1
|
Wu J, Han M, Tan X, Zeng L, Yang Z, Zhong H, Jiang X, Yao S, Liu W, Li W, Liu X, Wu W. Green synthesis of neuroprotective spirocyclic chalcone derivatives and their role in protecting against traumatic optic nerve injury. Eur J Med Chem 2024; 280:116933. [PMID: 39368262 DOI: 10.1016/j.ejmech.2024.116933] [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: 06/15/2024] [Revised: 08/30/2024] [Accepted: 10/01/2024] [Indexed: 10/07/2024]
Abstract
For clinically prevalent traumatic optic neuropathy (TON) and other retinal and optic nerve injuries lacking effective therapeutic agents, there is an urgent clinical demand for developing highly efficient and safe neuroprotective agents. Here, we have integrated naturally sourced chalcone with isatin through a catalyst-free green synthesis method, reporting a series of spirocyclic chalcone derivatives with significantly lower cytotoxicity than chalcone itself. Following in vitro cell protection assays in models of hydrogen peroxide and glutamic acid-induced damage, multiple active compounds capable of combating both forms of damage were identified. Among these, candidate compound X38 demonstrated promising neuroprotective prospects: in vitro, it attenuated glutamate-induced cell apoptosis, while in vivo, it effectively ameliorated retinal thinning and loss of optic nerve electrophysiological function induced by optic nerve injury. Preliminary mechanistic studies suggest that X38 exerts its neuroprotective effects by mitigating intracellular ROS accumulation, inhibiting JNK phosphorylation, and alleviating oxidative stress. Additionally, acute toxicity studies (intraperitoneal injection, 500 mg/kg) underscored the favorable in vivo safety profile of X38. Taken together, this study has designed a class of safe, neuroprotective spirocyclic chalcone derivatives that can be synthesized using green methods, offering an attractive candidate for treating retinal and optic nerve injuries.
Collapse
Affiliation(s)
- Jianzhang Wu
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325027, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine Vision and Brain Health), Wenzhou, Zhejiang, 325000, China.
| | - Meiting Han
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine Vision and Brain Health), Wenzhou, Zhejiang, 325000, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xiangpeng Tan
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine Vision and Brain Health), Wenzhou, Zhejiang, 325000, China
| | - Ling Zeng
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhenzhen Yang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Hongliang Zhong
- The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xiaohui Jiang
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Shuang Yao
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Weibin Liu
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325027, China
| | - Wulan Li
- The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xin Liu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine Vision and Brain Health), Wenzhou, Zhejiang, 325000, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China.
| | - Wencan Wu
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325027, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine Vision and Brain Health), Wenzhou, Zhejiang, 325000, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China.
| |
Collapse
|
2
|
Odugbemi AI, Nyirenda C, Christoffels A, Egieyeh SA. Artificial intelligence in antidiabetic drug discovery: The advances in QSAR and the prediction of α-glucosidase inhibitors. Comput Struct Biotechnol J 2024; 23:2964-2977. [PMID: 39148608 PMCID: PMC11326494 DOI: 10.1016/j.csbj.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 08/17/2024] Open
Abstract
Artificial Intelligence is transforming drug discovery, particularly in the hit identification phase of therapeutic compounds. One tool that has been instrumental in this transformation is Quantitative Structure-Activity Relationship (QSAR) analysis. This computer-aided drug design tool uses machine learning to predict the biological activity of new compounds based on the numerical representation of chemical structures against various biological targets. With diabetes mellitus becoming a significant health challenge in recent times, there is intense research interest in modulating antidiabetic drug targets. α-Glucosidase is an antidiabetic target that has gained attention due to its ability to suppress postprandial hyperglycaemia, a key contributor to diabetic complications. This review explored a detailed approach to developing QSAR models, focusing on strategies for generating input variables (molecular descriptors) and computational approaches ranging from classical machine learning algorithms to modern deep learning algorithms. We also highlighted studies that have used these approaches to develop predictive models for α-glucosidase inhibitors to modulate this critical antidiabetic drug target.
Collapse
Affiliation(s)
- Adeshina I Odugbemi
- South African Medical Research Council Bioinformatics Unit, South African National Bioinformatics Institute, University of the Western Cape, Bellville, Cape Town 7535, South Africa
- School of Pharmacy, University of the Western Cape, Bellville, Cape Town 7535, South Africa
- National Institute for Theoretical and Computational Sciences (NITheCS), South Africa
| | - Clement Nyirenda
- Department of Computer Science, University of the Western Cape, Cape Town 7535, South Africa
| | - Alan Christoffels
- South African Medical Research Council Bioinformatics Unit, South African National Bioinformatics Institute, University of the Western Cape, Bellville, Cape Town 7535, South Africa
- Africa Centres for Disease Control and Prevention, African Union, Addis Ababa, Ethiopia
| | - Samuel A Egieyeh
- School of Pharmacy, University of the Western Cape, Bellville, Cape Town 7535, South Africa
- National Institute for Theoretical and Computational Sciences (NITheCS), South Africa
| |
Collapse
|
3
|
Garg P, Singhal G, Kulkarni P, Horne D, Salgia R, Singhal SS. Artificial Intelligence-Driven Computational Approaches in the Development of Anticancer Drugs. Cancers (Basel) 2024; 16:3884. [PMID: 39594838 PMCID: PMC11593155 DOI: 10.3390/cancers16223884] [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: 10/21/2024] [Revised: 11/13/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
The integration of AI has revolutionized cancer drug development, transforming the landscape of drug discovery through sophisticated computational techniques. AI-powered models and algorithms have enhanced computer-aided drug design (CADD), offering unprecedented precision in identifying potential anticancer compounds. Traditionally, cancer drug design has been a complex, resource-intensive process, but AI introduces new opportunities to accelerate discovery, reduce costs, and optimize efficiency. This manuscript delves into the transformative applications of AI-driven methodologies in predicting and developing anticancer drugs, critically evaluating their potential to reshape the future of cancer therapeutics while addressing their challenges and limitations.
Collapse
Affiliation(s)
- Pankaj Garg
- Department of Chemistry, GLA University, Mathura 281406, Uttar Pradesh, India
| | - Gargi Singhal
- Department of Medical Sciences, S.N. Medical College, Agra 282002, Uttar Pradesh, India
| | - Prakash Kulkarni
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - David Horne
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Sharad S. Singhal
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
4
|
Jung J, Moon JO, Ahn SI, Lee H. Predicting antioxidant activity of compounds based on chemical structure using machine learning methods. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2024; 28:527-537. [PMID: 39467716 PMCID: PMC11519722 DOI: 10.4196/kjpp.2024.28.6.527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 10/30/2024]
Abstract
Oxidative stress is a well-established risk factor for numerous chronic diseases, emphasizing the need for efficient identification of potent antioxidants. Conventional methods for assessing antioxidant properties are often time-consuming and resource-intensive, typically relying on laborious biochemical assays. In this study, we investigated the applicability of machine learning (ML) algorithms for predicting the antioxidant activity of compounds based solely on their molecular structure. We evaluated the performance of five ML algorithms, Support Vector Machine (SVM), Logistic Regression (LR), XGBoost, Random Forest (RF), and Deep Neural Network (DNN), using a dataset of over 1,900 compounds with experimentally determined antioxidant activity. Both RF and SVM achieved the best overall performance, exhibiting high accuracy (> 0.9) and effectively distinguishing active and inactive compounds with high structural similarity. External validation using natural product data from the BATMAN database confirmed the generalizability of the RF and SVM models. Our results suggest that ML models serve as powerful tools to expedite the discovery of novel antioxidant candidates, potentially streamlining the development of future therapeutic interventions.
Collapse
Affiliation(s)
- Jinwoo Jung
- Department of Pharmacy, College of Pharmacy and Research Institute for Drug Development, Busan 46241, Korea
- School of Mechanical Engineering, Pusan National University, Busan 46241, Korea
| | - Jeon-Ok Moon
- Department of Pharmacy, College of Pharmacy and Research Institute for Drug Development, Busan 46241, Korea
| | - Song Ih Ahn
- School of Mechanical Engineering, Pusan National University, Busan 46241, Korea
| | - Haeseung Lee
- Department of Pharmacy, College of Pharmacy and Research Institute for Drug Development, Busan 46241, Korea
| |
Collapse
|
5
|
Dhanabalan AK, Devadasan V, Haribabu J, Krishnasamy G. Machine learning models to identify lead compound and substitution optimization to have derived energetics and conformational stability through docking and MD simulations for sphingosine kinase 1. Mol Divers 2024:10.1007/s11030-024-10997-4. [PMID: 39417979 DOI: 10.1007/s11030-024-10997-4] [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: 08/14/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
Sphingosine kinases (SphKs) are a group of important enzymes that circulate at low micromolar concentrations in mammals and have received considerable attention due to the roles they play in a broad array of biological processes including apoptosis, mutagenesis, lymphocyte migration, radio- and chemo-sensitization, and angiogenesis. In the present study, we constructed three classification models by four machine learning (ML) algorithms including naive bayes (NB), support vector machine (SVM), logistic regression, and random forest from 395 compounds. The generated ML models were validated by fivefold cross validation. Five different scaffold hit fragments resulted from SVM model-based virtual screening and docking results indicate that all the five fragments exhibit common hydrogen bond interaction a catalytic residue of SphK1. Further, molecular dynamics (MD) simulations and binding free energy calculation had been carried out with the identified five fragment leads and three cocrystal inhibitors. The best 15 fragments were selected. Molecular dynamics (MD) simulations showed that among these compounds, 7 compounds have favorable binding energy compared with cocrystal inhibitors. Hence, the study showed that the present lead fragments could act as potential inhibitors against therapeutic target of cancers and neurodegenerative disorders.
Collapse
Affiliation(s)
- Anantha Krishnan Dhanabalan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Velmurugan Devadasan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India.
| | - Jebiti Haribabu
- Facultad de Medicina, Universidad de Atacama, Los Carreras 1579, 1532502, Copiapó, Chile
- Chennai Institute of Technology (CIT), Chennai, Tamil Nadu, 600069, India
| | - Gunasekaran Krishnasamy
- Centre of Advanced Study in Crystallography and Biophysics, University of Madras, Guindy Campus, Chennai, Tamil Nadu, 600025, India.
| |
Collapse
|
6
|
Chakraborty C, Bhattacharya M, Pal S, Islam MA. Generative AI in drug discovery and development: the next revolution of drug discovery and development would be directed by generative AI. Ann Med Surg (Lond) 2024; 86:6340-6343. [PMID: 39359753 PMCID: PMC11444559 DOI: 10.1097/ms9.0000000000002438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 07/29/2024] [Indexed: 10/04/2024] Open
Affiliation(s)
- Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal
| | | | - Soumen Pal
- School of Mechanical Engineering, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Md Aminul Islam
- COVID-19 Diagnostic Lab, Department of Microbiology, Noakhali Science and Technology University, Noakhali
- Advanced Molecular Lab, Department of Microbiology, President Abdul Hamid Medical College, Karimganj, Kishoreganj, Bangladesh
| |
Collapse
|
7
|
Imam MA, Alandijany TA, Felemban HR, Attar RM, Faizo AA, Gattan HS, Dwivedi VD, Azhar EI. Machine learning, network pharmacology, and molecular dynamics reveal potent cyclopeptide inhibitors against dengue virus proteins. Mol Divers 2024:10.1007/s11030-024-10975-w. [PMID: 39227512 DOI: 10.1007/s11030-024-10975-w] [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: 07/12/2024] [Accepted: 08/20/2024] [Indexed: 09/05/2024]
Abstract
The dengue virus is a major global health hazard responsible for an estimated 390 million diseases yearly. This study focused on identifying cyclopeptide inhibitors for envelope structural proteins E, NS1, NS3, and NS5. Additionally, 5579 cyclopeptides were individually screened against the four target proteins using a machine learning-based quantitative structure-activity relationship model. Subsequently, the best 10 cyclopeptides from each protein were selected for molecular docking with their corresponding proteins. Moreover, the protein-peptide complexes with the highest affinity were subjected to a 100-ns molecular dynamics simulation. The protein-protein complexes exhibited superior structural stability and binding interactions. Based on the results of the MD simulation analyses, which included checking values for Root Mean Square Deviation, Root Mean Square Fluctuation, Principal Component Analysis (PCA), free energy landscapes, and energetic components, it was found that NS5-CP03714 complex is more stable and has stronger binding interactions than NS3-CP02054. PCA and free energy landscape plots have confirmed the higher conformational stability of NS5-CP03714. Analysis of the energetic components revealed that NS5-CP03714 (total binding energy = - 47.19 kcal/mol) exhibits more favorable interaction energies and overall binding energy compared to NS3-CP02054 (total binding energy = - 27.36 kcal/mol), suggesting a stronger and more stable formation of the complex. In addition, the drug-target network of two specific peptides (CP02950 and CP05582) and their associated target proteins were analyzed. This analysis revealed valuable information about their ability to target several proteins and their potential for broad-spectrum activity. Additional experimental investigations are necessary to validate these computational results and assess the efficacy of identified peptide inhibitors in biological systems.
