1
|
Yu X, Chen Y, Chen L, Li W, Wang Y, Tang Y, Liu G. GCLmf: A Novel Molecular Graph Contrastive Learning Framework Based on Hard Negatives and Application in Toxicity Prediction. Mol Inform 2024:e202400169. [PMID: 39421969 DOI: 10.1002/minf.202400169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024]
Abstract
In silico methods for prediction of chemical toxicity can decrease the cost and increase the efficiency in the early stage of drug discovery. However, due to low accessibility of sufficient and reliable toxicity data, constructing robust and accurate prediction models is challenging. Contrastive learning, a type of self-supervised learning, leverages large unlabeled data to obtain more expressive molecular representations, which can boost the prediction performance on downstream tasks. While molecular graph contrastive learning has gathered growing attentions, current models neglect the quality of negative data set. Here, we proposed a self-supervised pretraining deep learning framework named GCLmf. We first utilized molecular fragments that meet specific conditions as hard negative samples to boost the quality of the negative set and thus increase the difficulty of the proxy tasks during pre-training to learn informative representations. GCLmf has shown excellent predictive power on various molecular property benchmarks and demonstrates high performance in 33 toxicity tasks in comparison with multiple baselines. In addition, we further investigated the necessity of introducing hard negatives in model building and the impact of the proportion of hard negatives on the model.
Collapse
Affiliation(s)
- Xinxin Yu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Yuanting Chen
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Long Chen
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Weihua Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Yuhao Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Yun Tang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| | - Guixia Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, China
| |
Collapse
|
2
|
Yadav A, Verma A, Singh SK, Prakash R, Srivastava S, Sethi A, Pratap Singh R. Palladium catalysed cross coupling reactions on 2,3-isoxazol-17α-ethynyltestosterone, their anti-cancer activity, molecular docking studies and ADMET analysis. Steroids 2024; 212:109515. [PMID: 39307446 DOI: 10.1016/j.steroids.2024.109515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/26/2024]
Abstract
In the current study, the Sonogashira coupling reaction of danazol with aryl halides was carried out, yielding new aryl substituted danazol derivatives. The synthetic compounds were examined for anti-cancer potential on the HeLa human cervical cancer cell line, and they showed promising cytotoxic action. Synthesized compounds 2, 4 and 5 inhibited the growth of HeLa cervical cancer cells, potentially making them effective anti-cancer drugs in the future. Furthermore, molecular docking studies were performed to evaluate the inhibitory impact of danazol derivatives on the Human Papillomavirus (HPV) target protein (1F9F). The docking results showed a significant inhibitory action against the cervical cancer protein (1F9F). The binding energy (ΔG) values of 1, 2, 3, 4 and 5 against the protein 1F9F were -8.01, -8.70, -9.43, -9.58 and -9.75 kcal/mol, indicating a high affinity of the synthesized compounds to bind with the HPV target proteins compared to their parent compound danazol (1). ADMET analyses of all derivatives have also been carried out.
Collapse
Affiliation(s)
- Astha Yadav
- Department of Chemistry, University of Lucknow, Lucknow 226007, India
| | - Anmol Verma
- Department of Chemistry, University of Lucknow, Lucknow 226007, India
| | | | - Rohit Prakash
- Department of Applied Science & Humanities, Institute of Engineering & Technology, Lucknow 226021 India
| | - Sanjay Srivastava
- Department of Applied Science & Humanities, Institute of Engineering & Technology, Lucknow 226021 India
| | - Arun Sethi
- Department of Chemistry, University of Lucknow, Lucknow 226007, India
| | - Ranvijay Pratap Singh
- Department of Applied Science & Humanities, Faculty of Engineering & Technology, University of Lucknow, Lucknow 226031, India.
| |
Collapse
|
3
|
Tan Z, Zhao Y, Lin K, Zhou T. Multi-task pretrained language model with novel application domains enables more comprehensive health and ecological toxicity prediction. JOURNAL OF HAZARDOUS MATERIALS 2024; 477:135265. [PMID: 39038381 DOI: 10.1016/j.jhazmat.2024.135265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/29/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
In silico models for screening substances of healthy and ecological concern are essential for effective chemical management. However, current data-driven toxicity prediction models confront formidable challenges related to expressive capacity, data scarcity, and reliability issues. Thus, this study introduces TOX-BERT, a SMILES-based pretrained model for screening health and ecological toxicity. Results show that masked atom recovery pretraining and multi-task learning offer promising solutions to enhance model capacity and address data scarcity issues. Two novel application domain (AD) parameters, termed PCA-AD and LDS, were proposed to improve prediction reliability of TOX-BERT with accuracy surpassing 90 % and mean absolute error (MAE) below 0.52. TOX-BERT was applied to 18,905 IECSC chemicals, revealing distinct toxicity relationships that align with experimental studies such as those between cardiotoxicity and acute ecotoxicity. In addition to previous PBT screening, 156 potential high-risk chemicals for specific endpoint were identified covering 7 categories. Furthermore, a SMILES-based toxicity site detection approach was developed for structural toxicity analysis. These advancements carry profound implications to address challenges faced by current data-driven toxicity prediction models. TOX-BERT emerges as a valuable tool for more comprehensive, reliable, and applicable predictions of health and ecological toxicity in chemical risk assessment and management.
Collapse
Affiliation(s)
- Zhichao Tan
- The State Key Laboratory of Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai 200092, China; Shanghai Institute of Pollution Control and Ecological Security, 1515 North Zhongshan Rd. (No. 2), Shanghai 200092, PR China.
| | - Youcai Zhao
- The State Key Laboratory of Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai 200092, China; Shanghai Institute of Pollution Control and Ecological Security, 1515 North Zhongshan Rd. (No. 2), Shanghai 200092, PR China.
| | - Kunsen Lin
- The State Key Laboratory of Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai 200092, China; Shanghai Institute of Pollution Control and Ecological Security, 1515 North Zhongshan Rd. (No. 2), Shanghai 200092, PR China.
| | - Tao Zhou
- The State Key Laboratory of Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai 200092, China; Shanghai Institute of Pollution Control and Ecological Security, 1515 North Zhongshan Rd. (No. 2), Shanghai 200092, PR China.
| |
Collapse
|
4
|
Yu J, Liu JM, Chen HY, Xiong WM. Interaction mechanism of oseltamivir phosphate with bovine serum albumin: multispectroscopic and molecular docking study. BMC Chem 2024; 18:126. [PMID: 38970054 PMCID: PMC11227190 DOI: 10.1186/s13065-024-01232-0] [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: 02/05/2024] [Accepted: 06/24/2024] [Indexed: 07/07/2024] Open
Abstract
Oseltamivir phosphate (OP) is an antiviral drug with potential risks to human health due to overuse, leading to serious consequences such as gastrointestinal disturbances, abnormal neuropsychiatric symptoms, and sudden death. Therefore, gaining an in-depth understanding of its interaction with proteins is crucial. We investigated the interaction between OP and bovine serum albumin (BSA) utilizing multispectral methods (i.e., fluorescence, ultraviolet absorption, circular dichroism) combined with molecular docking techniques. Fluorescence spectroscopy indicated that OP quenched BSA fluorescence by forming the OP-BSA complex. The Stern-Volmer constants (KSV) between OP and BSA were determined to be 3.06 × 103 L/mol, 2.36 × 103 L/mol, and 1.86 × 103 L/mol at 293 K, 298 K, and 303 K, respectively. OP occupies exclusively one binding site on BSA, and the fluorescent probe displacement measurements revealed that this is BSA site I. Thermodynamic data (∆H, ∆S, and ∆G) obtained by fitting the van't Hoff equation were - 77.49 kJ/mol, -176.54 J/(mol∙K), and - 24.88 kJ/mol, respectively, suggesting that hydrogen bonding and van der Waals forces mainly participate in OP-BSA complex stabilization. Moreover, the reaction occurs spontaneously at room temperature. Synchronous fluorescence spectra indicated that OP interacts with tryptophan residue of BSA. The results of ultraviolet (UV) and 3D fluorescence spectroscopy indicated that the OP-BSA complex formation altered the microenvironment around amino acid residues. Circular dichroism spectra revealed that the addition of OP decreased the α-helix content of BSA by 7.13%. Docking analysis confirmed that OP binds to BSA site I through hydrogen bonding with amino acids VAL342, SER453, and ASP450. Finally, ADMET studies were conducted to explore the pharmacokinetics of OP as an antiviral drug.
Collapse
Affiliation(s)
- Jing Yu
- School of Chemistry and Civil Engineering, Shaoguan University, Shaoguan, 512005, China
| | - Jian-Ming Liu
- School of Chemistry and Civil Engineering, Shaoguan University, Shaoguan, 512005, China
| | - Hui-Yi Chen
- School of Chemistry and Civil Engineering, Shaoguan University, Shaoguan, 512005, China
| | - Wei-Ming Xiong
- School of Physical Science & Technology, Guangxi University, Nanning, 530004, China.
| |
Collapse
|
5
|
Syaifie PH, Ibadillah D, Jauhar MM, Reninta R, Ningsih S, Ramadhan D, Arda AG, Ningrum DWC, Kaswati NMN, Rochman NT, Mardliyati E. Phytochemical Profile, Antioxidant, Enzyme Inhibition, Acute Toxicity, In Silico Molecular Docking and Dynamic Analysis of Apis Mellifera Propolis as Antidiabetic Supplement. Chem Biodivers 2024; 21:e202400433. [PMID: 38584139 DOI: 10.1002/cbdv.202400433] [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: 02/19/2024] [Revised: 03/31/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
This study aims to identify the phytochemical profile of Apis mellifera propolis and explore the potential of its anti-diabetic activity through inhibition of α-amylase (α-AE), α-glucosidase(α-GE), as well as novel antidiabetic compounds of propolis. Apis mellifera propolis extract (AMPE) exhibited elevated polyphenol 33.26±0.17 (mg GAE/g) and flavonoid (15.45±0.13 mg RE/g). It also indicated moderate strong antioxidant activity (IC50 793.09±1.94 μg/ml). This study found that AMPE displayed promising α-AE and α-GE inhibition through in vitro study. Based on LC-MS/MS screening, 18 unique AMPE compounds were identified, with majorly belonging to anthraquinone and flavonoid compounds. Furthermore, in silico study determined that 8 compounds of AMPE exhibited strong binding to α-AE that specifically interacted with its catalytic residue of ASP197. Moreover, 2 compounds exhibit potential inhibition of α-GE, by interacting with crucial amino acids of ARG315, ASP352, and ASP69. Finally, we suggested that 2,7-Dihydroxy-1-(p-hydroxybenzyl)-4-methoxy-9,10-dihydrophenanthrene and 3(3-(3,4-Dihydroxybenzyl)-7-hydroxychroman-4-one as novel inhibitors of α-AE and α-GE. Notably, these compounds were initially discovered from Apis mellifera propolis in this study. The molecular dynamic analysis confirmed their stable binding with both enzymes over 100 ns simulations. The in vivo acute toxicity assay reveals AMPE as a practically non-toxic product with an LD50 value of 16,050 mg/kg. Therefore, this propolis may serve as a promising natural product for diabetes mellitus treatment.
