1
|
Wang Q, Wang X, Cai D, Yu J, Chen X, Niu W, Wang S, Liu X, Zhou D, Yin F. Hydrolysis and Transport Characteristics of Phospholipid Complex of Alkyl Gallates: Potential Sustained Release of Alkyl Gallate and Gallic Acid. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:2145-2153. [PMID: 38226868 DOI: 10.1021/acs.jafc.3c05731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
Phospholipid complexes of alkyl gallates (A-GAs) including ethyl gallate (EG), propyl gallate (PG), and butyl gallate (BG) were successfully prepared by the thin film dispersion method. HPLC-UV analysis in an everted rat gut sac model indicated that A-GAs can be liberated from phospholipid complexes, which were further hydrolyzed by intestinal lipase to generate free gallic acid (GA). Both A-GAs and GA are able to cross the membrane, and the hydrolysis rate of A-GAs and the transport rate of GA are positively correlated with the alkyl chain length. Especially, compared with the corresponding physical mixtures, the phospholipid complexes exhibit slower sustained-release of A-GAs and GA. Therefore, the formation of phospholipid complexes is an effective approach to prolong the residence time in vivo and additionally enhance the bioactivities of A-GAs and GA. More importantly, through regulating the carbon skeleton lengths, controlled-release of alkyl gallates and gallic acid from phospholipid complexes will be achieved.
Collapse
Affiliation(s)
- Qian Wang
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Liaoning Province Key Laboratory for Marine Food Science and Technology, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, People's Republic of China
| | - Xinmiao Wang
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Liaoning Province Key Laboratory for Marine Food Science and Technology, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, People's Republic of China
| | - Dong Cai
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Liaoning Province Key Laboratory for Marine Food Science and Technology, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, People's Republic of China
| | - Jinghan Yu
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Liaoning Province Key Laboratory for Marine Food Science and Technology, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, People's Republic of China
| | - Xuan Chen
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, People's Republic of China
| | - Weiyuan Niu
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Liaoning Province Key Laboratory for Marine Food Science and Technology, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, People's Republic of China
| | - Siya Wang
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Liaoning Province Key Laboratory for Marine Food Science and Technology, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, People's Republic of China
| | - Xiaoyang Liu
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Liaoning Province Key Laboratory for Marine Food Science and Technology, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, People's Republic of China
| | - Dayong Zhou
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Liaoning Province Key Laboratory for Marine Food Science and Technology, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, People's Republic of China
| | - Fawen Yin
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Liaoning Province Key Laboratory for Marine Food Science and Technology, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, People's Republic of China
| |
Collapse
|
2
|
Mumtaz L, Farid A, Yousef Alomar S, Ahmad N, Nawaz A, Andleeb S, Amin A. Assesment of polyphenolic compounds against biofilms produced by clinical Acinetobacter baumannii strains using in silico and in vitro models. Saudi J Biol Sci 2023; 30:103743. [PMID: 37564783 PMCID: PMC10410175 DOI: 10.1016/j.sjbs.2023.103743] [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: 05/27/2023] [Revised: 07/05/2023] [Accepted: 07/14/2023] [Indexed: 08/12/2023] Open
Abstract
Several types of microbial infections are caused by Acinetobacter baumanii that has developed resistance to antimicrobial agents. We therefore investigated the role of plant polyphenols against A. baumannii using in silico and in vitro models. The clinical strains of A. baumannii were investigated for determination of resistance pattern and resistance mechanisms including efflux pump, extended spectrum beta lactamase, phenotype detection of AmpC production, and Metallo-β-lactamase. The polyphenolic compounds were docked against transcription regulator BfmR (PDB ID 6BR7) and antimicrobial, antibiofilm, and anti-quorum sensing activities were performed. The antibiogram studies showed that all isolated strains were resistant. Strain A77 was positive in Metallo-β-lactamase production. Similarly, none of strains were producers of AmpC, however, A77, A76, A75 had active efflux pumps. Molecular docking studies confirmed a strong binding affinity of Rutin and Catechin towards transcription regulator 6BR7. A significant antimicrobial activity was recorded in case of quercetin and syringic acid (MIC 3.1 µg/mL) followed by vanillic acid and caffeic acid (MIC 12.5 µg/mL). All tested compounds presented a strong antibiofilm activity against A. baumanii strain A77 (65 to 90%). It was concluded that all tested polyphenols samples posess antimicrobial and antibiofilm activities, and hence they may be utilized to treat multidrug resistance A. baumannii infections.
