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 -- 1605 2023-08-17 07:34:15 |
2 format correct -1 word(s) 1604 2023-08-17 12:09:10 |

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.
Rohilla, D.; Srivastava, A.K.; Singh, R.P.; Yadav, P.; Singh, S.K.; Kumar, D.; Bhardwaj, N.; Kesawat, M.S.; Pandey, K.D.; Kumar, A. Antiviral Properties of Algal Polysaccharides. Encyclopedia. Available online: https://encyclopedia.pub/entry/48150 (accessed on 01 July 2024).
Rohilla D, Srivastava AK, Singh RP, Yadav P, Singh SK, Kumar D, et al. Antiviral Properties of Algal Polysaccharides. Encyclopedia. Available at: https://encyclopedia.pub/entry/48150. Accessed July 01, 2024.
Rohilla, Dikshansha, Akhileshwar Kumar Srivastava, Rahul Prasad Singh, Priya Yadav, Sandeep Kumar Singh, Dharmendra Kumar, Nikunj Bhardwaj, Mahipal Singh Kesawat, Kapil Deo Pandey, Ajay Kumar. "Antiviral Properties of Algal Polysaccharides" Encyclopedia, https://encyclopedia.pub/entry/48150 (accessed July 01, 2024).
Rohilla, D., Srivastava, A.K., Singh, R.P., Yadav, P., Singh, S.K., Kumar, D., Bhardwaj, N., Kesawat, M.S., Pandey, K.D., & Kumar, A. (2023, August 17). Antiviral Properties of Algal Polysaccharides. In Encyclopedia. https://encyclopedia.pub/entry/48150
Rohilla, Dikshansha, et al. "Antiviral Properties of Algal Polysaccharides." Encyclopedia. Web. 17 August, 2023.
Antiviral Properties of Algal Polysaccharides
Edit

Polysaccharides are a group of either similar or different saccharides that are connected with glycosidic bonds. These polysaccharide molecules inhibit viral replication by interfering in any stage of the viral life cycle, which generally takes place in phases such as the adsorption of the virus by the host cells, penetration into the host cell, uncoating of capsids, assembly and release of viral particles, or via inactivating virions before infection. The life cycle of viruses varies from species to species; thus, the action mechanisms of the algal polysaccharides also varies with the nature of the virus species.

algal polysaccharides alginic acid COVID-19 SARS-CoV-2 antiviral activity

1. Introduction

For the last three years, the world has faced an unexpected pandemic of COVID-19, caused by the novel coronavirus (SARS-CoV-2). The outbreak of this virus affects the lives and survivability of human beings. Although, in the last three years, significant progress has been made by the scientific community in developing vaccines that control the virus spread to a certain extent [1][2]. Although the administration of vaccines enhances the immune response of the body to cope against the infection caused by the ever-changing shape of the mutant RNA virus, the identification of different mutant strains always poses a threat, which necessitates the search for an alternative that can enhance the immune response of the body [2]. In this regard, new experiments have been executed regularly to meet the requirement of a novel, efficient, economic, and nontoxic antiviral candidate.
In last two decades, significant progress has been made with algal metabolites in developing different types of drugs to cure human viral or chronic diseases [3][4][5]. Algae are a diverse group of photosynthetic organisms found in both fresh and marine aquatic systems. The ubiquitous nature and survivability of algae under different harsh conditions, such as hot, cold, and intense light conditions, confirms the presence of some specific metabolites in their cells. These metabolites are a rich source of high-value foods and important pharmaceutical compounds [6]. The beneficial secondary metabolites, such as phycocyanin, polysaccharides, lutein, vitamin B12, vitamin E, vitamin K, polyphenols, polyunsaturated fatty acids, and polysaccharides, are well documented in a previous study [7]. In addition, antimicrobial, anti-inflammatory, anticancer, immunosuppressive, and other pharmacologically significant properties of these secondary metabolites have been investigated [8][9][10].
