Anti-COVID-19 Potential of Ellagic Acid and Polyphenols of Punica granatum L.
Abstract
:1. Introduction
2. COVID-19 Infection Pathology
3. Antioxidant and Anti-Inflammatory Activity of Pomegranate Extract
4. Antioxidant and Anti-Inflammatory Effects of EA and Its Metabolites
5. Antiviral Activity of Pomegranate Polyphenolic Extracts and Ellagic Acid
Compound Tested | Viral Target | Molecular Mechanism | References |
---|---|---|---|
ellagic acid | influenza A | synergistic effect on antioxidant defenses with oseltamivir and isoprinosine | [21] |
pomegranate polyphenol extract, punicalagin | influenza A influenza B | synergistic effect on viral proliferation inhibition with oseltamivir | [22] |
pomegranate leaf ethanolic extract | HSV-2 Zika | reduces viral proliferation in cells | [24] |
pomegranate peel extract and fruit juice | HCV | inhibition of NS3/4A protease activity | [26] |
pomegranate peel extract, punicalin | SARS-CoV-2 | binds to SARS-CoV-2 S-glycoprotein and inhibits binding to ACE2 | [91] |
Rhodiola rosea extract | Ebola | inhibits viral entry in cells | [20] |
punicalagin and Zn(II) | SARS-CoV-2 | inhibition of 3CL protease, synergistic effect with Zn(II) | [92] |
chebulagic acid, punicalagin | SARS-CoV-2 | non-competitive inhibition of 3CL protease | [93] |
HSV-1 | inhibits viral entry in cells and cell-to-cell spread via viral glycoprotein and host glucosaminoglycans interaction | [25] | |
HCMV HCV DENV MV RSV | inhibits viral attachment to cells | [94] | |
geraniin | SARS-CoV-2 | binds SARS-CoV-2 S-glycoprotein receptor binding domain | [95] |
corilagin | SARS-CoV-2 | binds to SARS-CoV-2 S-glycoprotein and inhibits binding to ACE2 | [96] |
SARS-CoV-2 | inhibits activity of RNA-dependent RNA polymerase nsp12 | [97] | |
ellagic acid | Zika | hypothetical interaction with cell surface to prevent viral infection | [24] |
HIV-1 | blocks viral integrase but not protease | [98] | |
HRV2 HRV3 | reduces viral proliferation in cells | [23] | |
HBV | blocks HBeAg secretion from cells | [99] | |
Ebola | inhibits viral entry in cells | [20] |
6. Binding of Ellagitannins and Ellagic Acid to Components of SARS-CoV-2 and Human Host
7. Considerations for the In Vivo Administration of EA-Containing Extracts
8. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- De Leon, V.N.O.; Manzano, J.A.H.; Pilapil, D.Y.H.; Fernandez, R.A.T.; Ching, J.K.A.R.; Quimque, M.T.J.; Agbay, J.C.M.; Notarte, K.I.R.; Macabeo, A.P.G. Anti-HIV reverse transcriptase plant polyphenolic natural products with in silico inhibitory properties on seven non-structural proteins vital in SARS-CoV-2 pathogenesis. J. Genet. Eng. Biotechnol. 2021, 19, 104. [Google Scholar] [CrossRef] [PubMed]
- Fakhri, S.; Piri, S.; Majnooni, M.B.; Farzaei, M.H.; Echeverría, J. Targeting Neurological Manifestations of Coronaviruses by Candidate Phytochemicals: A Mechanistic Approach. Front. Pharm. 2020, 11, 621099. [Google Scholar] [CrossRef] [PubMed]
- Santhi, V.P.; Masilamani, P.; Sriramavaratharajan, V.; Murugan, R.; Gurav, S.S.; Sarasu, V.P.; Parthiban, S.; Ayyanar, M. Therapeutic potential of phytoconstituents of edible fruits in combating emerging viral infections. J. Food Biochem. 2021, 45, e13851. [Google Scholar] [CrossRef] [PubMed]
- David, A.B.; Diamant, E.; Dor, E.; Barnea, A.; Natan, N.; Levin, L.; Chapman, S.; Mimran, L.C.; Epstein, E.; Zichel, R.; et al. Identification of SARS-CoV-2 Receptor Binding Inhibitors by In Vitro Screening of Drug Libraries. Molecules 2021, 26, 3213. [Google Scholar] [CrossRef] [PubMed]
- Puttaswamy, H.; Gowtham, H.G.; Ojha, M.D.; Yadav, A.; Choudhir, G.; Raguraman, V.; Kongkham, B.; Selvaraju, K.; Shareef, S.; Gehlot, P.; et al. In silico studies evidenced the role of structurally diverse plant secondary metabolites in reducing SARS-CoV-2 pathogenesis. Sci. Rep. 2020, 10, 20584. [Google Scholar] [CrossRef]
- Ali, S.; Alam, M.; Khatoon, F.; Fatima, U.; Elasbali, A.M.; Adnan, M.; Islam, A.; Hassan, M.I.; Snoussi, M.; De Feo, V. Natural products can be used in therapeutic management of COVID-19: Probable mechanistic insights. Biomed. Pharm. 2022, 147, 112658. [Google Scholar] [CrossRef]
- Wu, S.; Tian, L. Diverse Phytochemicals and Bioactivities in the Ancient Fruit and Modern Functional Food Pomegranate (Punica granatum). Molecules 2017, 22, 1606. [Google Scholar] [CrossRef]
- Lim, S.K.; Othman, R.; Yusof, R.; Heh, C.H. Rational drug discovery: Ellagic acid as a potent dual-target inhibitor against hepatitis C virus genotype 3 (HCV G3) NS3 enzymes. Chem. Biol. Drug Des. 2021, 97, 28–40. [Google Scholar] [CrossRef]
- AbouAitah, K.; Allayh, A.K.; Wojnarowicz, J.; Shaker, Y.M.; Swiderska-Sroda, A.; Lojkowski, W. Nanoformulation Composed of Ellagic Acid and Functionalized Zinc Oxide Nanoparticles Inactivates DNA and RNA Viruses. Pharmaceutics 2021, 13, 2174. [Google Scholar] [CrossRef]
- Arunkumar, J.; Rajarajan, S. Study on antiviral activities, drug-likeness and molecular docking of bioactive compounds of Punica granatum L. to Herpes simplex virus—2 (HSV-2). Microb. Pathog. 2018, 118, 301–309. [Google Scholar] [CrossRef]
