1
|
Ha SV, Jaensch S, Freitas LGA, Herman D, Czodrowski P, Ceulemans H. Low concentration cell painting images enable the identification of highly potent compounds. Sci Rep 2024; 14:24403. [PMID: 39420056 PMCID: PMC11487191 DOI: 10.1038/s41598-024-75401-5] [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: 05/23/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024] Open
Abstract
Image-based models that use features extracted from cell microscopy images can estimate the activity of small molecules in various biological assays. Typically, models are trained on images stained by an optimized protocol (e.g. Cell Painting) after exposure to a fairly high small molecule concentration (referred to as 'image concentration') of 10 μ M or higher. Low concentration images (e.g. 0.16 μM, 0.8 μM, 4 μM) tend to yield models with worse performance. In this work, we nevertheless report a practical use for low image concentration data. We propose the combination of well-performing models trained at higher image concentrations, with lower image concentration for inference to identify more potent compounds. We show that this approach improves on the conventional method (directly training a high-potency model) in 65 % of assays investigated in terms of AUC-ROC, and 75 % of assays in terms of RIPtoP-corrected AUC-PR.
Collapse
Affiliation(s)
- Son V Ha
- Janssen Pharmaceutica, N.V., a Johnson & Johnson company, 2340, Beerse, Belgium
- Department of Chemistry, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Steffen Jaensch
- Janssen Pharmaceutica, N.V., a Johnson & Johnson company, 2340, Beerse, Belgium.
| | - Lorena G A Freitas
- Janssen Pharmaceutica, N.V., a Johnson & Johnson company, 2340, Beerse, Belgium
| | - Dorota Herman
- Janssen Pharmaceutica, N.V., a Johnson & Johnson company, 2340, Beerse, Belgium
| | - Paul Czodrowski
- Department of Chemistry, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Hugo Ceulemans
- Janssen Pharmaceutica, N.V., a Johnson & Johnson company, 2340, Beerse, Belgium
| |
Collapse
|
2
|
Mufti IU, Ain QU, Malik A, Shahid I, Alzahrani AR, Ijaz B, Rehman S. Exploring antiviral activity of Betanin and Glycine Betaine against dengue virus type-2 in transfected Hela cells. Microb Pathog 2024; 195:106894. [PMID: 39214424 DOI: 10.1016/j.micpath.2024.106894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/17/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Dengue virus (DENV) infection is a worldwide public health concern infecting approximately 400 million individuals and about 40,000 mortalities yearly. Despite this, no licensed or readily available antiviral medication is currently available specifically for DENV infection, and therapy is typically symptomatic. Therefore, the objective of the study was to investigate the antiviral activity of Beta vulgaris L. phytoconstituents against DENV-2 targeting NS3 protein. The antiviral activity of phytochemicals was examined through virtual ligand-based screening, antiviral inhibition and dosage response assays, western blotting analysis and MD simulations. We conducted toxicological, and pharmacokinetic analysis to assess plant-based natural compound's efficacy, safety, and non-toxic doses. Molecular docking and MD simulation results revealed that the nonstructural protein-3 (NS3) might prove as a funamental target for Betanin and Glycine Betaine against Dengue virus. Betanin and Glycine betaine were initially studied for their non-toxic doses in HeLa, CHO, and Vero cells via MTT assay. HeLa cells were transiently transfected with cloned vector pcDNA3.1/Zeo(+)/DENV-2 NS3 along with non-toxic doses (80 μM-10 μM) of selected phytochemicals. The dose-response assay illustrated downregulated expression of DENV-2 NS3 gene after administration of Betanin (IC50 = 4.35 μM) and Glycine Betaine (IC50 = 4.49 μM). Dose response analysis of Betanin (80 μM-10 μM) depicted the significant inhibition of NS3 protein expression as well. These results suggested downregulated expression of DENV-2 NS3 at mRNA and protein level portraying the DENV replication inhibition. Based on our study findings, NS3 protease is depicted as distinctive DENV-2 inhibitor target. We will channel our study further into in vitro characterization employing the mechanistic study to understand the role of host factors in anti-flavi therapeutic.
Collapse
Affiliation(s)
- Isra Umbreen Mufti
- Department of Biosciences, COMSATS University Islamabad (CUI), Park Road, Islamabad, 45550, Pakistan
| | - Qurrat Ul Ain
- Department of Biosciences, COMSATS University Islamabad (CUI), Park Road, Islamabad, 45550, Pakistan; Department of Medical Laboratory Technology, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Ayesha Malik
- Center of Excellence in Molecular Biology, University of the Punjab, 87 West Canal Rd, Thoker Niaz Baig, Lahore, Punjab, 53700, Pakistan
| | - Imran Shahid
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, AlAbidiyah, P.O. Box 13578, Makkah, 21955, Saudi Arabia
| | - Abdullah R Alzahrani
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, AlAbidiyah, P.O. Box 13578, Makkah, 21955, Saudi Arabia
| | - Bushra Ijaz
- Center of Excellence in Molecular Biology, University of the Punjab, 87 West Canal Rd, Thoker Niaz Baig, Lahore, Punjab, 53700, Pakistan.
| | - Sidra Rehman
- Department of Biosciences, COMSATS University Islamabad (CUI), Park Road, Islamabad, 45550, Pakistan.
| |
Collapse
|
3
|
Fayaz A, Yousuf M, Segda A, Atia-Tul-Wahab, Zafar H, Kamran M, Meda RNT, Wang Y, Choudhary MI. Phytochemical investigation of Chrysanthellum americanum Vatke and its constituents- a targeted approach for the treatment of leishmaniasis. Fitoterapia 2024; 178:106192. [PMID: 39187029 DOI: 10.1016/j.fitote.2024.106192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/03/2024] [Accepted: 08/23/2024] [Indexed: 08/28/2024]
Abstract
The present study is focused on the isolation and identification of new therapeutic candidates from Chrysanthellum americanum Vatke., and their efficacy against pteridine reductase-1 (PTR1), a valid chemotherapeutic target in the Leishmania parasite. Henceforth, a new compound, chrysanamerine (1), along with 7 known compounds, polyacetylene 2, and flavonoids 3-8, were isolated from C. americanum. Their structures were determined by chemical and spectroscopic analyses and compared with the reported spectroscopic data. All compounds were evaluated for their anti-leishmanial activity against PTR1 via biochemical mechanism-based assay. The in vitro results showed five potential hits including a new compound, chrysanamerine (1), and four known compounds against the PTR1 enzyme. Among them, compound 1 showed a potent enzyme inhibition with an IC50 of 31.02 ± 2.36 μM, whereas a moderate inhibition was observed in cases of compounds 5 and 6 (IC50 = 59.86 ± 3.32, and 45.32 ± 3.5 μM, respectively). Whereas, compounds 3 and 8 showed mild inhibition (IC50 = 72.12 ± 1.12, and 97.18 ± 1.23 μM, respectively) against PTR1, compared with trimethoprim (positive control) (IC50 = 21.07 ± 1.6 μM). Moreover, the results were further validated via molecular docking and molecular dynamics (MD) simulations. Compound 1 showed a strong affinity to the binding site with a docking score of -11.83, along with the formation of a stable protein-ligand complex over the trajectory of 100 ns. Besides, compounds 1-8 were found to be non-cytotoxic on BJ (human fibroblast) cells.
Collapse
Affiliation(s)
- Aneela Fayaz
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Muhammad Yousuf
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Abdoulaye Segda
- Laboratory of Research and Teaching in Animal Health and Biotechnology, Natural Sciences and Agronomy Postgraduate Division, Nazi Boni University, Bobo-Dioulasso 01 BP 1091, Burkina Faso
| | - Atia-Tul-Wahab
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Humaira Zafar
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| | - Muhammad Kamran
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Roland Nâg-Tiéro Meda
- Laboratory of Research and Teaching in Animal Health and Biotechnology, Natural Sciences and Agronomy Postgraduate Division, Nazi Boni University, Bobo-Dioulasso 01 BP 1091, Burkina Faso
| | - Yan Wang
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| | - M Iqbal Choudhary
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
4
|
Ieque AL, Palomo CT, Gabriela de Freitas Spanhol V, Fróes da Motta Dacome ML, Júnior do Carmo Pereira J, Candido FC, Caleffi-Ferracioli KR, Dias Siqueira VL, Cardoso RF, Vandresen F, Alves-Olher VG, de Lima Scodro RB. Preclinical tests for salicylhydrazones derivatives to explore their potential for new antituberculosis agents. Tuberculosis (Edinb) 2024; 148:102545. [PMID: 39079220 DOI: 10.1016/j.tube.2024.102545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 09/09/2024]
Abstract
PURPOSE This study target the synthesis of 22 salicylhydrazones derivatives to apply in vitro screening to explore their potential in the search for new anti-TB prototypes drugs. METHODS The minimum inhibitory concentration (MIC) were evaluated against Mycobacterium tuberculosis (Mtb) H37Rv and clinical isolates. Drug combination assay, cytotoxicity assay, ethidium bromide accumulation assay (EtBr) and in silico analysis regarding the absorption, distribution, metabolism, excretion and toxicity (ADMET) and pharmacological properties were also performed. RESULTS Three most promising compounds were selected (10, 11 and 18) to proceed with screening tests. Compound 18 presented the lowest MIC value (0.49 μg/mL) against Mtb H37Rv strain, followed by compounds 11 (3.9 μg/mL) and 10 (7.8 μg/mL). All compounds showed activity against drug susceptible and resistant clinical isolates. Cytotoxicity results were promising for all salicylhydrazones, with SI values up to 4,205 for compound 18. The derivative 10 was the only one that demonstrated a non-promising cytotoxicity scenario for a single cell line. All derivatives showed an additive effect (FICI >0.5 to 4.0) in combination with isoniazid, ethambutol and rifampicin. CONCLUSION All salicylhydrazones showed potential in the screening tests performed in this study and compound 18 stood out due to its activity against susceptible and resistant bacilli at low concentrations and low cytotoxicity.
Collapse
Affiliation(s)
- Andressa Lorena Ieque
- Postgraduate Program in Health Sciences, State University of Maringá, Maringá, Paraná, 87020-900, Brazil.
| | - Carolina Trevisolli Palomo
- Postgraduate Program in Health Sciences, State University of Maringá, Maringá, Paraná, 87020-900, Brazil.
| | | | | | | | | | - Katiany Rizzieri Caleffi-Ferracioli
- Postgraduate Program in Bioscience and Physiopathology, State University of Maringá, Maringá, Paraná, 87020-900, Brazil; Laboratory of Medical Bacteriology, Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Paraná, 87020-900, Brazil.
| | - Vera Lucia Dias Siqueira
- Postgraduate Program in Bioscience and Physiopathology, State University of Maringá, Maringá, Paraná, 87020-900, Brazil; Laboratory of Medical Bacteriology, Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Paraná, 87020-900, Brazil.
| | - Rosilene Fressatti Cardoso
- Postgraduate Program in Health Sciences, State University of Maringá, Maringá, Paraná, 87020-900, Brazil; Postgraduate Program in Bioscience and Physiopathology, State University of Maringá, Maringá, Paraná, 87020-900, Brazil; Laboratory of Medical Bacteriology, Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Paraná, 87020-900, Brazil.
| | - Fábio Vandresen
- Departament of Chemistry, Federal Technological University of Paraná, Londrina, Paraná, 86036-370, Brazil.
| | | | - Regiane Bertin de Lima Scodro
- Postgraduate Program in Health Sciences, State University of Maringá, Maringá, Paraná, 87020-900, Brazil; Laboratory of Medical Bacteriology, Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Paraná, 87020-900, Brazil.
| |
Collapse
|
5
|
Ekins S, Lane TR, Urbina F, Puhl AC. In silico ADME/tox comes of age: twenty years later. Xenobiotica 2024; 54:352-358. [PMID: 37539466 PMCID: PMC10850432 DOI: 10.1080/00498254.2023.2245049] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/05/2023]
Abstract
In the early 2000s pharmaceutical drug discovery was beginning to use computational approaches for absorption, distribution, metabolism, excretion and toxicity (ADME/Tox, also known as ADMET) prediction. This emphasis on prediction was an effort to reduce the risk of later stage failures from ADME/Tox.Much has been written in the intervening twenty plus years and significant expenditure has occurred in companies developing these in silico capabilities which can be gleaned from publications. It is therefore an appropriate time to briefly reflect on what was proposed then and what the reality is today.20 years ago, we tended to optimise bioactivity and perhaps one ADME/Tox property at a time. Previously pharmaceutical companies needed a whole infrastructure for models - in silico and in vitro experts, IT, champions on a project team, educators and management support. Now we are in the age of generative de novo design where bioactivity and many ADME/Tox properties can be optimised and large language model technologies are available.There are also some challenges such as the focus on very large molecules which may be outside of current ADME/Tox models.We provide an opportunity to look forward with the increasing public data for ADME/Tox as well as expanded types of algorithms available.
