research, diagnosis, and treatment, with an emphasis on individualized care predicated upon patients' genetic and molecular profiles. The review highlights the significance of CDC25 phosphatases in the advancement of cancer and identifies them as promising candidates for therapeutic intervention. This statement underscores the significance of doing thorough molecular profiling in order to uncover the complex molecular characteristics of cancer cells.
Collapse
Affiliation(s)
- Ibraheem Dakilah
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Amani Harb
- Department of Basic Sciences, Faculty of Arts and Sciences, Al-Ahliyya Amman University, Amman, Jordan
| | - Eman Abu-Gharbieh
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Waseem El-Huneidi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Jalal Taneera
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Rifat Hamoudi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
| | - Mohammed H Semreen
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Yasser Bustanji
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- School of Pharmacy, The University of Jordan, Amman, Jordan
| |
Collapse
|
65
|
Nazli A, Qiu J, Tang Z, He Y. Recent Advances and Techniques for Identifying Novel Antibacterial Targets. Curr Med Chem 2024; 31:464-501. [PMID: 36734893 DOI: 10.2174/0929867330666230123143458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 10/30/2022] [Accepted: 11/11/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND With the emergence of drug-resistant bacteria, the development of new antibiotics is urgently required. Target-based drug discovery is the most frequently employed approach for the drug development process. However, traditional drug target identification techniques are costly and time-consuming. As research continues, innovative approaches for antibacterial target identification have been developed which enabled us to discover drug targets more easily and quickly. METHODS In this review, methods for finding drug targets from omics databases have been discussed in detail including principles, procedures, advantages, and potential limitations. The role of phage-driven and bacterial cytological profiling approaches is also discussed. Moreover, current article demonstrates the advancements being made in the establishment of computational tools, machine learning algorithms, and databases for antibacterial target identification. RESULTS Bacterial drug targets successfully identified by employing these aforementioned techniques are described as well. CONCLUSION The goal of this review is to attract the interest of synthetic chemists, biologists, and computational researchers to discuss and improve these methods for easier and quicker development of new drugs.
Collapse
Affiliation(s)
- Adila Nazli
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China
| | - Jingyi Qiu
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, 266 Fangzheng Avenue, Chongqing, 400714, P. R. China
| | - Ziyi Tang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, 266 Fangzheng Avenue, Chongqing, 400714, P. R. China
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China
| |
Collapse
|
66
|
Provasi D, Filizola M. Enhancing Opioid Bioactivity Predictions through Integration of Ligand-Based and Structure-Based Drug Discovery Strategies with Transfer and Deep Learning Techniques. J Phys Chem B 2023; 127:10691-10699. [PMID: 38084046 PMCID: PMC11252170 DOI: 10.1021/acs.jpcb.3c05306] [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] [Indexed: 12/22/2023]
Abstract
The opioid epidemic has cast a shadow over public health, necessitating immediate action to address its devastating consequences. To effectively combat this crisis, it is crucial to discover better opioid drugs with reduced addiction potential. Artificial intelligence-based and other machine learning tools, particularly deep learning models, have garnered significant attention in recent years for their potential to advance drug discovery. However, using these tools poses challenges, especially when training samples are insufficient to achieve adequate prediction performance. In this study, we investigate the effectiveness of transfer learning in building robust deep learning models to enhance ligand bioactivity prediction for each individual opioid receptor (OR) subtype. This is achieved by leveraging knowledge obtained from pretraining a model using supervised learning on a larger data set of bioactivity data combined with ligand-based and structure-based molecular descriptors related to the entire OR subfamily. Our studies hold the potential to advance opioid research by enabling the rapid identification of novel chemical probes with specific bioactivities, which can aid in the study of receptor function and contribute to the future development of improved opioid therapeutics.
Collapse
Affiliation(s)
- Davide Provasi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York 10029, United States
| | - Marta Filizola
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York 10029, United States
| |
Collapse
|
67
|
Cai L, Han F, Ji B, He X, Wang L, Niu T, Zhai J, Wang J. In Silico Screening of Natural Flavonoids against 3-Chymotrypsin-like Protease of SARS-CoV-2 Using Machine Learning and Molecular Modeling. Molecules 2023; 28:8034. [PMID: 38138524 PMCID: PMC10745665 DOI: 10.3390/molecules28248034] [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: 11/07/2023] [Revised: 11/30/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
The "Long-COVID syndrome" has posed significant challenges due to a lack of validated therapeutic options. We developed a novel multi-step virtual screening strategy to reliably identify inhibitors against 3-chymotrypsin-like protease of SARS-CoV-2 from abundant flavonoids, which represents a promising source of antiviral and immune-boosting nutrients. We identified 57 interacting residues as contributors to the protein-ligand binding pocket. Their energy interaction profiles constituted the input features for Machine Learning (ML) models. The consensus of 25 classifiers trained using various ML algorithms attained 93.9% accuracy and a 6.4% false-positive-rate. The consensus of 10 regression models for binding energy prediction also achieved a low root-mean-square error of 1.18 kcal/mol. We screened out 120 flavonoid hits first and retained 50 drug-like hits after predefined ADMET filtering to ensure bioavailability and safety profiles. Furthermore, molecular dynamics simulations prioritized nine bioactive flavonoids as promising anti-SARS-CoV-2 agents exhibiting both high structural stability (root-mean-square deviation < 5 Å for 218 ns) and low MM/PBSA binding free energy (<-6 kcal/mol). Among them, KB-2 (PubChem-CID, 14630497) and 9-O-Methylglyceofuran (PubChem-CID, 44257401) displayed excellent binding affinity and desirable pharmacokinetic capabilities. These compounds have great potential to serve as oral nutraceuticals with therapeutic and prophylactic properties as care strategies for patients with long-COVID syndrome.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Junmei Wang
- School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; (L.C.); (F.H.); (B.J.); (X.H.); (L.W.); (T.N.); (J.Z.)
| |
Collapse
|
68
|
Zhu Z, Yao Z, Zheng X, Qi G, Li Y, Mazur N, Gao X, Gong Y, Cong B. Drug-target affinity prediction method based on multi-scale information interaction and graph optimization. Comput Biol Med 2023; 167:107621. [PMID: 37907030 DOI: 10.1016/j.compbiomed.2023.107621] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 11/02/2023]
Abstract
Drug-target affinity (DTA) prediction as an emerging and effective method is widely applied to explore the strength of drug-target interactions in drug development research. By predicting these interactions, researchers can assess the potential efficacy and safety of candidate drugs at an early stage, narrowing down the search space for therapeutic targets and accelerating the discovery and development of new drugs. However, existing DTA prediction models mainly use graphical representations of drug molecules, which lack information on interactions between individual substructures, thus affecting prediction accuracy and model interpretability. Therefore, transformer and diffusion on drug graphs in DTA prediction (TDGraphDTA) are introduced to predict drug-target interactions using multi-scale information interaction and graph optimization. An interactive module is integrated into feature extraction of drug and target features at different granularity levels. A diffusion model-based graph optimization module is proposed to improve the representation of molecular graph structures and enhance the interpretability of graph representations while obtaining optimal feature representations. In addition, TDGraphDTA improves the accuracy and reliability of predictions by capturing relationships and contextual information between molecular substructures. The performance of the proposed TDGraphDTA in DTA prediction was verified on three publicly available benchmark datasets (Davis, Metz, and KIBA). Compared with state-of-the-art baseline models, it achieved better results in terms of consistency index, R-squared, etc. Furthermore, compared with some existing methods, the proposed TDGraphDTA is demonstrated to have better structure capturing capabilities by visualizing the feature capturing capabilities of the model using Grad-AAM toxicity labels in the ToxCast dataset. The corresponding source codes are available at https://github.com/Lamouryz/TDGraph.
Collapse
Affiliation(s)
- Zhiqin Zhu
- College of Automation, Chongqing University of Posts and Telecommunications, Chongqing, 400065, China.
| | - Zheng Yao
- College of Automation, Chongqing University of Posts and Telecommunications, Chongqing, 400065, China.
| | - Xin Zheng
- College of Automation, Chongqing University of Posts and Telecommunications, Chongqing, 400065, China.
| | - Guanqiu Qi
- Computer Information Systems Department, State University of New York at Buffalo State, Buffalo, NY 14222, USA.
| | - Yuanyuan Li
- College of Automation, Chongqing University of Posts and Telecommunications, Chongqing, 400065, China.
| | - Neal Mazur
- Computer Information Systems Department, State University of New York at Buffalo State, Buffalo, NY 14222, USA.
| | - Xinbo Gao
- College of Automation, Chongqing University of Posts and Telecommunications, Chongqing, 400065, China.
| | - Yifei Gong
- Faculty of applied science & engineering, the Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto at Toronto, ON M5S, Canada.
| | - Baisen Cong
- Diagnostics Digital, DH(Shanghai) Diagnostics Co, Ltd, a Danaher company, Shanghai, 200335, China.
| |
Collapse
|
69
|
Meng Y, Wang Y, Xu J, Lu C, Tang X, Peng T, Zhang B, Tian G, Yang J. Drug repositioning based on weighted local information augmented graph neural network. Brief Bioinform 2023; 25:bbad431. [PMID: 38019732 PMCID: PMC10686358 DOI: 10.1093/bib/bbad431] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 10/13/2023] [Accepted: 11/05/2023] [Indexed: 12/01/2023] Open
Abstract
Drug repositioning, the strategy of redirecting existing drugs to new therapeutic purposes, is pivotal in accelerating drug discovery. While many studies have engaged in modeling complex drug-disease associations, they often overlook the relevance between different node embeddings. Consequently, we propose a novel weighted local information augmented graph neural network model, termed DRAGNN, for drug repositioning. Specifically, DRAGNN firstly incorporates a graph attention mechanism to dynamically allocate attention coefficients to drug and disease heterogeneous nodes, enhancing the effectiveness of target node information collection. To prevent excessive embedding of information in a limited vector space, we omit self-node information aggregation, thereby emphasizing valuable heterogeneous and homogeneous information. Additionally, average pooling in neighbor information aggregation is introduced to enhance local information while maintaining simplicity. A multi-layer perceptron is then employed to generate the final association predictions. The model's effectiveness for drug repositioning is supported by a 10-times 10-fold cross-validation on three benchmark datasets. Further validation is provided through analysis of the predicted associations using multiple authoritative data sources, molecular docking experiments and drug-disease network analysis, laying a solid foundation for future drug discovery.
