1
|
Al-Shuhaib MBS, Al-Shuhaib JMB. Assessing Therapeutic Value and Side Effects of Key Botanical Compounds for Optimized Medical Treatments. Chem Biodivers 2025; 22:e202401754. [PMID: 39316731 DOI: 10.1002/cbdv.202401754] [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: 07/19/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 09/26/2024]
Abstract
Due to the significance of variable chemical groups across a wide spectrum of modern medicine, it is imperative to determine what is the most widely used group in medical applications with the fewest side effects. Ten compounds from ten chemical groups that are most commonly known for their medical uses were compared in terms of their therapeutic potential and side effects. The comparison among the selected compounds indicated the superiority of the flavonoids over other groups in the multitude of their utilizations and the lower side effects. Kaempferol and quercetin showed higher medical utilization with lower side effects. Whereas alkaloid compounds showed the lowest levels of medical use and the highest levels of side effects. Based on the comparison conducted, it is concluded to give priority to flavonoid compounds being used in medical applications because they exhibit the highest medical uses with the lowest side effects. Within flavonoids, kaempferol and quercetin are the two compounds that are highly recommended to be used in the widest range of medical applications. Serious caution should be considered before applying alkaloids to any medical service. Understanding the characteristics of these compounds can aid in developing safer and more effective treatments for medicinal plants.
Collapse
Affiliation(s)
- Mohammed Baqur S Al-Shuhaib
- Department of Animal Production, College of Agriculture, Al-Qasim Green University, 8 Babil, Al-Qasim, 51013, Iraq
| | | |
Collapse
|
2
|
Visvanathan R, Le DT, Dhital S, Rali T, Davis RA, Williamson G. Inhibition of Human Salivary and Pancreatic α-Amylase by Resveratrol Oligomers. J Med Chem 2024; 67:18753-18763. [PMID: 39501642 PMCID: PMC11571111 DOI: 10.1021/acs.jmedchem.4c01042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 11/15/2024]
Abstract
A key strategy to mitigate postprandial hyperglycemia involves inhibiting α-amylases, which commence the starch digestion process in the gut. This study examined the inhibitory effects of resveratrol and stilbenoid tetramers, vaticanol B, (-)-hopeaphenol, and vatalbinoside A on human salivary and pancreatic α-amylases experimentally and through molecular docking studies. Vaticanol B demonstrated the most potent inhibition with IC50 values of 5.3 ± 0.3 μM for salivary and 6.1 ± 0.5 μM for pancreatic α-amylase (compared to acarbose with IC50 values of 1.2 ± 0.1 μM and 0.5 ± 0.0 μM, respectively). Kinetic analysis suggested a competitive inhibition mode for vaticanol B. Resveratrol and vatalbinoside A were poor inhibitors of human α-amylases, while (-)-hopeaphenol exhibited moderate inhibition. Molecular docking supported the inhibition data, and several aspects of the structural configurations explained the stronger inhibition exerted by vaticanol B. Overall, vaticanol B shows promise as a natural alternative to acarbose for inhibiting α-amylase.
Collapse
Affiliation(s)
- Rizliya Visvanathan
- Molecular
Nutrition Group, Department of Nutrition, Dietetics and Food, Monash University, Victorian Heart Institute, Victoria
Heart Hospital, 631 Blackburn
Road, Clayton, VIC 3168, Australia
| | - Dang Truong Le
- Molecular
Nutrition Group, Department of Nutrition, Dietetics and Food, Monash University, Victorian Heart Institute, Victoria
Heart Hospital, 631 Blackburn
Road, Clayton, VIC 3168, Australia
- Bioresource
Processing Research Institute of Australia (BioPRIA), Department of
Chemical and Biological Engineering, Monash
University, Clayton, VIC 3800, Australia
| | - Sushil Dhital
- Bioresource
Processing Research Institute of Australia (BioPRIA), Department of
Chemical and Biological Engineering, Monash
University, Clayton, VIC 3800, Australia
| | - Topul Rali
- School
of Natural and Physical Sciences, The University
of Papua New Guinea, Port Moresby, Papua New Guinea
| | - Rohan A. Davis
- Institute
for Biomedicine and Glycomics, Griffith
University, Brisbane, QLD 4111, Australia
| | - Gary Williamson
- Molecular
Nutrition Group, Department of Nutrition, Dietetics and Food, Monash University, Victorian Heart Institute, Victoria
Heart Hospital, 631 Blackburn
Road, Clayton, VIC 3168, Australia
| |
Collapse
|
3
|
Poli ANR, Tietjen I, Nandwana NK, Cassel J, Messick TE, Register ET, Keeney F, Rajaiah R, Verma AK, Pandey K, Acharya A, Byrareddy SN, Montaner LJ, Salvino JM. Design of novel and highly selective SARS-CoV-2 main protease inhibitors. Antimicrob Agents Chemother 2024; 68:e0056224. [PMID: 39225484 PMCID: PMC11459967 DOI: 10.1128/aac.00562-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 08/04/2024] [Indexed: 09/04/2024] Open
Abstract
We have synthesized a novel and highly selective severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main protease peptide mimetic inhibitor mimicking the replicase 1ab recognition sequence -Val-Leu-Gln- and utilizing a cysteine selective acyloxymethyl ketone as the electrophilic warhead to target the active site Cys145. Utilizing a constrained cyclic peptide that locks the conformation between the P3 (Val) and P2 (Leu) residues, we identified a highly selective inhibitor that fills the P2 pocket occupied by the leucine residue sidechain of PF-00835231 and the dimethyl-3-azabicyclo-hexane motif in nirmatrelvir (PF-07321332). This strategy resulted in potent and highly selective Mpro inhibitors without inhibiting essential host cathepsin cysteine or serine proteases. The lead prototype compound 1 (MPro IC50 = 230 ± 18 nM) also inhibits the replication of multiple SARS-CoV-2 variants in vitro, including SARS-CoV-2 variants of concern, and can synergize at lower concentrations with the viral RNA polymerase inhibitor, remdesivir, to inhibit replication. It also reduces SARS-CoV-2 replication in SARS-CoV-2 Omicron-infected Syrian golden hamsters without obvious toxicities, demonstrating in vivo efficacy. This novel lead structure provides the basis for optimization of improved agents targeting evolving SARS-CoV-2 drug resistance that can selectively act on Mpro versus host proteases and are less likely to have off-target effects due to non-specific targeting. Developing inhibitors against the active site of the main protease (Mpro), which is highly conserved across coronaviruses, is expected to impart a higher genetic barrier to evolving SARS-CoV-2 drug resistance. Drugs that selectively inhibit the viral Mpro are less likely to have off-target effects warranting efforts to improve this therapy.
Collapse
Affiliation(s)
- Adi N. R. Poli
- Medicinal Chemistry, The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Ian Tietjen
- HIV-1 Program in the Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Nitesh K. Nandwana
- Medicinal Chemistry, The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Joel Cassel
- The Wistar Cancer Center Molecular Screening, The Wistar Institute, Philadelphia, Pennsylvania, USA
| | | | - Emery T. Register
- HIV-1 Program in the Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Frederick Keeney
- HIV-1 Program in the Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Rajesh Rajaiah
- Department of Pharmacology & Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Atul K. Verma
- Department of Pharmacology & Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Kabita Pandey
- Department of Pharmacology & Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Arpan Acharya
- Department of Pharmacology & Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Siddappa N. Byrareddy
- Department of Pharmacology & Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Luis J. Montaner
- HIV-1 Program in the Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Joseph M. Salvino
- Medicinal Chemistry, The Wistar Institute, Philadelphia, Pennsylvania, USA
- The Wistar Cancer Center Molecular Screening, The Wistar Institute, Philadelphia, Pennsylvania, USA
- Molecular and Cellular Oncogenesis (MCO) Program, The Wistar Institute, Philadelphia, Pennsylvania, USA
| |
Collapse
|
4
|
Dávid CZ, Kúsz N, Agbadua OG, Berkecz R, Kincses A, Spengler G, Hunyadi A, Hohmann J, Vasas A. Phytochemical Investigation of Carex praecox Schreb. and ACE-Inhibitory Activity of Oligomer Stilbenes of the Plant. Molecules 2024; 29:3427. [PMID: 39065005 PMCID: PMC11280411 DOI: 10.3390/molecules29143427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/17/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Phenolic compounds are the main special metabolites of Cyperaceae species from phytochemical, pharmacological, and chemotaxonomical points of view. The present study focused on the isolation, structure determination, and pharmacological investigation of constituents from Carex praecox. Twenty-six compounds, including lignans, stilbenes, flavonoids, megastigmanes, chromenes, and phenylpropanoids, were identified from the methanol extract of the plant. Five of these compounds, namely, carexines A-E, are previously undescribed natural products. All compounds were isolated for the first time from C. praecox. The ACE-inhibitory activity of seven stilbenoid compounds was tested, and (-)-hopeaphenol proved to be the most active (IC50 7.7 ± 0.9 μM). The enzyme-kinetic studies revealed a mixed-type inhibition; therefore, domain-specific studies were also conducted. The in silico docking of (-)-hopeaphenol to the ACE affirmed some favorable interactions. In addition, the antiproliferative and antibacterial effects of some compounds were also evaluated.
