323
Views
27
CrossRef citations to date
0
Altmetric
Original Research

Hepatitis C Virus NS3 Protease and Helicase Inhibitors from Red Sea Sponge (Amphimedon) Species in Green Synthesized Silver Nanoparticles Assisted by in Silico Modeling and Metabolic Profiling

, ORCID Icon, , , ORCID Icon, , , ORCID Icon, , & show all
Pages 3377-3389 | Published online: 12 May 2020

References

  • Zhao T, Sun R, Yu S, et al. Size-controlled preparation of silver nanoparticles by a modified polyol method. COLLOID SURF a PHYSICOCHEM ENG ASP. 2010;366(1–3):197–202. doi:10.1016/j.colsurfa.2010.06.005
  • Sun Y, Mayers B, Herricks T, Xia Y. Polyol synthesis of uniform silver nanowires: a plausible growth mechanism and the supporting evidence. Nano Lett. 2003;3(7):955–960. doi:10.1021/nl034312m
  • Du W-L, Niu -S-S, Xu Y-L, Xu Z-R, Fan C-L. Antibacterial activity of chitosan tripolyphosphate nanoparticles loaded with various metal ions. Carbohydr. 2009;75(3):385–389. doi:10.1016/j.carbpol.2008.07.039
  • Sinha S, Pan I, Chanda P, Sen SK. Nanoparticles fabrication using ambient biological resources. J Appl Biosci. 2009;19:1113–1130.
  • Amin M, Anwar F, Janjua MRSA, Iqbal MA, Rashid U. Green synthesis of silver nanoparticles through reduction with solanum xanthocarpum L. berry extract: characterization, antimicrobial and urease inhibitory activities against helicobacter pylori. Int J Mol Sci. 2012;13(8):9923–9941. doi:10.3390/ijms1308992322949839
  • Pastoriza-Santos I, Liz-Marzán LM. Formation of PVP-protected metal nanoparticles in DMF. Langmuir. 2002;18(7):2888–2894. doi:10.1021/la015578g
  • Zou J, Zhang F, Huang J, Chang PR, Su Z, Yu J. Effects of starch nanocrystals on structure and properties of waterborne polyurethane-based composites. Carbohydr. 2011;85(4):824–831. doi:10.1016/j.carbpol.2011.04.006
  • Mondal AK, Mondal S, Samanta S, Mallick S. Synthesis of ecofriendly silver nanoparticle from plant latex used as an important taxonomic tool for phylogenetic interrelationship advances in bioresearch vol. 2. Synthesis. 2011;31:33.
  • Sekhar EC, Rao K, Rao KMS, Alisha SB. A simple biosynthesis of silver nanoparticles from syzygium cumini stem bark aqueous extract and their spectrochemical and antimicrobial studies. J Appl Pharm. 2018;8(01):073–079.
  • Singh M, Kalaivani R, Manikandan S, Sangeetha N, Kumaraguru AK. Facile green synthesis of variable metallic gold nanoparticle using Padina gymnospora, a brown marine macroalga. Appl Nanoscience. 2013;3(2):145–151. doi:10.1007/s13204-012-0115-7
  • Haggag EG, Elshamy AM, Rabeh MA, et al. Antiviral potential of green synthesized silver nanoparticles of lampranthus coccineus and malephora lutea. Int J Nanomedicine. 2019;14:6217–6229. doi:10.2147/IJN.S21417131496682
  • El-Nour KMA, Eftaiha A, Al-Warthan A, Ammar RA. Synthesis and applications of silver nanoparticles. Arab J Chem. 2010;3(3):135–140. doi:10.1016/j.arabjc.2010.04.008
  • Shady NH, Fouad MA, Ahmed S, et al. A new antitrypanosomal alkaloid from the Red Sea marine sponge Hyrtios sp. J Antibiot (Tokyo). 2018;71(12):1036–1039. doi:10.1038/s41429-018-0092-530181571
  • Srividhya S, Chellaram C. Role of marine life in nanomedicine. Ind J Innov Develop. 2012;1(S8):31–33.
  • Kulkarni SR, Dikshit M. Indian marine pharmacology: a sneak peek into the ecosystem. Proc Indian Natl Sci Acad B. 2018;84(1):281–300.