Collapse
Affiliation(s)
- Yajie Meng
- Center of Applied Mathematics & Interdisciplinary Science, School of Mathematical & Physical Sciences, Wuhan Textile University, No. 1, Yangguang Avenue, Jiangxia District, Wuhan City, Hubei Province 430200, China
| | - Yi Wang
- Center of Applied Mathematics & Interdisciplinary Science, School of Mathematical & Physical Sciences, Wuhan Textile University, No. 1, Yangguang Avenue, Jiangxia District, Wuhan City, Hubei Province 430200, China
| | - Junlin Xu
- College of Computer Science and Electronic Engineering, Hunan University, Lushan Road (S), Yuelu District, Changsha, Hunan Province 410082, China
| | - Changcheng Lu
- College of Computer Science and Electronic Engineering, Hunan University, Lushan Road (S), Yuelu District, Changsha, Hunan Province 410082, China
| | - Xianfang Tang
- Center of Applied Mathematics & Interdisciplinary Science, School of Mathematical & Physical Sciences, Wuhan Textile University, No. 1, Yangguang Avenue, Jiangxia District, Wuhan City, Hubei Province 430200, China
| | - Tao Peng
- Center of Applied Mathematics & Interdisciplinary Science, School of Mathematical & Physical Sciences, Wuhan Textile University, No. 1, Yangguang Avenue, Jiangxia District, Wuhan City, Hubei Province 430200, China
| | - Bengong Zhang
- Center of Applied Mathematics & Interdisciplinary Science, School of Mathematical & Physical Sciences, Wuhan Textile University, No. 1, Yangguang Avenue, Jiangxia District, Wuhan City, Hubei Province 430200, China
| | - Geng Tian
- Geneis Beijing Co., Ltd, No. 31, New North Road, Laiguanying, Chaoyang District, Beijing 100102, China
| | - Jialiang Yang
- Geneis Beijing Co., Ltd, No. 31, New North Road, Laiguanying, Chaoyang District, Beijing 100102, China
| |
Collapse
|
70
|
van Heerden A, Turon G, Duran-Frigola M, Pillay N, Birkholtz LM. Machine Learning Approaches Identify Chemical Features for Stage-Specific Antimalarial Compounds. ACS OMEGA 2023; 8:43813-43826. [PMID: 38027377 PMCID: PMC10666252 DOI: 10.1021/acsomega.3c05664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/18/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023]
Abstract
Efficacy data from diverse chemical libraries, screened against the various stages of the malaria parasite Plasmodium falciparum, including asexual blood stage (ABS) parasites and transmissible gametocytes, serve as a valuable reservoir of information on the chemical space of compounds that are either active (or not) against the parasite. We postulated that this data can be mined to define chemical features associated with the sole ABS activity and/or those that provide additional life cycle activity profiles like gametocytocidal activity. Additionally, this information could provide chemical features associated with inactive compounds, which could eliminate any future unnecessary screening of similar chemical analogs. Therefore, we aimed to use machine learning to identify the chemical space associated with stage-specific antimalarial activity. We collected data from various chemical libraries that were screened against the asexual (126 374 compounds) and sexual (gametocyte) stages of the parasite (93 941 compounds), calculated the compounds' molecular fingerprints, and trained machine learning models to recognize stage-specific active and inactive compounds. We were able to build several models that predict compound activity against ABS and dual activity against ABS and gametocytes, with Support Vector Machines (SVM) showing superior abilities with high recall (90 and 66%) and low false-positive predictions (15 and 1%). This allowed the identification of chemical features enriched in active and inactive populations, an important outcome that could be mined for essential chemical features to streamline hit-to-lead optimization strategies of antimalarial candidates. The predictive capabilities of the models held true in diverse chemical spaces, indicating that the ML models are therefore robust and can serve as a prioritization tool to drive and guide phenotypic screening and medicinal chemistry programs.
Collapse
Affiliation(s)
- Ashleigh van Heerden
- Department
of Biochemistry, Genetics and Microbiology, Institute for Sustainable
Malaria Control, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Gemma Turon
- Ersilia
Open Source Initiative, 28 Belgrave Road, Cambridge CB1 3DE, U.K.
| | | | - Nelishia Pillay
- Department
of Computer Science, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Lyn-Marié Birkholtz
- Department
of Biochemistry, Genetics and Microbiology, Institute for Sustainable
Malaria Control, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| |
Collapse
|
71
|
Slabaugh G, Beltran L, Rizvi H, Deloukas P, Marouli E. Applications of machine and deep learning to thyroid cytology and histopathology: a review. Front Oncol 2023; 13:958310. [PMID: 38023130 PMCID: PMC10661921 DOI: 10.3389/fonc.2023.958310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
This review synthesises past research into how machine and deep learning can improve the cyto- and histopathology processing pipelines for thyroid cancer diagnosis. The current gold-standard preoperative technique of fine-needle aspiration cytology has high interobserver variability, often returns indeterminate samples and cannot reliably identify some pathologies; histopathology analysis addresses these issues to an extent, but it requires surgical resection of the suspicious lesions so cannot influence preoperative decisions. Motivated by these issues, as well as by the chronic shortage of trained pathologists, much research has been conducted into how artificial intelligence could improve current pipelines and reduce the pressure on clinicians. Many past studies have indicated the significant potential of automated image analysis in classifying thyroid lesions, particularly for those of papillary thyroid carcinoma, but these have generally been retrospective, so questions remain about both the practical efficacy of these automated tools and the realities of integrating them into clinical workflows. Furthermore, the nature of thyroid lesion classification is significantly more nuanced in practice than many current studies have addressed, and this, along with the heterogeneous nature of processing pipelines in different laboratories, means that no solution has proven itself robust enough for clinical adoption. There are, therefore, multiple avenues for future research: examine the practical implementation of these algorithms as pathologist decision-support systems; improve interpretability, which is necessary for developing trust with clinicians and regulators; and investigate multiclassification on diverse multicentre datasets, aiming for methods that demonstrate high performance in a process- and equipment-agnostic manner.
Collapse
Affiliation(s)
- Greg Slabaugh
- Digital Environment Research Institute, Queen Mary University of London, London, United Kingdom
| | - Luis Beltran
- Barts Health NHS Trust, The Royal London Hospital, London, United Kingdom
| | - Hasan Rizvi
- Barts Health NHS Trust, The Royal London Hospital, London, United Kingdom
| | - Panos Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Eirini Marouli
- Digital Environment Research Institute, Queen Mary University of London, London, United Kingdom
- Barts Health NHS Trust, The Royal London Hospital, London, United Kingdom
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
72
|
Liu H, Yu S, Li X, Wang X, Qi D, Pan F, Chai X, Wang Q, Pan Y, Zhang L, Liu Y. Integration of Deep Learning and Sequential Metabolism to Rapidly Screen Dipeptidyl Peptidase (DPP)-IV Inhibitors from Gardenia jasminoides Ellis. Molecules 2023; 28:7381. [PMID: 37959800 PMCID: PMC10649927 DOI: 10.3390/molecules28217381] [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: 10/01/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Traditional Chinese medicine (TCM) possesses unique advantages in the management of blood glucose and lipids. However, there is still a significant gap in the exploration of its pharmacologically active components. Integrated strategies encompassing deep-learning prediction models and active validation based on absorbable ingredients can greatly improve the identification rate and screening efficiency in TCM. In this study, the affinity prediction of 11,549 compounds from the traditional Chinese medicine system's pharmacology database (TCMSP) with dipeptidyl peptidase-IV (DPP-IV) based on a deep-learning model was firstly conducted. With the results, Gardenia jasminoides Ellis (GJE), a food medicine with homologous properties, was selected as a model drug. The absorbed components of GJE were subsequently identified through in vivo intestinal perfusion and oral administration. As a result, a total of 38 prototypical absorbed components of GJE were identified. These components were analyzed to determine their absorption patterns after intestinal, hepatic, and systemic metabolism. Virtual docking and DPP-IV enzyme activity experiments were further conducted to validate the inhibitory effects and potential binding sites of the common constituents of deep learning and sequential metabolism. The results showed a significant DPP-IV inhibitory activity (IC50 53 ± 0.63 μg/mL) of the iridoid glycosides' potent fractions, which is a novel finding. Genipin 1-gentiobioside was screened as a promising new DPP-IV inhibitor in GJE. These findings highlight the potential of this innovative approach for the rapid screening of active ingredients in TCM and provide insights into the molecular mechanisms underlying the anti-diabetic activity of GJE.
Collapse
Affiliation(s)
- Huining Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (H.L.); (S.Y.); (X.L.); (X.W.); (D.Q.); (F.P.); (X.C.); (Q.W.)
| | - Shuang Yu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (H.L.); (S.Y.); (X.L.); (X.W.); (D.Q.); (F.P.); (X.C.); (Q.W.)
| | - Xueyan Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (H.L.); (S.Y.); (X.L.); (X.W.); (D.Q.); (F.P.); (X.C.); (Q.W.)
| | - Xinyu Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (H.L.); (S.Y.); (X.L.); (X.W.); (D.Q.); (F.P.); (X.C.); (Q.W.)
| | - Dongying Qi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (H.L.); (S.Y.); (X.L.); (X.W.); (D.Q.); (F.P.); (X.C.); (Q.W.)
| | - Fulu Pan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (H.L.); (S.Y.); (X.L.); (X.W.); (D.Q.); (F.P.); (X.C.); (Q.W.)
| | - Xiaoyu Chai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (H.L.); (S.Y.); (X.L.); (X.W.); (D.Q.); (F.P.); (X.C.); (Q.W.)
| | - Qianqian Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (H.L.); (S.Y.); (X.L.); (X.W.); (D.Q.); (F.P.); (X.C.); (Q.W.)
| | - Yanli Pan
- Institute of Information on Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Lei Zhang
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Yang Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (H.L.); (S.Y.); (X.L.); (X.W.); (D.Q.); (F.P.); (X.C.); (Q.W.)
| |
Collapse
|
73
|
Jabin T, Biswas S, Islam S, Sarker S, Afroze M, Paul GK, Razu MH, Monirruzzaman M, Huda M, Rahman M, Kundu NK, Kamal S, Karmakar P, Islam MA, Saleh MA, Khan M, Zaman S. Effects of gamma-radiation on microbial, nutritional, and functional properties of Katimon mango peels: A combined biochemical and in silico studies. Heliyon 2023; 9:e21556. [PMID: 38027912 PMCID: PMC10665690 DOI: 10.1016/j.heliyon.2023.e21556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Gamma radiation has notable impacts on the flesh of mangoes. In this research, Katimon mangoes were subjected to different levels of irradiation (0.5, 1.0, 1.5, and 2.0 kGy) using a60Co irradiator. The results showed that irradiation significantly reduced the microbial population in the mango peels, with the 1.5 kGy dose showing the most significant reduction. Irradiation also delayed ripening and extended the shelf life of the mango peels. The total fat, protein, ash, moisture, and sugar content of the mango peels were all affected by irradiation. The total protein content, ash content and moisture content increased after irradiation, while the fat content remained relatively unchanged. The sugar content increased in all samples after storage, but the non-irradiated samples had higher sugar levels than the irradiated ones. The dietary fiber content of the mango peels was not significantly affected by irradiation. The vitamin C content decreased in all samples after storage. The titratable acidity and total soluble solids content of the mango peels increased after storage, but there were no significant differences between the irradiated and non-irradiated samples. Antioxidant activity and cytotoxicity assessment highlighted the antioxidant potential and reduced toxicity of irradiated samples. Additionally, the antimicrobial effectiveness of irradiated mango peels was evaluated. The most substantial inhibitory zones (measuring 16.90 ± 0.35) against Pseudomonas sp. were observed at a radiation dose of 1.5 kGy with 150 μg/disc. To identify potential antimicrobial agents, the volatile components of mangoes irradiated with 1.5 kGy were analyzed through GC-MS. Subsequently, these compounds were subjected to in silico studies against a viable protein, TgpA, of Pseudomonas sp. (PDB ID: 6G49). Based on molecular dynamic simulations and ADMET properties, (-)-Carvone (-6.2), p-Cymene (-6.1), and Acetic acid phenylmethyl ester (-6.1) were identified as promising compounds for controlling Pseudomonas sp.