Collapse
Affiliation(s)
- Putri Hawa Syaifie
- Center of Excellece Life Sciences, Nano Center Indonesia, Jl. PUSPIPTEK, South Tangerang, 15314, Banten, Indonesia
| | - Delfritama Ibadillah
- Center of Excellece Life Sciences, Nano Center Indonesia, Jl. PUSPIPTEK, South Tangerang, 15314, Banten, Indonesia
| | - Muhammad Miftah Jauhar
- Center of Excellece Life Sciences, Nano Center Indonesia, Jl. PUSPIPTEK, South Tangerang, 15314, Banten, Indonesia
- Biomedical Engineering, Graduate School of Universitas Gadjah Mada, Sleman, 55281, Yogyakarta, Indonesia
| | - Rikania Reninta
- Research Center for Applied Botany, National Research and Innovation Agency (BRIN), Cibinong, 16911, Indonesia
| | - Sri Ningsih
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), Cibinong, 16911, Indonesia
| | - Donny Ramadhan
- Center of Excellece Life Sciences, Nano Center Indonesia, Jl. PUSPIPTEK, South Tangerang, 15314, Banten, Indonesia
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), Cibinong, 16911, Indonesia
| | - Adzani Gaisani Arda
- Center of Excellece Life Sciences, Nano Center Indonesia, Jl. PUSPIPTEK, South Tangerang, 15314, Banten, Indonesia
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, H-4032, Hungary
| | - Dhecella Winy Cintya Ningrum
- Center of Excellece Life Sciences, Nano Center Indonesia, Jl. PUSPIPTEK, South Tangerang, 15314, Banten, Indonesia
| | - Nofa Mardia Ningsih Kaswati
- Center of Excellece Life Sciences, Nano Center Indonesia, Jl. PUSPIPTEK, South Tangerang, 15314, Banten, Indonesia
| | - Nurul Taufiqu Rochman
- Research Center for Advanced Materials, National Research and Innovation Agency (BRIN), South Tangerang, 15314, Indonesia
| | - Etik Mardliyati
- Research Center for Vaccine and Drugs, National Research and Innovation Agency (BRIN), Cibinong, 16911, Indonesia
| |
Collapse
|
6
|
Schieferdecker S, Rottach F, Vock E. In Silico Prediction of Oral Acute Rodent Toxicity Using Consensus Machine Learning. J Chem Inf Model 2024; 64:3114-3122. [PMID: 38498695 DOI: 10.1021/acs.jcim.4c00056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
Acute oral toxicity (AOT) is required for the classification and labeling of chemicals according to the global harmonized system (GHS). Acute oral toxicity studies are optimized to minimize the use of animals. However, with the advent of the three Rs principles and machine learning in toxicology, alternative in silico methods became a reasonable alternative approach for addressing the AOT of new chemical matter. Here, we describe the compilation of AOT data from a commercial database and the development of a consensus classification model after evaluating different combinations of molecular representations and machine learning algorithms. The model shows significantly better performance compared to publicly available AOT models. Its performance was evaluated on an external validation data set, which was compiled from the literature, and an applicability domain was deduced.
Collapse
Affiliation(s)
| | - Florian Rottach
- Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach, Germany
| | - Esther Vock
- Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach, Germany
| |
Collapse
|
7
|
Sonker P, Tamang R, Mehata AK, Nidhar M, Sharma VP, Kumar V, Muthu MS, Koch B, Tewari AK. PTSA-induced synthesis, in silico and nano study of novel ethylquinolin-thiazolo-triazole in cervical cancer. Future Med Chem 2024; 16:751-767. [PMID: 38596902 PMCID: PMC11221538 DOI: 10.4155/fmc-2023-0344] [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: 11/24/2023] [Accepted: 02/23/2024] [Indexed: 04/11/2024] Open
Abstract
Aim: p-Toluenesulfonic acid-(PTSA) and grinding-induced novel synthesis of ethylquinolin-thiazolo-triazole derivatives was performed using green chemistry. Materials & methods: Development of a nanoconjugate drug-delivery system of ethylquinolin-thiazolo-triazole was carried out with D-α-tocopheryl polyethylene glycol succinate (TPGS) and the formulation was further characterized by transmission electron microscopy, atomic force microscopy, dynamic light scattering and in vitro drug release assay. The effect of 3a nanoparticles was assessed against a cervical cancer cell line (HeLa) through the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, and the effect on apoptosis was determined. Results & discussion: The 3a nanoparticles triggered the apoptotic mode of cell death after increasing the intracellular reactive oxygen level by enhancing cellular uptake of micelles. Furthermore, in silico studies revealed higher absorption, distribution, metabolism, elimination and toxicity properties and bioavailability of the enzyme tyrosine protein kinase. Conclusion: The 3a nanoparticles enhanced the therapeutic potential and have higher potential for targeted drug delivery against cervical cancer.
Collapse
Affiliation(s)
- Priyanka Sonker
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Rupen Tamang
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Abhishesh K Mehata
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, Uttar Pradesh, India
| | - Manisha Nidhar
- Amrita school of pharmacy, Amrita Vishwa Vidhyapeetham, AIMS, Health Science Campus, Kochi, 682041, India
| | - Vishal P Sharma
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Vipin Kumar
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Madaswamy S Muthu
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, Uttar Pradesh, India
| | - Biplob Koch
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Ashish K Tewari
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| |
Collapse
|
8
|
Zhang R, Wang B, Li L, Li S, Guo H, Zhang P, Hua Y, Cui X, Li Y, Mu Y, Huang X, Li X. Modeling and insights into the structural characteristics of endocrine-disrupting chemicals. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 263:115251. [PMID: 37451095 DOI: 10.1016/j.ecoenv.2023.115251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 07/03/2023] [Accepted: 07/09/2023] [Indexed: 07/18/2023]
Abstract
Endocrine-disrupting chemicals (EDCs) can cause serious harm to human health and the environment; therefore, it is important to rapidly and correctly identify EDCs. Different computational models have been proposed for the prediction of EDCs over the past few decades, but the reported models are not always easily available, and few studies have investigated the structural characteristics of EDCs. In the present study, we have developed a series of artificial intelligence models targeting EDC receptors: the androgen receptor (AR); estrogen receptor (ER); and pregnane X receptor (PXR). The consensus models achieved good predictive results for validation sets with balanced accuracy values of 87.37%, 90.13%, and 79.21% for AR, ER, and PXR binding assays, respectively. Analysis of the physical-chemical properties suggested that several chemical properties were significantly (p < 0.05) different between EDCs and non-EDCs. We also identified structural alerts that can indicate an EDC, which were integrated into the web server SApredictor. These models and structural characteristics can provide useful tools and information in the discrimination and mechanistic understanding of EDCs in drug discovery and environmental risk assessment.
Collapse
Affiliation(s)
- Ruiqiu Zhang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan 250014, China
| | - Bailun Wang
- Department of Anesthesiology and perioperative medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Anesthesia and Respiratory Intensive Care Medicine, Jinan 250014, China
| | - Ling Li
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan 250014, China
| | - Shengjie Li
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
| | - Huizhu Guo
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan 250014, China
| | - Pei Zhang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan 250014, China
| | - Yuqing Hua
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan 250014, China
| | - Xueyan Cui
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan 250014, China
| | - Yan Li
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan 250014, China
| | - Yan Mu
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan 250014, China
| | - Xin Huang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan 250014, China
| | - Xiao Li
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan 250014, China.
| |
Collapse
|
9
|
Hu Y, Ren Q, Liu X, Gao L, Xiao L, Yu W. In Silico Prediction of Human Organ Toxicity via Artificial Intelligence Methods. Chem Res Toxicol 2023. [PMID: 37300507 DOI: 10.1021/acs.chemrestox.2c00411] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Unpredicted human organ level toxicity remains one of the major reasons for drug clinical failure. There is a critical need for cost-efficient strategies in the early stages of drug development for human toxicity assessment. At present, artificial intelligence methods are popularly regarded as a promising solution in chemical toxicology. Thus, we provided comprehensive in silico prediction models for eight significant human organ level toxicity end points using machine learning, deep learning, and transfer learning algorithms. In this work, our results showed that the graph-based deep learning approach was generally better than the conventional machine learning models, and good performances were observed for most of the human organ level toxicity end points in this study. In addition, we found that the transfer learning algorithm could improve model performance for skin sensitization end point using source domain of in vivo acute toxicity data and in vitro data of the Tox21 project. It can be concluded that our models can provide useful guidance for the rapid identification of the compounds with human organ level toxicity for drug discovery.
Collapse
Affiliation(s)
- Yuxuan Hu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Qiuhan Ren
- School of Science, China Pharmaceutical University, Nanjing 211198, China
| | - Xintong Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Liming Gao
- School of Science, China Pharmaceutical University, Nanjing 211198, China
| | - Lecheng Xiao
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Wenying Yu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
10
|
Tan Z, Zhao Y, Zhou T, Lin K. Hi-MGT: A hybrid molecule graph transformer for toxicity identification. JOURNAL OF HAZARDOUS MATERIALS 2023; 457:131808. [PMID: 37307723 DOI: 10.1016/j.jhazmat.2023.131808] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/18/2023] [Accepted: 06/07/2023] [Indexed: 06/14/2023]
Abstract
Conventional toxicity testing methods that rely on animal experimentation are resource-intensive, time-consuming, and ethically controversial. Therefore, the development of alternative non-animal testing approaches is crucial. This study proposes a novel hybrid graph transformer architecture, termed Hi-MGT, for the toxicity identification. An innovative aggregation strategy, referred to as GNN-GT combination, enables Hi-MGT to simultaneously and comprehensively aggregate local and global structural information of molecules, thus elucidating more informative toxicity information hidden in molecule graphs. The results show that the state-of-the-art model outperforms current baseline CML and DL models on a diverse range of toxicity endpoints and is even comparable to large-scale pretrained GNNs with geometry enhancement. Additionally, the impact of hyperparameters on model performance is investigated, and a systematic ablation study is conducted to demonstrate the effectiveness of the GNN-GT combination. Moreover, this study provides valuable insights into the learning process on molecules and proposes a novel similarity-based method for toxic site detection, which could potentially facilitate toxicity identification and analysis. Overall, the Hi-MGT model represents a significant advancement in the development of alternative non-animal testing approaches for toxicity identification, with promising implications for enhancing human safety in the use of chemical compounds.
Collapse
Affiliation(s)
- Zhichao Tan
- The State Key Laboratory of Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai 200092, China; Shanghai Institute of Pollution Control and Ecological Security, 1515 North Zhongshan Rd. (No. 2), Shanghai 200092, China
| | - Youcai Zhao
- The State Key Laboratory of Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai 200092, China; Shanghai Institute of Pollution Control and Ecological Security, 1515 North Zhongshan Rd. (No. 2), Shanghai 200092, China
| | - Tao Zhou
- The State Key Laboratory of Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai 200092, China; Shanghai Institute of Pollution Control and Ecological Security, 1515 North Zhongshan Rd. (No. 2), Shanghai 200092, China.
| | - Kunsen Lin
- The State Key Laboratory of Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai 200092, China; Shanghai Institute of Pollution Control and Ecological Security, 1515 North Zhongshan Rd. (No. 2), Shanghai 200092, China.
| |
Collapse
|
11
|
Sharma B, Chenthamarakshan V, Dhurandhar A, Pereira S, Hendler JA, Dordick JS, Das P. Accurate clinical toxicity prediction using multi-task deep neural nets and contrastive molecular explanations. Sci Rep 2023; 13:4908. [PMID: 36966203 PMCID: PMC10039880 DOI: 10.1038/s41598-023-31169-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 03/07/2023] [Indexed: 03/27/2023] Open
Abstract
Explainable machine learning for molecular toxicity prediction is a promising approach for efficient drug development and chemical safety. A predictive ML model of toxicity can reduce experimental cost and time while mitigating ethical concerns by significantly reducing animal and clinical testing. Herein, we use a deep learning framework for simultaneously modeling in vitro, in vivo, and clinical toxicity data. Two different molecular input representations are used; Morgan fingerprints and pre-trained SMILES embeddings. A multi-task deep learning model accurately predicts toxicity for all endpoints, including clinical, as indicated by the area under the Receiver Operator Characteristic curve and balanced accuracy. In particular, pre-trained molecular SMILES embeddings as input to the multi-task model improved clinical toxicity predictions compared to existing models in MoleculeNet benchmark. Additionally, our multitask approach is comprehensive in the sense that it is comparable to state-of-the-art approaches for specific endpoints in in vitro, in vivo and clinical platforms. Through both the multi-task model and transfer learning, we were able to indicate the minimal need of in vivo data for clinical toxicity predictions. To provide confidence and explain the model's predictions, we adapt a post-hoc contrastive explanation method that returns pertinent positive and negative features, which correspond well to known mutagenic and reactive toxicophores, such as unsubstituted bonded heteroatoms, aromatic amines, and Michael receptors. Furthermore, toxicophore recovery by pertinent feature analysis captures more of the in vitro (53%) and in vivo (56%), rather than of the clinical (8%), endpoints, and indeed uncovers a preference in known toxicophore data towards in vitro and in vivo experimental data. To our knowledge, this is the first contrastive explanation, using both present and absent substructures, for predictions of clinical and in vivo molecular toxicity.