Collapse
Affiliation(s)
- Csilla Zsuzsanna Dávid
- Department of Pharmacognosy, University of Szeged, 6720 Szeged, Hungary; (C.Z.D.); (N.K.); (O.G.A.); (A.K.); (A.H.); (J.H.)
| | - Norbert Kúsz
- Department of Pharmacognosy, University of Szeged, 6720 Szeged, Hungary; (C.Z.D.); (N.K.); (O.G.A.); (A.K.); (A.H.); (J.H.)
| | - Orinamhe Godwin Agbadua
- Department of Pharmacognosy, University of Szeged, 6720 Szeged, Hungary; (C.Z.D.); (N.K.); (O.G.A.); (A.K.); (A.H.); (J.H.)
| | - Róbert Berkecz
- Institute of Pharmaceutical Analysis, University of Szeged, 6720 Szeged, Hungary;
| | - Annamária Kincses
- Department of Pharmacognosy, University of Szeged, 6720 Szeged, Hungary; (C.Z.D.); (N.K.); (O.G.A.); (A.K.); (A.H.); (J.H.)
- HUN-REN-USZ Biologically Active Natural Products Research Group, University of Szeged, Eötvös u. 6, 6720 Szeged, Hungary
| | - Gabriella Spengler
- Department of Medical Microbiology, Albert Szent-Györgyi Health Center, Albert Szent-Györgyi Medical School, University of Szeged, 6725 Szeged, Hungary;
| | - Attila Hunyadi
- Department of Pharmacognosy, University of Szeged, 6720 Szeged, Hungary; (C.Z.D.); (N.K.); (O.G.A.); (A.K.); (A.H.); (J.H.)
- HUN-REN-USZ Biologically Active Natural Products Research Group, University of Szeged, Eötvös u. 6, 6720 Szeged, Hungary
| | - Judit Hohmann
- Department of Pharmacognosy, University of Szeged, 6720 Szeged, Hungary; (C.Z.D.); (N.K.); (O.G.A.); (A.K.); (A.H.); (J.H.)
- HUN-REN-USZ Biologically Active Natural Products Research Group, University of Szeged, Eötvös u. 6, 6720 Szeged, Hungary
| | - Andrea Vasas
- Department of Pharmacognosy, University of Szeged, 6720 Szeged, Hungary; (C.Z.D.); (N.K.); (O.G.A.); (A.K.); (A.H.); (J.H.)
- HUN-REN-USZ Biologically Active Natural Products Research Group, University of Szeged, Eötvös u. 6, 6720 Szeged, Hungary
| |
Collapse
|
5
|
Matos-Hernández ML, Samples R, Dyer G, Casimir Montán VM, Morales-Colón CA, Salvino JM, Montaner LJ, Cassel JA, Messick TE, Tietjen I, Caro-Diaz EJE. Metabolomic Analysis and Antiviral Screening of a Marine Algae Library Yield Jobosic Acid (2,5-Dimethyltetradecanoic Acid) as a Selective Inhibitor of SARS-CoV-2. JOURNAL OF NATURAL PRODUCTS 2024; 87:1513-1520. [PMID: 38781491 DOI: 10.1021/acs.jnatprod.3c01071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Current small-molecule-based SARS-CoV-2 treatments have limited global accessibility and pose the risk of inducing viral resistance. Therefore, a marine algae and cyanobacteria extract library was screened for natural products that could inhibit two well-defined and validated COVID-19 drug targets, disruption of the spike protein/ACE-2 interaction and the main protease (Mpro) of SARS-CoV-2. Following initial screening of 86 extracts, we performed an untargeted metabolomic analysis of 16 cyanobacterial extracts. This approach led to the isolation of an unusual saturated fatty acid, jobosic acid (2,5-dimethyltetradecanoic acid, 1). We confirmed that 1 demonstrated selective inhibitory activity toward both viral targets while retaining some activity against the spike-RBD/ACE-2 interaction of the SARS-CoV-2 omicron variant. To initially explore its structure-activity relationship (SAR), the methyl and benzyl ester derivatives of 1 were semisynthetically accessed and demonstrated acute loss of bioactivity in both SARS-CoV-2 biochemical assays. Our efforts have provided copious amounts of a fatty acid natural product that warrants further investigation in terms of SAR, unambiguous determination of its absolute configuration, and understanding of its specific mechanisms of action and binding site toward new therapeutic avenues for SARS-CoV-2 drug development.
Collapse
Affiliation(s)
- Marie L Matos-Hernández
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Puerto Rico-Medical Sciences Campus, San Juan, Puerto Rico 00935, United States
| | - Robert Samples
- Center for Mass Spectrometry, Smith College, Northampton, Massachusetts 01063, United States
| | - Grayce Dyer
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Puerto Rico-Medical Sciences Campus, San Juan, Puerto Rico 00935, United States
| | - Victoria M Casimir Montán
- Department of Chemistry, Natural Sciences College, University of Puerto Rico-Rio Piedras Campus, San Juan, Puerto Rico 00925, United States
| | - Chris A Morales-Colón
- Department of Chemistry, Natural Sciences College, University of Puerto Rico-Rio Piedras Campus, San Juan, Puerto Rico 00925, United States
| | - Joseph M Salvino
- The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Luis J Montaner
- The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Joel A Cassel
- The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Troy E Messick
- The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Ian Tietjen
- The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Eduardo J E Caro-Diaz
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Puerto Rico-Medical Sciences Campus, San Juan, Puerto Rico 00935, United States
| |
Collapse
|
6
|
Shayo MJ, Samwel B, Shadrack DM, Cassel J, Salvino JM, Montaner LJ, Deogratias G, Tietjen I, Kiruri L, Hilonga S, Innocent E. Drug repositioning identifies salvinorin A and deacetylgedunin (DCG) enriched plant extracts as novel inhibitors of Mpro, RBD-ACE2 and TMPRRS2 proteins. RSC Adv 2024; 14:21203-21212. [PMID: 38966817 PMCID: PMC11223729 DOI: 10.1039/d4ra02593h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 06/21/2024] [Indexed: 07/06/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) has spread worldwide with severe health, social, and economic repercussions. Although vaccines have significantly reduced the severity of symptoms and deaths, alternative medications derived from natural products (NPs) are vital to further decrease fatalities, especially in regions with low vaccine uptake. When paired with the latest computational developments, NPs, which have been used to cure illnesses and infections for thousands of years, constitute a renewed resource for drug discovery. In the present report, a combination of computational and in vitro methods reveals the repositioning of NPs and identifies salvinorin A and deacetylgedunin (DCG) as having potential anti-SARS-CoV-2 activities. Salvinorin A was found both in silico and in vitro to inhibit both SARS-CoV-2 spike/host ACE2 protein interactions, consistent with blocking viral cell entry, and well as live virus replication. Plant extracts from Azadirachta indica and Cedrela odorata, which contain high levels of DCG, inhibited viral cell replication by targeting the main protease (Mpro) and/or inhibited viral cell entry by blocking the interaction between spike RBD-ACE2 protein at concentrations lower than salvinorin A. Our findings suggest that salvinorin A represent promising chemical starting points where further optimization may result in effective natural product-derived and potent anti-SARS-CoV-2 inhibitors to supplement vaccine efforts.