Collapse
Affiliation(s)
- Tabassum Jabin
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Bangladesh
- Bangladesh Reference Institute for Chemical Measurements (BRiCM), Dhaka, Bangladesh
| | - Suvro Biswas
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Bangladesh
| | - Shirmin Islam
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Bangladesh
| | - Swagotom Sarker
- Bangladesh Reference Institute for Chemical Measurements (BRiCM), Dhaka, Bangladesh
| | - Mirola Afroze
- Bangladesh Reference Institute for Chemical Measurements (BRiCM), Dhaka, Bangladesh
| | - Gobindo Kumar Paul
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Bangladesh
| | - Mamudul Hasan Razu
- Bangladesh Reference Institute for Chemical Measurements (BRiCM), Dhaka, Bangladesh
| | - Md Monirruzzaman
- Bangladesh Reference Institute for Chemical Measurements (BRiCM), Dhaka, Bangladesh
| | - Mainul Huda
- Bangladesh Reference Institute for Chemical Measurements (BRiCM), Dhaka, Bangladesh
| | - Mashiur Rahman
- Bangladesh Reference Institute for Chemical Measurements (BRiCM), Dhaka, Bangladesh
| | - Nayan Kumer Kundu
- Bangladesh Reference Institute for Chemical Measurements (BRiCM), Dhaka, Bangladesh
| | - Sabiha Kamal
- Bangladesh Reference Institute for Chemical Measurements (BRiCM), Dhaka, Bangladesh
| | - Pranab Karmakar
- Bangladesh Reference Institute for Chemical Measurements (BRiCM), Dhaka, Bangladesh
| | - Md Ariful Islam
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Bangladesh
| | - Md Abu Saleh
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Bangladesh
| | - Mala Khan
- Bangladesh Reference Institute for Chemical Measurements (BRiCM), Dhaka, Bangladesh
| | - Shahriar Zaman
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Bangladesh
| |
Collapse
|
74
|
Lu Y, Chen Z, Pan Y, Qi F. Identification of Drug Compounds for Capsular Contracture Based on Text Mining and Deep Learning. Plast Reconstr Surg 2023; 152:779e-790e. [PMID: 36862957 DOI: 10.1097/prs.0000000000010350] [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] [Indexed: 03/04/2023]
Abstract
BACKGROUND Capsular contracture is a common and unpredictable complication after breast implant placement. Currently, the pathogenesis of capsular contracture is unclear, and the effectiveness of nonsurgical treatment is still doubtful. The authors' study aimed to investigate new drug therapies for capsular contracture by using computational methods. METHODS Genes related to capsular contracture were identified by text mining and GeneCodis. Then, the candidate key genes were selected through protein-protein interaction analysis in Search Tool for the Retrieval of Interacting Genes/Proteins and Cytoscape. Drugs targeting the candidate genes with relation to capsular contracture were screened out in Pharmaprojects. Based on the drug-target interaction analysis by DeepPurpose, candidate drugs with highest predicted binding affinity were obtained eventually. RESULTS The authors' study identified 55 genes related to capsular contracture. Gene set enrichment analysis and protein-protein interaction analysis generated eight candidate genes. One hundred drugs targeting the candidate genes were selected. The seven candidate drugs with the highest predicted binding affinity were determined by DeepPurpose, including tumor necrosis factor alpha antagonist, estrogen receptor agonist, insulin-like growth factor 1 receptor, tyrosine kinase inhibitor, and matrix metallopeptidase 1 inhibitor. CONCLUSION Text mining and DeepPurpose can be used as a promising tool for drug discovery in exploring nonsurgical treatment to capsular contracture. CLINICAL QUESTION/LEVEL OF EVIDENCE Therapeutic, V.
Collapse
Affiliation(s)
- Yeheng Lu
- From the Department of Plastic Surgery, Zhongshan Hospital
| | - Zhiwei Chen
- Big Data and Artificial Intelligence Center, Zhongshan Hospital, Fudan University
| | - Yuyan Pan
- From the Department of Plastic Surgery, Zhongshan Hospital
| | - Fazhi Qi
- From the Department of Plastic Surgery, Zhongshan Hospital
| |
Collapse
|
75
|
Shan W, Chen L, Xu H, Zhong Q, Xu Y, Yao H, Lin K, Li X. GcForest-based compound-protein interaction prediction model and its application in discovering small-molecule drugs targeting CD47. Front Chem 2023; 11:1292869. [PMID: 37927570 PMCID: PMC10623438 DOI: 10.3389/fchem.2023.1292869] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
Identifying compound-protein interaction plays a vital role in drug discovery. Artificial intelligence (AI), especially machine learning (ML) and deep learning (DL) algorithms, are playing increasingly important roles in compound-protein interaction (CPI) prediction. However, ML relies on learning from large sample data. And the CPI for specific target often has a small amount of data available. To overcome the dilemma, we propose a virtual screening model, in which word2vec is used as an embedding tool to generate low-dimensional vectors of SMILES of compounds and amino acid sequences of proteins, and the modified multi-grained cascade forest based gcForest is used as the classifier. This proposed method is capable of constructing a model from raw data, adjusting model complexity according to the scale of datasets, especially for small scale datasets, and is robust with few hyper-parameters and without over-fitting. We found that the proposed model is superior to other CPI prediction models and performs well on the constructed challenging dataset. We finally predicted 2 new inhibitors for clusters of differentiation 47(CD47) which has few known inhibitors. The IC50s of enzyme activities of these 2 new small molecular inhibitors targeting CD47-SIRPα interaction are 3.57 and 4.79 μM respectively. These results fully demonstrate the competence of this concise but efficient tool for CPI prediction.
Collapse
Affiliation(s)
- Wenying Shan
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Lvqi Chen
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hao Xu
- Institute of Chemical Industry of Forest Products, Chinese Academy of Forestry, Nanjing, China
- National Engineering Laboratory for Biomass Chemical Utilization, Nanjing, China
| | - Qinghao Zhong
- School of Humanities and Social Sciences, The Chinese University of Hong Kong, Shenzhen, China
| | - Yinqiu Xu
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hequan Yao
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Kejiang Lin
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xuanyi Li
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
76
|
Cui S, Gao Y, Huang Y, Shen L, Zhao Q, Pan Y, Zhuang S. Advances and applications of machine learning and deep learning in environmental ecology and health. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 335:122358. [PMID: 37567408 DOI: 10.1016/j.envpol.2023.122358] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/02/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
Machine learning (ML) and deep learning (DL) possess excellent advantages in data analysis (e.g., feature extraction, clustering, classification, regression, image recognition and prediction) and risk assessment and management in environmental ecology and health (EEH). Considering the rapid growth and increasing complexity of data in EEH, it is of significance to summarize recent advances and applications of ML and DL in EEH. This review summarized the basic processes and fundamental algorithms of the ML and DL modeling, and indicated the urgent needs of ML and DL in EEH. Recent research hotspots such as environmental ecology and restoration, environmental fate of new pollutants, chemical exposures and risks, chemical hazard identification and control were highlighted. Various applications of ML and DL in EEH demonstrate their versatility and technological revolution, and present some challenges. The perspective of ML and DL in EEH were further outlined to promote the innovative analysis and cultivation of the ML-driven research paradigm.
Collapse
Affiliation(s)
- Shixuan Cui
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China; Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Yuchen Gao
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yizhou Huang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Lilai Shen
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qiming Zhao
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yaru Pan
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Shulin Zhuang
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China; Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China.
| |
Collapse
|
77
|
Omar A, Abd El-Hafeez T. Quantum computing and machine learning for Arabic language sentiment classification in social media. Sci Rep 2023; 13:17305. [PMID: 37828056 PMCID: PMC10570340 DOI: 10.1038/s41598-023-44113-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 10/03/2023] [Indexed: 10/14/2023] Open
Abstract
With the increasing amount of digital data generated by Arabic speakers, the need for effective and efficient document classification techniques is more important than ever. In recent years, both quantum computing and machine learning have shown great promise in the field of document classification. However, there is a lack of research investigating the performance of these techniques on the Arabic language. This paper presents a comparative study of quantum computing and machine learning for two datasets of Arabic language document classification. In the first dataset of 213,465 Arabic tweets, both classic machine learning (ML) and quantum computing approaches achieve high accuracy in sentiment analysis, with quantum computing slightly outperforming classic ML. Quantum computing completes the task in approximately 59 min, slightly faster than classic ML, which takes around 1 h. The precision, recall, and F1 score metrics indicate the effectiveness of both approaches in predicting sentiment in Arabic tweets. Classic ML achieves precision, recall, and F1 score values of 0.8215, 0.8175, and 0.8121, respectively, while quantum computing achieves values of 0.8239, 0.8199, and 0.8147, respectively. In the second dataset of 44,000 tweets, both classic ML (using the Random Forest algorithm) and quantum computing demonstrate significantly reduced processing times compared to the first dataset, with no substantial difference between them. Classic ML completes the analysis in approximately 2 min, while quantum computing takes approximately 1 min and 53 s. The accuracy of classic ML is higher at 0.9241 compared to 0.9205 for quantum computing. However, both approaches achieve high precision, recall, and F1 scores, indicating their effectiveness in accurately predicting sentiment in the dataset. Classic ML achieves precision, recall, and F1 score values of 0.9286, 0.9241, and 0.9249, respectively, while quantum computing achieves values of 0.92456, 0.9205, and 0.9214, respectively. The analysis of the metrics indicates that quantum computing approaches are effective in identifying positive instances and capturing relevant sentiment information in large datasets. On the other hand, traditional machine learning techniques exhibit faster processing times when dealing with smaller dataset sizes. This study provides valuable insights into the strengths and limitations of quantum computing and machine learning for Arabic document classification, emphasizing the potential of quantum computing in achieving high accuracy, particularly in scenarios where traditional machine learning techniques may encounter difficulties. These findings contribute to the development of more accurate and efficient document classification systems for Arabic data.