Collapse
Affiliation(s)
- Laraib Mumtaz
- Gomal Centre of Biochemistry and Biotechnology(GCBB), Gomal University, KPK, 29050 D.I.Khan, Pakistan
- Gomal Centre of Pharmaceutical Sciences, Faculty of Pharmacy, Gomal University, D.I.Khan 29050, Pakistan
| | - Arshad Farid
- Gomal Centre of Biochemistry and Biotechnology(GCBB), Gomal University, KPK, 29050 D.I.Khan, Pakistan
| | - Suliman Yousef Alomar
- Doping Research Chair, Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Naushad Ahmad
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Asif Nawaz
- Gomal Centre of Pharmaceutical Sciences, Faculty of Pharmacy, Gomal University, D.I.Khan 29050, Pakistan
| | - Saadia Andleeb
- Atta Ur Rehman School of Biological Sciences, National University of Science and Technology, Islamabad Pakistan
| | - Adnan Amin
- Gomal Centre of Pharmaceutical Sciences, Faculty of Pharmacy, Gomal University, D.I.Khan 29050, Pakistan
| |
Collapse
|
3
|
Andrade MA, Mottin M, Sousa BKDP, Barbosa JARG, Dos Santos Azevedo C, Lasse Silva C, Gonçalves de Andrade M, Motta FN, Maulay-Bailly C, Amand S, Santana JMD, Horta Andrade C, Grellier P, Bastos IMD. Identification of novel Zika virus NS3 protease inhibitors with different inhibition modes by integrative experimental and computational approaches. Biochimie 2023; 212:143-152. [PMID: 37088408 DOI: 10.1016/j.biochi.2023.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/14/2023] [Accepted: 04/07/2023] [Indexed: 04/25/2023]
Abstract
Zika virus (ZIKV) infection is associated with severe neurological disorders and congenital malformation. Despite efforts to eradicate the disease, there is still neither vaccine nor approved drugs to treat ZIKV infection. The NS2B-NS3 protease is a validated drug target since it is essential to polyprotein virus maturation. In the present study, we describe an experimental screening of 2,320 compounds from the chemical library of the Muséum National d'Histoire Naturelle of Paris on ZIKV NS2B-NS3 protease. A total of 96 hits were identified with 90% or more of inhibitory activity at 10 μM. Amongst the most active compounds, five were analyzed for their inhibitory mechanisms by kinetics assays and computational approaches such as molecular docking. 2-(3-methoxyphenoxy) benzoic acid (compound 945) show characteristics of a competitive inhibition (Ki = 0.49 μM) that was corroborated by its molecular docking at the active site of the NS2B-NS3 protease. Taxifolin (compound 2292) behaves as an allosteric inhibitor whereas 3,8,9-trihydroxy-2-methyl-1H-phenalen-1-one (compound 128), harmol (compound 368) and anthrapurpurin (compound 1499) show uncompetitive inhibitions. These new NS2B-NS3 protease inhibitors are valuable hits to further hit-to-lead optimization.
Collapse
Affiliation(s)
- Milene Aparecida Andrade
- Pathogen-Host Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| | - Melina Mottin
- Pathogen-Host Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia, Brazil; Laboratory for Molecular Modeling and Drug Design - LabMol, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Bruna K de P Sousa
- Laboratory for Molecular Modeling and Drug Design - LabMol, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | | | - Clênia Dos Santos Azevedo
- Pathogen-Host Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| | - Camila Lasse Silva
- Pathogen-Host Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| | | | - Flávia Nader Motta
- Pathogen-Host Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia, Brazil; Faculdade de Ceilândia, Universidade de Brasília, Brasília, Brazil
| | - Christine Maulay-Bailly
- UMR 7245 MCAM, Muséum National d'Histoire Naturelle, Centre National de la Recherche Scientifique, Paris, France
| | - Séverine Amand
- UMR 7245 MCAM, Muséum National d'Histoire Naturelle, Centre National de la Recherche Scientifique, Paris, France
| | - Jaime Martins de Santana
- Pathogen-Host Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| | - Carolina Horta Andrade
- Laboratory for Molecular Modeling and Drug Design - LabMol, Faculdade de Farmácia, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Philippe Grellier
- UMR 7245 MCAM, Muséum National d'Histoire Naturelle, Centre National de la Recherche Scientifique, Paris, France.
| | - Izabela M D Bastos
- Pathogen-Host Interface Laboratory, Department of Cell Biology, University of Brasilia, Brasilia, Brazil.