Collapse
Affiliation(s)
- Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| | - Thomas R. Lane
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| | - Fabio Urbina
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| | - Ana C. Puhl
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| |
Collapse
|
6
|
Ries B, Alibay I, Swenson DWH, Baumann HM, Henry MM, Eastwood JRB, Gowers RJ. Kartograf: A Geometrically Accurate Atom Mapper for Hybrid-Topology Relative Free Energy Calculations. J Chem Theory Comput 2024; 20:1862-1877. [PMID: 38330251 PMCID: PMC10941767 DOI: 10.1021/acs.jctc.3c01206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 02/10/2024]
Abstract
Relative binding free energy (RBFE) calculations have emerged as a powerful tool that supports ligand optimization in drug discovery. Despite many successes, the use of RBFEs can often be limited by automation problems, in particular, the setup of such calculations. Atom mapping algorithms are an essential component in setting up automatic large-scale hybrid-topology RBFE calculation campaigns. Traditional algorithms typically employ a 2D subgraph isomorphism solver (SIS) in order to estimate the maximum common substructure. SIS-based approaches can be limited by time-intensive operations and issues with capturing geometry-linked chemical properties, potentially leading to suboptimal solutions. To overcome these limitations, we have developed Kartograf, a geometric-graph-based algorithm that uses primarily the 3D coordinates of atoms to find a mapping between two ligands. In free energy approaches, the ligand conformations are usually derived from docking or other previous modeling approaches, giving the coordinates a certain importance. By considering the spatial relationships between atoms related to the molecule coordinates, our algorithm bypasses the computationally complex subgraph matching of SIS-based approaches and reduces the problem to a much simpler bipartite graph matching problem. Moreover, Kartograf effectively circumvents typical mapping issues induced by molecule symmetry and stereoisomerism, making it a more robust approach for atom mapping from a geometric perspective. To validate our method, we calculated mappings with our novel approach using a diverse set of small molecules and used the mappings in relative hydration and binding free energy calculations. The comparison with two SIS-based algorithms showed that Kartograf offers a fast alternative approach. The code for Kartograf is freely available on GitHub (https://github.com/OpenFreeEnergy/kartograf). While developed for the OpenFE ecosystem, Kartograf can also be utilized as a standalone Python package.
Collapse
Affiliation(s)
- Benjamin Ries
- Medicinal
Chemistry, Boehringer Ingelheim Pharma GmbH
& Co KG, Birkendorfer Str 65, 88397 Biberach an der Riss, Germany
- Open
Free Energy, Open Molecular Software Foundation, Davis, 95616 California, United States
| | - Irfan Alibay
- Open
Free Energy, Open Molecular Software Foundation, Davis, 95616 California, United States
| | - David W. H. Swenson
- Open
Free Energy, Open Molecular Software Foundation, Davis, 95616 California, United States
| | - Hannah M. Baumann
- Open
Free Energy, Open Molecular Software Foundation, Davis, 95616 California, United States
| | - Michael M. Henry
- Open
Free Energy, Open Molecular Software Foundation, Davis, 95616 California, United States
- Computational
and Systems Biology Program, Sloan Kettering
Institute, Memorial Sloan Kettering Cancer Center, New York, 1275 New York, United States
| | - James R. B. Eastwood
- Open
Free Energy, Open Molecular Software Foundation, Davis, 95616 California, United States
| | - Richard J. Gowers
- Open
Free Energy, Open Molecular Software Foundation, Davis, 95616 California, United States
| |
Collapse
|
7
|
Murugan R, Tamil Selvan S, Dharmalingam Jothinathan MK, Srinivasan GP, Rajan Renuka R, Prasad M. Molecular Docking and Absorption, Distribution, Metabolism, and Excretion (ADME) Analysis: Examining the Binding Modes and Affinities of Myricetin With Insulin Receptor, Glycogen Synthase Kinase, and Glucokinase. Cureus 2024; 16:e53810. [PMID: 38465169 PMCID: PMC10924184 DOI: 10.7759/cureus.53810] [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: 01/05/2024] [Accepted: 02/07/2024] [Indexed: 03/12/2024] Open
Abstract
Aim By using molecular docking analysis (MDA) to examine its interactions with important regulatory proteins linked to diabetes, such as glycogen synthase kinase 3 beta (GSK3β), insulin receptor (IR), and glucose kinase (GCK), this study seeks to explore the therapeutic potential of myricetin, a naturally occurring flavonoid. Objective The main goal is to determine potential effects on insulin signalling, GSK3β activity, and glucose metabolism by evaluating the binding affinities of myricetin with GCK, IR, and GSK3β through MDA. In order to assess the drug affinity of myricetin, the study also intends to perform absorption, distribution, metabolism, and excretion (ADME) studies. Materials and methods To model the interaction between myricetin and the target proteins (GCK, IR, and GSK3β), we used molecular docking analysis with computational tools. ADME studies were also included in the study to evaluate drug affinity. Identification of binding sites, essential residues, and interaction stability were all part of the structural analysis. Results As evidence of possible interactions with these regulatory proteins, myricetin showed positive binding affinities with GCK, IR, and GSK3β. Strong interactions with important ligand recognition residues were seen in the docking into IR, indicating a potential impact on insulin signalling. Moreover, a strong binding affinity for GCK indicated potential effects on the metabolism of glucose. Studies using ADME confirmed the high drug affinity of myricetin. Conclusion This work sheds light on the multi-target potential of myricetin in the regulation of diabetes. It appears that it has the ability to influence glucose metabolism, suppress GSK3β activity, and regulate insulin signalling based on its interactions with IR, GSK3β, and GCK. Although these computational results show promise, more experimental work is necessary to confirm and fully understand the precise mechanisms that underlie myricetin's effects on the regulation of diabetes.
Collapse
Affiliation(s)
- Ramadurai Murugan
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, IND
| | - Silambarasan Tamil Selvan
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, IND
| | | | - Guru Prasad Srinivasan
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, IND
| | - Remya Rajan Renuka
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, IND
| | - Monisha Prasad
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, IND
| |
Collapse
|
8
|
Khairnar SI, Kulkarni YA, Murugesan S, Singh K. Effects of Acute and Repeated Dose Toxicity Profiling of Chelidonic Acid in Rats: in Silico and in Vivo Evidence. Chem Biodivers 2023; 20:e202301241. [PMID: 37983725 DOI: 10.1002/cbdv.202301241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/29/2023] [Indexed: 11/22/2023]
Abstract
Chelidonic acid is a phytoconstituent found in rhizomes of the perennial plant celandine. The current study aims to evaluate the acute and repeated dose oral toxicity study of chelidonic acid as per the OECD guidelines 425 and 407. The pharmacokinetic and toxicity profile of chelidonic acid was predicted using online servers and tools. A single dose of chelidonic acid (2000 mg/kg) was administered to female Wistar rats in an acute toxicity study, and the animals were monitored for 14 days. We studied the toxicity profile of chelidonic acid at 10, 20, and 40 mg/kg doses in Wistar rats for repeated dose toxicity (28 days). Clinical biochemistry, haematological, and urine parameters were estimated. A gross necropsy and histopathology were performed. A single oral dose of chelidonic acid (2000 mg/kg) showed no signs of toxicity or mortality. The Administration of chelidonic acid showed no significant alterations in haematological, biochemical, and urine parameters. The histopathology showed normal structure and architecture in all the vital organs. A gross necropsy of vital organs showed no signs of toxicity. The chelidonic acid was found to be safe at all selected dose levels in the acute and repeated dose toxicity study in rats.
Collapse
Affiliation(s)
- Shraddha I Khairnar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai, 400 056, India
| | - Yogesh A Kulkarni
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai, 400 056, India
| | - S Murugesan
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, India
| | - Kavita Singh
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai, 400 056, India
| |
Collapse
|
9
|
Gull H, Ikram A, Khalil AA, Ahmed Z, Nemat A. Assessing the multitargeted antidiabetic potential of three pomegranate peel-specific metabolites: An in silico and pharmacokinetics study. Food Sci Nutr 2023; 11:7188-7205. [PMID: 37970376 PMCID: PMC10630828 DOI: 10.1002/fsn3.3644] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/09/2023] [Accepted: 08/13/2023] [Indexed: 11/17/2023] Open
Abstract
Diabetes is a chronic metabolic disorder that occurs due to impaired secretion of insulin, insulin resistance, or both. Recent studies show that the antidiabetic drugs used to control hyperglycemic levels are associated with undesirable adverse effects. Therefore, developing a safe and effective medicine with antidiabetic potential is needed. In this context, in silico studies are considered a rapid, effectual, and cost-effective method in drug discovery procedures. It is evident from the literature that plant-based natural components have shown promising outcomes in drug development to alleviate various diseases and hence have diversified the screening of potential antidiabetic agents. Purposely, in the present study, an in silico approach was performed on three Punica granatum peel metabolites (punicalin, punicalagin, and ellagic acid). All these three compounds were docked against nine protein targets involved in glucose metabolism (GFAT, PTP1β, PPAR-ᵞ, TKIR, RBP4, α-amylase, α-glucosidase, GCK, and AQP-2). These three pomegranate-specific compounds demonstrated significant interactions with GFAT, PTP1β, PPAR-ᵞ, TKIR, RBP4, α-amylase, α-glucosidase, GCK, and AQP-2 protein targets. Specifically, punicalin, punicalagin, and ellagic acid revealed significant binding scores (-9.2, -9.3, -8.1, -9.1, -8.5, -11.3, -9.2, -9.5, -10.1 kcal/mol; -10, -9.9, -8.5, -8.9, -10.4, -9.0, -10.2, -9.4, -9.0 kcal/mol; and -8.1, -8.0, -8.0, -6.8, -8.7, -7.8, -8.3, -8.1, -8.1 kcal/mol, respectively), with nine protein targets mentioned above. Hence, punicalin, punicalagin, and ellagic acid can be promising candidates in drug discovery to manage diabetes. Furthermore, in vivo and clinical trials must be conducted to validate the outcomes of the current study.
Collapse
Affiliation(s)
- Hina Gull
- Faculty of Sciences, Institute of Molecular Biology and BiotechnologyThe University of LahoreLahorePakistan
| | - Aqsa Ikram
- Faculty of Sciences, Institute of Molecular Biology and BiotechnologyThe University of LahoreLahorePakistan
| | - Anees Ahmed Khalil
- Faculty of Allied Health Sciences, University Institute of Diet and Nutritional SciencesThe University of LahoreLahorePakistan
| | - Zahoor Ahmed
- School of Food and Biological EngineeringJiangsu UniversityZhenjiangChina
| | - Arash Nemat
- Department of MicrobiologyKabul University of Medical SciencesKabulAfghanistan
| |
Collapse
|
10
|
Du Y, Guo J, Zhou Y, Yan S, Xu B, Wang Y, Lu D, Ma Z, Chen Q, Tang Q, Zhang W, Zhu J, Huang Y, Yang C. Revealing the Mechanisms of Byu dMar 25 in the Treatment of Alzheimer's Disease through Network Pharmacology, Molecular Docking, and In Vivo Experiment. ACS OMEGA 2023; 8:25066-25080. [PMID: 37483184 PMCID: PMC10357573 DOI: 10.1021/acsomega.3c01683] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/25/2023] [Indexed: 07/25/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common neurodegenerative disease, severely reducing the cognitive level and life quality of patients. Byu dMar 25 (BM25) has been proved to have a therapeutic effect on AD. However, the pharmacological mechanism is still unclear. Therefore, this study aims to reveal the potential mechanism of BM25 affecting AD from the perspective of network pharmacology and experimental validation. METHODS The potential active ingredients of BM25 were obtained from the TCMSP database and literature. Possible targets were predicted using SwissTargetPrediction tools. AD-related genes were identified by using GeneCards, OMIM, DisGeNET, and Drugbank databases. The candidate genes were obtained by extraction of the intersection network. Additionally, the "drug-target-disease" network was constructed by Cytoscape 3.7.2 for visualization. The PPI network was constructed by the STRING database, and the core network modules were filtered by Cytoscape 3.7.2. Enrichment analysis of GO and KEGG was carried out in the Metascape platform. Ledock software was used to dock the critical components with the core target. Furthermore, protein levels were evaluated by immunohistochemistry. RESULTS In this study, 112 active components, 1112 disease candidate genes, 3084 GO functions, and 277 KEGG pathways were obtained. Molecular docking showed that the effective components of BM25 in treating AD were β-asarone and hydroxysafflor yellow A. The most important targets were APP, PIK3R1, and PIK3CA. Enrichment analysis indicated that the Golgi genetic regulation, peroxidase activity regulation, phosphatidylinositol 3-kinase complex IA, 5-hydroxytryptamine receptor complexes, cancer pathways, and neuroactive ligand-receptor interactions played vital roles against AD. The rat experiment verified that BM25 affected PI3K-Akt pathway activation in AD. CONCLUSIONS This study reveals the mechanism of BM25 in treating AD with network pharmacology, which provides a foundation for further study on the molecular mechanism of AD treatment.