- Živković, I.; Šavikin, K.; Živković, J.; Zdunić, G.; Janković, T.; Lazić, D.; Radin, D. Antiviral Effects of Pomegranate Peel Extracts on Human Norovirus in Food Models and Simulated Gastrointestinal Fluids. Plant Foods Hum. Nutr. 2021, 76, 203–209. [Google Scholar] [CrossRef] [PubMed]
- Moradi, M.-T.; Karimi, A.; Shahrani, M.; Hashemi, L.; Ghaffari-Goosheh, M.-S. Anti-Influenza Virus Activity and Phenolic Content of Pomegranate (Punica granatum L.) Peel Extract and Fractions. Avicenna J. Med. Biotechnol. 2019, 11, 285–291. [Google Scholar] [PubMed]
- Yang, Y.; Xiu, J.; Zhang, L.; Qin, C.; Liu, J. Antiviral activity of punicalagin toward human enterovirus 71 in vitro and in vivo. Phytomedicine 2012, 20, 67–70. [Google Scholar] [CrossRef] [PubMed]
- Kang, E.H.; Kown, T.Y.; Oh, G.T.; Park, W.F.; Park, S.I.; Park, S.K.; Lee, Y.I. The flavonoid ellagic acid from a medicinal herb inhibits host immune tolerance induced by the hepatitis B virus-e antigen. Antivir. Res. 2006, 72, 100–106. [Google Scholar] [CrossRef]
- Houston, D.M.J.; Robins, B.; Bugert, J.J.; Denyer, S.P.; Heard, C.M. In vitro permeation and biological activity of punicalagin and zinc (II) across skin and mucous membranes prone to Herpes simplex virus infection. Eur. J. Pharm. Sci. 2017, 96, 99–106. [Google Scholar] [CrossRef]
- Karimi, A.; Moradi, M.T.; Rabiei, M.; Alidadi, S. In vitro anti-adenoviral activities of ethanol extract, fractions, and main phenolic compounds of pomegranate (Punica granatum L.) peel. Antivir. Chem. Chemother. 2020, 28. [Google Scholar] [CrossRef]
- Salles, T.S.; Meneses, M.D.F.; Caldas, L.A.; Sá-Guimarães, T.E.; de Oliveira, D.M.; Ventura, J.A.; Azevedo, R.C.; Kuster, R.M.; Soares, M.R.; Ferreira, D.F. Virucidal and antiviral activities of pomegranate (Punica granatum) extract against the mosquito-borne Mayaro virus. Parasites Vectors 2021, 14, 433. [Google Scholar] [CrossRef]
- Li, P.; Du, R.; Chen, Z.; Wang, Y.; Zhan, P.; Liu, X.; Kang, D.; Chen, Z.; Zhao, X.; Wang, L.; et al. Punicalagin is a neuraminidase inhibitor of influenza viruses. J. Med. Virol. 2021, 93, 3465–3472. [Google Scholar] [CrossRef]
- Serkedjieva, J.; Stefanova, T.; Krumova, E.; Tancheva, L. Protective effect of polyphenol-rich extract on acute lung injury in influenza virus infected mice. Biotechnol. Biotechnol. Equip. 2009, 23, 1355–1359. [Google Scholar] [CrossRef]
- Cui, Q.; Du, R.; Anantpadma, M.; Schafer, A.; Hou, L.; Tian, J.; Davey, R.A.; Cheng, H.; Rong, L. Identification of ellagic acid from plant rhodiola rosea l. as an anti-ebola virus entry inhibitor. Viruses 2018, 10, 152. [Google Scholar] [CrossRef]
- Pavlova, E.L.; Simeonova, L.S.; Gegova, G.A. Combined efficacy of oseltamivir, isoprinosine and ellagic acid in influenza A(H3N2)-infected mice. Biomed. Pharmacother. 2018, 98, 29–35. [Google Scholar] [CrossRef] [PubMed]
- Haidari, M.; Ali, M.; Ward Casscells, S.; Madjid, M. Pomegranate (Punica granatum) purified polyphenol extract inhibits influenza virus and has a synergistic effect with oseltamivir. Phytomedicine 2009, 16, 1127–1136. [Google Scholar] [CrossRef] [PubMed]
- Park, S.W.; Kwon, M.J.; Yoo, J.Y.; Choi, H.J.; Ahn, Y.J. Antiviral activity and possible mode of action of ellagic acid identified in Lagerstroemia speciosa leaves toward human rhinoviruses. BMC Complement. Altern. Med. 2014, 14, 171. [Google Scholar] [CrossRef] [PubMed]
- Acquadro, S.; Civra, A.; Cagliero, C.; Marengo, A.; Rittà, M.; Francese, R.; Sanna, C.; Bertea, C.; Sgorbini, B.; Lembo, D.; et al. Punica granatum Leaf Ethanolic Extract and Ellagic Acid as Inhibitors of Zika Virus Infection. Planta Med. 2020, 86, 1363–1374. [Google Scholar] [CrossRef]
- Lin, L.T.; Chen, T.Y.; Chung, C.Y.; Noyce, R.S.; Grindley, T.B.; McCormick, C.; Lin, T.C.; Wang, G.H.; Lin, C.C.; Richardson, C.D. Hydrolyzable Tannins (Chebulagic Acid and Punicalagin) Target Viral Glycoprotein-Glycosaminoglycan Interactions to Inhibit Herpes Simplex Virus 1 Entry and Cell-to-Cell Spread. J. Virol. 2011, 85, 4386–4398. [Google Scholar] [CrossRef]
- Reddy, B.U.; Mullick, R.; Kumar, A.; Sudha, G.; Srinivasan, N.; Das, S. Small molecule inhibitors of HCV replication from Pomegranate. Sci. Rep. 2014, 4, 5411. [Google Scholar] [CrossRef]
- Abarova, S.; Tancheva, L.; Nikolov, R.; Serkedjieva, J.; Pavlova, E.; Bramanti, A.; Nicoletti, F.; Tzvetkov, N.T. Preventive effect of a polyphenol-rich extract from Geranium sanguineum L. On hepatic drug metabolism in influenza infected mice. Sci. Pharm. 2020, 88, 45. [Google Scholar] [CrossRef]
- Tancheva, L. Drug metabolism and oxidative stress during Influenza Virus Infection. In Experimental Approaches for Antioxidant Protection; Publisher Eagle: Silistra, Bulgaria, 2019; p. 111. [Google Scholar]
- Serkedjieva, J.; Krumova, E.; Stefanova, T.; Tancheva, L. Pulmonary protection of a plant polyphenol extract in influenza virus-infected mice. J. Infect. 2009, 59, S426. [Google Scholar] [CrossRef]
