Generic placeholder image

Letters in Drug Design & Discovery

Editor-in-Chief

ISSN (Print): 1570-1808
ISSN (Online): 1875-628X

Perspective Article

Chitosan: A Promising Protective Component Against SARS-CoV-2 and Influenza Virus

Author(s): Mohammad Reza Rahbar, Hadi Esmaeili Gouvarchin Galeh, Saeed Khalili and Abolfazl Jahangiri*

Volume 18, Issue 5, 2021

Published on: 10 November, 2020

Page: [418 - 421] Pages: 4

DOI: 10.2174/1570180817999201110114446

Abstract

The recent pandemic caused by a novel coronavirus known as SARS-CoV-2 has caught the international community by surprise. There is still no effective vaccine or treatment option against this virus. In this perspective, we discussed the potential protective and therapeutic effects of chitosan, as an FDA-approved biomolecule, against COVID-19 and influenza viruses.

Keywords: Chitosan, COVID-19, immune response, innate Immunity, biotherapeutic, macromolecule.

[1]
Kaur, S.P.; Gupta, V. COVID-19 Vaccine: A comprehensive status report. Virus Res., 2020, 288.
[http://dx.doi.org/10.1016/j.virusres.2020.198114] [PMID: 32800805]
[2]
Zhu, F-C.; Guan, X-H.; Li, Y-H.; Huang, J-Y.; Jiang, T.; Hou, L-H.; Li, J.X.; Yang, B.F.; Wang, L.; Wang, W.J.; Wu, S.P.; Wang, Z.; Wu, X.H.; Xu, J.J.; Zhang, Z.; Jia, S.Y.; Wang, B.S.; Hu, Y.; Liu, J.J.; Zhang, J.; Qian, X.A.; Li, Q.; Pan, H.X.; Jiang, H.D.; Deng, P.; Gou, J.B.; Wang, X.W.; Wang, X.H.; Chen, W. Immunogenicity and safety of a recombinant adenovirus type-5-vectored COVID-19 vaccine in healthy adults aged 18 years or older: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet, 2020, 396(10249), 479-488.
[http://dx.doi.org/10.1016/S0140-6736(20)31605-6] [PMID: 32702299]
[3]
Hashemi, S.A.; Safamanesh, S.; Ghafouri, M.; Taghavi, M.R.; Mohajer Zadeh Heydari, M.S.; Namdar Ahmadabad, H.; Ghasemzadeh-Moghaddam, H.; Azimian, A. Co-infection with COVID-19 and influenza A virus in two died patients with acute respiratory syndrome, Bojnurd, Iran. J. Med. Virol., 2020, 92(11), 2319-2321.
[http://dx.doi.org/10.1002/jmv.26014] [PMID: 32410338]
[4]
Cuadrado-Payán, E.; Montagud-Marrahi, E.; Torres-Elorza, M.; Bodro, M.; Blasco, M.; Poch, E.; Soriano, A.; Piñeiro, G.J. SARS-CoV-2 and influenza virus co-infection. Lancet, 2020, 395(10236), e84.
[http://dx.doi.org/10.1016/S0140-6736(20)31052-7] [PMID: 32423586]
[5]
Geoghegan, S.; O’Callaghan, K.P.; Offit, P.A. Vaccine Safety: Myths and Misinformation. Front. Microbiol., 2020, 11, 372.
[http://dx.doi.org/10.3389/fmicb.2020.00372] [PMID: 32256465]
[6]
Wang, B.; Shao, X.; Wang, D.; Xu, D.; Zhang, J.A. Vaccinations and risk of systemic lupus erythematosus and rheumatoid arthritis: A systematic review and meta-analysis. Autoimmun. Rev., 2017, 16(7), 756-765.
[http://dx.doi.org/10.1016/j.autrev.2017.05.012] [PMID: 28483543]
[7]
Shi, Y.; Wang, Y.; Shao, C.; Huang, J.; Gan, J.; Huang, X. COVID-19 infection: the perspectives on immune responses; Nature Publishing Group, 2020.
[8]
Periayah, M.H.; Halim, A.S.; Saad, A.Z.M. Chitosan: A promising marine polysaccharide for biomedical research. Pharmacogn. Rev., 2016, 10(19), 39-42.
[http://dx.doi.org/10.4103/0973-7847.176545] [PMID: 27041872]
[9]
Peluso, G.; Petillo, O.; Ranieri, M.; Santin, M.; Ambrosio, L.; Calabró, D.; Avallone, B.; Balsamo, G. Chitosan-mediated stimulation of macrophage function. Biomaterials, 1994, 15(15), 1215-1220.
