Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2243 word(s) 2243 2021-05-13 08:07:16 |
2 format correct -21 word(s) 2222 2021-07-02 11:12:59 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Musarra Pizzo, M.; Sciortino, M.T. Natural Compounds against RNA Viruses. Encyclopedia. Available online: https://encyclopedia.pub/entry/11607 (accessed on 25 April 2024).
Musarra Pizzo M, Sciortino MT. Natural Compounds against RNA Viruses. Encyclopedia. Available at: https://encyclopedia.pub/entry/11607. Accessed April 25, 2024.
Musarra Pizzo, Maria, Maria Teresa Sciortino. "Natural Compounds against RNA Viruses" Encyclopedia, https://encyclopedia.pub/entry/11607 (accessed April 25, 2024).
Musarra Pizzo, M., & Sciortino, M.T. (2021, July 02). Natural Compounds against RNA Viruses. In Encyclopedia. https://encyclopedia.pub/entry/11607
Musarra Pizzo, Maria and Maria Teresa Sciortino. "Natural Compounds against RNA Viruses." Encyclopedia. Web. 02 July, 2021.
Natural Compounds against RNA Viruses
Edit

Natural products from plants or other organisms are a rich source of structurally novel chemical compounds including antivirals. Indeed, in traditional medicine, many pathological conditions have been treated using plant-derived medicines. Thus, the identification of novel alternative antiviral agents is of critical importance.

viral infections natural bioactive compounds novel antiviral drugs

1. Human Immunodeficiency Virus (HIV)

Human immunodeficiency viruses 1 and 2 (HIV-1 and HIV-2) infection leads to immunological failure and Acquired Immunodeficiency Syndrome (AIDS). HIV is a member of the lentivirus genus, which includes retroviruses that possess complex genomes. All lentiviruses are enveloped by a lipid bilayer that is derived from the membrane of the host cell. HIV-1 particles bind specifically to cells bearing the CD4 receptor, lymphocytes, and cause their destruction with a half-life of fewer than two days [1]. This leads to the fusion of HIV-1 to the host cell, thereby leading to the release of viral RNA into the cell. The reverse transcriptase enzyme presents in the virus (HIV-1) converts single-stranded viral RNA to double-stranded viral DNA. The formed viral DNA enters the host cell nucleus and incorporates the viral DNA within the host cell’s DNA by the viral enzyme integrase. This integrated viral DNA as provirus could reproduce few or no copies or remain inactive for many years. Since the introduction of highly active antiretroviral therapy (HAART), as well as the impact of preventive measures, the prevalence and incidence of HIV, have declined globally over the last decade except for parts of Eastern Europe and Central Asia [2]. However, the present therapy finds its limitations in the emergence of multidrug resistance as well as HIV persistence within latent cellular reservoirs. Compounds derived from plant, marine, and other natural products have been found to combat HIV infection and/or target HIV reservoirs, and these discoveries have substantially guided current HIV therapy-based studies. Accordingly, finding new drugs and novel targets is needed to treat infected people and to eliminate HIV reservoirs in order to ultimately block HIV infection. Recently, several anti-HIV compounds obtained from natural products have been extensively reported [3]. However, a limited number of those are in the advanced development stage associated with well-known mechanisms of action [4] (Table 1).
Table 1. Natural compounds and their antiviral targets against Immunodeficiency virus.
Natural Source Compound Immunodeficiency Virus Target CC50 EC50-IC50 SI Reference
Griffithsia sp. Griffithsin HIV Entry inhibitors   0.043–0.63 nM   [5]
Nostoc ellipsosporum Ascyanovirin-N HIV       [6]
Siliquariaspongia mirabilis
Stelletta clavosa
Mirabamide-A HIV   40–140 nM   [7]
Syzygium claviflorum Betulinic acid Dihydro betulinic HIV Maturation inhibitors   1.4 µM
0.9 µM
9.3
14
[8]
Synthetic derivative of betulinic acid Bevirimat HIV 25 μM 7.8 nM >2500 [9]
Rheum palmatum Sennoside A HIV Reverse transcriptase and Integrase inhibitors       [10]
Morus nigra Kuwanon-L         [11]
Justica gendarussa Patentiflorin A HIV   24−37 nM   [12]
Calophyllum lanigerum Calanolides HIV   0.1–0.4 µM   [13]
Euphorbia kansui Ingenol
Bryostatin
Prostratin
HIV Latency-reversing agents (LRAs)       [14]
Theobroma cacao Procyanidin C1-flavonoids HIV       [15]
CC50: Half maximal cytotoxic concentration; EC50-IC50: Half maximal inhibitory concentration; SI: Selectivity index = CC50/IC50.