Collapse
Affiliation(s)
- Mariana J Shayo
- Department of Biological and Pre-clinical Studies, Institute of Traditional Medicine, Muhimbili University of Health and Allied Sciences P.O.Box 65001 Dar es Salaam Tanzania
| | - Baraka Samwel
- Department of Natural Products, Institute of Traditional Medicines, Muhimbili University of Health and Allied Sciences P.O.Box 65001 Dar es Salaam Tanzania
| | - Daniel M Shadrack
- Department of Chemistry, Faculty of Natural and Applied Sciences, St. John's University of Tanzania P.O.Box 47 Dodoma Tanzania
- School of Life Science and Bio-engineering, Nelson Mandela African Institute of Science and Technology P.O.Box 447 Arusha Tanzania
| | - Joel Cassel
- The Wistar Institute 3601 Spruce Street Philadelphia PA 19104 USA
| | - Joseph M Salvino
- The Wistar Institute 3601 Spruce Street Philadelphia PA 19104 USA
| | - Luis J Montaner
- The Wistar Institute 3601 Spruce Street Philadelphia PA 19104 USA
| | - Geradius Deogratias
- Chemistry Department, College of Natural and Applied Sciences, University of Dar es Salaam P.O.Box 35061 Dar es Salaam Tanzania
| | - Ian Tietjen
- The Wistar Institute 3601 Spruce Street Philadelphia PA 19104 USA
| | - Lucy Kiruri
- Kenyata University, Department of Chemistry P.O.Box 43844-00100 Nairobi Kenya
| | - Samson Hilonga
- Department of Medical Botany, Plant Breeding and Agronomy, Institute of Traditional Medicine, Muhimbili University of Health and Allied Sciences P.O.Box 65001 Dar es Salaam Tanzania
| | - Ester Innocent
- Department of Biological and Pre-clinical Studies, Institute of Traditional Medicine, Muhimbili University of Health and Allied Sciences P.O.Box 65001 Dar es Salaam Tanzania
| |
Collapse
|
7
|
Voget R, Breidenbach J, Claff T, Hingst A, Sylvester K, Steinebach C, Vu LP, Weiße RH, Bartz U, Sträter N, Müller CE, Gütschow M. Development of an active-site titrant for SARS-CoV-2 main protease as an indispensable tool for evaluating enzyme kinetics. Acta Pharm Sin B 2024; 14:2349-2357. [PMID: 38799620 PMCID: PMC11121168 DOI: 10.1016/j.apsb.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/25/2024] [Accepted: 02/27/2024] [Indexed: 05/29/2024] Open
Abstract
A titrant for the SARS-CoV-2 main protease (Mpro) was developed that enables, for the first time, the exact determination of the concentration of the enzymatically active Mpro by active-site titration. The covalent binding mode of the tetrapeptidic titrant was elucidated by the determination of the crystal structure of the enzyme-titrant complex. Four fluorogenic substrates of Mpro, including a prototypical, internally quenched Dabcyl-EDANS peptide, were compared in terms of solubility under typical assay conditions. By exploiting the new titrant, key kinetic parameters for the Mpro-catalyzed cleavage of these substrates were determined.
Collapse
Affiliation(s)
- Rabea Voget
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn 53121, Germany
| | - Julian Breidenbach
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn 53121, Germany
| | - Tobias Claff
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn 53121, Germany
| | - Alexandra Hingst
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn 53121, Germany
| | - Katharina Sylvester
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn 53121, Germany
| | - Christian Steinebach
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn 53121, Germany
| | - Lan Phuong Vu
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn 53121, Germany
| | - Renato H. Weiße
- Institute of Bioanalytical Chemistry, Center for Biotechnology and Biomedicine, Leipzig University, Leipzig 04103, Germany
| | - Ulrike Bartz
- Department of Natural Sciences, University of Applied Sciences Bonn-Rhein-Sieg, Rheinbach 53359, Germany
| | - Norbert Sträter
- Institute of Bioanalytical Chemistry, Center for Biotechnology and Biomedicine, Leipzig University, Leipzig 04103, Germany
| | - Christa E. Müller
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn 53121, Germany
| | - Michael Gütschow
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn 53121, Germany
| |
Collapse
|
8
|
Zhang C, Lum KY, White JM, Duffy S, Lock AM, Avery VM, Davis RA. Chemical and Antiplasmodial Investigations on Eremophila-Derived Alkaloids and Semisynthetic Ether Analogues. JOURNAL OF NATURAL PRODUCTS 2024; 87:849-854. [PMID: 38416027 DOI: 10.1021/acs.jnatprod.3c01072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
Microthecaline A (1), the known antiplasmodial quinoline serrulatane alkaloid from the roots of Eremophila microtheca F. Muell. ex Benth. (Scrophulariaceae), was targeted for isolation and subsequent use in the generation of a semisynthetic ether library. A large-scale extraction and isolation yielded the previously undescribed quinoline serrulatane microthecaline B (2), along with crystalline 1 that enabled the first X-ray crystallographic analysis to be undertaken on this rare alkaloid structure class. The X-ray diffraction analysis of 1 supported the absolute configuration assignment of microthecaline A, which was originally assigned by ECD data analysis. Microthecaline A (1) was converted into 10 new semisynthetic ether derivatives (3-12) using a diverse series of commercially available alkyl halides. Chemical structures of the new serrulatane alkaloid and semisynthetic ether analogues were assigned by spectroscopic and spectrometric analyses. Antiplasmodial evaluations of 1-12 showed that the semisynthetic derivative 5 elicited the most potent activity with an IC50 value of 7.2 μM against Plasmodium falciparum 3D7 (drug-sensitive) strain.
Collapse
Affiliation(s)
- Chen Zhang
- Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Brisbane, QLD 4111, Australia
| | - Kah Yean Lum
- Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Brisbane, QLD 4111, Australia
| | - Jonathan M White
- School of Chemistry and Bio21 Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Sandra Duffy
- Discovery Biology, Centre for Cellular Phenomics, School of Environment and Science, Griffith University, Brisbane, QLD 4111, Australia
| | - Aaron M Lock
- Discovery Biology, Centre for Cellular Phenomics, School of Environment and Science, Griffith University, Brisbane, QLD 4111, Australia
| | - Vicky M Avery
- Discovery Biology, Centre for Cellular Phenomics, School of Environment and Science, Griffith University, Brisbane, QLD 4111, Australia
| | - Rohan A Davis
- Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Brisbane, QLD 4111, Australia
- NatureBank, Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia
| |
Collapse
|
9
|
Shellenberger BM, Basile ON, Cassel J, Olsen MR, Salvino JM, Montaner LJ, Tietjen I, Henry GE. Synthesis, SARS-CoV-2 main protease inhibition, molecular docking and in silico ADME studies of furanochromene-quinoline hydrazone derivatives. Bioorg Med Chem Lett 2024; 102:129679. [PMID: 38423371 DOI: 10.1016/j.bmcl.2024.129679] [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: 12/24/2023] [Revised: 02/06/2024] [Accepted: 02/26/2024] [Indexed: 03/02/2024]
Abstract
Seven furanochromene-quinoline derivatives containing a hydrazone linker were synthesized by condensing a furanochromene hydrazide with quinoline 2-, 3-, 4-, 5-, 6-, and 8-carbaldehydes, including 8-hydroxyquinoline-2-carbaldehye. Structure-activity correlations were investigated to determine the influence of the location of the hydrazone linker on the quinoline unit on SARS-CoV-2 Mpro enzyme inhibition. The 3-, 5-, 6- and 8-substituted derivatives showed moderate inhibition of SARS-CoV-2 Mpro with IC50 values ranging from 16 to 44 μM. Additionally, all of the derivatives showed strong interaction with the SARS-CoV-2 Mpro substrate binding pocket, with docking energy scores ranging from -8.0 to -8.5 kcal/mol. These values are comparable to that of N3 peptide (-8.1 kcal/mol) and more favorable than GC-373 (-7.6 kcal/mol) and ML-188 (-7.5 kcal/mol), all of which are known SARS-CoV-2 Mpro inhibitors. Furthermore, in silico absorption, distribution, metabolism, and excretion (ADME) profiles indicate that the derivatives have good drug-likeness properties. Overall, this study highlights the potential of the furanochromene-quinoline hydrazone scaffold as a SARS-CoV-2 Mpro inhibitor.