| |
Collapse
|
4
|
Gu Y, Liu M, Staker BL, Buchko GW, Quinn RJ. Drug-Repurposing Screening Identifies a Gallic Acid Binding Site on SARS-CoV-2 Non-structural Protein 7. ACS Pharmacol Transl Sci 2023; 6:578-586. [PMID: 37082753 PMCID: PMC10111621 DOI: 10.1021/acsptsci.2c00225] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Indexed: 03/09/2023]
Abstract
SARS-CoV-2 is the agent responsible for acute respiratory disease COVID-19 and the global pandemic initiated in early 2020. While the record-breaking development of vaccines has assisted the control of COVID-19, there is still a pressing global demand for antiviral drugs to halt the destructive impact of this disease. Repurposing clinically approved drugs provides an opportunity to expediate SARS-CoV-2 treatments into the clinic. In an effort to facilitate drug repurposing, an FDA-approved drug library containing 2400 compounds was screened against the SARS-CoV-2 non-structural protein 7 (nsp7) using a native mass spectrometry-based assay. Nsp7 is one of the components of the SARS-CoV-2 replication/transcription complex essential for optimal viral replication, perhaps serving to off-load RNA from nsp8. From this library, gallic acid was identified as a compound that bound tightly to nsp7, with an estimated K d of 15 μM. NMR chemical shift perturbation experiments were used to map the ligand-binding surface of gallic acid on nsp7, indicating that the compound bound to a surface pocket centered on one of the protein's four α-helices (α2). The identification of the gallic acid-binding site on nsp7 may allow development of a SARS-CoV-2 therapeutic via artificial-intelligence-based virtual docking and other strategies.
Collapse
Affiliation(s)
- Yushu Gu
- Griffith
Institute for Drug Discovery, Griffith University, Brisbane 4111, Australia
| | - Miaomiao Liu
- Griffith
Institute for Drug Discovery, Griffith University, Brisbane 4111, Australia
| | - Bart L. Staker
- Seattle
Children’s Research Institute, Seattle, Washington 98101, United States
| | - Garry W. Buchko
- Earth
and Biological Sciences Directorate, Pacific
Northwest National Laboratory, Richland, Washington 99354, United States
- School of
Molecular Biosciences, Washington State
University, Pullman, Washington 99164, United States
| | - Ronald J. Quinn
- Griffith
Institute for Drug Discovery, Griffith University, Brisbane 4111, Australia
| |
Collapse
|
5
|
Ang D, Kendall R, Atamian HS. Virtual and In Vitro Screening of Natural Products Identifies Indole and Benzene Derivatives as Inhibitors of SARS-CoV-2 Main Protease (M pro). BIOLOGY 2023; 12:biology12040519. [PMID: 37106720 PMCID: PMC10135783 DOI: 10.3390/biology12040519] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/25/2023] [Accepted: 03/26/2023] [Indexed: 04/29/2023]
Abstract
The rapid spread of the coronavirus disease 2019 (COVID-19) resulted in serious health, social, and economic consequences. While the development of effective vaccines substantially reduced the severity of symptoms and the associated deaths, we still urgently need effective drugs to further reduce the number of casualties associated with SARS-CoV-2 infections. Machine learning methods both improved and sped up all the different stages of the drug discovery processes by performing complex analyses with enormous datasets. Natural products (NPs) have been used for treating diseases and infections for thousands of years and represent a valuable resource for drug discovery when combined with the current computation advancements. Here, a dataset of 406,747 unique NPs was screened against the SARS-CoV-2 main protease (Mpro) crystal structure (6lu7) using a combination of ligand- and structural-based virtual screening. Based on 1) the predicted binding affinities of the NPs to the Mpro, 2) the types and number of interactions with the Mpro amino acids that are critical for its function, and 3) the desirable pharmacokinetic properties of the NPs, we identified the top 20 candidates that could potentially inhibit the Mpro protease function. A total of 7 of the 20 top candidates were subjected to in vitro protease inhibition assay and 4 of them (4/7; 57%), including two beta carbolines, one N-alkyl indole, and one Benzoic acid ester, had significant inhibitory activity against Mpro protease. These four NPs could be developed further for the treatment of COVID-19 symptoms.