Collapse
Affiliation(s)
- Mohammed A Imam
- Department of Medical Microbiology and Parasitology, Qunfudah Faculty of Medicine, Umm Al-Qura University, Al-Qunfudah, 21961, Saudi Arabia
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, 21362, Jeddah, Saudi Arabia
| | - Thamir A Alandijany
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, 21362, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, 21362, Jeddah, Saudi Arabia
| | - Hashim R Felemban
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, 21362, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, 21362, Jeddah, Saudi Arabia
| | - Roba M Attar
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, 21362, Jeddah, Saudi Arabia
- Department of Biological Sciences/Microbiology, Faculty of Science, University of Jeddah, , 21959, Jeddah, Saudi Arabia
| | - Arwa A Faizo
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, 21362, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, 21362, Jeddah, Saudi Arabia
| | - Hattan S Gattan
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, 21362, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, 21362, Jeddah, Saudi Arabia
| | - Vivek Dhar Dwivedi
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College and Hospitals, Saveetha University, Chennai, India.
- Bioinformatics Research Division, Quanta Calculus, Greater Noida, India.
| | - Esam I Azhar
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, 21362, Jeddah, Saudi Arabia.
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, 21362, Jeddah, Saudi Arabia.
| |
Collapse
|
8
|
Alkhatabi HA, Naemi FMA, Alsolami R, Alatyb HN. Computational Design and Optimization of Peptide Inhibitors for SIRT2. Pharmaceuticals (Basel) 2024; 17:1120. [PMID: 39338285 PMCID: PMC11435109 DOI: 10.3390/ph17091120] [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: 07/10/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 09/30/2024] Open
Abstract
Sirtuin 2 (SIRT2), an NAD+-dependent deacetylase, is crucial for regulating vital physiological processes, including aging, DNA repair, and cell cycle progression. Its abnormal activity is linked to diseases such as Parkinson's disease, cancer, and metabolic disorders, making it a potential target for therapeutic intervention. While small molecule inhibitors have been studied, peptide-based inhibitors offer a promising alternative due to their selectivity and bioavailability. This study explores the effects of converting the naturally occurring cyclic inhibitor peptide of SIRT2 (S2iL5) into a non-cyclic form by replacing a residue with FAK (LYS + CF3CO-). The new peptide sequence, Tyr-His-Thr-Tyr-His-Val-FAK (LYS)-Arg-Arg-Thr-Asn-Tyr-Tyr-Cys, was modeled to confirm its stable conformation. Docking studies and MM/GBSA calculations showed that the non-cyclic peptide had a better binding free energy (-50.66 kcal/mol) compared to the cyclic S2iL5 (-49.44 kcal/mol). Further mutations generated 160,000 unique peptides, screened using a machine learning-based QSAR model. Three promising peptides (Peptide 1: YGGNNVKRRTNYYC, Peptide 2: YMGEWVKRRTNYYC, and Peptide 3: YGGNGVKRRTNYYC) were selected and further modeled. Molecular dynamics (MD) analyses demonstrated that Peptide 1 and Peptide 2 had significant potential as SIRT2 inhibitors, showing moderate stability and some structural flexibility. Their best binding free energies were -59.07 kcal/mol and -46.01 kcal/mol, respectively. The study aimed to enhance peptide flexibility and binding affinity, suggesting that optimized peptide-based inhibitors can interact effectively with SIRT2. However, further experimental validation is necessary to confirm these computational predictions and evaluate the therapeutic potential of the identified peptides.
Collapse
Affiliation(s)
- Heba A Alkhatabi
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Hematology Research Unit (HRU), King Fahd Medical Research Center (KFMRC), King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Center of Artificial Intelligence for Precision Medicine (CAIPM), King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Fatmah M A Naemi
- Histocompatibility and Immunogenetics Laboratory, Prince Abdulmajed Dialysis Center, King Fahd General Hospital, Ministry of Health, Jeddah 21589, Saudi Arabia
| | - Reem Alsolami
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Hematology Research Unit (HRU), King Fahd Medical Research Center (KFMRC), King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Center of Artificial Intelligence for Precision Medicine (CAIPM), King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hisham N Alatyb
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
9
|
Altayb HN, Alatawi HA. Employing Machine Learning-Based QSAR for Targeting Zika Virus NS3 Protease: Molecular Insights and Inhibitor Discovery. Pharmaceuticals (Basel) 2024; 17:1067. [PMID: 39204173 PMCID: PMC11359100 DOI: 10.3390/ph17081067] [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: 07/15/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 09/03/2024] Open
Abstract
Zika virus infection is a mosquito-borne viral disease that has become a global health concern recently. Zika virus belongs to the Flavivirus genus and is primarily transmitted by Aedes mosquitoes. Prevention of Zika virus infection involves avoiding mosquito bites by using repellent, wearing protective clothing, and staying in screened areas, especially for pregnant women. Treatment focuses on managing symptoms with rest, fluids, and acetaminophen, with close monitoring for pregnant women. Currently, there is no specific antiviral treatment or vaccine for the Zika virus, highlighting the importance of prevention strategies to control its spread. Therefore, in this study, the Zika virus non-structural protein NS3 was targeted to inhibit Zika infection by identifying the novel inhibitor through an in silico approach. Here, 2864 natural compounds were screened using a machine learning-based QSAR model, and later docking was performed to select the potential target. Subsequently, Tanimoto similarity and clustering were performed to obtain the potential target. The three most potential compounds were obtained: (a) 5297, (b) 432449, and (c) 85137543. The protein-ligand complex's stability and flexibility were then investigated by dynamic modelling. The 300 ns simulation showed that 5297 exhibited the steadiest deviation and constant creation of hydrogen bonds. Compared to the other compounds, 5297 demonstrated a superior binding free energy (ΔG = -20.81 kcal/mol) with the protein when the MM/GBSA technique was used. The study determined that 5297 showed significant therapeutic potential and justifies further experimental investigation as a possible inhibitor of the NS2B-NS3 protease target implicated in Zika virus infection.
Collapse
Affiliation(s)
- Hisham N. Altayb
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hanan Ali Alatawi
- Department of Biological Sciences, University Collage of Haqel, University of Tabuk, Tabuk 71491, Saudi Arabia;
| |
Collapse
|
10
|
Chihomvu P, Ganesan A, Gibbons S, Woollard K, Hayes MA. Phytochemicals in Drug Discovery-A Confluence of Tradition and Innovation. Int J Mol Sci 2024; 25:8792. [PMID: 39201478 PMCID: PMC11354359 DOI: 10.3390/ijms25168792] [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: 06/12/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 09/02/2024] Open
Abstract
Phytochemicals have a long and successful history in drug discovery. With recent advancements in analytical techniques and methodologies, discovering bioactive leads from natural compounds has become easier. Computational techniques like molecular docking, QSAR modelling and machine learning, and network pharmacology are among the most promising new tools that allow researchers to make predictions concerning natural products' potential targets, thereby guiding experimental validation efforts. Additionally, approaches like LC-MS or LC-NMR speed up compound identification by streamlining analytical processes. Integrating structural and computational biology aids in lead identification, thus providing invaluable information to understand how phytochemicals interact with potential targets in the body. An emerging computational approach is machine learning involving QSAR modelling and deep neural networks that interrelate phytochemical properties with diverse physiological activities such as antimicrobial or anticancer effects.
Collapse
Affiliation(s)
- Patience Chihomvu
- Compound Synthesis and Management, Discovery Sciences, Biopharmaceuticals R&D, AstraZeneca, 431 83 Mölndal, Sweden
| | - A. Ganesan
- School of Chemistry, Pharmacy & Pharmacology, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK;
| | - Simon Gibbons
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat Al Mawz 616, Oman;
| | - Kevin Woollard
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolic, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB21 6GH, UK;
| | - Martin A. Hayes
- Compound Synthesis and Management, Discovery Sciences, Biopharmaceuticals R&D, AstraZeneca, 431 83 Mölndal, Sweden
| |
Collapse
|
11
|
Adhikari N, Ayyannan SR. Development and validation of machine learning models for the prediction of SH-2 containing protein tyrosine phosphatase 2 inhibitors. Mol Divers 2024; 28:1889-1905. [PMID: 37552436 DOI: 10.1007/s11030-023-10710-x] [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: 05/30/2023] [Accepted: 07/31/2023] [Indexed: 08/09/2023]
Abstract
Discovery and development of a new drug to the market is a highly challenging and resource consuming process. Although, modern drug discovery technologies have enabled the rapid identification of lead compounds, translation of the lead compounds into successful clinical candidates remains a big challenge. In recent years, the availability of massive structural and biological data of diverse small molecules and macromolecules has helped the researchers to deep mine the multidimensional data with the help of artificial intelligence-based predictive tools to draw useful insights on the structural features of biological or therapeutic significance. The aim of this study was to utilize the available data on small molecule (SH2)-containing protein tyrosine phosphatase 2 (SHP2) inhibitors to build and develop machine learning (ML) models that can predict the SHP2 inhibitory potential of new compounds. The dataset contained 2739 unique small molecule SHP2 inhibitors obtained from the BindingDB, ChEMBL and recent literature. After curation of the data, the predictive models such as XGBoost, K nearest neighbours, neural networks were developed and validated through a tenfold cross-validation testing procedure. Out of the seven models developed, the XGBoost model showed an excellent performance with ROC AUC score of 0.96 and accuracy of 0.97 on the test data. Moreover, the Shapley Additive Explanations method was applied to assess a more in-depth understanding of the influence of variables on the model's predictions. In summary, the XGBoost model developed in this study can be useful in the identification of novel SHP2 inhibitors and therefore, can accelerate the discovery of novel therapeutics for cancer therapy.
Collapse
Affiliation(s)
- Nilanjan Adhikari
- Pharmaceutical Chemistry Research Laboratory II, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, UP, 221005, India
| | - Senthil Raja Ayyannan
- Pharmaceutical Chemistry Research Laboratory II, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, UP, 221005, India.
| |
Collapse
|
12
|
Banerjee S, Jana S, Jha T, Ghosh B, Adhikari N. An assessment of crucial structural contributors of HDAC6 inhibitors through fragment-based non-linear pattern recognition and molecular dynamics simulation approaches. Comput Biol Chem 2024; 110:108051. [PMID: 38520883 DOI: 10.1016/j.compbiolchem.2024.108051] [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: 12/31/2023] [Revised: 02/28/2024] [Accepted: 03/08/2024] [Indexed: 03/25/2024]
Abstract
Amidst the Zn2+-dependant isoforms of the HDAC family, HDAC6 has emerged as a potential target associated with an array of diseases, especially cancer and neuronal disorders like Rett's Syndrome, Alzheimer's disease, Huntington's disease, etc. Also, despite the availability of a handful of HDAC inhibitors in the market, their non-selective nature has restricted their use in different disease conditions. In this situation, the development of selective and potent HDAC6 inhibitors will provide efficacious therapeutic agents to treat different diseases. In this context, this study has been carried out to evaluate the potential structural contributors of quinazoline-cap-containing HDAC6 inhibitors via machine learning (ML), conventional classification-dependant QSAR, and MD simulation-based binding mode of interaction analysis toward HDAC6 binding. This combined conventional and modern molecular modeling study has revealed the significance of the quinazoline moiety, substitutions present at the quinazoline cap group, as well as the importance of molecular property, number of hydrogen bond donor-acceptor functions, carbon-chlorine distance that significantly affects the HDAC6 binding of these inhibitors, subsequently affecting their potency . Interestingly, the study also revealed that the substitutions such as the chloroethyl group, and bulky quinazolinyl cap group can affect the binding of the cap function with the amino acid residues present in the loops proximal to the catalytic site of HDAC6. Such contributions of cap groups can lead to both stabilization and destabilization of the cap function after occupying the hydrophobic catalytic site by the aryl hydroxamate linker-ZBG functions.