[http://dx.doi.org/10.1016/0142-9612(94)90272-0] [PMID: 7703317]
[10]
Knorr, D. Use of chitinous polymers in food: a challenge for food research and development; Food Technology: USA, 1984.
[11]
Kurita, K. Chemistry and application of chitin and chitosan. Polym. Degrad. Stabil., 1998, 59, 117-120.
[http://dx.doi.org/10.1016/S0141-3910(97)00160-2]
[12]
Razdan, A.; Pettersson, D. Effect of chitin and chitosan on nutrient digestibility and plasma lipid concentrations in broiler chickens. Br. J. Nutr., 1994, 72(2), 277-288.
[http://dx.doi.org/10.1079/BJN19940029] [PMID: 7947645]
[13]
Croisier, F.; Jérôme, C. Chitosan-based biomaterials for tissue engineering. Eur. Polym. J., 2013, 49, 780-792.
[http://dx.doi.org/10.1016/j.eurpolymj.2012.12.009]
[14]
Shariatinia, Z. Pharmaceutical applications of chitosan. Adv. Colloid Interface Sci., 2019, 263, 131-194.
[http://dx.doi.org/10.1016/j.cis.2018.11.008] [PMID: 30530176]
[15]
Muzzarelli, R.; Mattioli-Belmonte, M.; Muzzarelli, B. Advances in Chitin. SciLyon. Andru., 1998, 2, 219-224.
[16]
Papineau, A.M.; Hoover, D.G.; Knorr, D.; Farkas, D.F. Antimicrobial effect of water‐soluble chitosans with high hydrostatic pressure. Food Biotechnol., 1991, 5, 45-57.
[http://dx.doi.org/10.1080/08905439109549790]
[17]
Andres, Y.; Giraud, L.; Gerente, C.; Le Cloirec, P. Antibacterial effects of chitosan powder: mechanisms of action. Environ. Technol., 2007, 28(12), 1357-1363.
[http://dx.doi.org/10.1080/09593332808618893] [PMID: 18341146]
[18]
Moran, H.B.T.; Turley, J.L.; Andersson, M.; Lavelle, E.C. Immunomodulatory properties of chitosan polymers. Biomaterials, 2018, 184, 1-9.
[http://dx.doi.org/10.1016/j.biomaterials.2018.08.054] [PMID: 30195140]
[19]
Iriti, M.; Varoni, E.M. Chitosan-induced antiviral activity and innate immunity in plants. Environ. Sci. Pollut. Res. Int., 2015, 22(4), 2935-2944.
[http://dx.doi.org/10.1007/s11356-014-3571-7] [PMID: 25226839]
[20]
Sun, Q.S.; Zhang, J.L.; Han, D.Q.; Yang, Y.B.; Zhu, L.; Yu, L. Characterization and immunological evaluation of chitosan nanoparticles as adjuvants for bovine coronavirus N protein; Publ, T., Ed.; Applied Mechanics and Materials, 2012, pp. 113-120.
[21]
Carroll, E.C.; Jin, L.; Mori, A.; Muñoz-Wolf, N.; Oleszycka, E.; Moran, H.B.T.; Mansouri, S.; McEntee, C.P.; Lambe, E.; Agger, E.M.; Andersen, P.; Cunningham, C.; Hertzog, P.; Fitzgerald, K.A.; Bowie, A.G.; Lavelle, E.C. The vaccine adjuvant chitosan promotes cellular immunity via DNA sensor cGAS-STING-dependent induction of type I interferons. Immunity, 2016, 44(3), 597-608.
[http://dx.doi.org/10.1016/j.immuni.2016.02.004] [PMID: 26944200]
[22]
Yang, Y.; Xing, R.; Liu, S.; Qin, Y.; Li, K.; Yu, H.; Li, P. Chitosan, hydroxypropyltrimethyl ammonium chloride chitosan and sulfated chitosan nanoparticles as adjuvants for inactivated Newcastle disease vaccine. Carbohydr. Polym., 2020, 229, 115423.
[http://dx.doi.org/10.1016/j.carbpol.2019.115423] [PMID: 31826462]
[23]
AbdelAllah, N.H.; Gaber, Y.; Rashed, M.E.; Azmy, A.F.; Abou-Taleb, H.A.; AbdelGhani, S. Alginate-coated chitosan nanoparticles act as effective adjuvant for hepatitis A vaccine in mice. Int. J. Biol. Macromol., 2020, 152, 904-912.