Collapse
Affiliation(s)
- Yikuan Du
- Central
Laboratory, The Tenth Affiliated Hospital
of Southern Medical University, Dongguan 523059, China
| | - Jinyan Guo
- Dongguan
Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523000, China
- Dongguan
Key Laboratory of stem cell and regenerative tissue engineering, Guangdong Medical University, Dongguan 523808, China
| | - Yuqi Zhou
- Dongguan
Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523000, China
- Dongguan
Key Laboratory of stem cell and regenerative tissue engineering, Guangdong Medical University, Dongguan 523808, China
| | - Simin Yan
- Dongguan
Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523000, China
- Dongguan
Key Laboratory of stem cell and regenerative tissue engineering, Guangdong Medical University, Dongguan 523808, China
| | - Bijun Xu
- Dongguan
Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523000, China
- Dongguan
Key Laboratory of stem cell and regenerative tissue engineering, Guangdong Medical University, Dongguan 523808, China
| | - Yuni Wang
- Dongguan
Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523000, China
- Dongguan
Key Laboratory of stem cell and regenerative tissue engineering, Guangdong Medical University, Dongguan 523808, China
| | - Duoduo Lu
- Dongguan
Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523000, China
- Dongguan
Key Laboratory of stem cell and regenerative tissue engineering, Guangdong Medical University, Dongguan 523808, China
| | - Zhendong Ma
- Dongguan
Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523000, China
- Dongguan
Key Laboratory of stem cell and regenerative tissue engineering, Guangdong Medical University, Dongguan 523808, China
| | - Qianwen Chen
- Dongguan
Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523000, China
- Dongguan
Key Laboratory of stem cell and regenerative tissue engineering, Guangdong Medical University, Dongguan 523808, China
| | - Qibin Tang
- Dongguan
Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523000, China
- Dongguan
Key Laboratory of stem cell and regenerative tissue engineering, Guangdong Medical University, Dongguan 523808, China
| | - Weichui Zhang
- Dongguan
Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523000, China
- Dongguan
Key Laboratory of stem cell and regenerative tissue engineering, Guangdong Medical University, Dongguan 523808, China
| | - Jinfeng Zhu
- Dongguan
Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523000, China
- Dongguan
Key Laboratory of stem cell and regenerative tissue engineering, Guangdong Medical University, Dongguan 523808, China
| | - Yixing Huang
- Dongguan
Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523000, China
- Dongguan
Key Laboratory of stem cell and regenerative tissue engineering, Guangdong Medical University, Dongguan 523808, China
| | - Chun Yang
- Dongguan
Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523000, China
- Dongguan
Key Laboratory of stem cell and regenerative tissue engineering, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
11
|
Herman D, Kańduła MM, Freitas LGA, van Dongen C, Le Van T, Mesens N, Jaensch S, Gustin E, Micholt L, Lardeau CH, Varsakelis C, Reumers J, Zoffmann S, Will Y, Peeters PJ, Ceulemans H. Leveraging Cell Painting Images to Expand the Applicability Domain and Actively Improve Deep Learning Quantitative Structure-Activity Relationship Models. Chem Res Toxicol 2023. [PMID: 37327474 DOI: 10.1021/acs.chemrestox.2c00404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The search for chemical hit material is a lengthy and increasingly expensive drug discovery process. To improve it, ligand-based quantitative structure-activity relationship models have been broadly applied to optimize primary and secondary compound properties. Although these models can be deployed as early as the stage of molecule design, they have a limited applicability domain─if the structures of interest differ substantially from the chemical space on which the model was trained, a reliable prediction will not be possible. Image-informed ligand-based models partly solve this shortcoming by focusing on the phenotype of a cell caused by small molecules, rather than on their structure. While this enables chemical diversity expansion, it limits the application to compounds physically available and imaged. Here, we employ an active learning approach to capitalize on both of these methods' strengths and boost the model performance of a mitochondrial toxicity assay (Glu/Gal). Specifically, we used a phenotypic Cell Painting screen to build a chemistry-independent model and adopted the results as the main factor in selecting compounds for experimental testing. With the additional Glu/Gal annotation for selected compounds we were able to dramatically improve the chemistry-informed ligand-based model with respect to the increased recognition of compounds from a 10% broader chemical space.
Collapse
Affiliation(s)
- Dorota Herman
- In-Silico Discovery, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Maciej M Kańduła
- Discovery Technology and Molecular Pharmacology, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Lorena G A Freitas
- In-Silico Discovery, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | | | - Thanh Le Van
- Discovery Technology and Molecular Pharmacology, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Natalie Mesens
- Predictive, Investigative and Translational Toxicology, PSTS, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Steffen Jaensch
- Discovery Technology and Molecular Pharmacology, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Emmanuel Gustin
- Discovery Technology and Molecular Pharmacology, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Liesbeth Micholt
- Discovery Technology and Molecular Pharmacology, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Charles-Hugues Lardeau
- Discovery Technology and Molecular Pharmacology, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Christos Varsakelis
- In-Silico Discovery, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Joke Reumers
- Discovery Technology and Molecular Pharmacology, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Sannah Zoffmann
- Discovery Technology and Molecular Pharmacology, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Yvonne Will
- Predictive, Investigative and Translational Toxicology, PSTS, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Pieter J Peeters
- Discovery Technology and Molecular Pharmacology, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| | - Hugo Ceulemans
- In-Silico Discovery, Janssen Research & Development, Pharmaceutical Companies of Johnson & Johnson, Beerse B-2340, Belgium
| |
Collapse
|
12
|
Moharana M, Pattanayak SK, Khan F. Molecular recognition of bio-active triterpenoids from Swertia chirayita towards hepatitis Delta antigen: a mechanism through docking, dynamics simulation, Gibbs free energy landscape. J Biomol Struct Dyn 2023; 41:14651-14664. [PMID: 36856037 DOI: 10.1080/07391102.2023.2184173] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 02/18/2023] [Indexed: 03/02/2023]
Abstract
Medicinal plants the underpinning of indigenous herbal serve, are the possible source of key compounds for the development of new drugs. Hepatitis D, one of the most widespread infectious diseases associated with global public health issues. Therefore, we aim to screen natural compounds to find out potent inhibitor towards hepatitis delta antigen. Through ADMET investigation, we have screened twenty phytochemicals for this study. Additionally, using molecular docking, these phytochemicals were docked with the HDV protease which signifies the phytochemicals beta-amyrin, chiratenol, episwertenol and swertanone have a significant capability to bind with hepatitis D virus protein. The docking study was further accompanied by analyzes RMSD, RMSF, Rg, SASA, Hbond number, and principal component analysis through 100 ns MD simulations. Based on our principal component analysis, beta-amyrin, chiratenol, episwertenol and swertanone phytochemicals can be a potential drug candidates for inhibition of hepatitis D. The above observation is also supported by our Gibbs free energy landscape study. The potential therapeutic characteristics of the phytochemicals against hepatitis D inhibition offer additional support for the in vitro and in vivo studies in future.
Collapse
Affiliation(s)
- Maheswata Moharana
- Department of Chemistry, National Institute of Technology, Raipur, India
| | | | - Fahmida Khan
- Department of Chemistry, National Institute of Technology, Raipur, India
| |
Collapse
|
13
|
Kumari S, Chakraborty S, Ahmad M, Kumar V, Tailor PB, Biswal BK. Identification of probable inhibitors for the DNA polymerase of the Monkeypox virus through the virtual screening approach. Int J Biol Macromol 2023; 229:515-528. [PMID: 36584781 PMCID: PMC9794403 DOI: 10.1016/j.ijbiomac.2022.12.252] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/09/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022]
Abstract
Given the paucity of antiviral treatments for monkeypox disease, caused by the Monkeypox virus (MPXV), there is a pressing need for the development/identification of new drugs to treat the infection. MPXV possesses a linear dsDNA genome that is replicated by a DNA replication complex of which DNA polymerase (DPol) forms an important component. Owing to the importance of DPol in the viral life cycle, identifying/designing small molecules abolishing its function could yield new antivirals. In this study, we first used the AlphaFold artificial intelligence program to model the 3D structure of the MPXV DPol; like the fold of DPol from other organisms, the MPXV DPol structure has the characteristic exonuclease, thumb, palm, and fingers sub-domains arrangement. Subsequently, we have identified several inhibitors through virtual screening of ZINC and antiviral libraries. Molecules with phenyl scaffold along with alanine-based and tetrazole-based molecules showed the best docking score of -8 to -10 kcal/mol. These molecules bind in the palm and fingers sub-domains interface region, which partially overlaps with the DNA binding path. The delineation of DPol/inhibitor interactions showed that majorly active site residues ASP549, ASP753, TYR550, ASN551, SER552, and ASN665 interact with the inhibitors. These compounds exhibit good Absorption, Distribution, Metabolism and Excretion properties.
Collapse
Affiliation(s)
- Swati Kumari
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Sayan Chakraborty
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Mohammed Ahmad
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Varun Kumar
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | | | - Bichitra K Biswal
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| |
Collapse
|
14
|
Chang Y, Hawkins BA, Du JJ, Groundwater PW, Hibbs DE, Lai F. A Guide to In Silico Drug Design. Pharmaceutics 2022; 15:pharmaceutics15010049. [PMID: 36678678 PMCID: PMC9867171 DOI: 10.3390/pharmaceutics15010049] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/16/2022] [Accepted: 12/17/2022] [Indexed: 12/28/2022] Open
Abstract
The drug discovery process is a rocky path that is full of challenges, with the result that very few candidates progress from hit compound to a commercially available product, often due to factors, such as poor binding affinity, off-target effects, or physicochemical properties, such as solubility or stability. This process is further complicated by high research and development costs and time requirements. It is thus important to optimise every step of the process in order to maximise the chances of success. As a result of the recent advancements in computer power and technology, computer-aided drug design (CADD) has become an integral part of modern drug discovery to guide and accelerate the process. In this review, we present an overview of the important CADD methods and applications, such as in silico structure prediction, refinement, modelling and target validation, that are commonly used in this area.
Collapse
Affiliation(s)
- Yiqun Chang
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Bryson A. Hawkins
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Jonathan J. Du
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Paul W. Groundwater
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - David E. Hibbs
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Felcia Lai
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
- Correspondence:
| |
Collapse
|
15
|
Khan J, Sakib SA, Mahmud S, Khan Z, Islam MN, Sakib MA, Emran TB, Simal-Gandara J. Identification of potential phytochemicals from Citrus Limon against main protease of SARS-CoV-2: molecular docking, molecular dynamic simulations and quantum computations. J Biomol Struct Dyn 2022; 40:10741-10752. [PMID: 34278965 DOI: 10.1080/07391102.2021.1947893] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The outbreak of coronavirus disease (COVID-19) caused by a novel RNA virus emerged at the end of 2019. Most of the patient's symptoms are mild to moderate, and influenza, acute respiratory distress syndrome (ARDS) and multi-organ failure are common. The disease is mild to moderate in most patients and is reported in many cases such as pneumonia, ARDS and multi-organ dysfunction. This study's objective is to evaluate 25 natural compounds from Citrus limon (CL) used by comprehensive molecular docking, density functional theory (DFT) and molecular dynamics analysis against SARS-CoV-2 main protease (Mpro). Among all the experimental compounds, diosmetin has shown the best docking values against the Mpro of SARS-CoV-2 compared to the standard antiviral drug. In DFT calculations, the order associated with biochemical reactivity is as follows: eriodictoyl > quercetin > spinacetin > diosmetin > luteolin > apigenin, whereas the regions of oxygen and hydrogen atoms from the selected isolated compounds are appropriate for electrophilic and nucleophilic attacks, respectively. Also, HOMO-LUMO and global descriptors values indicated a promising result of these compounds. Moreover, a molecular dynamics simulation study revealed the stable conformation and binding pattern in a stimulating environment of natural compounds CL. Considering molecular docking, simulation, and DFT analysis of the selected compounds, notably eriodictoyl, quercetin, and diosmetin showed good potential against SARS-CoV-2 Mpro. Our in silico study revealed promising antiviral activity, which may be considered a potential key factor or a therapeutic target for COVID-19.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Jishan Khan
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, Bangladesh
| | - Shahenur Alam Sakib
- Department of Theoretical and Computational Chemistry, University of Dhaka, Dhaka
| | - Shafi Mahmud
- Microbiology Laboratory, Bioinformatics Division, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, Bangladesh
| | - Zidan Khan
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, Bangladesh
| | - Mohammad Nazmul Islam
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, Bangladesh
| | - Mahfuz Ahmed Sakib
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Department of Analytical and Food Chemistry, Faculty of Food Science and Technology, University of Vigo-Ourense Campus, Ourense, Spain
| |
Collapse
|
16
|
Tangmanussukum P, Kawichai T, Suratanee A, Plaimas K. Heterogeneous network propagation with forward similarity integration to enhance drug-target association prediction. PeerJ Comput Sci 2022; 8:e1124. [PMID: 36262151 PMCID: PMC9575853 DOI: 10.7717/peerj-cs.1124] [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: 06/17/2022] [Accepted: 09/14/2022] [Indexed: 06/16/2023]
Abstract
Identification of drug-target interaction (DTI) is a crucial step to reduce time and cost in the drug discovery and development process. Since various biological data are publicly available, DTIs have been identified computationally. To predict DTIs, most existing methods focus on a single similarity measure of drugs and target proteins, whereas some recent methods integrate a particular set of drug and target similarity measures by a single integration function. Therefore, many DTIs are still missing. In this study, we propose heterogeneous network propagation with the forward similarity integration (FSI) algorithm, which systematically selects the optimal integration of multiple similarity measures of drugs and target proteins. Seven drug-drug and nine target-target similarity measures are applied with four distinct integration methods to finally create an optimal heterogeneous network model. Consequently, the optimal model uses the target similarity based on protein sequences and the fused drug similarity, which combines the similarity measures based on chemical structures, the Jaccard scores of drug-disease associations, and the cosine scores of drug-drug interactions. With an accuracy of 99.8%, this model significantly outperforms others that utilize different similarity measures of drugs and target proteins. In addition, the validation of the DTI predictions of this model demonstrates the ability of our method to discover missing potential DTIs.