2. Influenza Viruses

Influenza viruses are negative-stranded, segmented RNA viruses, and are members of the Orthomyxoviridae family. Influenza viruses comprise three types (A, B, and C), while type A is divided into different subtypes which are distinguishable by antigenicity of their surface glycoproteins, haemagglutinin (HA), and neuraminidase (NA) [16]. Influenza viruses, based on their genetic mutations can be categorized into two different entities: seasonal or pandemic representing a major public health problem, with high rates of morbidity and mortality [17]. Airway epithelial cells lining the respiratory mucosa are the primary target of influenza infection. The recognition of influenza virus antigens through antigen-presenting cells and pattern recognition receptors (PRRs) can consequently upregulate several correspondent downstream molecules including interleukin-6 (IL-6), IL-1β, and tumour necrosis factor α (TNF-α) which causes influenza mediated signs and symptoms [18]. To date, anti-influenza drugs include M2 ion channel inhibitors and neuraminidase inhibitors [19]. M2 ion channel inhibitors, such as amantadine and rimantadine, act as inhibitors of the uncoating process, which is essential for the release of the virus into the cytoplasm. Neuraminidase inhibitors, including oseltamivir and zanamivir, are directed against the enzymatic activity of neuraminidase, which assures the release of progeny viruses from infected cells. It has been suggested that herbal medicines might be beneficial in the prevention or management of seasonal or pandemic influenza (Table 2). An in vitro study showed that oligonol extracted from lychee fruit (Litchi chinensis) inhibits proliferation of influenza virus H3N2 by blocking reactive oxygen species (ROS)-dependent ERK (extracellular-signal-regulated kinases) phosphorylation [20]. Another in vitro investigation showed that green tea catechins possess higher inhibitory effects on the endonuclease activity of influenza A virus RNA polymerase [21]. Slaine and collaborators showed that the macrolide pateamine A (from Mycale hentscheli) and the rocagalte silvestrol (from Aglaia), two inhibitors of the eukaryotic initiation factor-4A (eIF4A), caused the block of influenza A viral protein synthesis as well as the failure of the viral genome replication [22]. The inhibitory effect of silvestrol was fully reversible while the pateamine A irreversibly binds to eIF4A and caused the inhibition of genetically divergent influenza A strains replication [22].
Table 2. Natural products against Influenza viruses.
Natural Source Compound Influenza
Viruses
Target CC50 EC50-IC50 SI Reference
Litchi chinensis Oligonol H3N2 Blocking (ROS)-dependent ERK
phosphorylation
      [20]
Green tea Catechins H1N1 Inhibiting RNA
polymerase
      [21]
Aglaia Silvestrol H1N1 Inhibitors of the
cellular factor eEIF4A
      [22]
Mycale hentscheli Pateamine A H1N1
H3N2
     
Curcuma longa L. Curcumin H1N1
H6N1
Haemagglutinin
inhibitors
43 µM 0.47 µM 92.5 [23]
Cistus incanus Polyphenol rich extract A549   50 μg/mL   [24]
Punica granatum Punicalagin H3N2       [25]
Green tea Epigallocatechin gallate H1N1       [26]

CC50: Half maximal cytotoxic concentration; EC50-IC50: Half maximal inhibitory concentration; SI: Selectivity index = CC50/IC50.

Natural Products as Hemagglutinin Inhibitors

Given the rapid emergence of drug-resistant influenza virus strains, hemagglutinin (HA) is a promising target for developing anti-influenza drugs. HA is an envelope protein that plays a critical role in viral binding, fusion and entry processes. Curcumin showed anti-influenza activity against influenza viruses PR8, H1N1, and H6N1. The results showed more than a 90% reduction in virus yield in cell culture using 30 μM of curcumin. The plaque reduction test elicited the approximate EC50 of 0.47 μM for curcumin against influenza viruses [23]. In H1N1 and also H6N1 subtypes, the inhibition of HA interaction reflected the direct effect of curcumin on infectivity of viral particles and this has proved by the time of the drug addiction experiment [23]. An in vitro investigation indicated that Cistus incanus, a member of the Cistaceae family, has anti-influenza virus activity in A549 (human lung epithelial cell) or MadinDarby canine kidney (MDCK) cell cultures infected with prototype avian and human influenza strains of different subtypes by the reduction of progeny virus titers of up to two logs without any toxicity [24]. Furthermore, the binding of the polyphenol components of the extract to the virus surface showed protective effects through inhibition of HA binding to cellular receptors [24]. In another study, Cistus incanus exhibited antiviral activity against a highly pathogenic avian influenza A virus (H7N7) in both cell cultures and a mouse infection model [27]. Haidari and colleagues indicated that punicalagin from Punica granatum polyphenol-rich extract had anti-influenza properties in MDCK and chicken red blood cells (cRBC) infected by human influenza A (H3N2) through inhibiting the virus replication as well as inhibiting virus-induced agglutination of cRBCs [25]. Furthermore, an investigation showed that epigallocatechin gallate (EGCG) and theaflavin digallate (TF3) from green tea and black tea respectively, inhibit the infectivity of both influenza A and B viruses in MDCK cells through binding to virus HA and prevention of virus adsorption to MDCK cells [26].

