1
|
Marinho MM, da Rocha MN, Magalhães EP, Ribeiro LR, Roberto CHA, de Queiroz Almeida-Neto FW, Monteiro ML, Nunes JVS, de Menezes RRPPB, Marinho ES, de Lima Neto P, Martins AMC, Dos Santos HS. Insights of potential trypanocidal effect of the synthetic derivative (2E)-1-(4-aminophenyl)-3-(2,4-dichlorophenyl)prop-2-en-1-one: in vitro assay, MEV analysis, quantum study, molecular docking, molecular dynamics, MPO analysis, and predictive ADMET. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7797-7818. [PMID: 38722342 DOI: 10.1007/s00210-024-03138-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 04/30/2024] [Indexed: 10/04/2024]
Abstract
This study aims to evaluate the antitrypanosomiasis activity of a synthetic dichloro-substituted aminochalcone via in vitro assays against infected cell cultures, as well as a theoretical characterization of pharmacokinetics and pharmacodynamics against the protein targets of the evolutionary cycle of T. cruzi. The in vitro evaluation of parasite proliferation inhibition was performed via cytotoxicity analysis on mammalian host cells, effect on epimastigote and trypomastigote forms, and cell death analysis, while computer simulations characterized the electronic structure of (2E)-1-(4-aminophenyl)-3-(2,4-dichlorophenyl)prop-2-en-1-one (DCl), the mechanism of action against the proteins of the evolutionary cycle of T. cruzi: Cruzain, Trypanothione reductase, TcGAPDH, and CYP51 by molecular docking and dynamics and predictive pharmacokinetics by MPO-based ADMET. The in vitro tests showed that the DCl LC50 in order of 178.9 ± 23.9 was similar to the BZN, evidencing the effectiveness of chalcone against Trypomastigotes. Molecular docking and dynamics simulations suggest that DCl acts on the active site of the CYP51 receptor, with hydrogen interactions that showed a high degree of occupation, establishing a stable complex with the target. MPO analysis and ADMET prediction tests suggest that the compound presents an alignment between permeability and hepatic clearance, although it presents low metabolic stability. Chalcone showed stable pharmacodynamics against the CYP51 target, but can form reactive metabolites from N-conjugation and C = C epoxidation, as an indication of controlled oral dose, although the estimated LD50 rate > 500 mg/kg is a indicative of low incidence of lethality by ingestion, constituting a promising therapeutic strategy.
Collapse
Affiliation(s)
- Márcia Machado Marinho
- Department of Biological Chemistry, Regional University of Cariri, Crato, CE, Brazil
- Center for Exact Sciences and Technology, State University of Vale do Acaraú, Sobral, CE, Brazil
| | - Matheus Nunes da Rocha
- Center for Science and Technology, Postgraduate Program in Natural Sciences, State University of Ceará, Fortaleza, CE, Brazil
| | - Emanuel Paula Magalhães
- Department of Clinical and Toxicological Analysis, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Lyanna Rodrigues Ribeiro
- Department of Clinical and Toxicological Analysis, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Caio Henrique Alexandre Roberto
- Center for Science and Technology, Postgraduate Program in Natural Sciences, State University of Ceará, Fortaleza, CE, Brazil
| | | | - Marília Lopes Monteiro
- Department of Clinical and Toxicological Analysis, Federal University of Ceará, Fortaleza, CE, Brazil
| | - João Victor Serra Nunes
- Department of Clinical and Toxicological Analysis, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - Emmanuel Silva Marinho
- Center for Science and Technology, Postgraduate Program in Natural Sciences, State University of Ceará, Fortaleza, CE, Brazil
| | - Pedro de Lima Neto
- Department of Analytical Chemistry and Physical Chemistry, Federal University of Ceará, Campus do Pici, Fortaleza, CE, Brazil
| | - Alice Maria Costa Martins
- Department of Clinical and Toxicological Analysis, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Hélcio Silva Dos Santos
- Center for Exact Sciences and Technology, State University of Vale do Acaraú, Sobral, CE, Brazil.
| |
Collapse
|
2
|
Gabidia Torres OL, Loroño M, Paz Rojas JL, Garrido Schaeffer CJA, Linares Fuentes TC, Cordova Sintjago TC. Computational Study of the Kinetics and Mechanisms of Gas-Phase Decomposition of N-Diacetamides Using Density Functional Theory. Molecules 2024; 29:3833. [PMID: 39202912 PMCID: PMC11356786 DOI: 10.3390/molecules29163833] [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: 07/16/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024] Open
Abstract
In this research work, we examined the decomposition mechanisms of N-substituted diacetamides. We focused on the substituent effect on the nitrogen lone-pair electron delocalization, with electron-withdrawing and electron donor groups. DFT functionals used the following: B1LYP, B3PW91, CAMB3LYP, LC-BLYP, and X3LYP. Dispersion corrections (d3bj) with Becke-Johnson damping were applied when necessary to improve non-covalent interactions in the transition state. Pople basis sets with higher angular moments and def2-TZVP basis sets were also applied and were crucial for obtaining consistent thermodynamic parameters. The proposed mechanism involves a six-membered transition state with the extraction of an α hydrogen. Several conformers of N-diacetamides were used to account for the decrease in entropy in the transition state in the rate-determining state. All calculations, including natural bond orbital (NBO) analyses, were performed using the Gaussian16 computational package and its GaussView 6.0 visualizer, along with VMD and GNUPLOT software. The isosurfaces and IBSIs were calculated using MultiWFN and IGMPlot, respectively.