Collapse
Affiliation(s)
- Piyanut Tangmanussukum
- Advanced Virtual and Intelligent Computing (AVIC) Center, Department of Mathematics and Computer Science, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Thitipong Kawichai
- Department of Mathematics and Computer Science, Academic Division, Chulachomklao Royal Military Academy, Nakhon Nayok, Thailand
| | - Apichat Suratanee
- Department of Mathematics, Faculty of Applied Science, King Mongkut’s University of Technology North Bangkok, Bangkok, Thailand
- Intelligent and Nonlinear Dynamics Innovations Research Center, Science and Technology Research Institute, King Mongkut’s University of Technology North Bangkok, Bangkok, Thailand
| | - Kitiporn Plaimas
- Advanced Virtual and Intelligent Computing (AVIC) Center, Department of Mathematics and Computer Science, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Omics Science and Bioinformatics Center, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
17
|
Shekh S, Moi S, Gowd KH. Virtual screening of sulfur compounds of Allium against coronavirus proteases: E-Ajoene is a potential dual protease targeting covalent inhibitor. J Sulphur Chem 2022. [DOI: 10.1080/17415993.2022.2119086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Shamasoddin Shekh
- Department of Chemistry, School of Chemical Sciences, Central University of Karnataka, Kalaburagi, India
| | - Smriti Moi
- Department of Chemistry, School of Chemical Sciences, Central University of Karnataka, Kalaburagi, India
| | - Konkallu Hanumae Gowd
- Department of Chemistry, School of Chemical Sciences, Central University of Karnataka, Kalaburagi, India
| |
Collapse
|
18
|
Identification of Novel Inhibitors Targeting SGK1 via Ensemble-Based Virtual Screening Method, Biological Evaluation and Molecular Dynamics Simulation. Int J Mol Sci 2022; 23:ijms23158635. [PMID: 35955763 PMCID: PMC9369041 DOI: 10.3390/ijms23158635] [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: 07/05/2022] [Revised: 07/26/2022] [Accepted: 08/02/2022] [Indexed: 11/17/2022] Open
Abstract
Serum and glucocorticoid-regulated kinase 1 (SGK1), as a serine threonine protein kinase of the AGC family, regulates different enzymes, transcription factors, ion channels, transporters, and cell proliferation and apoptosis. Inhibition of SGK1 is considered as a valuable approach for the treatment of various metabolic diseases. In this investigation, virtual screening methods, including pharmacophore models, Bayesian classifiers, and molecular docking, were combined to discover novel inhibitors of SGK1 from the database with 29,158 compounds. Then, the screened compounds were subjected to ADME/T, PAINS and drug-likeness analysis. Finally, 28 compounds with potential inhibition activity against SGK1 were selected for biological evaluation. The kinase inhibition activity test revealed that among these 28 hits, hit15 exhibited the highest inhibition activity against SGK1, which gave 44.79% inhibition rate at the concentration of 10 µM. In order to further investigate the interaction mechanism of hit15 and SGK1 at simulated physiological conditions, a molecular dynamics simulation was performed. The molecular dynamics simulation demonstrated that hit15 could bind to the active site of SGK1 and form stable interactions with key residues, such as Tyr178, ILE179, and VAL112. The binding free energy of the SGK1-hit15 was −48.90 kJ mol−1. Therefore, the identified hit15 with novel scaffold may be a promising lead compound for development of new SGK1 inhibitors for various diseases treatment.
Collapse
|
19
|
Saraswat J, Riaz U, Patel R. In-silico study for the screening and preparation of ionic liquid-AVDs conjugate to combat COVID-19 surge. J Mol Liq 2022; 359:119277. [PMID: 35530033 PMCID: PMC9061583 DOI: 10.1016/j.molliq.2022.119277] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 04/13/2022] [Accepted: 04/26/2022] [Indexed: 11/17/2022]
Abstract
The pandemic due to COVID-19 caused by SARS-CoV-2 has led to the recorded deaths worldwide and is still a matter of concern for scientists to find an effective counteragent. The combination therapy is always been a successful attempt in treating various threatful diseases. Recently, Ionic liquids (ILs) are known for their antiviral activity. Fascinating tunable properties of ILs make them a potential candidate for designing the therapeutic agent. The concern while using ILs in biomedical field remains is toxicity therefore, choline-based ILs were used in the study as they are considered to be greener as compared to other ILs. In the present study strategically, we performed the blind molecular docking of antiviral drug (Abacavir, Acyclovir, and Galidesivir)-choline based ILs conjugates with the target protein (Mpro protease). The molecules were screened on the basis of binding energy. The data suggested that the combination of AVDs-ILs have greater antiviral potential as compared to the drugs and ILs alone. Further, the ADME properties and toxicity analysis of the screened conjugates was done which revealed the non-toxicity of the conjugates. Additionally, the energetic profiling of the ILs drugs and their conjugates was done using DFT calculations which revealed the stability of the conjugates and have a better option to be developed as a therapeutic agent. Also, from molecular dynamic simulation was done and results showed the stability of the complex formed between target protein and the designed conjugates of AVDs and ILs.
Collapse
Affiliation(s)
- Juhi Saraswat
- Biophysical Chemistry Laboratory, Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Ufana Riaz
- Department of Chemistry, Jamia Millia Islamia, New Delhi 110025, India
| | - Rajan Patel
- Biophysical Chemistry Laboratory, Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| |
Collapse
|
20
|
Lanka G, Bhargavi M, Bathula R, Potlapally SR. Targeting tribbles homolog 3 (TRIB3) protein against type 2 diabetes for the identification of potential inhibitors by in silico screening. J INDIAN CHEM SOC 2022. [DOI: 10.1016/j.jics.2022.100531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
21
|
A single-molecule with multiple investigations: Synthesis, characterization, computational methods, inhibitory activity against Alzheimer's disease, toxicity, and ADME studies. Comput Biol Med 2022; 146:105514. [DOI: 10.1016/j.compbiomed.2022.105514] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/01/2022] [Accepted: 04/09/2022] [Indexed: 01/18/2023]
|
22
|
Zheng J, Xiao X, Qiu WR. DTI-BERT: Identifying Drug-Target Interactions in Cellular Networking Based on BERT and Deep Learning Method. Front Genet 2022; 13:859188. [PMID: 35754843 PMCID: PMC9213727 DOI: 10.3389/fgene.2022.859188] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/25/2022] [Indexed: 11/20/2022] Open
Abstract
Drug–target interactions (DTIs) are regarded as an essential part of genomic drug discovery, and computational prediction of DTIs can accelerate to find the lead drug for the target, which can make up for the lack of time-consuming and expensive wet-lab techniques. Currently, many computational methods predict DTIs based on sequential composition or physicochemical properties of drug and target, but further efforts are needed to improve them. In this article, we proposed a new sequence-based method for accurately identifying DTIs. For target protein, we explore using pre-trained Bidirectional Encoder Representations from Transformers (BERT) to extract sequence features, which can provide unique and valuable pattern information. For drug molecules, Discrete Wavelet Transform (DWT) is employed to generate information from drug molecular fingerprints. Then we concatenate the feature vectors of the DTIs, and input them into a feature extraction module consisting of a batch-norm layer, rectified linear activation layer and linear layer, called BRL block and a Convolutional Neural Networks module to extract DTIs features further. Subsequently, a BRL block is used as the prediction engine. After optimizing the model based on contrastive loss and cross-entropy loss, it gave prediction accuracies of the target families of G Protein-coupled receptors, ion channels, enzymes, and nuclear receptors up to 90.1, 94.7, 94.9, and 89%, which indicated that the proposed method can outperform the existing predictors. To make it as convenient as possible for researchers, the web server for the new predictor is freely accessible at: https://bioinfo.jcu.edu.cn/dtibert or http://121.36.221.79/dtibert/. The proposed method may also be a potential option for other DITs.
Collapse
Affiliation(s)
- Jie Zheng
- Computer Department, Jing-De-Zhen Ceramic Institute, Jing-De-Zhen, China
| | - Xuan Xiao
- Computer Department, Jing-De-Zhen Ceramic Institute, Jing-De-Zhen, China
| | - Wang-Ren Qiu
- Computer Department, Jing-De-Zhen Ceramic Institute, Jing-De-Zhen, China
| |
Collapse
|
23
|
Yeh SJ, Yeh TY, Chen BS. Systems Drug Discovery for Diffuse Large B Cell Lymphoma Based on Pathogenic Molecular Mechanism via Big Data Mining and Deep Learning Method. Int J Mol Sci 2022; 23:ijms23126732. [PMID: 35743172 PMCID: PMC9224183 DOI: 10.3390/ijms23126732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/10/2022] [Accepted: 06/15/2022] [Indexed: 02/01/2023] Open
Abstract
Diffuse large B cell lymphoma (DLBCL) is an aggressive heterogeneous disease. The most common subtypes of DLBCL include germinal center b-cell (GCB) type and activated b-cell (ABC) type. To learn more about the pathogenesis of two DLBCL subtypes (i.e., DLBCL ABC and DLBCL GCB), we firstly construct a candidate genome-wide genetic and epigenetic network (GWGEN) by big database mining. With the help of two DLBCL subtypes’ genome-wide microarray data, we identify their real GWGENs via system identification and model order selection approaches. Afterword, the core GWGENs of two DLBCL subtypes could be extracted from real GWGENs by principal network projection (PNP) method. By comparing core signaling pathways and investigating pathogenic mechanisms, we are able to identify pathogenic biomarkers as drug targets for DLBCL ABC and DLBCL GCD, respectively. Furthermore, we do drug discovery considering drug-target interaction ability, drug regulation ability, and drug toxicity. Among them, a deep neural network (DNN)-based drug-target interaction (DTI) model is trained in advance to predict potential drug candidates holding higher probability to interact with identified biomarkers. Consequently, two drug combinations are proposed to alleviate DLBCL ABC and DLBCL GCB, respectively.
Collapse
|
24
|
Nadh AG, Revikumar A, Sudhakaran P, Nair AS. Identification of potential lead compounds against BACE1 through in-silico screening of phytochemicals of Medhya rasayana plants for Alzheimer's disease management. Comput Biol Med 2022; 145:105422. [DOI: 10.1016/j.compbiomed.2022.105422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/11/2022] [Accepted: 03/17/2022] [Indexed: 11/03/2022]
|
25
|
Tahir T, Tabassum R, Javed Q, Ali A, Ashfaq M, Shahzad MI. Synthesis, kinetics, structure-activity relationship and in silico ADME studies of new diazenyl azo-phenol derivatives against urease, SARS-CoV-2 main protease (Mpro) and ribosomal protein S1 (RpsA) of Mycobacterium tuberculosis. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
26
|
Valverde TL, Sampiron EG, Montaholi DC, Baldin VP, Insaurralde DD, Alves-Olher VG, Siqueira VL, Caleffi-Ferracioli KR, Cardoso RF, Vandresen F, Scodro RB. 3,5-dinitrobenzoylhydrazone derivatives as a scaffold for antituberculosis drug development. Future Microbiol 2022; 17:267-280. [PMID: 35164529 DOI: 10.2217/fmb-2021-0119] [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: 11/21/2022] Open
Abstract
Background: The development of drugs is essential to eradicate tuberculosis. Materials & methods: Sixteen 3,5-dinitrobenzoylhydrazone (2-17) derivatives and their synthetic precursors 3,5-dinitrobenzoylhydrazide (1) and methyl ester (18) were screened for their anti-Mycobacterium tuberculosis (Mtb) potential. Results: Twelve compounds had minimum inhibitory concentration (MIC) ranging from 0.24 to 7.8 μg/ml against the Mtb strain. The activity was maintained in multidrug-resistant Mtb clinical isolates. Only compound (17) showed activity against nontuberculous mycobacteria. The compounds exhibited a limited spectrum of activity, with an MIC >500 μg/ml against Gram-positive and -negative bacteria. Compounds (2), (5) and (11) showed a synergistic effect with rifampicin. An excellent selectivity index value was found, with values reaching 583.33. Conclusion: 3,5-dinitrobenzoylhydrazone derivatives could be considered as a scaffold for the development of antituberculosis drugs.