Algal groups are a rich source of secondary metabolites, which have been synthesized during the different phases of growth stages via various metabolic processes [11][12]. However, the cultivation practices and growth conditions are limiting factors during metabolite production [13][14][15]. Although like other metabolites, algal metabolites also constitute different functional groups, including polyphenols, carotenoids, vitamins, lipids, and polysaccharides, which possess a diverse range of medicinal properties, as documented in the previous study [16][17]. For example, calcium spirulan (Ca-SP), a derivative of Spirulina platensis, is reported to inhibit the replication and penetration into the host cells of different viruses such as herpes simplex virus (HSV) type 1, measles, mumps, influenza A virus, and human immunodeficiency virus HIV-1 [18]. Sharaf et al. [19] evaluated the anti-herpetic activity of Arthrospira fusiformi and found the application of A. fusiformis extract inhibits the multiplication of the herpes virus before and after host infection. Similarly, Silva et al. [20] reported cyanobacterial extract significantly inhibits replication of the influenza virus. In another study, Shih et al. [21] evaluated the potential of allophycocyanin isolated from Spirulina platensis against enterovirus and found it had the potential to delay RNA synthesis in the infected cell. Cyanovirin-N, proteins derived from Nostoc ellipsosporum, showed broad-spectrum activity against HIV [22], hepatitis C [23], and influenza [24]. Lectins, a protein derived from algae, are also considered the largest antiviral chemical class, which generally target glycoproteins of the virus envelope and help to control viruses’ entry into the host cells [25][26]. The most effective antiviral drugs include sulfated polysaccharides, phenolic chemicals, and organic acids [27]. The structural composition of COVID-19 possesses similar structural proteins, and therefore preventive measures can be followed by previous research conducted on algal-derived metabolites [28][29].
In last two decades, several virus outbreaks such as swine influenza, avian influenza, and Ebola have emerged [30][31] and the bioactive chemicals generated from algae have been used as a promising antiviral candidate. Nostoc is the genus within the Nostocales order that produces the most metabolites with antibacterial and antiviral activity. Furthermore, Nostoc sp. can withstand a wide range of environmental circumstances, allowing them to thrive in various environments [32][33]. Different marine microalgal species extracts advocated to have thousands of novel bioactive compounds with therapeutic potential could be exploited to produce therapeutic agents against some common human viral diseases [34][35]. Notwithstanding the Brazilian sea, algal crude extracts have been more effective against HSV-1 than HSV-2 [36]. Furthermore, it is warranted to mention that an aqueous extract of Laurencia obtuse showed significant inhibitory potential against influenza virus replication during an in vitro experiment [37]. Furthermore, Zaid et al. [38] demonstrated inhibition in the multiplication of the Coxsackie B4 virus, hepatitis A virus, HSV-1, and HSV-2 by employing seaweed extracts of Ulva Lactuca and Cystoseira myrica in an in vitro study. Thus, the use of different algal metabolites and products offers a way to combat newly emerged viral diseases [39][40].
In the recent past, in silico approaches have been widely used to screen drugs against various diseases in limited time and with less cost [9]. Computational approaches offer several important tools in each step of drug exploration. Various bioinformatics tools are capable of aiding researchers in the recognition and investigation of new drug compounds. The important parameters of in silico methods include virtual screening, molecular docking studies, molecular dynamics simulations, and the determination of ADMET (absorption, distribution, metabolism, excretion, and toxicity) properties [10]. Hence, the current study based on bioinformatic approaches emphasizes the exploration of the potential antiviral activity of algal polysaccharides alginic acid and carrageenan against SARS-CoV-2 targeting the S-RBD protein.

2. The Potential of Algal Polysaccharides as Antiviral Agents

In the recent past, various polysaccharides, such as carrageenan, alginate, ulvans, and laminarins, have been isolated from both fresh and marine water alga and these polysaccharides have been reported for their potential antiviral properties [41]. These polysaccharides, particularly the sulfated polysaccharides, are conferred with strong polyanionic characteristics and have the ability to block the cationic charge upon the surface of cells in order to prevent virus infection or even adsorption [42]. These sulfated polysaccharides have significant therapeutic potential (Table 1); because they can mimic glycosaminoglycans, sugar-rich molecules that are prevalent in cell membranes [43]. Thus, in this research, algal polysaccharides, carrageenans, and alginates are considered to study their impact on SARS-CoV-2.
Table 1. Some common algal polysaccharides used as antiviral agents and their mechanism of action.