Collapse
Affiliation(s)
- Dony Ang
- Computational and Data Sciences Program, Chapman University, Orange, CA 92866, USA
- Schmid College of Science and Technology, Chapman University, Orange, CA 92866, USA
| | - Riley Kendall
- Computational and Data Sciences Program, Chapman University, Orange, CA 92866, USA
- Schmid College of Science and Technology, Chapman University, Orange, CA 92866, USA
| | - Hagop S Atamian
- Schmid College of Science and Technology, Chapman University, Orange, CA 92866, USA
- Biological Sciences Program, Chapman University, Orange, CA 92866, USA
| |
Collapse
|
6
|
Ayipo YO, Ahmad I, Najib YS, Sheu SK, Patel H, Mordi MN. Molecular modelling and structure-activity relationship of a natural derivative of o-hydroxybenzoate as a potent inhibitor of dual NSP3 and NSP12 of SARS-CoV-2: in silico study. J Biomol Struct Dyn 2023; 41:1959-1977. [PMID: 35037841 DOI: 10.1080/07391102.2022.2026818] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The nsp3 macrodomain and nsp12 (RdRp) enzymes are strongly implicated in the virulent regulation of the host immune response and viral replication of SARS-CoV-2, making them plausible therapeutic targets for mitigating infectivity. Remdesivir remains the only FDA-approved small-molecule inhibitor of the nsp12 in clinical conditions while none has been approved yet for the nsp3 macrodomain. In this study, 69,067 natural compounds from the IBScreen database were screened for efficacious potentials with mechanistic multitarget-directed inhibitory pharmacology against the dual targets using in silico approaches. Standard and extra precision (SP and XP) Maestro glide docking analyses were employed to evaluate their inhibitory interactions against the enzymes. Four compounds, STOCK1N-45901, 03804, 83408, 08377 consistently showed high XP scores against the respective targets and interacted strongly with pharmacologically essential amino acid and RNA residues, in better terms than the standard, co-crystallized inhibitors, GS-441524 and remdesivir. Further assessments through the predictions of ADMET and mutagenicity distinguished STOCK1N-45901, a natural derivative of o-hydroxybenzoate as the most promising candidate. The ligand maintained a good conformational and thermodynamic stability in complex with the enzymes throughout the trajectories of 100 ns molecular dynamics, indicated by RMSD, RMSF and radius of gyration plots. Its binding free energy, MM-GBSA was recorded as -54.24 and -31.77 kcal/mol against the respective enzyme, while its structure-activity relationships confer high probabilities as active antiviral, anti-inflammatory, antiinfection, antitussive and peroxidase inhibitor. The IBScreen database natural product, STOCK1N-45901 (2,3,4,5,6-pentahydroxyhexyl o-hydroxybenzoate) is thus recommended as a potent inhibitor of dual nsp3 and nsp12 of SARS-CoV-2 for further study. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Yusuf Oloruntoyin Ayipo
- Centre for Drug Research, Universiti Sains Malaysia, USM, Pulau Pinang, Malaysia
- Department of Chemistry and Industrial Chemistry, Kwara State University, Malete, Ilorin, Nigeria
| | - Iqrar Ahmad
- Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, India
| | - Yahaya Sani Najib
- Centre for Drug Research, Universiti Sains Malaysia, USM, Pulau Pinang, Malaysia
- Department of Pharmaceutical and Medicinal Chemistry, Bayero University, Kano, Nigeria
| | - Sikirat Kehinde Sheu
- Department of Chemistry and Industrial Chemistry, Kwara State University, Malete, Ilorin, Nigeria
| | - Harun Patel
- Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, India
| | - Mohd Nizam Mordi
- Centre for Drug Research, Universiti Sains Malaysia, USM, Pulau Pinang, Malaysia
| |
Collapse
|
7
|
Pilaquinga F, Bosch R, Morey J, Bastidas-Caldes C, Torres M, Toscano F, Debut A, Pazmiño-Viteri K, Nieves Piña MDL. High in vitroactivity of gold and silver nanoparticles from Solanum mammosum L. against SARS-CoV-2 surrogate Phi6 and viral model PhiX174. NANOTECHNOLOGY 2023; 34:175705. [PMID: 36689773 DOI: 10.1088/1361-6528/acb558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 01/23/2023] [Indexed: 06/17/2023]
Abstract
The search for new strategies to curb the spread of the SARS-CoV-2 coronavirus, which causes COVID-19, has become a global priority. Various nanomaterials have been proposed as ideal candidates to inactivate the virus; however, because of the high level of biosecurity required for their use, alternative models should be determined. This study aimed to compare the effects of two types of nanomaterials gold (AuNPs) and silver nanoparticles (AgNPs), recognized for their antiviral activity and affinity with the coronavirus spike protein using PhiX174 and enveloped Phi6 bacteriophages as models. To reduce the toxicity of nanoparticles, a species known for its intermediate antiviral activity,Solanum mammosumL. (Sm), was used. NPs prepared with sodium borohydride (NaBH4) functioned as the control. Antiviral activity against PhiX174 and Phi6 was analyzed using its seed, fruit, leaves, and essential oil; the leaves were the most effective on Phi6. Using the aqueous extract of the leaves, AuNPs-Sm of 5.34 ± 2.25 nm and AgNPs-Sm of 15.92 ± 8.03 nm, measured by transmission electron microscopy, were obtained. When comparing NPs with precursors, both gold(III) acetate and silver nitrate were more toxic than their respective NPs (99.99% at 1 mg ml-1). The AuNPs-Sm were less toxic, reaching 99.30% viral inactivation at 1 mg ml-1, unlike the AgNPs-Sm, which reached 99.94% at 0.01 mg ml-1. In addition, cell toxicity was tested in human adenocarcinoma alveolar basal epithelial cells (A549) and human foreskin fibroblasts. Gallic acid was the main component identified in the leaf extract using high performance liquid chromatography with diode array detection (HPLC-DAD). The FT-IR spectra showed the presence of a large proportion of polyphenolic compounds, and the antioxidant analysis confirmed the antiradical activity. The control NPs showed less antiviral activity than the AuNPs-Sm and AgNPs-Sm, which was statistically significant; this demonstrates that both theS. mammosumextract and its corresponding NPs have a greater antiviral effect on the surrogate Phi bacteriophage, which is an appropriate model for studying SARS-CoV-2.