- Encheva, E.; Tancheva, L.; Abarova, S.; Serkedjieva, J.; Ivantcheva, S. Modulation of hepatic drug metabolism in influenza infected mice. Preventive effect of polyphenols isolated from Geranium sanguineum L. Toxicol. Lett. 2010, 196, 723. [Google Scholar] [CrossRef]
- Moreno Fernández-Ayala, D.J.; Navas, P.; López-Lluch, G. Age-related mitochondrial dysfunction as a key factor in COVID-19 disease. Exp. Gerontol. 2020, 142, 111147. [Google Scholar] [CrossRef]
- Raman, B.; Cassar, M.P.; Tunnicliffe, E.M.; Filippini, N.; Griffanti, L.; Alfaro-Almagro, F.; Okell, T.; Sheerin, F.; Xie, C.; Mahmod, M.; et al. Medium-term effects of SARS-CoV-2 infection on multiple vital organs, exercise capacity, cognition, quality of life and mental health, post-hospital discharge. EClinicalMedicine 2021, 31, 100683. [Google Scholar] [CrossRef] [PubMed]
- Peng, R.; Wu, L.A.; Wang, Q.; Qi, J.; Gao, G.F. Cell entry by SARS-CoV-2. Trends Biochem. Sci. 2021, 46, 848–860. [Google Scholar] [CrossRef] [PubMed]
- Basile, M.S.; Cavalli, E.; McCubrey, J.; Hernández-Bello, J.; Muñoz-Valle, J.F.; Fagone, P.; Nicoletti, F. The PI3K/Akt/mTOR pathway: A potential pharmacological target in COVID-19. Drug Discov. Today 2022, 27, 848–856. [Google Scholar] [CrossRef] [PubMed]
- Rahman, M.R.; Islam, T.; Shahjaman, M.; Islam, M.R.; Lombardo, S.D.; Bramanti, P.; Ciurleo, R.; Bramanti, A.; Tchorbanov, A.; Fisicaro, F.; et al. Discovering common pathogenetic processes between COVID-19 and diabetes mellitus by differential gene expression pattern analysis. Brief. Bioinform. 2021, 22. [Google Scholar] [CrossRef] [PubMed]
- Cavalli, E.; Petralia, M.C.; Basile, M.S.; Bramanti, A.; Bramanti, P.; Nicoletti, F.; Spandidos, D.A.; Shoenfeld, Y.; Fagone, P. Transcriptomic analysis of COVID-19 lungs and bronchoalveolar lavage fluid samples reveals predominant B cell activation responses to infection. Int. J. Mol. Med. 2020, 46, 1266–1273. [Google Scholar] [CrossRef] [PubMed]
- Fagone, P.; Ciurleo, R.; Lombardo, S.D.; Iacobello, C.; Palermo, C.I.; Shoenfeld, Y.; Bendtzen, K.; Bramanti, P.; Nicoletti, F. Transcriptional landscape of SARS-CoV-2 infection dismantles pathogenic pathways activated by the virus, proposes unique sex-specific differences and predicts tailored therapeutic strategies. Autoimmun. Rev. 2020, 19, 102571. [Google Scholar] [CrossRef]
- Tancheva, L.; Petralia, M.C.; Miteva, S.; Dragomanova, S.; Solak, A.; Kalfin, R.; Lazarova, M.; Yarkov, D.; Ciurleo, R.; Cavalli, E.; et al. Emerging neurological and psychobiological aspects of COVID-19 infection. Brain Sci. 2020, 10, 852. [Google Scholar] [CrossRef]
- Harrison, A.G.; Lin, T.; Wang, P. Mechanisms of SARS-CoV-2 Transmission and Pathogenesis. Trends Immunol. 2020, 41, 1100–1115. [Google Scholar] [CrossRef]
- Cavalli, E.; Bramanti, A.; Ciurleo, R.; Tchorbanov, A.I.; Giordano, A.; Fagone, P.; Belizna, C.; Bramanti, P.; Shoenfeld, Y.; Nicoletti, F. Entangling COVID-19 associated thrombosis into a secondary antiphospholipid antibody syndrome: Diagnostic and therapeutic perspectives (Review). Int. J. Mol. Med. 2020, 46, 903–912. [Google Scholar] [CrossRef]
- Laforge, M.; Elbim, C.; Frère, C.; Hémadi, M.; Massaad, C.; Nuss, P.; Benoliel, J.J.; Becker, C. Tissue damage from neutrophil-induced oxidative stress in COVID-19. Nat. Rev. Immunol. 2020, 20, 515–516. [Google Scholar] [CrossRef]
- Schönrich, G.; Raftery, M.J.; Samstag, Y. Devilishly radical NETwork in COVID-19: Oxidative stress, neutrophil extracellular traps (NETs), and T cell suppression. Adv. Biol. Regul. 2020, 77, 100741. [Google Scholar] [CrossRef] [PubMed]
- Fratta Pasini, A.M.; Stranieri, C.; Cominacini, L.; Mozzini, C. Potential Role of Antioxidant and Anti-Inflammatory Therapies to Prevent Severe SARS-CoV-2 Complications. Antioxidants 2021, 10, 272. [Google Scholar] [CrossRef] [PubMed]
- Lin, C.W.; Lin, K.H.; Hsieh, T.H.; Shiu, S.Y.; Li, J.Y. Severe acute respiratory syndrome coronavirus 3C-like protease-induced apoptosis. FEMS Immunol. Med. Microbiol. 2006, 46, 375–380. [Google Scholar] [CrossRef] [PubMed]
- Hameedi, M.A.T.; Prates, E.; Garvin, M.R.; Mathews, I.I.; Amos, B.K.; Demerdash, O.; Bechthold, M.; Iyer, M.; Rahighi, S.; Kneller, D.W.; et al. Structural and functional characterization of NEMO cleavage by SARS-CoV-2 3CLpro. Nat. Commun. 2022, 13, 5285. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Guo, M.; Tian, X.; Liu, C.; Wang, X.; Yang, X.; Wu, P.; Xiao, Z.; Qu, Y.; Yin, Y.; et al. Virus-host interactome and proteomic survey of PMBCs from COVID-19 patients reveal potential virulence factors influencing SARS-CoV-2 pathogenesis. bioRxiv 2020, 2, 99–112.e7. [Google Scholar] [CrossRef]
- Khomich, O.A.; Kochetkov, S.N.; Bartosch, B.; Ivanov, A.V. Redox biology of respiratory viral infections. Viruses 2018, 10, 392. [Google Scholar] [CrossRef]
- Camini, F.C.; da Silva Caetano, C.C.; Almeida, L.T.; de Brito Magalhães, C.L. Implications of oxidative stress on viral pathogenesis. Arch. Virol. 2017, 162, 907–917. [Google Scholar] [CrossRef]