Algal Polysaccharides Source Viral Diseases Mechanism of Actions
Carrageenan Red algae Influenza virus
Human immunodeficiency virus
Herpes simplex virus
The compound inhibits the binding or entry of the virus to the host cell
Alginate Brown algae Human immunodeficiency virus
Hepatitis B Virus
Compounds inhibit adhesion of virus to the host cell and also inhibit replication inside the cell
Fucan Brown algae Human immunodeficiency virus
Herpes simplex virus
By blockage of reverse transcriptase
Agar Red algae Influenza virus By the partial blockage of the adhesion to the endothelial cells
Laminaran Brown algae Human immunodeficiency virus By the blockage of reverse transcriptase
Galactan Red algae Human immunodeficiency virus
Herpes simplex virus
By inhibiting adhesion of the virus to the host cells and inhibition of replication
Ulvans Green algae Human and avian influenza viruses Inhibit viral reproduction
Carrageenans (CGs) are sulfated linear polysaccharides that alternately contain (1→3)-β-D-galactopyranoses and (1→4)-α-D-galactopyranoses substituted with sulfate esters at the several positions [44]. The position of sulfation determines the active properties of the carrageenans. ICGs have been previously well reported in various algal groups, such as Agardhiella, Eucheuma, Chondrus crispus, Furcellaria, Hypnea, Iridaea, Gigartina, Solieria, and Sarconema, accounting for 30–75% of their dry weight [45][46][47]. The multifunctional qualities of the CGs, such as biocompatibility, the absence of toxic effects, biodegradable nature, and emulsifying, gelling, and stabilizing abilities, make them popular in the food, pharmaceutical and cosmetic industries [48]. The presence of antioxidant, antiviral, anti-cancerous, and excellent drug-transport properties make CGs a suitable or multipurpose agent for the pharmaceutical industry [46].
In previous studies, various authors have reported the pharmaceutical properties of CGs against different viral diseases such as the herpes virus types 1 and 2 [49][50], varicella zoster virus [51], cytomegalovirus [52], HIV [53], human metapneumovirus [54], and influenza virus (IAV) [55]. CG has reportedly been found to be most efficient against enveloped viruses. The antiviral effect of the CG structures help in attaching the virus to its receptor [56][57]. In the latest studies, carrageenans such as ι and λ showed strong inhibitory effects against the SARS-CoV-2 virus [58][59][60]. Similarly, Song et al. [61] reported the ability of sulfated polysaccharides, ι -CG, to prevent the binding and penetration of SARS-CoV-2 into host cells.
Alginates, the salts of alginic acid, are polysaccharides comprising 13-(1→4)-D-mannosyluronic acid (M), o~-(1→4)-L-glucosyluronic acid (G), and alternating (MG) blocks [62]. In previously published reports, extraction of alginic acid from various algal groups such as Laminaria, Mcrocystis, and Ascophyllum was well documented [63][64]. Like the CGs, alginates have some specific characteristics such as absence of toxic effect and biocompatible nature, which also make them popular in medical science [47]. Alginates have been used generally in the pharmaceutical industry for microencapsulation, wound dressing, and drug delivery [65]. Numerous authors have reported the use of alginic acid in the treatment of various human viral diseases. For example, Mastromarino et al. [66] reported the antiviral activity of alginic acid against the Vero cells of the enveloped group IV rubella virus. Similarly, Bandyopadhyay et al. [67] reported the antiviral activity of alginate hydrogels against the HSV type-1. Sinha et al. [68] reported an anti-HSV-1 effect of sulfonated alginate, and found that antiviral activity enhanced with increasing sulfate ester content. In addition, different authors have reported the anti-HIV-1 and anti-HSV-1 properties of alginic acid [69][70]. These studies provide a primary clue to the use of these algal polysaccharides in the treatment and prevention SARS-CoV-2.

References

  1. Kumar, A.; Singh, R.P.; Kumar, I.; Yadav, P.; Singh, S.K.; Kaushalendra; Singh, P.K.; Gupta, R.K.; Singh, S.M.; Kesawat, M.S.; et al. Algal Metabolites Can Be an Immune Booster against COVID-19 Pandemic. Antioxidants 2022, 11, 452.
  2. Prabhu, S.; Vijayakumar, S.; Praseetha, P. Cyanobacterial metabolites as novel drug candidates in corona viral therapies: A review. Chronic. Dis. Transl. Med. 2022, 8, 172–183.