Collapse
Affiliation(s)
- Fernanda Pilaquinga
- Laboratory of Nanotechnology, School of Chemistry Sciences, Pontificia Universidad Católica del Ecuador, Avenida 12 de octubre 1076 y Roca, Quito, Ecuador
- Department of Chemistry, University of the Balearic Islands, Cra. de Valldemossa Km. 7.5, 07122 Palma de Mallorca, Spain
| | - Rafael Bosch
- Environmental Microbiology, IMEDEA (CSIC-UIB); and Microbiology, Department of Biology, University of Balearic Islands, Palma de Mallorca, Spain
| | - Jeroni Morey
- Department of Chemistry, University of the Balearic Islands, Cra. de Valldemossa Km. 7.5, 07122 Palma de Mallorca, Spain
| | - Carlos Bastidas-Caldes
- One Health Research Group, Facultad de Ingeniería y Ciencias Aplicadas, Biotecnología, Universidad de las Américas, Redondel del Ciclista, Antigua Vía a Nayón, Quito, Ecuador
- Programa de Doctorado en Salud Pública y Animal, Universidad de Extremadura, Plaza de Caldereros, s/n, Extremadura, Spain
| | - Marbel Torres
- Departamento de Ciencias de la Vida y la Agricultura, Laboratorio de Inmunología y Virología, Centro de Nanociencia y Nanotecnología, Universidad de las Fuerzas Armadas ESPE, Sangolquí, Ecuador
| | - Fernanda Toscano
- Departamento de Ciencias de la Vida y la Agricultura, Laboratorio de Inmunología y Virología, Centro de Nanociencia y Nanotecnología, Universidad de las Fuerzas Armadas ESPE, Sangolquí, Ecuador
| | - Alexis Debut
- Centro de Nanociencia y Nanotecnología CENCINAT, Universidad de las Fuerzas Armadas ESPE, Sangolquí 170501, Ecuador Universidad de las Fuerzas Armadas ESPE, Sangolquí 170501, Ecuador
| | - Katherine Pazmiño-Viteri
- Centro de Nanociencia y Nanotecnología CENCINAT, Universidad de las Fuerzas Armadas ESPE, Sangolquí 170501, Ecuador Universidad de las Fuerzas Armadas ESPE, Sangolquí 170501, Ecuador
| | - María de Las Nieves Piña
- Department of Chemistry, University of the Balearic Islands, Cra. de Valldemossa Km. 7.5, 07122 Palma de Mallorca, Spain
| |
Collapse
|
8
|
Mousavi S, Zare S, Mirzaei M, Feizi A. Novel Drug Design for Treatment of COVID-19: A Systematic Review of Preclinical Studies. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2022; 2022:2044282. [PMID: 36199815 PMCID: PMC9527439 DOI: 10.1155/2022/2044282] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 05/23/2022] [Accepted: 08/03/2022] [Indexed: 11/27/2022]
Abstract
Background Since the beginning of the novel coronavirus (SARS-CoV-2) disease outbreak, there has been an increasing interest in discovering potential therapeutic agents for this disease. In this regard, we conducted a systematic review through an overview of drug development (in silico, in vitro, and in vivo) for treating COVID-19. Methods A systematic search was carried out in major databases including PubMed, Web of Science, Scopus, EMBASE, and Google Scholar from December 2019 to March 2021. A combination of the following terms was used: coronavirus, COVID-19, SARS-CoV-2, drug design, drug development, In silico, In vitro, and In vivo. A narrative synthesis was performed as a qualitative method for the data synthesis of each outcome measure. Results A total of 2168 articles were identified through searching databases. Finally, 315 studies (266 in silico, 34 in vitro, and 15 in vivo) were included. In studies with in silico approach, 98 article study repurposed drug and 91 studies evaluated herbal medicine on COVID-19. Among 260 drugs repurposed by the computational method, the best results were observed with saquinavir (n = 9), ritonavir (n = 8), and lopinavir (n = 6). Main protease (n = 154) following spike glycoprotein (n = 62) and other nonstructural protein of virus (n = 45) was among the most studied targets. Doxycycline, chlorpromazine, azithromycin, heparin, bepridil, and glycyrrhizic acid showed both in silico and in vitro inhibitory effects against SARS-CoV-2. Conclusion The preclinical studies of novel drug design for COVID-19 focused on main protease and spike glycoprotein as targets for antiviral development. From evaluated structures, saquinavir, ritonavir, eucalyptus, Tinospora cordifolia, aloe, green tea, curcumin, pyrazole, and triazole derivatives in in silico studies and doxycycline, chlorpromazine, and heparin from in vitro and human monoclonal antibodies from in vivo studies showed promised results regarding efficacy. It seems that due to the nature of COVID-19 disease, finding some drugs with multitarget antiviral actions and anti-inflammatory potential is valuable and some herbal medicines have this potential.