- Beck, M.A.; Handy, J.; Levander, O.A. The Role of Oxidative Stress in Viral Infections. Ann. N. Y. Acad. Sci. 2000, 917, 906–912. [Google Scholar] [CrossRef]
- Checconi, P.; De Angelis, M.; Marcocci, M.E.; Fraternale, A.; Magnani, M.; Palamara, A.T.; Nencioni, L. Redox-modulating agents in the treatment of viral infections. Int. J. Mol. Sci. 2020, 21, 4084. [Google Scholar] [CrossRef]
- Crump, K.E.; Langston, P.K.; Rajkarnikar, S.; Grayson, J.M. Antioxidant treatment regulates the humoral immune response during acute viral infection. J. Virol. 2013, 87, 2577–2586. [Google Scholar] [CrossRef]
- Mendonca, P.; Soliman, K.F.A. Flavonoids Activation of the Transcription Factor Nrf2 as a Hypothesis Approach for the Prevention and Modulation of SARS-CoV-2 Infection Severity. Antioxidants 2020, 9, 659. [Google Scholar] [CrossRef] [PubMed]
- Shi, C.S.; Nabar, N.R.; Huang, N.N.; Kehrl, J.H. SARS-Coronavirus Open Reading Frame-8b triggers intracellular stress pathways and activates NLRP3 inflammasomes. Cell Death Discov. 2019, 5, 101. [Google Scholar] [CrossRef] [PubMed]
- D’Amico, D.; Andreux, P.A.; Valdés, P.; Singh, A.; Rinsch, C.; Auwerx, J. Impact of the Natural Compound Urolithin A on Health, Disease, and Aging. Trends Mol. Med. 2021, 27, 687–699. [Google Scholar] [CrossRef]
- Ganji, R.; Reddy, P.H. Impact of COVID-19 on Mitochondrial-Based Immunity in Aging and Age-Related Diseases. Front. Aging Neurosci. 2021, 12, 614650. [Google Scholar] [CrossRef] [PubMed]
- Pastorino, S.; Baldassari, S.; Ailuno, G.; Zuccari, G.; Drava, G.; Petretto, A.; Cossu, V.; Marini, C.; Alfei, S.; Florio, T.; et al. Two Novel PET Radiopharmaceuticals for Endothelial Vascular Cell Adhesion Molecule-1 (VCAM-1) Targeting. Pharmaceutics 2021, 13, 1025. [Google Scholar] [CrossRef] [PubMed]
- Wang, T.; Rumbaugh, J.A.; Nath, A. Viruses and the Brain: From Inflammation to Dementia. Clin. Sci. 2006, 110, 393–407. [Google Scholar] [CrossRef] [PubMed]
- Savoia, C.; Schiffrin, E.L. Inflammation in Hypertension. Curr. Opin. Nephrol. Hypertens. 2006, 15, 152–158. [Google Scholar] [CrossRef]
- Benchagra, L.; Berrougui, H.; Islam, M.O.; Ramchoun, M.; Boulbaroud, S.; Hajjaji, A.; Fulop, T.; Ferretti, G.; Khalil, A. Antioxidant effect of moroccan pomegranate (Punica granatum L. sefri variety) extracts rich in punicalagin against the oxidative stress process. Foods 2021, 10, 2219. [Google Scholar] [CrossRef]
- Tzulker, R.; Glazer, I.; Bar-Ilan, I.; Holland, D.; Aviram, M.; Amir, R. Antioxidant activity, polyphenol content, and related compounds in different fruit juices and homogenates prepared from 29 different pomegranate accessions. J. Agric. Food Chem. 2007, 55, 9559–9570. [Google Scholar] [CrossRef]
- Braidy, N.; Selvaraju, S.; Essa, M.M.; Vaishnav, R.; Al-Adawi, S.; Al-Asmi, A.; Al-Senawi, H.; Abd Alrahman Alobaidy, A.; Lakhtakia, R.; Guillemin, G.J. Neuroprotective effects of a variety of pomegranate juice extracts against MPTP-induced cytotoxicity and oxidative stress in human primary neurons. Oxid. Med. Cell Longev. 2013, 2013, 685909. [Google Scholar] [CrossRef]
- Saeed, M.; Naveed, M.; BiBi, J.; Kamboh, A.A.; Arain, M.A.; Shah, Q.A.; Alagawany, M.; El-Hack, M.E.A.; Abdel-Latif, M.A.; Yatoo, M.I.; et al. The Promising Pharmacological Effects and Therapeutic/Medicinal Applications of Punica Granatum L. (Pomegranate) as a Functional Food in Humans and Animals. Recent Pat. Inflamm. Allergy Drug Discov. 2018, 12, 24–38. [Google Scholar] [CrossRef] [PubMed]
- DaSilva, N.A.; Nahar, P.P.; Ma, H.; Eid, A.; Wei, Z.; Meschwitz, S.; Zawia, N.H.; Slitt, A.L.; Seeram, N.P. Pomegranate ellagitannin-gut microbial-derived metabolites, urolithins, inhibit neuroinflammation in vitro. Nutr. Neurosci. 2019, 22, 185–195. [Google Scholar] [CrossRef] [PubMed]
- De Oliveira, J.F.F.; Garreto, D.V.; Da Silva, M.C.P.; Fortes, T.S.; De Oliveira, R.B.; Nascimento, F.R.F.; Da Costa, F.B.; Grisotto, M.A.G.; Nicolete, R. Therapeutic potential of biodegradable microparticles containing Punica granatum L. (pomegranate) in murine model of asthma. Inflamm. Res. 2013, 62, 971–980. [Google Scholar] [CrossRef]
- Guo, Y.J.; Zhao, L.; Li, X.F.; Mei, Y.W.; Zhang, S.L.; Tao, J.Y.; Zhou, Y.; Dong, J.H. Effect of Corilagin on anti-inflammation in HSV-1 encephalitis and HSV-1 infected microglias. Eur. J. Pharmacol. 2010, 635, 79–86. [Google Scholar] [CrossRef] [PubMed]
- Husari, A.; Khayat, A.; Bitar, H.; Hashem, Y.; Rizkallah, A.; Zaatari, G.; El Sabban, M. Antioxidant activity of pomegranate juice reduces acute lung injury secondary to hyperoxia in an animal model. BMC Res. Notes 2014, 7, 664. [Google Scholar] [CrossRef] [PubMed]
- Peng, J.; Wei, D.; Fu, Z.; Li, D.; Tan, Y.; Xu, T.; Zhou, J.; Zhang, T. Punicalagin Ameliorates Lipopolysaccharide-Induced Acute Respiratory Distress Syndrome in Mice. Inflammation 2015, 38, 493–499. [Google Scholar] [CrossRef] [PubMed]
- Shen, P.X.; Li, X.; Deng, S.Y.; Zhao, L.; Zhang, Y.Y.; Deng, X.; Han, B.; Yu, J.; Li, Y.; Wang, Z.Z.; et al. Urolithin A ameliorates experimental autoimmune encephalomyelitis by targeting aryl hydrocarbon receptor. EBioMedicine 2021, 64, 103227. [Google Scholar] [CrossRef]