3. Hepatitis C Virus

Hepatitis C virus (HCV) is an enveloped, positive-sense single-stranded RNA virus belonging to the Flaviviridae family. The HCV genomic RNA encodes a polyprotein that is then cleaved by both the host and virus proteases into mature proteins. The nonstructural proteins; NS2, NS3, NS4A, NS4B, NS5A, and NS5B, core proteins, glycoproteins E1, and E2; the ion channel p7 (Banerjee et al. 2010). The (NS5B), an RNA-dependent RNA polymerase (RdRp) is responsible for replicating the viral RNA genome [28]. Infected individuals have treated with standard treatment, consisting of PEGylated (PEG)-interferon (IFN)-α in combination with ribavirin (RBV) for over a decade. Recently, several protease inhibitors such as boceprevir and telaprevir have been approved as treatments for hepatitis C. However, these new inhibitors are associated with drug toxicity and the development of resistant mutants [29]. Based on this, many phytochemical constituents have been identified that display considerable inhibition of the HCV life cycle at different steps (Table 3).
Table 3. Natural products against the Hepatitis C virus.
Natural Source Compound HCV Target CC50
(µg/mL)
EC50-IC50 (µg/mL) SI or TI Reference
Trichilia dregeana Root extract HCV Inhibition of viral entry   16.6 37 [30]
Detarium
microcarpum
Stem bark extract   1.42 211
Phragmanthera
capitata
Leave extract   13.17  
Bupleurum kaoi Saikosaponin B2 740.4 ± 28.35 µM 16.13 ± 2.41 µM 45.9 [31]
Bupleurum kaoi Methanolic extract 16.82 ± 1.89 215.4 ± 10.7 12.8
Anthocyanidin Delphinidin   3.7 ± 0.8 μM   [32]
Alloeocomatella polycladia Ethyl acetate-
soluble fraction
Suppression of the
helicase activity of
HCV NS3
  11.7 ± 0.7   [33]
Fusarium equiseti Crude extracts Inhibition of HCV NS3/4A protease   19–77 µM   [34]
Eclipta alba Aqueous extract Inhibition of HCV
NS5B replicase activity
  11   [35]
Taraxacum officinale Flavonoids   [36]
Swietenia macrophylla 3-hydroxy caruilignan C Reduction of HCV
protein and HCV- RNA levels
  10.5 ± 1.2μM   [37]
Entada africana Methylene
chloride-methanol (MCM) stem
bark crude extract
Broad antiviral activity   453 ± 0.00117   [38]
Grapeseed Phenolic
compounds
Suppression of HCV-
induced Cox-2
  7.5 ± 0.3   [39]
Flavanone Naringenin Release/Assembly   109 μM   [40]
CC50: Half maximal cytotoxic concentration; EC50-IC50: Half maximal inhibitory concentration; SI: Selectivity index = CC50/IC50.

4. Picornaviruses

The family Picornaviridae currently contains 147 species grouped into 63 genera. Viruses in the family Picornaviridae have non-enveloped particles with a single-stranded RNA (ssRNA) genome and include numerous human pathogens such as poliovirus, enterovirus 71, foot and mouth disease virus (FMDV), hepatitis A virus and rhinovirus. The broadly studied and most well-characterized group is represented by an enterovirus, including enterovirus A71 (EV71), coxsackievirus, poliovirus, and rhinovirus. The viral infection initiates by attaching to a receptor on the host cell plasma membrane and viruses belonging to different genera use different receptors to bind to and infect cells, thus exhibiting a different tissue tropism [41]. The antiviral drugs used in the picornaviruses treatment target virus entry (pleconaril, WIN54954 and CAR-Fc), the viral translation and/or transcription (antisense oligodeoxynucleotide and short interfering RNA) or intracellular signalling pathway (immune response activators) but do not completely eradicate the infection. To date, the antiviral activity of several natural products and herbal medicines have been tested against various picornavirus (Table 4).
Table 4. Natural compounds and their antiviral targets against Picornaviruses.
Natural Source Compound Picornaviruses Target CC50
(µg/mL)
EC50-IC50
(µg/mL)
SI Reference
Lagerstroemia
speciosa L.
Orobol 7-O-d-glucoside (O7G) Human rhinovirus A
Human rhinovirus B
Broad
spectrum
antiviral
activity
100 0.58-8.80 12 [42]
Ocimum basilicum Crude aqueous extracts Coxsackievirus B1
Enterovirus 71
1469.3 105.7 ± 2.6
200.2 ± 3.2
13.9
7.3
[43]
Ocimum basilicum Ethanolic extracts Coxsackievirus B1
Enterovirus 71
684.8 146.3 ± 2.9
198.9 ± 1.8
4.7
3.4
Woodfordia fruticosa Gallic acid Enterovirus 71 100 0.76 132 [44]
Raoulia australis Raoulic acid Human rhinovirus 2
Human rhinovirus 3
Coxsackievirus B3
Coxsackievirus B4
Enterovirus 71
201.78
65.86
0.1
0.19
0.33
0.40
0.1
  [45][46]
Ocimum basilicum Ursolic acid Coxsackievirus B1
Enterovirus 71
Targets viral structures and inhibits viral infection and
replication
process
100.5 0.4 ± 0.1
0.5 ± 0.2
251
201
[43]
Silybum marianum Silymarin Enterovirus 71 160.20 ± 1.56 7.99 ± 3.0 20.05 [47]
Macaranga barteri DCM fraction Echoviruses E7
Echoviruses E19
0.18 7.54 × 10−6
1.75 × 10−6
19.9
8581.24
[48]
Syzygium
brazzavillense
Aqueous extract Coxsackievirus B4 2800 0.8   [49]
Rheum palmatum Ethanol extract Coxsackievirus B3   4 10 [50]
Lagerstroemia
speciosa L.
Quercetin-7-glucoside (Q7G) Human rhinovirus 2 >100 4.85–0.59 >20.62 [51]
Salvia miltiorrhiza Rosmarinic acid Enterovirus A71 327.68 ± 14.43 31.57–114 2.87–10.36 [52]
Green tea Epigallocatechin-3-gallate (EGCG) Hepatitis virus A       [53]
Vitis vinifera Grapeseed
extract (GSE)
Hepatitis virus A       [54][55]
Lagerstroemia
speciosa L.
Tannin ellagic acid Human rhinovirus 2
Human rhinovirus 3
Human rhinovirus 4
Targets host cellular factors >100 38 ± 3.2
31 ± 5.2
29 ± 2.5
>2.6
>3.2
>3.4
[56]
Bupleurum kaoi Roots extract Coxsackievirus B1 883.56 50.93   [57]
Mix of seven
medicinal herbs
Xiao chai hu tang Coxsackievirus B1 945.75 50.93 18.92 [58]
Ornithogalum
saundersiae
Orsaponin (OSW-1) Enterovirus 71, Coxsackievirus A21 Human rhinovirus 2 >100 nM 2.4–9.4 nM   [59][60]
Panax ginseng Ginsenosides Hepatitis virus A       [61]