Collapse
Affiliation(s)
- Suvankar Banerjee
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Sandeep Jana
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Tarun Jha
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Shamirpet, Hyderabad 500078, India
| | - Nilanjan Adhikari
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India.
| |
Collapse
|
13
|
Bosch B, DeJesus MA, Schnappinger D, Rock JM. Weak links: Advancing target-based drug discovery by identifying the most vulnerable targets. Ann N Y Acad Sci 2024; 1535:10-19. [PMID: 38595325 DOI: 10.1111/nyas.15139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Mycobacterium tuberculosis remains the most common infectious killer worldwide despite decades of antitubercular drug development. Effectively controlling the tuberculosis (TB) pandemic will require innovation in drug discovery. In this review, we provide a brief overview of the two main approaches to discovering new TB drugs-phenotypic screens and target-based drug discovery-and outline some of the limitations of each method. We then explore recent advances in genetic tools that aim to overcome some of these limitations. In particular, we highlight a novel metric to prioritize essential targets, termed vulnerability. Stratifying targets based on their vulnerability presents new opportunities for future target-based drug discovery campaigns.
Collapse
Affiliation(s)
- Barbara Bosch
- Laboratory of Host-Pathogen Biology, The Rockefeller University, New York, New York, USA
| | - Michael A DeJesus
- Laboratory of Host-Pathogen Biology, The Rockefeller University, New York, New York, USA
| | - Dirk Schnappinger
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, New York, USA
| | - Jeremy M Rock
- Laboratory of Host-Pathogen Biology, The Rockefeller University, New York, New York, USA
| |
Collapse
|
14
|
Kumar N, Acharya V. Advances in machine intelligence-driven virtual screening approaches for big-data. Med Res Rev 2024; 44:939-974. [PMID: 38129992 DOI: 10.1002/med.21995] [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: 09/12/2022] [Revised: 07/15/2023] [Accepted: 10/29/2023] [Indexed: 12/23/2023]
Abstract
Virtual screening (VS) is an integral and ever-evolving domain of drug discovery framework. The VS is traditionally classified into ligand-based (LB) and structure-based (SB) approaches. Machine intelligence or artificial intelligence has wide applications in the drug discovery domain to reduce time and resource consumption. In combination with machine intelligence algorithms, VS has emerged into revolutionarily progressive technology that learns within robust decision orders for data curation and hit molecule screening from large VS libraries in minutes or hours. The exponential growth of chemical and biological data has evolved as "big-data" in the public domain demands modern and advanced machine intelligence-driven VS approaches to screen hit molecules from ultra-large VS libraries. VS has evolved from an individual approach (LB and SB) to integrated LB and SB techniques to explore various ligand and target protein aspects for the enhanced rate of appropriate hit molecule prediction. Current trends demand advanced and intelligent solutions to handle enormous data in drug discovery domain for screening and optimizing hits or lead with fewer or no false positive hits. Following the big-data drift and tremendous growth in computational architecture, we presented this review. Here, the article categorized and emphasized individual VS techniques, detailed literature presented for machine learning implementation, modern machine intelligence approaches, and limitations and deliberated the future prospects.
Collapse
Affiliation(s)
- Neeraj Kumar
- Artificial Intelligence for Computational Biology Lab (AICoB), Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Vishal Acharya
- Artificial Intelligence for Computational Biology Lab (AICoB), Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| |
Collapse
|
15
|
Dhoble S, Wu TH, Kenry. Decoding Nanomaterial-Biosystem Interactions through Machine Learning. Angew Chem Int Ed Engl 2024; 63:e202318380. [PMID: 38687554 DOI: 10.1002/anie.202318380] [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: 11/30/2023] [Indexed: 05/02/2024]
Abstract
The interactions between biosystems and nanomaterials regulate most of their theranostic and nanomedicine applications. These nanomaterial-biosystem interactions are highly complex and influenced by a number of entangled factors, including but not limited to the physicochemical features of nanomaterials, the types and characteristics of the interacting biosystems, and the properties of the surrounding microenvironments. Over the years, different experimental approaches coupled with computational modeling have revealed important insights into these interactions, although many outstanding questions remain unanswered. The emergence of machine learning has provided a timely and unique opportunity to revisit nanomaterial-biosystem interactions and to further push the boundary of this field. This minireview highlights the development and use of machine learning to decode nanomaterial-biosystem interactions and provides our perspectives on the current challenges and potential opportunities in this field.
Collapse
Affiliation(s)
- Sagar Dhoble
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Tzu-Hsien Wu
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Kenry
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85721, USA
- BIO5 Institute, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
16
|
Mao J, Wang J, Zeb A, Cho KH, Jin H, Kim J, Lee O, Wang Y, No KT. Transformer-Based Molecular Generative Model for Antiviral Drug Design. J Chem Inf Model 2024; 64:2733-2745. [PMID: 37366644 PMCID: PMC11005037 DOI: 10.1021/acs.jcim.3c00536] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Indexed: 06/28/2023]
Abstract
Since the Simplified Molecular Input Line Entry System (SMILES) is oriented to the atomic-level representation of molecules and is not friendly in terms of human readability and editable, however, IUPAC is the closest to natural language and is very friendly in terms of human-oriented readability and performing molecular editing, we can manipulate IUPAC to generate corresponding new molecules and produce programming-friendly molecular forms of SMILES. In addition, antiviral drug design, especially analogue-based drug design, is also more appropriate to edit and design directly from the functional group level of IUPAC than from the atomic level of SMILES, since designing analogues involves altering the R group only, which is closer to the knowledge-based molecular design of a chemist. Herein, we present a novel data-driven self-supervised pretraining generative model called "TransAntivirus" to make select-and-replace edits and convert organic molecules into the desired properties for design of antiviral candidate analogues. The results indicated that TransAntivirus is significantly superior to the control models in terms of novelty, validity, uniqueness, and diversity. TransAntivirus showed excellent performance in the design and optimization of nucleoside and non-nucleoside analogues by chemical space analysis and property prediction analysis. Furthermore, to validate the applicability of TransAntivirus in the design of antiviral drugs, we conducted two case studies on the design of nucleoside analogues and non-nucleoside analogues and screened four candidate lead compounds against anticoronavirus disease (COVID-19). Finally, we recommend this framework for accelerating antiviral drug discovery.
Collapse
Affiliation(s)
- Jiashun Mao
- The
Interdisciplinary Graduate Program in Integrative Biotechnology and
Translational Medicine, Yonsei University, Incheon 21983, Republic of Korea
| | - Jianmin Wang
- The
Interdisciplinary Graduate Program in Integrative Biotechnology and
Translational Medicine, Yonsei University, Incheon 21983, Republic of Korea
| | - Amir Zeb
- Faculty
of Natural and Basic Sciences, University
of Turbat, Balochistan 92600, Pakistan
| | - Kwang-Hwi Cho
- School
of Systems Biomedical Science, Soongsil
University, Seoul 06978, Republic of Korea
| | - Haiyan Jin
- The
Interdisciplinary Graduate Program in Integrative Biotechnology and
Translational Medicine, Yonsei University, Incheon 21983, Republic of Korea
| | - Jongwan Kim
- Department
of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
- Bioinformatics
and Molecular Design Research Center (BMDRC), Incheon 21983, Republic of Korea
| | - Onju Lee
- The
Interdisciplinary Graduate Program in Integrative Biotechnology and
Translational Medicine, Yonsei University, Incheon 21983, Republic of Korea
| | - Yunyun Wang
- School
of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and
Molecular Drug Target, Nantong University, Nantong 226001, Jiangsu, P. R. China
| | - Kyoung Tai No
- The
Interdisciplinary Graduate Program in Integrative Biotechnology and
Translational Medicine, Yonsei University, Incheon 21983, Republic of Korea
| |
Collapse
|
17
|
Nandi S, Bhaduri S, Das D, Ghosh P, Mandal M, Mitra P. Deciphering the Lexicon of Protein Targets: A Review on Multifaceted Drug Discovery in the Era of Artificial Intelligence. Mol Pharm 2024; 21:1563-1590. [PMID: 38466810 DOI: 10.1021/acs.molpharmaceut.3c01161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Understanding protein sequence and structure is essential for understanding protein-protein interactions (PPIs), which are essential for many biological processes and diseases. Targeting protein binding hot spots, which regulate signaling and growth, with rational drug design is promising. Rational drug design uses structural data and computational tools to study protein binding sites and protein interfaces to design inhibitors that can change these interactions, thereby potentially leading to therapeutic approaches. Artificial intelligence (AI), such as machine learning (ML) and deep learning (DL), has advanced drug discovery and design by providing computational resources and methods. Quantum chemistry is essential for drug reactivity, toxicology, drug screening, and quantitative structure-activity relationship (QSAR) properties. This review discusses the methodologies and challenges of identifying and characterizing hot spots and binding sites. It also explores the strategies and applications of artificial-intelligence-based rational drug design technologies that target proteins and protein-protein interaction (PPI) binding hot spots. It provides valuable insights for drug design with therapeutic implications. We have also demonstrated the pathological conditions of heat shock protein 27 (HSP27) and matrix metallopoproteinases (MMP2 and MMP9) and designed inhibitors of these proteins using the drug discovery paradigm in a case study on the discovery of drug molecules for cancer treatment. Additionally, the implications of benzothiazole derivatives for anticancer drug design and discovery are deliberated.
Collapse
Affiliation(s)
- Suvendu Nandi
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| | - Soumyadeep Bhaduri
- Centre for Computational and Data Sciences, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| | - Debraj Das
- Centre for Computational and Data Sciences, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| | - Priya Ghosh
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| | - Pralay Mitra
- Department of Computer Science and Engineering, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| |
Collapse
|
18
|
Srisongkram T, Tookkane D. Insights into the structure-activity relationship of pyrimidine-sulfonamide analogues for targeting BRAF V600E protein. Biophys Chem 2024; 307:107179. [PMID: 38241826 DOI: 10.1016/j.bpc.2024.107179] [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: 11/23/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 01/21/2024]
Abstract
B-rapidly accelerated fibrosarcoma (BRAF) V600E plays a crucial role in the progression of cutaneous melanoma. Core structures of BRAF V600E inhibitors are based on pyrimidine-sulfonamide scaffolds. Exploring the QSAR of these structures can improve our understanding of BRAF V600E inhibitor drug design. This study utilized machine learning-based QSAR to elucidate chemical substructures of pyrimidine-sulfonamide analogues that correlated to the BRAF V600E inhibitory activity. The findings indicate that the support vector regression (SVR) combined with 15 fingerprints achieved the highest statistical performances in terms of goodness-of-fit, robustness, and predictability. Nine key fingerprints from pyrimidine-sulfonamide analogues were identified to exert the BRAF V600E inhibitory activity. These key fingerprints were validated using network-based activity cliff landscape and molecular docking. Together, the developed algorithm can serve as a screening tool for designing BRAF V600E inhibitors. To further utilize this model, we deployed our developed algorithm at https://qsarlabs.com/#braf.
Collapse
Affiliation(s)
- Tarapong Srisongkram
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, 40002, Thailand.
| | - Dheerapat Tookkane
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, 40002, Thailand
| |
Collapse
|
19
|
Lee KH, Won SJ, Oyinloye P, Shi L. Unlocking the Potential of High-Quality Dopamine Transporter Pharmacological Data: Advancing Robust Machine Learning-Based QSAR Modeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.06.583803. [PMID: 38558976 PMCID: PMC10979915 DOI: 10.1101/2024.03.06.583803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The dopamine transporter (DAT) plays a critical role in the central nervous system and has been implicated in numerous psychiatric disorders. The ligand-based approaches are instrumental to decipher the structure-activity relationship (SAR) of DAT ligands, especially the quantitative SAR (QSAR) modeling. By gathering and analyzing data from literature and databases, we systematically assemble a diverse range of ligands binding to DAT, aiming to discern the general features of DAT ligands and uncover the chemical space for potential novel DAT ligand scaffolds. The aggregation of DAT pharmacological activity data, particularly from databases like ChEMBL, provides a foundation for constructing robust QSAR models. The compilation and meticulous filtering of these data, establishing high-quality training datasets with specific divisions of pharmacological assays and data types, along with the application of QSAR modeling, prove to be a promising strategy for navigating the pertinent chemical space. Through a systematic comparison of DAT QSAR models using training datasets from various ChEMBL releases, we underscore the positive impact of enhanced data set quality and increased data set size on the predictive power of DAT QSAR models.