- Abdelazeem, K.N.M.; Kalo, M.Z.; Beer-Hammer, S.; Lang, F. The gut microbiota metabolite urolithin A inhibits NF-κB activation in LPS stimulated BMDMs. Sci. Rep. 2021, 11, 7117. [Google Scholar] [CrossRef]
- Du, L.; Li, J.; Zhang, X.; Wang, L.; Zhang, W.; Yang, M.; Hou, C. Pomegranate peel polyphenols inhibits inflammation in LPS-induced RAW264.7 macrophages via the suppression of TLR4/NF-κB pathway activation. Food Nutr. Res. 2019, 63, 3392. [Google Scholar] [CrossRef]
- Bobowska, A.; Granica, S.; Filipek, A.; Melzig, M.F.; Moeslinger, T.; Zentek, J.; Kruk, A.; Piwowarski, J.P. Comparative studies of urolithins and their phase II metabolites on macrophage and neutrophil functions. Eur. J. Nutr. 2021, 60, 1957–1972. [Google Scholar] [CrossRef]
- Busto, R.; Serna, J.; Perianes-Cachero, A.; Quintana-Portillo, R.; García-Seisdedos, D.; Canfrán-Duque, A.; Paino, C.L.; Lerma, M.; Casado, M.E.; Martín-Hidalgo, A.; et al. Ellagic acid protects from myelin-associated sphingolipid loss in experimental autoimmune encephalomyelitis. Biochim. Biophys. Acta. Mol. Cell Biol. Lipids 2018, 1863, 958–967. [Google Scholar] [CrossRef] [PubMed]
- Esselun, C.; Theyssen, E.; Eckert, G.P. Effects of Urolithin A on Mitochondrial Parameters in a Cellular Model of Early Alzheimer Disease. Int. J. Mol. Sci. 2021, 22, 8333. [Google Scholar] [CrossRef] [PubMed]
- Alfei, S.; Marengo, B.; Zuccari, G. Oxidative stress, antioxidant capabilities, and bioavailability: Ellagic acid or urolithins? Antioxidants 2020, 9, 707. [Google Scholar] [CrossRef] [PubMed]
- Cortés-Martín, A.; Selma, M.V.; Tomás-Barberán, F.A.; González-Sarrías, A.; Espín, J.C. Where to Look into the Puzzle of Polyphenols and Health? The Postbiotics and Gut Microbiota Associated with Human Metabotypes. Mol. Nutr. Food Res. 2020, 64, e1900952. [Google Scholar] [CrossRef]
- Cortés-Martín, A.; García-Villalba, R.; González-Sarrías, A.; Romo-Vaquero, M.; Loria-Kohen, V.; Ramírez-De-Molina, A.; Tomás-Barberán, F.A.; Selma, M.V.; Espín, J.C. The gut microbiota urolithin metabotypes revisited: The human metabolism of ellagic acid is mainly determined by aging. Food Funct. 2018, 9, 4100–4106. [Google Scholar] [CrossRef]
- Selma, M.V.; González-Sarrías, A.; Salas-Salvadó, J.; Andrés-Lacueva, C.; Alasalvar, C.; Örem, A.; Tomás-Barberán, F.A.; Espín, J.C. The gut microbiota metabolism of pomegranate or walnut ellagitannins yields two urolithin-metabotypes that correlate with cardiometabolic risk biomarkers: Comparison between normoweight, overweight-obesity and metabolic syndrome. Clin. Nutr. 2018, 37, 897–905. [Google Scholar] [CrossRef]
- Li, Y.; Yu, Z.; Zhao, D.; Han, D. Corilagin alleviates hypertrophic scars via inhibiting the transforming growth factor (TGF)-β/Smad signal pathway. Life Sci. 2021, 277, 119483. [Google Scholar] [CrossRef]
- Wei, Y.; Kim, T.J.; Peng, D.H.; Duan, D.; Gibbons, D.L.; Yamauchi, M.; Jackson, J.R.; Le Saux, C.J.; Calhoun, C.; Peters, J.; et al. Fibroblast-specific inhibition of TGF-β1 signaling attenuates lung and tumor fibrosis. J. Clin. Investig. 2017, 127, 3675–3688. [Google Scholar] [CrossRef]
- Papoutsi, Z.; Kassi, E.; Chinou, I.; Halabalaki, M.; Skaltsounis, L.A.; Moutsatsou, P. Walnut extract (Juglans regia L.) and its component ellagic acid exhibit anti-inflammatory activity in human aorta endothelial cells and osteoblastic activity in the cell line KS483. Br. J. Nutr. 2008, 99, 715–722. [Google Scholar] [CrossRef]
- Ding, Y.; Zhang, B.; Zhou, K.; Chen, M.; Wang, M.; Jia, Y.; Song, Y.; Li, Y.; Wen, A. Dietary ellagic acid improves oxidant-induced endothelial dysfunction and atherosclerosis: Role of Nrf2 activation. Int. J. Cardiol. 2014, 175, 508–514. [Google Scholar] [CrossRef]
- Anderson, M.; Turchi, J. Targeting of Non-Structural Protein 9 as a Novel Therapeutic Target for the Treatment of SARS-CoV-2. Proc. IMPRS 2020, 3, 24502. [Google Scholar] [CrossRef] [PubMed]
- Mertens-Talcott, S.U.; Jilma-Stohlawetz, P.; Rios, J.; Hingorani, L.; Derendorf, H. Absorption, metabolism, and antioxidant effects of pomegranate (Punica granatum L.) polyphenols after ingestion of a standardized extract in healthy human volunteers. J. Agric. Food Chem. 2006, 54, 8956–8961. [Google Scholar] [CrossRef] [PubMed]
- Yuan, T.; Ma, H.; Liu, W.; Niesen, D.B.; Shah, N.; Crews, R.; Rose, K.N.; Vattem, D.A.; Seeram, N.P. Pomegranate’s Neuroprotective Effects against Alzheimer’s Disease Are Mediated by Urolithins, Its Ellagitannin-Gut Microbial Derived Metabolites. ACS Chem Neurosci. 2016, 7, 26–33. [Google Scholar] [CrossRef] [PubMed]
- Huang, T.H.; Yang, Q.; Harada, M.; Li, G.Q.; Yamahara, J.; Roufogalis, B.D.; Li, Y. Pomegranate flower extract diminishes cardiac fibrosis in Zucker diabetic fatty rats: Modulation of cardiac endothelin-1 and nuclear factor-kappaB pathways. J. Cardiovasc. Pharmacol. 2005, 46, 856–862. [Google Scholar] [CrossRef]