[http://dx.doi.org/10.1016/j.ijbiomac.2020.02.287] [PMID: 32114177]
[24]
Eslam, E.D.; Astaneh, S.D.A.; Rasooli, I.; Nazarian, S.; Jahangiri, A. Passive immunization with chitosan-loaded biofilm-associated protein against Acinetobacter Baumannii murine infection model. Gene Rep., 2020, 20, 100708.
[http://dx.doi.org/10.1016/j.genrep.2020.100708]
[25]
Shibata, Y.; Foster, L.A.; Metzger, W.J.; Myrvik, Q.N. Alveolar macrophage priming by intravenous administration of chitin particles, polymers of N-acetyl-D-glucosamine, in mice. Infect. Immun., 1997, 65(5), 1734-1741.
[http://dx.doi.org/10.1128/IAI.65.5.1734-1741.1997] [PMID: 9125555]
[26]
Pyrc, K.; Milewska, A.; Nowakowska, M.; Szczubialka, K.; Kaminski, K. The use of chitosan polymer in the treatment and prevention of infections caused by coronaviruses; Google Patents, 2015.
[27]
Alitongbieke, G.; Li, X-M.; Wu, Q-C.; Lin, Z-C.; Huang, J-F.; Xue, Y. Study on β-Chitosan against the binding of SARS-CoV-2S-RBD/ACE2. bioRxiv, 2020.
[28]
Zheng, M.; Qu, D.; Wang, H.; Sun, Z.; Liu, X.; Chen, J.; Li, C.; Li, X.; Chen, Z. Intranasal administration of chitosan against influenza A (H7N9) virus infection in a mouse model. Sci. Rep., 2016, 6, 28729.
[http://dx.doi.org/10.1038/srep28729] [PMID: 27353250]
[29]
Wen, Z-S.; Xu, Y-L.; Zou, X-T.; Xu, Z-R. Chitosan nanoparticles act as an adjuvant to promote both Th1 and Th2 immune responses induced by ovalbumin in mice. Mar. Drugs, 2011, 9(6), 1038-1055.
[http://dx.doi.org/10.3390/md9061038] [PMID: 21747747]
[30]
Sheppard, P.; Kindsvogel, W.; Xu, W.; Henderson, K.; Schlutsmeyer, S.; Whitmore, T.E.; Kuestner, R.; Garrigues, U.; Birks, C.; Roraback, J.; Ostrander, C.; Dong, D.; Shin, J.; Presnell, S.; Fox, B.; Haldeman, B.; Cooper, E.; Taft, D.; Gilbert, T.; Grant, F.J.; Tackett, M.; Krivan, W.; McKnight, G.; Clegg, C.; Foster, D.; Klucher, K.M. IL-28, IL-29 and their class II cytokine receptor IL-28R. Nat. Immunol., 2003, 4(1), 63-68.
[http://dx.doi.org/10.1038/ni873] [PMID: 12469119]
[31]
Isorce, N.; Testoni, B.; Locatelli, M.; Fresquet, J.; Rivoire, M.; Luangsay, S.; Zoulim, F.; Durantel, D. Antiviral activity of various interferons and pro-inflammatory cytokines in non-transformed cultured hepatocytes infected with hepatitis B virus. Antiviral Res., 2016, 130, 36-45.
[http://dx.doi.org/10.1016/j.antiviral.2016.03.008] [PMID: 26971407]
[32]
Kang, S.; Brown, H.M.; Hwang, S. Direct antiviral mechanisms of interferon-gamma. Immune Netw., 2018, 18(5), e33.
[http://dx.doi.org/10.4110/in.2018.18.e33] [PMID: 30402328]
[33]
Zhao, J.; Zhao, J.; Mangalam, A.K.; Channappanavar, R.; Fett, C.; Meyerholz, D.K.; Agnihothram, S.; Baric, R.S.; David, C.S.; Perlman, S. Airway memory CD4+ T cells mediate protective immunity against emerging respiratory coronaviruses. Immunity, 2016, 44(6), 1379-1391.
[http://dx.doi.org/10.1016/j.immuni.2016.05.006] [PMID: 27287409]
[34]
Damjanovic, D.; Divangahi, M.; Kugathasan, K.; Small, C-L.; Zganiacz, A.; Brown, E.G.; Hogaboam, C.M.; Gauldie, J.; Xing, Z. Negative regulation of lung inflammation and immunopathology by TNF-α during acute influenza infection. Am. J. Pathol., 2011, 179(6), 2963-2976.
[http://dx.doi.org/10.1016/j.ajpath.2011.09.003] [PMID: 22001698]
[35]