Collapse
Affiliation(s)
- Oswaldo Luis Gabidia Torres
- Departamento Académico de Fisicoquímica, Facultad de Química e Ingeniería Química, Universidad Nacional Mayor de San Marcos, Lima 15081, Peru
| | - Marcos Loroño
- Departamento Académico de Fisicoquímica, Facultad de Química e Ingeniería Química, Universidad Nacional Mayor de San Marcos, Lima 15081, Peru
| | - Jose Luis Paz Rojas
- Departamento Académico de Química Inorgánica, Facultad de Química e Ingeniería Química, Universidad Nacional Mayor de San Marcos, Lima 15081, Peru;
| | | | - Thais Cleofe Linares Fuentes
- Departamento Académico de Química Orgánica, Facultad de Química e Ingeniería Química, Universidad Nacional Mayor de San Marcos, Lima 15081, Peru;
| | | |
Collapse
|
3
|
Boonserm P, Khunrae P, Sutthibutpong T. A computational study on the molecular mechanisms of panduratin A as a potential inhibitor on SARS-CoV-2 protein targets. Heliyon 2023; 9:e12780. [PMID: 36628324 PMCID: PMC9816080 DOI: 10.1016/j.heliyon.2022.e12780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 12/24/2022] [Accepted: 12/30/2022] [Indexed: 01/08/2023] Open
Abstract
Panduratin A from Boesebergia rotunda was recently reported as a potent anti-SARS-CoV-2 compound. However, the molecular mechanisms underlying the inhibition by Panduratin A and its target remained unclear. Molecular docking calculations were performed between panduratin A and five important proteins, i.e., main protease (Mpro), papain-like protease (PLpro), receptor binding domain (RBD) of spike proteins, RNA-dependent-RNA-polymerase (RdRp), and 2'-O-methyltransferase (MTase). The estimated binding free energy and the interaction networks extracted from the best docking mode for each complex suggested that MTase was the most probable target for panduratin A inhibition. To further validate the ability of panduratin A to inhibit MTase, molecular dynamics (MD) simulations and binding free energy calculations were performed for panduratin A-MTase complex, in comparison with another MTase complex with sinefungin as a positive control. Chemical features of panduratin A and sinefungin were compared for their contribution in MTase binding. It was found that both molecules could bind to the S-Adenosyl methionine (SAM) binding pocket and prevent the SAM entrance co-substrate, which could eventually halt the function of MTase. Despite a slightly weaker binding free energy, the equilibrated positional binding of panduratin A was found at a closer distance to the active sites. Therefore, this study proposed MTase as a possible target of panduratin A, along with the mechanisms of inhibition, prompting another future in vitro study as a verification.
Collapse
Affiliation(s)
- Patamalai Boonserm
- Department of Microbiology, Faculty of Science, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
| | - Pongsak Khunrae
- Department of Microbiology, Faculty of Science, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
| | - Thana Sutthibutpong
- Department of Physics, Faculty of Science, King Mongkut's University of Technology Thonburi, Bangkok, Thailand,Center of Excellence in Theoretical and Computational Science (TACS-CoE), Faculty of Science, King Mongkut's University of Technology Thonburi, Bangkok, Thailand,Corresponding author. Theoretical and Computational Science Center (TaCS), Science Laboratory Building, Faculty of Science, King Mongkut's University of Technology Thonburi (KMUTT), 126 Pracha-Uthit Road, Bang Mod, Thrung Khru, Bangkok, Thailand.,
| |
Collapse
|
4
|
da Silva AW, Ferreira MKA, Pereira LR, Rebouças EL, Coutinho MR, Dos J, Lima R, Guedes MIF, Bandeira PN, Magalhães FEA, Menezes JESAD, Marinho MM, Teixeira AMR, Salles Trevisan MT, Dos Santos HS, Marinho ES. Combretum lanceolatum extract reverses anxiety and seizure behavior in adult zebrafish through GABAergic neurotransmission: an in vivo and in silico study. J Biomol Struct Dyn 2022; 40:9801-9814. [PMID: 34121622 DOI: 10.1080/07391102.2021.1935322] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Combretaceae are reported in the literature for presenting neuroprotective and anxiolytic effects in animal models. Combretum lanceolatum Pohl. has few scientific reports on its pharmacological effects. The aim of this study was to evaluate the anxiolytic and anticonvulsant effects of the ethanol extract from the leaves of C. lanceolatum Pohl. (EtFoCl) and its possible mechanism of GABAergic action in adult zebrafish. EtFoCl was subjected to determination of the total phenol concentration, identification of phytochemical flavonoids by HPLC and in vitro antioxidant