Collapse
Affiliation(s)
- Ahmed Omar
- Department of Computer Science, Faculty of Science, Minia University, EL-Minia, Egypt.
| | - Tarek Abd El-Hafeez
- Department of Computer Science, Faculty of Science, Minia University, EL-Minia, Egypt.
- Computer Science Unit, Deraya University, EL-Minia, Egypt.
| |
Collapse
|
78
|
Sabei FY, Y Safhi A, Almoshari Y, Salawi A, H Sultan M, Ali Bakkari M, Alsalhi A, A Madkhali O, M Jali A, Ahsan W. Structure-based virtual screening of natural compounds as inhibitors of HCV using molecular docking and molecular dynamics simulation studies. J Biomol Struct Dyn 2023; 42:11574-11585. [PMID: 37776007 DOI: 10.1080/07391102.2023.2263588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 08/28/2023] [Indexed: 10/01/2023]
Abstract
The hepatitis C virus (HCV), which causes hepatitis C, is a viral infection that damages the liver and causes inflammation in the liver. New potentially effective antiviral drugs are required for its treatment owing to various issues associated with the existing medications, including moderate to severe adverse effects, higher costs, and the emergence of drug-resistant strains. The objective of the current study was to utilize computational techniques to assess the anti-HCV efficacy of certain phytochemicals against tetraspanin (CD81) and claudin 1 (CLDN1) entry proteins. A 200-nanosecond molecular dynamics (MD) simulation was employed to examine the stability of the lead-protein complexes. Free binding energy and molecular docking calculations were conducted utilizing MM/GBSA method, and the selectivity of hit compounds for CD81 and CLDN1 was determined. Five significant CD81 and CLDN1 inhibitors were identified: Petasiphenone, Silibinin, Tanshinone IIA, Taxifolin, and Topaquinone. The MM/GBSA analysis of the compounds revealed high free binding energies. All the identified compounds were stable within the CD81 and CLDN1 binding pockets. This study indicated the promising inhibitory potential of the identified compounds against CD81 and CLDN1 receptors and might develop into potential viral entry inhibitors. However, to validate the chemotherapeutic capabilities of the discovered leads extensive preclinical research is required.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Fahad Y Sabei
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Awaji Y Safhi
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Yosif Almoshari
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Ahmad Salawi
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Muhammad H Sultan
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Mohammed Ali Bakkari
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Abdullah Alsalhi
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Osama A Madkhali
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Abdulmajeed M Jali
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Waquar Ahsan
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
79
|
Wang Z, Sun L, Xu Y, Liang P, Xu K, Huang J. Discovery of novel JAK1 inhibitors through combining machine learning, structure-based pharmacophore modeling and bio-evaluation. J Transl Med 2023; 21:579. [PMID: 37641144 PMCID: PMC10464202 DOI: 10.1186/s12967-023-04443-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND Janus kinase 1 (JAK1) plays a critical role in most cytokine-mediated inflammatory, autoimmune responses and various cancers via the JAK/STAT signaling pathway. Inhibition of JAK1 is therefore an attractive therapeutic strategy for several diseases. Recently, high-performance machine learning techniques have been increasingly applied in virtual screening to develop new kinase inhibitors. Our study aimed to develop a novel layered virtual screening method based on machine learning (ML) and pharmacophore models to identify the potential JAK1 inhibitors. METHODS Firstly, we constructed a high-quality dataset comprising 3834 JAK1 inhibitors and 12,230 decoys, followed by establishing a series of classification models based on a combination of three molecular descriptors and six ML algorithms. To further screen potential compounds, we constructed several pharmacophore models based on Hiphop and receptor-ligand algorithms. We then used molecular docking to filter the recognized compounds. Finally, the binding stability and enzyme inhibition activity of the identified compounds were assessed by molecular dynamics (MD) simulations and in vitro enzyme activity tests. RESULTS The best performance ML model DNN-ECFP4 and two pharmacophore models Hiphop3 and 6TPF 08 were utilized to screen the ZINC database. A total of 13 potentially active compounds were screened and the MD results demonstrated that all of the above molecules could bind with JAK1 stably in dynamic conditions. Among the shortlisted compounds, the four purchasable compounds demonstrated significant kinase inhibition activity, with Z-10 being the most active (IC50 = 194.9 nM). CONCLUSION The current study provides an efficient and accurate integrated model. The hit compounds were promising candidates for the further development of novel JAK1 inhibitors.
Collapse
Affiliation(s)
- Zixiao Wang
- Department of Pharmacy, Honghui Hospital, Xi' an Jiaotong University, Xi' an, 710054, China.
| | - Lili Sun
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yu Xu
- State Key Laboratory of Natural Medicines,Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery,China Pharmaceutical University, Nanjing, 210009, China
| | - Peida Liang
- Department of Pharmacy, Honghui Hospital, Xi' an Jiaotong University, Xi' an, 710054, China
| | - Kaiyan Xu
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Jing Huang
- Department of Pharmacy, Honghui Hospital, Xi' an Jiaotong University, Xi' an, 710054, China.
| |
Collapse
|
80
|
Li X, Yang Q, Luo G, Xu L, Dong W, Wang W, Dong S, Wang K, Xuan P, Gao X. SAGDTI: self-attention and graph neural network with multiple information representations for the prediction of drug-target interactions. BIOINFORMATICS ADVANCES 2023; 3:vbad116. [PMID: 38282612 PMCID: PMC10818136 DOI: 10.1093/bioadv/vbad116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/31/2023] [Accepted: 08/24/2023] [Indexed: 01/30/2024]
Abstract
Motivation Accurate identification of target proteins that interact with drugs is a vital step in silico, which can significantly foster the development of drug repurposing and drug discovery. In recent years, numerous deep learning-based methods have been introduced to treat drug-target interaction (DTI) prediction as a classification task. The output of this task is binary identification suggesting the absence or presence of interactions. However, existing studies often (i) neglect the unique molecular attributes when embedding drugs and proteins, and (ii) determine the interaction of drug-target pairs without considering biological interaction information. Results In this study, we propose an end-to-end attention-derived method based on the self-attention mechanism and graph neural network, termed SAGDTI. The aim of this method is to overcome the aforementioned drawbacks in the identification of DTI. SAGDTI is the first method to sufficiently consider the unique molecular attribute representations for both drugs and targets in the input form of the SMILES sequences and three-dimensional structure graphs. In addition, our method aggregates the feature attributes of biological information between drugs and targets through multi-scale topologies and diverse connections. Experimental results illustrate that SAGDTI outperforms existing prediction models, which benefit from the unique molecular attributes embedded by atom-level attention and biological interaction information representation aggregated by node-level attention. Moreover, a case study on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) shows that our model is a powerful tool for identifying DTIs in real life. Availability and implementation The data and codes underlying this article are available in Github at https://github.com/lixiaokun2020/SAGDTI.
Collapse
Affiliation(s)
- Xiaokun Li
- School of Computer Science and Technology, Heilongjiang University, Harbin 150080, China
- Postdoctoral Program of Heilongjiang Hengxun Technology Co., Ltd., Harbin 150090, China
| | - Qiang Yang
- School of Computer Science and Technology, Heilongjiang University, Harbin 150080, China
- Postdoctoral Program of Heilongjiang Hengxun Technology Co., Ltd., Harbin 150090, China
| | - Gongning Luo
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Long Xu
- School of Computer Science and Technology, Heilongjiang University, Harbin 150080, China
- Postdoctoral Program of Heilongjiang Hengxun Technology Co., Ltd., Harbin 150090, China
| | - Weihe Dong
- Postdoctoral Program of Heilongjiang Hengxun Technology Co., Ltd., Harbin 150090, China
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
| | - Wei Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Suyu Dong
- College of Computer and Control Engineering, Northeast Forestry University, Harbin 150040, China
| | - Kuanquan Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Ping Xuan
- School of Computer Science and Technology, Heilongjiang University, Harbin 150080, China
- Department of Computer Science, School of Engineering, Shantou University, Shantou 515063, China
| | - Xin Gao
- Computer, Electrical and Mathematical Sciences & Engineering Division, King Abdullah University of Science and Technology, 4700 KAUST, Thuwal 23955, Saudi Arabia
| |
Collapse
|
81
|
Provasi D, Filizola M. Enhancing Opioid Bioactivity Predictions through Integration of Ligand-Based and Structure-Based Drug Discovery Strategies with Transfer and Deep Learning Techniques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.04.552065. [PMID: 37609329 PMCID: PMC10441297 DOI: 10.1101/2023.08.04.552065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
The opioid epidemic has cast a shadow over public health, necessitating immediate action to address its devastating consequences. To effectively combat this crisis, it is crucial to discover better opioid drugs with reduced addiction potential. Artificial intelligence-based and other machine learning tools, particularly deep learning models, have garnered significant attention in recent years for their potential to advance drug discovery. However, utilizing these tools poses challenges, especially when training samples are insufficient to achieve adequate prediction performance. In this study, we investigate the effectiveness of transfer learning using combined ligand-based and structure-based molecular descriptors from the entire opioid receptor (OR) subfamily in building robust deep learning models for enhanced bioactivity prediction of opioid ligands at each individual OR subtype. Our studies hold the potential to greatly advance opioid research by enabling the rapid identification of novel chemical probes with specific bioactivities, which can aid in the study of receptor function and contribute to the future development of improved opioid therapeutics.
Collapse
|
82
|
Binatlı OC, Gönen M. MOKPE: drug-target interaction prediction via manifold optimization based kernel preserving embedding. BMC Bioinformatics 2023; 24:276. [PMID: 37407927 DOI: 10.1186/s12859-023-05401-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/25/2023] [Indexed: 07/07/2023] Open
Abstract
BACKGROUND In many applications of bioinformatics, data stem from distinct heterogeneous sources. One of the well-known examples is the identification of drug-target interactions (DTIs), which is of significant importance in drug discovery. In this paper, we propose a novel framework, manifold optimization based kernel preserving embedding (MOKPE), to efficiently solve the problem of modeling heterogeneous data. Our model projects heterogeneous drug and target data into a unified embedding space by preserving drug-target interactions and drug-drug, target-target similarities simultaneously. RESULTS We performed ten replications of ten-fold cross validation on four different drug-target interaction network data sets for predicting DTIs for previously unseen drugs. The classification evaluation metrics showed better or comparable performance compared to previous similarity-based state-of-the-art methods. We also evaluated MOKPE on predicting unknown DTIs of a given network. Our implementation of the proposed algorithm in R together with the scripts that replicate the reported experiments is publicly available at https://github.com/ocbinatli/mokpe .
