1
|
Tuohudaali W, Ji TF, Ding WT, Li CY, Bianba JS, Ci R, Zhao J. Urolithin B inhibits LPS-induced macrophage M1 polarization via miR155-5p mediated MAPK/NF-кB pathway. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024:1-10. [PMID: 39671344 DOI: 10.1080/10286020.2024.2435984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/15/2024]
Abstract
This study investigated inhibiting mechanisms of Urolithin B (Uro B) on macrophage M1 polarization. Uro B (50 μM) could inhibit the PGE2, COX-2, NO, iNOS, TNF-α, IL-1β and IL-6 levels compared with model group (P < 0.05) as well as the CD86 and F4/80 expression. The miR155-5p overexpression could increase the p38 MAPK, JNK, ERK mRNA activities (P < 0.05), Uro B (50 μM) could reverse changes in these indicators (P < 0.05). Moreover, Uro B (50 μM) could inhibit the TLR4, Src, IκBα, NF-κBp65 and their phosphorylated protein expression (P < 0.05). Therefore, Uro B may inhibit macrophage M1 polarization via miR155-5p mediated MAPK/NF-кB pathway.
Collapse
Affiliation(s)
| | - Teng-Fei Ji
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wan-Ting Ding
- School of Pharmacy, Xinjiang Medical University, Urumqi 830046, China
| | - Chen-Yang Li
- Key Laboratory for Uighur Medicine, Institute of Materia Medica of Xinjiang, Urumqi 830004, China
| | | | - Ren Ci
- Hospital of Tibetan Traditional Medicine, Lhasa 850002, China
| | - Jun Zhao
- School of Pharmacy, Xinjiang Medical University, Urumqi 830046, China
- Key Laboratory for Uighur Medicine, Institute of Materia Medica of Xinjiang, Urumqi 830004, China
| |
Collapse
|
2
|
Liu ZQ. How many organic small molecules might be used to treat COVID-19? From natural products to synthetic agents. Eur J Med Chem 2024; 278:116788. [PMID: 39236494 DOI: 10.1016/j.ejmech.2024.116788] [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: 07/04/2024] [Revised: 08/05/2024] [Accepted: 08/19/2024] [Indexed: 09/07/2024]
Abstract
A large scale of pandemic coronavirus disease (COVID-19) in the past five years motivates a great deal of endeavors donating to the exploration on therapeutic drugs against COVID-19 as well as other diseases caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Herein is an overview on the organic small molecules that are potentially employed to treat COVID-19 and other SARS-CoV-2-related diseases. These organic small molecules are accessed from both natural resources and synthetic strategies. Notably, typical natural products presented herein consist of polyphenols, lignans, alkaloids, terpenoids, and peptides, which exert an advantage for the further discovery of novel anti-COVID-19 drugs from plant herbs. On the other hand, synthetic prodrugs are composed of a series of inhibitors towards RNA-dependent RNA polymerase (RdRp), main protease (Mpro), 3-chymotrypsin-like cysteine protease (3CLpro), spike protein, papain-like protease (PLpro) of the SARS-CoV-2 as well as the angiotensin-converting enzyme 2 (ACE2) in the host cells. Synthetic strategies are worth taken into consideration because they are beneficial for designing novel anti-COVID-19 drugs in the coming investigations. Although examples collected herein are just a drop in the bucket, developments of organic small molecules against coronavirus infections are believed to pave a promising way for the discovery of multi-targeted therapeutic drugs against not only COVID-19 but also other virus-mediated diseases.
Collapse
Affiliation(s)
- Zai-Qun Liu
- Department of Organic Chemistry, College of Chemistry, Jilin University, No.2519 Jiefang Road, Changchun, 130021, People's Republic of China.
| |
Collapse
|
3
|
Yuan J, Wang J, Chen Z, Chang Y, Chen L, Gao Z, Crommen J, Zhang T, Jiang Z. Establishment of an at-line nanofractionation-based screening platform by coupling HPLC-MS/MS with high-throughput fluorescence polarization bioassay for natural SARS-CoV-2 fusion inhibitors. J Chromatogr A 2024; 1728:464986. [PMID: 38797137 DOI: 10.1016/j.chroma.2024.464986] [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: 03/21/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024]
Abstract
In this study, a novel at-line nanofractionation platform was established for screening SARS-CoV-2 fusion inhibitors from natural products for the first time by combining HPLC-MS/MS with high-throughput fluorescence polarization (FP) bioassay. A time-course FP bioassay in 384 well-plates was conducted in parallel with MS/MS to simultaneously obtain chemical and biological information of potential fusion inhibitors in Lonicerae Japonicae Flos (LJF) and Lianhua Qingwen capsules (LHQW). Semi-preparative liquid chromatography and orthogonal HPLC separation were employed to enrich and better identify the co-eluted components. After comprehensive evaluation and validation, 28 potential SARS-CoV-2 fusion inhibitors were screened out and identified. Several compounds at low micromolar activity were validated by in vitro inhibitory assay, molecular docking, cytotoxicity test, and pseudovirus assay. Moreover, four potential dual-target inhibitors against influenza and COVID-19 were discovered from LJF using this method, offering novel insights for the development of future pharmaceuticals targeting epidemic respiratory diseases.