  • Arya G, Sharma N, Mankamna R, Nimesh S. Antimicrobial silver nanoparticles: future of nanomaterials In: Microbial Nanobionics. Springer 2019;89–119.
  • El-Gaied HAAA. Antiviral evaluation of secondary metabolites derived from actinomycetes conjugated to silver nanoparticles. CU Theses. 2018.
  • Ryoo S-R, Jang H, Kim K-S, et al. Functional delivery of DNAzyme with iron oxide nanoparticles for hepatitis C virus gene knockdown. Biomaterials. 2012;33(9):2754–2761. doi:10.1016/j.biomaterials.2011.12.01522206595
  • Schiering N, D’Arcy A, Villard F, et al. A macrocyclic HCV NS3/4A protease inhibitor interacts with protease and helicase residues in the complex with its full-length target. PNAS. 2011;108(52):21052–21056. doi:10.1073/pnas.111053410822160684
  • Lauer GM, Walker BD. Hepatitis C virus infection. N Engl J Med. 2001;345(1):41–52. doi:10.1056/NEJM20010705345010711439948
  • Chlibek R, Smetana J, Sosovickova R, et al. Prevalence of hepatitis C virus in adult population in the Czech Republic - time for birth cohort screening. PLoS One. 2017;12(4):e0175525–e0175525. doi:10.1371/journal.pone.017552528406947
  • Licata A, Minissale MG, Montalto FA, Soresi M. Is vitamin D deficiency predictor of complications development in patients with HCV-related cirrhosis? Intern Emerg Med. 2019;14(5):735–737. doi:10.1007/s11739-019-02072-w30879232
  • Gaballah AM, Esawy MM. Comparison of 2 different antibody assay methods, Elecsys Anti-HCVII (Roche) and Vidas Anti-HCV (Biomerieux), for the detection of antibody to hepatitis C virus in Egypt. Diagn Microbiol Infect Dis. 2018;92(2):107–111. doi:10.1016/j.diagmicrobio.2018.05.01329925467
  • Fujimoto Y, Salam KA, Furuta A, et al. Inhibition of both protease and helicase activities of hepatitis C virus NS3 by an ethyl acetate extract of marine sponge Amphimedon sp. PLoS One. 2012;7(11):e48685. doi:10.1371/journal.pone.004868523144928
  • Moriishi K, Matsuura Y. Exploitation of lipid components by viral and host proteins for hepatitis C virus infection. Front Microbiol. 2012;3:54. doi:10.3389/fmicb.2012.0005422347882
  • Hong TT, Dat TTH, Cuc NTK, Cuong PV. Mini-review protease inhibitor (PI) and Pis from sponge-associated microorganisms. Vietnam J Sci Technol. 2018;56(4):405. doi:10.15625/2525-2518/56/4/10911
  • Belon CA, Frick DN. Helicase Inhibitors as Specifically Targeted Antiviral Therapy for Hepatitis C. 2009.