Collapse
Affiliation(s)
- Blake M Shellenberger
- Department of Chemistry, Susquehanna University, 514 University Avenue, Selinsgrove, PA 17870, USA
| | - Olivia N Basile
- Department of Chemistry, Susquehanna University, 514 University Avenue, Selinsgrove, PA 17870, USA
| | - Joel Cassel
- The Wistar Institute, Philadelphia, PA 19104, USA
| | - Morgan R Olsen
- Department of Chemistry, Bucknell University, One Dent Drive, Lewisburg, PA 17837, USA
| | | | | | - Ian Tietjen
- The Wistar Institute, Philadelphia, PA 19104, USA.
| | - Geneive E Henry
- Department of Chemistry, Susquehanna University, 514 University Avenue, Selinsgrove, PA 17870, USA.
| |
Collapse
|
10
|
Majoumo-Mbe F, Sangbong NA, Tadjong Tcho A, Namba-Nzanguim CT, Simoben CV, Eni DB, Alhaji Isa M, Poli ANR, Cassel J, Salvino JM, Montaner LJ, Tietjen I, Ntie-Kang F. 5-chloro-3-(2-(2,4-dinitrophenyl) hydrazono)indolin-2-one: synthesis, characterization, biochemical and computational screening against SARS-CoV-2. CHEMICKE ZVESTI 2024; 78:3431-3441. [PMID: 38685970 PMCID: PMC11055700 DOI: 10.1007/s11696-023-03274-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 12/04/2023] [Indexed: 05/02/2024]
Abstract
Chemical prototypes with broad-spectrum antiviral activity are important toward developing new therapies that can act on both existing and emerging viruses. Binding of the SARS-CoV-2 spike protein to the host angiotensin-converting enzyme 2 (ACE2) receptor is required for cellular entry of SARS-CoV-2. Toward identifying new chemical leads that can disrupt this interaction, including in the presence of SARS-CoV-2 adaptive mutations found in variants like omicron that can circumvent vaccine, immune, and therapeutic antibody responses, we synthesized 5-chloro-3-(2-(2,4-dinitrophenyl)hydrazono)indolin-2-one (H2L) from the condensation reaction of 5-chloroisatin and 2,4-dinitrophenylhydrazine in good yield. H2L was characterised by elemental and spectral (IR, electronic, Mass) analyses. The NMR spectrum of H2L indicated a keto-enol tautomerism, with the keto form being more abundant in solution. H2L was found to selectively interfere with binding of the SARS-CoV-2 spike receptor-binding domain (RBD) to the host angiotensin-converting enzyme 2 receptor with a 50% inhibitory concentration (IC50) of 0.26 μM, compared to an unrelated PD-1/PD-L1 ligand-receptor-binding pair with an IC50 of 2.06 μM in vitro (Selectivity index = 7.9). Molecular docking studies revealed that the synthesized ligand preferentially binds within the ACE2 receptor-binding site in a region distinct from where spike mutations in SARS-CoV-2 variants occur. Consistent with these models, H2L was able to disrupt ACE2 interactions with the RBDs from beta, delta, lambda, and omicron variants with similar activities. These studies indicate that H2L-derived compounds are potential inhibitors of multiple SARS-CoV-2 variants, including those capable of circumventing vaccine and immune responses. Supplementary Information The online version contains supplementary material available at 10.1007/s11696-023-03274-5.
Collapse
Affiliation(s)
- Felicite Majoumo-Mbe
- Department of Chemistry, Faculty of Science, University of Buea, P. O. Box 63, Buea, Cameroon
| | - Neba Abongwa Sangbong
- Department of Chemistry, Faculty of Science, University of Buea, P. O. Box 63, Buea, Cameroon
| | - Alain Tadjong Tcho
- Department of Chemistry, Faculty of Science, University of Buea, P. O. Box 63, Buea, Cameroon
| | - Cyril T. Namba-Nzanguim
- Department of Chemistry, Faculty of Science, University of Buea, P. O. Box 63, Buea, Cameroon
- Center for Drug Discovery, Faculty of Science, University of Buea, P. O. Box 63, Buea, Cameroon
| | - Conrad V. Simoben
- Center for Drug Discovery, Faculty of Science, University of Buea, P. O. Box 63, Buea, Cameroon
| | - Donatus B. Eni
- Department of Chemistry, Faculty of Science, University of Buea, P. O. Box 63, Buea, Cameroon
- Center for Drug Discovery, Faculty of Science, University of Buea, P. O. Box 63, Buea, Cameroon
| | - Mustafa Alhaji Isa
- Department of Microbiology, Faculty of Sciences, University of Maiduguri, PMB 1069, Maiduguri, Borno State Nigeria
| | | | - Joel Cassel
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104 USA
| | - Joseph M. Salvino
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104 USA
| | - Luis J. Montaner
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104 USA
| | - Ian Tietjen
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104 USA
| | - Fidele Ntie-Kang
- Department of Chemistry, Faculty of Science, University of Buea, P. O. Box 63, Buea, Cameroon
- Center for Drug Discovery, Faculty of Science, University of Buea, P. O. Box 63, Buea, Cameroon
- Institute of Pharmacy, Martin-Luther University Halle-Wittenberg, Kurt-Mothes-Strasse 3, 06120 Halle (Saale), Germany
| |
Collapse
|
11
|
Eni DB, Cassel J, Namba-Nzanguim CT, Simoben CV, Tietjen I, Akunuri R, Salvino JM, Ntie-Kang F. Design, synthesis, and biochemical and computational screening of novel oxindole derivatives as inhibitors of Aurora A kinase and SARS-CoV-2 spike/host ACE2 interaction. Med Chem Res 2024; 33:620-634. [PMID: 38646411 PMCID: PMC11024012 DOI: 10.1007/s00044-024-03201-7] [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: 12/30/2023] [Accepted: 02/09/2024] [Indexed: 04/23/2024]
Abstract
Isatin (indol-2,3-dione), a secondary metabolite of tryptophan, has been used as the core structure to design several compounds that have been tested and identified as potent inhibitors of apoptosis, potential antitumor agents, anticonvulsants, and antiviral agents. In this work, several analogs of isatin hybrids have been synthesized and characterized, and their activities were established as inhibitors of both Aurora A kinase and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike/host angiotensin-converting enzyme II (ACE2) interactions. Amongst the synthesized isatin hybrids, compounds 6a, 6f, 6g, and 6m exhibited Aurora A kinase inhibitory activities (with IC50 values < 5 μ M), with GScore values of -7.9, -7.6, -8.2 and -7.7 kcal/mol, respectively. Compounds 6g and 6i showed activities in blocking SARS-CoV-2 spike/ACE2 binding (with IC50 values in the range < 30 μ M), with GScore values of -6.4 and -6.6 kcal/mol, respectively. Compounds 6f, 6g, and 6i were both capable of inhibiting spike/ACE2 binding and blocking Aurora A kinase. Pharmacophore profiling indicated that compound 6g tightly fits Aurora A kinase and SARS-CoV-2 pharmacophores, while 6d fits SARS-CoV-2 and 6l fits Aurora A kinase pharmacophore. This work is a proof of concept that some existing cancer drugs may possess antiviral properties. Molecular modeling showed that the active compound for each protein adopted different binding modes, hence interacting with a different set of amino acid residues in the binding site. The weaker activities against spike/ACE2 could be explained by the small sizes of the ligands that fail to address the important interactions for binding to the ACE2 receptor site.