CC50: Half maximal cytotoxic concentration; EC50-IC50: Half maximal inhibitory concentration; SI: Selectivity index = CC50/IC50.

5. Norovirus

Human noroviruses (HuNoVs) are the leading cause of gastroenteritis and severe childhood diarrhea worldwide [62]. Noroviruses belong to the family Caliciviridae, members of which have a small, single-stranded positive-sense RNA genome. Norovirus utilizes cell surface molecules as mediators for binding and cellular entry. Murine norovirus (MNV-1) and feline calicivirus (FCV) were successfully grown and served as a surrogate model system for HuNoV [63]. Several medicinal plants and herb extracts were screened for antiviral activity against MVN-1 and FCV as a surrogate of norovirus (Table 5). The work of Lee and colleagues showed that the components of Morus alba L. possess antiviral effects against foodborne enteric virus surrogates. It was found that Morus alba juice and its fractions inhibit the internalization and replication of MNV-1 as well as the internalization of FCV-F9 virions [64]. Black raspberry juice (Rubus coreanus) was found to decrease MNV-1 plaque formation by blocking viral entry into the cell and inhibiting its internalization or through direct effects on viral particles or host cell receptors [65]. Green tea polyphenolic chatechins from Camellia sinensis exhibited anti-FCV-F9 antiviral activity with epigallocatechin gallate showing the best combination of antiviral activity and low cytotoxicity [66]. The essential oil from oregano (Origanum vulgare) decreased FCV-F9 and MNV-1 replication in a dose-dependent manner. Besides, it has been shown that oregano essential oil and its primary component carvacrol caused the loss of viral capsid integrity of MNV-1 virions as determined by transmission electron microscopy experiments [67]. Persimmon (Diospyros kaki) extracts containing persimmon tannin was found to reduce noroviral genome replication with no cytotoxicity effect [68].
Table 5. Natural products against Noroviruses.
Natural Source Compound Noroviruses Target CC50 EC50-IC50 SI Reference
Morus alba L Juice MNV-1
FCV-F9
Inhibiting
internalization and replication
>0.1%
>2.5%
0.005%
0.25%
20
10
[64]
Camellia sinensis Epigallocatechin gallate FCV-F9   12 mg/mL   [66]
Rubus coreanus Juice MNV-1       [65]
Origanum vulgare Carvacrol MNV-1       [67]
Diospyros kaki Persimmon tannin HuNoV Reduce genome
replication
      [68]

CC50: Half maximal cytotoxic concentration; EC50-IC50: Half maximal inhibitory concentration; SI: Selectivity index = CC50/IC50.