Collapse
Affiliation(s)
- Oğuz C Binatlı
- Graduate School of Sciences and Engineering, Koç University, 34450, Istanbul, Turkey
| | - Mehmet Gönen
- Department of Industrial Engineering, College of Engineering, Koç University, 34450, Istanbul, Turkey.
- School of Medicine, Koç University, 34450, Istanbul, Turkey.
| |
Collapse
|
83
|
Qureshi R, Irfan M, Gondal TM, Khan S, Wu J, Hadi MU, Heymach J, Le X, Yan H, Alam T. AI in drug discovery and its clinical relevance. Heliyon 2023; 9:e17575. [PMID: 37396052 PMCID: PMC10302550 DOI: 10.1016/j.heliyon.2023.e17575] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/17/2023] [Accepted: 06/21/2023] [Indexed: 07/04/2023] Open
Abstract
The COVID-19 pandemic has emphasized the need for novel drug discovery process. However, the journey from conceptualizing a drug to its eventual implementation in clinical settings is a long, complex, and expensive process, with many potential points of failure. Over the past decade, a vast growth in medical information has coincided with advances in computational hardware (cloud computing, GPUs, and TPUs) and the rise of deep learning. Medical data generated from large molecular screening profiles, personal health or pathology records, and public health organizations could benefit from analysis by Artificial Intelligence (AI) approaches to speed up and prevent failures in the drug discovery pipeline. We present applications of AI at various stages of drug discovery pipelines, including the inherently computational approaches of de novo design and prediction of a drug's likely properties. Open-source databases and AI-based software tools that facilitate drug design are discussed along with their associated problems of molecule representation, data collection, complexity, labeling, and disparities among labels. How contemporary AI methods, such as graph neural networks, reinforcement learning, and generated models, along with structure-based methods, (i.e., molecular dynamics simulations and molecular docking) can contribute to drug discovery applications and analysis of drug responses is also explored. Finally, recent developments and investments in AI-based start-up companies for biotechnology, drug design and their current progress, hopes and promotions are discussed in this article.
Collapse
Affiliation(s)
- Rizwan Qureshi
- College of Science and Engineering, Hamad Bin Khalifa University, Doha, Qatar
- Department of Imaging Physics, MD Anderson Cancer Center, The University of Texas, Houston, USA
| | - Muhammad Irfan
- Faculty of Electrical Engineering, Ghulam Ishaq Khan Institute of Engineering Sciences and Technology, Swabi, Pakistan
| | | | - Sheheryar Khan
- School of Professional Education & Executive Development, The Hong Kong Polytechnic University, Hong Kong
| | - Jia Wu
- Department of Imaging Physics, MD Anderson Cancer Center, The University of Texas, Houston, USA
| | | | - John Heymach
- Department of Thoracic Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas, MD Anderson Cancer Center, Houston, USA
| | - Xiuning Le
- Department of Thoracic Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas, MD Anderson Cancer Center, Houston, USA
| | - Hong Yan
- Department of Electrical Engineering, City University of Hong Kong, Kowloon, Hong Kong
| | - Tanvir Alam
- College of Science and Engineering, Hamad Bin Khalifa University, Doha, Qatar
| |
Collapse
|
84
|
Dalkıran A, Atakan A, Rifaioğlu AS, Martin MJ, Atalay RÇ, Acar AC, Doğan T, Atalay V. Transfer learning for drug-target interaction prediction. Bioinformatics 2023; 39:i103-i110. [PMID: 37387156 DOI: 10.1093/bioinformatics/btad234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/19/2023] [Indexed: 07/01/2023] Open
Abstract
MOTIVATION Utilizing AI-driven approaches for drug-target interaction (DTI) prediction require large volumes of training data which are not available for the majority of target proteins. In this study, we investigate the use of deep transfer learning for the prediction of interactions between drug candidate compounds and understudied target proteins with scarce training data. The idea here is to first train a deep neural network classifier with a generalized source training dataset of large size and then to reuse this pre-trained neural network as an initial configuration for re-training/fine-tuning purposes with a small-sized specialized target training dataset. To explore this idea, we selected six protein families that have critical importance in biomedicine: kinases, G-protein-coupled receptors (GPCRs), ion channels, nuclear receptors, proteases, and transporters. In two independent experiments, the protein families of transporters and nuclear receptors were individually set as the target datasets, while the remaining five families were used as the source datasets. Several size-based target family training datasets were formed in a controlled manner to assess the benefit provided by the transfer learning approach. RESULTS Here, we present a systematic evaluation of our approach by pre-training a feed-forward neural network with source training datasets and applying different modes of transfer learning from the pre-trained source network to a target dataset. The performance of deep transfer learning is evaluated and compared with that of training the same deep neural network from scratch. We found that when the training dataset contains fewer than 100 compounds, transfer learning outperforms the conventional strategy of training the system from scratch, suggesting that transfer learning is advantageous for predicting binders to under-studied targets. AVAILABILITY AND IMPLEMENTATION The source code and datasets are available at https://github.com/cansyl/TransferLearning4DTI. Our web-based service containing the ready-to-use pre-trained models is accessible at https://tl4dti.kansil.org.
Collapse
Affiliation(s)
- Alperen Dalkıran
- Department of Computer Engineering, Middle East Technical University, Ankara 06800, Turkey
- Department of Computer Engineering, Adana Alparslan Türkeş Science and Technology University, Adana 01250, Turkey
| | - Ahmet Atakan
- Department of Computer Engineering, Middle East Technical University, Ankara 06800, Turkey
- Department of Computer Engineering, Erzincan Binali Yıldırım University, Erzincan 24002, Turkey
| | - Ahmet S Rifaioğlu
- Department of Computer Engineering, Iskenderun Technical University, Hatay 31200, Turkey
- Faculty of Medicine, Institute for Computational Biomedicine, Heidelberg University and Heidelberg University Hospital, Heidelberg 69120, Germany
| | - Maria J Martin
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, Hinxton CB10 1SD, United Kingdom
| | - Rengül Çetin Atalay
- Faculty of Pulmonary and Critical Care Medicine, the University of Chicago, Chicago, IL, 60637, United States
| | - Aybar C Acar
- Cancer Systems Biology Laboratory (Kansil), Middle East Technical University, Ankara 06800, Turkey
| | - Tunca Doğan
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, Hinxton CB10 1SD, United Kingdom
- Department of Computer Engineering, Hacettepe University, Ankara 06800, Turkey
| | - Volkan Atalay
- Department of Computer Engineering, Middle East Technical University, Ankara 06800, Turkey
| |
Collapse
|
85
|
Kamaraj C, Satish Kumar RC, Al-Ghanim KA, Nicoletti M, Sathiyamoorthy V, Sarvesh S, Ragavendran C, Govindarajan M. Novel Essential Oils Blend as a Repellent and Toxic Agent against Disease-Transmitting Mosquitoes. TOXICS 2023; 11:517. [PMID: 37368617 DOI: 10.3390/toxics11060517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/22/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023]
Abstract
Bio-insecticidal research has focused on long-term vector control using essential oils (EOs). This study examined the larvicidal, oviposition-deterrent, and repellent properties of five medicinal herb-based EO formulations (EOFs) on mosquitoes that are vectors of dengue, filariasis, and malaria. EOFs were significantly more toxic to the larvae and pupae of Culex quinquefasciatus, Anopheles stephensi, and Aedes aegypti with LC50 = 9.23, 12.85, and 14.46 ppm, as well with 10.22, 11.39, and 12.81 ppm, with oviposition active indexes of -0.84, -0.95, and -0.92, respectively. Oviposition-deterrent repellence was found in 91.39%, 94.83%, and 96.09%. EOs and N, N-Diethyl-3-methylbenzamide (DEET) were prepared at various concentrations for time duration repellent bioassays (6.25-100 ppm). Ae. aegypti, An. stephensi, and Cx. quinquefasciatus were monitored for 300, 270, and 180 min, respectively. At 100 ppm, EOs and DEET had comparable repellence in terms of test durations. EOF's primary components d-limonene (12.9%), 2,6-octadienal, 3,7-dimethyl, (Z) (12.2%), acetic acid, phenylmethyl ester (19.6%), verbenol (7.6%), and benzyl benzoate (17.4%) may be combined to make a mosquito larvicidal and repellant equivalent to synthetic repellent lotions. In the molecular dynamics simulations, limonene (-6.1 kcal/mol) and benzyl benzoate (-7.5 kcal/mol) had a positive chemical association with DEET (-6.3 kcal/mol) and interacted with the OBP binding pocket with high affinity and stability. This research will help local herbal product manufacturers and the cosmetics industry in developing 100% herbal insect repellent products to combat mosquito-borne diseases, including dengue, malaria, and filariasis.
Collapse
Affiliation(s)
- Chinnaperumal Kamaraj
- Interdisciplinary Institute of Indian System of Medicine (IIISM), Directorate of Research, SRM Institute Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Rajappan Chandra Satish Kumar
- Interdisciplinary Institute of Indian System of Medicine (IIISM), Directorate of Research, SRM Institute Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Khalid A Al-Ghanim
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Marcello Nicoletti
- Department of Environmental Biology, Foundation in Unam Sapientiam, Sapienza University of Rome, 00185 Rome, Italy
| | - V Sathiyamoorthy
- Ayurvedic Manufacturing, Kancheepuram 631 501, Tamil Nadu, India
| | - Sabarathinam Sarvesh
- Interdisciplinary Institute of Indian System of Medicine (IIISM), Directorate of Research, SRM Institute Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Chinnasamy Ragavendran
- Department of Conservative Dentistry and Endodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 600 077, Tamil Nadu, India
| | - Marimuthu Govindarajan
- Unit of Vector Control, Phytochemistry and Nanotechnology, Department of Zoology, Annamalai University, Annamalainagar 608 002, Tamil Nadu, India
- Unit of Natural Products and Nanotechnology, Department of Zoology, Government College for Women (Autonomous), Kumbakonam 612 001, Tamil Nadu, India
| |
Collapse
|
86
|
Malachowska B, Yang WL, Qualman A, Muro I, Boe DM, Lampe JN, Kovacs EJ, Idrovo JP. Transcriptomics, metabolomics, and in-silico drug predictions for liver damage in young and aged burn victims. Commun Biol 2023; 6:597. [PMID: 37268765 DOI: 10.1038/s42003-023-04964-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 05/22/2023] [Indexed: 06/04/2023] Open
Abstract
Burn induces a systemic response affecting multiple organs, including the liver. Since the liver plays a critical role in metabolic, inflammatory, and immune events, a patient with impaired liver often exhibits poor outcomes. The mortality rate after burns in the elderly population is higher than in any other age group, and studies show that the liver of aged animals is more susceptible to injury after burns. Understanding the aged-specific liver response to burns is fundamental to improving health care. Furthermore, no liver-specific therapy exists to treat burn-induced liver damage highlighting a critical gap in burn injury therapeutics. In this study, we analyzed transcriptomics and metabolomics data from the liver of young and aged mice to identify mechanistic pathways and in-silico predict therapeutic targets to prevent or reverse burn-induced liver damage. Our study highlights pathway interactions and master regulators that underlie the differential liver response to burn injury in young and aged animals.