Collapse
Affiliation(s)
- Jiaming Yuan
- Institute of Pharmaceutical Analysis, College of Pharmacy / Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research / International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Jincai Wang
- Institute of Pharmaceutical Analysis, College of Pharmacy / Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research / International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Zhixu Chen
- Institute of Pharmaceutical Analysis, College of Pharmacy / Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research / International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Yuexiang Chang
- Institute of Pharmaceutical Analysis, College of Pharmacy / Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research / International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Litong Chen
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Marine Sciences, Sun Yat-sen University, Zhuhai, 519080, China
| | - Zhizeng Gao
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Marine Sciences, Sun Yat-sen University, Zhuhai, 519080, China
| | - Jacques Crommen
- Laboratory of Analytical Pharmaceutical Chemistry, Department of Pharmaceutical Sciences, CIRM, University of Liege, CHU B36, B-4000, Liege, Belgium
| | - Tingting Zhang
- Institute of Pharmaceutical Analysis, College of Pharmacy / Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research / International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China.
| | - Zhengjin Jiang
- Institute of Pharmaceutical Analysis, College of Pharmacy / Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research / International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
4
|
Fan J, Xi P, Liu H, Song X, Zhao X, Zhou X, Zou Y, Fu Y, Li L, Jia R, Yin Z. Myricetin inhibits transmissible gastroenteritis virus replication by targeting papain-like protease deubiquitinating enzyme activity. Front Microbiol 2024; 15:1433664. [PMID: 39050632 PMCID: PMC11266173 DOI: 10.3389/fmicb.2024.1433664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Myricetin, a natural flavonoid found in various foods, was investigated for its antiviral effect against transmissible gastroenteritis virus (TGEV). This α-coronavirus causes significant economic losses in the global swine industry. The study focused on the papain-like protease (PLpro), which plays a crucial role in coronavirus immune evasion by mediating deubiquitination. Targeting PLpro could potentially disrupt viral replication and enhance antiviral responses. The results demonstrated that myricetin effectively inhibited TGEV-induced cytopathic effects in a dose-dependent manner, with an EC50 value of 31.19 μM. Myricetin significantly reduced TGEV viral load within 48 h after an 8-h co-incubation period. Further investigations revealed that myricetin at a concentration of 100 μM directly inactivated TGEV and suppressed its intracellular replication stage. Moreover, pretreatment with 100 μM myricetin conferred a protective effect on PK-15 cells against TGEV infection. Myricetin competitively inhibited PLpro with an IC50 value of 6.563 μM. Molecular docking experiments show that myricetin binds to the Cys102 residue of PLpro through conventional hydrogen bonds, Pi-sulfur, and Pi-alkyl interactions. This binding was confirmed through site-directed mutagenesis experiments, indicating myricetin as a potential candidate for preventing and treating TGEV infection.
Collapse
Affiliation(s)
- Jiahao Fan
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Pengyuan Xi
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Huimao Liu
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xu Song
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xinghong Zhao
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xun Zhou
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yuanfeng Zou
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yuping Fu
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lixia Li
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Renyong Jia
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Zhongqiong Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
5
|
Ferreira GC, Manzato VDM, Okamoto DN, Fernandes LR, Santos DM, Costa GCA, Silva FAA, Torquato RJS, Palmisano G, Juliano MA, Tanaka AS. Sunflower Trypsin Monocyclic Inhibitor Selected for the Main Protease of SARS-CoV-2 by Phage Display. Biol Pharm Bull 2024; 47:1813-1822. [PMID: 39522974 DOI: 10.1248/bpb.b24-00369] [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: 11/16/2024]
Abstract
Main protease (Mpro), also known as 3-chymotrypsin-like protease (3CLpro), is a nonstructural protein (NSP5) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) responsible for the cleavage of virus polyproteins during viral replication at 11 sites, which generates 12 functional proteins. Mpro is a cysteine protease that presents specificity for the amino acid residue glutamine (Gln) at the P1 position of the substrate. Due to its essential role in processing the viral polyprotein for viral particle formation (assembly), Mpro inhibition has become an important tool to control coronavirus disease 2019 (COVID-19), since Mpro inhibitors act as antivirals. In this work, we proposed to identify specific inhibitors of the Mpro of SARS-CoV-2 using a monocyclic peptide (sunflower trypsin inhibitor (SFTI)) phage display library. Initially, we expressed, purified and activated recombinant Mpro. The screening of the mutant SFTI phage display library using recombinant Mpro as a receptor resulted in the five most frequent SFTI mutant sequences. Synthetized mutant SFTIs did not inhibit Mpro protease using the fluorogenic substrate. However, the mutant SFTI 4 efficiently decreased the cleavage of recombinant human prothrombin as a substrate by Mpro, as confirmed by sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis (PAGE). Additionally, SFTI 4 presented a dissociation constant (KD) of 21.66 ± 6.66 µM for Mpro by surface plasmon resonance. Finally, 0.1 µM SFTI 4 reduced VERO cell infection by SARS-CoV-2 wt after 24 and 48 h. In conclusion, we successfully screened a monocyclic peptide library using phage display for the Mpro of SARS-CoV-2, suggesting that this methodology can be useful in identifying new inhibitors of viral enzymes.