Collapse
Affiliation(s)
- Kuo Hao Lee
- Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Sung Joon Won
- Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Precious Oyinloye
- Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Lei Shi
- Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
20
|
Tripathi T, Singh DB, Tripathi T. Computational resources and chemoinformatics for translational health research. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 139:27-55. [PMID: 38448138 DOI: 10.1016/bs.apcsb.2023.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
The integration of computational resources and chemoinformatics has revolutionized translational health research. It has offered a powerful set of tools for accelerating drug discovery. This chapter overviews the computational resources and chemoinformatics methods used in translational health research. The resources and methods can be used to analyze large datasets, identify potential drug candidates, predict drug-target interactions, and optimize treatment regimens. These resources have the potential to transform the drug discovery process and foster personalized medicine research. We discuss insights into their various applications in translational health and emphasize the need for addressing challenges, promoting collaboration, and advancing the field to fully realize the potential of these tools in transforming healthcare.
Collapse
Affiliation(s)
- Tripti Tripathi
- Molecular and Structural Biophysics Laboratory, Department of Biochemistry, North-Eastern Hill University, Shillong, India
| | - Dev Bukhsh Singh
- Department of Biotechnology, Siddharth University, Kapilvastu, Siddharth Nagar, India
| | - Timir Tripathi
- Molecular and Structural Biophysics Laboratory, Department of Zoology, North-Eastern Hill University, Shillong, India.
| |
Collapse
|
21
|
Singh AV, Shelar A, Rai M, Laux P, Thakur M, Dosnkyi I, Santomauro G, Singh AK, Luch A, Patil R, Bill J. Harmonization Risks and Rewards: Nano-QSAR for Agricultural Nanomaterials. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:2835-2852. [PMID: 38315814 DOI: 10.1021/acs.jafc.3c06466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
This comprehensive review explores the emerging landscape of Nano-QSAR (quantitative structure-activity relationship) for assessing the risk and potency of nanomaterials in agricultural settings. The paper begins with an introduction to Nano-QSAR, providing background and rationale, and explicitly states the hypotheses guiding the review. The study navigates through various dimensions of nanomaterial applications in agriculture, encompassing their diverse properties, types, and associated challenges. Delving into the principles of QSAR in nanotoxicology, this article elucidates its application in evaluating the safety of nanomaterials, while addressing the unique limitations posed by these materials. The narrative then transitions to the progression of Nano-QSAR in the context of agricultural nanomaterials, exemplified by insightful case studies that highlight both the strengths and the limitations inherent in this methodology. Emerging prospects and hurdles tied to Nano-QSAR in agriculture are rigorously examined, casting light on important pathways forward, existing constraints, and avenues for research enhancement. Culminating in a synthesis of key insights, the review underscores the significance of Nano-QSAR in shaping the future of nanoenabled agriculture. It provides strategic guidance to steer forthcoming research endeavors in this dynamic field.
Collapse
Affiliation(s)
- Ajay Vikram Singh
- Department of Chemical and Product Safety, German Federal Institute of Risk Assessment (BfR), Maxdohrnstrasse 8-10, 10589 Berlin, Germany
| | - Amruta Shelar
- Department of Technology, Savitribai Phule Pune University, Pune 411007, India
| | - Mansi Rai
- Department of Microbiology, Central University of Rajasthan NH-8, Bandar Sindri, Dist-Ajmer-305817, Rajasthan, India
| | - Peter Laux
- Department of Chemical and Product Safety, German Federal Institute of Risk Assessment (BfR), Maxdohrnstrasse 8-10, 10589 Berlin, Germany
| | - Manali Thakur
- Uniklinik Köln, Kerpener Strasse 62, 50937 Köln Germany
| | - Ievgen Dosnkyi
- Institute of Chemistry and Biochemistry Department of Organic ChemistryFreie Universität Berlin Takustr. 3 14195 Berlin, Germany
| | - Giulia Santomauro
- Institute for Materials Science, Department of Bioinspired Materials, University of Stuttgart, 70569, Stuttgart, Germany
| | - Alok Kumar Singh
- Department of Plant Molecular Biology & Genetic Engineering, ANDUA&T, Ayodhya 224229, Uttar Pradesh, India
| | - Andreas Luch
- Department of Chemical and Product Safety, German Federal Institute of Risk Assessment (BfR), Maxdohrnstrasse 8-10, 10589 Berlin, Germany
| | - Rajendra Patil
- Department of Technology, Savitribai Phule Pune University, Pune 411007, India
| | - Joachim Bill
- Institute for Materials Science, Department of Bioinspired Materials, University of Stuttgart, 70569, Stuttgart, Germany
| |
Collapse
|
22
|
Djoumbou-Feunang Y, Wilmot J, Kinney J, Chanda P, Yu P, Sader A, Sharifi M, Smith S, Ou J, Hu J, Shipp E, Tomandl D, Kumpatla SP. Cheminformatics and artificial intelligence for accelerating agrochemical discovery. Front Chem 2023; 11:1292027. [PMID: 38093816 PMCID: PMC10716421 DOI: 10.3389/fchem.2023.1292027] [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: 09/10/2023] [Accepted: 11/09/2023] [Indexed: 10/17/2024] Open
Abstract
The global cost-benefit analysis of pesticide use during the last 30 years has been characterized by a significant increase during the period from 1990 to 2007 followed by a decline. This observation can be attributed to several factors including, but not limited to, pest resistance, lack of novelty with respect to modes of action or classes of chemistry, and regulatory action. Due to current and projected increases of the global population, it is evident that the demand for food, and consequently, the usage of pesticides to improve yields will increase. Addressing these challenges and needs while promoting new crop protection agents through an increasingly stringent regulatory landscape requires the development and integration of infrastructures for innovative, cost- and time-effective discovery and development of novel and sustainable molecules. Significant advances in artificial intelligence (AI) and cheminformatics over the last two decades have improved the decision-making power of research scientists in the discovery of bioactive molecules. AI- and cheminformatics-driven molecule discovery offers the opportunity of moving experiments from the greenhouse to a virtual environment where thousands to billions of molecules can be investigated at a rapid pace, providing unbiased hypothesis for lead generation, optimization, and effective suggestions for compound synthesis and testing. To date, this is illustrated to a far lesser extent in the publicly available agrochemical research literature compared to drug discovery. In this review, we provide an overview of the crop protection discovery pipeline and how traditional, cheminformatics, and AI technologies can help to address the needs and challenges of agrochemical discovery towards rapidly developing novel and more sustainable products.
Collapse
Affiliation(s)
| | - Jeremy Wilmot
- Corteva Agriscience, Crop Protection Discovery and Development, Indianapolis, IN, United States
| | - John Kinney
- Corteva Agriscience, Farming Solutions and Digital, Indianapolis, IN, United States
| | - Pritam Chanda
- Corteva Agriscience, Farming Solutions and Digital, Indianapolis, IN, United States
| | - Pulan Yu
- Corteva Agriscience, Crop Protection Discovery and Development, Indianapolis, IN, United States
| | - Avery Sader
- Corteva Agriscience, Crop Protection Discovery and Development, Indianapolis, IN, United States
| | - Max Sharifi
- Corteva Agriscience, Regulatory and Stewardship, Indianapolis, IN, United States
| | - Scott Smith
- Corteva Agriscience, Farming Solutions and Digital, Indianapolis, IN, United States
| | - Junjun Ou
- Corteva Agriscience, Crop Protection Discovery and Development, Indianapolis, IN, United States
| | - Jie Hu
- Corteva Agriscience, Farming Solutions and Digital, Indianapolis, IN, United States
| | - Elizabeth Shipp
- Corteva Agriscience UK Limited, Regulation Innovation Center, Abingdon, United Kingdom
| | | | | |
Collapse
|
23
|
Samanipour S, O’Brien JW, Reid MJ, Thomas KV, Praetorius A. From Molecular Descriptors to Intrinsic Fish Toxicity of Chemicals: An Alternative Approach to Chemical Prioritization. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:17950-17958. [PMID: 36480454 PMCID: PMC10666547 DOI: 10.1021/acs.est.2c07353] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/27/2022] [Accepted: 11/28/2022] [Indexed: 06/17/2023]
Abstract
The European and U.S. chemical agencies have listed approximately 800k chemicals about which knowledge of potential risks to human health and the environment is lacking. Filling these data gaps experimentally is impossible, so in silico approaches and prediction are essential. Many existing models are however limited by assumptions (e.g., linearity and continuity) and small training sets. In this study, we present a supervised direct classification model that connects molecular descriptors to toxicity. Categories can be driven by either data (using k-means clustering) or defined by regulation. This was tested via 907 experimentally defined 96 h LC50 values for acute fish toxicity. Our classification model explained ≈90% of the variance in our data for the training set and ≈80% for the test set. This strategy gave a 5-fold decrease in the frequency of incorrect categorization compared to a quantitative structure-activity relationship (QSAR) regression model. Our model was subsequently employed to predict the toxicity categories of ≈32k chemicals. A comparison between the model-based applicability domain (AD) and the training set AD was performed, suggesting that the training set-based AD is a more adequate way to avoid extrapolation when using such models. The better performance of our direct classification model compared to that of QSAR methods makes this approach a viable tool for assessing the hazards and risks of chemicals.
Collapse
Affiliation(s)
- Saer Samanipour
- Van
’t Hoff Institute for Molecular Sciences (HIMS), University of Amsterdam (UvA), 1090 GDAmsterdam, The Netherlands
- UvA
Data Science Center, University of Amsterdam, 1090 GDAmsterdam, The Netherlands
- Queensland
Alliance for Environmental Health Sciences (QAEHS), The University of Queensland, Brisbane, QLD4072, Australia
| | - Jake W. O’Brien
- Van
’t Hoff Institute for Molecular Sciences (HIMS), University of Amsterdam (UvA), 1090 GDAmsterdam, The Netherlands
- Queensland
Alliance for Environmental Health Sciences (QAEHS), The University of Queensland, Brisbane, QLD4072, Australia
| | - Malcolm J. Reid
- Norwegian
Institute for Water Research (NIVA), NO-0579Oslo, Norway
| | - Kevin V. Thomas
- Queensland
Alliance for Environmental Health Sciences (QAEHS), The University of Queensland, Brisbane, QLD4072, Australia
| | - Antonia Praetorius
- Institute
for Biodiversity and Ecosystem Dynamics (IBED), University of Amsterdam, 1090 GDAmsterdam, The Netherlands
| |
Collapse
|
24
|
Piochi LF, Preto AJ, Moreira IS. DELFOS-drug efficacy leveraging forked and specialized networks-benchmarking scRNA-seq data in multi-omics-based prediction of cancer sensitivity. Bioinformatics 2023; 39:btad645. [PMID: 37862234 PMCID: PMC10627353 DOI: 10.1093/bioinformatics/btad645] [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: 02/20/2023] [Revised: 09/28/2023] [Accepted: 10/19/2023] [Indexed: 10/22/2023] Open
Abstract
MOTIVATION Cancer is currently one of the most notorious diseases, with over 1 million deaths in the European Union alone in 2022. As each tumor can be composed of diverse cell types with distinct genotypes, cancer cells can acquire resistance to different compounds. Moreover, anticancer drugs can display severe side effects, compromising patient well-being. Therefore, novel strategies for identifying the optimal set of compounds to treat each tumor have become an important research topic in recent decades. RESULTS To address this challenge, we developed a novel drug response prediction algorithm called Drug Efficacy Leveraging Forked and Specialized networks (DELFOS). Our model learns from multi-omics data from over 65 cancer cell lines, as well as structural data from over 200 compounds, for the prediction of drug sensitivity. We also evaluated the benefits of incorporating single-cell expression data to predict drug response. DELFOS was validated using datasets with unseen cell lines or drugs and compared with other state-of-the-art algorithms, achieving a high prediction performance on several correlation and error metrics. Overall, DELFOS can effectively leverage multi-omics data for the prediction of drug responses in thousands of drug-cell line pairs. AVAILABILITY AND IMPLEMENTATION The DELFOS pipeline and associated data are available at github.com/MoreiraLAB/delfos.