Collapse
Affiliation(s)
| | | | | | - Shiranee Pereira
- ICARE, International Center for Alternatives in Research and Education, Chennai, India
| | | | | | - Payel Das
- IBM Research, Yorktown Heights, NY, USA.
| |
Collapse
|
12
|
Thirunavukkarasu M, Prabakaran P, Saral A, Alharbi NS, Kadaikunnan S, Kazachenko AS, Muthu S. Molecular level Solvent interaction (microscopic), Electronic, Covalent assembly (RDG, AIM & ELF), ADMET prediction and anti-cancer activity of 1-(4-Fluorophenyl)-1-propanone): cytotoxic agent. J Mol Liq 2023. [DOI: 10.1016/j.molliq.2023.121714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2023]
|
13
|
Al-Shuaeeb RAA, Yassin AA, Ibrahim MAA, Abd El-Mageed HR, Ghandour MA, Khalil MM. Computer-based identification of olive oil components as a potential inhibitor of neirisaral adhesion a regulatory protein. J Biomol Struct Dyn 2023; 41:1553-1560. [PMID: 34974817 DOI: 10.1080/07391102.2021.2022535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In silico methods such as molecular docking and molecular dynamic (MD) simulations have significant interest due to their ability to identify the protein-ligand interactions at the atomic level. In this work, different computational methods were used to elucidate the ability of some olive oil components to act as Neisseria adhesion A Regulatory protein (NadR) inhibitors. The frontier molecular orbitals (FMOs) and the global properties such as global hardness, electronegativity, and global softness of ten olive oil components (α-Tocopherol, Erythrodiol, Hydroxytyrosol, Linoleic acid, Apigenin, Luteolin, Oleic acid, Oleocanthal, Palmitic acid, and Tyrosol) were reported using Density Functional Theory (DFT) methods. Among all investigated compounds, Erythrodiol, Apigenin, and Luteolin demonstrated the highest binding affinities (-8.72, -7.12, and -8.24 kcal/mol, respectively) against NadR, compared to -8.21 kcal/mol of the native ligand based on molecular docking calculations. ADMET properties and physicochemical features showed that Erythrodiol, Apigenin, and Luteolin have good physicochemical features and can act as drugs candidate. Molecular dynamics (MD) simulations demonstrated that Erythrodiol, Apigenin, and Luteolin show stable binding affinity and molecular interaction with NadR. Further Molecular Mechanics Poisson-Boltzmann Surface Area (MM-PBSA) analyses using the MD trajectories also demonstrated the higher binding affinity of Erythrodiol, Apigenin and Luteolin inside NadR protein. The overall study provides a rationale to use Erythrodiol, Apigenin, and Luteolin in the drug development as anti-adhesive drugs lead. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | - A A Yassin
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Mahmoud A A Ibrahim
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, Egypt
| | - H R Abd El-Mageed
- Micro-Analysis, Environmental Research and Community Affairs Center (MAESC), Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - M A Ghandour
- Chemistry Department, Faculty of Science, Assuit University, Asyut, Egypt
| | - M M Khalil
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
14
|
Lane TR, Harris J, Urbina F, Ekins S. Comparing LD 50/LC 50 Machine Learning Models for Multiple Species. ACS CHEMICAL HEALTH & SAFETY 2023. [DOI: 10.1021/acs.chas.2c00088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Affiliation(s)
- Thomas R. Lane
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Joshua Harris
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Fabio Urbina
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| |
Collapse
|
15
|
Li X, Liu G, Wang Z, Zhang L, Liu H, Ai H. Ensemble multiclassification model for aquatic toxicity of organic compounds. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 255:106379. [PMID: 36587517 DOI: 10.1016/j.aquatox.2022.106379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 12/04/2022] [Accepted: 12/19/2022] [Indexed: 06/17/2023]
Abstract
With environmental pollution becoming increasingly serious, organic compounds have become the main hazard of environmental pollution and exert substantial negative impacts on aquatic organisms. In research pertaining to the acute toxicity of organic compounds, traditional biological experimental methods are time-consuming and expensive. In addition, computer-aided binary classification models cannot accurately classify acute toxicity. Therefore, the multiclassication model is necessary for more accurate classification of acute toxicity. In this study, median lethal concentrations of 373 organic compounds in the environmental toxicology datasets ECOTOX and EAT5 were used. These chemicals were classified into four categories based on the European Economic Community criteria. Then the random forest, support vector machine, extreme gradient boosting, adaptive gradient boosting, and C5.0 decision tree algorithms and eight molecular fingerprints were used to build a multiclassification base model for the acute toxicity of organic compounds. The base models were repeated 100 times with fivefold cross-validation and external validation. The ensemble model was obtained by the voting method. The best base classifier was ExtendFP-C5.0, which had an accuracy, sensitivity and specificity values of 87.30%, 87.32% and 95.76% for external validation, and the voting ensemble model performance of 96.92%, 96.93% and 98.97%, respectively. The ensemble model achieved a higher accuracy than previously reported studies. Our study will help to further classify the acute toxicity of organic compounds to aquatic organisms and predict the hazard classes of organic compounds.
Collapse
Affiliation(s)
- Xinran Li
- College of Life Science, Liaoning University, Shenyang, 110036, China
| | - Gaohua Liu
- College of Life Science, Liaoning University, Shenyang, 110036, China
| | - Zhibo Wang
- College of Life Science, Liaoning University, Shenyang, 110036, China
| | - Li Zhang
- College of Life Science, Liaoning University, Shenyang, 110036, China; China Research Center for Computer Simulating and Information Processing of Bio-macromolecules of Shenyang, China
| | - Hongsheng Liu
- China Research Center for Computer Simulating and Information Processing of Bio-macromolecules of Shenyang, China; College of Pharmacy, Liaoning University, Shenyang, 110036, China
| | - Haixin Ai
- College of Life Science, Liaoning University, Shenyang, 110036, China; China Research Center for Computer Simulating and Information Processing of Bio-macromolecules of Shenyang, China.
| |
Collapse
|
16
|
Wang Y, Huang M, Deng H, Li W, Wu Z, Tang Y, Liu G. Identification of vital chemical information via visualization of graph neural networks. Brief Bioinform 2023; 24:6936421. [PMID: 36537081 DOI: 10.1093/bib/bbac577] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/02/2022] [Accepted: 11/25/2022] [Indexed: 12/24/2022] Open
Abstract
Qualitative or quantitative prediction models of structure-activity relationships based on graph neural networks (GNNs) are prevalent in drug discovery applications and commonly have excellently predictive power. However, the network information flows of GNNs are highly complex and accompanied by poor interpretability. Unfortunately, there are relatively less studies on GNN attributions, and their developments in drug research are still at the early stages. In this work, we adopted several advanced attribution techniques for different GNN frameworks and applied them to explain multiple drug molecule property prediction tasks, enabling the identification and visualization of vital chemical information in the networks. Additionally, we evaluated them quantitatively with attribution metrics such as accuracy, sparsity, fidelity and infidelity, stability and sensitivity; discussed their applicability and limitations; and provided an open-source benchmark platform for researchers. The results showed that all attribution techniques were effective, while those directly related to the predicted labels, such as integrated gradient, preferred to have better attribution performance. These attribution techniques we have implemented could be directly used for the vast majority of chemical GNN interpretation tasks.
Collapse
Affiliation(s)
- Yimeng Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Mengting Huang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Hua Deng
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Weihua Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Zengrui Wu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yun Tang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Guixia Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| |
Collapse
|
17
|
Guerraoui A, Goudjil M, Direm A, Guerraoui A, Şengün İY, Parlak C, Djedouani A, Chelazzi L, Monti F, Lunedei E, Boumaza A. A rhodanine derivative as a potential antibacterial and anticancer agent: crystal structure, spectral characterization, DFT calculations, Hirshfeld surface analysis, in silico molecular docking and ADMET studies. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.135025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
18
|
Elseginy SA. Virtual screening and structure-based 3D pharmacophore approach to identify small-molecule inhibitors of SARS-CoV-2 M pro. J Biomol Struct Dyn 2022; 40:13658-13674. [PMID: 34676801 DOI: 10.1080/07391102.2021.1993341] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The outbreak caused by a coronavirus 2 has required quick and potential treatment strategies. The main protease enzyme Mpro plays an important role in the viral replication which renders it an important target for discovering SARS-CoV-2 inhibitors. In this study, 3D pharmacophore structure-based virtual screening and molecular docking were conducted using MOE and Bristol University Docking Engine (BUDE). Around 400,000 molecules of ZINC15 database were docked against the crystal structure of main protease, followed by 3D pharmacophore filtration. Six top-ranked hits (ZINC58717986, ZINC60399606, ZINC58662884, ZINC45988635, ZINC54706757 and ZINC17320595) were identified based on their strong spatial affinity and forming H-bonds with key residues H41, E166, Q189 and T190 of the binding pocket of Mpro SARS-CoV-2. The 6 hits subjected to molecular dynamics simulations for 100 ns followed by binding free energy calculations using MM-PBSA technique. Interestingly, three hits showed free binding energy (ΔGbinding) lower than tert-butyl N-[1-[(2S)-1-[[(2S)-4-(benzylamino)-3,4-dioxo-1-[(3S)-2-oxopyrrolidin-3-yl]butan-2-yl]amino]-3-cyclopropyl-1-oxopropan-2-yl]-2-oxopyridin-3-yl]carbamate (α-ketoamide 13 b) (ΔGbinding) -76.67 ± 0.5 kJ/mol which suggested their potential against SARS-CoV-2. The best binding free energy candidates, ZINC58717986 (ΔGbinding) -98.41 ± 0.7 kJ/mol. The second-best hit candidate, ZINC54706757 (ΔGbinding) -83.4 ± 0.6 kJ/mol and the third one ZINC17320595 (ΔGbinding) -78.85 ± 0.5 kJ/mol. Per residue decomposition free energy indicates H41, S46, H164, E166, D187, Q189 and Q192 are hot spot residues while residues M49, M165, L167 and P168 contribute to the hydrophobic interactions. The pharmacokinetic study suggests that the selected 6 hits possess drug-like properties. The 3D pharmacophore virtual screening, molecular dynamics and MM-PBSA approaches facilitated identification 3 promising hits with low free binding energy as SARS-CoV-2 inhibitors.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Samia A Elseginy
- Molecular Modelling Lab., Biochemistry School, Bristol University, Bristol, UK.,Green Chemistry Department, Chemical Industries Research Division, National Research Centre, Egypt
| |
Collapse
|
19
|
Cavasotto CN, Scardino V. Machine Learning Toxicity Prediction: Latest Advances by Toxicity End Point. ACS OMEGA 2022; 7:47536-47546. [PMID: 36591139 PMCID: PMC9798519 DOI: 10.1021/acsomega.2c05693] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/28/2022] [Indexed: 05/29/2023]
Abstract
Machine learning (ML) models to predict the toxicity of small molecules have garnered great attention and have become widely used in recent years. Computational toxicity prediction is particularly advantageous in the early stages of drug discovery in order to filter out molecules with high probability of failing in clinical trials. This has been helped by the increase in the number of large toxicology databases available. However, being an area of recent application, a greater understanding of the scope and applicability of ML methods is still necessary. There are various kinds of toxic end points that have been predicted in silico. Acute oral toxicity, hepatotoxicity, cardiotoxicity, mutagenicity, and the 12 Tox21 data end points are among the most commonly investigated. Machine learning methods exhibit different performances on different data sets due to dissimilar complexity, class distributions, or chemical space covered, which makes it hard to compare the performance of algorithms over different toxic end points. The general pipeline to predict toxicity using ML has already been analyzed in various reviews. In this contribution, we focus on the recent progress in the area and the outstanding challenges, making a detailed description of the state-of-the-art models implemented for each toxic end point. The type of molecular representation, the algorithm, and the evaluation metric used in each research work are explained and analyzed. A detailed description of end points that are usually predicted, their clinical relevance, the available databases, and the challenges they bring to the field are also highlighted.