  3. Silva, S.C.; Ferreira, I.C.F.R.; Dias, M.M.; Barreiro, M.F. Microalgae-Derived Pigments: A 10-Year Bibliometric Review and Industry and Market Trend Analysis. Molecules 2020, 25, 3406.
  4. Chakdar, H.; Jadhav, S.D.; Dhar, D.W.; Pabbi, S. Potential applications of blue green algae. J. Sci. Ind. Res. 2012, 71, 13–20.
  5. Novoveská, L.; Ross, M.E.; Stanley, M.S.; Pradelles, R.; Wasiolek, V.; Sassi, J.F. Microalgal carotenoids: A review of production, current markets, regulations, and future direction. Mar. Drugs 2019, 17, 640.
  6. Chin, Y.W.; Balunas, M.J.; Chai, H.B.; Kinghorn, A.D. Drug discovery from natural sources. AAPS J. 2006, 8, E239–E253.
  7. Ibrahim, T.N.B.T.; Feisal, N.A.S.; Kamaludin, N.H.; Cheah, W.Y.; How, V.; Bhatnagar, A.; Ma, Z.; Show, P.L. Biological active metabolites from microalgae for healthcare and pharmaceutical industries: A comprehensive review. Bioresour. Technol. 2023, 21, 128661.
  8. Yeo, C.; Kaushal, S.; Yeo, D. Enteric involvement of coronaviruses: Is faecal-oral transmission of SARS-CoV-2 possible? Lancet Gastroenterol. Hepatol. 2020, 5, 335–337.
  9. Dini, I. The Potential of Algae in the Nutricosmetic Sector. Molecules 2023, 28, 4032.
  10. Brogi, S. Computational Approaches for Drug Discovery. Molecules 2019, 24, 3061.
  11. Kaur, M.; Bhatia, S.; Gupta, U.; Decker, E.; Tak, Y.; Bali, M.; Gupta, V.K.; Dar, R.A.; Bala, S. Microalgal bioactive metabolites as promising implements in nutraceuticals and pharmaceuticals: Inspiring therapy for health benefits. Phytochem. Rev. 2023, 14, 1–31.
  12. Perumal, P.K.; Dong, C.D.; Chauhan, A.S.; Anisha, G.S.; Kadri, M.S.; Chen, C.W.; Singhania, R.R.; Patel, A.K. Advances in oligosaccharides production from algal sources and potential applications. Biotechnol. Adv. 2023, 67, 108195.
  13. Kumari, A.; GarimaBharadvaja, N. A comprehensive review on algal nutraceuticals as prospective therapeutic agent for different diseases. 3 Biotech 2023, 13, 44.
  14. Dewi, I.C.; Falaise, C.; Hellio, C.; Bourgougnon, N.; Mouget, J.L. Anticancer, antiviral, antibacterial, and antifungal properties in microalgae. In Microalgae in Health and Disease Prevention. In Microalgae in Health and Disease Prevention; Academic Press: Cambridge, MA, USA, 2018; pp. 235–261.
  15. Joshi, V.; Bharadvaja, N. Current Prospects and Clinical Status of Microalgae Derived Chemotherapeutics. Rev. Bras. Farmacogn. 2023, 33, 445–470.
  16. Singh, U.; Gandhi, H.A.; Nikita; Bhattacharya, J.; Tandon, R.; Tiwari, G.L.; Tandon, R. Cyanometabolites: Molecules with immense antiviral potential. Arch. Microbiol. 2023, 205, 164.
  17. Ibañez, E.; Cifuentes, A. Benefits of using algae as natural sources of functional ingredients. J. Sci. Food Agric. 2013, 93, 703–709.
  18. Ganesan, A.R.; Tiwari, U.; Rajauria, G. Seaweed nutraceuticals and their therapeutic role in disease prevention. Food Sci. Hum. Wellness. 2019, 8, 252–263.
  19. Sharaf, M.; Amara, A.; Aboul-Enein, A.; Helmi, S.; Ballot, A.; Astani, A.; Schnitzler, P. Molecular authentication and characterization of the antiherpetic activity of the cyanobacterium Arthrospira fusiformis. Die Pharm. Int. J. Pharm. Sci. 2010, 65, 132–136.