Collapse
Affiliation(s)
- Tamires L Valverde
- Postgraduate Program in Health Sciences, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
| | - Eloísa G Sampiron
- Postgraduate Program in Health Sciences, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
| | - Débora C Montaholi
- Postgraduate Program in Health Sciences, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
| | - Vanessa P Baldin
- Postgraduate Program in Bioscience & Physiopathology, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
| | - Diego Dr Insaurralde
- Department of Chemistry, Federal Technological University of Paraná, Londrina, Paraná, 86036-370, Brazil
| | - Vanessa G Alves-Olher
- Department of Chemistry, Federal Institute of Paraná, Paranavaí, Paraná, 87703-536, Brazil
| | - Vera Ld Siqueira
- Postgraduate Program in Bioscience & Physiopathology, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
| | - Katiany R Caleffi-Ferracioli
- Postgraduate Program in Bioscience & Physiopathology, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
| | - Rosilene F Cardoso
- Postgraduate Program in Health Sciences, State University of Maringá, Maringá, Paraná, 87020-900, Brazil.,Postgraduate Program in Bioscience & Physiopathology, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
| | - Fábio Vandresen
- Department of Chemistry, Federal Technological University of Paraná, Londrina, Paraná, 86036-370, Brazil
| | - Regiane Bl Scodro
- Postgraduate Program in Health Sciences, State University of Maringá, Maringá, Paraná, 87020-900, Brazil
| |
Collapse
|
27
|
Bois FY, Tebby C, Brochot C. PBPK Modeling to Simulate the Fate of Compounds in Living Organisms. Methods Mol Biol 2022; 2425:29-56. [PMID: 35188627 DOI: 10.1007/978-1-0716-1960-5_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Pharmacokinetics study the fate of xenobiotics in a living organism. Physiologically based pharmacokinetic (PBPK) models provide realistic descriptions of xenobiotics' absorption, distribution, metabolism, and excretion processes. They model the body as a set of homogeneous compartments representing organs, and their parameters refer to anatomical, physiological, biochemical, and physicochemical entities. They offer a quantitative mechanistic framework to understand and simulate the time-course of the concentration of a substance in various organs and body fluids. These models are well suited for performing extrapolations inherent to toxicology and pharmacology (e.g., between species or doses) and for integrating data obtained from various sources (e.g., in vitro or in vivo experiments, structure-activity models). In this chapter, we describe the practical development and basic use of a PBPK model from model building to model simulations, through implementation with an easily accessible free software.
Collapse
Affiliation(s)
| | - Cleo Tebby
- INERIS, Unit of Experimental Toxicology and Modelling, Verneuil en Halatte, France
| | - Céline Brochot
- INERIS, Unit of Experimental Toxicology and Modelling, Verneuil en Halatte, France
| |
Collapse
|
28
|
Discovery of thiazolidin-4-one analogue as selective GSK-3β inhibitor through structure based virtual screening. Bioorg Med Chem Lett 2021; 52:128375. [PMID: 34560262 DOI: 10.1016/j.bmcl.2021.128375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 09/05/2021] [Accepted: 09/15/2021] [Indexed: 11/21/2022]
Abstract
GSK-3β directly phosphorylate tubulin binding site of tau protein, indicating its importance in tau aggregation and, therefore, in Alzheimer's disease pathology. New GSK-3β inhibitors were identified using a structure-based screening, ADMET analysis. These studies revealed that ZINC09036109, ZINC72371723, ZINC72371725, and ZINC01373165 approached optimal ADMET properties along with good MM-GBSA dG binding. Protein kinase assays of these compounds against eight disease-relevant kinases were performed. During disease-relevant kinase profiling, ZINC09036109 ((E)-2-((3,4-dimethylphenyl)imino)-5-(3-methoxy-4-(naphthalen-2-ylmethoxy)benzyl)thiazolidin-4-one) emerged as a selective GSK-3β inhibitor with more than 10-fold selectivity over other disease-relevant kinases. Molecular dynamics study of ZINC09036109 molecule revealed interactions with Ile62, Phe67, Val135, Leu188, Asp200 amino acid residues of the binding site of GSK-3β, which were highly comparable to the co-crystallized molecule and hence validating comparative better activity of this compound compared to overall screened molecules.
Collapse
|
29
|
Jebasingh Kores J, Antony Danish I, Sasitha T, Gershom Stuart J, Jimla Pushpam E, Winfred Jebaraj J. Spectral, NBO, NLO, NCI, aromaticity and charge transfer analyses of anthracene-9,10-dicarboxaldehyde by DFT. Heliyon 2021; 7:e08377. [PMID: 34825087 PMCID: PMC8605071 DOI: 10.1016/j.heliyon.2021.e08377] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/10/2021] [Accepted: 11/10/2021] [Indexed: 11/25/2022] Open
Abstract
Anthracene-9,10-dicarboxaldehyde (ADCA) is a polynuclear aromatic compound that has a planar structure with double bonds which are in conjugation. The molecule is subjected to theoretical investigation with DFT/B3LYP/6-311++G(d,p) basis set to find out the electronic structural properties and stability. Theoretical and experimental vibrational analyses are carried out. NBO studies predict that the molecule has high stability. NCI interaction studies reveal that Van der Waals force and steric effect are seen in the molecule. A shaded surface map with a projection of LOL analysis pointed out that electron depletion area is seen in this molecule. The tunnelling current is more in the boundary rings than the central ring. It is docked with the protein 4COF and the binding energy is found to be -6.6 kcal/mol. Electrons excitation analysis is performed and found that local excitation takes place for the lowest five excitations. The aromaticity of the molecule is also thoroughly investigated.
Collapse
Affiliation(s)
- J. Jebasingh Kores
- Department of Physics, Pope's College (Autonomous), Sawyerpuram, 628251, Tamilnadu, India
| | - I. Antony Danish
- Department of Chemistry, Sadakathullah Appa College (Autonomous), Tirunelveli, 627011, Tamilnadu, India
| | - T. Sasitha
- Department of Chemistry, St. John's College, Tirunelveli, 627002, Tamilnadu, India
| | - J. Gershom Stuart
- Department of Chemistry, St. John's College, Tirunelveli, 627002, Tamilnadu, India
| | - E. Jimla Pushpam
- Department of Chemistry, St. John's College, Tirunelveli, 627002, Tamilnadu, India
| | - J. Winfred Jebaraj
- Department of Chemistry, St. John's College, Tirunelveli, 627002, Tamilnadu, India
| |
Collapse
|
30
|
Alghamdi SS, Suliman RS, Alsaeed AS, Almutairi KK, Aljammaz NA, Altolayyan A, Ali R, Alhallaj A. Novel Anti-Tubulin Compounds from Trigonella foenum-graecum Seeds; Insights into In-vitro and Molecular Docking Studies. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:4195-4211. [PMID: 34675483 PMCID: PMC8502543 DOI: 10.2147/dddt.s320793] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/22/2021] [Indexed: 01/08/2023]
Abstract
Background Fenugreek, also known as Trigonella foenum-graecum L, is a natural plant that belongs to the Fabaceae family and has been known as a promising source of bioactive compounds. It has been widely used as traditional medicine since it has shown to lower blood glucose, manage cholesterol levels and further aid in the prevention and treatment of cancer. Herein, we aim to evaluate the anticancer activity of methanolic fenugreek seed extract against several cancer cell lines. Methods We sought to investigate the phytochemical classes present in multiple fenugreek seeds extracts using HPLC-DAD followed by LC/MS, predict and investigate anticancer activity using PASS online webserver, the CellTiter-Glo assay, evaluate ADME properties, and perform molecular docking for all bioactive compounds via Maestro software. Results Multiple extracts exhibited distinct phytochemical classes that demonstrated different biological activities. Fenugreek methanolic extract contains flavonoid chemical class, which showed the highest anticancer activity against the HCT8 cell line of colorectal cancer (IC50 of 8.83 μg/mL), followed by KAIMRC1 breast cancer cell line (IC50 of 35.06 μg/mL), HL60 leukemia cell line (37.80 μg/mL), MDA-MB-231 breast cancer cell line (38.51 μg/mL), and lastly, HCT116 colorectal cancer cell line with IC50 of 56.03 μg/mL. In contrast, the chloroform extract was inactive. The molecular docking study for all the bioactive compounds suggested that flavonoids F6 (−9.713 and −12.132), F7 (−10.166 and −12.411), and F11 (−10.084 and −13.516) possess the highest docking scores through SP and XP scores, respectively. Conclusion The obtained results confirm that the bioactive compounds present in fenugreek seeds exhibit anticancer activity against several cancer cells that can mediate via tubulin polymerization inhibition. Although our study has evaluated the anticancer potential of Trigonella foenum-graecum as a promising natural source for new anticancer agents, fenugreek biological activity needs further research and investigations on their mechanism of action and toxicity profile.
Collapse
Affiliation(s)
- Sahar Saleh Alghamdi
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Kingdom of Saudi Arabia.,Medical Research Core Facility and Platforms, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Rasha Saad Suliman
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Kingdom of Saudi Arabia.,Medical Research Core Facility and Platforms, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Amjad Sulaiman Alsaeed
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Kingdom of Saudi Arabia
| | - Khlood Khaled Almutairi
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Kingdom of Saudi Arabia
| | - Norah Abdulaziz Aljammaz
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Kingdom of Saudi Arabia
| | - Abdulelah Altolayyan
- Medical Research Core Facility and Platforms, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Rizwan Ali
- Medical Research Core Facility and Platforms, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Alshaimaa Alhallaj
- Medical Research Core Facility and Platforms, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
31
|
Pantaleão SQ, Fernandes PO, Gonçalves JE, Maltarollo VG, Honorio KM. Recent Advances in the Prediction of Pharmacokinetics Properties in Drug Design Studies: A Review. ChemMedChem 2021; 17:e202100542. [PMID: 34655454 DOI: 10.1002/cmdc.202100542] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/07/2021] [Indexed: 12/11/2022]
Abstract
This review presents the main aspects related to pharmacokinetic properties, which are essential for the efficacy and safety of drugs. This topic is very important because the analysis of pharmacokinetic aspects in the initial design stages of drug candidates can increase the chances of success for the entire process. In this scenario, experimental and in silico techniques have been widely used. Due to the difficulties encountered with the use of some experimental tests to determine pharmacokinetic properties, several in silico tools have been developed and have shown promising results. Therefore, in this review, we address the main free tools/servers that have been used in this area, as well as some cases of application. Finally, we present some studies that employ a multidisciplinary approach with synergy between in silico, in vitro, and in vivo techniques to assess ADME properties of bioactive substances, achieving successful results in drug discovery and design.
Collapse
Affiliation(s)
- Simone Q Pantaleão
- Centro de Ciências Naturais e Humanas, Institution Universidade Federal do ABC, 09210-580, Santo André, SP, Brazil
| | - Philipe O Fernandes
- Departamento de Produtos Farmacêuticos, Universidade Federal de Minas Gerais, 31270-901, Pampulha, MG, Brazil
| | - José Eduardo Gonçalves
- Departamento de Produtos Farmacêuticos, Universidade Federal de Minas Gerais, 31270-901, Pampulha, MG, Brazil
| | - Vinícius G Maltarollo
- Departamento de Produtos Farmacêuticos, Universidade Federal de Minas Gerais, 31270-901, Pampulha, MG, Brazil
| | - Kathia Maria Honorio
- Centro de Ciências Naturais e Humanas, Institution Universidade Federal do ABC, 09210-580, Santo André, SP, Brazil.,Escola de Artes, Ciências e Humanidades, Universidade de São Paulo, 03828-000, São Paulo, SP, Brazil
| |
Collapse
|
32
|
Alzahrani AY, Shaaban MM, Elwakil BH, Hamed MT, Rezki N, Aouad MR, Zakaria MA, Hagar M. Anti-COVID-19 activity of some benzofused 1,2,3-triazolesulfonamide hybrids using in silico and in vitro analyses. CHEMOMETRICS AND INTELLIGENT LABORATORY SYSTEMS : AN INTERNATIONAL JOURNAL SPONSORED BY THE CHEMOMETRICS SOCIETY 2021; 217:104421. [PMID: 34538993 PMCID: PMC8434689 DOI: 10.1016/j.chemolab.2021.104421] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/14/2021] [Accepted: 09/06/2021] [Indexed: 05/26/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a pandemic fatal infection with no known treatment. The severity of the disease and the fast viral mutations forced the scientific community to search for potential solution. Here in the present manuscript, some benzofused1,2,3triazolesulfonamide hybrids were synthesized and evaluated for their anti- SARS-CoV-2 activity using in silico prediction then the most potent compounds were assessed using in-Vitro analysis. The in-Silico study was assessed against RNA dependent RNA polymerase, Spike protein S1, Main protease (3CLpro) and 2'-O-methyltransferase (nsp16). It was found that 4b and 4c showed high binding scores against RNA dependent RNA polymerase reached -8.40 and -8.75 kcal/mol, respectively compared to the approved antiviral (remdesivir -6.77 kcal/mol). Upon testing the binding score with SARS-CoV-2 Spike protein it was revealed that 4c exhibited the highest score (-7.22 kcal/mol) compared to the reference antibacterial drug Ceftazidime (-6.36 kcal/mol). Surprisingly, the two compounds 4b and 4c showed the highest binding scores against SARS-CoV-2 3CLpro (-8.75, -8.48 kcal/mol, respectively) and nsp16 (- 8.84 and - 8.89 kcal/mol, respectively) displaying many types of interaction with all the enzymes binding sites. The derivatives 4b and 4c were examined in vitro for their potential anti-SARS-CoV-2 and it was revealed that 4c was the most promising compound with IC50 reached 758.8108 mM and complete (100%) inhibition of the binding of SARS-CoV-2 virus to human ACE2 can be accomplished by using 0.01 mg.