Collapse
Affiliation(s)
| | | | - Debora Noma Okamoto
- Institute of Environmental, Chemical and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, Federal University of São Paulo
| | | | | | | | | | | | - Giuseppe Palmisano
- Institute of Biomedical Sciences, University of São Paulo
- School of Natural Sciences, Macquarie University
| | | | - Aparecida Sadae Tanaka
- Department of Biochemistry, Paulista School of Medicine, Federal University of São Paulo
- National Institute of Science and Technology for Molecular Entomology (INCT-EM), Institute of Medical Biochemistry-Federal University of Rio de Janeiro
| |
Collapse
|
6
|
Singh Dagur H, Behmard E, Rajakumara E, Barzegari E. Identifying potent inhibitory phytocompounds from Lagerstroemia speciosa against SARS-Coronavirus-2: structure-based virtual screening. J Biomol Struct Dyn 2024; 42:806-818. [PMID: 37170794 DOI: 10.1080/07391102.2023.2205942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 03/20/2023] [Indexed: 05/13/2023]
Abstract
The ongoing spillover of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) calls for expedited countermeasure through developing therapeutics from natural reservoirs and/or the use of less time-consuming drug discovery methodologies. This study aims to apply these approaches to identify potential blockers of the virus from the longstanding medicinal herb, Lagerstroemia speciosa, through comprehensive computational-based screening. Nineteen out of 22 L. speciosa phytochemicals were selected on the basis of their pharmacokinetic properties. SARS-CoV-2 Main protease (Mpro), RNA-directed RNA polymerase (RdRp), Envelope viroporin protein (Evp) and receptor-binding domain of Spike glycoprotein (S-RBD), as well as the human receptor Angiotensin-converting enzyme-2 (hACE2) were chosen as targets. The screening was performed by molecular docking, followed by 100-ns molecular dynamic simulations and free energy calculations. 24-Methylene cycloartanol acetate (24MCA) was found as the best inhibitor for both Evp and RdRp, and sitosterol acetate (SA) as the best hit for Mpro, S-RBD and hACE2. Dynamic simulations, binding mode analyses, free energy terms and share of key amino acids in protein-drug interactions confirmed the stable binding of these phytocompounds to the hotspot sites on the target proteins. With their possible multi-targeting capability, the introduced phytoligands might offer promising lead compounds for persistent fight with the rapidly evolving coronavirus. Therefore, experimental verification of their safety and efficacy is recommended.
Collapse
Affiliation(s)
- Hanuman Singh Dagur
- Macromolecular Structural Biology Lab, Department of Biotechnology, Indian Institute of Technology Hyderabad, Sangareddy, Telangana, India
| | - Esmaeil Behmard
- School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Eerappa Rajakumara
- Macromolecular Structural Biology Lab, Department of Biotechnology, Indian Institute of Technology Hyderabad, Sangareddy, Telangana, India
| | - Ebrahim Barzegari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
7
|
Haapakoski M, Emelianov A, Reshamwala D, Laajala M, Tienaho J, Kilpeläinen P, Liimatainen J, Jyske T, Pettersson M, Marjomäki V. Antiviral functionalization of cellulose using tannic acid and tannin-rich extracts. Front Microbiol 2023; 14:1287167. [PMID: 38125579 PMCID: PMC10731304 DOI: 10.3389/fmicb.2023.1287167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/13/2023] [Indexed: 12/23/2023] Open
Abstract
Due to seasonally appearing viruses and several outbreaks and present pandemic, we are surrounded by viruses in our everyday life. In order to reduce viral transmission, functionalized surfaces that inactivate viruses are in large demand. Here the endeavor was to functionalize cellulose-based materials with tannic acid (TA) and tannin-rich extracts by using different binding polymers to prevent viral infectivity of both non-enveloped coxsackievirus B3 (CVB3) and enveloped human coronavirus OC43 (HCoV-OC43). Direct antiviral efficacy of TA and spruce bark extract in solution was measured: EC50 for CVB3 was 0.12 and 8.41 μg/ml and for HCoV-OC43, 78.16 and 95.49 μg/ml, respectively. TA also led to an excellent 5.8- to 7-log reduction of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus infectivity. TA functionalized materials reduced infectivity already after 5-min treatment at room temperature. All the tested methods to bind TA showed efficacy on paperboard with 0.1 to 1% (w/v) TA concentrations against CVB3 whereas material hydrophobicity decreased activities. Specific signatures for TA and HCoV-OC43 were discovered by Raman spectroscopy and showed clear co-localization on the material. qPCR study suggested efficient binding of CVB3 to the TA functionalized cellulose whereas HCoV-OC43 was flushed out from the surfaces more readily. In conclusion, the produced TA-materials showed efficient and broadly acting antiviral efficacy. Additionally, the co-localization of TA and HCoV-OC43 and strong binding of CVB3 to the functionalized cellulose demonstrates an interaction with the surfaces. The produced antiviral surfaces thus show promise for future use to increase biosafety and biosecurity by reducing pathogen persistence.