- Tancheva, L.P.; Lazarova, M.I.; Alexandrova, A.V.; Dragomanova, S.T.; Nicoletti, F.; Tzvetanova, E.R.; Hodzhev, Y.K.; Kalfin, R.E.; Miteva, S.A.; Mazzon, E.; et al. Neuroprotective Mechanisms of Three Natural Antioxidants on a Rat Model of Parkinson’s Disease: A Comparative Study. Antioxidants 2020, 9, 49. [Google Scholar] [CrossRef]
- Bachoual, R.; Talmoudi, W.; Boussetta, T.; Braut, F.; El-Benna, J. An aqueous pomegranate peel extract inhibits neutrophil myeloperoxidase in vitro and attenuates lung inflammation in mice. Food Chem. Toxicol. 2011, 49, 1224–1228. [Google Scholar] [CrossRef]
- Lee, S.I.; Kim, B.S.; Kim, K.S.; Lee, S.; Shin, K.S.; Lim, J.S. Immune-suppressive activity of punicalagin via inhibition of NFAT activation. Biochem. Biophys. Res. Commun. 2008, 371, 799–803. [Google Scholar] [CrossRef]
- Romier, B.; Van De Walle, J.; During, A.; Larondelle, Y.; Schneider, Y.J. Modulation of signalling nuclear factor-κB activation pathway by polyphenols in human intestinal Caco-2 cells. Br. J. Nutr. 2008, 100, 542–551. [Google Scholar] [CrossRef]
- Singh, R.; Chandrashekharappa, S.; Bodduluri, S.R.; Baby, B.V.; Hegde, B.; Kotla, N.G.; Hiwale, A.A.; Saiyed, T.; Patel, P.; Vijay-Kumar, M.; et al. Enhancement of the gut barrier integrity by a microbial metabolite through the Nrf2 pathway. Nat. Commun. 2019, 10, 89. [Google Scholar] [CrossRef]
- Suručić, R.; Travar, M.; Petković, M.; Tubić, B.; Stojiljković, M.P.; Grabež, M.; Šavikin, K.; Zdunić, G.; Škrbić, R. Pomegranate peel extract polyphenols attenuate the SARS-CoV-2 S-glycoprotein binding ability to ACE2 Receptor: In silico and in vitro studies. Bioorganic. Chem. 2021, 114, 105145. [Google Scholar] [CrossRef]
- Saadh, M.J.; Almaaytah, A.M.; Alaraj, M.; Dababneh, M.F.; Sa’adeh, I.; Aldalaen, S.M.; Kharshid, A.M.; Alboghdadly, A.; Hailat, M.; Khaleel, A.; et al. Punicalagin and zinc (II) ions inhibit the activity of SARS-CoV-2 3CL-protease in vitro. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 3908–3913. [Google Scholar] [CrossRef] [PubMed]
- Du, R.; Cooper, L.; Chen, Z.; Lee, H.; Rong, L.; Cui, Q. Discovery of chebulagic acid and punicalagin as novel allosteric inhibitors of SARS-CoV-2 3CLpro. Antivir. Res. 2021, 190, 105075. [Google Scholar] [CrossRef] [PubMed]
- Lin, L.T.; Chen, T.Y.; Lin, S.C.; Chung, C.Y.; Lin, T.C.; Wang, G.H.; Anderson, R.; Lin, C.C.; Richardson, C.D. Broad-spectrum antiviral activity of chebulagic acid and punicalagin against viruses that use glycosaminoglycans for entry. BMC Microbiol. 2013, 13, 187. [Google Scholar] [CrossRef]
- Kim, Y.S.; Chung, H.S.; Noh, S.G.; Lee, B.; Chung, H.Y.; Choi, J.G. Geraniin inhibits the entry of SARS-CoV-2 by blocking the interaction between spike protein RBD and human ACE2 receptor. Int. J. Mol. Sci. 2021, 22, 8604. [Google Scholar] [CrossRef] [PubMed]
- Yang, L.J.; Chen, R.H.; Hamdoun, S.; Coghi, P.; Ng, J.P.L.; Zhang, D.W.; Guo, X.; Xia, C.; Law, B.Y.K.; Wong, V.K.W. Corilagin prevents SARS-CoV-2 infection by targeting RBD-ACE2 binding. Phytomedicine 2021, 87, 153591. [Google Scholar] [CrossRef] [PubMed]
- Li, Q.; Yi, D.; Lei, X.; Zhao, J.; Zhang, Y.; Cui, X.; Xiao, X.; Jiao, T.; Dong, X.; Zhao, X.; et al. Corilagin inhibits SARS-CoV-2 replication by targeting viral RNA-dependent RNA polymerase. Acta Pharm. Sin. B 2021, 11, 1555–1567. [Google Scholar] [CrossRef] [PubMed]
- Promsong, A.; Chuenchitra, T.; Saipin, K.; Tewtrakul, S.; Panichayupakaranant, P.; Satthakarn, S.; Nittayananta, W. Ellagic acid inhibits HIV-1 infection in vitro: Potential role as a novel microbicide. Oral Dis. 2018, 24, 249–252. [Google Scholar] [CrossRef] [PubMed]
- Shin, M.S.; Kang, E.H.; Lee, Y.I. A flavonoid from medicinal plants blocks hepatitis B virus-e antigen secretion in HBV-infected hepatocytes. Antivir. Res. 2005, 67, 163–168. [Google Scholar] [CrossRef]
- Giordano, D.; Facchiano, A.; Carbone, V. Food Plant Secondary Metabolites Antiviral Activity and Their Possible Roles in SARS-CoV-2 Treatment: An Overview. Molecules 2023, 8, 2470. [Google Scholar] [CrossRef]
- Antoine, T.E.; Park, P.J.; Shukla, D. Glycoprotein targeted therapeutics: A new era of anti-herpes simplex virus-1 therapeutics. Rev. Med. Virol. 2013, 23, 194–208. [Google Scholar] [CrossRef]
- Roa-Linares, V.C.; Escudero-Flórez, M.; Vicente-Manzanares, M.; Gallego-Gómez, J.C. Host Cell Targets for Unconventional Antivirals against RNA Viruses. Viruses 2023, 15, 776. [Google Scholar] [CrossRef] [PubMed]
- Muhammad, S.; Hassan, S.H.; Al-Sehemi, A.G.; Shakir, H.A.; Khan, M.; Irfan, M.; Iqbal, J. Exploring the new potential antiviral constituents of Moringa oliefera for SARS-CoV-2 pathogenesis: An in silico molecular docking and dynamic studies. Chem. Phys. Lett. 2021, 767, 138379. [Google Scholar] [CrossRef] [PubMed]
- Bahun, M.; Jukić, M.; Oblak, D.; Kranjc, L.; Bajc, G.; Butala, M.; Bozovičar, K.; Bratkovič, T.; Podlipnik, Č.; Poklar Ulrih, N. Inhibition of the SARS-CoV-2 3CL(pro) main protease by plant polyphenols. Food Chem. 2022, 373, 131594. [Google Scholar] [CrossRef] [PubMed]