Collapse
Affiliation(s)
- Donatus B. Eni
- Center for Drug Discovery, Faculty of Science, University of Buea, Buea, Cameroon
- Department of Chemistry, Faculty of Science, University of Buea, Buea, Cameroon
| | | | - Cyril T. Namba-Nzanguim
- Center for Drug Discovery, Faculty of Science, University of Buea, Buea, Cameroon
- Department of Chemistry, Faculty of Science, University of Buea, Buea, Cameroon
| | - Conrad V. Simoben
- Center for Drug Discovery, Faculty of Science, University of Buea, Buea, Cameroon
| | | | | | | | - Fidele Ntie-Kang
- Center for Drug Discovery, Faculty of Science, University of Buea, Buea, Cameroon
- Department of Chemistry, Faculty of Science, University of Buea, Buea, Cameroon
- Institute of Pharmacy, Martin-Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
12
|
Ramage KS, Lock A, White JM, Ekins MG, Kiefel MJ, Avery VM, Davis RA. Semisynthesis and Cytotoxic Evaluation of an Ether Analogue Library Based on a Polyhalogenated Diphenyl Ether Scaffold Isolated from a Lamellodysidea Sponge. Mar Drugs 2024; 22:33. [PMID: 38248658 PMCID: PMC10817568 DOI: 10.3390/md22010033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 12/20/2023] [Accepted: 12/23/2023] [Indexed: 01/23/2024] Open
Abstract
The known oxygenated polyhalogenated diphenyl ether, 2-(2',4'-dibromophenoxy)-3,5-dibromophenol (1), with previously reported activity in multiple cytotoxicity assays was isolated from the sponge Lamellodysidea sp. and proved to be an amenable scaffold for semisynthetic library generation. The phenol group of 1 was targeted to generate 12 ether analogues in low-to-excellent yields, and the new library was fully characterized by NMR, UV, and MS analyses. The chemical structures for 2, 8, and 9 were additionally determined via single-crystal X-ray diffraction analysis. All natural and semisynthetic compounds were evaluated for their ability to inhibit the growth of DU145, LNCaP, MCF-7, and MDA-MB-231 cancer cell lines. Compound 3 was shown to have near-equivalent activity compared to scaffold 1 in two in vitro assays, and the activity of the compounds with an additional benzyl ring appeared to be reliant on the presence and position of additional halogens.
Collapse
Affiliation(s)
- Kelsey S. Ramage
- Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Brisbane, QLD 4111, Australia; (K.S.R.); (M.G.E.)
| | - Aaron Lock
- Discovery Biology, School of Environment and Science, Griffith University, Brisbane, QLD 4111, Australia; (A.L.); (V.M.A.)
| | - Jonathan M. White
- School of Chemistry and Bio21 Institute, The University of Melbourne, Parkville, VIC 3010, Australia;
| | - Merrick G. Ekins
- Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Brisbane, QLD 4111, Australia; (K.S.R.); (M.G.E.)
- Queensland Museum, South Brisbane, QLD 4101, Australia
| | - Milton J. Kiefel
- Institute for Glycomics, School of Environment and Science, Griffith University, Gold Coast, QLD 4222, Australia;
| | - Vicky M. Avery
- Discovery Biology, School of Environment and Science, Griffith University, Brisbane, QLD 4111, Australia; (A.L.); (V.M.A.)
| | - Rohan A. Davis
- Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Brisbane, QLD 4111, Australia; (K.S.R.); (M.G.E.)
- NatureBank, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia
| |
Collapse
|
13
|
Wang Z, Song XQ, Xu W, Lei S, Zhang H, Yang L. Stand Up to Stand Out: Natural Dietary Polyphenols Curcumin, Resveratrol, and Gossypol as Potential Therapeutic Candidates against Severe Acute Respiratory Syndrome Coronavirus 2 Infection. Nutrients 2023; 15:3885. [PMID: 37764669 PMCID: PMC10535599 DOI: 10.3390/nu15183885] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
The COVID-19 pandemic has stimulated collaborative drug discovery efforts in academia and the industry with the aim of developing therapies and vaccines that target SARS-CoV-2. Several novel therapies have been approved and deployed in the last three years. However, their clinical application has revealed limitations due to the rapid emergence of viral variants. Therefore, the development of next-generation SARS-CoV-2 therapeutic agents with a high potency and safety profile remains a high priority for global health. Increasing awareness of the "back to nature" approach for improving human health has prompted renewed interest in natural products, especially dietary polyphenols, as an additional therapeutic strategy to treat SARS-CoV-2 patients, owing to its good safety profile, exceptional nutritional value, health-promoting benefits (including potential antiviral properties), affordability, and availability. Herein, we describe the biological properties and pleiotropic molecular mechanisms of dietary polyphenols curcumin, resveratrol, and gossypol as inhibitors against SARS-CoV-2 and its variants as observed in in vitro and in vivo studies. Based on the advantages and disadvantages of dietary polyphenols and to obtain maximal benefits, several strategies such as nanotechnology (e.g., curcumin-incorporated nanofibrous membranes with antibacterial-antiviral ability), lead optimization (e.g., a methylated analog of curcumin), combination therapies (e.g., a specific combination of plant extracts and micronutrients), and broad-spectrum activities (e.g., gossypol broadly inhibits coronaviruses) have also been emphasized as positive factors in the facilitation of anti-SARS-CoV-2 drug development to support effective long-term pandemic management and control.
Collapse
Affiliation(s)
- Zhonglei Wang
- Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China; (W.X.); (S.L.); (H.Z.)
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus, Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China
| | - Xian-qing Song
- General Surgery Department, Baoan Central Hospital, Affiliated Baoan Central Hospital of Guangdong Medical University, Shenzhen 518000, China
| | - Wenjing Xu
- Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China; (W.X.); (S.L.); (H.Z.)
| | - Shizeng Lei
- Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China; (W.X.); (S.L.); (H.Z.)
| | - Hao Zhang
- Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, China; (W.X.); (S.L.); (H.Z.)
| | - Liyan Yang
- School of Physics and Physical Engineering, Qufu Normal University, Qufu 273165, China
| |
Collapse
|
14
|
Davis RA, Cervin G, Beattie KD, Rali T, Fauchon M, Hellio C, Bodin Åkerlund L, Pavia H, Svenson J. Evaluation of natural resveratrol multimers as marine antifoulants. BIOFOULING 2023; 39:775-784. [PMID: 37822262 DOI: 10.1080/08927014.2023.2263374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 09/18/2023] [Indexed: 10/13/2023]
Abstract
In the current study we investigate the antifouling potential of three polyphenolic resveratrol multimers (-)-hopeaphenol, vaticanol B and vatalbinoside A, isolated from two species of Anisoptera found in the Papua New Guinean rainforest. The compounds were evaluated against the growth and settlement of eight marine microfoulers and against the settlement and metamorphosis of Amphibalanus improvisus barnacle cyprids. The two isomeric compounds (-)-hopeaphenol and vaticanol B displayed a high inhibitory potential against the cyprid larvae metamorphosis at 2.8 and 1.1 μM. (-)-Hopeaphenol was also shown to be a strong inhibitor of both microalgal and bacterial adhesion at submicromolar concentrations with low toxicity. Resveratrol displayed a lower antifouling activity compared to the multimers and had higher off target toxicity against MCR-5 fibroblasts. This study illustrates the potential of natural products as a valuable source for the discovery of novel antifouling leads with low toxicity.