activity test, open field test and 96-hour acute toxicity in zebrafish. Anxiolytic doses were tested for pentylenetetrazole-induced seizures in adult zebrafish. To study the mechanisms of action, molecular docking simulations were performed between the main phytochemicals and the GABAA receptor (anxiolytic activity) and carbonic anhydrase II (anticonvulsant). The non-toxic doses that caused motor impairment were assessed in acute and chronic anxiety using the light and dark test. EtFoCl had altered the animals' locomotion, presenting an effect similar to the anxiolytic and anticonvulsant. These effects were prevented with flumazenil (GABAA antagonist). The phytochemicals homoorientin and quercetin-3-O-galactoside coupling in a region close to that of the inhibitor diazepam (GABAA receptor). Regarding the anticonvulsant mechanism, Homoorientina and Isovitexina were identified as the most favorable for the complex form with the carbonic anhydrase enzyme. C. lanceolatum has pharmacological potential for the treatment of acute and chronic anxiety and seizures, which can be partially explained by an interaction with the GABAA receptor.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Antonio Wlisses da Silva
- Northeast Biotechnology Network, Graduate Program of Biotechnology, State University of Ceará, Fortaleza, Ceará, Brazil.,Itaperi Campus, Laboratory of Natural Products Chemistry - LQPN-S, State University of Ceará, Science and Technology Center (CCT), Fortaleza, Ceará, Brazil
| | - Maria Kueirislene A Ferreira
- Itaperi Campus, Laboratory of Natural Products Chemistry - LQPN-S, State University of Ceará, Science and Technology Center (CCT), Fortaleza, Ceará, Brazil
| | - Lucas Ramos Pereira
- Department of Organic and Inorganic Chemistry, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Emanuela L Rebouças
- Northeast Biotechnology Network, Graduate Program of Biotechnology, State University of Ceará, Fortaleza, Ceará, Brazil.,Itaperi Campus, Laboratory of Biotechnology and Molecular Biology, State University of Ceará, Health Sciences Center (CCS), Fortaleza, Ceará, Brazil
| | - Marnielle Rodrigues Coutinho
- Itaperi Campus, Laboratory of Biotechnology and Molecular Biology, State University of Ceará, Health Sciences Center (CCS), Fortaleza, Ceará, Brazil
| | | | - Reis Lima
- Itaperi Campus, Laboratory of Natural Products Chemistry - LQPN-S, State University of Ceará, Science and Technology Center (CCT), Fortaleza, Ceará, Brazil
| | - Maria Izabel Florindo Guedes
- Itaperi Campus, Laboratory of Biotechnology and Molecular Biology, State University of Ceará, Health Sciences Center (CCS), Fortaleza, Ceará, Brazil
| | - Paulo N Bandeira
- Postgraduate Program in Biological Chemistry, Department of Biological Chemistry, Regional University of Cariri, Crato, Ceará, Brazil
| | - Francisco Ernani A Magalhães
- Itaperi Campus, Laboratory of Natural Products Chemistry - LQPN-S, State University of Ceará, Science and Technology Center (CCT), Fortaleza, Ceará, Brazil.,Itaperi Campus, Laboratory of Biotechnology and Molecular Biology, State University of Ceará, Health Sciences Center (CCS), Fortaleza, Ceará, Brazil.,Department of Chemistry, Laboratory of Natural Products Bioprospecting and Biotechnology, CECITEC Campus, State University of Ceará, Tauá, Ceará, Brazil
| | - Jane Eire S A de Menezes
- Itaperi Campus, Laboratory of Natural Products Chemistry - LQPN-S, State University of Ceará, Science and Technology Center (CCT), Fortaleza, Ceará, Brazil
| | - Marcia Machado Marinho
- Faculty of Education, Science and Letters of Iguatu, State University of Ceará, Iguatu, Ceara, Brazil
| | - Alexandre Magno Rodrigues Teixeira
- Northeast Biotechnology Network, Graduate Program of Biotechnology, State University of Ceará, Fortaleza, Ceará, Brazil.,Postgraduate Program in Biological Chemistry, Department of Biological Chemistry, Regional University of Cariri, Crato, Ceará, Brazil
| | | | - Hélcio S Dos Santos
- Northeast Biotechnology Network, Graduate Program of Biotechnology, State University of Ceará, Fortaleza, Ceará, Brazil.,Itaperi Campus, Laboratory of Natural Products Chemistry - LQPN-S, State University of Ceará, Science and Technology Center (CCT), Fortaleza, Ceará, Brazil.,Postgraduate Program in Biological Chemistry, Department of Biological Chemistry, Regional University of Cariri, Crato, Ceará, Brazil
| | - Emmanuel Silva Marinho
- Faculty of Philosophy Dom Aureliano Matos, State University of Ceará, Limoeiro do Norte, Ceará, Brasil
| |
Collapse
|
5
|