Collapse
Affiliation(s)
- Marjo Haapakoski
- Department of Biological and Environmental Sciences/Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Aleksei Emelianov
- Department of Chemistry/Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Dhanik Reshamwala
- Department of Biological and Environmental Sciences/Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Mira Laajala
- Department of Biological and Environmental Sciences/Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Jenni Tienaho
- Production Systems Unit, Natural Resources Institute Finland (Luke), Helsinki, Finland
| | - Petri Kilpeläinen
- Production Systems Unit, Natural Resources Institute Finland (Luke), Helsinki, Finland
| | - Jaana Liimatainen
- Production Systems Unit, Natural Resources Institute Finland (Luke), Helsinki, Finland
| | - Tuula Jyske
- Production Systems Unit, Natural Resources Institute Finland (Luke), Helsinki, Finland
| | - Mika Pettersson
- Department of Chemistry/Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Varpu Marjomäki
- Department of Biological and Environmental Sciences/Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| |
Collapse
|
8
|
Bianconi E, Gidari A, Souma M, Sabbatini S, Grifagni D, Bigiotti C, Schiaroli E, Comez L, Paciaroni A, Cantini F, Francisci D, Macchiarulo A. The hope and hype of ellagic acid and urolithins as ligands of SARS-CoV-2 Nsp5 and inhibitors of viral replication. J Enzyme Inhib Med Chem 2023; 38:2251721. [PMID: 37638806 PMCID: PMC10464554 DOI: 10.1080/14756366.2023.2251721] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 08/29/2023] Open
Abstract
Non-structural protein 5 (Nsp5) is a cysteine protease that plays a key role in SARS-CoV-2 replication, suppressing host protein synthesis and promoting immune evasion. The investigation of natural products as a potential strategy for Nsp5 inhibition is gaining attention as a means of developing antiviral agents. In this work, we have investigated the physicochemical properties and structure-activity relationships of ellagic acid and its gut metabolites, urolithins A-D, as ligands of Nsp5. Results allow us to identify urolithin D as promising ligand of Nsp5, with a dissociation constant in the nanomolar range of potency. Although urolithin D is able to bind to the catalytic cleft of Nsp5, the appraisal of its viral replication inhibition against SARS-CoV-2 in Vero E6 assay highlights a lack of activity. While these results are discussed in the framework of the available literature reporting conflicting data on polyphenol antiviral activity, they provide new clues for natural products as potential viral protease inhibitors.
Collapse
Affiliation(s)
- Elisa Bianconi
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Anna Gidari
- Department of Medicine and Surgery, Clinic of Infectious Diseases, University of Perugia, Perugia, Italy
| | - Maria Souma
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Samuele Sabbatini
- Medical Microbiology Section, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Deborah Grifagni
- Centre for Magnetic Resonance, University of Florence, Sesto Fiorentino, Italy
- Department of Chemistry, University of Florence, Sesto Fiorentino, Italy
| | - Carlo Bigiotti
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Elisabetta Schiaroli
- Department of Medicine and Surgery, Clinic of Infectious Diseases, University of Perugia, Perugia, Italy
| | - Lucia Comez
- Istituto Officina dei Materiali-IOM, National Research Council-CNR, Perugia, Italy
| | | | - Francesca Cantini
- Centre for Magnetic Resonance, University of Florence, Sesto Fiorentino, Italy
- Department of Chemistry, University of Florence, Sesto Fiorentino, Italy
| | - Daniela Francisci
- Department of Medicine and Surgery, Clinic of Infectious Diseases, University of Perugia, Perugia, Italy
| | - Antonio Macchiarulo
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| |
Collapse
|
9
|
Alexova R, Alexandrova S, Dragomanova S, Kalfin R, Solak A, Mehan S, Petralia MC, Fagone P, Mangano K, Nicoletti F, Tancheva L. Anti-COVID-19 Potential of Ellagic Acid and Polyphenols of Punica granatum L. Molecules 2023; 28:molecules28093772. [PMID: 37175181 PMCID: PMC10180134 DOI: 10.3390/molecules28093772] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Pomegranate (Punica granatum L.) is a rich source of polyphenols, including ellagitannins and ellagic acid. The plant is used in traditional medicine, and its purified components can provide anti-inflammatory and antioxidant activity and support of host defenses during viral infection and recovery from disease. Current data show that pomegranate polyphenol extract and its ellagitannin components and metabolites exert their beneficial effects by controlling immune cell infiltration, regulating the cytokine secretion and reactive oxygen and nitrogen species production, and by modulating the activity of the NFκB pathway. In vitro, pomegranate extracts and ellagitannins interact with and inhibit the infectivity of a range of viruses, including SARS-CoV-2. In silico docking studies show that ellagitannins bind to several SARS-CoV-2 and human proteins, including a number of proteases. This warrants further exploration of polyphenol-viral and polyphenol-host interactions in in vitro and in vivo studies. Pomegranate extracts, ellagitannins and ellagic acid are promising agents to target the SARS-CoV-2 virus and to restrict the host inflammatory response to viral infections, as well as to supplement the depleted host antioxidant levels during the stage of recovery from COVID-19.