  20. Silva, T.; Salomon, P.S.; Hamerski, L.; Walter, J.; Menezes, R.B.; Siqueira, J.E.; Santos, A.; Santos, J.A.M.; Ferme, N.; Guimarães, T.; et al. Inhibitory effect of microalgae and cyanobacteria extracts on influenza virus replication and neuraminidase activity. Peer J. 2018, 6, e5716.
  21. Shih, S.R.; Tsai, K.N.; Li, Y.S.; Chueh, C.C.; Chan, E.C. Inhibition of enterovirus 71-induced apoptosis by allophycocyanin isolated from a blue-green alga Spirulina platensis. J. Med. Virol. 2003, 70, 119–125.
  22. Dey, B.; Lerner, D.L.; Lusso, P.; Boyd, M.R.; Elder, J.H.; Berger, E.A. Multiple antiviral activities of cyanovirin-N: Blocking of human immunodeficiency virus type 1 gp120 interaction with CD4 and co-receptor and inhibition of diverse enveloped viruses. J. Virol. 2000, 74, 4562–4569.
  23. Kachko, A.; Loesgen, S.; Shahzad-ul-Hussan, S.; Tan, W.; Zubkova, I.; Takeda, K.; Wells, F.; Rubin, S.; Bewley, C.A.; Major, M.E. Inhibition of hepatitis C virus by the cyanobacterial protein Microcystis viridis lectin: Mechanistic differences between the high-mannose specific lectins MVL, CV-N., and GNA. Mol. Pharm. 2013, 10, 4590–4602.
  24. O’Keefe, B.R.; Smee, D.F.; Turpin, J.A.; Saucedo, C.J.; Gustafson, K.R.; Mori, T.; Blakeslee, D.; Buckheit, R.; Boyd, M.R. Potent anti-influenza activity of cyanovirin-N and interactions with viral hemagglutinin. Antimicrob. Agents Chemother. 2003, 47, 2518–2525.
  25. Singh, R.; Parihar, P.; Singh, M.; Bajguz, A.; Kumar, J.; Singh, S.; Singh, V.P.; Prasad, S.M. Uncovering potential applications of cyanobacteria and algal metabolites in biology, agriculture and medicine: Current status and future prospects. Front. Microbiol. 2017, 8, 459–537.
  26. Elaya Perumal, U.; Sundararaj, R. Algae: A potential source to prevent and cure the novel coronavirus–A review. Int. J. Emerg. Technol. 2020, 11, 479–483.
  27. Khan, R.J.; Jha, R.K.; Amera, G.M.; Jain, M.; Singh, E.; Pathak, A.; Singh, R.P.; Muthukumaran, J.; Singh, A.K. Targeting SARS-CoV-2: A systematic drug repurposing approach to identify promising inhibitors against 3C-like proteinase and 2′-O-ribose methyltransferase. J. Biomol. Struct. Dyn. 2021, 39, 2679–2692.
  28. Hui, D.S.; Azhar, E.I.; Madani, T.A.; Ntoumi, F.; Kock, R.; Dar, O.; Ippolito, G.; Mchugh, T.D.; Memish, Z.A.; Drosten, C.; et al. The continuing 2019-nCoV epidemic threat of novel coronaviruses to global health—The latest 2019 novel coronavirus outbreak in Wuhan, China. Int. J. Infec. Dis. 2020, 91, 264–266.
  29. Sahin, A.R.; Erdogan, A.; Agaoglu, P.M.; Dineri, Y.; Cakirci, A.Y.; Senel, M.E.; Okyay, R.A.; Tasdogan, A.M. 2019 novel coronavirus (COVID-19) outbreak: A review of the current literature. EJMO 2020, 4, 1–7.
  30. Grubaugh, N.D.; Ladner, J.T.; Lemey, P.; Pybus, O.G.; Rambaut, A.; Holmes, E.C.; Andersen, K.G. Tracking virus outbreaks in the twenty-first century. Nat. Microbiol. 2019, 4, 10–19.
  31. Guan, W.J.; Ni, Z.Y.; Hu, Y.; Liang, W.H.; Ou, C.Q.; He, J.X.; Liu, L.; Shan, H.; Lei, C.L.; Hui, D.S.; et al. Clinical characteristics of coronavirus disease 2019 in China. N. Engl. J. Med. 2020, 382, 1708–1720.