Collapse
Affiliation(s)
- Sarah Mousavi
- Department of Clinical Pharmacy and Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shima Zare
- School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahmoud Mirzaei
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Awat Feizi
- Department of Epidemiology and Biostatistics, School of Health, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
9
|
Ribaudo G, Orian L. From a Molecule to a Drug: Chemical Features Enhancing Pharmacological Potential. Molecules 2022; 27:molecules27134144. [PMID: 35807388 PMCID: PMC9268219 DOI: 10.3390/molecules27134144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 06/27/2022] [Indexed: 02/05/2023] Open
Abstract
Health is a fundamental human right and is a global goal to which extensive research effort is devoted in all fields [...]
Collapse
Affiliation(s)
- Giovanni Ribaudo
- Dipartimento di Medicina Molecolare e Traslazionale, Università degli Studi di Brescia, Viale Europa 11, 25123 Brescia, Italy;
| | - Laura Orian
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, Via Marzolo 1, 35131 Padova, Italy
- Correspondence:
| |
Collapse
|
10
|
Zhao Z, Ishida M, Onodera T, Toko K. Effect of Hydroxybenzoic Acids on Caffeine Detection Using Taste Sensor with Lipid/Polymer Membranes. SENSORS 2022; 22:s22041607. [PMID: 35214507 PMCID: PMC8876041 DOI: 10.3390/s22041607] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/10/2022] [Accepted: 02/16/2022] [Indexed: 02/01/2023]
Abstract
A taste sensor with lipid/polymer membranes can objectively evaluate taste. As previously reported, caffeine can be detected electrically using lipid/polymer membranes modified with hydroxybenzoic acids (HBAs). However, a systematic understanding of how HBAs contribute to caffeine detection is still lacking. In this study, we used various HBAs such as 2,6–dihydroxybenzoic acid (2,6–DHBA) to modify lipid/polymer membranes, and we detected caffeine using a taste sensor with the modified membranes. The effect of the concentrations of the HBAs on caffeine detection was also discussed. The results of the caffeine detection indicated that the response to caffeine and the reference potential measured in a reference solution were affected by the log P and pKa of HBAs. Furthermore, the taste sensor displayed high sensitivity to caffeine when the reference potential was adjusted to an appropriate range by modification with 2,6–DHBA, where the slope of the change in reference potential with increasing 2,6–DHBA concentration was steep. This is helpful in order to improve the sensitivity of taste sensors to other taste substances, such as theophylline and theobromine, in the future.
Collapse
Affiliation(s)
- Zeyu Zhao
- Graduate School of Information Science and Electrical Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; (M.I.); (T.O.)
- Correspondence:
| | - Misaki Ishida
- Graduate School of Information Science and Electrical Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; (M.I.); (T.O.)
| | - Takeshi Onodera
- Graduate School of Information Science and Electrical Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; (M.I.); (T.O.)
| | - Kiyoshi Toko
- Institute for Advanced Study, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan;
- Research and Development Center for Five-Sense Devices, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| |
Collapse
|
11
|
Świątek Ł, Sieniawska E, Sinan KI, Maciejewska-Turska M, Boguszewska A, Polz-Dacewicz M, Senkardes I, Guler GO, Bibi Sadeer N, Mahomoodally MF, Zengin G. LC-ESI-QTOF-MS/MS Analysis, Cytotoxic, Antiviral, Antioxidant, and Enzyme Inhibitory Properties of Four Extracts of Geranium pyrenaicum Burm. f.: A Good Gift from the Natural Treasure. Int J Mol Sci 2021; 22:7621. [PMID: 34299238 PMCID: PMC8307321 DOI: 10.3390/ijms22147621] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 02/01/2023] Open
Abstract
This study focused on the biological evaluation and chemical characterization of Geranium pyrenaicum Burm. f. Different solvent extracts (hexane, ethyl acetate, methanol, and water extracts) were prepared. The phytochemical profile, antioxidant, and enzyme inhibitory activity were investigated. Cytotoxicity was assessed using VERO, FaDu, HeLa and RKO cells. The antiviral activity was carried out against HSV-1 (Herpes simplex virus 1) propagated in VERO cell line. The aqueous extract, possessing high phenolic content (170.50 mg gallic acid equivalent/g extract), showed the highest reducing capacity (613.27 and 364.10 mg Trolox equivalent/g extract, for cupric reducing antioxidant capacity and ferric reducing antioxidant power, respectively), radical scavenging potential (469.82 mg Trolox equivalent/g extract, against 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid)), metal chelating ability (52.39 mg ethylenediaminetetraacetic acid equivalent/g extract) and total antioxidant capacity (3.15 mmol Trolox equivalent/g extract). Liquid chromatography-electrospray ionization-quadrupole time-of-flight-mass spectrometry (LC-ESI-QTOF-MS/MS) alloved to tentatively identify a total of 56 compounds in the extracts, including ellagitannins, gallic acid and galloyl derivatives amongst others. The ethyl acetate extracts substantially depressed cholinesterase enzymes (4.49 and 12.26 mg galantamine equivalent/g extract against AChE and BChE, respectively) and α-amylase enzyme (1.04 mmol acarbose equivalent/g extract). On the other hand, the methanolic extract inhibited tyrosinase (121.42 mg kojic acid equivalent/g extract) and α-glucosidase (2.39 mmol acarbose equivalent/g extract) activities. The highest selectivity towards all cancer cell lines (SI 4.5-10.8) was observed with aqueous extract with the FaDu cells being the most sensitive (CC50 40.22 µg/mL). It can be concluded that the presence of certain bioactive antiviral molecules may be related to the high anti HSV-1 activity of the methanolic extract. This work has generated vital scientific data on this medicinal plant, which is a prospective candidate for the creation of innovative phyto-pharmaceuticals.