Collapse
Affiliation(s)
- Ralitza Alexova
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Medical University-Sofia, Zdrave Str. 2, 1431 Sofia, Bulgaria
| | - Simona Alexandrova
- Department of Biological Effects of Natural and Synthetic Substances, Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. Georgi Bonchev Str., Block 23, 1113 Sofia, Bulgaria
| | - Stela Dragomanova
- Department of Pharmacology, Toxicology and Pharmacotherapy, Faculty of Pharmacy, Medical University, Marin Drinov Str. 55, 9002 Varna, Bulgaria
| | - Reni Kalfin
- Department of Biological Effects of Natural and Synthetic Substances, Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. Georgi Bonchev Str., Block 23, 1113 Sofia, Bulgaria
- Department of Healthcare, South-West University "Neofit Rilski", Ivan Mihailov Str. 66, 2700 Blagoevgrad, Bulgaria
| | - Ayten Solak
- Institute of Cryobiology and Food Technologies, Cherni Vrah Blvd. 5, 1407 Sofia, Bulgaria
| | - Sidharth Mehan
- Department of Pharmacology, Division of Neuroscience, ISF College of Pharmacy, Moga 142001, India
| | - Maria Cristina Petralia
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy
| | - Lyubka Tancheva
- Department of Biological Effects of Natural and Synthetic Substances, Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. Georgi Bonchev Str., Block 23, 1113 Sofia, Bulgaria
| |
Collapse
|
10
|
Xu W, Ning Y, Cao S, Wu G, Sun H, Chai L, Wu S, Li J, Luo D. Insight into the interaction between tannin acid and bovine serum albumin from a spectroscopic and molecular docking perspective. RSC Adv 2023; 13:10592-10599. [PMID: 37025671 PMCID: PMC10071303 DOI: 10.1039/d3ra00375b] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/20/2023] [Indexed: 04/08/2023] Open
Abstract
In this study, the interaction mechanism of bovine serum albumin (BSA) with tannic acid (TA) was investigated by spectroscopic and computational approaches and further validated using circular dichroism (CD), differential scanning calorimetry (DSC) and molecular docking techniques. The fluorescence spectra showed that TA bound to BSA and underwent static quenching at a single binding site, which was consistent with the molecular docking results. And the fluorescence quenching of BSA by TA was dose-dependent. Thermodynamic analysis indicated that hydrophobic forces dominated the interaction of BSA with TA. The results of circular dichroism showed that the secondary structure of BSA was slightly changed after coupling with TA. Differential scanning calorimetry showed that the interaction between BSA and TA improved the stability of the BSA-TA complex, and the melting temperature increased to 86.67 °C and the enthalpy increased to 264.1 J g-1 when the ratio of TA to BSA was 1.2 : 1. Molecular docking techniques revealed specific amino acid binding sites for the BSA-TA complex with a docking energy of -12.9 kcal mol-1, which means the TA is non-covalently bound to the BSA active site.
Collapse
Affiliation(s)
- Wei Xu
- College of Life Science, Xinyang Normal University Xinyang 464000 China
| | - Yuli Ning
- College of Life Science, Xinyang Normal University Xinyang 464000 China
| | - Shiwan Cao
- College of Life Science, Xinyang Normal University Xinyang 464000 China
| | - Guanchen Wu
- College of Life Science, Xinyang Normal University Xinyang 464000 China
| | - Haomin Sun
- College of Food and Bioengineering, Henan University of Science and Technology Luoyang 471023 China
| | - Liwen Chai
- College of Food and Bioengineering, Henan University of Science and Technology Luoyang 471023 China
| | - Shuping Wu
- College of Food and Bioengineering, Henan University of Science and Technology Luoyang 471023 China
| | - Jingyi Li
- College of Life Science, Xinyang Normal University Xinyang 464000 China
| | - Denglin Luo
- College of Food and Bioengineering, Henan University of Science and Technology Luoyang 471023 China
| |
Collapse
|
11
|
Wang Q, Song Y, Kim M, Hahn SK, Jiang G. Effect of chitooligosaccharide on the inhibition of SARS-CoV-2 main protease. Biomater Res 2023; 27:13. [PMID: 36797775 PMCID: PMC9935244 DOI: 10.1186/s40824-023-00351-4] [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: 11/16/2022] [Accepted: 02/05/2023] [Indexed: 02/18/2023] Open
Abstract
BACKGROUND The main protease (Mpro) is a crucial target for severe acute respiratory syndrome coronavirus (SARS-CoV-2). Chitooligosaccharide (CS) has broad-spectrum antiviral activity and can effectively inhibit the activity of SARS-CoV. Here, based on the high homology between SARS-CoV-2 and SARS-CoV, this study explores the effect and mechanism of CS with various molecular weights on the activity of SARS-CoV-2 Mpro. METHODS We used fluorescence resonance energy transfer (FRET), UV-Vis, synchronous fluorescence spectroscopy, circular dichroism (CD) spectroscopy and computational simulation to investigate the molecular interaction and the interaction mechanism between CS and SARS-CoV-2 Mpro. RESULTS Four kinds of CS with different molecular weights significantly inhibited the activity of Mpro by combining the hydrogen bonding and the salt bridge interaction to form a stable complex. Glu166 appeared to be the key amino acid. Among them, chitosan showed the highest inhibition effect on Mpro enzyme activity and the greatest impact on the spatial structure of protein. Chitosan would be one of the most potential anti-viral compounds. CONCLUSION This study provides the theoretical basis to develop targeted Mpro inhibitors for the screening and application of anti-novel coronavirus drugs.