Collapse
Affiliation(s)
- Abdullah Y Alzahrani
- Department of Chemistry, Faculty of Science and Arts, King Khalid University, Mohail, Assir, Saudi Arabia
| | - Marwa M Shaaban
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Bassma H Elwakil
- Department of Medical Laboratory Technology, Faculty of Applied Health Sciences Technology, Pharos University in Alexandria, Alexandria, Egypt
| | - Moaaz T Hamed
- Industrial Microbiology and Applied Chemistry Program, Department of Botany & Microbiology, Faculty of Science, Alexandria University, Alexandria, 21568, Egypt
| | - Nadjet Rezki
- Department of Chemistry, College of Science, Taibah University, Al-Madinah Al-Munawarah, 30002, Saudi Arabia
| | - Mohamed R Aouad
- Department of Chemistry, College of Science, Taibah University, Al-Madinah Al-Munawarah, 30002, Saudi Arabia
| | - Mohamed A Zakaria
- Department of Chemistry, College of Sciences, Taibah University, Yanbu, 30799, Saudi Arabia
| | - Mohamed Hagar
- Department of Chemistry, College of Sciences, Taibah University, Yanbu, 30799, Saudi Arabia
- Department of Chemistry, Faculty of Science, Alexandria University, Alexandria, 21321, Egypt
| |
Collapse
|
33
|
Ramirez DA, Federici MF, Altamirano JC, Camargo AB, Luco JM. Permeability Data of Organosulfur Garlic Compounds Estimated by Immobilized Artificial Membrane Chromatography: Correlation Across Several Biological Barriers. Front Chem 2021; 9:690707. [PMID: 34616711 PMCID: PMC8488277 DOI: 10.3389/fchem.2021.690707] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/27/2021] [Indexed: 01/11/2023] Open
Abstract
Among healthy vegetables, those of the genus Allium stand out. Antioxidant and anti-inflammatory properties have been associated with these vegetables, attributed mainly to organosulfur compounds (OSCs). In turn, they are linked to a protective effect counteracting cardiovascular disease development. Now, to really ensure the bioactive efficacy of the said compounds once consumed, it is necessary to previously evaluate the ADME (absorption, distribution, metabolism, and excretion) profile. Alternatively, in vitro and in silico methods attempt to avoid or reduce experimental animals' use and provide preliminary information on drugs' ability to overcome the various biological barriers inherent in the ADME process. In this sense, in silico methods serve to provide primary information on drugs' bioavailability mechanisms. High-performance liquid chromatography (HPLC) using a stationary phase composed of phospholipids, the so-called immobilized artificial membrane (IAM), has been widely recognized as a valuable alternative method to extract and quantify information about the structure and physicochemical properties of organic compounds which are extensively used in studies of quantitative structure-activity relationships (QSARs). In the present study, the chromatographic capacity factors (log k' (IAM)) for 28 OSCs were determined by IAM-HPLC. In order to evaluate the ability of the IAM phase in assessing lipophilicity of the compounds under study, several quantitative structure-retention relationships (QSRRs) were derived from exploring fundamental intermolecular interactions that govern the retention of compounds under study on IAM phases. As expected, the hydrophobic factors are of prime importance for the IAM retention of these compounds. However, the molecular flexibility and specific polar interactions expressed by several electronic descriptors (relative negative charge, RNCG, and Mulliken electronegativity) are also involved. We also evaluated the IAM phase ability to assess several ADME parameters for the OSCs under study obtained using the SwissADME web tool integrated into the SwissDrugDesign workspace and the PreADMET web tool. The human gastrointestinal absorption (HIA), blood-brain barrier (BBB) permeation, and skin permeability were investigated through QSAR modeling, using several chemometric approaches. The ADME properties under study are strongly dependent on hydrophobic factors as expressed by log k'(IAM), which provide evidence for the great potential of the IAM phases in the development of QSAR models.
Collapse
Affiliation(s)
- Daniela Andrea Ramirez
- Instituto de Biología Agrícola de Mendoza (IBAM), CONICET-Mendoza, Mendoza, Argentina
- Laboratorio de Cromatografía para Agroalimentos, Facultad de Ciencias Agrarias, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - María Fernanda Federici
- Laboratorio de Cromatografía para Agroalimentos, Facultad de Ciencias Agrarias, Universidad Nacional de Cuyo, Mendoza, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Jorgelina Cecilia Altamirano
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina
- Instituto Argentino de Nivología, Glaciología y Ciencias Ambientales (IANIGLA), CONICET-Mendoza, Mendoza, Argentina
| | - Alejandra Beatriz Camargo
- Instituto de Biología Agrícola de Mendoza (IBAM), CONICET-Mendoza, Mendoza, Argentina
- Laboratorio de Cromatografía para Agroalimentos, Facultad de Ciencias Agrarias, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Juan María Luco
- Área de Química Analítica, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| |
Collapse
|
34
|
Dwivedy A, Mariadasse R, Ahmad M, Chakraborty S, Kar D, Tiwari S, Bhattacharyya S, Sonar S, Mani S, Tailor P, Majumdar T, Jeyakanthan J, Biswal BK. Characterization of the NiRAN domain from RNA-dependent RNA polymerase provides insights into a potential therapeutic target against SARS-CoV-2. PLoS Comput Biol 2021; 17:e1009384. [PMID: 34516563 PMCID: PMC8478224 DOI: 10.1371/journal.pcbi.1009384] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 09/28/2021] [Accepted: 08/26/2021] [Indexed: 12/14/2022] Open
Abstract
Apart from the canonical fingers, palm and thumb domains, the RNA dependent RNA polymerases (RdRp) from the viral order Nidovirales possess two additional domains. Of these, the function of the Nidovirus RdRp associated nucleotidyl transferase domain (NiRAN) remains unanswered. The elucidation of the 3D structure of RdRp from the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), provided the first ever insights into the domain organisation and possible functional characteristics of the NiRAN domain. Using in silico tools, we predict that the NiRAN domain assumes a kinase or phosphotransferase like fold and binds nucleoside triphosphates at its proposed active site. Additionally, using molecular docking we have predicted the binding of three widely used kinase inhibitors and five well characterized anti-microbial compounds at the NiRAN domain active site along with their drug-likeliness. For the first time ever, using basic biochemical tools, this study shows the presence of a kinase like activity exhibited by the SARS-CoV-2 RdRp. Interestingly, a well-known kinase inhibitor- Sorafenib showed a significant inhibition and dampened viral load in SARS-CoV-2 infected cells. In line with the current global COVID-19 pandemic urgency and the emergence of newer strains with significantly higher infectivity, this study provides a new anti-SARS-CoV-2 drug target and potential lead compounds for drug repurposing against SARS-CoV-2. The on-going coronavirus disease 2019 (COVID-19) pandemic caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is significantly affecting the world health. Unfortunately, over 180 million cases of COVID-19 resulting in nearly 4 million deaths have been reported till June, 2021. In this study, using a combination of bioinformatics, biochemical and mass spectrometry methods, we show that the Nidovirus RdRp associated Nucleotidyl transferase (NiRAN) domain of the RNA-dependent RNA polymerase (RdRp) of SARS-CoV-2 exhibits a kinase like activity. Additionally, we also show that few broad spectrum anti-cancer and anti-microbial drugs dampen this kinase like activity. Of note, Sorafenib, an FDA approved anti-cancer kinase inhibiting drug significantly reduces the SARS-CoV-2 load in cell lines. Our study suggests that NiRAN domain of the SARS-CoV-2 RdRp is indispensible for the successful viral life cycle and shows that abolishing this enzymatic function of RdRp by small molecule inhibitors may open novel avenues for COVID-19 therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sudipta Sonar
- Translational Health Science and Technology Institute, Faridabad, India
| | - Shailendra Mani
- Translational Health Science and Technology Institute, Faridabad, India
| | | | - Tanmay Majumdar
- National Institute of Immunology, New Delhi, India
- * E-mail: (TM); (JJ); (BKB)
| | - Jeyaraman Jeyakanthan
- Department of Bioinformatics, Alagappa University, Tamil Nadu, India
- * E-mail: (TM); (JJ); (BKB)
| | | |
Collapse
|
35
|
Novel oxazolones incorporated azo dye: Design, synthesis photophysical-DFT aspects and antimicrobial assessments with In-silico and In-vitro surveys. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY 2021. [DOI: 10.1016/j.jpap.2021.100032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
36
|
Yadav R, Imran M, Dhamija P, Suchal K, Handu S. Virtual screening and dynamics of potential inhibitors targeting RNA binding domain of nucleocapsid phosphoprotein from SARS-CoV-2. J Biomol Struct Dyn 2021; 39:4433-4448. [PMID: 32568013 PMCID: PMC7332875 DOI: 10.1080/07391102.2020.1778536] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 05/29/2020] [Indexed: 12/23/2022]
Abstract
The emergence of the coronavirus disease-2019 pandemic has led to an outbreak in the world. The SARS-CoV-2 is seventh and latest in coronavirus family with unique exonucleases for repairing any mismatches in newly transcribed genetic material. Therefore, drugs with novel additional mechanisms are required to simultaneously target and eliminate the virus. Thus, a newly deciphered N protein is taken as a target that belongs to SARS-CoV-2. They play a vital role in RNA transcription, viral replication and new virion formation. This study used virtual screening, molecular modeling and docking of the 8987 ligands from Asinex and PubChem databases against this novel target protein. Three hotspot sites having DScore ≥1 (Site 1, Site 2 and Site 3) for ligand binding were selected. Subsequently, high throughput screening, standard precision and extra precision docking process and molecular dynamics concluded three best drugs from two libraries. Two antiviral moieties from Asinex databases (5817 and 6799) have docking scores of -10.29 and -10.156; along with their respective free binding energies (ΔG bind) of -51.96 and -64.36 on Site 3. The third drug, Zidovudine, is from PubChem database with docking scores of -9.75 with its binding free energies (ΔG bind) of -59.43 on Site 3. The RMSD and RMSF were calculated for all the three drugs through molecular dynamics simulation studies for 50 ns. Zidovudine shows a very stable interaction with fluctuation starting at 2.4 Å on 2 ns and remained stable at 3 Å from 13 to 50 ns. Thus, paving the way for further biological validation as a potential treatment.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Rohitash Yadav
- Department of Pharmacology, All India Institute of Medical Sciences, Rishikesh, India
| | - Mohammed Imran
- Department of Pharmacology, College of Medicine, Shaqra University, Shaqra, Kingdom of Saudi Arabia
| | - Puneet Dhamija
- Department of Pharmacology, All India Institute of Medical Sciences, Rishikesh, India
| | - Kapil Suchal
- School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Shailendra Handu
- Department of Pharmacology, All India Institute of Medical Sciences, Rishikesh, India
| |
Collapse
|
37
|
Choudhury S, Jena S, Sahoo DK, Shekh S, Kar RK, Dhakad A, Gowd KH, Biswal HS. Gram-Scale Synthesis of 1,8-Naphthyridines in Water: The Friedlander Reaction Revisited. ACS OMEGA 2021; 6:19304-19313. [PMID: 34337267 PMCID: PMC8320145 DOI: 10.1021/acsomega.1c02798] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 06/29/2021] [Indexed: 06/13/2023]
Abstract
The products of the Friedlander reaction, i.e., 1,8-naphthyridines, have far-reaching impacts in materials science, chemical biology, and medicine. The reported synthetic methodologies elegantly orchestrate the diverse synthetic routes of naphthyridines but require harsh reaction conditions, organic solvents, and expensive metal catalysts. Here, we introduce gram-scale synthesis of 1,8-naphthyridines in water using an inexpensive and biocompatible ionic liquid (IL) as a catalyst. This is the first-ever report on the synthesis of naphthyridines in water. This is a one-step reaction, and the product separation is relatively easy. The choline hydroxide (ChOH) is used as a metal-free, nontoxic, and water-soluble catalyst. In comparison to other catalysts reported in the literature, ChOH has the advantage of forming an additional hydrogen bond with the reactants, which is the vital step for the reaction to happen in water. Density functional theory (DFT) and noncovalent interaction (NCI) plot index analysis provide the plausible reaction mechanism for the catalytic cycle and confirm that hydrogen bonds with the IL catalyst are pivotal to facilitate the reaction. Molecular docking and molecular dynamics (MD) simulations are also performed to demonstrate the potentialities of the newly synthesized products as drugs. Through MD simulations, it was established that the tetrahydropyrido derivative of naphthyridine (10j) binds to the active sites of the ts3 human serotonin transporter (hSERT) (PDB ID: 6AWO) without perturbing the secondary structure, suggesting that 10j can be a potential preclinical drug candidate for hSERT inhibition and depression treatment.