Collapse
Affiliation(s)
- Rohan A Davis
- Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Brisbane, QLD, Australia
| | - Gunnar Cervin
- Department of Marine Sciences - Tjärnö, University of Gothenburg, Strömstad, Sweden
| | - Karren D Beattie
- Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Brisbane, QLD, Australia
| | - Topul Rali
- School of Natural and Physical Sciences, The University of Papua New Guinea, Port Moresby, Papua New Guinea
| | - Marilyne Fauchon
- Laboratoire des Sciences de l'Environnement MARin (LEMAR), CNRS, IRD, IFREMER, University of Brest, Brest, France
| | - Claire Hellio
- Laboratoire des Sciences de l'Environnement MARin (LEMAR), CNRS, IRD, IFREMER, University of Brest, Brest, France
| | - Lovisa Bodin Åkerlund
- Department of Biological Function, RISE Research Institutes of Sweden, Borås, Sweden
| | - Henrik Pavia
- Department of Marine Sciences - Tjärnö, University of Gothenburg, Strömstad, Sweden
| | | |
Collapse
|
15
|
Romani A, Sergi D, Zauli E, Voltan R, Lodi G, Vaccarezza M, Caruso L, Previati M, Zauli G. Nutrients, herbal bioactive derivatives and commensal microbiota as tools to lower the risk of SARS-CoV-2 infection. Front Nutr 2023; 10:1152254. [PMID: 37324739 PMCID: PMC10267353 DOI: 10.3389/fnut.2023.1152254] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/15/2023] [Indexed: 06/17/2023] Open
Abstract
The SARS-CoV-2 outbreak has infected a vast population across the world, causing more than 664 million cases and 6.7 million deaths by January 2023. Vaccination has been effective in reducing the most critical aftermath of this infection, but some issues are still present regarding re-infection prevention, effectiveness against variants, vaccine hesitancy and worldwide accessibility. Moreover, although several old and new antiviral drugs have been tested, we still lack robust and specific treatment modalities. It appears of utmost importance, facing this continuously growing pandemic, to focus on alternative practices grounded on firm scientific bases. In this article, we aim to outline a rigorous scientific background and propose complementary nutritional tools useful toward containment, and ultimately control, of SARS-CoV-2 infection. In particular, we review the mechanisms of viral entry and discuss the role of polyunsaturated fatty acids derived from α-linolenic acid and other nutrients in preventing the interaction of SARS-CoV-2 with its entry gateways. In a similar way, we analyze in detail the role of herbal-derived pharmacological compounds and specific microbial strains or microbial-derived polypeptides in the prevention of SARS-CoV-2 entry. In addition, we highlight the role of probiotics, nutrients and herbal-derived compounds in stimulating the immunity response.
Collapse
Affiliation(s)
- Arianna Romani
- Department of Environmental and Prevention Sciences and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Domenico Sergi
- Department of Translational Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Enrico Zauli
- Department of Translational Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Rebecca Voltan
- Department of Environmental and Prevention Sciences and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Giada Lodi
- Department of Environmental and Prevention Sciences and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Mauro Vaccarezza
- Curtin Medical School & Curtin Health Innovation Research Institute (CHIRI), Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Lorenzo Caruso
- Department of Environmental and Prevention Sciences and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Maurizio Previati
- Department of Translational Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Giorgio Zauli
- Research Department, King Khaled Eye Specialistic Hospital, Riyadh, Saudi Arabia
| |
Collapse
|
16
|
Egbewande FA, Schwartz BD, Duffy S, Avery VM, Davis RA. Synthesis and Antimalarial Evaluation of Halogenated Analogues of Thiaplakortone A. Mar Drugs 2023; 21:md21050317. [PMID: 37233511 DOI: 10.3390/md21050317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/12/2023] [Accepted: 05/17/2023] [Indexed: 05/27/2023] Open
Abstract
The incorporation of bromine, iodine or fluorine into the tricyclic core structure of thiaplakortone A (1), a potent antimalarial marine natural product, is reported. Although yields were low, it was possible to synthesise a small nine-membered library using the previously synthesised Boc-protected thiaplakortone A (2) as a scaffold for late-stage functionalisation. The new thiaplakortone A analogues (3-11) were generated using N-bromosuccinimide, N-iodosuccinimide or a Diversinate™ reagent. The chemical structures of all new analogues were fully characterised by 1D/2D NMR, UV, IR and MS data analyses. All compounds were evaluated for their antimalarial activity against Plasmodium falciparum 3D7 (drug-sensitive) and Dd2 (drug-resistant) strains. Incorporation of halogens at positions 2 and 7 of the thiaplakortone A scaffold was shown to reduce antimalarial activity compared to the natural product. Of the new compounds, the mono-brominated analogue (compound 5) displayed the best antimalarial activity with IC50 values of 0.559 and 0.058 μM against P. falciparum 3D7 and Dd2, respectively, with minimal toxicity against a human cell line (HEK293) observed at 80 μM. Of note, the majority of the halogenated compounds showed greater efficacy against the P. falciparum drug-resistant strain.
Collapse
Affiliation(s)
- Folake A Egbewande
- Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Nathan, QLD 4111, Australia
| | - Brett D Schwartz
- Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Nathan, QLD 4111, Australia
| | - Sandra Duffy
- Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Nathan, QLD 4111, Australia
- Discovery Biology, Centre for Cellular Phenomics, Griffith University, Nathan, QLD 4111, Australia
| | - Vicky M Avery
- Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Nathan, QLD 4111, Australia
- Discovery Biology, Centre for Cellular Phenomics, Griffith University, Nathan, QLD 4111, Australia
| | - Rohan A Davis
- Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Nathan, QLD 4111, Australia
- NatureBank, Griffith University, Nathan, QLD 4111, Australia
| |
Collapse
|
17
|
Tietjen I, Schonhofer C, Sciorillo A, Naidu ME, Haq Z, Kannan T, Kossenkov AV, Rivera-Ortiz J, Mounzer K, Hart C, Gyampoh K, Yuan Z, Beattie KD, Rali T, Shuda McGuire K, Davis RA, Montaner LJ. The Natural Stilbenoid (-)-Hopeaphenol Inhibits HIV Transcription by Targeting Both PKC and NF-κB Signaling and Cyclin-Dependent Kinase 9. Antimicrob Agents Chemother 2023; 67:e0160022. [PMID: 36975214 PMCID: PMC10112218 DOI: 10.1128/aac.01600-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/27/2023] [Indexed: 03/29/2023] Open
Abstract
Despite effective combination antiretroviral therapy (cART), people living with HIV (PLWH) continue to harbor replication-competent and transcriptionally active virus in infected cells, which in turn can lead to ongoing viral antigen production, chronic inflammation, and increased risk of age-related comorbidities. To identify new agents that may inhibit postintegration HIV beyond cART, we screened a library of 512 pure compounds derived from natural products and identified (-)-hopeaphenol as an inhibitor of HIV postintegration transcription at low to submicromolar concentrations without cytotoxicity. Using a combination of global RNA sequencing, plasmid-based reporter assays, and enzyme activity studies, we document that hopeaphenol inhibits protein kinase C (PKC)- and downstream NF-κB-dependent HIV transcription as well as a subset of PKC-dependent T-cell activation markers, including interleukin-2 (IL-2) cytokine and CD25 and HLA-DRB1 RNA production. In contrast, it does not substantially inhibit the early PKC-mediated T-cell activation marker CD69 production of IL-6 or NF-κB signaling induced by tumor necrosis factor alpha (TNF-α). We further show that hopeaphenol can inhibit cyclin-dependent kinase 9 (CDK9) enzymatic activity required for HIV transcription. Finally, it inhibits HIV replication in peripheral blood mononuclear cells (PBMCs) infected in vitro and dampens viral reactivation in CD4+ cells from PLWH. Our study identifies hopeaphenol as a novel inhibitor that targets a subset of PKC-mediated T-cell activation pathways in addition to CDK9 to block HIV expression. Hopeaphenol-based therapies could complement current antiretroviral therapy otherwise not targeting cell-associated HIV RNA and residual antigen production in PLWH.