Alves Borges Leal AL, Fonseca Bezerra C, Ferreira E Silva AK, Everson da Silva L, Bezerra LL, Almeida-Neto FW, Marinho EM, Celedonio Fernandes CF, Nunes da Rocha M, Marinho MM, Coutinho HDM, Barreto HM, Rafaela Freitas Dotto A, Amaral WD, Santos HSD, Lima-Neto PD, Marinho ES. Seasonal variation of the composition of essential oils from Piper cernuum Vell and Piper rivinoides Kunth, ADMET study, DFT calculations, molecular docking and dynamics studies of major components as potent inhibitors of the heterodimer methyltransferase complex NSP16-NSP10 SARS COV-2 protein. J Biomol Struct Dyn 2022:1-19. [PMID: 35943030 DOI: 10.1080/07391102.2022.2107072] [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: 10/15/2022]
Abstract
Coronavirus disease (COVID-19) has the virus that causes the SARS-CoV-2 severe acute respiratory syndrome, which has reached a pandemic proportion, with thousands of deaths worldwide already registered. It has no standardized effective clinical treatment, arousing the urgent need for the discovery of bioactive compounds for the treatment of symptoms of COVID-19. In this context, the present study aimed to evaluate the influence of seasonality on the yield and chemical composition of the essential oils of Piper cernuum and Piper rivinoides as well as to evaluate the anti-SARS-CoV-2 potential of the major components of each oil by molecular docking and quantum chemical calculation (Density Functional Theory method), being possible indicate that the winter and autumn periods, the seasons of the year where it is possible to obtain the highest percentage of Piper cernuum and Piper rivinoides oils, respectively. Regarding the anti-SARS-Cov-2 potential, the present work showed that the dihydroagarofuran present in Piper cernuum, presented a strong interaction with amino acid residues from Mpro, presenting a potential similar to Remdesivir, a drug for clinical use. Regarding methyltransferase, dihydroagarofuran (Piper cernuum) and myristicin (Piper rivinoids) showed better affinity, with important interactions at the active site of the inhibitor Sinefugin, suggesting a potential inhibitory effect of the heterodimer methyltransferase complex NSP16-NSP10 SARS Cov-2. Molecular docking and molecular dynamics studies represent an initial step, being indicative for future in vitro studies of dihydroagarofuran and myristicin, as possible pharmacological tools for COVID-19.
Collapse
Affiliation(s)
- Antonio Linkoln Alves Borges Leal
- Department of Biological Chemistry, Regional University of Cariri, Crato, Ceará, Brazil.,Departament of Parasitology and Microbial, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Camila Fonseca Bezerra
- Department of Antibiotics, Drug Planning and Synthesis Laboratory - LPSF, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | | | - Luiz Everson da Silva
- Postgraduate Program in Sustainable Territorial Development, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - Lucas Lima Bezerra
- Department of Analytical Chemistry and Physical Chemistry - UFC, PICI Campus, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Francisco Wagner Almeida-Neto
- Department of Analytical Chemistry and Physical Chemistry - UFC, PICI Campus, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Emanuelle Machado Marinho
- Department of Analytical Chemistry and Physical Chemistry - UFC, PICI Campus, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Carla Freire Celedonio Fernandes
- Multi-User Laboratory for Research and Development, Antibody and Nanocorp Platform, Foundation Oswaldo Cruz-Fiocruz, Eusébio, Ceará, Brazil
| | - Matheus Nunes da Rocha
- Department of Chemistry, Group of Theoretical Chemistry and Electrochemistry, FAFIDAM Campus, State University of Ceará, Limoeiro do Norte, Ceará, Brazil
| | - Marcia Machado Marinho
- Department of Biological Chemistry, Regional University of Cariri, Crato, Ceará, Brazil.,Chemistry Course, Laboratory of Natural Products and Synthesis and of Organic Compounds - LBPNSB, Betânia Campus, State University of Vale do Acaraú, Sobral, Ceará, Brazil
| | - Henrique D M Coutinho
- Department of Biological Chemistry, Regional University of Cariri, Crato, Ceará, Brazil
| | | | - Ana Rafaela Freitas Dotto
- Postgraduate Program in Sustainable Territorial Development, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - Wanderlei do Amaral
- Department of Chemical Engineering, Curitiba, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - Hélcio Silva Dos Santos
- Department of Biological Chemistry, Regional University of Cariri, Crato, Ceará, Brazil.,Chemistry Course, Laboratory of Natural Products and Synthesis and of Organic Compounds - LBPNSB, Betânia Campus, State University of Vale do Acaraú, Sobral, Ceará, Brazil