Collapse
Affiliation(s)
- Łukasz Świątek
- Department of Virology with SARS Laboratory, Medical University of Lublin, Chodzki 1, 20-093 Lublin, Poland; (A.B.); (M.P.-D.)
| | - Elwira Sieniawska
- Department of Pharmacognosy, Medical University of Lublin, Chodzki 1, 20-093 Lublin, Poland; (E.S.); (M.M.-T.)
| | - Kouadio Ibrahime Sinan
- Physiology and Biochemistry Research Laboratory, Department of Biology, Science Faculty, Selcuk University, Konya 42130, Turkey;
| | | | - Anastazja Boguszewska
- Department of Virology with SARS Laboratory, Medical University of Lublin, Chodzki 1, 20-093 Lublin, Poland; (A.B.); (M.P.-D.)
| | - Małgorzata Polz-Dacewicz
- Department of Virology with SARS Laboratory, Medical University of Lublin, Chodzki 1, 20-093 Lublin, Poland; (A.B.); (M.P.-D.)
| | - Ismail Senkardes
- Department of Pharmaceutical Botany, Faculty of Pharmacy, Marmara University, Istanbul 34854, Turkey;
| | - Gokalp Ozmen Guler
- Department of Biological Education, Ahmet Kelesoglu Education Faculty, Necmettin Erbakan University, Konya 42090, Turkey;
| | - Nabeelah Bibi Sadeer
- Department of Health Sciences, Faculty of Medicine and Health Sciences, University of Mauritius, Réduit 80837, Mauritius; (N.B.S.); (M.F.M.)
| | - Mohamad Fawzi Mahomoodally
- Department of Health Sciences, Faculty of Medicine and Health Sciences, University of Mauritius, Réduit 80837, Mauritius; (N.B.S.); (M.F.M.)
| | - Gokhan Zengin
- Physiology and Biochemistry Research Laboratory, Department of Biology, Science Faculty, Selcuk University, Konya 42130, Turkey;
| |
Collapse
|
12
|
Bieberich AA, Rajwa B, Irvine A, Fatig RO, Fekete A, Jin H, Kutlina E, Urban L. TEMPORARY REMOVAL: Acute cell stress screen with supervised machine learning predicts cytotoxicity of excipients. J Pharmacol Toxicol Methods 2021; 111:107088. [PMID: 34144174 DOI: 10.1016/j.vascn.2021.107088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/17/2021] [Accepted: 06/12/2021] [Indexed: 11/18/2022]
Abstract
The publisher regrets that this article has been temporarily removed. A replacement will appear as soon as possible in which the reason for the removal of the article will be specified, or the article will be reinstated. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Andrew A Bieberich
- AsedaSciences Inc., 1281 Win Hentschel Boulevard, West Lafayette, IN 47906, USA.
| | - Bartek Rajwa
- Bindley Bioscience Center, Purdue University, West Lafayette, IN 47906, USA.
| | - Allison Irvine
- AsedaSciences Inc., 1281 Win Hentschel Boulevard, West Lafayette, IN 47906, USA.
| | - Raymond O Fatig
- AsedaSciences Inc., 1281 Win Hentschel Boulevard, West Lafayette, IN 47906, USA.
| | - Alexander Fekete
- Preclinical Safety, Translational Medicine, Novartis Institutes of Biomedical Research, Cambridge, MA, 02139, USA.
| | - Hong Jin
- Preclinical Safety, Translational Medicine, Novartis Institutes of Biomedical Research, Cambridge, MA, 02139, USA.