Collapse
Affiliation(s)
- Claudio N. Cavasotto
- Computational
Drug Design and Biomedical Informatics Laboratory, Instituto de Investigaciones
en Medicina Traslacional (IIMT), CONICET-Universidad
Austral, Pilar, B1629AHJ Buenos Aires, Argentina
- Austral
Institute for Applied Artificial Intelligence, Universidad Austral, Pilar, B1629AHJ Buenos Aires, Argentina
- Facultad
de Ciencias Biomédicas, Facultad de Ingenierá, Universidad Austral, Pilar, B1630FHB Buenos
Aires, Argentina
| | - Valeria Scardino
- Austral
Institute for Applied Artificial Intelligence, Universidad Austral, Pilar, B1629AHJ Buenos Aires, Argentina
- Meton
AI, Inc., Wilmington, Delaware 19801, United
States
| |
Collapse
|
20
|
Basnet S, Marahatha R, Shrestha A, Bhattarai S, Katuwal S, Sharma KR, Marasini BP, Dahal SR, Basnyat RC, Patching SG, Parajuli N. In Vitro and In Silico Studies for the Identification of Potent Metabolites of Some High-Altitude Medicinal Plants from Nepal Inhibiting SARS-CoV-2 Spike Protein. Molecules 2022; 27:8957. [PMID: 36558090 PMCID: PMC9786757 DOI: 10.3390/molecules27248957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/01/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Despite ongoing vaccination programs against COVID-19 around the world, cases of infection are still rising with new variants. This infers that an effective antiviral drug against COVID-19 is crucial along with vaccinations to decrease cases. A potential target of such antivirals could be the membrane components of the causative pathogen, SARS-CoV-2, for instance spike (S) protein. In our research, we have deployed in vitro screening of crude extracts of seven ethnomedicinal plants against the spike receptor-binding domain (S1-RBD) of SARS-CoV-2 using an enzyme-linked immunosorbent assay (ELISA). Following encouraging in vitro results for Tinospora cordifolia, in silico studies were conducted for the 14 reported antiviral secondary metabolites isolated from T. cordifolia-a species widely cultivated and used as an antiviral drug in the Himalayan country of Nepal-using Genetic Optimization for Ligand Docking (GOLD), Molecular Operating Environment (MOE), and BIOVIA Discovery Studio. The molecular docking and binding energy study revealed that cordifolioside-A had a higher binding affinity and was the most effective in binding to the competitive site of the spike protein. Molecular dynamics (MD) simulation studies using GROMACS 5.4.1 further assayed the interaction between the potent compound and binding sites of the spike protein. It revealed that cordifolioside-A demonstrated better binding affinity and stability, and resulted in a conformational change in S1-RBD, hence hindering the activities of the protein. In addition, ADMET analysis of the secondary metabolites from T. cordifolia revealed promising pharmacokinetic properties. Our study thus recommends that certain secondary metabolites of T. cordifolia are possible medicinal candidates against SARS-CoV-2.
Collapse
Affiliation(s)
- Saroj Basnet
- Center for Drug Design and Molecular Simulation Division, Kathmandu 44600, Nepal
| | - Rishab Marahatha
- Central Department of Chemistry, Tribhuvan University, Kathmandu 44618, Nepal
- Department of Chemistry, Oklahoma State University, Still Water, OK 74078, USA
| | - Asmita Shrestha
- Central Department of Chemistry, Tribhuvan University, Kathmandu 44618, Nepal
| | - Salyan Bhattarai
- Paraza Pharma, Inc., 2525 Avenue Marie-Curie, Montreal, QC H4S 2E1, Canada
| | - Saurav Katuwal
- Central Department of Chemistry, Tribhuvan University, Kathmandu 44618, Nepal
| | - Khaga Raj Sharma
- Central Department of Chemistry, Tribhuvan University, Kathmandu 44618, Nepal
| | | | - Salik Ram Dahal
- Department of Chemistry, Oklahoma State University, Still Water, OK 74078, USA
- Oakridge National Laboratory, Bethel Valley Rd, Oak Ridge, TN 37830, USA
| | - Ram Chandra Basnyat
- Central Department of Chemistry, Tribhuvan University, Kathmandu 44618, Nepal
| | | | - Niranjan Parajuli
- Central Department of Chemistry, Tribhuvan University, Kathmandu 44618, Nepal
| |
Collapse
|
21
|
Zhao X, Sun Y, Zhang R, Chen Z, Hua Y, Zhang P, Guo H, Cui X, Huang X, Li X. Machine Learning Modeling and Insights into the Structural Characteristics of Drug-Induced Neurotoxicity. J Chem Inf Model 2022; 62:6035-6045. [PMID: 36448818 DOI: 10.1021/acs.jcim.2c01131] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Neurotoxicity can be resulted from many diverse clinical drugs, which has been a cause of concern to human populations across the world. The detection of drug-induced neurotoxicity (DINeurot) potential with biological experimental methods always required a lot of budget and time. In addition, few studies have addressed the structural characteristics of neurotoxic chemicals. In this study, we focused on the computational modeling for drug-induced neurotoxicity with machine learning methods and the insights into the structural characteristics of neurotoxic chemicals. Based on the clinical drug data with neurotoxicity effects, we developed 35 different classifiers by combining five different machine learning methods and seven fingerprint packages. The best-performing model achieved good results on both 5-fold cross-validation (balanced accuracy of 76.51%, AUC value of 0.83, and MCC value of 0.52) and external validation (balanced accuracy of 83.63%, AUC value of 0.87, and MCC value of 0.67). The model can be freely accessed on the web server DINeuroTpredictor (http://dineurot.sapredictor.cn/). We also analyzed the distribution of several key molecular properties between neurotoxic and non-neurotoxic structures. The results indicated that several physicochemical properties were significantly different between the neurotoxic and non-neurotoxic compounds, including molecular polar surface area (MPSA), AlogP, the number of hydrogen bond acceptors (nHAcc) and donors (nHDon), the number of rotatable bonds (nRotB), and the number of aromatic rings (nAR). In addition, 18 structural alerts responsible for chemical neurotoxicity were identified. The structural alerts have been integrated with our web server SApredictor (http://www.sapredictor.cn). The results of this study could provide useful information for the understanding of the structural characteristics and computational prediction for chemical neurotoxicity.
Collapse
Affiliation(s)
- Xia Zhao
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, Shandong250014, China
| | - Yuhao Sun
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, Shandong250014, China
| | - Ruiqiu Zhang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, Shandong250014, China
| | - Zhaoyang Chen
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, Shandong250014, China
| | - Yuqing Hua
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, Shandong250014, China
| | - Pei Zhang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, Shandong250014, China
| | - Huizhu Guo
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, Shandong250014, China
| | - Xueyan Cui
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, Shandong250014, China
| | - Xin Huang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, Shandong250014, China
| | - Xiao Li
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, Shandong250014, China
| |
Collapse
|
22
|
Isopropyl Gallate, a Gallic Acid Derivative: In Silico and In Vitro Investigation of Its Effects on Leishmania major. Pharmaceutics 2022; 14:pharmaceutics14122701. [PMID: 36559198 PMCID: PMC9787715 DOI: 10.3390/pharmaceutics14122701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/30/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Isopropyl gallate (IPG) is a polyphenol obtained from alterations in the gallic acid molecule via acid catalysis with previously reported leishmanicidal and trypanocidal activities. The present study aims to evaluate in silico binding activity towards some targets for antileishmanial chemotherapy against Leishmania major species, and ADMET parameters for IPG, as well as in vitro antileishmanial and cytotoxic effects. Molecular docking was performed using AutoDockVina and BIOVIA Discovery Studio software, whereas in silico analysis used SwissADME, PreADMET and admetSAR software. In vitro antileishmanial activity on promastigotes and amastigotes of Leishmania major, cytotoxicity and macrophages activation were assessed. IPG exhibited affinity for pteridine reductase (PTR1; -8.2 kcal/mol) and oligopeptidase B (OPB; -8.0 kcal/mol) enzymes. ADMET assays demonstrated good lipophilicity, oral bioavailability, and skin permeability, as well as non-mutagenic, non-carcinogenic properties and low risk of cardiac toxicity for IPG. Moreover, IPG inhibited the in vitro growth of promastigotes (IC50 = 90.813 µM), presented significant activity against amastigotes (IC50 = 13.45 μM), promoted low cytotoxicity in macrophages (CC50 = 1260 μM), and increased phagocytic capacity. These results suggest IPG is more selectively toxic to the parasite than to mammalian cells. IPG demonstrated acceptable in silico pharmacokinetics parameters, and reduced infection and infectivity in parasitized macrophages, possibly involving macrophage activation pathways and inhibition of leishmania enzymes.