Collapse
Affiliation(s)
- Qian Wang
- grid.440706.10000 0001 0175 8217Bioengineering College, Dalian University, 10 Xuefu Street, Jinzhou District, Dalian, 116600 Liaoning China
| | - Yuanyuan Song
- grid.440706.10000 0001 0175 8217Bioengineering College, Dalian University, 10 Xuefu Street, Jinzhou District, Dalian, 116600 Liaoning China
| | - Mungu Kim
- grid.49100.3c0000 0001 0742 4007Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, 790-784 Gyeongbuk Korea
| | - Sei Kwang Hahn
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, 790-784, Gyeongbuk, Korea.
| | - Ge Jiang
- Bioengineering College, Dalian University, 10 Xuefu Street, Jinzhou District, Dalian, 116600, Liaoning, China.
| |
Collapse
|
12
|
Liu C, Puopolo T, Li H, Cai A, Seeram NP, Ma H. Identification of SARS-CoV-2 Main Protease Inhibitors from a Library of Minor Cannabinoids by Biochemical Inhibition Assay and Surface Plasmon Resonance Characterized Binding Affinity. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27186127. [PMID: 36144858 PMCID: PMC9502466 DOI: 10.3390/molecules27186127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 01/08/2023]
Abstract
The replication of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is mediated by its main protease (Mpro), which is a plausible therapeutic target for coronavirus disease 2019 (COVID-19). Although numerous in silico studies reported the potential inhibitory effects of natural products including cannabis and cannabinoids on SARS-CoV-2 Mpro, their anti-Mpro activities are not well validated by biological experimental data. Herein, a library of minor cannabinoids belonging to several chemotypes including tetrahydrocannabinols, cannabidiols, cannabigerols, cannabichromenes, cannabinodiols, cannabicyclols, cannabinols, and cannabitriols was evaluated for their anti-Mpro activity using a biochemical assay. Additionally, the binding affinities and molecular interactions between the active cannabinoids and the Mpro protein were studied by a biophysical technique (surface plasmon resonance; SPR) and molecular docking, respectively. Cannabinoids tetrahydrocannabutol and cannabigerolic acid were the most active Mpro inhibitors (IC50 = 3.62 and 14.40 μM, respectively) and cannabigerolic acid had a binding affinity KD=2.16×10-4 M). A preliminary structure and activity relationship study revealed that the anti-Mpro effects of cannabinoids were influenced by the decarboxylation of cannabinoids and the length of cannabinoids' alkyl side chain. Findings from the biochemical, biophysical, and computational assays support the growing evidence of cannabinoids' inhibitory effects on SARS-CoV-2 Mpro.
Collapse
Affiliation(s)
- Chang Liu
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
- Cannabis Research Collaborative, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Tess Puopolo
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Huifang Li
- 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
| | - Navindra P. Seeram
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
- Cannabis Research Collaborative, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
- Correspondence: (N.P.S.); (H.M.); Tel.: +1-(401)-874-9367 (N.P.S.); +1-(401)-874-2711 (H.M.)
| | - Hang Ma
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
- Cannabis Research Collaborative, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
- Correspondence: (N.P.S.); (H.M.); Tel.: +1-(401)-874-9367 (N.P.S.); +1-(401)-874-2711 (H.M.)
| |
Collapse
|
13
|
Jin YH, Lee J, Jeon S, Kim S, Min JS, Kwon S. Natural Polyphenols, 1,2,3,4,6-O-Pentagalloyglucose and Proanthocyanidins, as Broad-Spectrum Anticoronaviral Inhibitors Targeting Mpro and RdRp of SARS-CoV-2. Biomedicines 2022; 10:biomedicines10051170. [PMID: 35625907 PMCID: PMC9138959 DOI: 10.3390/biomedicines10051170] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 02/04/2023] Open
Abstract
The natural plant dietary polyphenols 1,2,3,4,6-O-Pentagalloylglucose (PGG) and proanthocyanidin (PAC) have potent antioxidant activity and a variety of pharmacological activities, including antiviral activity. In this study, we examined the inhibitory effect of PGG and PAC on SARS-CoV-2 virus infection, and elucidated its mode of action. PGG and PAC have dose-dependent inhibitory activity against SARS-CoV-2 infection in Vero cells. PGG has a lower IC50 (15.02 ± 0.75 μM) than PAC (25.90 ± 0.81 μM), suggesting that PGG has better inhibitory activity against SARS-CoV-2 than PAC. The PGG and PAC inhibit similar Mpro activities in a protease activity assay, with IC50 values of 25–26 μM. The effects of PGG and PAC on the activity of the other essential SARS-CoV-2 viral protein, RdRp, were analyzed using a cell-based activity assay system. The activity of RdRp is inhibited by PGG and PAC, and PGG has a lower IC50 (5.098 ± 1.089 μM) than PAC (21.022 ± 1.202 μM), which is consistent with their inhibitory capacity of SARS-CoV-2 infection. PGG and PAC also inhibit infection by SARS-CoV and MERS-CoV. These data indicate that PGG and PAC may be candidate broad-spectrum anticoronaviral therapeutic agents, simultaneously targeting the Mpro and RdRp proteins of SARS-CoV-2.