| | - Pedro de Lima-Neto
- Department of Analytical Chemistry and Physical Chemistry - UFC, PICI Campus, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Emmanuel Silva Marinho
- Department of Chemistry, Group of Theoretical Chemistry and Electrochemistry, FAFIDAM Campus, State University of Ceará, Limoeiro do Norte, Ceará, Brazil
| |
Collapse
|
6
|
Geromichalou EG, Trafalis DT, Dalezis P, Malis G, Psomas G, Geromichalos GD. In silico study of potential antiviral activity of copper(II) complexes with non-steroidal anti-inflammatory drugs on various SARS-CoV-2 target proteins. J Inorg Biochem 2022; 231:111805. [PMID: 35334392 PMCID: PMC8930182 DOI: 10.1016/j.jinorgbio.2022.111805] [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: 11/16/2021] [Revised: 03/08/2022] [Accepted: 03/14/2022] [Indexed: 11/21/2022]
Abstract
In silico molecular docking studies, in vitro toxicity and in silico predictions on the biological activity profile, pharmacokinetic properties, drug-likeness, ADMET (absorption, distribution, metabolism, excretion, and toxicity) physicochemical pharmacokinetic data, and target proteins and toxicity predictions were performed on six copper(II) complexes with the non-steroidal anti-inflammatory drugs ibuprofen, loxoprofen, fenoprofen and clonixin as ligands, in order to investigate the ability of these complexes to interact with the key therapeutic target proteins of SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) 3C-like cysteine main protease (3CLpro/Mpro), viral papain-like protease (PLpro), RNA-dependent RNA polymerase (RdRp), and non-structural proteins (Nsps) Nsp16-Nsp10 2'-O-methyltransferase complex, and their capacity to act as antiviral agents, contributing thus to understanding the role they can play in the context of coronavirus 2019 (COVID-19) pandemic. Cytotoxic activity against five human cancer and normal cell lines were also evaluated.
Collapse
Affiliation(s)
- Elena G Geromichalou
- Laboratory of Pharmacology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Athens 11527, Greece
| | - Dimitrios T Trafalis
- Laboratory of Pharmacology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Athens 11527, Greece
| | - Panagiotis Dalezis
- Laboratory of Pharmacology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Athens 11527, Greece
| | - Georgios Malis
- Department of General and Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, GR 54124 Thessaloniki, Greece
| | - George Psomas
- Department of General and Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, GR 54124 Thessaloniki, Greece.
| | - George D Geromichalos
- Department of General and Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, GR 54124 Thessaloniki, Greece.
| |
Collapse
|
7
|
Fischer TR, Meidner L, Schwickert M, Weber M, Zimmermann RA, Kersten C, Schirmeister T, Helm M. Chemical biology and medicinal chemistry of RNA methyltransferases. Nucleic Acids Res 2022; 50:4216-4245. [PMID: 35412633 PMCID: PMC9071492 DOI: 10.1093/nar/gkac224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/17/2022] [Accepted: 04/08/2022] [Indexed: 12/24/2022] Open
Abstract
RNA methyltransferases (MTases) are ubiquitous enzymes whose hitherto low profile in medicinal chemistry, contrasts with the surging interest in RNA methylation, the arguably most important aspect of the new field of epitranscriptomics. As MTases become validated as drug targets in all major fields of biomedicine, the development of small molecule compounds as tools and inhibitors is picking up considerable momentum, in academia as well as in biotech. Here we discuss the development of small molecules for two related aspects of chemical biology. Firstly, derivates of the ubiquitous cofactor S-adenosyl-l-methionine (SAM) are being developed as bioconjugation tools for targeted transfer of functional groups and labels to increasingly visible targets. Secondly, SAM-derived compounds are being investigated for their ability to act as inhibitors of RNA MTases. Drug development is moving from derivatives of cosubstrates towards higher generation compounds that may address allosteric sites in addition to the catalytic centre. Progress in assay development and screening techniques from medicinal chemistry have led to recent breakthroughs, e.g. in addressing human enzymes targeted for their role in cancer. Spurred by the current pandemic, new inhibitors against coronaviral MTases have emerged at a spectacular rate, including a repurposed drug which is now in clinical trial.