Collapse
|
23
|
Hao Y, Fan T, Sun G, Li F, Zhang N, Zhao L, Zhong R. Environmental toxicity risk evaluation of nitroaromatic compounds: Machine learning driven binary/multiple classification and design of safe alternatives. Food Chem Toxicol 2022; 170:113461. [PMID: 36243219 DOI: 10.1016/j.fct.2022.113461] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/11/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022]
|
24
|
Limbu S, Zakka C, Dakshanamurthy S. Predicting Dose-Range Chemical Toxicity using Novel Hybrid Deep Machine-Learning Method. TOXICS 2022; 10:706. [PMID: 36422913 PMCID: PMC9692315 DOI: 10.3390/toxics10110706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 06/16/2023]
Abstract
Humans are exposed to thousands of chemicals, including environmental chemicals. Unfortunately, little is known about their potential toxicity, as determining the toxicity remains challenging due to the substantial resources required to assess a chemical in vivo. Here, we present a novel hybrid neural network (HNN) deep learning method, called HNN-Tox, to predict chemical toxicity at different doses. To develop a hybrid HNN-Tox method, we combined two neural network frameworks, the Convolutional Neural Network (CNN) and the multilayer perceptron (MLP)-type feed-forward neural network (FFNN). Combining the CNN and FCNN in the field of environmental chemical toxicity prediction is a novel approach. We developed several binary and multiclass classification models to assess dose-range chemical toxicity that is trained based on thousands of chemicals with known toxicity. The performance of the HNN-Tox was compared with other machine-learning methods, including Random Forest (RF), Bootstrap Aggregation (Bagging), and Adaptive Boosting (AdaBoost). We also analyzed the model performance dependency on varying features, descriptors, dataset size, route of exposure, and toxic dose. The HNN-Tox model, trained on 59,373 chemicals annotated with known LD50 and routes of exposure, maintained its predictive ability with an accuracy of 84.9% and 84.1%, even after reducing the descriptor size from 318 to 51, and the area under the ROC curve (AUC) was 0.89 and 0.88, respectively. Further, we validated the HNN-Tox with several external toxic chemical datasets on a large scale. The HNN-Tox performed optimally or better than the other machine-learning methods for diverse chemicals. This study is the first to report a large-scale prediction of dose-range chemical toxicity with varying features. The HNN-Tox has broad applicability in predicting toxicity for diverse chemicals and could serve as an alternative methodology approach to animal-based toxicity assessment.
Collapse
Affiliation(s)
- Sarita Limbu
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Cyril Zakka
- Faculty of Medicine, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon
| | - Sivanesan Dakshanamurthy
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
25
|
Rzepka Z, Bębenek E, Chrobak E, Wrześniok D. Synthesis and Anticancer Activity of Indole-Functionalized Derivatives of Betulin. Pharmaceutics 2022; 14:2372. [PMID: 36365190 PMCID: PMC9694481 DOI: 10.3390/pharmaceutics14112372] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 09/01/2023] Open
Abstract
Pentacyclic triterpenes, including betulin, are widespread natural products with various pharmacological effects. These compounds are the starting material for the synthesis of substances with promising anticancer activity. The chemical modification of the betulin scaffold that was carried out as part of the research consisted of introducing the indole moiety at the C-28 position. The synthesized new 28-indole-betulin derivatives were evaluated for anticancer activity against seven human cancer lines (A549, MDA-MB-231, MCF-7, DLD-1, HT-29, A375, and C32). It was observed that MCF-7 breast cancer cells were most sensitive to the action of the 28-indole-betulin derivatives. The study shows that the lup-20(29)-ene-3-ol-28-yl 2-(1H-indol-3-yl)acetate caused the MCF-7 cells to arrest in the G1 phase, preventing the cells from entering the S phase. The performed cytometric analysis of DNA fragmentation indicates that the mechanism of EB355A action on the MCF-7 cell line is related to the induction of apoptosis. An in silico ADMET profile analysis of EB355A and EB365 showed that both compounds are bioactive molecules characterized by good intestinal absorption. In addition, the in silico studies indicate that the 28-indole-betulin derivatives are substances of relatively low toxicity.
Collapse
Affiliation(s)
- Zuzanna Rzepka
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska Str., 41-200 Sosnowiec, Poland
| | - Ewa Bębenek
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska Str., 41-200 Sosnowiec, Poland
| | - Elwira Chrobak
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska Str., 41-200 Sosnowiec, Poland
| | - Dorota Wrześniok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska Str., 41-200 Sosnowiec, Poland
| |
Collapse
|
26
|
Li L, Lu Z, Liu G, Tang Y, Li W. In Silico Prediction of Human and Rat Liver Microsomal Stability via Machine Learning Methods. Chem Res Toxicol 2022; 35:1614-1624. [PMID: 36053050 DOI: 10.1021/acs.chemrestox.2c00207] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Liver microsomal stability is an important property considered for the screening of drug candidates in the early stage of drug development. Determination of hepatic metabolic stability can be performed by an in vitro assay, but it requires quite a few resources and time. In recent years, machine learning methods have made much progress. Therefore, development of computational models to predict liver microsomal stability is highly desirable in the drug discovery process. In this study, the in silico classification models for the prediction of the metabolic stability of compounds in rat and human liver microsomes were constructed by the conventional machine learning and deep learning methods. The performance of the models was evaluated using the test and external sets. For the rat liver microsomes (RLM) stability, the best model yielded the AUC values of 0.84 and 0.71 on the test and external validation sets, respectively. For the human liver microsome (HLM) stability, the best model exhibited the AUC values of 0.86 and 0.77 on the test and external validation sets, respectively. In addition, several important substructure fragments were detected using information gain and frequency substructure analysis methods. The applicability domain of the models was defined using the Euclidean distance-based method. We anticipate that our results would be helpful for the prediction of liver microsomal stability of compounds in the early stage of drug discovery.
Collapse
Affiliation(s)
- Longqiang Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Zhou Lu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Guixia Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yun Tang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Weihua Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| |
Collapse
|
27
|
Yang P, Henle EA, Fern XZ, Simon CM. Classifying the toxicity of pesticides to honey bees via support vector machines with random walk graph kernels. J Chem Phys 2022; 157:034102. [DOI: 10.1063/5.0090573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Pesticides benefit agriculture by increasing crop yield, quality, and security. However, pesticides may inadvertently harm bees, which are valuable as pollinators. Thus, candidate pesticides in development pipelines must be assessed for toxicity to bees. Leveraging a dataset of 382 molecules with toxicity labels from honey bee exposure experiments, we train a support vector machine (SVM) to predict the toxicity of pesticides to honey bees. We compare two representations of the pesticide molecules: (i) a random walk feature vector listing counts of length- L walks on the molecular graph with each vertex- and edge-label sequence and (ii) the Molecular ACCess System (MACCS) structural key fingerprint (FP), a bit vector indicating the presence/absence of a list of pre-defined subgraph patterns in the molecular graph. We explicitly construct the MACCS FPs but rely on the fixed-length- L random walk graph kernel (RWGK) in place of the dot product for the random walk representation. The L-RWGK-SVM achieves an accuracy, precision, recall, and F1 score (mean over 2000 runs) of 0.81, 0.68, 0.71, and 0.69, respectively, on the test data set—with L = 4 being the mode optimal walk length. The MACCS-FP-SVM performs on par/marginally better than the L-RWGK-SVM, lends more interpretability, but varies more in performance. We interpret the MACCS-FP-SVM by illuminating which subgraph patterns in the molecules tend to strongly push them toward the toxic/non-toxic side of the separating hyperplane.
Collapse
Affiliation(s)
- Ping Yang
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, Oregon 97331, USA
| | - E. Adrian Henle
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, Oregon 97331, USA
| | - Xiaoli Z. Fern
- School of Electrical Engineering and Computer Science, Oregon State University, Corvallis, Oregon 97331, USA
| | - Cory M. Simon
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, Oregon 97331, USA
| |
Collapse
|
28
|
Hua Y, Cui X, Liu B, Shi Y, Guo H, Zhang R, Li X. SApredictor: An Expert System for Screening Chemicals Against Structural Alerts. Front Chem 2022; 10:916614. [PMID: 35910729 PMCID: PMC9326022 DOI: 10.3389/fchem.2022.916614] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
The rapid and accurate evaluation of chemical toxicity is of great significance for estimation of chemical safety. In the past decades, a great number of excellent computational models have been developed for chemical toxicity prediction. But most machine learning models tend to be “black box”, which bring about poor interpretability. In the present study, we focused on the identification and collection of structural alerts (SAs) responsible for a series of important toxicity endpoints. Then, we carried out effective storage of these structural alerts and developed a web-server named SApredictor (www.sapredictor.cn) for screening chemicals against structural alerts. People can quickly estimate the toxicity of chemicals with SApredictor, and the specific key substructures which cause the chemical toxicity will be intuitively displayed to provide valuable information for the structural optimization by medicinal chemists.
Collapse
Affiliation(s)
- Yuqing Hua
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Xueyan Cui
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Bo Liu
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yinping Shi
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Huizhu Guo
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Ruiqiu Zhang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - Xiao Li
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
- Department of Clinical Pharmacy, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
- *Correspondence: Xiao Li, , , orcid.org/0000-0002-1148-9898
| |
Collapse
|
29
|
Jeong J, Choi J. Artificial Intelligence-Based Toxicity Prediction of Environmental Chemicals: Future Directions for Chemical Management Applications. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:7532-7543. [PMID: 35666838 DOI: 10.1021/acs.est.1c07413] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Recently, research on the development of artificial intelligence (AI)-based computational toxicology models that predict toxicity without the use of animal testing has emerged because of the rapid development of computer technology. Various computational toxicology techniques that predict toxicity based on the structure of chemical substances are gaining attention, including the quantitative structure-activity relationship. To understand the recent development of these models, we analyzed the databases, molecular descriptors, fingerprints, and algorithms considered in recent studies. Based on a selection of 96 papers published since 2014, we found that AI models have been developed to predict approximately 30 different toxicity end points using more than 20 toxicity databases. For model development, molecular access system and extended-connectivity fingerprints are the most commonly used molecular descriptors. The most used algorithm among the machine learning techniques is the random forest, while the most used algorithm among the deep learning techniques is a deep neural network. The use of AI technology in the development of toxicity prediction models is a new concept that will aid in achieving a scientific accord and meet regulatory applications. The comprehensive overview provided in this study will provide a useful guide for the further development and application of toxicity prediction models.
Collapse
Affiliation(s)
- Jaeseong Jeong
- School of Environmental Engineering, University of Seoul, 163 Seoulsiripdae-ro, Dongdaemun-gu, Seoul 02504, South Korea
| | - Jinhee Choi
- School of Environmental Engineering, University of Seoul, 163 Seoulsiripdae-ro, Dongdaemun-gu, Seoul 02504, South Korea
| |
Collapse
|
30
|
Xu M, Yang H, Liu G, Tang Y, Li W. In Silico Prediction of Chemical Aquatic Toxicity by Multiple Machine Learning and Deep Learning Approaches. J Appl Toxicol 2022; 42:1766-1776. [PMID: 35653511 DOI: 10.1002/jat.4354] [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: 04/12/2022] [Revised: 05/16/2022] [Accepted: 05/31/2022] [Indexed: 11/08/2022]
Abstract
Fish is one of the model animals used to evaluate the adverse effects of a chemical exposed to the ecosystem. However, its low throughput and relevantly high expense make it impossible to test all new chemicals in manufacture. Hence using in silico models to prioritize compounds to be tested has been widely applied in environmental risk assessment and drug discovery. In this study, we constructed the local predictive models for four fish species, including bluegill sunfish, rainbow trout, fathead minnow, and sheepshead minnow, and the global models with all four fish data. A total of 1874 unique compounds with their labels, i.e. toxic (LC50 < 10 ppm) or nontoxic were collected from ECOTOX and literature. Both conventional machine learning methods and the deep learning architecture, graph convolutional network (GCN), were used to build predictive models. The classification accuracy of the best local model for each fish species was higher than 0.83. For the global models, two strategies including consistency prediction and probability threshold were adopted to improve the predictive capability at the cost of limiting applicability domain. For 63% of compounds in domain, the accuracy was around 0.97. By comparison of the deep learning and machine learning methods, we found that the single-task GCN showed specific advantages in performance and multi-task GCN showed no advantages over the conventional machine learning methods. The data and models are available on GitHub (https://github.com/ChemPredict/ChemicalAquaticToxicity).