Collapse
Affiliation(s)
- Luiz Felipe Piochi
- Department of Life Sciences, University of Coimbra, Coimbra 3000-456, Portugal
- CNC—Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, Coimbra 3004-504, Portugal
| | - António J Preto
- CNC—Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, Coimbra 3004-504, Portugal
- PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra 3030-789, Portugal
| | - Irina S Moreira
- Department of Life Sciences, University of Coimbra, Coimbra 3000-456, Portugal
- CNC—Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, Coimbra 3004-504, Portugal
| |
Collapse
|
25
|
Fedorov R, Gryn’ova G. Unlocking the Potential: Predicting Redox Behavior of Organic Molecules, from Linear Fits to Neural Networks. J Chem Theory Comput 2023; 19:4796-4814. [PMID: 37463673 PMCID: PMC10414033 DOI: 10.1021/acs.jctc.3c00355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Indexed: 07/20/2023]
Abstract
Redox-active organic molecules, i.e., molecules that can relatively easily accept and/or donate electrons, are ubiquitous in biology, chemical synthesis, and electronic and spintronic devices, such as solar cells and rechargeable batteries, etc. Choosing the best candidates from an essentially infinite chemical space for experimental testing in a target application requires efficient screening approaches. In this Review, we discuss modern in silico techniques for predicting reduction and oxidation potentials of organic molecules that go beyond conventional first-principles computations and thermodynamic cycles. Approaches ranging from simple linear fits based on molecular orbital energy approximation and energy difference approximation to advanced regression and neural network machine learning algorithms employing complex descriptors of molecular compositions, geometries, and electronic structures are examined in conjunction with relevant literature examples. We discuss the interplay between ab initio data and machine learning (ML), i.e., whether it is better to base predictions on low-level quantum-chemical results corrected with ML or to bypass first-principles computations entirely and instead rely on elaborate deep learning architectures. Finally, we list currently available data sets of redox-active organic molecules and their experimental and/or computed properties to facilitate the development of screening platforms and rational design of redox-active organic molecules.
Collapse
Affiliation(s)
- Rostislav Fedorov
- Heidelberg
Institute for Theoretical Studies (HITS gGmbH), 69118 Heidelberg, Germany
- Interdisciplinary
Center for Scientific Computing, Heidelberg
University, 69120 Heidelberg, Germany
| | - Ganna Gryn’ova
- Heidelberg
Institute for Theoretical Studies (HITS gGmbH), 69118 Heidelberg, Germany
- Interdisciplinary
Center for Scientific Computing, Heidelberg
University, 69120 Heidelberg, Germany
| |
Collapse
|
26
|
Srisongkram T, Khamtang P, Weerapreeyakul N. Prediction of KRAS G12C inhibitors using conjoint fingerprint and machine learning-based QSAR models. J Mol Graph Model 2023; 122:108466. [PMID: 37058997 DOI: 10.1016/j.jmgm.2023.108466] [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: 09/30/2022] [Revised: 03/19/2023] [Accepted: 03/29/2023] [Indexed: 04/16/2023]
Abstract
Kirsten rat sarcoma virus G12C (KRASG12C) is the major protein mutation associated with non-small cell lung cancer (NSCLC) severity. Inhibiting KRASG12C is therefore one of the key therapeutic strategies for NSCLC patients. In this paper, a cost-effective data driven drug design employing machine learning-based quantitative structure-activity relationship (QSAR) analysis was built for predicting ligand affinities against KRASG12C protein. A curated and non-redundant dataset of 1033 compounds with KRASG12C inhibitory activity (pIC50) was used to build and test the models. The PubChem fingerprint, Substructure fingerprint, Substructure fingerprint count, and the conjoint fingerprint-a combination of PubChem fingerprint and Substructure fingerprint count-were used to train the models. Using comprehensive validation methods and various machine learning algorithms, the results clearly showed that the XGBoost regression (XGBoost) achieved the highest performance in term of goodness of fit, predictivity, generalizability and model robustness (R2 = 0.81, Q2CV = 0.60, Q2Ext = 0.62, R2 - Q2Ext = 0.19, R2Y-Random = 0.31 ± 0.03, Q2Y-Random = -0.09 ± 0.04). The top 13 molecular fingerprints that correlated with the predicted pIC50 values were SubFPC274 (aromatic atoms), SubFPC307 (number of chiral-centers), PubChemFP37 (≥1 Chlorine), SubFPC18 (Number of alkylarylethers), SubFPC1 (number of primary carbons), SubFPC300 (number of 1,3-tautomerizables), PubChemFP621 (N-C:C:C:N structure), PubChemFP23 (≥1 Fluorine), SubFPC2 (number of secondary carbons), SubFPC295 (number of C-ONS bonds), PubChemFP199 (≥4 6-membered rings), PubChemFP180 (≥1 nitrogen-containing 6-membered ring), and SubFPC180 (number of tertiary amine). These molecular fingerprints were virtualized and validated using molecular docking experiments. In conclusion, this conjoint fingerprint and XGBoost-QSAR model demonstrated to be useful as a high-throughput screening tool for KRASG12C inhibitor identification and drug design.
Collapse
Affiliation(s)
- Tarapong Srisongkram
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, 40002, Thailand.
| | | | - Natthida Weerapreeyakul
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, 40002, Thailand
| |
Collapse
|
27
|
Chen X, Wei Q, Si F, Wang F, Lu Q, Guo Z, Chai Y, Zhu R, Xing G, Jin Q, Zhang G. Design and Identification of a Novel Antiviral Affinity Peptide against Fowl Adenovirus Serotype 4 (FAdV-4) by Targeting Fiber2 Protein. Viruses 2023; 15:v15040821. [PMID: 37112802 PMCID: PMC10146638 DOI: 10.3390/v15040821] [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: 02/22/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Outbreaks of hydropericardium hepatitis syndrome caused by fowl adenovirus serotype 4 (FAdV-4) with a novel genotype have been reported in China since 2015, with significant economic losses to the poultry industry. Fiber2 is one of the important structural proteins on FAdV-4 virions. In this study, the C-terminal knob domain of the FAdV-4 Fiber2 protein was expressed and purified, and its trimer structure (PDB ID: 7W83) was determined for the first time. A series of affinity peptides targeting the knob domain of the Fiber2 protein were designed and synthesized on the basis of the crystal structure using computer virtual screening technology. A total of eight peptides were screened using an immunoperoxidase monolayer assay and RT-qPCR, and they exhibited strong binding affinities to the knob domain of the FAdV-4 Fiber2 protein in a surface plasmon resonance assay. Treatment with peptide number 15 (P15; WWHEKE) at different concentrations (10, 25, and 50 μM) significantly reduced the expression level of the Fiber2 protein and the viral titer during FAdV-4 infection. P15 was found to be an optimal peptide with antiviral activity against FAdV-4 in vitro with no cytotoxic effect on LMH cells up to 200 μM. This study led to the identification of a class of affinity peptides designed using computer virtual screening technology that targeted the knob domain of the FAdV-4 Fiber2 protein and may be developed as a novel potential and effective antiviral strategy in the prevention and control of FAdV-4.
Collapse
Affiliation(s)
- Xiao Chen
- College of Veterinary Medicine, Northwest A&F University, Xianyang 712100, China
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Qiang Wei
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Fusheng Si
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China
| | - Fangyu Wang
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Qingxia Lu
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Zhenhua Guo
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Yongxiao Chai
- College of Veterinary Medicine, Northwest A&F University, Xianyang 712100, China
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Rongfang Zhu
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
- College of Life Science, Henan Agricultural University, Zhengzhou 450002, China
| | - Guangxu Xing
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Qianyue Jin
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Gaiping Zhang
- College of Veterinary Medicine, Northwest A&F University, Xianyang 712100, China
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
28
|
Kour S, Biswas I, Sheoran S, Arora S, Sheela P, Duppala SK, Murthy DK, Pawar SC, Singh H, Kumar D, Prabhu D, Vuree S, Kumar R. Artificial intelligence and nanotechnology for cervical cancer treatment: Current status and future perspectives. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
|
29
|
Zaslavsky J, Bannigan P, Allen C. Re-envisioning the design of nanomedicines: harnessing automation and artificial intelligence. Expert Opin Drug Deliv 2023; 20:241-257. [PMID: 36644850 DOI: 10.1080/17425247.2023.2167978] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
INTRODUCTION Interest in nanomedicines has surged in recent years due to the critical role they have played in the COVID-19 pandemic. Nanoformulations can turn promising therapeutic cargo into viable products through improvements in drug safety and efficacy profiles. However, the developmental pathway for such formulations is non-trivial and largely reliant on trial-and-error. Beyond the costly demands on time and resources, this traditional approach may stunt innovation. The emergence of automation, artificial intelligence (AI) and machine learning (ML) tools, which are currently underutilized in pharmaceutical formulation development, offers a promising direction for an improved path in the design of nanomedicines. AREAS COVERED the potential of harnessing experimental automation and AI/ML to drive innovation in nanomedicine development. The discussion centers on the current challenges in drug formulation research and development, and the major advantages afforded through the application of data-driven methods. EXPERT OPINION The development of integrated workflows based on automated experimentation and AI/ML may accelerate nanomedicine development. A crucial step in achieving this is the generation of high-quality, accessible datasets. Future efforts to make full use of these tools can ultimately contribute to the development of more innovative nanomedicines and improved clinical translation of formulations that rely on advanced drug delivery systems.
Collapse
Affiliation(s)
- Jonathan Zaslavsky
- Leslie Dan Faculty of Pharmacy, University of Toronto, M5S 3M2, Toronto, ON, Canada
| | - Pauric Bannigan
- Leslie Dan Faculty of Pharmacy, University of Toronto, M5S 3M2, Toronto, ON, Canada
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy, University of Toronto, M5S 3M2, Toronto, ON, Canada
| |
Collapse
|
30
|
Prathima TS, Ahmad MG, Karuppasamy R, Chanda K, Balamurali MM. Investigation on Phyto‐active Constituent of
Clerodendrum paniculatum
as Therapeutic Agent against Viral Diseases. ChemistrySelect 2023. [DOI: 10.1002/slct.202203932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- T. S. Prathima
- Division of Chemistry School of Advanced Sciences Vellore Institute of Technology Chennai Tamil Nadu India 600027
| | - Md. Gulzar Ahmad
- Department of Chemistry School of Advanced Sciences Vellore Institute of Technology Vellore Tamil Nadu India 632014
| | - Ramanathan Karuppasamy
- Department of Biotechnology School of BioSciences and Technology Vellore Institute of Technology Vellore Tamil Nadu India 632014
| | - Kaushik Chanda
- Department of Chemistry School of Advanced Sciences Vellore Institute of Technology Vellore Tamil Nadu India 632014
| | - M. M. Balamurali
- Division of Chemistry School of Advanced Sciences Vellore Institute of Technology Chennai Tamil Nadu India 600027
| |
Collapse
|
31
|
Belfield SJ, Cronin MTD, Enoch SJ, Firman JW. Guidance for good practice in the application of machine learning in development of toxicological quantitative structure-activity relationships (QSARs). PLoS One 2023; 18:e0282924. [PMID: 37163504 PMCID: PMC10171609 DOI: 10.1371/journal.pone.0282924] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 02/26/2023] [Indexed: 05/12/2023] Open
Abstract
Recent years have seen a substantial growth in the adoption of machine learning approaches for the purposes of quantitative structure-activity relationship (QSAR) development. Such a trend has coincided with desire to see a shifting in the focus of methodology employed within chemical safety assessment: away from traditional reliance upon animal-intensive in vivo protocols, and towards increased application of in silico (or computational) predictive toxicology. With QSAR central amongst techniques applied in this area, the emergence of algorithms trained through machine learning with the objective of toxicity estimation has, quite naturally, arisen. On account of the pattern-recognition capabilities of the underlying methods, the statistical power of the ensuing models is potentially considerable-appropriate for the handling even of vast, heterogeneous datasets. However, such potency comes at a price: this manifesting as the general practical deficits observed with respect to the reproducibility, interpretability and generalisability of the resulting tools. Unsurprisingly, these elements have served to hinder broader uptake (most notably within a regulatory setting). Areas of uncertainty liable to accompany (and hence detract from applicability of) toxicological QSAR have previously been highlighted, accompanied by the forwarding of suggestions for "best practice" aimed at mitigation of their influence. However, the scope of such exercises has remained limited to "classical" QSAR-that conducted through use of linear regression and related techniques, with the adoption of comparatively few features or descriptors. Accordingly, the intention of this study has been to extend the remit of best practice guidance, so as to address concerns specific to employment of machine learning within the field. In doing so, the impact of strategies aimed at enhancing the transparency (feature importance, feature reduction), generalisability (cross-validation) and predictive power (hyperparameter optimisation) of algorithms, trained upon real toxicity data through six common learning approaches, is evaluated.