References

  1. Ho, D.D.; Neumann, A.U.; Perelson, A.S.; Chen, W.; Leonard, J.M.; Markowitz, M. Rapid turnover of plasma virions and CD4 lymphocytes in HIV-1 infection. Nature 1995, 373, 123–126.
  2. Kumari, G.; Singh, R.K. Highly Active Antiretroviral Therapy for treatment of HIV/AIDS patients: Current status and future prospects and the Indian scenario. HIV AIDS Rev. 2012, 11, 5–14.
  3. Cary, D.C.; Peterlin, B.M. Natural Products and HIV/AIDS. AIDS Res. Hum. Retrovir. 2018, 34, 31–38.
  4. Andersen, R.J.; Ntie-Kang, F.; Tietjen, I. Natural product-derived compounds in HIV suppression, remission, and eradication strategies. Antivir. Res. 2018, 158, 63–77.
  5. Lusvarghi, S.; Bewley, C.A. Griffithsin: An Antiviral Lectin with Outstanding Therapeutic Potential. Viruses 2016, 8, 296.
  6. Gandhi, M.J.; Boyd, M.R.; Yi, L.; Yang, G.G.; Vyas, G.N. Properties of Cyanovirin-N (CV-N): Inactivation of HIV-1 by Sessile Cyanovirin-N (SCV-N). Dev. Biol. 2000, 102, 141–148.
  7. Plaza, A.; Gustchina, E.; Baker, H.L.; Kelly, M.; Bewley, C.A. Mirabamides A–D, Depsipeptides from the SpongeSiliquariaspongia mirabilisThat Inhibit HIV-1 Fusion. J. Nat. Prod. 2007, 70, 1753–1760.
  8. Kashiwada, Y.; Hashimoto, F.; Cosentino, L.M.; Chen, C.-H.; Garrett, A.P.E.; Lee, K.-H. Betulinic Acid and Dihydrobetulinic Acid Derivatives as Potent Anti-HIV Agents1. J. Med. Chem. 1996, 39, 1016–1017.
  9. Martin, D.E.; Salzwedel, K.; Allaway, G.P. Bevirimat: A Novel Maturation Inhibitor for the Treatment of HIV-1 Infection. Antivir. Chem. Chemother. 2008, 19, 107–113.
  10. Esposito, F.; Carli, I.; Del Vecchio, C.; Xu, L.; Corona, A.; Grandi, N.; Piano, D.; Maccioni, E.; Distinto, S.; Parolin, C.; et al. Sennoside A, derived from the traditional chinese medicine plant Rheum L., is a new dual HIV-1 inhibitor effective on HIV-1 replication. Phytomedicine 2016, 23, 1383–1391.
  11. Martini, R.; Esposito, F.; Corona, A.; Ferrarese, R.; Ceresola, E.R.; Visconti, L.; Tintori, C.; Barbieri, A.; Calcaterra, A.; Iovine, V.; et al. Natural Product Kuwanon-L Inhibits HIV-1 Replication through Multiple Target Binding. ChemBioChem 2017, 18, 374–377.
  12. Zhang, H.-J.; Rumschlag-Booms, E.; Guan, Y.-F.; Wang, D.-Y.; Liu, K.-L.; Li, W.-F.; Nguyen, V.H.; Cuong, N.M.; Soejarto, D.D.; Fong, H.H.S.; et al. Potent Inhibitor of Drug-Resistant HIV-1 Strains Identified from the Medicinal PlantJusticia gendarussa. J. Nat. Prod. 2017, 80, 1798–1807.
  13. Kashman, Y.; Gustafson, K.R.; Fuller, R.W.; Ii, J.H.C.; McMahon, J.B.; Currens, M.J.; Buckheit, R.W., Jr.; Hughes, S.H.; Cragg, G.M.; Boyd, M.R. HIV inhibitory natural products. Part 7. The calanolides, a novel HIV-inhibitory class of coumarin derivatives from the tropical rainforest tree, Calophyllum lanigerum. J. Med. Chem. 1992, 35, 2735–2743.
  14. Cary, D.C.; Fujinaga, K.; Peterlin, B.M. Euphorbia Kansui Reactivates Latent HIV. PLoS ONE 2016, 11, e0168027.
  15. Hori, T.; Barnor, J.; Huu, T.N.; Morinaga, O.; Hamano, A.; Ndzinu, J.; Frimpong, A.; Minta-Asare, K.; Amoa-Bosompem, M.; Brandful, J.; et al. Procyanidin trimer C1 derived from Theobroma cacao reactivates latent human immunodeficiency virus type 1 provirus. Biochem. Biophys. Res. Commun. 2015, 459, 288–293.
  16. Medina, R.A.; García-Sastre, A. Influenza A viruses: New research developments. Nat. Rev. Genet. 2011, 9, 590–603.
  17. Diceinson, G.T. Epidemic and Endemic Influenza. Can. Med. Assoc. J. 1962, 86, 588–589.
  18. Bahadoran, A.; Lee, S.H.; Wang, S.M.; Manikam, R.; Rajarajeswaran, J.; Raju, C.S.; Sekaran, S.D. Immune Responses to Influenza Virus and Its Correlation to Age and Inherited Factors. Front. Microbiol. 2016, 7, 1841.
  19. De Clercq, E. Antiviral agents active against influenza A viruses. Nat. Rev. Drug Discov. 2006, 5, 1015–1025.