Collapse
Affiliation(s)
- Minjie Xu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Hongbin Yang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Guixia Liu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yun Tang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Weihua Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
31
|
Sethi A, Yadav P, Singh RP, Kumar S, Parveen S, Singh A, Yadav A, Banerjee M. Pregnenolone derivatives as potential anti‐lung cancer agents: A combined in silico and in vitro approach. J CHIN CHEM SOC-TAIP 2022. [DOI: 10.1002/jccs.202200040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Arun Sethi
- Department of Chemistry University of Lucknow Lucknow India
| | - Priyanka Yadav
- Department of Chemistry University of Lucknow Lucknow India
| | - Ranvijay Pratap Singh
- Department of Applied Science & Humanities, Faculty of Engineering & Technology University of Lucknow Lucknow India
| | - Saurabh Kumar
- Department of Zoology University of Lucknow Lucknow India
| | - Shama Parveen
- Department of Zoology University of Lucknow Lucknow India
| | - Asmita Singh
- Department of Chemistry University of Lucknow Lucknow India
| | - Astha Yadav
- Department of Chemistry University of Lucknow Lucknow India
| | | |
Collapse
|
32
|
Panda M, Purohit P, Meher BR. Structure-based virtual screening, ADMET profiling, and molecular dynamics simulation studies on HIV-1 protease for identification of active phytocompounds as potential anti-HIV agents. MOLECULAR SIMULATION 2022. [DOI: 10.1080/08927022.2022.2060968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Madhusmita Panda
- Computational Biology and Bioinformatics Laboratory, PG Department of Botany, Berhampur University, Berhampur, India
| | - Priyanka Purohit
- Computational Biology and Bioinformatics Laboratory, PG Department of Botany, Berhampur University, Berhampur, India
| | - Biswa Ranjan Meher
- Computational Biology and Bioinformatics Laboratory, PG Department of Botany, Berhampur University, Berhampur, India
| |
Collapse
|
33
|
Rudrapal M, Celik I, Khan J, Ansari MA, Alomary MN, Yadav R, Sharma T, Tallei TE, Pasala PK, Sahoo RK, Khairnar SJ, Bendale AR, Zothantluanga JH, Chetia D, Walode SG. Identification of bioactive molecules from Triphala (Ayurvedic herbal formulation) as potential inhibitors of SARS-CoV-2 main protease (Mpro) through computational investigations. JOURNAL OF KING SAUD UNIVERSITY. SCIENCE 2022; 34:101826. [PMID: 35035181 PMCID: PMC8744360 DOI: 10.1016/j.jksus.2022.101826] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/20/2021] [Accepted: 01/05/2022] [Indexed: 05/28/2023]
Abstract
Severe acute respiratory syndrome coronavirus disease (SARS-CoV-2) induced coronavirus disease 2019 (COVID-19) pandemic is the present worldwide health emergency. The global scientific community faces a significant challenge in developing targeted therapies to combat the SARS-CoV-2 infection. Computational approaches have been critical for identifying potential SARS-CoV-2 inhibitors in the face of limited resources and in this time of crisis. Main protease (Mpro) is an intriguing drug target because it processes the polyproteins required for SARS-CoV-2 replication. The application of Ayurvedic knowledge from traditional Indian systems of medicine may be a promising strategy to develop potential inhibitor for different target proteins of SARS-CoV-2. With this endeavor, we docked bioactive molecules from Triphala, an Ayurvedic formulation, against Mpro followed by molecular dynamics (MD) simulation (100 ns) to investigate their inhibitory potential against SARS-CoV-2. The top four best docked molecules (terflavin A, chebulagic acid, chebulinic acid, and corilagin) were selected for MD simulation study and the results obtained were compared to native ligand X77. From docking and MD simulation studies, the selected molecules showed promising binding affinity with the formation of stable complexes at the active binding pocket of Mpro and exhibited negative binding energy during MM-PBSA calculations, indication their strong binding affinity with the target protein. The identified bioactive molecules were further analyzed for drug-likeness by Lipinski's filter, ADMET and toxicity studies. Computational (in silico) investigations identified terflavin A, chebulagic acid, chebulinic acid, and corilagin from Triphala formulation as promising inhibitors of SARS-CoV-2 Mpro, suggesting experimental (in vitro/in vivo) studies to further explore their inhibitory mechanisms.
Collapse
Affiliation(s)
- Mithun Rudrapal
- Department of Pharmaceutical Chemistry, Rasiklal M. Dhariwal Institute of Pharmaceutical Education & Research, Pune 411019, Maharashtra, India
| | - Ismail Celik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, Kayseri 38039, Turkey
| | - Johra Khan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia
- Health and Basic Sciences Research Center, Majmaah University, Al Majmaah 11952, Saudi Arabia
| | - Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabaia
| | - Mohammad N Alomary
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah 11952, Saudi Arabia
- Health and Basic Sciences Research Center, Majmaah University, Al Majmaah 11952, Saudi Arabia
- National Centre for Biotechnology, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Rohitash Yadav
- Department of Pharmacology, All India Institute of Medical Sciences, Rishikesh 249203, India
| | - Tripti Sharma
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Siksha O Anusandhan Deemed to be University, Bhubaneswar 751003, Odisha, India
| | - Trina Ekawati Tallei
- Department of Biology, Faculty of Mathematics and Natural Sciences, Sam Ratulangi University, Manado 95115, North Sulawesi, Indonesia
- The University Center of Excellence for Biotechnology and Conservation of Wallacea, Sam Ratulangi University, Manado, North Sulawesi 95115, Indonesia
| | | | - Ranjan Kumar Sahoo
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Bhubaneswar 752050, Odisha, India
| | | | - Atul R Bendale
- Sandip Institute of Pharmaceutical Sciences, Nashik 422213, India
| | - James H Zothantluanga
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India
| | - Dipak Chetia
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India
| | - Sanjay G Walode
- Department of Pharmaceutical Chemistry, Rasiklal M. Dhariwal Institute of Pharmaceutical Education & Research, Pune 411019, Maharashtra, India
| |
Collapse
|
34
|
In Silico Screening of Cinnamon (Cinnamomum burmannii) Bioactive Compounds as Acetylcholinesterase Inhibitors. JURNAL KIMIA SAINS DAN APLIKASI 2022. [DOI: 10.14710/jksa.25.3.97-107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Alzheimer’s is a progressive and neurodegenerative disease that mainly affects people aged 65 years and older. The pathophysiology of Alzheimer’s is possibly related to the depletion of the neurotransmitter acetylcholine (ACh) due to beta-amyloid plaques and neurofibrillary tangles. Secondary metabolites found in cinnamon bark (Cinnamomum burmannii) have the potential as anticholinesterases to treat Alzheimer’s symptoms. This study aimed to identify the potency of bioactive compounds from cinnamon bark as AChE inhibitors in silico through analysis of binding energy, inhibition constants, and types of interactions. The research was conducted by screening virtually 60 test ligands using the PyRx program and molecular docking using the Autodock Tools program. The results of the ligand-receptor interaction analysis showed that 12 of the 15 tested ligands had potential as AChE inhibitors. Epicatechin and medioresinol are the ligands with the best potential for AChE inhibition with affinity close to the natural ligand or donepezil. Epicatechin has a binding energy of −10.0 kcal/mol and inhibition constant of 0.0459 M, with four hydrogen bonds and seven hydrophobic bonds. Meanwhile, medioresinol has −9.9 kcal/mol binding energy and inhibition constant of 0.0543 M, with one hydrogen bond and thirteen hydrophobic bonds.
Collapse
|
35
|
In silico prediction models for thyroid peroxidase inhibitors and their application to synthetic flavors. Food Sci Biotechnol 2022; 31:483-495. [PMID: 35464247 PMCID: PMC8994803 DOI: 10.1007/s10068-022-01041-y] [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: 10/13/2021] [Revised: 01/22/2022] [Accepted: 02/02/2022] [Indexed: 11/27/2022] Open
Abstract
AbstractSystematic toxicity tests are often waived for the synthetic flavors as they are added in a very small amount in foods. However, their safety for some endpoints such as endocrine disruption should be concerned as they are likely to be active in low levels. In this case, structure–activity-relationship (SAR) models are good alternatives. In this study, therefore, binary, ternary, and quaternary prediction models were designed using simple or complex machine-learning methods. Overall, hard-voting classifiers outperformed other methods. The test scores for the best binary, ternary, and quaternary models were 0.6635, 0.5083, and 0.5217, respectively. Along with model development, some substructures including primary aromatic amine, (enol)ether, phenol, heterocyclic sulfur, and heterocyclic nitrogen, dominantly occurred in the most highly active compounds. The best predicting models were applied to synthetic flavors, and 22 agents appeared to have a strong inhibitory potential towards TPO activities.
Collapse
|
36
|
Synthesis, molecular modeling, quantum mechanical calculations and ADME estimation studies of benzimidazole-oxadiazole derivatives as potent antifungal agents. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.132095] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
37
|
Hamre JR, Jafri MS. Optimizing peptide inhibitors of SARS-Cov-2 nsp10/nsp16 methyltransferase predicted through molecular simulation and machine learning. INFORMATICS IN MEDICINE UNLOCKED 2022; 29:100886. [PMID: 35252541 PMCID: PMC8883729 DOI: 10.1016/j.imu.2022.100886] [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: 12/23/2021] [Revised: 02/04/2022] [Accepted: 02/16/2022] [Indexed: 11/30/2022] Open
Abstract
Coronaviruses, including the recent pandemic strain SARS-Cov-2, use a multifunctional 2'-O-methyltransferase (2'-O-MTase) to restrict the host defense mechanism and to methylate RNA. The nonstructural protein 16 2'-O-MTase (nsp16) becomes active when nonstructural protein 10 (nsp10) and nsp16 interact. Novel peptide drugs have shown promise in the treatment of numerous diseases and new research has established that nsp10 derived peptides can disrupt viral methyltransferase activity via interaction of nsp16. This study had the goal of optimizing new analogous nsp10 peptides that have the ability to bind nsp16 with equal to or higher affinity than those naturally occurring. The following research demonstrates that in silico molecular simulations can shed light on peptide structures and predict the potential of new peptides to interrupt methyltransferase activity via the nsp10/nsp16 interface. The simulations suggest that misalignments at residues F68, H80, I81, D94, and Y96 or rotation at H80 abrogate MTase function. We develop a new set of peptides based on conserved regions of the nsp10 protein in the Coronaviridae species and test these to known MTase variant values. This results in the prediction that the H80R variant is a solid new candidate for potential new testing. We envision that this new lead is the beginning of a reputable foundation of a new computational method that combats coronaviruses and that is beneficial for new peptide drug development.