  • Jensen DM. A new era of hepatitis C therapy begins. N Engl J Med. 2011;364(13):1272–1274. doi:10.1056/NEJMe110082921449791
  • Li B, Li L, Peng Z, et al. Harzianoic acids A and B, new natural scaffolds with inhibitory effects against hepatitis C virus. Bioorg Med Chem. 2019;27(3):560–567. doi:10.1016/j.bmc.2018.12.03830606673
  • Lange CM, Sarrazin C, Zeuzem S. specifically targeted anti‐viral therapy for hepatitis C–a new era in therapy. Aliment Pharmacol Ther. 2010;32(1):14–28. doi:10.1111/j.1365-2036.2010.04317.x20374226
  • Feld JJ, Hoofnagle JH. Mechanism of action of interferon and ribavirin in treatment of hepatitis C. Nature. 2005;436(7053):967. doi:10.1038/nature0408216107837
  • Kjellin M, Wesslén T, Löfblad E, Lennerstrand J, Lannergård A. The effect of the first-generation HCV-protease inhibitors boceprevir and telaprevir and the relation to baseline NS3 resistance mutations in genotype 1: experience from a small Swedish cohort. Ups J Med Sci. 2018;123(1):50–56. doi:10.1080/03009734.2018.144192829536805
  • Chen C, Qiu H, Gong J, et al. (−)-Epigallocatechin-3-gallate inhibits the replication cycle of hepatitis C virus. Arch Virol. 2012;157(7):1301–1312. doi:10.1007/s00705-012-1304-022491814
  • Gonzalez O, Fontanes V, Raychaudhuri S, et al. The heat shock protein inhibitor Quercetin attenuates hepatitis C virus production. Hepatology. 2009;50(6):1756–1764. doi:10.1002/hep.2323219839005
  • Bachmetov L, Gal‐Tanamy M, Shapira A, et al. Suppression of hepatitis C virus by the flavonoid quercetin is mediated by inhibition of NS3 protease activity. J Viral Hepatitis. 2012;19(2):e81–e88. doi:10.1111/j.1365-2893.2011.01507.x
  • Li Y, Yu S, Liu D, Proksch P, Lin W. Inhibitory effects of polyphenols toward HCV from the mangrove plant Excoecaria agallocha L. Bioorg Med Chem Lett. 2012;22(2):1099–1102. doi:10.1016/j.bmcl.2011.11.10922196120
  • Sahuc M-E, Sahli R, Rivière C, et al. Dehydrojuncusol, a natural phenanthrene compound extracted from juncus maritimus is a new inhibitor of hepatitis C virus RNA replication. J Virol. 2019;JVI:02009–02018.
  • Sepčić K, Kauferstein S, Mebs D, Turk T. Biological activities of aqueous and organic extracts from tropical marine sponges. Mar Drugs. 2010;8(5):1550. doi:10.3390/md805155020559488
  • Shady N, El-Hossary E, Fouad M, Gulder T, Kamel M, Abdelmohsen U. Bioactive natural products of marine sponges from the genus Hyrtios. Molecules. 2017;22(5):781. doi:10.3390/molecules22050781
  • Abdelmohsen UR, Balasubramanian S, Oelschlaeger TA, et al. Potential of marine natural products against drug-resistant fungal, viral, and parasitic infections. Lancet Infect Dis. 2017;17(2):e30–e41. doi:10.1016/S1473-3099(16)30323-127979695
  • Liu M, El-Hossary EM, Oelschlaeger TA, Donia MS, Quinn RJ, Abdelmohsen UR. Potential of marine natural products against drug-resistant bacterial infections. Lancet Infect Dis. 2019.
  • Ahmed EF, Rateb ME, Abou El-Kassem LT, Hawas UW. Anti-HCV protease of diketopiperazines produced by the Red Sea sponge-associated fungus Aspergillus versicolor. Appl Biochem Biotechnol. 2017;53(1):101–106.
  • Na M, Ding Y, Wang B, et al. Anti-infective discorhabdins from a deep-water Alaskan sponge of the genus Latrunculia. Indian J Nat Prod Resour. 2009;73(3):383–387. doi:10.1021/np900281r
  • Cheung RCF, Wong JH, Pan WL, et al. Antifungal and antiviral products of marine organisms. Appl Microbiol Biotechnol. 2014;98(8):3475–3494. doi:10.1007/s00253-014-5575-024562325