  32. Nowruz, B.; Haghighat, S.; Fahimi, H.; Mohammadi, E. Nostoc cyanobacteria species: A new and rich source of novel bioactive compounds with pharmaceutical potential. J. Pharm. Health Serv. Res. 2018, 9, 5–12.
  33. Thuan, N.H.; An, T.T.; Shrestha, A.; Canh, N.X.; Sohng, J.K.; Dhakal, D. Recent advances in exploration and biotechnological production of bioactive compounds in three cyanobacterial genera: Nostoc, Lyngbya, and Microcystis. Front. Chem. 2019, 7, 604.
  34. de Jesus Raposo, M.F.; de Morais, R.M.S.C.; de Morais, A.M.M.B. Health applications of bioactive compounds from marine microalgae. Life Sci. 2013, 93, 479–486.
  35. Mayer, A.; Guerrero, A.J.; Rodríguez, A.D.; Taglialatela-Scafati, O.; Nakamura, F.; Fusetani, N. Marine pharmacology in 2014–2015: Marine compounds with antibacterial, antidiabetic, antifungal, anti-inflammatory, antiprotozoal, antituberculosis, antiviral, and anthelmintic activities; affecting the immune and nervous systems, and other miscellaneous mechanisms of action. Mar. Drugs 2020, 18, 5.
  36. Soares, A.R.; Robaina, M.; Mendes, G.S.; Silva, T.S.; Gestinari, L.; Pamplona, O.S.; Yoneshigue-Valentin, Y.; Kaiser, C.R.; Romanos, M.T.V. Antiviral activity of extracts from Brazilian seaweeds against herpes simplex virus. Rev. Bras. Farmacogn. 2012, 22, 714–723.
  37. Pérez-Riverol, A.; Ramos, A.P.; Díaz, L.F.M.; López, Y.T.; Llanes, D.M.; del Barrio Alonso, G. Antiviral activity of an aqueous extract from the red alga Laurencia obtusa against influenza A and B viruses. Rev. Cubana. Med. Trop. 2014, 66, 273–285.
  38. Zaid, S.A.A.L.; Hamed, N.N.E.D.; Abdel-Wahab, K.S.E.D.; Abo El-Magd, E.K.; Salah El-Din, R.A.L. Antiviral activities and phytochemical constituents of Egyptian marine seaweeds (Cystoseira myrica (SG Gmelin) C. Agardh and Ulva Lactuca Linnaeus) aqueous extract. Egypt. J. Hosp. Med. 2016, 64, 422–429.
  39. El-Sheekh, M.M.; Shabaan, M.T.; Hassan, L.; Morsi, H.H. Antiviral activity of algae biosynthesized silver and gold nanoparticles against Herpes Simplex (HSV-1) virus in-vitro using cell-line culture technique. Int. J. Environ. Health Res. 2020, 32, 616–627.
  40. Pinto, A.M.V.; Leite, J.P.G.; Ferreira, W.J.; Cavalcanti, D.N.; Villaça, R.C.; Giongo, V.; Teixeira, V.L.; Paixão, I.C.N.D.P. Marine natural seaweed products as potential antiviral drugs against bovine viral diarrhea virus. Rev. Bras. Farmacogn. 2012, 22, 813–817.
  41. Pradhan, B.; Ki, J.S. Biological activity of algal derived carrageenan: A comprehensive review in light of human health and disease. Int. J. Biol. Macromol. 2023, 21, 124085.
  42. Al-Horani, R.A.; Kar, S.; Aliter, K.F. Potential Anti-COVID-19 Therapeutics that Block the Early Stage of the Viral Life Cycle: Structures, Mechanisms, and Clinical Trials. Int. J. Mol. Sci. 2020, 21, 5224.
  43. Wang, W.; Wang, S.X.; Guan, H.S. The antiviral activities and mechanisms of marine polysaccharides: An overview. Mar. Drugs 2012, 10, 2795–2816.
  44. Cosenza, V.A.; Navarro, D.A.; Pujol, C.A.; Damonte, E.B.; Stortz, C.A. Partial and total C-6 oxidation of gelling carrageenans. Modulation of the antiviral activity with the anionic character. Carbohydr. Polym. 2015, 128, 199–206.