Collapse
Affiliation(s)
- Samuel J Belfield
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Mark T D Cronin
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Steven J Enoch
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - James W Firman
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| |
Collapse
|
32
|
Köse E, Erkan Köse M, Güneşdoğdu Sağdınç S. Principal component analysis of quantum mechanical descriptors data to reveal the pharmacological activities of oxindole derivatives. RESULTS IN CHEMISTRY 2023. [DOI: 10.1016/j.rechem.2023.100905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
|
33
|
Application of a deep generative model produces novel and diverse functional peptides against microbial resistance. Comput Struct Biotechnol J 2022; 21:463-471. [PMID: 36618982 PMCID: PMC9804011 DOI: 10.1016/j.csbj.2022.12.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022] Open
Abstract
Antimicrobial resistance could threaten millions of lives in the immediate future. Antimicrobial peptides (AMPs) are an alternative to conventional antibiotics practice against infectious diseases. Despite the potential contribution of AMPs to the antibiotic's world, their development and optimization have encountered serious challenges. Cutting-edge methods with novel and improved selectivity toward resistant targets must be established to create AMPs-driven treatments. Here, we present AMPTrans-lstm, a deep generative network-based approach for the rational design of AMPs. The AMPTrans-lstm pipeline involves pre-training, transfer learning, and module identification. The AMPTrans-lstm model has two sub-models, namely, (long short-term memory) LSTM sampler and Transformer converter, which can be connected in series to make full use of the stability of LSTM and the novelty of Transformer model. These elements could generate AMPs candidates, which can then be tailored for specific applications. By analyzing the generated sequence and trained AMPs, we prove that AMPTrans-lstm can expand the design space of the trained AMPs and produce reasonable and brand-new AMPs sequences. AMPTrans-lstm can generate functional peptides for antimicrobial resistance with good novelty and diversity, so it is an efficient AMPs design tool.
Collapse
|
34
|
Tullius Scotti M, Herrera-Acevedo C, Barros de Menezes RP, Martin HJ, Muratov EN, Ítalo de Souza Silva Á, Faustino Albuquerque E, Ferreira Calado L, Coy-Barrera E, Scotti L. MolPredictX: Online Biological Activity Predictions by Machine Learning Models. Mol Inform 2022; 41:e2200133. [PMID: 35961924 DOI: 10.1002/minf.202200133] [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: 06/06/2022] [Accepted: 08/12/2022] [Indexed: 01/05/2023]
Abstract
Here we report the development of MolPredictX, an innovate and freely accessible web interface for biological activity predictions of query molecules. MolPredictX utilizes in-house QSAR models to provide 27 qualitative predictions (active or inactive), and quantitative probabilities for bioactivity against parasitic (Trypanosoma and Leishmania), viral (Dengue, Sars-CoV and Hepatitis C), pathogenic yeast (Candida albicans), bacterial (Salmonella enterica and Escherichia coli), and Alzheimer disease enzymes. In this article, we introduce the methodology and usability of this webtool, highlighting its potential role in the development of new drugs against a variety of diseases. MolPredictX is undergoing continuous development and is freely available at https://www.molpredictx.ufpb.br/.
Collapse
Affiliation(s)
- Marcus Tullius Scotti
- Programa de Pós-Graduação de Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, 58051-900, João Pessoa-PB, Brazil
| | - Chonny Herrera-Acevedo
- Programa de Pós-Graduação de Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, 58051-900, João Pessoa-PB, Brazil.,Department of Chemical Engineering, Universidad ECCI, Carrera 19 # 49-20, 111311, Bogotá D.C., Colombia
| | - Renata Priscila Barros de Menezes
- Programa de Pós-Graduação de Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, 58051-900, João Pessoa-PB, Brazil
| | - Holli-Joi Martin
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Eugene N Muratov
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Ávilla Ítalo de Souza Silva
- Programa de Pós-Graduação de Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, 58051-900, João Pessoa-PB, Brazil
| | - Emmanuella Faustino Albuquerque
- Programa de Pós-Graduação de Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, 58051-900, João Pessoa-PB, Brazil
| | - Lucas Ferreira Calado
- Programa de Pós-Graduação de Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, 58051-900, João Pessoa-PB, Brazil
| | - Ericsson Coy-Barrera
- Bioorganic Chemistry Laboratory, Facultad de Ciencias Básicas y Aplicadas, Universidad Militar Nueva Granada, Cajicá, 250247, Colombia
| | - Luciana Scotti
- Programa de Pós-Graduação de Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, 58051-900, João Pessoa-PB, Brazil
| |
Collapse
|
35
|
Liu R, Laxminarayan S, Reifman J, Wallqvist A. Enabling data-limited chemical bioactivity predictions through deep neural network transfer learning. J Comput Aided Mol Des 2022; 36:867-878. [PMID: 36272041 DOI: 10.1007/s10822-022-00486-x] [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: 08/06/2022] [Accepted: 10/17/2022] [Indexed: 01/07/2023]
Abstract
The main limitation in developing deep neural network (DNN) models to predict bioactivity properties of chemicals is the lack of sufficient assay data to train the network's classification layers. Focusing on feedforward DNNs that use atom- and bond-based structural fingerprints as input, we examined whether layers of a fully trained DNN based on large amounts of data to predict one property could be used to develop DNNs to predict other related or unrelated properties based on limited amounts of data. Hence, we assessed if and under what conditions the dense layers of a pre-trained DNN could be transferred and used for the development of another DNN associated with limited training data. We carried out a quantitative study employing more than 400 pairs of assay datasets, where we used fully trained layers from a large dataset to augment the training of a small dataset. We found that the higher the correlation r between two assay datasets, the more efficient the transfer learning is in reducing prediction errors associated with the smaller dataset DNN predictions. The reduction in mean squared prediction errors ranged from 10 to 20% for every 0.1 increase in r2 between the datasets, with the bulk of the error reductions associated with transfers of the first dense layer. Transfer of other dense layers did not result in additional benefits, suggesting that deeper, dense layers conveyed more specialized and assay-specific information. Importantly, depending on the dataset correlation, training sample size could be reduced by up to tenfold without any loss of prediction accuracy.
Collapse
Affiliation(s)
- Ruifeng Liu
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, FCMR-TT, 504 Scott Street, Fort Detrick, MD, 21702-5012, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Srinivas Laxminarayan
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, FCMR-TT, 504 Scott Street, Fort Detrick, MD, 21702-5012, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Jaques Reifman
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, FCMR-TT, 504 Scott Street, Fort Detrick, MD, 21702-5012, USA
| | - Anders Wallqvist
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, FCMR-TT, 504 Scott Street, Fort Detrick, MD, 21702-5012, USA.
| |
Collapse
|
36
|
Machine learning and structure-based modeling for the prediction of UDP-glucuronosyltransferase inhibition. iScience 2022; 25:105290. [PMID: 36304105 PMCID: PMC9593791 DOI: 10.1016/j.isci.2022.105290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/05/2022] [Accepted: 10/03/2022] [Indexed: 11/23/2022] Open
Abstract
UDP-glucuronosyltransferases (UGTs) are responsible for 35% of the phase II drug metabolism. In this study, we focused on UGT1A1, which is a key UGT isoform. Strong inhibition of UGT1A1 may trigger adverse drug/herb-drug interactions, or result in disorders of endobiotic metabolism. Most of the current machine learning methods predicting the inhibition of drug metabolizing enzymes neglect protein structure and dynamics, both being essential for the recognition of various substrates and inhibitors. We performed molecular dynamics simulations on a homology model of the human UGT1A1 structure containing both the cofactor- (UDP-glucuronic acid) and substrate-binding domains to explore UGT conformational changes. Then, we created models for the prediction of UGT1A1 inhibitors by integrating information on UGT1A1 structure and dynamics, interactions with diverse ligands, and machine learning. These models can be helpful for further prediction of drug-drug interactions of drug candidates and safety treatments. UGTs are responsible for 35% of the phase II drug metabolism reactions We created machine learning models for prediction of UGT1A1 inhibitors Our simulations suggested key residues of UGT1A1 involved in the substrate binding
Collapse
|
37
|
Pal R, Patra SG, Chattaraj PK. Quantitative Structure-Toxicity Relationship in Bioactive Molecules from a Conceptual DFT Perspective. Pharmaceuticals (Basel) 2022; 15:1383. [PMID: 36355555 PMCID: PMC9695291 DOI: 10.3390/ph15111383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/01/2022] [Accepted: 11/07/2022] [Indexed: 10/29/2023] Open
Abstract
The preclinical drug discovery stage often requires a large amount of costly and time-consuming experiments using huge sets of chemical compounds. In the last few decades, this process has undergone significant improvements by the introduction of quantitative structure-activity relationship (QSAR) modelling that uses a certain percentage of experimental data to predict the biological activity/property of compounds with similar structural skeleton and/or containing a particular functional group(s). The use of machine learning tools along with it has made life even easier for pharmaceutical researchers. Here, we discuss the toxicity of certain sets of bioactive compounds towards Pimephales promelas and Tetrahymena pyriformis in terms of the global conceptual density functional theory (CDFT)-based descriptor, electrophilicity index (ω). We have compared the results with those obtained by using the commonly used hydrophobicity parameter, logP (where P is the n-octanol/water partition coefficient), considering the greater ease of computing the ω descriptor. The Human African trypanosomiasis (HAT) curing activity of 32 pyridyl benzamide derivatives is also studied against Tryphanosoma brucei. In this review article, we summarize these multiple linear regression (MLR)-based QSAR studies in terms of electrophilicity (ω, ω2) and hydrophobicity (logP, (logP)2) parameters.
Collapse
Affiliation(s)
- Ranita Pal
- Advanced Technology Development Centre, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Shanti Gopal Patra
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Pratim Kumar Chattaraj
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| |
Collapse
|
38
|
Yang L, Chen P, He K, Wang R, Chen G, Shan G, Zhu L. Predicting bioconcentration factor and estrogen receptor bioactivity of bisphenol a and its analogues in adult zebrafish by directed message passing neural networks. ENVIRONMENT INTERNATIONAL 2022; 169:107536. [PMID: 36152365 DOI: 10.1016/j.envint.2022.107536] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/23/2022] [Accepted: 09/19/2022] [Indexed: 06/16/2023]
Abstract
The bioconcentration factor (BCF) is a key parameter for bioavailability assessment of environmental pollutants in regulatory frameworks. The comparative toxicology and mechanism of action of congeners are also of concern. However, there are limitations to acquire them by conducting field and laboratory experiments while machinelearning is emerging as a promising predictive tool to fill the gap. In this study, the Direct Message Passing Neural Network (DMPNN) was applied to predict logBCFs of bisphenol A (BPA) and its four analogues (bisphenol AF (BPAF), bisphenol B (BPB), bisphenol F (BPF) and bisphenol S (BPS)). For the test set, the Pearson correlation coefficient (PCC) and mean square error (MSE) were 0.85 and 0.52 respectively, suggesting a good predictive performance. The predicted logBCFs values by the DMPNN ranging from 0.35 (BPS) to 2.14 (BPAF) coincided well with those by the classical EPI Suite (BCFBAF model). Besides, estrogen receptor α (ERα) bioactivity of these bisphenols was also predicted well by the DMPNN, with a probability of 97.0 % (BPB) to 99.7 % (BPAF), which was validated by the extent of vitellogenin (VTG) induction in male zebrafish as a biomarker except BPS. Thus, with little need for expert knowledge, DMPNN is confirmed to be a useful tool to accurately predict logBCF and screen for estrogenic activity from molecular structures. Moreover, a gender difference was noted in the changes of three endpoints (logBCF, ER binding affinity and VTG levels), the rank order of which was BPAF > BPB > BPA > BPF > BPS consistently, and abnormal amino acid metabolism is featured as an omics signature of abnormal hormone protein expression.