Collapse
Affiliation(s)
- Tim R Fischer
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, 55128Mainz, Germany
| | - Laurenz Meidner
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, 55128Mainz, Germany
| | - Marvin Schwickert
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, 55128Mainz, Germany
| | - Marlies Weber
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, 55128Mainz, Germany
| | - Robert A Zimmermann
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, 55128Mainz, Germany
| | - Christian Kersten
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, 55128Mainz, Germany
| | - Tanja Schirmeister
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, 55128Mainz, Germany
| | - Mark Helm
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, 55128Mainz, Germany
| |
Collapse
|
8
|
Soni U, Singh P, Gupta OP, Gupta S, Singh SP, Singh P, Singh S, Mishra K. Lichen planus drugs re-purposing as potential anti COVID-19 therapeutics through molecular docking and molecular dynamics simulation approach. J Clin Transl Res 2022; 8:127-146. [PMID: 35475273 PMCID: PMC9036082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/03/2022] [Accepted: 01/08/2022] [Indexed: 10/26/2022] Open
Abstract
Background and Aim The present study intends to investigate COVID-19 by targeting their main proteins with 17 selected drugs used for treating Oral Lichen Planus (OLP) which is a chronic muco-cutaneous disorder. Here, an attempt is made to gain better insight into the structure of various drugs targeting specific proteins which will be helpful in developing drugs useful for therapeutic and preventive measures. Method In silico studies, molecular docking and molecular dynamic simulations were performed to repurpose the therapeutic drugs (n = 17) which were used to treat OLP against COVID-19. In addition, the maximum binding affinities of the key protein spike glycoprotein, main-protease (Mpro) of coronavirus, and Angiotensin-Converting Enzyme-2 (ACE-2) in the human body were evaluated with the selected drugs. Results Epigallocatechin-3-gallate (EGCG) showed the highest docking values among the drugs selected for repurposing. Among the target proteins, EGCG has shown maximum binding affinity with ACE-2 receptor. Further, according to the molecular dynamic simulation studies, EGCG has shown the least conformational fluctuations with Mpro. Conclusion EGCG can be a potential inhibitor drug which can bind with ACE-2 receptor thus inhibiting the interaction of mainly Mpro protein and spike glycoprotein of SARS-CoV-2. Relevance for Patients EGCG, a natural compound shows antiviral potential having considerably high affinity and stability with SARS-CoV-2. It might be further employed as a lead drug against selective inhibitors of SARS-CoV-2 for the therapeutic management of COVID-19 patients after necessary clinical trials.
Collapse
Affiliation(s)
- Unnati Soni
- 1Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, Uttar Pradesh, India
| | - Pratyush Singh
- 2Department of Oral Pathology and Microbiology, MaharanaPratap Dental College, Kanpur, Uttar Pradesh, India
| | - Om Prakash Gupta
- 3Department of General Surgery, Career Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Shalini Gupta
- 4Department of Oral Pathology and Microbiology, King George’s Medical University, Lucknow, Uttar Pradesh, India,Corresponding author: Shalini Gupta Department of Oral Pathology and Microbiology, King George’s Medical University, Lucknow, Uttar Pradesh - 226 003, India.
| | - Saurabh Pratap Singh
- 4Department of Oral Pathology and Microbiology, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Prerna Singh
- 4Department of Oral Pathology and Microbiology, King George’s Medical University, Lucknow, Uttar Pradesh, India
| | - Sangeeta Singh
- 1Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, Uttar Pradesh, India
| | - Krishna Mishra
- 1Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, Uttar Pradesh, India
| |
Collapse
|
9
|
Molecular docking and molecular dynamic simulation approaches for drug development and repurposing of drugs for severe acute respiratory syndrome-Coronavirus-2. COMPUTATIONAL APPROACHES FOR NOVEL THERAPEUTIC AND DIAGNOSTIC DESIGNING TO MITIGATE SARS-COV-2 INFECTION 2022. [PMCID: PMC9300476 DOI: 10.1016/b978-0-323-91172-6.00007-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
10
|
Li H, Xu F, Liu C, Cai A, Dain JA, Li D, Seeram NP, Cho BP, Ma H. Inhibitory Effects and Surface Plasmon Resonance-Based Binding Affinities of Dietary Hydrolyzable Tannins and Their Gut Microbial Metabolites on SARS-CoV-2 Main Protease. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:12197-12208. [PMID: 34586788 PMCID: PMC8491554 DOI: 10.1021/acs.jafc.1c03521] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 05/16/2023]
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV-2) main protease (Mpro) inhibitors are considered as potential treatments for coronavirus disease 2019, and dietary polyphenols show promise in SARS-CoV-2 Mpro inhibition based on in silico studies. In the present study, we utilize a combination of biochemical-, surface plasmon resonance-, and docking-based assays to evaluate the inhibition and binding affinities of a series of tannins and their gut microbial metabolites on SARS-CoV-2 Mpro. The tested compounds (2-50 μM) were hydrolyzable tannins, including ellagitannins (punicalagin and ellagic acid) and gallotannins (tannic acid, pentagalloyl glucose, ginnalin A, and gallic acid), and their gut microbial metabolites, urolithins and pyrogallol, respectively. They inhibited SARS-CoV-2 Mpro (by 6.6-100.0% at 50 μM) and bound directly to the Mpro protein (with dissociation constants from 1.1 × 10-6 to 5.3 × 10-5 M). This study sheds light on the inhibitory effects of tannins and their metabolites on SARS-CoV-2 Mpro.