Collapse
Affiliation(s)
- Shubhranshu
Shekhar Choudhury
- School
of Chemical Sciences, National Institute
of Science Education and Research (NISER), PO-Bhimpur-Padanpur, Via-Jatni,
Khurda, 752050 Bhubaneswar, India
- Homi
Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Subhrakant Jena
- School
of Chemical Sciences, National Institute
of Science Education and Research (NISER), PO-Bhimpur-Padanpur, Via-Jatni,
Khurda, 752050 Bhubaneswar, India
- Homi
Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Dipak Kumar Sahoo
- School
of Chemical Sciences, National Institute
of Science Education and Research (NISER), PO-Bhimpur-Padanpur, Via-Jatni,
Khurda, 752050 Bhubaneswar, India
- Homi
Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Shamasoddin Shekh
- Department
of Chemistry, Central University of Karnataka, Kalaburagi 585367, Karnataka, India
| | - Rajiv K. Kar
- Fritz
Haber Center for Molecular Dynamics, Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Ambuj Dhakad
- School
of Chemical Sciences, National Institute
of Science Education and Research (NISER), PO-Bhimpur-Padanpur, Via-Jatni,
Khurda, 752050 Bhubaneswar, India
- Homi
Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| | - Konkallu Hanumae Gowd
- Department
of Chemistry, Central University of Karnataka, Kalaburagi 585367, Karnataka, India
| | - Himansu S. Biswal
- School
of Chemical Sciences, National Institute
of Science Education and Research (NISER), PO-Bhimpur-Padanpur, Via-Jatni,
Khurda, 752050 Bhubaneswar, India
- Homi
Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400094, India
| |
Collapse
|
38
|
Rajalakshmi R, Lalitha P, Sharma SC, Rajiv A, Chithambharan A, Ponnusamy A. In silico studies: Physicochemical properties, drug score, toxicity predictions and molecular docking of organosulphur compounds against Diabetes mellitus. J Mol Recognit 2021; 34:e2925. [PMID: 34302410 DOI: 10.1002/jmr.2925] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/07/2021] [Accepted: 06/17/2021] [Indexed: 11/09/2022]
Abstract
Diabetes mellitus (DM) is a significant common metabolic disorder seen all over the world. In 2020, according to the International Diabetes Federation (IDF), out of 463 million people who have diabetes all over the world, 77 million belong to India. As per the statistical prediction, the affected numbers are probably expected to rise to 642 million by 2040. The commercially available anti-diabetic drugs in the market include metformin, sulphonyl urea, meglitinides, miglitol, acarbose, biguanides, and thiazolidinediones cause side effects like hypoglycaemia, dizziness, liver cell injury, digestive discomfort, neurological defects, etc. Hence, bioactive organosulphur based functional ligands are chosen in this study to arrive at a newer drug for DM. In this work, in silico analysis of organosulphur molecular descriptors like physicochemical properties, solubility, drug score, and toxicity predictions are evaluated using OSIRIS and Toxtree freeware. The essential parameters for discovering drugs for biopharmaceutical formulations viz the solubility of drugs and toxicity have been calculated. The protein target Dipeptidyl peptidase DPP4 (PID: 2RIP) was docked against energy minimised sulphur compounds using Hex 6.3. The results indicate that the drug likeliness of the molecule 4, that is, N-[(3,3-dimethyl piperidin-2-yl) methyl]-4-ethyl sulphonyl aniline is active with decreasing binding energy score (-212.24 Kcal mol-1 ) with no toxicity and also few sulphur compounds are active against diabetes compared to standard drug metformin (-158.33 Kcal mol-1 ). The best drug-like ligand N-[(3,3-dimethyl piperidin-2-yl) methyl]-4-ethyl sulphonyl aniline, was docked using commercial Maestro Schrodinger software to predict the results.
Collapse
Affiliation(s)
- Ravimoorthy Rajalakshmi
- Department of Chemistry, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, India
| | - Pottail Lalitha
- Associate Professor, Department of Chemistry and Coordinator, Bharat Ratna Prof. C.N.R. Rao Research Centre, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, India
| | | | - Asha Rajiv
- Department of Physics, Director IQAC, School of Science, SoS, B-II, Jain (Deemed-to-be-University), Bangalore, India
| | - Akhila Chithambharan
- Department of Chemistry, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, India
| | - Aruna Ponnusamy
- Department of Chemistry, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, India
| |
Collapse
|
39
|
Hu S, Xia D, Su B, Chen P, Wang B, Li J. A Convolutional Neural Network System to Discriminate Drug-Target Interactions. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:1315-1324. [PMID: 31514149 DOI: 10.1109/tcbb.2019.2940187] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Biological targets are most commonly proteins such as enzymes, ion channels, and receptors. They are anything within a living organism to bind with some other entities (like an endogenous ligand or a drug), resulting in change in their behaviors or functions. Exploring potential drug-target interactions (DTIs) are crucial for drug discovery and effective drug development. Computational methods were widely applied in drug-target interactions, since experimental methods are extremely time-consuming and resource-intensive. In this paper, we proposed a novel deep learning-based prediction system, with a new negative instance generation, to identify DTIs. As a result, our method achieved an accuracy of 0.9800 on our created dataset. Another dataset derived from DrugBank was used to further assess the generalization of the model, which yielded a good performance with accuracy of 0.8814 and AUC value of 0.9527 on the dataset. The outcome of our experimental results indicated that the proposed method, involving the credible negative generation, can be employed to discriminate the interactions between drugs and targets. Website: http://www.dlearningapp.com/web/DrugCNN.htm.
Collapse
|
40
|
Daoui O, Elkhattabi S, Chtita S, Elkhalabi R, Zgou H, Benjelloun AT. QSAR, molecular docking and ADMET properties in silico studies of novel 4,5,6,7-tetrahydrobenzo[D]-thiazol-2-Yl derivatives derived from dimedone as potent anti-tumor agents through inhibition of C-Met receptor tyrosine kinase. Heliyon 2021; 7:e07463. [PMID: 34296007 PMCID: PMC8282965 DOI: 10.1016/j.heliyon.2021.e07463] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/14/2021] [Accepted: 06/29/2021] [Indexed: 02/09/2023] Open
Abstract
A quantitative structure-activity relationship (QSAR) study is performed on 48 novel 4,5,6,7-tetrahydrobenzo[D]-thiazol-2 derivatives as anticancer agents capable of inhibiting c-Met receptor tyrosine kinase. The present study is conducted using multiple linear regression, multiple nonlinear regression and artificial neural networks. Three QSAR models are developed after partitioning the database into two sets (training and test) via the k-means method. The obtained values of the correlation coefficients by the three developed QSAR models are 0.90, 0.91 and 0.92, respectively. The resulting models are validated by using the external validation, leave-one-out cross-validation, Y-randomization test, and applicability domain methods. Moreover, we evaluated the drug-likeness properties of seven selected molecules based on their observed high activity to inhibit the c-Met receptor. The results of the evaluation showed that three of the seven compounds present drug-like characteristics. In order to identify the important active sites for the inhibition of the c-Met receptor responsible for the development of cancer cell lines, the crystallized form of the Crizotinib-c-Met complex (PDB code: 2WGJ) is used. These sites are used as references in the molecular docking test of the three selected molecules to identify the most suitable molecule for use as a new c-Met inhibitor. A comparative study is conducted based on the evaluation of the predicted properties of ADMET in silico between the candidate molecule and the Crizotinib inhibitor. The comparison results show that the selected molecule can be used as new anticancer drug candidates.
Collapse
Affiliation(s)
- Ossama Daoui
- Laboratory of Engineering, Systems and Applications, National School of Applied Sciences, Sidi Mohamed Ben Abdellah-Fez University, Fez, Morocco
| | - Souad Elkhattabi
- Laboratory of Engineering, Systems and Applications, National School of Applied Sciences, Sidi Mohamed Ben Abdellah-Fez University, Fez, Morocco
| | - Samir Chtita
- Laboratory of Physical Chemistry of Materials, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, Casablanca P.O. Box 7955, Morocco
| | - Rachida Elkhalabi
- Laboratory of Applied Organic Chemistry, Faculty of Sciences and Technologies, Sidi Mohamed Ben Abdellah-Fez University, Fez, Morocco
| | - Hsaine Zgou
- Polydisciplinary Faculty of Ouarzazate, Ibn Zohr University, Agadir, Morocco
| | - Adil Touimi Benjelloun
- LIMAS, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdallah University, Fez, Morocco
| |
Collapse
|
41
|
Sampiron EG, Costacurta GF, Calsavara LL, Baldin VP, Silva GVD, Alves Olher VG, Ferraretto LH, Caleffi-Ferraciolli KR, Cardoso RF, Siqueira VLD, Vandresen F, Scodro RBDL. In Vitro and In Silico Evaluations of Anti- Mycobacterium tuberculosis Activity of Benzohydrazones Compounds. Microb Drug Resist 2021; 27:1564-1577. [PMID: 33913749 DOI: 10.1089/mdr.2020.0392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Tuberculosis is a disease caused by Mycobacterium tuberculosis, with high mortality rates and an extended treatment that causes severe adverse effects, besides the emergence of resistant bacteria. Therefore, the search for new compounds with anti-M. tuberculosis activity has considerably increased in recent years. In this context, benzohydrazones are significant compounds that have antifungal and antibacterial action. This study aimed at evaluating the in vitro activity of 18 benzohydrazones against M. tuberculosis. Compounds' cytotoxicity, inhibition of M. tuberculosis efflux pumps, and in silico absorption, distribution, metabolism, excretion, and toxicity (ADMET) assays were also performed. In general, the minimum inhibitory concentration values for the standard M. tuberculosis H37Rv strain ranged from 7.8 to 250 μg/mL, and some compounds were not toxic to any of the cells tested (IC50 ranged from 18.0 to 302.5 μg/mL). In addition, compounds (4) and (7) showed to be possible efflux pump inhibitors. In ADMET assays, all benzohydrazones had high gastrointestinal absorption. Most of the compounds were able to overcome the blood-brain barrier, and no compounds had irritant or tumorigenic effects. Compounds (1), (3), (9), (12), and (15) stood out for showing good activities, both in vitro and in silico assays.
Collapse
Affiliation(s)
- Eloísa Gibin Sampiron
- Postgraduate Program in Health Sciences, State University of Maringá, Maringá, Brazil
| | | | - Leonora Lacerda Calsavara
- Postgraduate Program in Bioscience and Physiopathology, State University of Maringá, Maringá, Brazil
| | - Vanessa Pietrowski Baldin
- Postgraduate Program in Bioscience and Physiopathology, State University of Maringá, Maringá, Brazil
| | - Gabrielle Vaz da Silva
- Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Brazil
| | | | | | - Katiany Rizzieri Caleffi-Ferraciolli
- Postgraduate Program in Bioscience and Physiopathology, State University of Maringá, Maringá, Brazil.,Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Brazil
| | - Rosilene Fressatti Cardoso
- Postgraduate Program in Health Sciences, State University of Maringá, Maringá, Brazil.,Postgraduate Program in Bioscience and Physiopathology, State University of Maringá, Maringá, Brazil.,Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Brazil
| | - Vera Lucia Dias Siqueira
- Postgraduate Program in Bioscience and Physiopathology, State University of Maringá, Maringá, Brazil.,Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Brazil
| | - Fábio Vandresen
- Department of Chemistry, Federal Technologic University of Paraná, Londrina, Brazil
| | - Regiane Bertin de Lima Scodro
- Postgraduate Program in Health Sciences, State University of Maringá, Maringá, Brazil.,Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Brazil
| |
Collapse
|
42
|
Fayed EA, Bayoumi AH, Saleh AS, Ezz Al-Arab EM, Ammar YA. In vivo and in vitro anti-inflammatory, antipyretic and ulcerogenic activities of pyridone and chromenopyridone derivatives, physicochemical and pharmacokinetic studies. Bioorg Chem 2021; 109:104742. [PMID: 33647742 DOI: 10.1016/j.bioorg.2021.104742] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 01/07/2023]
Abstract
Throughout this study, we present the victorious synthesis of a novel class of 2(1H)-pyridone molecules, bearing a 4-hydroxyphenyl moiety through a one-pot reaction of 2-cyano-N-(4-hydroxyphenyl)acetamide with cyanoacetamide, acetylacetone or ethyl acetoacetate, and their corresponding aldehydes. In addition, the chromene moiety was introduced into the pyridine skeleton through the cyclization of the cyanoacetamide 2 with salicylaldehyde, followed by treatment with malononitrile, ethyl cyanoacetate, and cyanoacetamide, in order to improve their biological behaviour. Due to their anti-inflammatory, ulcerogenic, and antipyretic characters, the target molecules have undergone in-vitro and in-vivo examination, that display promising results. Moreover, in order to predict the physicochemical and ADME traits of all synthesized compounds and standard reference drugs, paracetamol and phenylbutazone, the in-silico prediction methodology was provided.
Collapse
Affiliation(s)
- Eman A Fayed
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11754, Egypt.
| | - Ashraf H Bayoumi
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11754, Egypt
| | - Aya S Saleh
- National Organization for Drug Control and Research, Cairo, Egypt
| | | | - Yousry A Ammar
- Chemistry Department, Faculty of Science (Boys), Al-Azhar University, Cairo 11754, Egypt.
| |
Collapse
|
43
|
Sharma P, Singh M, Sharma S. Molecular docking analysis of pyruvate kinase M2 with a potential inhibitor from the ZINC database. Bioinformation 2021; 17:139-146. [PMID: 34393429 PMCID: PMC8340708 DOI: 10.6026/97320630017139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/31/2020] [Accepted: 01/26/2021] [Indexed: 12/02/2022] Open
Abstract
The pyruvate kinase M2 isoform (PKM2) is linked with cancer. Therefore, it is of interest to document the molecular docking analysis of Pyruvate Kinase M2 (PDB ID: 4G1N) with potential activators from the ZINC database. Thus, we document the optimal molecular docking features of a compound having ID ZINC000034285235 with PKM2 for further consideration.