  • Hirano K, Kubota T, Tsuda M, Mikami Y, Kobayashi J. Pyrinodemins BD, potent cytotoxic bis-pyridine alkaloids from marine sponge Amphimedon sp. Chem Pharm Bull. 2000;48(7):974–977. doi:10.1248/cpb.48.97410923825
  • Kubota T, Kamijyo Y, Takahashi-Nakaguchi A, Fromont J, Gonoi T, Kobayashi J. Zamamiphidin A, a new manzamine related alkaloid from an Okinawan marine sponge Amphimedon sp. Org Lett. 2013;15(3):610–612. doi:10.1021/ol303427423343122
  • Sakurada T, Gill MB, Frausto S, et al. Novel N-methylated 8-oxoisoguanines from Pacific sponges with diverse neuroactivities. Eur J Med Chem. 2010;53(16):6089–6099. doi:10.1021/jm100490m
  • Ovenden SP, Capon RJ, Lacey E, Gill JH, Friedel T, Wadsworth D. Amphilactams A− D: novel nematocides from Southern Australian Marine sponges of the Genus Amphimedon. JOC. 1999;64(4):1140–1144. doi:10.1021/jo981377e
  • Emura C, Higuchi R, Miyamoto T, Van Soest RW. Amphimelibiosides A− F, six new ceramide dihexosides isolated from a Japanese Marine Sponge Amphimedon sp. JOC. 2005;70(8):3031–3038. doi:10.1021/jo048635u
  • Nemoto T, Yoshino G, Ojika M, Sakagami Y. Amphimic acids and related long-chain fatty acids as DNA topoisomerase I inhibitors from an Australian sponge, Amphimedon sp.: isolation, structure, synthesis, and biological evaluation. Tetrahedron. 1997;53(49):16699–16710. doi:10.1016/S0040-4020(97)10099-0
  • Shady NH, Fouad MA, Salah Kamel M, Schirmeister T, Abdelmohsen UR. Natural product repertoire of the Genus Amphimedon. Mar Drugs. 2018;17(1):19. doi:10.3390/md17010019
  • O’Rourke A. Bioprospecting of Red Sea Sponges for Novel Antiviral Pharmacophores. 2015.
  • Costa FG, BRdS N, Gonçalves RL, et al. Alkaloids as Inhibitors of malate synthase from span class=“named-content genus-species” id=“named-content-1”paracoccidioides span spp.: receptor-ligand interaction-based virtual screening and molecular docking studies, antifungal activity, and the adhesion process. Antimicrob Agents Chemother. 2015;59(9):5581–5594. doi:10.1128/AAC.04711-1426124176
  • Montefiori DC. Evaluating neutralizing antibodies against HIV, SIV, and SHIV in luciferase reporter gene assays. Curr Protoc Immunol. 2004;64(1):12.11.11–12.11. 17. doi:10.1002/0471142735.im1211s64
  • Inc. CCG. Molecular Operating Environment (MOE), 2012.10. 1010 Sherbrooke St.west, Suite #910. Montr. QC, Canada, H3A 2R7; 2012.
  • Nasr T, Bondock S, Eid S. Design, synthesis, antimicrobial evaluation and molecular docking studies of some new thiophene, pyrazole and pyridone derivatives bearing sulfisoxazole moiety. Eur J Med Chem. 2014;84:491–504. doi:10.1016/j.ejmech.2014.07.05225050881
  • Avilala J, Golla N. Antibacterial and antiviral properties of silver nanoparticles synthesized by marine actinomycetes. Int J Pharm Sci & Res. 2019;10(3):1223–1228.
  • Tsuda M, Inaba K, Kawasaki N, Honma K, Kobayashi J. Chiral resolution of (±)-keramaphidin B and isolation of manzamine L, a new β-carboline alkaloid from a sponge Amphimedon sp. Tetrahedron. 1996;52(7):2319–2324. doi:10.1016/0040-4020(95)01057-2
  • Tsuda M, Watanabe D, Kobayashi J. Ma’eganedin A, a new manzamine alkaloid from Amphimedon sponge. Tetrahedron Lett. 1998;39(10):1207–1210. doi:10.1016/S0040-4039(97)10842-5
  • Kobayashi J, Watanabe D, Kawasaki N, Tsuda M. Nakadomarin A, a novel hexacyclic manzamine-related alkaloid from Amphimedon sponge. JOC. 1997;62(26):9236–9239. doi:10.1021/jo9715377
  • Jeong S-J, Inagaki M, Higuchi R, et al. 1, 3-Dimethylisoguaninium, an antiangiogenic purine analog from the sponge amphimedon paraviridis. Chem Pharm Bull. 2003;51(6):731–733. doi:10.1248/cpb.51.73112808256