- Suručić, R.; Tubić, B.; Stojiljković, M.P.; Djuric, D.M.; Travar, M.; Grabež, M.; Šavikin, K.; Škrbić, R. Computational study of pomegranate peel extract polyphenols as potential inhibitors of SARS-CoV-2 virus internalization. Mol. Cell Biochem. 2021, 476, 1179–1193. [Google Scholar] [CrossRef]
- Arokiyaraj, S.; Stalin, A.; Kannan, B.S.; Shin, H. Geranii Herba as a Potential Inhibitor of SARS-CoV-2 Main 3CL(pro), Spike RBD, and Regulation of Unfolded Protein Response: An In Silico Approach. Antibiotics 2020, 9, 863. [Google Scholar] [CrossRef]
- Umar, A.K.; Zothantluanga, J.H.; Aswin, K.; Maulana, S.; Sulaiman Zubair, M.; Lalhlenmawia, H.; Rudrapal, M.; Chetia, D. Antiviral phytocompounds “ellagic acid” and “(+)-sesamin” of Bridelia retusa identified as potential inhibitors of SARS-CoV-2 3CL pro using extensive molecular docking, molecular dynamics simulation studies, binding free energy calculations, and bioactivity prediction. Struct. Chem. 2022, 33, 1445–1465. [Google Scholar] [CrossRef]
- Pandey, A.K.; Verma, S. An in-silico evaluation of dietary components for structural inhibition of SARS-CoV-2 main protease. J. Biomol. Struct. Dyn. 2022, 40, 136–142. [Google Scholar] [CrossRef]
- Falade, V.A.; Adelusi, T.I.; Adedotun, I.O.; Abdul-Hammed, M.; Lawal, T.A.; Agboluaje, S.A. In silico investigation of saponins and tannins as potential inhibitors of SARS-CoV-2 main protease (M(pro)). In Silico Pharmacol. 2021, 9, 9. [Google Scholar] [CrossRef]
- Gupta, A.; Chauhan, S.S.; Gaur, A.S.; Parthasarathi, R. Computational screening for investigating the synergistic regulatory potential of drugs and phytochemicals in combination with 2-deoxy-D-glucose against SARS-CoV-2. Struct. Chem. 2022, 33, 2179–2193. [Google Scholar] [CrossRef]
- Adelusi, T.I.; Oyedele, A.Q.K.; Monday, O.E.; Boyenle, I.D.; Idris, M.O.; Ogunlana, A.T.; Ayoola, A.M.; Fatoki, J.O.; Kolawole, O.E.; David, K.B.; et al. Dietary polyphenols mitigate SARS-CoV-2 main protease (Mpro)–Molecular dynamics, molecular mechanics, and density functional theory investigations. J. Mol. Struct. 2022, 1250, 131879. [Google Scholar] [CrossRef]
- Monteiro, N.; Monteiro, V.; Lima, L.; Karolline, A.; Machado, R. In Silico Prediction of Inhibitory Potential of a Punicalagin β-anomer Against SARS-CoV-2 Main Protease (Mpro). Quimica Nova 2021, 45, 1230–1235. [Google Scholar] [CrossRef]
- Li, H.; Xu, F.; Liu, C.; Cai, A.; Dain, J.A.; Li, D.; Seeram, N.P.; Cho, B.P.; Ma, H. Inhibitory Effects and Surface Plasmon Resonance-Based Binding Affinities of Dietary Hydrolyzable Tannins and Their Gut Microbial Metabolites on SARS-CoV-2 Main Protease. J. Agric. Food Chem. 2021, 69, 12197–12208. [Google Scholar] [CrossRef] [PubMed]
- Khalifa, I.; Zhu, W.; Mohammed, H.H.H.; Dutta, K.; Li, C. Tannins inhibit SARS-CoV-2 through binding with catalytic dyad residues of 3CLpro: An in silico approach with 19 structural different hydrolysable tannins. J. Food Biochem. 2020, 44, e13412. [Google Scholar] [CrossRef] [PubMed]
- Loschwitz, J.; Jäckering, A.; Keutmann, M.; Olagunju, M.; Eberle, R.J.; Coronado, M.A.; Olubiyi, O.O.; Strodel, B. Novel inhibitors of the main protease enzyme of SARS-CoV-2 identified via molecular dynamics simulation-guided in vitro assay. Bioorg. Chem. 2021, 111, 104862. [Google Scholar] [CrossRef] [PubMed]
- Tito, A.; Colantuono, A.; Pirone, L.; Pedone, E.; Intartaglia, D.; Giamundo, G.; Conte, I.; Vitaglione, P.; Apone, F. Pomegranate Peel Extract as an Inhibitor of SARS-CoV-2 Spike Binding to Human ACE2 Receptor (in vitro): A Promising Source of Novel Antiviral Drugs. Front. Chem. 2021, 9, 638187. [Google Scholar] [CrossRef] [PubMed]
- Vardhan, S.; Sahoo, S.K. Virtual screening by targeting proteolytic sites of furin and TMPRSS2 to propose potential compounds obstructing the entry of SARS-CoV-2 virus into human host cells. J. Tradit. Complement. Med. 2022, 12, 6–15. [Google Scholar] [CrossRef]
- Fraser, B.J.; Beldar, S.; Seitova, A.; Hutchinson, A.; Mannar, D.; Li, Y.; Kwon, D.; Tan, R.; Wilson, R.P.; Leopold, K.; et al. Structure and activity of human TMPRSS2 protease implicated in SARS-CoV-2 activation. Nat. Chem. Biol. 2022, 18, 963–971. [Google Scholar] [CrossRef]
- Vankadari, N.; Ketavarapu, V.; Mitnala, S.; Vishnubotla, R.; Reddy, D.N.; Ghosal, D. Structure of Human TMPRSS2 in Complex with SARS-CoV-2 Spike Glycoprotein and Implications for Potential Therapeutics. J. Phys. Chem. Lett. 2022, 13, 5324–5333. [Google Scholar] [CrossRef]
- Vora, A.; Londhe, V.; Pandita, N. Herbosomes Enhance the in Vivo Antioxidant Activity and Bioavailability of Punicalagins from Standardized Pomegranate Extract. J. Funct. Foods 2015, 12, 540–548. [Google Scholar] [CrossRef]
- Zuccari, G.; Baldassari, S.; Ailuno, G.; Turrini, F.; Alfei, S.; Caviglioli, G. Formulation Strategies to Improve Oral Bioavailability of Ellagic Acid. Appl. Sci. 2020, 10, 3353. [Google Scholar] [CrossRef]
Compound Tested | Experimental System | Findings | References |
---|---|---|---|
pomegranate extract | human consumption of capsules | ↑ antioxidant capacity of plasma (ORAC) within 30 min | [83] |
Alzheimer’s disease transgenic R1.40 mice model | non-significant ↓ TNFα, IL-1 and COX2 | [84] | |