Collapse
Affiliation(s)
- John R Hamre
- School of Systems Biology, George Mason University, Fairfax, VA, 22030, USA
| | - M Saleet Jafri
- School of Systems Biology, George Mason University, Fairfax, VA, 22030, USA
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| |
Collapse
|
38
|
das Chagas Almeida A, Meinel RS, Leal YL, Silva TP, Glanzmann N, Mendonça DVC, Perin L, Cunha-Júnior EF, Coelho EAF, Melo RCN, da Silva AD, Coimbra ES. Functionalized 1,2,3-triazolium salts as potential agents against visceral leishmaniasis. Parasitol Res 2022; 121:1389-1406. [PMID: 35169883 DOI: 10.1007/s00436-022-07431-9] [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: 08/16/2021] [Accepted: 01/07/2022] [Indexed: 10/19/2022]
Abstract
Visceral leishmaniasis (VL) is the most severe clinical form of leishmaniasis, being fatal if untreated. In search of a more effective treatment for VL, one of the main strategies is the development and screening of new antileishmanial compounds. Here, we reported the synthesis of seven new acetyl functionalized 1,2,3-triazolium salts, together with four 1,2,3-triazole precursors, and investigated their effect against different strains of L. infantum from dogs and humans. The 1,2,3-triazolium salts exhibited better activity than the 1,2,3-triazole derivatives with IC50 range from 0.12 to 8.66 μM and, among them, compound 5 showed significant activity against promastigotes (IC50 from 4.55 to 5.28 μM) and intracellular amastigotes (IC50 from 5.36 to 7.92 μM), with the best selective index (SI ~ 6-9) and reduced toxicity. Our findings, using biochemical and ultrastructural approaches, demonstrated that compound 5 targets the mitochondrion of L. infantum promastigotes, leading to the formation of reactive oxygen species (ROS), increase of the mitochondrial membrane potential, and mitochondrial alteration. Moreover, quantitative transmission electron microscopy (TEM) revealed that compound 5 induces the reduction of promastigote size and cytoplasmic vacuolization. Interestingly, the effect of compound 5 was not associated with apoptosis or necrosis of the parasites but, instead, seems to be mediated through a pathway involving autophagy, with a clear detection of autophagic vacuoles in the cytoplasm by using both a fluorescent marker and TEM. As for the in vivo studies, compound 5 showed activity in a mouse model of VL at 20 mg/kg, reducing the parasite load in both spleen and liver (59.80% and 26.88%, respectively). Finally, this compound did not induce hepatoxicity or nephrotoxicity and was able to normalize the altered biochemical parameters in the infected mice. Thus, our findings support the use of 1,2,3-triazolium salts as potential agents against visceral leishmaniasis.
Collapse
Affiliation(s)
- Ayla das Chagas Almeida
- Núcleo de Pesquisas Em Parasitologia, Departamento de Parasitologia, Microbiologia e Imunologia, I.C.B., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Raíssa Soares Meinel
- SINTBIOMOL, Departamento de Química, I.C.E., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Yasmim Lopes Leal
- SINTBIOMOL, Departamento de Química, I.C.E., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Thiago P Silva
- Laboratório de Biologia Celular, Departamento de Biologia, Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Nícolas Glanzmann
- SINTBIOMOL, Departamento de Química, I.C.E., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Débora Vasconcelos Costa Mendonça
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luísa Perin
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Edézio Ferreira Cunha-Júnior
- Laboratório de Imunoparasitologia, Unidade Integrada de Pesquisa Em Produtos Bioativos e Biociências, Universidade Federal Do Rio de Janeiro, Campus UFRJ-Macaé, Macaé, Brazil
| | - Eduardo A F Coelho
- Programa de Pós-Graduação Em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Rossana C N Melo
- Laboratório de Biologia Celular, Departamento de Biologia, Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Adilson David da Silva
- SINTBIOMOL, Departamento de Química, I.C.E., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil
| | - Elaine Soares Coimbra
- Núcleo de Pesquisas Em Parasitologia, Departamento de Parasitologia, Microbiologia e Imunologia, I.C.B., Universidade Federal de Juiz de Fora, Campus Universitário, Juiz de Fora, Minas Gerais, Brazil.
| |
Collapse
|
39
|
Swain SS, Pati S, Hussain T. Quinoline heterocyclic containing plant and marine candidates against drug-resistant Mycobacterium tuberculosis: A systematic drug-ability investigation. Eur J Med Chem 2022; 232:114173. [DOI: 10.1016/j.ejmech.2022.114173] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/30/2022] [Accepted: 02/02/2022] [Indexed: 12/22/2022]
|
40
|
Xing Y, Wang Z, Li X, Hou C, Chai J, Li X, Su J, Gao J, Xu H. A new method for predicting the acute toxicity of carbamate pesticides based on the perspective of binding information with carrier protein. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 264:120188. [PMID: 34358782 DOI: 10.1016/j.saa.2021.120188] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/08/2021] [Accepted: 07/12/2021] [Indexed: 06/13/2023]
Abstract
Toxicity is one of the most important factors limiting the success of new drug development. In this paper, we built a fast and convenient new method (Carrier protein binding information-toxicity relationship, CPBITR) for predicting drug acute toxicity based on the perspective of binding information with carrier protein. First, we studied the binding information between carbamate pesticides and human serum albumin (HSA) through various spectroscopic methods and molecular docking. Then a total of 16 models were established to clarify the relationship between binding information with HSA and drug toxicity. The results showed that the binding information was related to toxicity. Finally we obtained the effective toxicity prediction model for carbamate pesticides. And the "Platform for Predicting Drug Toxicity Based on the Information of Binding with Carrier Protein" was established with the Back-propagation neural network model. We proposed and proved that it was feasible to predict drug toxicity from this new perspective: binding with carrier protein. According to this new perspective, toxicity prediction model of other drugs can also be established. This new method has the advantages of convenience and fast, and can be used to screen out low-toxic drugs quickly in the early stage. It is helpful for drug research and development.
Collapse
Affiliation(s)
- Yue Xing
- Engineering Research Center of Pesticide of Heilongjiang Province, College of Advanced Agriculture and Ecological Environment, Heilongjiang University, Harbin 150080, China
| | - Zishi Wang
- Engineering Research Center of Pesticide of Heilongjiang Province, College of Advanced Agriculture and Ecological Environment, Heilongjiang University, Harbin 150080, China
| | - Xiangshuai Li
- Engineering Research Center of Pesticide of Heilongjiang Province, College of Advanced Agriculture and Ecological Environment, Heilongjiang University, Harbin 150080, China
| | - Chenxin Hou
- Engineering Research Center of Pesticide of Heilongjiang Province, College of Advanced Agriculture and Ecological Environment, Heilongjiang University, Harbin 150080, China
| | - Jiashuang Chai
- Engineering Research Center of Pesticide of Heilongjiang Province, College of Advanced Agriculture and Ecological Environment, Heilongjiang University, Harbin 150080, China
| | - Xiangfen Li
- Engineering Research Center of Pesticide of Heilongjiang Province, College of Advanced Agriculture and Ecological Environment, Heilongjiang University, Harbin 150080, China
| | - Jing Su
- Engineering Research Center of Pesticide of Heilongjiang Province, College of Advanced Agriculture and Ecological Environment, Heilongjiang University, Harbin 150080, China
| | - Jinsheng Gao
- Engineering Research Center of Pesticide of Heilongjiang Province, College of Advanced Agriculture and Ecological Environment, Heilongjiang University, Harbin 150080, China.
| | - Hongliang Xu
- Engineering Research Center of Pesticide of Heilongjiang Province, College of Advanced Agriculture and Ecological Environment, Heilongjiang University, Harbin 150080, China.
| |
Collapse
|
41
|
Król SK, Bębenek E, Dmoszyńska-Graniczka M, Sławińska-Brych A, Boryczka S, Stepulak A. Acetylenic Synthetic Betulin Derivatives Inhibit Akt and Erk Kinases Activity, Trigger Apoptosis and Suppress Proliferation of Neuroblastoma and Rhabdomyosarcoma Cell Lines. Int J Mol Sci 2021; 22:12299. [PMID: 34830180 PMCID: PMC8624615 DOI: 10.3390/ijms222212299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 12/12/2022] Open
Abstract
Neuroblastoma (NB) and rhabdomyosarcoma (RMS), the most common pediatric extracranial solid tumors, still represent an important clinical challenge since no effective treatment is available for metastatic and recurrent disease. Hence, there is an urgent need for the development of new chemotherapeutics to improve the outcome of patients. Betulin (Bet), a triterpenoid from the bark of birches, demonstrated interesting anti-cancer potential. The modification of natural phytochemicals with evidenced anti-tumor activity, including Bet, is one of the methods of receiving new compounds for potential implementation in oncological treatment. Here, we showed that two acetylenic synthetic Bet derivatives (ASBDs), EB5 and EB25/1, reduced the viability and proliferation of SK-N-AS and TE671 cells, as measured by MTT and BrdU tests, respectively. Moreover, ASBDs were also more cytotoxic than temozolomide (TMZ) and cisplatin (cis-diaminedichloroplatinum [II], CDDP) in vitro, and the combination of EB5 with CDDP enhanced anti-cancer effects. We also showed the slowdown of cell cycle progression at S/G2 phases mediated by EB5 using FACS flow cytometry. The decreased viability and proliferation of pediatric cancers cells after treatment with ASBDs was linked to the reduced activity of kinases Akt, Erk1/2 and p38 and the induction of apoptosis, as investigated using Western blotting and FACS. In addition, in silico analyses of the ADMET profile found EB5 to be a promising anti-cancer drug candidate that would benefit from further investigation.
Collapse
Affiliation(s)
- Sylwia K. Król
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Medical University of Lublin, Chodźki 1, 20-093 Lublin, Poland; (M.D.-G.); (A.S.)
| | - Ewa Bębenek
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland; (E.B.); (S.B.)
| | - Magdalena Dmoszyńska-Graniczka
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Medical University of Lublin, Chodźki 1, 20-093 Lublin, Poland; (M.D.-G.); (A.S.)
| | - Adrianna Sławińska-Brych
- Department of Cell Biology, Faculty of Biology and Biotechnology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Akademicka 19, 20-033 Lublin, Poland;
| | - Stanisław Boryczka
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland; (E.B.); (S.B.)
| | - Andrzej Stepulak
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Medical University of Lublin, Chodźki 1, 20-093 Lublin, Poland; (M.D.-G.); (A.S.)
| |
Collapse
|
42
|
Synthesis, In Silico Study, and Anti-Cancer Activity of Thiosemicarbazone Derivatives. Biomedicines 2021; 9:biomedicines9101375. [PMID: 34680491 PMCID: PMC8533299 DOI: 10.3390/biomedicines9101375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/23/2021] [Accepted: 09/28/2021] [Indexed: 12/14/2022] Open
Abstract
Thiosemicarbazones are known for their biological and pharmacological activities. In this study, we have synthesized and characterized 3-Methoxybenzaldehyde thiosemicarbazone (3-MBTSc) and 4-Nitrobenzaldehyde thiosemicarbazone (4-NBTSc) using IR, 1HNMR and 13C NMR. The compound’s in vitro anticancer activities against different cell lines were evaluated. Molecular docking, Insilco ADMET, and drug-likeness prediction were also done. The test compounds showed a comparative IC50 and growth inhibition with the standard drug Doxorubicin. The IC50 ranges from 2.82 µg/mL to 14.25 µg/mL in 3-MBTSc and 2.80 µg/mL to 7.59 µg/mL in 4-NBTSc treated cells. The MTT assay result revealed, 3-MBTSc inhibits 50.42 and 50.31 percent of cell growth in B16-F0 and EAC cell lines, respectively. The gene expression showed that tumor suppressor genes such as PTEN and BRCA1 are significantly upregulated in 7.42 and 5.33 folds, and oncogenes, PKC, and RAS are downregulated −7.96 and −7.64 folds, respectively in treated cells. The molecular docking performed on the four targeted proteins (PARP, VEGFR-1, TGF-β1, and BRAFV600E) indicated that both 4-NBTSc and 3-MBTSc potentially bind to TGF-β1 with the best binding energy of −42.34 Kcal/mol and −32.13 Kcal/mol, respectively. In addition, the test compound possesses desirable ADMET and drug-likeness properties. Overall, both 3-MBTSc and 4-NBTSc have the potential to be multitargeting drug candidates for further study. Moreover, 3-MBTSc showed better activity than 4-NBTSc.