  • Tsukamoto S, Takahashi M, Matsunaga S, Fusetani N, Van Soest RW. Hachijodines A− G: seven new cytotoxic 3-alkylpyridine alkaloids from two marine sponges of the Genera Xestospongia and Amphimedon. J Nat Prod. 2000;63(5):682–684. doi:10.1021/np990576610843588
  • Nishi T, Kubota T, Fromont J, Sasaki T, Kobayashi J. Nakinadines B–F: new pyridine alkaloids with a β-amino acid moiety from sponge Amphimedon sp. Tetrahedron. 2008;64(14):3127–3132. doi:10.1016/j.tet.2008.01.111
  • Tsuda M, Kawasaki N, Kobayashi J. Ircinols A and B, first antipodes of manzamine-related alkaloids from an Okinawan marine sponge. Tetrahedron. 1994;50(27):7957–7960. doi:10.1016/S0040-4020(01)85280-7
  • Kobayashi J, Tsuda M, Kawasaki N, Sasaki T, Mikami Y. 6-Hydroxymanzamine A and 3, 4-dihydromanzamine A, new alkaloids from the Okinawan marine sponge Amphimedon Sp. J Nat Prod. 1994;57(12):1737–1740. doi:10.1021/np50114a0217714542
  • Watanabe D, Tsuda M, Kobayashi J. Three new manzamine congeners from amphimedon sponge. J Nat Prod. 1998;61(5):689–692. doi:10.1021/np970564p9599281
  • Carballeira NM, Restituyo J. Identification of the new 11, 15-icosadienoic acid and related acids in the sponge Amphimedon complanata. J Nat Prod. 1991;54(1):315–317. doi:10.1021/np50073a0432045822
  • Carballeira NM, Colón R, Emiliano A. Identification of 2-methoxyhexadecanoic acid in Amphimedon compressa. J Nat Prod. 1998;61(5):675–676. doi:10.1021/np970578v9599277
  • Carballeira NM, Negrón V, Reyes ED. Novel monounsaturated fatty acids from the sponges Amphimedon compressa and Mycale laevis. J Nat Prod. 1992;55(3):333–339. doi:10.1021/np50081a009
  • Takekawa Y, Matsunaga S, van Soest RW, Fusetani N. Amphimedosides, 3-alkylpyridine glycosides from a marine sponge Amphimedon sp. J Nat Prod. 2006;69(10):1503–1505. doi:10.1021/np060122q17067172
  • Kobayashi J, Tsuda M, Kawasaki N, Matsumoto K, Adachi T. Keramaphidin B, a novel pentacyclic alkaloid from a marine sponge Amphimedon sp.: a plausible biogenetic precursor of manzamine alkaloids. Tetrahedron Lett. 1994;35(25):4383–4386. doi:10.1016/S0040-4039(00)73362-4
  • Schmitz FJ, Agarwal SK, Gunasekera SP, Schmidt PG, Shoolery JN. Amphimedine, new aromatic alkaloid from a pacific sponge, Amphimedon sp. carbon connectivity determination from natural abundance carbon-13-carbon-13 coupling constants. J Am Chem Soc. 1983;105(14):4835–4836. doi:10.1021/ja00352a052
  • Beran RK, Serebrov V, Pyle AM. The serine protease domain of hepatitis C viral NS3 activates RNA helicase activity by promoting the binding of RNA substrate. J Biol Chem. 2007;282(48):34913–34920. doi:10.1074/jbc.M70716520017921146
  • Beran RK, Pyle AM. Hepatitis C viral NS3-4A protease activity is enhanced by the NS3 helicase. J Biol Chem. 2008;283(44):29929–29937. doi:10.1074/jbc.M80406520018723512
  • Morgenstern KA, Landro JA, Hsiao K, et al. Polynucleotide modulation of the protease, nucleoside triphosphatase, and helicase activities of a hepatitis C virus NS3-NS4A complex isolated from transfected COS cells. J Virol. 1997;71(5):3767–3775. doi:10.1128/JVI.71.5.3767-3775.19979094652
  • Lipinski CA. Lead-and drug-like compounds: the rule-of-five revolution. Drug Discov Today Technol. 2004;1(4):337–341. doi:10.1016/j.ddtec.2004.11.00724981612
  • Ebejer J-P, Charlton MH, Finn PW. Are the physicochemical properties of antibacterial compounds really different from other drugs? J Cheminform. 2016;8(1):30. doi:10.1186/s13321-016-0143-527274770