Collapse
Affiliation(s)
- Young-Hee Jin
- KM Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Korea
- Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea;
- Correspondence: (Y.-H.J.); (S.K.); Tel.: +82-(42)-610-8850 (Y.-H.J.); +82-(42)-868-9675 (S.K.)
| | - Jihye Lee
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam 13488, Korea; (J.L.); (S.J.); (S.K.)
| | - Sangeun Jeon
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam 13488, Korea; (J.L.); (S.J.); (S.K.)
| | - Seungtaek Kim
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam 13488, Korea; (J.L.); (S.J.); (S.K.)
| | - Jung Sun Min
- Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea;
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea
| | - Sunoh Kwon
- Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea;
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea
- Correspondence: (Y.-H.J.); (S.K.); Tel.: +82-(42)-610-8850 (Y.-H.J.); +82-(42)-868-9675 (S.K.)
| |
Collapse
|
14
|
Liu C, Cai A, Li H, Deng N, Cho BP, Seeram NP, Ma H. Characterization of molecular interactions between cannabidiol and human plasma proteins (serum albumin and γ-globulin) by surface plasmon resonance, microcalorimetry, and molecular docking. J Pharm Biomed Anal 2022; 214:114750. [DOI: 10.1016/j.jpba.2022.114750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/30/2022] [Accepted: 04/01/2022] [Indexed: 01/22/2023]
|
15
|
Use of hyphenated analytical techniques to identify the bioactive constituents of Gunnera perpensa L., a South African medicinal plant, which potently inhibit SARS-CoV-2 spike glycoprotein-host ACE2 binding. Anal Bioanal Chem 2022; 414:3971-3985. [PMID: 35419694 PMCID: PMC9007697 DOI: 10.1007/s00216-022-04041-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/21/2022] [Accepted: 03/23/2022] [Indexed: 11/16/2022]
Abstract
SARS-CoV-2, the causative agent of COVID-19, continues to cause global morbidity and mortality despite the increasing availability of vaccines. Alongside vaccines, antivirals are urgently needed to combat SARS-CoV-2 infection and spread, particularly in resource-limited regions which lack access to existing therapeutics. Small molecules isolated from medicinal plants may be able to block cellular entry by SARS-CoV-2 by antagonising the interaction of the viral spike glycoprotein receptor-binding domain (RBD) with the host angiotensin-converting enzyme II (ACE2) receptor. As the medicinal plant Gunnera perpensa L. is being used by some South African traditional healers for SARS-CoV-2/COVID-19 management, we hypothesised that it may contain chemical constituents that inhibit the RBD-ACE2 interaction. Using a previously described AlphaScreen-based protein interaction assay, we show here that the DCM:MeOH extract of G. perpensa readily disrupts RBD (USA-WA1/2020)-ACE2 interactions with a half-maximal inhibition concentration (IC50) of < 0.001 µg/mL, compared to an IC50 of 0.025 µg/mL for the control neutralising antibody REGN10987. Employing hyphenated analytical techniques like UPLC-IMS-HRMS (method developed and validated as per the International Conference on Harmonization guidelines), we identified two ellagitannins, punicalin (2.12% w/w) and punicalagin (1.51% w/w), as plant constituents in the DCM:MeOH extract of G. perpensa which antagonised RBD-ACE2 binding with respective IC50s of 9 and 29 nM. This good potency makes both compounds promising leads for development of future entry-based SARS-CoV-2 antivirals. The results also highlight the advantages of combining reverse pharmacology (based on medicinal plant use) with hyphenated analytical techniques to expedite identification of urgently needed antivirals.
Collapse
|
16
|
Haddad M, Gaudreault R, Sasseville G, Nguyen PT, Wiebe H, Van De Ven T, Bourgault S, Mousseau N, Ramassamy C. Molecular Interactions of Tannic Acid with Proteins Associated with SARS-CoV-2 Infectivity. Int J Mol Sci 2022; 23:2643. [PMID: 35269785 PMCID: PMC8910432 DOI: 10.3390/ijms23052643] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/16/2022] [Accepted: 02/23/2022] [Indexed: 02/01/2023] Open
Abstract
The overall impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on our society is unprecedented. The identification of small natural ligands that could prevent the entry and/or replication of the coronavirus remains a pertinent approach to fight the coronavirus disease (COVID-19) pandemic. Previously, we showed that the phenolic compounds corilagin and 1,3,6-tri-O-galloyl-β-D-glucose (TGG) inhibit the interaction between the SARS-CoV-2 spike protein receptor binding domain (RBD) and angiotensin-converting enzyme 2 (ACE2), the SARS-CoV-2 target receptor on the cell membrane of the host organism. Building on these promising results, we now assess the effects of these phenolic ligands on two other crucial targets involved in SARS-CoV-2 cell entry and replication, respectively: transmembrane protease serine 2 (TMPRSS2) and 3-chymotrypsin like protease (3CLpro) inhibitors. Since corilagin, TGG, and tannic acid (TA) share many physicochemical and structural properties, we investigate the binding of TA to these targets. In this work, a combination of experimental methods (biochemical inhibition assays, surface plasmon resonance, and quartz crystal microbalance with dissipation monitoring) confirms the potential role of TA in the prevention of SARS-CoV-2 infectivity through the inhibition of extracellular RBD/ACE2 interactions and TMPRSS2 and 3CLpro activity. Moreover, molecular docking prediction followed by dynamic simulation and molecular mechanics Poisson-Boltzmann surface area (MMPBSA) free energy calculation also shows that TA binds to RBD, TMPRSS2, and 3CLpro with higher affinities than TGG and corilagin. Overall, these results suggest that naturally occurring TA is a promising candidate to prevent and inhibit the infectivity of SARS-CoV-2.