Collapse
Affiliation(s)
- Liping Yang
- Key Laboratory of Pollution Processes and Environmental Criteria, Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Pengyu Chen
- Key Laboratory of Pollution Processes and Environmental Criteria, Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China; College of Oceanography, Hohai University, Nanjing 210098, China
| | - Keyan He
- Key Laboratory of Pollution Processes and Environmental Criteria, Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Ruihan Wang
- College of Chemistry, Sichuan University, Chengdu, Sichuan 610064, China
| | - Geng Chen
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 330106, China
| | - Guoqiang Shan
- Key Laboratory of Pollution Processes and Environmental Criteria, Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China.
| | - Lingyan Zhu
- Key Laboratory of Pollution Processes and Environmental Criteria, Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| |
Collapse
|
39
|
Kanapeckaitė A, Mažeikienė A, Geris L, Burokienė N, Cottrell GS, Widera D. Computational pharmacology: New avenues for COVID-19 therapeutics search and better preparedness for future pandemic crises. Biophys Chem 2022; 290:106891. [PMID: 36137310 PMCID: PMC9464258 DOI: 10.1016/j.bpc.2022.106891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/03/2022] [Accepted: 09/04/2022] [Indexed: 01/07/2023]
Abstract
The COVID-19 pandemic created an unprecedented global healthcare emergency prompting the exploration of new therapeutic avenues, including drug repurposing. A large number of ongoing studies revealed pervasive issues in clinical research, such as the lack of accessible and organised data. Moreover, current shortcomings in clinical studies highlighted the need for a multi-faceted approach to tackle this health crisis. Thus, we set out to explore and develop new strategies for drug repositioning by employing computational pharmacology, data mining, systems biology, and computational chemistry to advance shared efforts in identifying key targets, affected networks, and potential pharmaceutical intervention options. Our study revealed that formulating pharmacological strategies should rely on both therapeutic targets and their networks. We showed how data mining can reveal regulatory patterns, capture novel targets, alert about side-effects, and help identify new therapeutic avenues. We also highlighted the importance of the miRNA regulatory layer and how this information could be used to monitor disease progression or devise treatment strategies. Importantly, our work bridged the interactome with the chemical compound space to better understand the complex landscape of COVID-19 drugs. Machine and deep learning allowed us to showcase limitations in current chemical libraries for COVID-19 suggesting that both in silico and experimental analyses should be combined to retrieve therapeutically valuable compounds. Based on the gathered data, we strongly advocate for taking this opportunity to establish robust practices for treating today's and future infectious diseases by preparing solid analytical frameworks.
Collapse
Affiliation(s)
- Austė Kanapeckaitė
- AK Consulting, Laisvės g. 7, LT 12007 Vilnius, Lithuania,Corresponding author
| | - Asta Mažeikienė
- Department of Physiology, Biochemistry, Microbiology and Laboratory Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, M. K. Čiurlionio g. 21, LT-03101 Vilnius, Lithuania
| | - Liesbet Geris
- Biomechanics Research Unit, GIGA In Silico Medicine, University of Liège, Quartier Hôpital, Avenue de l'Hôpital 11 (B34), Liège 4000, Belgium,Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Celestijnenlaan 300C (2419), Leuven 3001, Belgium,Skeletel Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, Herestraat 49 (813), Leuven 3000, Belgium
| | - Neringa Burokienė
- Clinics of Internal Diseases, Family Medicine and Oncology, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, M. K. Čiurlionio str. 21/27, LT-03101 Vilnius, Lithuania
| | - Graeme S. Cottrell
- University of Reading, School of Pharmacy, Hopkins Building, Reading RG6 6UB, United Kingdom
| | - Darius Widera
- University of Reading, School of Pharmacy, Hopkins Building, Reading RG6 6UB, United Kingdom
| |
Collapse
|
40
|
Goel H, Yu W, MacKerell AD. hERG Blockade Prediction by Combining Site Identification by Ligand Competitive Saturation and Physicochemical Properties. CHEMISTRY (BASEL, SWITZERLAND) 2022; 4:630-646. [PMID: 36712295 PMCID: PMC9881610 DOI: 10.3390/chemistry4030045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Human ether-a-go-go-related gene (hERG) potassium channel is well-known contributor to drug-induced cardiotoxicity and therefore an extremely important target when performing safety assessments of drug candidates. Ligand-based approaches in connection with quantitative structure active relationships (QSAR) analyses have been developed to predict hERG toxicity. Availability of the recent published cryogenic electron microscopy (cryo-EM) structure for the hERG channel opened the prospect for using structure-based simulation and docking approaches for hERG drug liability predictions. In recent time, the idea of combining structure- and ligand-based approaches for modeling hERG drug liability has gained momentum offering improvements in predictability when compared to ligand-based QSAR practices alone. The present article demonstrates uniting the structure-based SILCS (site-identification by ligand competitive saturation) approach in conjunction with physicochemical properties to develop predictive models for hERG blockade. This combination leads to improved model predictability based on Pearson's R and percent correct (represents rank-ordering of ligands) metric for different validation sets of hERG blockers involving diverse chemical scaffold and wide range of pIC50 values. The inclusion of the SILCS structure-based approach allows determination of the hERG region to which compounds bind and the contribution of different chemical moieties in the compounds to blockade, thereby facilitating the rational ligand design to minimize hERG liability.
Collapse
Affiliation(s)
- Himanshu Goel
- Computer Aided Drug Design Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn St. Baltimore, MD 21201, United States
| | - Wenbo Yu
- Computer Aided Drug Design Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn St. Baltimore, MD 21201, United States
| | - Alexander D. MacKerell
- Computer Aided Drug Design Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn St. Baltimore, MD 21201, United States
| |
Collapse
|
41
|
Okagu IU, Aham EC, Ezeorba TPC, Ndefo JC, Aguchem RN, Udenigwe CC. Osteo‐modulatory dietary proteins and peptides: A concise review. J Food Biochem 2022; 46:e14365. [DOI: 10.1111/jfbc.14365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/20/2022] [Accepted: 07/18/2022] [Indexed: 11/29/2022]
Affiliation(s)
| | - Emmanuel Chigozie Aham
- Department of Biochemistry, Faculty of Biological Sciences University of Nigeria Nsukka Nigeria
| | | | - Joseph Chinedum Ndefo
- Department of Science Laboratory Technology Faculty of Physical Sciences, University of Nigeria Nsukka Nigeria
| | - Rita Ngozi Aguchem
- Department of Biochemistry, Faculty of Biological Sciences University of Nigeria Nsukka Nigeria
| | - Chibuike C. Udenigwe
- School of Nutrition Sciences, Faculty of Health Sciences University of Ottawa Ottawa Ontario Canada
| |
Collapse
|
42
|
Mao J, Zeb A, Kim MS, Jeon HN, Wang J, Guan S, No KT. Development of an innovative data-driven system to generate descriptive prediction equation of dielectric constant on small sample sets. Heliyon 2022; 8:e10011. [PMID: 36016529 PMCID: PMC9396556 DOI: 10.1016/j.heliyon.2022.e10011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 04/13/2022] [Accepted: 07/15/2022] [Indexed: 11/29/2022] Open
Abstract
Dielectric constant (DC, ε) is a fundamental parameter in material sciences to measure polarizability of the system. In industrial processes, its value is an imperative indicator, which demonstrates the dielectric property of material and compiles information including separation information, chemical equilibrium, chemical reactivity analysis, and solubility modeling. Since, the available ε-prediction models are fairly primitive and frequently suffer from serious failures especially when deals with strong polar compounds. Therefore, we have developed a novel data-driven system to improve the efficiency and wide-range applicability of ε using in material sciences. This innovative scheme adopts the correlation distance and genetic algorithm to discriminate features’ combination and avoid overfitting. Herein, the prediction output of the single ML model as a coding to estimate the target value by simulating the layer-by-layer extraction in deep learning, and enabling instant search for the optimal combination of features is recruited. Our model established an improved correlation value of 0.956 with target as compared to the previously available best traditional ML result of 0.877. Our framework established a profound improvement, especially for material systems possessing ε value >50. In terms of interpretability, we have derived a conceptual computational equation from a minimum generating tree. Our innovative data-driven system is preferentially superior over other methods due to its application for the prediction of dielectric constants as well as for the prediction of overall micro and macro-properties of any multi-components complex.
Collapse
|
43
|
Wang J, Chu Y, Mao J, Jeon HN, Jin H, Zeb A, Jang Y, Cho KH, Song T, No KT. De novo molecular design with deep molecular generative models for PPI inhibitors. Brief Bioinform 2022; 23:6643455. [PMID: 35830870 DOI: 10.1093/bib/bbac285] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/14/2022] [Accepted: 06/20/2022] [Indexed: 12/27/2022] Open
Abstract
We construct a protein-protein interaction (PPI) targeted drug-likeness dataset and propose a deep molecular generative framework to generate novel drug-likeness molecules from the features of the seed compounds. This framework gains inspiration from published molecular generative models, uses the key features associated with PPI inhibitors as input and develops deep molecular generative models for de novo molecular design of PPI inhibitors. For the first time, quantitative estimation index for compounds targeting PPI was applied to the evaluation of the molecular generation model for de novo design of PPI-targeted compounds. Our results estimated that the generated molecules had better PPI-targeted drug-likeness and drug-likeness. Additionally, our model also exhibits comparable performance to other several state-of-the-art molecule generation models. The generated molecules share chemical space with iPPI-DB inhibitors as demonstrated by chemical space analysis. The peptide characterization-oriented design of PPI inhibitors and the ligand-based design of PPI inhibitors are explored. Finally, we recommend that this framework will be an important step forward for the de novo design of PPI-targeted therapeutics.
Collapse
Affiliation(s)
- Jianmin Wang
- The Interdisciplinary Graduate Program in Integrative Biotechnology and Translational Medicine, Yonsei University, Incheon 21983, Republic of Korea.,Bioinformatics and Molecular Design Research Center (BMDRC), Incheon 21983, Republic of Korea
| | - Yanyi Chu
- State Key Laboratory of Microbial Metabolism, Shanghai-Islamabad Belgrade Joint Innovation Center on Antibacterial Resistances, Joint International Research Laboratory of Metabolic & Developmental Sciences and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Jiashun Mao
- The Interdisciplinary Graduate Program in Integrative Biotechnology and Translational Medicine, Yonsei University, Incheon 21983, Republic of Korea.,Bioinformatics and Molecular Design Research Center (BMDRC), Incheon 21983, Republic of Korea
| | - Hyeon-Nae Jeon
- Bioinformatics and Molecular Design Research Center (BMDRC), Incheon 21983, Republic of Korea.,Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Haiyan Jin
- The Interdisciplinary Graduate Program in Integrative Biotechnology and Translational Medicine, Yonsei University, Incheon 21983, Republic of Korea.,Bioinformatics and Molecular Design Research Center (BMDRC), Incheon 21983, Republic of Korea
| | - Amir Zeb
- The Interdisciplinary Graduate Program in Integrative Biotechnology and Translational Medicine, Yonsei University, Incheon 21983, Republic of Korea.,Department of Natural and Basic Sciences, University of Turbat, 92600, Pakistan
| | - Yuil Jang
- The Interdisciplinary Graduate Program in Integrative Biotechnology and Translational Medicine, Yonsei University, Incheon 21983, Republic of Korea.,Bioinformatics and Molecular Design Research Center (BMDRC), Incheon 21983, Republic of Korea
| | - Kwang-Hwi Cho
- School of Systems Biomedical Science, Soongsil University, Seoul, Republic of Korea
| | - Tao Song
- School of Computer Science and Technology, China University of Petroleum, Qingdao, 266580, Shandong, China
| | - Kyoung Tai No
- The Interdisciplinary Graduate Program in Integrative Biotechnology and Translational Medicine, Yonsei University, Incheon 21983, Republic of Korea.,Bioinformatics and Molecular Design Research Center (BMDRC), Incheon 21983, Republic of Korea
| |
Collapse
|
44
|
Sun D, Gao W, Hu H, Zhou S. Why 90% of clinical drug development fails and how to improve it? Acta Pharm Sin B 2022; 12:3049-3062. [PMID: 35865092 PMCID: PMC9293739 DOI: 10.1016/j.apsb.2022.02.002] [Citation(s) in RCA: 473] [Impact Index Per Article: 157.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 02/03/2022] [Accepted: 02/06/2022] [Indexed: 12/14/2022] Open
Abstract
Ninety percent of clinical drug development fails despite implementation of many successful strategies, which raised the question whether certain aspects in target validation and drug optimization are overlooked? Current drug optimization overly emphasizes potency/specificity using structure‒activity-relationship (SAR) but overlooks tissue exposure/selectivity in disease/normal tissues using structure‒tissue exposure/selectivity-relationship (STR), which may mislead the drug candidate selection and impact the balance of clinical dose/efficacy/toxicity. We propose structure‒tissue exposure/selectivity-activity relationship (STAR) to improve drug optimization, which classifies drug candidates based on drug's potency/selectivity, tissue exposure/selectivity, and required dose for balancing clinical efficacy/toxicity. Class I drugs have high specificity/potency and high tissue exposure/selectivity, which needs low dose to achieve superior clinical efficacy/safety with high success rate. Class II drugs have high specificity/potency and low tissue exposure/selectivity, which requires high dose to achieve clinical efficacy with high toxicity and needs to be cautiously evaluated. Class III drugs have relatively low (adequate) specificity/potency but high tissue exposure/selectivity, which requires low dose to achieve clinical efficacy with manageable toxicity but are often overlooked. Class IV drugs have low specificity/potency and low tissue exposure/selectivity, which achieves inadequate efficacy/safety, and should be terminated early. STAR may improve drug optimization and clinical studies for the success of clinical drug development.