| | - Elena Kutlina
- Preclinical Safety, Translational Medicine, Novartis Institutes of Biomedical Research, Cambridge, MA, 02139, USA.
| | - Laszlo Urban
- Preclinical Safety, Translational Medicine, Novartis Institutes of Biomedical Research, Cambridge, MA, 02139, USA.
| |
Collapse
|
13
|
Kim CH. Anti-SARS-CoV-2 Natural Products as Potentially Therapeutic Agents. Front Pharmacol 2021; 12:590509. [PMID: 34122058 PMCID: PMC8194829 DOI: 10.3389/fphar.2021.590509] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 04/19/2021] [Indexed: 12/21/2022] Open
Abstract
Severe acute respiratory syndrome-related coronavirus-2 (SARS-CoV-2), a β-coronavirus, is the cause of the recently emerged pandemic and worldwide outbreak of respiratory disease. Researchers exchange information on COVID-19 to enable collaborative searches. Although there is as yet no effective antiviral agent, like tamiflu against influenza, to block SARS-CoV-2 infection to its host cells, various candidates to mitigate or treat the disease are currently being investigated. Several drugs are being screened for the ability to block virus entry on cell surfaces and/or block intracellular replication in host cells. Vaccine development is being pursued, invoking a better elucidation of the life cycle of the virus. SARS-CoV-2 recognizes O-acetylated neuraminic acids and also several membrane proteins, such as ACE2, as the result of evolutionary switches of O-Ac SA recognition specificities. To provide information related to the current development of possible anti-SARS-COV-2 viral agents, the current review deals with the known inhibitory compounds with low molecular weight. The molecules are mainly derived from natural products of plant sources by screening or chemical synthesis via molecular simulations. Artificial intelligence-based computational simulation for drug designation and large-scale inhibitor screening have recently been performed. Structure-activity relationship of the anti-SARS-CoV-2 natural compounds is discussed.
Collapse
Affiliation(s)
- Cheorl-Ho Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, Sungkyunkhwan University, Suwon, South Korea
| |
Collapse
|
14
|
Singab RA, Elleboudy NS, Elkhatib WF, Yassein MA, Hassouna NA. Improvement of caffeic acid biotransformation into para-hydroxybenzoic acid by Candida albicans CI-24 via gamma irradiation and model-based optimization. Biotechnol Appl Biochem 2021; 69:469-478. [PMID: 33576532 DOI: 10.1002/bab.2124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 02/09/2021] [Indexed: 11/08/2022]
Abstract
Para-hydroxybenzoic acid (PHBA) has great potential in biological applications due to its putative antiviral activity against SARS-CoV-2 and its antimicrobial activity in the face of the radically increasing number of multidrug-resistant pathogens. This is in addition to its antimutagenic, anti-inflammatory, antioxidant, hypoglycemic, antiestrogenic, and antiplatelet aggregating activities. In this study, an approximate sixfold increase in the production of PHBA was achieved via biotransformation of caffeic acid by Candida albicans. The improvement was performed in two steps: first, through mutation by gamma irradiation (5 KGy dose), resulting in the recovery of a mutant (CI-24), which produced approximately triple the amount of PHBA produced by the wild-type isolate. Then, biotransformation by this mutant was further optimized via response surface methodology model-based optimization. The maximum PHBA production (7.47 mg/mL) was obtained in a fermentation medium composed of 1% w/v yeast extract as a nitrogen source, with an initial pH of 6.6, incubated at 28 °C at an agitation rate of 250 rpm. To further enhance the performance and economics of the process, cells of the CI-24 mutant were immobilized in calcium alginate beads and could retain an equivalent biotransformation capacity after three successive biotransformation cycles.
Collapse
Affiliation(s)
- Raghda Abdelnasser Singab
- Microbiology and Immunology Department, Faculty of Pharmacy, Ain Shams University, African Union Organization St., Abbassia, Cairo, Egypt
| | - Nooran Sherif Elleboudy
- Microbiology and Immunology Department, Faculty of Pharmacy, Ain Shams University, African Union Organization St., Abbassia, Cairo, Egypt
| | - Walid Faisal Elkhatib
- Microbiology and Immunology Department, Faculty of Pharmacy, Ain Shams University, African Union Organization St., Abbassia, Cairo, Egypt.,Department of Microbiology and Immunology, Faculty of Pharmacy, Galala University, New Galala City, Suez, Egypt
| | - Mahmoud Abdulmegead Yassein
- Microbiology and Immunology Department, Faculty of Pharmacy, Ain Shams University, African Union Organization St., Abbassia, Cairo, Egypt
| | - Nadia Adelhaleem Hassouna
- Microbiology and Immunology Department, Faculty of Pharmacy, Ain Shams University, African Union Organization St., Abbassia, Cairo, Egypt
| |
Collapse
|