Collapse
Affiliation(s)
- Beata Malachowska
- Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Weng-Lang Yang
- Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Andrea Qualman
- Department of Surgery; Division of G.I., Trauma, and Endocrine Surgery, University of Colorado, Aurora, CO, 80045, USA
| | - Israel Muro
- Department of Surgery; Division of G.I., Trauma, and Endocrine Surgery, University of Colorado, Aurora, CO, 80045, USA
| | - Devin M Boe
- Department of Surgery; Division of G.I., Trauma, and Endocrine Surgery, University of Colorado, Aurora, CO, 80045, USA
- Graduate Program in Immunology, University of Colorado, Aurora, CO, 80045, USA
| | - Jed N Lampe
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Colorado, Aurora, CO, 80045, USA
| | - Elizabeth J Kovacs
- Department of Surgery; Division of G.I., Trauma, and Endocrine Surgery, University of Colorado, Aurora, CO, 80045, USA
- Graduate Program in Immunology, University of Colorado, Aurora, CO, 80045, USA
- Molecular Biology Program, University of Colorado, Aurora, CO, 80045, USA
| | - Juan-Pablo Idrovo
- Department of Surgery; Division of G.I., Trauma, and Endocrine Surgery, University of Colorado, Aurora, CO, 80045, USA.
| |
Collapse
|
87
|
Liu S, Qin HH, Ji XR, Gan JW, Sun MJ, Tao J, Tao ZQ, Zhao GN, Ma BX. Virtual Screening of Nrf2 Dietary-Derived Agonists and Safety by a New Deep-Learning Model and Verified In Vitro and In Vivo. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:8038-8049. [PMID: 37196215 DOI: 10.1021/acs.jafc.3c00867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is an essential regulatory target of antioxidants, but the lack of Nrf2 active site information has hindered discovery of new Nrf2 agonists from food-derived compounds by large-scale virtual screening. Two deep-learning models were separately trained to screen for Nrf2-agonists and safety. The trained models screened potentially active chemicals from approximately 70,000 dietary compounds within 5 min. Of the 169 potential Nrf2 agonists identified via deep-learning screening, 137 had not been reported before. Six compounds selected from the new Nrf2 agonists significantly increased (p < 0.05) the activity of Nrf2 on carbon tetrachloride (CCl4)-intoxicated HepG2 cells (nicotiflorin (99.44 ± 18.5%), artemetin (97.91 ± 8.22%), daidzin (87.73 ± 3.77%), linonin (74.27 ± 5.73%), sinensetin (72.74 ± 10.41%), and tectoridin (77.78 ± 4.80%)), and their safety were demonstrated by an MTT assay. The safety and Nrf2 agonistic activity of nicotiflorin, artemetin, and daidzin were also reconfirm by a single-dose acute oral toxicity study and CCl4-intoxicated rat assay.
Collapse
Affiliation(s)
- Song Liu
- Institute of Pharmaceutical Process, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Huan-Huan Qin
- Institute of Pharmaceutical Process, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Xin-Ran Ji
- Institute of Pharmaceutical Process, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Jian-Wen Gan
- Institute of Pharmaceutical Process, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Meng-Jia Sun
- Institute of Pharmaceutical Process, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Jin Tao
- Institute of Pharmaceutical Process, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Zhuo-Qi Tao
- Institute of Pharmaceutical Process, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Guang-Nian Zhao
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Bing-Xin Ma
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
88
|
Zhao M, Yuan M, Yang Y, Xu SX. CPGL: Prediction of Compound-Protein Interaction by Integrating Graph Attention Network With Long Short-Term Memory Neural Network. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:1935-1942. [PMID: 36445995 DOI: 10.1109/tcbb.2022.3225296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Recent advancements of artificial intelligence based on deep learning algorithms have made it possible to computationally predict compound-protein interaction (CPI) without conducting laboratory experiments. In this manuscript, we integrated a graph attention network (GAT) for compounds and a long short-term memory neural network (LSTM) for proteins, used end-to-end representation learning for both compounds and proteins, and proposed a deep learning algorithm, CPGL (CPI with GAT and LSTM) to optimize the feature extraction from compounds and proteins and to improve the model robustness and generalizability. CPGL demonstrated an excellent predictive performance and outperforms recently reported deep learning models. Based on 3 public CPI datasets, C.elegans, Human and BindingDB, CPGL represented 1 - 5% improvement compared to existing deep-learning models. Our method also achieves excellent results on datasets with imbalanced positive and negative proportions constructed based on the C.elegans and Human datasets. More importantly, using 2 label reversal datasets, GPCR and Kinase, CPGL showed superior performance compared to other existing deep learning models. The AUC were substantially improved by 20% on the Kinase dataset, indicative of the robustness and generalizability of CPGL.
Collapse
|
89
|
Boswell Z, Verga JU, Mackle J, Guerrero-Vazquez K, Thomas OP, Cray J, Wolf BJ, Choo YM, Croot P, Hamann MT, Hardiman G. In-Silico Approaches for the Screening and Discovery of Broad-Spectrum Marine Natural Product Antiviral Agents Against Coronaviruses. Infect Drug Resist 2023; 16:2321-2338. [PMID: 37155475 PMCID: PMC10122865 DOI: 10.2147/idr.s395203] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 03/16/2023] [Indexed: 05/10/2023] Open
Abstract
The urgent need for SARS-CoV-2 controls has led to a reassessment of approaches to identify and develop natural product inhibitors of zoonotic, highly virulent, and rapidly emerging viruses. There are yet no clinically approved broad-spectrum antivirals available for beta-coronaviruses. Discovery pipelines for pan-virus medications against a broad range of betacoronaviruses are therefore a priority. A variety of marine natural product (MNP) small molecules have shown inhibitory activity against viral species. Access to large data caches of small molecule structural information is vital to finding new pharmaceuticals. Increasingly, molecular docking simulations are being used to narrow the space of possibilities and generate drug leads. Combining in-silico methods, augmented by metaheuristic optimization and machine learning (ML) allows the generation of hits from within a virtual MNP library to narrow screens for novel targets against coronaviruses. In this review article, we explore current insights and techniques that can be leveraged to generate broad-spectrum antivirals against betacoronaviruses using in-silico optimization and ML. ML approaches are capable of simultaneously evaluating different features for predicting inhibitory activity. Many also provide a semi-quantitative measure of feature relevance and can guide in selecting a subset of features relevant for inhibition of SARS-CoV-2.
Collapse
Affiliation(s)
- Zachary Boswell
- School of Biological Sciences and Institute for Global Security, Queen's University, Belfast, Northern Ireland, UK
| | - Jacopo Umberto Verga
- School of Biological Sciences and Institute for Global Security, Queen's University, Belfast, Northern Ireland, UK
- Genomic Data Science, University of Galway, Galway, Ireland
| | - James Mackle
- School of Biological Sciences and Institute for Global Security, Queen's University, Belfast, Northern Ireland, UK
| | | | - Olivier P Thomas
- School of Biological and Chemical Sciences, Ryan Institute, University of Galway, Galway, H91TK33Ireland
| | - James Cray
- Department of Biomedical Education and Anatomy, College of Medicine and Division of Biosciences, College of Dentistry, Ohio State University, Columbus, OH, USA
| | - Bethany J Wolf
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Yeun-Mun Choo
- Department of Chemistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Peter Croot
- Irish Centre for Research in Applied Geoscience, Earth and Ocean Sciences and Ryan Institute, School of Natural Sciences, University of Galway, Galway, Ireland
| | - Mark T Hamann
- Departments of Drug Discovery and Biomedical Sciences and Public Health, Colleges of Pharmacy and Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Gary Hardiman
- School of Biological Sciences and Institute for Global Security, Queen's University, Belfast, Northern Ireland, UK
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
- Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
90
|
Tran TTV, Tayara H, Chong KT. Artificial Intelligence in Drug Metabolism and Excretion Prediction: Recent Advances, Challenges, and Future Perspectives. Pharmaceutics 2023; 15:1260. [PMID: 37111744 PMCID: PMC10143484 DOI: 10.3390/pharmaceutics15041260] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/07/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Drug metabolism and excretion play crucial roles in determining the efficacy and safety of drug candidates, and predicting these processes is an essential part of drug discovery and development. In recent years, artificial intelligence (AI) has emerged as a powerful tool for predicting drug metabolism and excretion, offering the potential to speed up drug development and improve clinical success rates. This review highlights recent advances in AI-based drug metabolism and excretion prediction, including deep learning and machine learning algorithms. We provide a list of public data sources and free prediction tools for the research community. We also discuss the challenges associated with the development of AI models for drug metabolism and excretion prediction and explore future perspectives in the field. We hope this will be a helpful resource for anyone who is researching in silico drug metabolism, excretion, and pharmacokinetic properties.
Collapse
Affiliation(s)
- Thi Tuyet Van Tran
- Department of Electronics and Information Engineering, Jeonbuk National University, Jeonju 54896, Republic of Korea;
- Faculty of Information Technology, An Giang University, Long Xuyen 880000, Vietnam
- Vietnam National University—Ho Chi Minh City, Ho Chi Minh 700000, Vietnam
| | - Hilal Tayara
- School of International Engineering and Science, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Kil To Chong
- Advances Electronics and Information Research Center, Jeonbuk National University, Jeonju 54896, Republic of Korea
| |
Collapse
|
91
|
Belchor MN, Costa CRDC, Roggero A, Moraes LLF, Samelo R, Annunciato I, de Oliveira MA, Sousa SF, Toyama MH. In Silico Evaluation of Quercetin Methylated Derivatives on the Interaction with Secretory Phospholipases A2 from Crotalus durissus terrificus and Bothrops jararacussu. Pharmaceuticals (Basel) 2023; 16:ph16040597. [PMID: 37111354 PMCID: PMC10143728 DOI: 10.3390/ph16040597] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/07/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Quercetin derivatives have already shown their anti-inflammatory potential, inhibiting essential enzymes involved in this process. Among diverse pro-inflammatory toxins from snake venoms, phospholipase A2 is one of the most abundant in some species, such as Crotalus durissus terrificus and Bothrops jararacussu from the Viperidae family. These enzymes can induce the inflammatory process through hydrolysis at the sn-2 position of glycerophospholipids. Hence, elucidating the main residues involved in the biological effects of these macromolecules can help to identify potential compounds with inhibitory activity. In silico tools were used in this study to evaluate the potential of quercetin methylated derivatives in the inhibition of bothropstoxin I (BthTX-I) and II (BthTX-II) from Bothrops jararacussu and phospholipase A2 from Crotalus durissus terrificus. The use of a transitional analogous and two classical inhibitors of phospholipase A2 guided this work to find the role of residues involved in the phospholipid anchoring and the subsequent development of the inflammatory process. First, main cavities were studied, revealing the best regions to be inhibited by a compound. Focusing on these regions, molecular docking assays were made to show main interactions between each compound. Results reveal that analogue and inhibitors, Varespladib (Var) and p-bromophenacyl bromide (BPB), guided quercetins derivatives analysis, revealing that Leu2, Phe5, Tyr28, glycine in the calcium-binding loop, His48, Asp49 of BthTX-II and Cdtspla2 were the main residues to be inhibited. 3MQ exhibited great interaction with the active site, similar to Var results, while Q anchored better in the BthTX-II active site. However, strong interactions in the C-terminal region, highlighting His120, seem to be crucial to decreasing contacts with phospholipid and BthTX-II. Hence, quercetin derivatives anchor differently with each toxin and further in vitro and in vivo studies are essential to elucidate these data.