Collapse
Affiliation(s)
- Huifang Li
- School of Biotechnology and Health Sciences, Wuyi University; International Healthcare Innovation Institute (Jiangmen), Jiangmen 529020, China
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Feng Xu
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550001, China
| | - Chang Liu
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Ang Cai
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
- Department of Chemistry, University of Rhode Island, Kingston, RI 02881, USA
| | - Joel A. Dain
- Department of Chemistry, University of Rhode Island, Kingston, RI 02881, USA
| | - Dongli Li
- Department of Chemistry, University of Rhode Island, Kingston, RI 02881, USA
| | - Navindra P. Seeram
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Bongsup P. Cho
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Hang Ma
- School of Biotechnology and Health Sciences, Wuyi University; International Healthcare Innovation Institute (Jiangmen), Jiangmen 529020, China
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| |
Collapse
|
11
|
Tumskiy RS, Tumskaia AV. Multistep rational molecular design and combined docking for discovery of novel classes of inhibitors of SARS-CoV-2 main protease 3CLpro. Chem Phys Lett 2021; 780:138894. [PMID: 34276059 PMCID: PMC8277558 DOI: 10.1016/j.cplett.2021.138894] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/08/2021] [Accepted: 07/11/2021] [Indexed: 12/16/2022]
Abstract
The main protease (3CLpro) of SARS-CoV and SARS-CoV-2 is a promising target for discovery of novel antiviral agents. In this paper, new possible inhibitors of 3CLpro with high predicted binding affinity were detected through multistep computer-aided molecular design and bioisosteric replacements. For discovery of prospective 3CLpro binders several virtual ligand libraries were created and combined docking was performed. Moreover, the molecular dynamics simulation was applied for evaluation of protein-ligand complexes stability. Besides, important molecular properties and ADMET pharmacokinetic profiles of possible 3CLpro inhibitors were assessed by in silico prediction.
Collapse
Affiliation(s)
- Roman S. Tumskiy
- Institute of Biochemistry and Physiology of Plants and Microorganisms (IBPPM RAS), Russian Academy of Sciences, 13 Prospekt Entuziastov, Saratov 410049, Russia,Corresponding author
| | - Anastasiia V. Tumskaia
- Chemistry Institute, Saratov State University, 83 Astrakhanskaya, Saratov 410012, Russia
| |
Collapse
|
12
|
de Oliveira VM, Marinho MM, Magalhães EP, de Menezes RRPPB, Sampaio TL, Costa Martins AM, dos Santos HS, Marinho ES. Molecular docking identification for the efficacy of natural limonoids against COVID-19 virus main protease. J INDIAN CHEM SOC 2021. [PMCID: PMC8442302 DOI: 10.1016/j.jics.2021.100157] [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] [Indexed: 11/05/2022]
Abstract
COVID-19 pandemic is the biggest public health problem of the century so far.The main protease (Mpro) is one of the main enzymes studied as a pharmacological target. In this context, the present work aimed to perform a virtual screening of possible inhibitors against the enzyme Mpro, having limonoids as the main object of research as supposed inhibitors. Molecular docking simulations indicated that limonoids have an affinity to complex with M-pro.However, Limonine and Nimoliciol showed nonspecific and low affinity interactions. In conclusion, Limonoids are substances of natural origin that can be used in the study of new pharmacological tools designed to combat and understand COVID-19.