Collapse
Affiliation(s)
- Mohamed Haddad
- Centre Armand-Frappier Santé Biotechnologie, 531 Boulevard des Prairies, Laval, QC H7V 1B7, Canada;
- Institute on Nutrition and Functional Foods, Laval University, Quebec City, QC G1V 0A6, Canada
| | - Roger Gaudreault
- Succursale Centre-Ville, Départment de Physique, Université de Montréal, Case Postale 6128, Montréal, QC H3C 3J7, Canada; (R.G.); (G.S.); (N.M.)
| | - Gabriel Sasseville
- Succursale Centre-Ville, Départment de Physique, Université de Montréal, Case Postale 6128, Montréal, QC H3C 3J7, Canada; (R.G.); (G.S.); (N.M.)
| | - Phuong Trang Nguyen
- Département de Chimie, Université du Québec à Montréal, 2101 Rue Jeanne-Mance, Montréal, QC H2X 2J6, Canada; (P.T.N.); (S.B.)
| | - Hannah Wiebe
- Département de Chimie, Université McGill, 3420 Rue University, Montréal, QC H3A 2A7, Canada; (H.W.); (T.V.D.V.)
| | - Theo Van De Ven
- Département de Chimie, Université McGill, 3420 Rue University, Montréal, QC H3A 2A7, Canada; (H.W.); (T.V.D.V.)
| | - Steve Bourgault
- Département de Chimie, Université du Québec à Montréal, 2101 Rue Jeanne-Mance, Montréal, QC H2X 2J6, Canada; (P.T.N.); (S.B.)
| | - Normand Mousseau
- Succursale Centre-Ville, Départment de Physique, Université de Montréal, Case Postale 6128, Montréal, QC H3C 3J7, Canada; (R.G.); (G.S.); (N.M.)
| | - Charles Ramassamy
- Centre Armand-Frappier Santé Biotechnologie, 531 Boulevard des Prairies, Laval, QC H7V 1B7, Canada;
- Institute on Nutrition and Functional Foods, Laval University, Quebec City, QC G1V 0A6, Canada
| |
Collapse
|
17
|
Vini R, Azeez JM, Remadevi V, Susmi TR, Ayswarya RS, Sujatha AS, Muraleedharan P, Lathika LM, Sreeharshan S. Urolithins: The Colon Microbiota Metabolites as Endocrine Modulators: Prospects and Perspectives. Front Nutr 2022; 8:800990. [PMID: 35187021 PMCID: PMC8849129 DOI: 10.3389/fnut.2021.800990] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 12/10/2021] [Indexed: 12/19/2022] Open
Abstract
Selective estrogen receptor modulators (SERMs) have been used in hormone related disorders, and their role in clinical medicine is evolving. Tamoxifen and raloxifen are the most commonly used synthetic SERMs, and their long-term use are known to create side effects. Hence, efforts have been directed to identify molecules which could retain the beneficial effects of estrogen, at the same time produce minimal side effects. Urolithins, the products of colon microbiota from ellagitannin rich foodstuff, have immense health benefits and have been demonstrated to bind to estrogen receptors. This class of compounds holds promise as therapeutic and nutritional supplement in cardiovascular disorders, osteoporosis, muscle health, neurological disorders, and cancers of breast, endometrium, and prostate, or, in essence, most of the hormone/endocrine-dependent diseases. One of our findings from the past decade of research on SERMs and estrogen modulators, showed that pomegranate, one of the indirect but major sources of urolithins, can act as SERM. The prospect of urolithins to act as agonist, antagonist, or SERM will depend on its structure; the estrogen receptor conformational change, availability and abundance of co-activators/co-repressors in the target tissues, and also the presence of other estrogen receptor ligands. Given that, urolithins need to be carefully studied for its SERM activity considering the pleotropic action of estrogen receptors and its numerous roles in physiological systems. In this review, we unveil the possibility of urolithins as a potent SERM, which we are currently investigating, in the hormone dependent tissues.
Collapse
Affiliation(s)
- Ravindran Vini
- Cancer Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Juberiya M. Azeez
- Cancer Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Viji Remadevi
- Cancer Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - T. R. Susmi
- Cancer Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - R. S. Ayswarya
- Cancer Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | | | | | - Lakshmi Mohan Lathika
- Cancer Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Sreeja Sreeharshan
- Cancer Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- *Correspondence: Sreeja Sreeharshan
| |
Collapse
|
18
|
Abstract
The emerging risk of viral diseases has triggered the search for preventive and therapeutic agents. Since the beginning of the COVID-19 pandemic, greater efforts have been devoted to investigating virus entry mechanisms into host cells. The feasibility of plasmonic sensing technologies for screening interactions of small molecules in real time, while providing the pharmacokinetic drug profiling of potential antiviral compounds, offers an advantageous approach over other biophysical methods. This review summarizes recent advancements in the drug discovery process of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) inhibitors using Surface Plasmon Resonance (SPR) biosensors. A variety of SPR assay formats are discussed according to the binding kinetics and drug efficacies of both natural products and repurposed drugs. Special attention has been given to the targeting of antiviral agents that block the receptor binding domain of the spike protein (RBD-S) and the main protease (3CLpro) of SARS-CoV-2. The functionality of plasmonic biosensors for high-throughput screening of entry virus inhibitors was also reviewed taking into account experimental parameters (binding affinities, selectivity, stability), potential limitations and future applications.
Collapse
|