Collapse
Affiliation(s)
- Mariana Novo Belchor
- Center of Natural and Human Sciences, Federal University of ABC (UFABC), Santo André 09210-580, SP, Brazil
- Biosciences Institute of Paulista Coast Campus (IB/CLP), University of São Paulo State (UNESP), São Vicente 11330-900, SP, Brazil
| | - Caroline Ramos da Cruz Costa
- Center of Natural and Human Sciences, Federal University of ABC (UFABC), Santo André 09210-580, SP, Brazil
- Biosciences Institute of Paulista Coast Campus (IB/CLP), University of São Paulo State (UNESP), São Vicente 11330-900, SP, Brazil
| | - Airam Roggero
- Biosciences Institute of Paulista Coast Campus (IB/CLP), University of São Paulo State (UNESP), São Vicente 11330-900, SP, Brazil
| | - Laila L F Moraes
- Biosciences Institute of Paulista Coast Campus (IB/CLP), University of São Paulo State (UNESP), São Vicente 11330-900, SP, Brazil
| | - Ricardo Samelo
- Biosciences Institute of Paulista Coast Campus (IB/CLP), University of São Paulo State (UNESP), São Vicente 11330-900, SP, Brazil
| | - Isabelly Annunciato
- Biosciences Institute of Paulista Coast Campus (IB/CLP), University of São Paulo State (UNESP), São Vicente 11330-900, SP, Brazil
| | - Marcos Antonio de Oliveira
- Center of Natural and Human Sciences, Federal University of ABC (UFABC), Santo André 09210-580, SP, Brazil
- Biosciences Institute of Paulista Coast Campus (IB/CLP), University of São Paulo State (UNESP), São Vicente 11330-900, SP, Brazil
| | - Sergio F Sousa
- Unit of Applied Biomolecular Sciences (UCIBIO), REQUIMTE-BioSIM-Medicine Faculty, Porto University, 4050-345 Porto, Portugal
| | - Marcos Hikari Toyama
- Center of Natural and Human Sciences, Federal University of ABC (UFABC), Santo André 09210-580, SP, Brazil
- Biosciences Institute of Paulista Coast Campus (IB/CLP), University of São Paulo State (UNESP), São Vicente 11330-900, SP, Brazil
| |
Collapse
|
92
|
Khusial R, Bies RR, Akil A. Deep Learning Methods Applied to Drug Concentration Prediction of Olanzapine. Pharmaceutics 2023; 15:pharmaceutics15041139. [PMID: 37111625 PMCID: PMC10145228 DOI: 10.3390/pharmaceutics15041139] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/17/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
Pharmacometrics and the utilization of population pharmacokinetics play an integral role in model-informed drug discovery and development (MIDD). Recently, there has been a growth in the application of deep learning approaches to aid in areas within MIDD. In this study, a deep learning model, LSTM-ANN, was developed to predict olanzapine drug concentrations from the CATIE study. A total of 1527 olanzapine drug concentrations from 523 individuals along with 11 patient-specific covariates were used in model development. The hyperparameters of the LSTM-ANN model were optimized through a Bayesian optimization algorithm. A population pharmacokinetic model using the NONMEM model was constructed as a reference to compare to the performance of the LSTM-ANN model. The RMSE of the LSTM-ANN model was 29.566 in the validation set, while the RMSE of the NONMEM model was 31.129. Permutation importance revealed that age, sex, and smoking were highly influential covariates in the LSTM-ANN model. The LSTM-ANN model showed potential in the application of drug concentration predictions as it was able to capture the relationships within a sparsely sampled pharmacokinetic dataset and perform comparably to the NONMEM model.
Collapse
Affiliation(s)
- Richard Khusial
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Robert R. Bies
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14214, USA
- Institute for Artificial Intelligence and Data Science, University at Buffalo, Buffalo, NY 14260, USA
| | - Ayman Akil
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| |
Collapse
|
93
|
Yang X, Wang S, Qi L, Chen S, Du K, Shang Y, Guo J, Fang S, Li J, Zhang H, Chang Y. An efficient method for qualitation and quantitation of multi-components of the herbal medicine Qingjin Yiqi Granules. J Pharm Biomed Anal 2023; 227:115288. [PMID: 36796275 DOI: 10.1016/j.jpba.2023.115288] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/08/2023] [Accepted: 02/08/2023] [Indexed: 02/13/2023]
Abstract
Qingjin Yiqi Granules (QJYQ) is a Traditional Chinese Medicines (TCMs) prescription for the patients with post-COVID-19 condition. It is essential to carry out the quality evaluation of QJYQ. A comprehensive investigation was conducted by establishing deep-learning assisted mass defect filter (deep-learning MDF) mode for qualitative analysis, ultra-high performance liquid chromatography and scheduled multiple reaction monitoring method (UHPLC-sMRM) for precise quantitation to evaluate the quality of QJYQ. Firstly, a deep-learning MDF was used to classify and characterize the whole phytochemical components of QJYQ based on the mass spectrum (MS) data of ultra-high performance liquid chromatography quadrupole time of flight tandem mass spectrometry (UHPLC-Q-TOF/MS). Secondly, the highly sensitive UHPLC-sMRM data-acquisition method was established to quantify the multi-ingredients of QJYQ. Totally, nine major types of phytochemical compounds in QJYQ were intelligently classified and 163 phytochemicals were initially identified. Furthermore, fifty components were rapidly quantified. The comprehensive evaluation strategy established in this study would provide an effective tool for accurately evaluating the quality of QJYQ as a whole.
Collapse
Affiliation(s)
- Xiaohua Yang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuangqi Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lina Qi
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shujing Chen
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Kunze Du
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ye Shang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jiading Guo
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shiming Fang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jin Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Han Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China.
| | - Yanxu Chang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Phytochemistry and Pharmaceutical Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
94
|
Abbasi Mesrabadi H, Faez K, Pirgazi J. Drug-target interaction prediction based on protein features, using wrapper feature selection. Sci Rep 2023; 13:3594. [PMID: 36869062 PMCID: PMC9984486 DOI: 10.1038/s41598-023-30026-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 02/14/2023] [Indexed: 03/05/2023] Open
Abstract
Drug-target interaction prediction is a vital stage in drug development, involving lots of methods. Experimental methods that identify these relationships on the basis of clinical remedies are time-taking, costly, laborious, and complex introducing a lot of challenges. One group of new methods is called computational methods. The development of new computational methods which are more accurate can be preferable to experimental methods, in terms of total cost and time. In this paper, a new computational model to predict drug-target interaction (DTI), consisting of three phases, including feature extraction, feature selection, and classification is proposed. In feature extraction phase, different features such as EAAC, PSSM and etc. would be extracted from sequence of proteins and fingerprint features from drugs. These extracted features would then be combined. In the next step, one of the wrapper feature selection methods named IWSSR, due to the large amount of extracted data, is applied. The selected features are then given to rotation forest classification, to have a more efficient prediction. Actually, the innovation of our work is that we extract different features; and then select features by the use of IWSSR. The accuracy of the rotation forest classifier based on tenfold on the golden standard datasets (enzyme, ion channels, G-protein-coupled receptors, nuclear receptors) is as follows: 98.12, 98.07, 96.82, and 95.64. The results of experiments indicate that the proposed model has an acceptable rate in DTI prediction and is compatible with the proposed methods in other papers.
Collapse
Affiliation(s)
- Hengame Abbasi Mesrabadi
- Faculty of Computer and Information Technology Engineering, Qazvin Branch, Islamic Azad University, Qazvin, Iran
| | - Karim Faez
- Department of Electrical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.
| | - Jamshid Pirgazi
- Department of Computer Engineering, University of Science and Technology of Mazandaran, Behshahr, Iran
| |
Collapse
|
95
|
Zhao Q, Duan G, Yang M, Cheng Z, Li Y, Wang J. AttentionDTA: Drug-Target Binding Affinity Prediction by Sequence-Based Deep Learning With Attention Mechanism. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:852-863. [PMID: 35471889 DOI: 10.1109/tcbb.2022.3170365] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The identification of drug-target relations (DTRs) is substantial in drug development. A large number of methods treat DTRs as drug-target interactions (DTIs), a binary classification problem. The main drawback of these methods are the lack of reliable negative samples and the absence of many important aspects of DTR, including their dose dependence and quantitative affinities. With increasing number of publications of drug-protein binding affinity data recently, DTRs prediction can be viewed as a regression problem of drug-target affinities (DTAs) which reflects how tightly the drug binds to the target and can present more detailed and specific information than DTIs. The growth of affinity data enables the use of deep learning architectures, which have been shown to be among the state-of-the-art methods in binding affinity prediction. Although relatively effective, due to the black-box nature of deep learning, these models are less biologically interpretable. In this study, we proposed a deep learning-based model, named AttentionDTA, which uses attention mechanism to predict DTAs. Different from the models using 3D structures of drug-target complexes or graph representation of drugs and proteins, the novelty of our work is to use attention mechanism to focus on key subsequences which are important in drug and protein sequences when predicting its affinity. We use two separate one-dimensional Convolution Neural Networks (1D-CNNs) to extract the semantic information of drug's SMILES string and protein's amino acid sequence. Furthermore, a two-side multi-head attention mechanism is developed and embedded to our model to explore the relationship between drug features and protein features. We evaluate our model on three established DTA benchmark datasets, Davis, Metz, and KIBA. AttentionDTA outperforms the state-of-the-art deep learning methods under different evaluation metrics. The results show that the attention-based model can effectively extract protein features related to drug information and drug features related to protein information to better predict drug target affinities. It is worth mentioning that we test our model on IC50 dataset, which provides the binding sites between drugs and proteins, to evaluate the ability of our model to locate binding sites. Finally, we visualize the attention weight to demonstrate the biological significance of the model. The source code of AttentionDTA can be downloaded from https://github.com/zhaoqichang/AttentionDTA_TCBB.