  20. Gangehei, L.; Ali, M.; Zhang, W.; Chen, Z.; Wakame, K.; Haidari, M. Oligonol a low molecular weight polyphenol of lychee fruit extract inhibits proliferation of influenza virus by blocking reactive oxygen species-dependent ERK phosphorylation. Phytomedicine 2010, 17, 1047–1056.
  21. Kuzuhara, T.; Iwai, Y.; Takahashi, H.; Hatakeyama, D.; Echigo, N. Green tea catechins inhibit the endonuclease activity of influenza A virus RNA polymerase. PLoS Curr. 2009, 1, RRN1052.
  22. Slaine, P.D.; Kleer, M.; Smith, N.K.; Khaperskyy, D.A.; McCormick, C. Stress Granule-Inducing Eukaryotic Translation Initiation Factor 4A Inhibitors Block Influenza A Virus Replication. Viruses 2017, 9, 388.
  23. Chen, D.-Y.; Shien, J.-H.; Tiley, L.; Chiou, S.-S.; Wang, S.-Y.; Chang, T.-J.; Lee, Y.-J.; Chan, K.-W.; Hsu, W.-L. Curcumin inhibits influenza virus infection and haemagglutination activity. Food Chem. 2010, 119, 1346–1351.
  24. Ehrhardt, C.; Hrincius, E.R.; Korte, V.; Mazur, I.; Droebner, K.; Poetter, A.; Dreschers, S.; Schmolke, M.; Planz, O.; Ludwig, S. A polyphenol rich plant extract, CYSTUS052, exerts anti influenza virus activity in cell culture without toxic side effects or the tendency to induce viral resistance. Antivir. Res. 2007, 76, 38–47.
  25. Haidari, M.; Ali, M.; Casscells, S.W.; Madjid, M. Pomegranate (Punica granatum) purified polyphenol extract inhibits influenza virus and has a synergistic effect with oseltamivir. Phytomedicine 2009, 16, 1127–1136.
  26. Nakayama, M.; Suzuki, K.; Toda, M.; Okubo, S.; Hara, Y.; Shimamura, T. Inhibition of the infectivity of influenza virus by tea polyphenols. Antivir. Res. 1993, 21, 289–299.
  27. Droebner, K.; Ehrhardt, C.; Poetter, A.; Ludwig, S.; Planz, O. CYSTUS052, a polyphenol-rich plant extract, exerts anti-influenza virus activity in mice. Antivir. Res. 2007, 76, 1–10.
  28. Butcher, S.J.; Grimes, J.M.; Makeyev, E.V.; Bamford, D.H.; Stuart, D.I. A mechanism for initiating RNA-dependent RNA polymerization. Nat. Cell Biol. 2001, 410, 235–240.
  29. Wyles, D.L.; Luetkemeyer, A.F. Understanding Hepatitis C Virus Drug Resistance: Clinical Implications for Current and Future Regimens. Top. Antivir. Med. 2017, 25, 103–109.
  30. Galani, B.R.T.; Sahuc, M.-E.; Njayou, F.N.; Deloison, G.; Mkounga, P.; Feudjou, W.F.; Brodin, P.; Rouillé, Y.; Nkengfack, A.E.; Moundipa, P.F.; et al. Plant extracts from Cameroonian medicinal plants strongly inhibit hepatitis C virus infection in vitro. Front. Microbiol. 2015, 6, 488.
  31. Lin, L.-T.; Chung, C.-Y.; Hsu, W.-C.; Chang, S.-P.; Hung, T.-C.; Shields, J.; Russell, R.S.; Lin, C.-C.; Liang-Tzung, L.; Yen, M.-H.; et al. Saikosaponin b2 is a naturally occurring terpenoid that efficiently inhibits hepatitis C virus entry. J. Hepatol. 2015, 62, 541–548.
  32. Calland, N.; Sahuc, M.-E.; Belouzard, S.; Pène, V.; Bonnafous, P.; Mesalam, A.A.; Deloison, G.; Descamps, V.; Sahpaz, S.; Wychowski, C.; et al. Polyphenols Inhibit Hepatitis C Virus Entry by a New Mechanism of Action. J. Virol. 2015, 89, 10053–10063.
  33. Yamashita, A.; Salam, K.A.; Furuta, A.; Matsuda, Y.; Fujita, O.; Tani, H.; Fujita, Y.; Fujimoto, Y.; Ikeda, M.; Kato, N.; et al. Inhibition of Hepatitis C Virus Replication and Viral Helicase by Ethyl Acetate Extract of the Marine Feather Star Alloeocomatella polycladia. Mar. Drugs 2012, 10, 744–761.
  34. Hawas, U.W.; Al-Farawati, R.; El-Kassem, L.T.A.; Turki, A.J. Different Culture Metabolites of the Red Sea Fungus Fusarium equiseti Optimize the Inhibition of Hepatitis C Virus NS3/4A Protease (HCV PR). Mar. Drugs 2016, 14, 190.