Collapse
Affiliation(s)
- Ian Tietjen
- The Wistar Institute, Philadelphia, Pennsylvania, USA
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Cole Schonhofer
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | | | - Maya E. Naidu
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Zahra Haq
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | | | | | | | - Karam Mounzer
- Jonathan Lax Immune Disorders Treatment Center, Philadelphia Field Initiating Group for HIV-1 Trials, Philadelphia, Pennsylvania, USA
| | - Colin Hart
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Kwasi Gyampoh
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Zhe Yuan
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Karren D. Beattie
- Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Brisbane, Queensland, Australia
| | - Topul Rali
- School of Natural and Physical Sciences, The University of Papua New Guinea, Port Moresby, Papua New Guinea
| | | | - Rohan A. Davis
- Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Brisbane, Queensland, Australia
| | | |
Collapse
|
18
|
Williams DE, Cassel J, Zhu JL, Yang JX, de Voogd NJ, Matainaho T, Salvino JM, Wang YA, Montaner LJ, Tietjen I, Andersen RJ. Thorectidiol A Isolated from the Marine Sponge Dactylospongia elegans Disrupts Interactions of the SARS-CoV-2 Spike Receptor Binding Domain with the Host ACE2 Receptor. JOURNAL OF NATURAL PRODUCTS 2023; 86:582-588. [PMID: 36657039 PMCID: PMC9885524 DOI: 10.1021/acs.jnatprod.2c01030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Indexed: 06/15/2023]
Abstract
Thorectidiols isolated from the marine sponge Dactylospongia elegans (family Thorectidae, order Dictyoceratida) collected in Papua New Guinea are a family of symmetrical and unsymmetrical dimeric biphenyl meroterpenoid stereoisomers presumed to be products of oxidative phenol coupling of a co-occurring racemic monomer, thorectidol (3). One member of the family, thorectidiol A (1), has been isolated in its natural form, and its structure has been elucidated by analysis of NMR, MS, and ECD data. Acetylation of the sponge extract facilitated isolation of additional thorectidiol diacetate stereoisomers and the isolation of the racemic monomer thorectidol acetate (6). Racemic thorectidiol A (1) showed selective inhibition of the SARS-CoV-2 spike receptor binding domain (RBD) interaction with the host ACE2 receptor with an IC50 = 1.0 ± 0.7 μM.
Collapse
Affiliation(s)
- David E Williams
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, B.C. Canada, V6T 1Z1
- Department of Earth, Ocean & Atmospheric Sciences, University of British Columbia, 2207 Main Mall, Vancouver, B.C. Canada, V6T 1Z4
| | - Joel Cassel
- The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Jin-Lin Zhu
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, B.C. Canada, V6T 1Z1
| | - Jian-Xiong Yang
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, B.C. Canada, V6T 1Z1
| | - Nicole J de Voogd
- Naturalis Biodiversity Center, P.O. Box 9517, 2300 RA Leiden, The Netherlands
| | - Teatulohi Matainaho
- University of Papua New Guinea, University National Capital District, 134, Papua New Guinea
| | - Joseph M Salvino
- The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Yan Alexander Wang
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, B.C. Canada, V6T 1Z1
| | - Luis J Montaner
- The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Ian Tietjen
- The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Raymond J Andersen
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, B.C. Canada, V6T 1Z1
- Department of Earth, Ocean & Atmospheric Sciences, University of British Columbia, 2207 Main Mall, Vancouver, B.C. Canada, V6T 1Z4
| |
Collapse
|
19
|
Yang Z, Cai X, Ye Q, Zhao Y, Li X, Zhang S, Zhang L. High-Throughput Screening for the Potential Inhibitors of SARS-CoV-2 with Essential Dynamic Behavior. Curr Drug Targets 2023; 24:532-545. [PMID: 36876836 DOI: 10.2174/1389450124666230306141725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 11/09/2022] [Accepted: 01/11/2023] [Indexed: 03/07/2023]
Abstract
Global health security has been challenged by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) pandemic. Due to the lengthy process of generating vaccinations, it is vital to reposition currently available drugs in order to relieve anti-epidemic tensions and accelerate the development of therapies for Coronavirus Disease 2019 (COVID-19), the public threat caused by SARS-CoV-2. High throughput screening techniques have established their roles in the evaluation of already available medications and the search for novel potential agents with desirable chemical space and more cost-effectiveness. Here, we present the architectural aspects of highthroughput screening for SARS-CoV-2 inhibitors, especially three generations of virtual screening methodologies with structural dynamics: ligand-based screening, receptor-based screening, and machine learning (ML)-based scoring functions (SFs). By outlining the benefits and drawbacks, we hope that researchers will be motivated to adopt these methods in the development of novel anti- SARS-CoV-2 agents.
Collapse
Affiliation(s)
- Zhiwei Yang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, Xi'an710049, China
| | - Xinhui Cai
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, Xi'an710049, China
| | - Qiushi Ye
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, Xi'an710049, China
| | - Yizhen Zhao
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, Xi'an710049, China
| | - Xuhua Li
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, Xi'an710049, China
| | - Shengli Zhang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, Xi'an710049, China
| | - Lei Zhang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, Xi'an710049, China
| |
Collapse
|
20
|
Brahmi F, Vejux A, Ghzaiel I, Ksila M, Zarrouk A, Ghrairi T, Essadek S, Mandard S, Leoni V, Poli G, Vervandier-Fasseur D, Kharoubi O, El Midaoui A, Atanasov AG, Meziane S, Latruffe N, Nasser B, Bouhaouala-Zahar B, Masmoudi-Kouki O, Madani K, Boulekbache-Makhlouf L, Lizard G. Role of Diet and Nutrients in SARS-CoV-2 Infection: Incidence on Oxidative Stress, Inflammatory Status and Viral Production. Nutrients 2022; 14:2194. [PMID: 35683996 PMCID: PMC9182601 DOI: 10.3390/nu14112194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 05/10/2022] [Accepted: 05/17/2022] [Indexed: 11/17/2022] Open
Abstract
Coronavirus illness (COVID-19) is an infectious pathology generated by intense severe respiratory syndrome coronavirus 2 (SARS-CoV-2). This infectious disease has emerged in 2019. The COVID-19-associated pandemic has considerably affected the way of life and the economy in the world. It is consequently crucial to find solutions allowing remedying or alleviating the effects of this infectious disease. Natural products have been in perpetual application from immemorial time given that they are attested to be efficient towards several illnesses without major side effects. Various studies have shown that plant extracts or purified molecules have a promising inhibiting impact towards coronavirus. In addition, it is substantial to understand the characteristics, susceptibility and impact of diet on patients infected with COVID-19. In this review, we recapitulate the influence of extracts or pure molecules from medicinal plants on COVID-19. We approach the possibilities of plant treatment/co-treatment and feeding applied to COVID-19. We also show coronavirus susceptibility and complications associated with nutrient deficiencies and then discuss the major food groups efficient on COVID-19 pathogenesis. Then, we covered emerging technologies using plant-based SARS-CoV-2 vaccine. We conclude by giving nutrient and plants curative therapy recommendations which are of potential interest in the COVID-19 infection and could pave the way for pharmacological treatments or co-treatments of COVID-19.
Collapse
Affiliation(s)
- Fatiha Brahmi
- Laboratory Biomathématique, Biochimie, Biophysique et Scientométrie, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia 06000, Algeria; (K.M.); (L.B.-M.)
| | - Anne Vejux
- Department of Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism, University of Bourgogne Franche-Comte, 21000 Dijon, France; (A.V.); (I.G.); (M.K.); (S.E.); (N.L.)
| | - Imen Ghzaiel
- Department of Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism, University of Bourgogne Franche-Comte, 21000 Dijon, France; (A.V.); (I.G.); (M.K.); (S.E.); (N.L.)
- Lab-NAFS ‘Nutrition-Functional Food & Vascular Health’, Faculty of Medicine, LR12ES05, University Monastir, Monastir 5000, Tunisia;
| | - Mohamed Ksila
- Department of Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism, University of Bourgogne Franche-Comte, 21000 Dijon, France; (A.V.); (I.G.); (M.K.); (S.E.); (N.L.)