Collapse
|
43
|
Syahputra G, Gustini N, Bustanussalam B, Hapsari Y, Sari M, Ardiansyah A, Bayu A, Putra MY. Molecular docking of secondary metabolites from Indonesian marine and terrestrial organisms targeting SARS-CoV-2 ACE-2, M pro, and PL pro receptors. PHARMACIA 2021. [DOI: 10.3897/pharmacia.68.e68432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
With the uncontrolled spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), development and distribution of antiviral drugs and vaccines have gained tremendous importance. This study focused on two viral proteases namely main protease (Mpro) and papain-like protease (PLpro) and human angiotensin-converting enzyme (ACE-2) to identify which of these are essential for viral replication. We screened 102 secondary metabolites against SARS-CoV-2 isolated from 36 terrestrial plants and 36 marine organisms from Indonesian biodiversity. These organisms are typically presumed to have antiviral effects, and some of them have been used as an immunomodulatory activity in traditional medicine. For the molecular docking procedure to obtain Gibbs free energy value (∆G), toxicity, ADME and Lipinski, AutoDock Vina was used. In this study, five secondary metabolites, namely corilagin, dieckol, phlorofucofuroeckol A, proanthocyanidins, and isovitexin, were found to inhibit ACE-2, Mpro, and PLpro receptors in SARS-CoV-2, with a high affinity to the same sites of ptilidepsin, remdesivir, and chloroquine as the control molecules. This study was delimited to molecular docking without any validation by simulations concerned with molecular dynamics. The interactions with two viral proteases and human ACE-2 may play a key role in developing antiviral drugs for five active compounds. In future, we intend to investigate antiviral drugs and the mechanisms of action by in vitro study.
Collapse
|
44
|
Reshad RAI, Alam S, Raihan HB, Meem KN, Rahman F, Zahid F, Rafid MI, Rahman SMO, Omit S, Ali MH. In silico investigations on curcuminoids from Curcuma longa as positive regulators of the Wnt/β-catenin signaling pathway in wound healing. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2021. [DOI: 10.1186/s43042-021-00182-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Abstract
Background
Curcuma longa (Turmeric) is a traditionally used herb in wound healing. The efficacy of fresh turmeric paste to heal wounds has already been investigated in multiple ethnobotanical studies. Wnt/β-catenin signaling pathway plays a significant role in wound healing and injury repair processes which has been evident in different in vitro studies. This study aims to analyze the potentiality of curcuminoids (curcumin I, curcumin II and curcumin III) from Curcuma longa to bind and enhance the activity of two intracellular signaling proteins- casein kinase-1 (CK1) and glycogen synthase kinase-3β (GSK3B) involved in Wnt/β-catenin signaling pathway. This study is largely based on a computer-based molecular docking program which mimics the in vivo condition and works on a specific algorithm to interpret the binding affinity and poses of a ligand molecule to a receptor. Subsequently, drug likeness property, ADME/Toxicity profile, pharmacological activity, and site of metabolism of the curcuminoids were also analyzed.
Results
Curcumin I showed better affinity of binding with CK1 (− 10.31 Kcal/mol binding energy) and curcumin II showed better binding affinity (− 7.55 Kcal/mol binding energy) for GSK3B. All of the ligand molecules showed quite similar pharmacological properties.
Conclusion
Curcumin has anti-oxidant, anti-carcinogenic, anti-mutagenic, anti-coagulant, and anti-infective properties. Curcumin has also anti-inflammatory and wound healing properties. It hastens wound healing by acting on different stages of the natural wound healing process. In this study, three curcumins from Curcuma longa were utilized in this experiment in a search for a drug to be used in wound healing and injury repair processes. Hopefully, this study will raise research interest among researchers.
Collapse
|
45
|
Wu Z, Jiang D, Wang J, Hsieh CY, Cao D, Hou T. Mining Toxicity Information from Large Amounts of Toxicity Data. J Med Chem 2021; 64:6924-6936. [PMID: 33961429 DOI: 10.1021/acs.jmedchem.1c00421] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Safety is a main reason for drug failures, and therefore, the detection of compound toxicity and potential adverse effects in the early stage of drug development is highly desirable. However, accurate prediction of many toxicity endpoints is extremely challenging due to low accessibility of sufficient and reliable toxicity data, as well as complicated and diversified mechanisms related to toxicity. In this study, we proposed the novel multitask graph attention (MGA) framework to learn the regression and classification tasks simultaneously. MGA has shown excellent predictive power on 33 toxicity data sets and has the capability to extract general toxicity features and generate customized toxicity fingerprints. In addition, MGA provides a new way to detect structural alerts and discover the relationship between different toxicity tasks, which will be quite helpful to mine toxicity information from large amounts of toxicity data.
Collapse
Affiliation(s)
- Zhenxing Wu
- Innovation Institute for Artificial Intelligence in Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058 Zhejiang, P. R. China.,State Key Lab of CAD&CG, Zhejiang University, Hangzhou 310058 Zhejiang, P. R. China
| | - Dejun Jiang
- Innovation Institute for Artificial Intelligence in Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058 Zhejiang, P. R. China
| | - Jike Wang
- Innovation Institute for Artificial Intelligence in Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058 Zhejiang, P. R. China.,National Engineering Research Center for Multimedia Software, School of Computer Science, Wuhan University, Wuhan 430072 Hubei, P. R. China
| | - Chang-Yu Hsieh
- Tencent Quantum Laboratory, Tencent, Shenzhen 518057 Guangdong, P. R. China
| | - Dongsheng Cao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410004 Hunan, P. R. China
| | - Tingjun Hou
- Innovation Institute for Artificial Intelligence in Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058 Zhejiang, P. R. China.,State Key Lab of CAD&CG, Zhejiang University, Hangzhou 310058 Zhejiang, P. R. China
| |
Collapse
|
46
|
Wu Z, Wang Q, Yang H, Wang J, Li W, Liu G, Yang Y, Zhao Y, Tang Y. Discovery of Natural Products Targeting NQO1 via an Approach Combining Network-Based Inference and Identification of Privileged Substructures. J Chem Inf Model 2021; 61:2486-2498. [PMID: 33955748 DOI: 10.1021/acs.jcim.1c00260] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
NAD(P)H:quinone oxidoreductase 1 (NQO1) has been shown to be a potential therapeutic target for various human diseases, such as cancer and neurodegenerative disorders. Recent advances in computational methods, especially network-based methods, have made it possible to identify novel compounds for a target with high efficiency and low cost. In this study, we designed a workflow combining network-based methods and identification of privileged substructures to discover new compounds targeting NQO1 from a natural product library. According to the prediction results, we purchased 56 compounds for experimental validation. Without the assistance of privileged substructures, 31 compounds (31/56 = 55.4%) showed IC50 < 100 μM, and 11 compounds (11/56 = 19.6%) showed IC50 < 10 μM. With the assistance of privileged substructures, the two success rates were increased to 61.8 and 26.5%, respectively. Seven natural products further showed inhibitory activity on NQO1 at the cellular level, which may serve as lead compounds for further development. Moreover, network analysis revealed that osthole may exert anticancer effects against multiple cancer types by inhibiting not only carbonic anhydrases IX and XII but also NQO1. Our workflow and computational methods can be easily applied in other targets and become useful tools in drug discovery and development.
Collapse
Affiliation(s)
- Zengrui Wu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Qiaohui Wang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.,Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Hongbin Yang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Jiye Wang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Weihua Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Guixia Liu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yi Yang
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuzheng Zhao
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.,Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yun Tang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| |
Collapse
|
47
|
Abstract
Toxicity analysis is a major challenge in drug design and discovery. Recently significant progress has been made through machine learning due to its accuracy, efficiency, and lower cost. US Toxicology in the 21st Century (Tox21) screened a large library of compounds, including approximately 12 000 environmental chemicals and drugs, for different mechanisms responsible for eliciting toxic effects. The Tox21 Data Challenge offered a platform to evaluate different computational methods for toxicity predictions. Inspired by the success of multiscale weighted colored graph (MWCG) theory in protein-ligand binding affinity predictions, we consider MWCG theory for toxicity analysis. In the present work, we develop a geometric graph learning toxicity (GGL-Tox) model by integrating MWCG features and the gradient boosting decision tree (GBDT) algorithm. The benchmark tests of the Tox21 Data Challenge are employed to demonstrate the utility and usefulness of the proposed GGL-Tox model. An extensive comparison with other state-of-the-art models indicates that GGL-Tox is an accurate and efficient model for toxicity analysis and prediction.
Collapse
Affiliation(s)
- Jian Jiang
- Research Center of Nonlinear Science, College of Mathematics and Computer Science, Engineering Research Center of Hubei Province for Clothing Information, Wuhan Textile University, Wuhan 430200, P R. China
| | - Rui Wang
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Guo-Wei Wei
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
48
|
Xu X, Zhao P, Wang Z, Zhang X, Wu Z, Li W, Tang Y, Liu G. In silico prediction of chemical acute contact toxicity on honey bees via machine learning methods. Toxicol In Vitro 2021; 72:105089. [DOI: 10.1016/j.tiv.2021.105089] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 01/06/2021] [Indexed: 01/30/2023]
|
49
|
Molecular Docking of Red Betel (Piper crocatum Ruiz & Pav) Bioactive Compounds as HMG-CoA Reductase Inhibitor. JURNAL KIMIA SAINS DAN APLIKASI 2021. [DOI: 10.14710/jksa.24.3.101-107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cholesterol plaque buildup in artery walls occurs due to oxidation of Low-Density Lipoprotein (LDL) molecules by free radicals, which are a risk factor for coronary heart disease. Piper crocatum contains active compounds that can act as HMG-CoA reductase inhibitors, such as flavonoids, alkaloids, polyphenols, tannins, and essential oils. This study aimed to predict the potential of Piper crocatum extract and fraction compounds as HMG-CoA reductase inhibitors by investigating the ligand affinity to the HMG-CoA reductase enzyme. Ligand and receptor preparation was conducted using BIOVIA Discovery Studio Visualizer v16.1.0.15350 and AutoDock Tools v.1.5.6. Molecular docking used AutoDock Vina, while ligand visualization and receptor binding used PyMOL(TM) 1.7.4.5.Edu. The receptor used was HMG-CoA reductase (PDB code: 1HWK) with atorvastatin as a control ligand. Catechin, schisandrin B, and CHEMBL216163 had the highest inhibition with affinity energies of -7.9 kcal/mol, -8.2 kcal/mol, -8.3 kcal/mol, respectively. Amino acid residues that played a role in ligand and receptor interactions were Ser684, Asp690, Lys691, Lys692.
Collapse
|
50
|
Abstract
Scale deposition is a pertinent challenge in the oil and gas industry. Scales formed from iron sulfide are one of the troublous scales, particularly pyrite. Moreover, the use of biodegradable environmentally friendly chemicals reduces the cost compared to the conventional removal process. In this work, the chelating abilities of four novel chemicals, designed using the in silico technique of density functional theory (DFT), are studied as potential iron sulfide scale removers. Only one of the chemicals containing a hydroxamate functional group had a good chelating ability with Fe2+. The chelating strength and ecotoxicological properties of this chemical were compared to diethylenetriaminepentaacetic acid (DTPA), an already established iron sulfide scale remover. The new promising chemical surpassed DTPA in being a safer chemical and having a greater binding affinity to Fe2+ upon optimization, hence, a better choice. The presence of oxime (-NHOH) and carbonyl (C=O) moieties in the new chemical showed that the bidentate form of chelation is favored. Moreover, the presence of an intramolecular hydrogen bond enhanced its chelating ability.
Collapse
|