  35. Singh, B.; Saxena, A.K.; Chandan, B.K.; Agarwal, S.G.; Bhatia, M.S.; Anand, K.K. Hepatoprotective effect of ethanolic extract ofEclipta alba on experimental liver damage in rats and mice. Phytother. Res. 1993, 7, 154–158.
  36. Rehman, S.; Ijaz, B.; Fatima, N.; Muhammad, S.A.; Riazuddin, S. Therapeutic potential of Taraxacum officinale against HCV NS5B polymerase: In-vitro and In silico study. Biomed. Pharmacother. 2016, 83, 881–891.
  37. Wu, S.-F.; Lin, C.-K.; Chuang, Y.-S.; Chang, F.-R.; Tseng, C.-K.; Wu, Y.-C.; Lee, J.-C. Anti-hepatitis C virus activity of 3-hydroxy caruilignan C from Swietenia macrophylla stems. J. Viral Hepat. 2011, 19, 364–370.
  38. Tietcheu, B.R.G.; Sass, G.; Njayou, N.F.; Mkounga, P.; Tiegs, G.; Moundipa, P.F. Anti-Hepatitis C Virus Activity of Crude Extract and Fractions of Entada africana in Genotype 1b Replicon Systems. Am. J. Chin. Med. 2014, 42, 853–868.
  39. Chen, W.-C.; Tseng, C.-K.; Chen, B.-H.; Lin, C.-K.; Lee, J.-C. Grape Seed Extract Attenuates Hepatitis C Virus Replication and Virus-Induced Inflammation. Front. Pharmacol. 2016, 7, 490.
  40. Nahmias, Y.; Goldwasser, J.; Casali, M.; Van Poll, D.; Wakita, T.; Chung, R.T.; Yarmush, M.L. Apolipoprotein B-dependent hepatitis C virus secretion is inhibited by the grapefruit flavonoid naringenin. Hepatology 2008, 47, 1437–1445.
  41. Wormser, G.P.; Rubin, D.H. Fundamental Virology, 4th Edition By David M. Knipe, Peter M. Howley, Diane E. Griffin, Robert A. Lamb, Malcolm A. Martin, Bernard Roizman, and Stephen E. Straus Philadelphia: Lippincott Williams & Wilkins, 2001. 1408 pp. $99.95 (cloth). Fields Virology, 4th Edition, Volumes I and II By David M. Knipe, Peter M. Howley, Diane E. Griffin, Robert A. Lamb, Malcolm A. Martin, Bernard Roizman, and Stephen E. Straus Philadelphia: Lippincott Williams & Wilkins, 2001. 3280 pp. $339.00 (cloth). Clin. Infect. Dis. 2002, 34, 1029–1030.
  42. Choi, H.; Bae, E.; Song, J.; Baek, S.; Kwon, D. Inhibitory effects of orobol 7-O-d-glucoside from banaba (Lagerstroemia speciosa L.) on human rhinoviruses replication. Lett. Appl. Microbiol. 2010, 51, 1–5.
  43. Chiang, L.-C.; Ng, L.-T.; Cheng, P.-W.; Chiang, W.; Lin, C.-C. Antiviral activities of extracts and selected pure constituents of Ocimum basilicum. Clin. Exp. Pharmacol. Physiol. 2005, 32, 811–816.
  44. Choi, H.; Song, J.; Park, K.; Baek, S. In vitroanti-enterovirus 71 activity of gallic acid fromWoodfordia fruticosaflowers. Lett. Appl. Microbiol. 2010, 50, 438–440.
  45. Choi, H.; Lim, C.; Song, J.; Baek, S.; Kwon, D. Antiviral activity of raoulic acid from Raoulia australis against Picornaviruses. Phytomedicine 2009, 16, 35–39.
  46. Choi, H.J.; Song, J.-H.; Lim, C.-H.; Baek, S.-H.; Kwon, D.-H. Anti-Human Rhinovirus Activity of Raoulic Acid fromRaoulia australis. J. Med. Food 2010, 13, 326–328.
  47. Lalani, S.S.; Anasir, M.I.; Poh, C.L. Antiviral activity of silymarin in comparison with baicalein against EV-A71. BMC Complement. Med. Ther. 2020, 20, 97.
  48. Ogbole, O.O.; Akinleye, T.E.; Segun, P.A.; Faleye, T.C.; Adeniji, A.J. In vitro antiviral activity of twenty-seven medicinal plant extracts from Southwest Nigeria against three serotypes of echoviruses. Virol. J. 2018, 15, 1–8.
  49. Badia-Boungou, F.; Sane, F.; Alidjinou, E.K.; Hennebelle, T.; Roumy, V.; Ngakegni-Limbili, A.C.; Nguimbi, E.; Moukassa, D.; Abena, A.A.; Hober, D.; et al. Aqueous extracts of Syzygium brazzavillense can inhibit the infection with coxsackievirus B4 in vitro. J. Med. Virol. 2019, 91, 1210–1216.
  50. Xiong, H.-R.; Shen, Y.-Y.; Lu, L.; Hou, W.; Luo, F.; Xiao, H.; Yang, Z.-Q. The Inhibitory Effect of Rheum palmatum Against Coxsackievirus B3 in Vitro and in Vivo. Am. J. Chin. Med. 2012, 40, 801–812.