- Laboratory of Neurophysiology, Cellular Physiopathology and Valorisation of Biomolecules, (LR18ES03), Department of Biology, Faculty of Sciences, University Tunis El Manar, Tunis 2092, Tunisia; (T.G.); (O.M.-K.)
| | - Amira Zarrouk
- Lab-NAFS ‘Nutrition-Functional Food & Vascular Health’, Faculty of Medicine, LR12ES05, University Monastir, Monastir 5000, Tunisia;
- Laboratory of Biochemistry, Faculty of Medicine, University of Sousse, Sousse 4000, Tunisia
| | - Taoufik Ghrairi
- Laboratory of Neurophysiology, Cellular Physiopathology and Valorisation of Biomolecules, (LR18ES03), Department of Biology, Faculty of Sciences, University Tunis El Manar, Tunis 2092, Tunisia; (T.G.); (O.M.-K.)
| | - Soukena Essadek
- Department of Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism, University of Bourgogne Franche-Comte, 21000 Dijon, France; (A.V.); (I.G.); (M.K.); (S.E.); (N.L.)
- Laboratory Neuroscience and Biochemistry, University of Hassan 1st, Settat 26000, Morocco;
| | - Stéphane Mandard
- Lipness Team and LipSTIC LabEx, UFR Sciences de Santé, INSERM/University of Bourgogne Franche-Comté LNC UMR1231, 21000 Dijon, France;
| | - Valerio Leoni
- Department of Laboratory Medicine, University of Milano-Bicocca, Azienda Socio Sanitaria Territoriale Brianza ASST-Brianza, Desio Hospital, Via Mazzini 1, 20833 Desio, Italy;
| | - Giuseppe Poli
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Turin, 10043 Orbassano (Turin), Italy;
| | - Dominique Vervandier-Fasseur
- Team OCS, Institute of Molecular Chemistry of University of Burgundy (ICMUB UMR CNRS 6302), University of Bourgogne Franche-Comté, 21000 Dijon, France;
| | - Omar Kharoubi
- Laboratory of Experimental Biotoxicology, Biodepollution and Phytoremediation, Faculty of Life and Natural Sciences, University Oran 1 ABB, Oran 31000, Algeria;
| | - Adil El Midaoui
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada;
- Faculty of Sciences and Techniques, Moulay Ismail University of Meknes, Errachidia 52000, Morocco
| | - Atanas G. Atanasov
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzebiec, 05-552 Magdalenka, Poland;
| | - Smail Meziane
- Institut Européen des Antioxydants, 1b Rue Victor de Lespinats, 54230 Neuves-Maison, France;
| | - Norbert Latruffe
- Department of Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism, University of Bourgogne Franche-Comte, 21000 Dijon, France; (A.V.); (I.G.); (M.K.); (S.E.); (N.L.)
| | - Boubker Nasser
- Laboratory Neuroscience and Biochemistry, University of Hassan 1st, Settat 26000, Morocco;
| | - Balkiss Bouhaouala-Zahar
- Laboratory of Biomolecules, Venoms and Theranostic Applications, Pasteur Institute of Tunis, University of Tunis El Manar, Tunis 1002, Tunisia;
| | - Olfa Masmoudi-Kouki
- Laboratory of Neurophysiology, Cellular Physiopathology and Valorisation of Biomolecules, (LR18ES03), Department of Biology, Faculty of Sciences, University Tunis El Manar, Tunis 2092, Tunisia; (T.G.); (O.M.-K.)
| | - Khodir Madani
- Laboratory Biomathématique, Biochimie, Biophysique et Scientométrie, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia 06000, Algeria; (K.M.); (L.B.-M.)
- Centre de Recherche en Technologie des Industries Agroalimentaires, Route de Targua Ouzemour, Bejaia 06000, Algeria
| | - Lila Boulekbache-Makhlouf
- Laboratory Biomathématique, Biochimie, Biophysique et Scientométrie, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia 06000, Algeria; (K.M.); (L.B.-M.)
| | - Gérard Lizard
- Department of Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism, University of Bourgogne Franche-Comte, 21000 Dijon, France; (A.V.); (I.G.); (M.K.); (S.E.); (N.L.)
| |
Collapse
|
21
|
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
|
22
|
Olas B. The Antioxidant, Anti-Platelet and Anti-Coagulant Properties of Phenolic Compounds, Associated with Modulation of Hemostasis and Cardiovascular Disease, and Their Possible Effect on COVID-19. Nutrients 2022; 14:nu14071390. [PMID: 35406002 PMCID: PMC9003312 DOI: 10.3390/nu14071390] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 01/14/2023] Open
Abstract
Patients affected by coronavirus disease 2019 (COVID-19) demonstrate a range of hemostasis dysfunctions, such as coagulation dysfunction and changes in blood platelet function, this being a major cause of death. These complications may also be associated with oxidative stress. Recently, various papers, including some reviews, have suggested that the use of dietary bioactive compounds, including phenolic compounds, may play a significant role in the treatment of COVID-19. However, while some phenolic compounds, such as curcumin, resveratrol, myricetin and scutellarian, have been found to have antiviral effects against COVID-19, recommendations regarding the use of such compounds to prevent or reduce the risk of CVDs during COVID-19 infection remain tentative. The present mini-review examines the antioxidant, anti-platelet and anticoagulant and antiviral activities of selected phenolic compounds and the possible implications for their use in treating CVDs associated with COVID-19. This review also examines whether these phenolic compounds can be promising agents in the modulation of hemostasis and CVDs during COVID-19. While their properties have been well documented in various in vitro and in vivo studies, particularly their positive role in the prophylaxis and treatment of CVDs, less is known regarding their prophylactic potential against CVDs during COVID-19, and no credible evidence exists for their efficiency in humans or animals. In such cases, no in vitro or in vivo studies are available. Therefore, it cannot be unequivocally stated whether treatment with these phenolic compounds offers benefits against CVDs in patients with COVID-19.
Collapse
Affiliation(s)
- Beata Olas
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/3, 90-236 Lodz, Poland
| |
Collapse
|
23
|
Urda L, Kreuter MH, Drewe J, Boonen G, Butterweck V, Klimkait T. The Petasites hybridus CO 2 Extract (Ze 339) Blocks SARS-CoV-2 Replication In Vitro. Viruses 2022; 14:v14010106. [PMID: 35062310 PMCID: PMC8781559 DOI: 10.3390/v14010106] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19), caused by a novel coronavirus (SARS-CoV-2), has spread worldwide, affecting over 250 million people and resulting in over five million deaths. Antivirals that are effective are still limited. The antiviral activities of the Petasites hybdridus CO2 extract Ze 339 were previously reported. Thus, to assess the anti-SARS-CoV-2 activity of Ze 339 as well as isopetasin and neopetasin as major active compounds, a CPE and plaque reduction assay in Vero E6 cells was used for viral output. Antiviral effects were tested using the original virus (Wuhan) and the Delta variant of SARS-CoV-2. The antiviral drug remdesivir was used as control. Pre-treatment with Ze 339 in SARS-CoV-2-infected Vero E6 cells with either virus variant significantly inhibited virus replication with IC50 values of 0.10 and 0.40 μg/mL, respectively. The IC50 values obtained for isopetasin ranged between 0.37 and 0.88 μM for both virus variants, and that of remdesivir ranged between 1.53 and 2.37 μM. In conclusion, Ze 339 as well as the petasins potently inhibited SARS-CoV-2 replication in vitro of the Wuhan and Delta variants. Since time is of essence in finding effective treatments, clinical studies will have to demonstrate if Ze339 can become a therapeutic option to treat SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Lorena Urda
- Department Biomedicine, University of Basel, Petersplatz 10, 4051 Basel, Switzerland
| | | | - Jürgen Drewe
- Medical Department, Max Zeller & Söhne AG, Seeblickstrasse 4, 8590 Romanshorn, Switzerland
| | - Georg Boonen
- Medical Department, Max Zeller & Söhne AG, Seeblickstrasse 4, 8590 Romanshorn, Switzerland
| | - Veronika Butterweck
- Medical Department, Max Zeller & Söhne AG, Seeblickstrasse 4, 8590 Romanshorn, Switzerland
| | - Thomas Klimkait
- Department Biomedicine, University of Basel, Petersplatz 10, 4051 Basel, Switzerland
| |
Collapse
|