Scheller, J.; Chalaris, A.; Schmidt-Arras, D.; Rose-John, S. The pro-and anti-inflammatory properties of the cytokine interleukin-6. Biochimica et Biophysica Acta (BBA)-. Molecular Cell Res., 2011, 1813, 878-888.
[PMID: 21296109]
[36]
Velazquez-Salinas, L.; Verdugo-Rodriguez, A.; Rodriguez, L.L.; Borca, M.V. The role of interleukin 6 during viral infections. Front. Microbiol., 2019, 10, 1057.
[http://dx.doi.org/10.3389/fmicb.2019.01057] [PMID: 31134045]
[37]
Hokeness, K.L.; Kuziel, W.A.; Biron, C.A.; Salazar-Mather, T.P. Monocyte chemoattractant protein-1 and CCR2 interactions are required for IFN-α/β-induced inflammatory responses and antiviral defense in liver. J. Immunol., 2005, 174(3), 1549-1556.
[http://dx.doi.org/10.4049/jimmunol.174.3.1549] [PMID: 15661915]
[38]
Sabnis, S.; Rege, P.; Block, L.H. Use of chitosan in compressed tablets of diclofenac sodium: inhibition of drug release in an acidic environment. Pharm. Dev. Technol., 1997, 2(3), 243-255.
[http://dx.doi.org/10.3109/10837459709031444] [PMID: 9552452]
[39]
Lehr, C-M.; Bouwstra, J.A.; Schacht, E.H.; Junginger, H.E. In vitro evaluation of mucoadhesive properties of chitosan and some other natural polymers. Int. J. Pharm., 1992, 78, 43-48.
[http://dx.doi.org/10.1016/0378-5173(92)90353-4]
[40]
Kotzé, A.; De Leeuw, B.; Lueßen, H.; De Boer, A.; Verhoef, J.; Junginger, H. Chitosans for enhanced delivery of therapeutic peptides across intestinal epithelia: In vitro evaluation in Caco-2 cell monolayers. Int. J. Pharm., 1997, 159, 243-253.
[http://dx.doi.org/10.1016/S0378-5173(97)00287-1]
[41]
Schipper, N.G.; Vårum, K.M.; Artursson, P. Chitosans as absorption enhancers for poorly absorbable drugs. 1: Influence of molecular weight and degree of acetylation on drug transport across human intestinal epithelial (Caco-2) cells. Pharm. Res., 1996, 13(11), 1686-1692.
[http://dx.doi.org/10.1023/A:1016444808000] [PMID: 8956335]
[42]
Keefe, D. Agency Response Letter GRAS Notice No. GRN000424. FDA ucm335743, 2012.
[43]
Abdel-Rahman, A.; Anyangwe, N.; Carlacci, L.; Casper, S.; Danam, R.P.; Enongene, E.; Erives, G.; Fabricant, D.; Gudi, R.; Hilmas, C.J.; Hines, F.; Howard, P.; Levy, D.; Lin, Y.; Moore, R.J.; Pfeiler, E.; Thurmond, T.S.; Turujman, S.; Walker, N.J. The safety and regulation of natural products used as foods and food ingredients. Toxicol. Sci., 2011, 123(2), 333-348.
[http://dx.doi.org/10.1093/toxsci/kfr198] [PMID: 21821733]
[44]
Gades, M.D.; Stern, J.S. Chitosan supplementation and fecal fat excretion in men. Obes. Res., 2003, 11(5), 683-688.
[http://dx.doi.org/10.1038/oby.2003.97] [PMID: 12740459]
[45]
Tapola, N.S.; Lyyra, M.L.; Kolehmainen, R.M.; Sarkkinen, E.S.; Schauss, A.G. Safety aspects and cholesterol-lowering efficacy of chitosan tablets. J. Am. Coll. Nutr., 2008, 27(1), 22-30.
[http://dx.doi.org/10.1080/07315724.2008.10719671] [PMID: 18460478]
[46]
Ylitalo, R.; Lehtinen, S.; Wuolijoki, E.; Ylitalo, P.; Lehtimäki, T. Cholesterol-lowering properties and safety of chitosan. Arzneimittelforschung, 2002, 52(1), 1-7.
[PMID: 11838268]
[47]
Mohammed, M.A.; Syeda, J.T.M.; Wasan, K.M.; Wasan, E.K. An overview of chitosan nanoparticles and its application in non-parenteral drug delivery. Pharmaceutics, 2017, 9(4), 53.
[http://dx.doi.org/10.3390/pharmaceutics9040053] [PMID: 29156634]

© 2024 Bentham Science Publishers | Privacy Policy