pomegranate flower extract | Zucker diabetic fatty rat | ↓ interstitial and perivascular collagen accumulation in heart, expression of collagen I, collagen III, fibronectin, ET1, ETA, ETB, x NFκB activity | [85] |
pomegranate juice | hyperoxia rat model | ↓ neutrophil infiltration, albumin leak, ROS, apoptotic bodies in lungs, IL-1β, IL-6 | [86] |
pomegranate leaf ethanolic extract | intranasal application in asthma mouse model | ↓ IL-1β, IL-5, inflammatory cell infiltration in lung, mucous glycoprotein secretion | [64] |
pomegranate peel extract | neutrophil culture and LPS-stimulated mice | x MPO activity in neutrophils, ↓ lung invasion of inflammatory cells | [87] |
LPS-induced RAW264.7 macrophages | ↓ TLR4 expression, ↓ IL-1β, IL-6, TNFα, NO, PGE2, ROS production, x nuclear translocation of NFκB nuclear translocation | [70] | |
walnut methanolic extract | human aorta endothelial cells (HAEC) | ↓ TNFα-induced VCAM1 and ICAM1 expression | [80] |
KS483 osteoblastic cells line | nodule formation induced | ||
corilagin | HSV-1 infected MV-2 microglia cells | ↓ secretion of NO, TNFα, IL-1β, ↑ secretion of IL-10, cytochrome c, caspase-3, -8, -9 and -12 | [65] |
HSV-1 infected mice | ↓ numbers of inflammatory cells in the brain, ↓ neuronal degeneration and interstitial edema | ||
punicalagin | acute respiratory distress mouse model | ↓ inflammatory cell lung invasion, alveolar wall thickening, pulmonary congestion, ↓ TNFα, IL-1β, and IL-6 levels, MPO activity, TLR4 expression, x phosphorylation of IκBα and NFκB p65 | [67] |
Jurkat cells | T cell activation by NFAT | [88] | |
activated CD4+ murine splenic lymphocytes | ↓ IL-2 mRNA and protein | ||
PMA-induced ear edema in mice | ↓ hyperplasia and inflammatory cell infiltration | ||
LPS-induced RAW264.7 macrophages | ↓ TLR4 expression, ↓ IL-1β, IL-6, TNFα, NO, PGE2, ROS production, x nuclear translocation of NFκB nuclear translocation | [70] | |
ellagic acid | human aorta endothelial cells (HAEC) | ↓ TNFα-induced VCAM1 and ICAM1 expression | [80] |
KS483 osteoblastic cells line | nodule formation induced | ||
mice on high fat diet | ↓ aortic lesions, plasma cholesterol and triglyceride, ↓sICAM1 and E-selectin expression, ↑ Nrf2, HO-1 protein and aortic NOS activity | [81] | |
human umbilical vein endothelial cells (HUVEC) | Nrf2-mediated cytoprotection, ↑ HO-1 protein | ||
human Caco-2 intestinal cells | ↓ NFκB activation after LPS stimulation, ↑ IκB-α phosphorylation and IL-8 secretion after IL-1β stimulation | [89] | |
in combination with oseltamivir and isoprinosine in influenza A infected mice | ↑ glutathione reductase activity, ↓ TBARS in blood plasma and lungs during infection | [21] | |
LPS-induced RAW264.7 macrophages | ↓ TLR4 expression, ↓ IL-1β, IL-6, TNFα, NO, PGE2, ROS production, x nuclear translocation of NFκB nuclear translocation | [70] | |
Caco-2 and HT-29/B6 intestinal cells | ↑ transepithelial resistance, ↓ caludin-4, -7, -15 expression | [90] | |
urolithin A | experimental autoimmune encephalomyelitis | ↓ demyelination and inflammatory infiltrating cells, reduce severity of disease, ↓ activation of dendritic cells and CNS microglia | [68] |
bone marrow-derived dendritic cells and SIM-A9 microglia | ↓ IL-1β, IL-6, TNFα, ↑ IL-10 | ||
inflammatory bowel disease model LPS-stimulated BMDM | ↓IκB-α phosphorylation, IL-1β, IL-2, IL-6, IL-12, TNFα, NOS2, double-stranded DNA breaks, superoxide production, MAPK and PI3K activation, proinflammatory miRNAs | [69] | |
Caco-2 and HT-29/B6 intestinal cells | x TNF-α induced drop in transepithelial resistance | [31] | |
urolithins | LPS-stimulated BV2 microglia | ↓ NO, TNFα and IL-6, improved SH-SY5Y neuronal cell viability in H2O2 | [63] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Alexova, R.; Alexandrova, S.; Dragomanova, S.; Kalfin, R.; Solak, A.; Mehan, S.; Petralia, M.C.; Fagone, P.; Mangano, K.; Nicoletti, F.; et al. Anti-COVID-19 Potential of Ellagic Acid and Polyphenols of Punica granatum L. Molecules 2023, 28, 3772. https://doi.org/10.3390/molecules28093772
Alexova R, Alexandrova S, Dragomanova S, Kalfin R, Solak A, Mehan S, Petralia MC, Fagone P, Mangano K, Nicoletti F, et al. Anti-COVID-19 Potential of Ellagic Acid and Polyphenols of Punica granatum L. Molecules. 2023; 28(9):3772. https://doi.org/10.3390/molecules28093772
Chicago/Turabian StyleAlexova, Ralitza, Simona Alexandrova, Stela Dragomanova, Reni Kalfin, Ayten Solak, Sidharth Mehan, Maria Cristina Petralia, Paolo Fagone, Katia Mangano, Ferdinando Nicoletti, and et al. 2023. "Anti-COVID-19 Potential of Ellagic Acid and Polyphenols of Punica granatum L." Molecules 28, no. 9: 3772. https://doi.org/10.3390/molecules28093772
APA StyleAlexova, R., Alexandrova, S., Dragomanova, S., Kalfin, R., Solak, A., Mehan, S., Petralia, M. C., Fagone, P., Mangano, K., Nicoletti, F., & Tancheva, L. (2023). Anti-COVID-19 Potential of Ellagic Acid and Polyphenols of Punica granatum L. Molecules, 28(9), 3772. https://doi.org/10.3390/molecules28093772