  51. Song, J.H.; Park, K.S.; Kwon, D.H.; Choi, H.J. Anti–Human Rhinovirus 2 Activity and Mode of Action of Quercetin-7-Glucoside fromLagerstroemia speciosa. J. Med. Food 2013, 16, 274–279.
  52. Hsieh, C.-F.; Jheng, J.-R.; Lin, G.-H.; Chen, Y.-L.; Ho, J.-Y.; Liu, C.-J.; Hsu, K.-Y.; Chen, Y.-S.; Chan, Y.F.; Yu, H.-M.; et al. Rosmarinic acid exhibits broad anti-enterovirus A71 activity by inhibiting the interaction between the five-fold axis of capsid VP1 and cognate sulfated receptors. Emerg. Microbes Infect. 2020, 9, 1194–1205.
  53. Randazzo, W.; Falcó, I.; Aznar, R.; Sánchez, G. Effect of green tea extract on enteric viruses and its application as natural sanitizer. Food Microbiol. 2017, 66, 150–156.
  54. Patwardhan, M.; Morgan, M.T.; Dia, V.; D’Souza, D.H. Heat sensitization of hepatitis A virus and Tulane virus using grape seed extract, gingerol and curcumin. Food Microbiol. 2020, 90, 103461.
  55. Su, X.; D’Souza, D.H. Grape Seed Extract for Control of Human Enteric Viruses. Appl. Environ. Microbiol. 2011, 77, 3982–3987.
  56. 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.
  57. Cheng, P.-W.; Chiang, L.-C.; Yen, M.-H.; Lin, C.-C. Bupleurum kaoi inhibits Coxsackie B virus type 1 infection of CCFS-1 cells by induction of type I interferons expression. Food Chem. Toxicol. 2007, 45, 24–31.
  58. Cheng, P.-W.; Ng, L.-T.; Lin, C.-C. Xiao Chai Hu Tang inhibits CVB1 virus infection of CCFS-1 cells through the induction of Type I interferon expression. Int. Immunopharmacol. 2006, 6, 1003–1012.
  59. Albulescu, L.; Bigay, J.; Biswas, B.; Weber-Boyvat, M.; Dorobantu, C.M.; Delang, L.; Van Der Schaar, H.M.; Jung, Y.-S.; Neyts, J.; Olkkonen, V.M.; et al. Uncovering oxysterol-binding protein (OSBP) as a target of the anti-enteroviral compound TTP-8307. Antivir. Res. 2017, 140, 37–44.
  60. Burgett, A.W.G.; Poulsen, T.B.; Wangkanont, K.; Anderson, D.R.; Kikuchi, C.; Shimada, K.; Okubo, S.; Fortner, K.C.; Mimaki, Y.; Kuroda, M.; et al. Natural products reveal cancer cell dependence on oxysterol-binding proteins. Nat. Chem. Biol. 2011, 7, 639–647.
  61. Kulka, M.; Calvo, M.S.; Ngo, D.T.; Wales, S.Q.; Goswami, B.B. Activation of the 2-5OAS/RNase L pathway in CVB1 or HAV/18f infected FRhK-4 cells does not require induction of OAS1 or OAS2 expression. Virology 2009, 388, 169–184.
  62. Glass, R.I.; Parashar, U.D.; Estes, M.K. Norovirus Gastroenteritis. N. Engl. J. Med. 2009, 361, 1776–1785.
  63. Kniel, K.E. The makings of a good human norovirus surrogate. Curr. Opin. Virol. 2014, 4, 85–90.
  64. Lee, J.-H.; Bae, S.Y.; Oh, M.; Kim, K.H.; Chung, M.S. Antiviral Effects of Mulberry (Morus alba) Juice and Its Fractions on Foodborne Viral Surrogates. Foodborne Pathog. Dis. 2014, 11, 224–229.
  65. Oh, M.; Bae, S.Y.; Lee, J.-H.; Cho, K.J.; Kim, K.H.; Chung, M.S. Antiviral Effects of Black Raspberry (Rubus coreanus) Juice on Foodborne Viral Surrogates. Foodborne Pathog. Dis. 2012, 9, 915–921.
  66. Oh, E.-G.; Kim, K.-L.; Shin, S.B.; Son, K.-T.; Lee, H.-J.; Kim, T.H.; Kim, Y.-M.; Cho, E.-J.; Kim, D.-K.; Lee, E.-W.; et al. Antiviral activity of green tea catechins against feline calicivirus as a surrogate for norovirus. Food Sci. Biotechnol. 2013, 22, 593–598.
  67. Gilling, D.; Kitajima, M.; Torrey, J.; Bright, K. Antiviral efficacy and mechanisms of action of oregano essential oil and its primary component carvacrol against murine norovirus. J. Appl. Microbiol. 2014, 116, 1149–1163.
  68. Kamimoto, M.; Nakai, Y.; Tsuji, T.; Shimamoto, T.; Shimamoto, T. Antiviral Effects of Persimmon Extract on Human Norovirus and Its Surrogate, Bacteriophage MS2. J. Food Sci. 2014, 79, M941–M946.
More
Information
Subjects: Cell Biology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 438
Revisions: 2 times (View History)
Update Date: 02 Jul 2021
1000/1000