Collapse
Affiliation(s)
- Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Wei Gao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hongxiang Hu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Simon Zhou
- Translational Development and Clinical Pharmacology, Bristol Meyer Squibb Company, Summit, NJ, 07920, USA
| |
Collapse
|
45
|
Barros de Menezes RP, Fechine Tavares J, Kato MJ, da Rocha Coelho FA, Sousa Dos Santos AL, da Franca Rodrigues KA, Sessions ZL, Muratov EN, Scotti L, Tullius Scotti M. Natural Products from Annonaceae as Potential Antichagasic Agents. ChemMedChem 2022; 17:e202200196. [PMID: 35678042 DOI: 10.1002/cmdc.202200196] [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: 04/06/2022] [Revised: 06/06/2022] [Indexed: 11/12/2022]
Abstract
Chagas disease, a neglected tropical disease, is endemic in 21 Latin American countries and particularly prevalent in Brazil. Chagas disease has drawn more attention in recent years due to its expansion into non-endemic areas. The aim of this work was to computationally identify and experimentally validate the natural products from an Annonaceae family as antichagasic agents. Through the ligand-based virtual screening, we identified 57 molecules with potential activity against the epimastigote form of T. cruzi. Then, 16 molecules were analyzed in the in vitro study, of which, six molecules displayed previously unknown antiepimastigote activity. We also evaluated these six molecules for trypanocidal activity. We observed that all six molecules have potential activity against the amastigote form, but no molecules were active against the trypomastigote form. 13-Epicupressic acid seems to be the most promising, as it was predicted as an active compound in the in silico study against the amastigote form of T. cruzi, in addition to having in vitro activity against the epimastigote form.
Collapse
Affiliation(s)
- Renata Priscila Barros de Menezes
- Programa de Pós-Graduação de Produtos Naturais e Sintéticos Bioativos, Universidade federal da Paraíba, 58051-900, João Pessoa, PB, Brazil
| | - Josean Fechine Tavares
- Programa de Pós-Graduação de Produtos Naturais e Sintéticos Bioativos, Universidade federal da Paraíba, 58051-900, João Pessoa, PB, Brazil
| | - Massuo Jorge Kato
- Instituto de Química, Universidade de São Paulo, 05508-000, São Paulo, SP, Brazil
| | | | | | | | - Zoe L Sessions
- Molecular Modeling Lab, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, 27599, NC, USA
| | - Eugene N Muratov
- Molecular Modeling Lab, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, 27599, NC, USA
| | - Luciana Scotti
- Programa de Pós-Graduação de Produtos Naturais e Sintéticos Bioativos, Universidade federal da Paraíba, 58051-900, João Pessoa, PB, Brazil
| | - Marcus Tullius Scotti
- Programa de Pós-Graduação de Produtos Naturais e Sintéticos Bioativos, Universidade federal da Paraíba, 58051-900, João Pessoa, PB, Brazil
| |
Collapse
|
46
|
Huang HJ, Lee YH, Chou CL, Zheng CM, Chiu HW. Investigation of potential descriptors of chemical compounds on prevention of nephrotoxicity via QSAR approach. Comput Struct Biotechnol J 2022; 20:1876-1884. [PMID: 35521549 PMCID: PMC9052077 DOI: 10.1016/j.csbj.2022.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/02/2022] [Accepted: 04/11/2022] [Indexed: 11/15/2022] Open
Abstract
Drug-induced nephrotoxicity remains a common problem after exposure to medications and diagnostic agents, which may be heightened in the kidney microenvironment and deteriorate kidney function. In this study, the toxic effects of fourteen marked drugs with the individual chemical structure were evaluated in kidney cells. The quantitative structure-activity relationship (QSAR) approach was employed to investigate the potential structural descriptors of each drug-related to their toxic effects. The most reasonable equation of the QSAR model displayed that the estimated regression coefficients such as the number of ring assemblies, three-membered rings, and six-membered rings were strongly related to toxic effects on renal cells. Meanwhile, the chemical properties of the tested compounds including carbon atoms, bridge bonds, H-bond donors, negative atoms, and rotatable bonds were favored properties and promote the toxic effects on renal cells. Particularly, more numbers of rotatable bonds were positively correlated with strong toxic effects that displayed on the most toxic compound. The useful information discovered from our regression QSAR models may help to identify potential hazardous moiety to avoid nephrotoxicity in renal preventive medicine.
Collapse
Key Words
- AKI, acute kidney injury
- CKD, chronic kidney disease
- DIKD, drug-induced kidney disease
- ESRD, end‐stage renal disease
- GFA, genetic function approximation
- GFR, glomerular filtration rate
- Genetic algorithm
- KCSF, keratinocyte serum-free
- Nephrotoxicity
- PBS, phosphate buffered saline
- QSAR
- QSAR, quantitative structure-activity relationship
- SRB, sulforhodamine B
Collapse
Affiliation(s)
- Hung-Jin Huang
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Hsuan Lee
- Department of Cosmeceutics, China Medical University, Taichung, Taiwan
| | - Chu-Lin Chou
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taiwan
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Hsin Kuo Min Hospital, Taipei Medical University, Taoyuan City, Taiwan
| | - Cai-Mei Zheng
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taiwan
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
| | - Hui-Wen Chiu
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Medical Research, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| |
Collapse
|
47
|
Machine Learning Analysis of Essential Oils from Cuban Plants: Potential Activity against Protozoa Parasites. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27041366. [PMID: 35209156 PMCID: PMC8878085 DOI: 10.3390/molecules27041366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/10/2022] [Accepted: 02/15/2022] [Indexed: 12/04/2022]
Abstract
Essential oils (EOs) are a mixture of chemical compounds with a long history of use in food, cosmetics, perfumes, agricultural and pharmaceuticals industries. The main object of this study was to find chemical patterns between 45 EOs and antiprotozoal activity (antiplasmodial, antileishmanial and antitrypanosomal), using different machine learning algorithms. In the analyses, 45 samples of EOs were included, using unsupervised Self-Organizing Maps (SOM) and supervised Random Forest (RF) methodologies. In the generated map, the hit rate was higher than 70% and the results demonstrate that it is possible find chemical patterns using a supervised and unsupervised machine learning approach. A total of 20 compounds were identified (19 are terpenes and one sulfur-containing compound), which was compared with literature reports. These models can be used to investigate and screen for bioactivity of EOs that have antiprotozoal activity more effectively and with less time and financial cost.
Collapse
|
48
|
Matsuzaka Y, Uesawa Y. A Deep Learning-Based Quantitative Structure-Activity Relationship System Construct Prediction Model of Agonist and Antagonist with High Performance. Int J Mol Sci 2022; 23:ijms23042141. [PMID: 35216254 PMCID: PMC8877122 DOI: 10.3390/ijms23042141] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/12/2022] [Accepted: 02/14/2022] [Indexed: 01/27/2023] Open
Abstract
Molecular design and evaluation for drug development and chemical safety assessment have been advanced by quantitative structure–activity relationship (QSAR) using artificial intelligence techniques, such as deep learning (DL). Previously, we have reported the high performance of prediction models molecular initiation events (MIEs) on the adverse toxicological outcome using a DL-based QSAR method, called DeepSnap-DL. This method can extract feature values from images generated on a three-dimensional (3D)-chemical structure as a novel QSAR analytical system. However, there is room for improvement of this system’s time-consumption. Therefore, in this study, we constructed an improved DeepSnap-DL system by combining the processes of generating an image from a 3D-chemical structure, DL using the image as input data, and statistical calculation of prediction-performance. Consequently, we obtained that the three prediction models of agonists or antagonists of MIEs achieved high prediction-performance by optimizing the parameters of DeepSnap, such as the angle used in the depiction of the image of a 3D-chemical structure, data-split, and hyperparameters in DL. The improved DeepSnap-DL system will be a powerful tool for computer-aided molecular design as a novel QSAR system.
Collapse
Affiliation(s)
- Yasunari Matsuzaka
- Department of Medical Molecular Informatics, Meiji Pharmaceutical University, Kiyose 204-8588, Japan;
- Center for Gene and Cell Therapy, Division of Molecular and Medical Genetics, The Institute of Medical Science, University of Tokyo, Minato City 108-8639, Japan
| | - Yoshihiro Uesawa
- Department of Medical Molecular Informatics, Meiji Pharmaceutical University, Kiyose 204-8588, Japan;
- Correspondence: ; Tel.: +81-42-495-8983
| |
Collapse
|
49
|
Wang F, Hu S, Ma DQ, Li Q, Li HC, Liang JY, Chang S, Kong R. ER/AR Multi-Conformational Docking Server: A Tool for Discovering and Studying Estrogen and Androgen Receptor Modulators. Front Pharmacol 2022; 13:800885. [PMID: 35140614 PMCID: PMC8819068 DOI: 10.3389/fphar.2022.800885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 01/03/2022] [Indexed: 11/13/2022] Open
Abstract
The prediction of the estrogen receptor (ER) and androgen receptor (AR) activity of a compound is quite important to avoid the environmental exposures of endocrine-disrupting chemicals. The Estrogen and Androgen Receptor Database (EARDB, http://eardb.schanglab.org.cn/) provides a unique collection of reported ERα, ERβ, or AR protein structures and known small molecule modulators. With the user-uploaded query molecules, molecular docking based on multi-conformations of a single target will be performed. Moreover, the 2D similarity search against known modulators is also provided. Molecules predicted with a low binding energy or high similarity to known ERα, ERβ, or AR modulators may be potential endocrine-disrupting chemicals or new modulators. The server provides a tool to predict the endocrine activity for compounds of interests, benefiting for the ER and AR drug design and endocrine-disrupting chemical identification.
Collapse
Affiliation(s)
- Feng Wang
- Changzhou University Huaide College, Taizhou, China
| | - Shuai Hu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, School of Chemical and Environmental Engineering, Jiangsu University of Technology, Changzhou, China
| | - De-Qing Ma
- Changzhou University Huaide College, Taizhou, China
| | - Qiuye Li
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, School of Chemical and Environmental Engineering, Jiangsu University of Technology, Changzhou, China
| | - Hong-Cheng Li
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, School of Chemical and Environmental Engineering, Jiangsu University of Technology, Changzhou, China
| | - Jia-Yi Liang
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, School of Chemical and Environmental Engineering, Jiangsu University of Technology, Changzhou, China
| | - Shan Chang
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, School of Chemical and Environmental Engineering, Jiangsu University of Technology, Changzhou, China
| | - Ren Kong
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, School of Chemical and Environmental Engineering, Jiangsu University of Technology, Changzhou, China
| |
Collapse
|