Collapse
|
96
|
Odhar HA, Hashim AF, Ahjel SW, Humadi SS. Molecular docking and dynamics simulation analysis of the human FXIIa with compounds from the Mcule database. Bioinformation 2023; 19:160-166. [PMID: 37814681 PMCID: PMC10560304 DOI: 10.6026/97320630019160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/28/2023] [Accepted: 02/28/2023] [Indexed: 09/01/2023] Open
Abstract
The human factor XIIa is a serine protease enzyme that is implicated in the pathological thrombosis. This coagulation factor represents an interesting molecular target to design safer antithrombotic agents without adversely influencing physiological hemostasis. Therefore, it is of interest to virtually screen the human factor XIIa crystal with millions of compounds in Mcule database in order to identify potential inhibitors. For this purpose, both molecular docking and dynamics simulation were employed to identify potential hits. Also, various predictive approaches were utilized to estimate chemical, pharmacokinetics and toxicological features for the top hits. As such, we report here that compound 4 (1-(4-benzylpiperazin-1-yl)-2-[5-(3,5-dimethylpyrazol-1-yl)-1,2,3, 4-tetrazol-2-yl]ethanone) may be a potential ligand against the human factor XIIa for further consideration in the design and development of novel antithrombotic agents.
Collapse
Affiliation(s)
| | | | | | - Suhad Sami Humadi
- Department of pharmacy, Al-Zahrawi University College, Karbala, Iraq
| |
Collapse
|
97
|
Comparative Studies on Resampling Techniques in Machine Learning and Deep Learning Models for Drug-Target Interaction Prediction. Molecules 2023; 28:molecules28041663. [PMID: 36838652 PMCID: PMC9964614 DOI: 10.3390/molecules28041663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 02/12/2023] Open
Abstract
The prediction of drug-target interactions (DTIs) is a vital step in drug discovery. The success of machine learning and deep learning methods in accurately predicting DTIs plays a huge role in drug discovery. However, when dealing with learning algorithms, the datasets used are usually highly dimensional and extremely imbalanced. To solve this issue, the dataset must be resampled accordingly. In this paper, we have compared several data resampling techniques to overcome class imbalance in machine learning methods as well as to study the effectiveness of deep learning methods in overcoming class imbalance in DTI prediction in terms of binary classification using ten (10) cancer-related activity classes from BindingDB. It is found that the use of Random Undersampling (RUS) in predicting DTIs severely affects the performance of a model, especially when the dataset is highly imbalanced, thus, rendering RUS unreliable. It is also found that SVM-SMOTE can be used as a go-to resampling method when paired with the Random Forest and Gaussian Naïve Bayes classifiers, whereby a high F1 score is recorded for all activity classes that are severely and moderately imbalanced. Additionally, the deep learning method called Multilayer Perceptron recorded high F1 scores for all activity classes even when no resampling method was applied.
Collapse
|
98
|
Atas Guvenilir H, Doğan T. How to approach machine learning-based prediction of drug/compound-target interactions. J Cheminform 2023; 15:16. [PMID: 36747300 PMCID: PMC9901167 DOI: 10.1186/s13321-023-00689-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 01/30/2023] [Indexed: 02/08/2023] Open
Abstract
The identification of drug/compound-target interactions (DTIs) constitutes the basis of drug discovery, for which computational predictive approaches have been developed. As a relatively new data-driven paradigm, proteochemometric (PCM) modeling utilizes both protein and compound properties as a pair at the input level and processes them via statistical/machine learning. The representation of input samples (i.e., proteins and their ligands) in the form of quantitative feature vectors is crucial for the extraction of interaction-related properties during the artificial learning and subsequent prediction of DTIs. Lately, the representation learning approach, in which input samples are automatically featurized via training and applying a machine/deep learning model, has been utilized in biomedical sciences. In this study, we performed a comprehensive investigation of different computational approaches/techniques for protein featurization (including both conventional approaches and the novel learned embeddings), data preparation and exploration, machine learning-based modeling, and performance evaluation with the aim of achieving better data representations and more successful learning in DTI prediction. For this, we first constructed realistic and challenging benchmark datasets on small, medium, and large scales to be used as reliable gold standards for specific DTI modeling tasks. We developed and applied a network analysis-based splitting strategy to divide datasets into structurally different training and test folds. Using these datasets together with various featurization methods, we trained and tested DTI prediction models and evaluated their performance from different angles. Our main findings can be summarized under 3 items: (i) random splitting of datasets into train and test folds leads to near-complete data memorization and produce highly over-optimistic results, as a result, should be avoided, (ii) learned protein sequence embeddings work well in DTI prediction and offer high potential, despite interaction-related properties (e.g., structures) of proteins are unused during their self-supervised model training, and (iii) during the learning process, PCM models tend to rely heavily on compound features while partially ignoring protein features, primarily due to the inherent bias in DTI data, indicating the requirement for new and unbiased datasets. We hope this study will aid researchers in designing robust and high-performing data-driven DTI prediction systems that have real-world translational value in drug discovery.
Collapse
Affiliation(s)
- Heval Atas Guvenilir
- Biological Data Science Laboratory, Department of Computer Engineering, Hacettepe University, Ankara, Turkey
- Department of Health Informatics, Graduate School of Informatics, METU, Ankara, Turkey
| | - Tunca Doğan
- Biological Data Science Laboratory, Department of Computer Engineering, Hacettepe University, Ankara, Turkey.
- Institute of Informatics, Hacettepe University, Ankara, Turkey.
- Department of Bioinformatics, Graduate School of Health Sciences, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
99
|
Hu L, Fu C, Ren Z, Cai Y, Yang J, Xu S, Xu W, Tang D. SSELM-neg: spherical search-based extreme learning machine for drug-target interaction prediction. BMC Bioinformatics 2023; 24:38. [PMID: 36737694 PMCID: PMC9896467 DOI: 10.1186/s12859-023-05153-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The experimental verification of a drug discovery process is expensive and time-consuming. Therefore, efficiently and effectively identifying drug-target interactions (DTIs) has been the focus of research. At present, many machine learning algorithms are used for predicting DTIs. The key idea is to train the classifier using an existing DTI to predict a new or unknown DTI. However, there are various challenges, such as class imbalance and the parameter optimization of many classifiers, that need to be solved before an optimal DTI model is developed. METHODS In this study, we propose a framework called SSELM-neg for DTI prediction, in which we use a screening approach to choose high-quality negative samples and a spherical search approach to optimize the parameters of the extreme learning machine. RESULTS The results demonstrated that the proposed technique outperformed other state-of-the-art methods in 10-fold cross-validation experiments in terms of the area under the receiver operating characteristic curve (0.986, 0.993, 0.988, and 0.969) and AUPR (0.982, 0.991, 0.982, and 0.946) for the enzyme dataset, G-protein coupled receptor dataset, ion channel dataset, and nuclear receptor dataset, respectively. CONCLUSION The screening approach produced high-quality negative samples with the same number of positive samples, which solved the class imbalance problem. We optimized an extreme learning machine using a spherical search approach to identify DTIs. Therefore, our models performed better than other state-of-the-art methods.
Collapse
Affiliation(s)
- Lingzhi Hu
- grid.411847.f0000 0004 1804 4300School of Medical Information Engineering, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| | - Chengzhou Fu
- grid.411847.f0000 0004 1804 4300School of Medical Information Engineering, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China ,Guangdong Province Precise Medicine Big Data of Traditional Chinese Medicine Engineering Technology Research Center, Guangzhou, People’s Republic of China
| | - Zhonglu Ren
- grid.411847.f0000 0004 1804 4300School of Medical Information Engineering, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| | - Yongming Cai
- grid.411847.f0000 0004 1804 4300School of Medical Information Engineering, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China ,Guangdong Province Precise Medicine Big Data of Traditional Chinese Medicine Engineering Technology Research Center, Guangzhou, People’s Republic of China
| | - Jin Yang
- grid.411847.f0000 0004 1804 4300School of Medical Information Engineering, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China ,Guangdong Province Precise Medicine Big Data of Traditional Chinese Medicine Engineering Technology Research Center, Guangzhou, People’s Republic of China
| | - Siwen Xu
- grid.411847.f0000 0004 1804 4300School of Medical Information Engineering, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| | - Wenhua Xu
- grid.411847.f0000 0004 1804 4300School of Medical Information Engineering, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| | - Deyu Tang
- grid.411847.f0000 0004 1804 4300School of Medical Information Engineering, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China ,grid.79703.3a0000 0004 1764 3838School of Computer Science and Engineering, South China University of Technology, Guangzhou, People’s Republic of China ,Guangdong Province Precise Medicine Big Data of Traditional Chinese Medicine Engineering Technology Research Center, Guangzhou, People’s Republic of China
| |
Collapse
|
100
|
Kumar M, Nguyen TPN, Kaur J, Singh TG, Soni D, Singh R, Kumar P. Opportunities and challenges in application of artificial intelligence in pharmacology. Pharmacol Rep 2023; 75:3-18. [PMID: 36624355 PMCID: PMC9838466 DOI: 10.1007/s43440-022-00445-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/23/2022] [Accepted: 12/25/2022] [Indexed: 01/11/2023]
Abstract
Artificial intelligence (AI) is a machine science that can mimic human behaviour like intelligent analysis of data. AI functions with specialized algorithms and integrates with deep and machine learning. Living in the digital world can generate a huge amount of medical data every day. Therefore, we need an automated and reliable evaluation tool that can make decisions more accurately and faster. Machine learning has the potential to learn, understand and analyse the data used in healthcare systems. In the last few years, AI is known to be employed in various fields in pharmaceutical science especially in pharmacological research. It helps in the analysis of preclinical (laboratory animals) and clinical (in human) trial data. AI also plays important role in various processes such as drug discovery/manufacturing, diagnosis of big data for disease identification, personalized treatment, clinical trial research, radiotherapy, surgical robotics, smart electronic health records, and epidemic outbreak prediction. Moreover, AI has been used in the evaluation of biomarkers and diseases. In this review, we explain various models and general processes of machine learning and their role in pharmacological science. Therefore, AI with deep learning and machine learning could be relevant in pharmacological research.
Collapse
Affiliation(s)
- Mandeep Kumar
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
| | - T P Nhung Nguyen
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
- Department of Pharmacy, Da Nang University of Medical Technology and Pharmacy, Da Nang, Vietnam
| | - Jasleen Kaur
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Lucknow, Uttar Pradesh, 226002, India
| | | | - Divya Soni
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Randhir Singh
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India.
| |
Collapse
|