Collapse
Affiliation(s)
- Pankaj Sharma
- Department of Biotechnology, UIET, Maharshi Dayanand University Rohtak Haryana, India
| | - Manvender Singh
- Department of Biotechnology, UIET, Maharshi Dayanand University Rohtak Haryana, India
| | | |
Collapse
|
44
|
Sumon TA, Hussain MA, Hasan MT, Hasan M, Jang WJ, Bhuiya EH, Chowdhury AAM, Sharifuzzaman SM, Brown CL, Kwon HJ, Lee EW. A Revisit to the Research Updates of Drugs, Vaccines, and Bioinformatics Approaches in Combating COVID-19 Pandemic. Front Mol Biosci 2021; 7:585899. [PMID: 33569389 PMCID: PMC7868442 DOI: 10.3389/fmolb.2020.585899] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/17/2020] [Indexed: 12/19/2022] Open
Abstract
A new strain of coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) responsible for the coronavirus disease 2019 (COVID-19) pandemic was first detected in the city of Wuhan in Hubei province, China in late December 2019. To date, more than 1 million deaths and nearly 57 million confirmed cases have been recorded across 220 countries due to COVID-19, which is the greatest threat to global public health in our time. Although SARS-CoV-2 is genetically similar to other coronaviruses, i.e., SARS and Middle East respiratory syndrome coronavirus (MERS-CoV), no confirmed therapeutics are yet available against COVID-19, and governments, scientists, and pharmaceutical companies worldwide are working together in search for effective drugs and vaccines. Repurposing of relevant therapies, developing vaccines, and using bioinformatics to identify potential drug targets are strongly in focus to combat COVID-19. This review deals with the pathogenesis of COVID-19 and its clinical symptoms in humans including the most recent updates on candidate drugs and vaccines. Potential drugs (remdesivir, hydroxychloroquine, azithromycin, dexamethasone) and vaccines [mRNA-1273; measles, mumps and rubella (MMR), bacille Calmette-Guérin (BCG)] in human clinical trials are discussed with their composition, dosage, mode of action, and possible release dates according to the trial register of US National Library of Medicines (clinicaltrials.gov), European Union (clinicaltrialsregister.eu), and Chinese Clinical Trial Registry (chictr.org.cn) website. Moreover, recent reports on in silico approaches like molecular docking, molecular dynamics simulations, network-based identification, and homology modeling are included, toward repurposing strategies for the use of already approved drugs against newly emerged pathogens. Limitations of effectiveness, side effects, and safety issues of each approach are also highlighted. This review should be useful for the researchers working to find out an effective strategy for defeating SARS-CoV-2.
Collapse
Affiliation(s)
- Tofael Ahmed Sumon
- Department of Fish Health Management, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Md. Ashraf Hussain
- Department of Fisheries Technology and Quality Control, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Md. Tawheed Hasan
- Department of Aquaculture, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Mahmudul Hasan
- Department of Pharmaceuticals and Industrial Biotechnology, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Won Je Jang
- Department of Biotechnology, Pukyong National University, Busan, South Korea
| | | | | | - S. M. Sharifuzzaman
- Institute of Marine Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Christopher Lyon Brown
- World Fisheries University Pilot Programme, Pukyong National University, Busan, South Korea
| | - Hyun-Ju Kwon
- Biopharmaceutical Engineering Major, Division of Applied Bioengineering, Dong-Eui University, Busan, South Korea
| | - Eun-Woo Lee
- Biopharmaceutical Engineering Major, Division of Applied Bioengineering, Dong-Eui University, Busan, South Korea
| |
Collapse
|
45
|
An JY, Meng FR, Yan ZJ. An efficient computational method for predicting drug-target interactions using weighted extreme learning machine and speed up robot features. BioData Min 2021; 14:3. [PMID: 33472664 PMCID: PMC7816443 DOI: 10.1186/s13040-021-00242-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 01/10/2021] [Indexed: 01/09/2023] Open
Abstract
Background Prediction of novel Drug–Target interactions (DTIs) plays an important role in discovering new drug candidates and finding new proteins to target. In consideration of the time-consuming and expensive of experimental methods. Therefore, it is a challenging task that how to develop efficient computational approaches for the accurate predicting potential associations between drug and target. Results In the paper, we proposed a novel computational method called WELM-SURF based on drug fingerprints and protein evolutionary information for identifying DTIs. More specifically, for exploiting protein sequence feature, Position Specific Scoring Matrix (PSSM) is applied to capturing protein evolutionary information and Speed up robot features (SURF) is employed to extract sequence key feature from PSSM. For drug fingerprints, the chemical structure of molecular substructure fingerprints was used to represent drug as feature vector. Take account of the advantage that the Weighted Extreme Learning Machine (WELM) has short training time, good generalization ability, and most importantly ability to efficiently execute classification by optimizing the loss function of weight matrix. Therefore, the WELM classifier is used to carry out classification based on extracted features for predicting DTIs. The performance of the WELM-SURF model was evaluated by experimental validations on enzyme, ion channel, GPCRs and nuclear receptor datasets by using fivefold cross-validation test. The WELM-SURF obtained average accuracies of 93.54, 90.58, 85.43 and 77.45% on enzyme, ion channels, GPCRs and nuclear receptor dataset respectively. We also compared our performance with the Extreme Learning Machine (ELM), the state-of-the-art Support Vector Machine (SVM) on enzyme and ion channels dataset and other exiting methods on four datasets. By comparing with experimental results, the performance of WELM-SURF is significantly better than that of ELM, SVM and other previous methods in the domain. Conclusion The results demonstrated that the proposed WELM-SURF model is competent for predicting DTIs with high accuracy and robustness. It is anticipated that the WELM-SURF method is a useful computational tool to facilitate widely bioinformatics studies related to DTIs prediction.
Collapse
Affiliation(s)
- Ji-Yong An
- Engineering Research Center of Mine Digitalization (China University of Mining and Technology), Ministry of Education, Xuzhou, China. .,School of Computer Science and Technology, China University of Mining and Technology, Xuzhou, 21116, Jiangsu, China.
| | - Fan-Rong Meng
- Engineering Research Center of Mine Digitalization (China University of Mining and Technology), Ministry of Education, Xuzhou, China.,School of Computer Science and Technology, China University of Mining and Technology, Xuzhou, 21116, Jiangsu, China
| | - Zi-Ji Yan
- Engineering Research Center of Mine Digitalization (China University of Mining and Technology), Ministry of Education, Xuzhou, China.,School of Computer Science and Technology, China University of Mining and Technology, Xuzhou, 21116, Jiangsu, China
| |
Collapse
|
46
|
Gao D, Chen Q, Zeng Y, Jiang M, Zhang Y. Applications of Machine Learning in Drug Target Discovery. Curr Drug Metab 2020; 21:790-803. [PMID: 32723266 DOI: 10.2174/1567201817999200728142023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 03/12/2020] [Accepted: 05/13/2020] [Indexed: 12/15/2022]
Abstract
Drug target discovery is a critical step in drug development. It is the basis of modern drug development because it determines the target molecules related to specific diseases in advance. Predicting drug targets by computational methods saves a great deal of financial and material resources compared to in vitro experiments. Therefore, several computational methods for drug target discovery have been designed. Recently, machine learning (ML) methods in biomedicine have developed rapidly. In this paper, we present an overview of drug target discovery methods based on machine learning. Considering that some machine learning methods integrate network analysis to predict drug targets, network-based methods are also introduced in this article. Finally, the challenges and future outlook of drug target discovery are discussed.
Collapse
Affiliation(s)
- Dongrui Gao
- School of Computer Science, Chengdu University of Information Technology, Chengdu 610225, China
| | - Qingyuan Chen
- School of Computer Science, Chengdu University of Information Technology, Chengdu 610225, China
| | - Yuanqi Zeng
- School of Computer Science, Chengdu University of Information Technology, Chengdu 610225, China
| | - Meng Jiang
- School of Mechanical Automotive Engineering, Nanyang Institute of Technology, Nanyang 473000, China
| | - Yongqing Zhang
- School of Computer Science, Chengdu University of Information Technology, Chengdu 610225, China
| |
Collapse
|
47
|
In vitro investigation of metabolic fate of α-mangostin and gartanin via skin permeation by LC-MS/MS and in silico evaluation of the metabolites by ADMET predictor™. BMC Complement Med Ther 2020; 20:359. [PMID: 33228689 PMCID: PMC7685627 DOI: 10.1186/s12906-020-03144-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/31/2020] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Mangosteen, Garciniam angostana L., is a juicy fruit commonly found in Thailand. The rinds of Garciniam angostana L.have been used as a traditional medicine for the treatment of trauma, diarrhea and skin infection. It is also used in dermatological product such as in cosmetics. The mangosteen pericarp can be used to extract valuable bioactive xanthone compounds such as α-mangostin and gartanin. This study is aimed to predict the metabolism of α-mangostin and gartanin using in silico and in vitro skin permeation strategies. METHODS Based on their 2D molecular structures, metabolites of those compounds were predicted in silico using ADMET Predictor™. The Km and Vmax, for 5 important recombinant CYP isozymes 1A2, 2C9, 2C19, 2D6 and 3A4 were predicted. Moreover, the in vitro investigation of metabolites produced during skin permeation using human epidermal keratinocyte cells, neonatal (HEKn cells) was performed by LC-MS/MS. RESULTS It was found that the results derived from in silico were in excellent alignment with those obtained from in vitro studies for both compounds. The prediction referred that gartanin and α-mangostin were the substrate of CYP1A2, 2C9, 2C19 and 3A. In the investigation of α-mangostin metabolites by LC-MS/MS system, the MW of the parent compound was increased from 411.200 to 459.185 Da. Therefore, α-mangostin might be metabolized via tri-oxidation process. The increased molecular weight of parent compound (397.200 to 477.157 Da) illustrated that gartanin might be conjugated to sulfated derivatives. CONCLUSIONS In all the studies, α-mangostin and gartanin were predicted to be. metabolized via phase I and phase II metabolism (sulfation), respectively.
Collapse
|
48
|
Li Y, Liu X, You Z, Li L, Guo J, Wang Z. A computational approach for predicting drug–target interactions from protein sequence and drug substructure fingerprint information. INT J INTELL SYST 2020. [DOI: 10.1002/int.22332] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Yang Li
- School of Computer Science & Cyberspace Security Hainan University Haikou China
| | - Xiao‐zhang Liu
- School of Computer Science & Cyberspace Security Hainan University Haikou China
| | - Zhu‐Hong You
- School of Information Engineering Xijing University Xi'an China
| | - Li‐Ping Li
- School of Information Engineering Xijing University Xi'an China
| | - Jian‐Xin Guo
- School of Information Engineering Xijing University Xi'an China
| | - Zheng Wang
- School of Information Engineering Xijing University Xi'an China
| |
Collapse
|
49
|
Ahmad M, Dwivedy A, Mariadasse R, Tiwari S, Kar D, Jeyakanthan J, Biswal BK. Prediction of Small Molecule Inhibitors Targeting the Severe Acute Respiratory Syndrome Coronavirus-2 RNA-dependent RNA Polymerase. ACS OMEGA 2020; 5:18356-18366. [PMID: 32743211 PMCID: PMC7391942 DOI: 10.1021/acsomega.0c02096] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/30/2020] [Indexed: 05/08/2023]
Abstract
The current COVID-19 outbreak warrants the design and development of novel anti-COVID therapeutics. Using a combination of bioinformatics and computational tools, we modelled the 3D structure of the RdRp (RNA-dependent RNA polymerase) of SARS-CoV2 (severe acute respiratory syndrome coronavirus-2) and predicted its probable GTP binding pocket in the active site. GTP is crucial for the formation of the initiation complex during RNA replication. This site was computationally targeted using a number of small molecule inhibitors of the hepatitis C RNA polymerase reported previously. Further optimizations suggested a lead molecule that may prove fruitful in the development of potent inhibitors against the RdRp of SARS-CoV2.
Collapse
Affiliation(s)
- Mohammed Ahmad
- National
Institute of Immunology, New Delhi 110067, India
| | | | - Richard Mariadasse
- Department
of Bioinformatics, Alagappa University, karaikudi 630004, Tamil Nadu, India
| | - Satish Tiwari
- National
Institute of Immunology, New Delhi 110067, India
| | - Deepsikha Kar
- National
Institute of Immunology, New Delhi 110067, India
| | - Jeyaraman Jeyakanthan
- Department
of Bioinformatics, Alagappa University, karaikudi 630004, Tamil Nadu, India
| | | |
Collapse
|
50
|
von Lilienfeld OA, Müller KR, Tkatchenko A. Exploring chemical compound space with quantum-based machine learning. Nat Rev Chem 2020; 4:347-358. [PMID: 37127950 DOI: 10.1038/s41570-020-0189-9] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2020] [Indexed: 12/16/2022]
Abstract
Rational design of compounds with specific properties requires understanding and fast evaluation of molecular properties throughout chemical compound space - the huge set of all potentially stable molecules. Recent advances in combining quantum-mechanical calculations with machine learning provide powerful tools for exploring wide swathes of chemical compound space. We present our perspective on this exciting and quickly developing field by discussing key advances in the development and applications of quantum-mechanics-based machine-learning methods to diverse compounds and properties, and outlining the challenges ahead. We argue that significant progress in the exploration and understanding of chemical compound space can be made through a systematic combination of rigorous physical theories, comprehensive synthetic data sets of microscopic and macroscopic properties, and modern machine-learning methods that account for physical and chemical knowledge.
Collapse
|