Collapse
|
13
|
de Oliveira VM, da Rocha MN, Magalhães EP, da Silva Mendes FR, Marinho MM, de Menezes RRPPB, Sampaio TL, Dos Santos HS, Martins AMC, Marinho ES. Computational approach towards the design of artemisinin-thymoquinone hybrids against main protease of SARS-COV-2. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2021; 7:185. [PMID: 34514004 PMCID: PMC8419828 DOI: 10.1186/s43094-021-00334-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/26/2021] [Indexed: 01/22/2023] Open
Abstract
Background The sanitary emergency installed in the world, generated by the pandemic of COVID-19, instigates the search for scientific strategies to mitigate the damage caused by the disease to different sectors of society. The disease caused by the coronavirus, SARS-CoV-2, reached 216 countries/territories, where about 199 million people were reported with the infection. Of these, more than 4 million died. In this sense, strategies involving the development of new antiviral molecules are extremely important. The main protease (Mpro) from SARS-CoV-2 is an important target, which has been widely studied for antiviral treatment. This work aims to perform a screening of pharmacodynamics and pharmacokinetics of synthetic hybrids from thymoquinone and artemisin (THY-ART) against COVID-19. Results Molecular docking studies indicated that hybrids of artemisinin and thymoquinone showed a relevant interaction with the active fraction of the enzyme Mpro, when compared to the reference drugs. Furthermore, hybrids show an improvement in the interaction of substances with the enzyme, mainly due to the higher frequency of interactions with the Thr199 residue. ADMET studies indicated that hybrids tend to permeate biological membranes, allowing good human intestinal absorption, with low partition to the central nervous system, potentiation for CYP-450 enzyme inhibitors, low risk of toxicity compared to commercially available drugs, considering mainly mutagenicity and cardiotoxicity, low capacity of hybrids to permeate the blood–brain barrier, high absorption and moderate permeability in Caco-2 cells. In addition, T1–T7 tend to have a better distribution of their available fractions to carry out diffusion and transport across cell membranes, as well as increase the energy of interaction with the SARS-CoV-2 target. Conclusions Hybrid products of artemisinin and thymoquinone have the potential to inhibit Mpro, with desirable pharmacokinetic and toxicity characteristics compared to commercially available drugs, being indicated for preclinical and subsequent clinical studies against SARS-CoV-2. Emphasizing the possibility of synergistic use with currently used drugs in order to increase half-life and generate a possible synergistic effect. This work represents an important step for the development of specific drugs against COVID-19.
Collapse
Affiliation(s)
- Victor Moreira de Oliveira
- Theoretical and Electrochemical Chemistry Research Group/FAFIDAM, State University of Ceará, Limoeiro do Norte, CE CEP 62930-000 Brazil
| | - Matheus Nunes da Rocha
- Theoretical and Electrochemical Chemistry Research Group/FAFIDAM, State University of Ceará, Limoeiro do Norte, CE CEP 62930-000 Brazil
| | - Emanuel Paula Magalhães
- Department of Clinical and Toxicological Analysis, Federal University of Ceara, Fortaleza, CE CEP 60430-172 Brazil
| | - Francisco Rogênio da Silva Mendes
- Theoretical and Electrochemical Chemistry Research Group/FAFIDAM, State University of Ceará, Limoeiro do Norte, CE CEP 62930-000 Brazil
| | - Márcia Machado Marinho
- Iguatu Faculty of Education, Science and Letters/FECLI, State University of Ceará, Iguatu, CE CEP 63502-253 Brazil
| | | | - Tiago Lima Sampaio
- Department of Clinical and Toxicological Analysis, Federal University of Ceara, Fortaleza, CE CEP 60430-172 Brazil
| | - Hélcio Silva Dos Santos
- Laboratory of Natural Products Chemistry, Synthesis and Biocatalysis of Organic Compounds - LBPNSB, State University of Vale do Acaraú, Sobral, CE CEP 62040370 Brazil
| | - Alice Maria Costa Martins
- Department of Clinical and Toxicological Analysis, Federal University of Ceara, Fortaleza, CE CEP 60430-172 Brazil
| | - Emmanuel Silva Marinho
- Theoretical and Electrochemical Chemistry Research Group/FAFIDAM, State University of Ceará, Limoeiro do Norte, CE CEP 62930-000 Brazil
| |
Collapse
|
14
|
Lin H, Cherukupalli S, Feng D, Gao S, Kang D, Zhan P, Liu X. SARS-CoV-2 Entry inhibitors targeting virus-ACE2 or virus-TMPRSS2 interactions. Curr Med Chem 2021; 29:682-699. [PMID: 33881969 DOI: 10.2174/0929867328666210420103021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/29/2021] [Accepted: 01/30/2021] [Indexed: 11/22/2022]
Abstract
COVID-19 is an infectious disease caused by SARS-CoV-2. The life cycle of SARS-CoV-2 includes the entry into the target cells, replicase translation, replicating and transcribing genomes, translating structural proteins, assembling and releasing new virions. Entering host cells is a crucial stage in the early life cycle of the virus, and blocking this stage can effectively prevent virus infection. SARS enters the target cells mediated by the interaction between the viral S protein and the target cell surface receptor angiotensin-converting enzyme 2 (ACE2), as well as the cleavage effect of type-II transmembrane serine protease (TMPRSS2) on the S protein. Therefore, the ACE2 receptor and TMPRSS2 are important targets for SARS-CoV-2 entry inhibitors. Herein, we provide a concise report/information on drugs with potential therapeutic value targeting virus-ACE2 or virus-TMPRSS2 interactions, to provide a reference for the design and discovery of potential entry inhibitors against SARS-CoV-2.
Collapse
Affiliation(s)
- Hao Lin
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, China
| | - Srinivasulu Cherukupalli
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, China
| | - Da Feng
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, China
| | - Shenghua Gao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, China
| | - Dongwei Kang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, China
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, China
| |
Collapse
|