  45. Zia, K.M.; Tabasum, S.; Nasif, M.; Sultan, N.; Aslam, N.; Noreen, A.; Zuber, M. A review on synthesis, properties and applications of natural polymer based carrageenan blends and composites. Int. J. Biol. Macromol. 2017, 96, 282–301.
  46. Pacheco-Quito, E.M.; Ruiz-Caro, R.; Veiga, M.D. Carrageenan: Drug delivery systems and other biomedical applications. Mar. Drugs 2020, 18, 583.
  47. Álvarez-Viñas, M.; Souto, S.; Flórez-Fernández, N.; Torres, M.D.; Bandín, I.; Domínguez, H. Antiviral activity of carrageenans and processing implications. Mar. Drugs 2021, 19, 437.
  48. Campo, V.L.; Kawano, D.F.; da Silva Jr, D.B.; Carvalho, I. Carrageenans: Biological properties, chemical modifications and structural analysis—A review. Carbohyd. Polym. 2009, 77, 167–180.
  49. Boulho, R.; Marty, C.; Freile-Pelegrín, Y.; Robledo, D.; Bourgougnon, N.; Bedoux, G. Antiherpetic (HSV-1) activity of carrageenans from the red seaweed Solieria chordalis (Rhodophyta, Gigartinales) extracted by microwave-assisted extraction (MAE). J. Appl. Phycol. 2017, 29, 2219–2228.
  50. Gomaa, H.H.A.; Elshoubaky, G.A. Antiviral activity of sulfated polysaccharides carrageenan from some marine seaweeds. Int. J. Curr. Pharm. Rev. Res. 2016, 7, 34–42.
  51. Abu-Galiyun, E.; Huleihel, M.; Levy-Ontman, O. Antiviral bioactivity of renewable polysaccharides against Varicella zoster. Cell Cycle. 2019, 18, 3540–3549.
  52. Baba, M.; Snoeck, R.; Pauwels, R.; De Clercq, E. Sulfated polysaccharides are potent and selective inhibitors of various enveloped viruses, including herpes simplex virus, cytomegalovirus, vesicular stomatitis virus, and human immunodeficiency virus. Antimicrob. Agents Chemother. 1988, 32, 1742–1745.
  53. Yamada, T.; Ogamo, A.; Saito, T.; Uchiyama, H.; Nakagawa, Y. Preparation of O-acylated low-molecular-weight carrageenans with potent anti-HIV activity and low anticoagulant effect. Carbohydr. Polym. 2000, 41, 115–120.
  54. Klimyte, E.M.; Smith, S.E.; Oreste, P.; Lembo, D.; Dutch, R.E. Inhibition of Human Metapneumovirus Binding to Heparan Sulfate Blocks Infection in Human Lung Cells and Airway Tissues. J. Virol. 2016, 90, 9237–9250.
  55. Besednova, N.N.; Zvyagintseva, T.N.; Kuznetsova, T.A.; Makarenkova, I.D.; Smolina, T.P.; Fedyanina, L.N.; Kryzhanovsky, S.P.; Zaporozhets, T.S. Marine algae metabolites as promising therapeutics for the prevention and treatment of HIV/AIDS. Metabolites 2019, 9, 87.
  56. Damonte, E.; Matulewicz, M.; Cerezo, A. Sulfated Seaweed Polysaccharides as Antiviral Agents. Curr. Med. Chem. 2012, 11, 2399–2419.
  57. Shi, Q.; Wang, A.; Lu, Z.; Qin, C.; Hu, J.; Yin, J. Overview on the antiviral activities and mechanisms of marine polysaccharides from seaweeds. Carbohydr. Res. 2017, 453, 1–9.
  58. Bansal, S.; Jonsson, C.B.; Taylor, S.L.; Manuel Figueroa, J.; Vanesa, A.; Palacios, C.; César Vega, J. Iota-carrageenan and Xylitol inhibit SARS-CoV-2 in cell culture. bioRxiv 2020.
  59. Jang, Y.; Shin, H.; Lee, M.K.; Kwon, O.S.; Shin, J.S.; Kim, Y.; Kim, C.W.; Lee, H.R.; Kim, M. Antiviral activity of lambda-carrageenan against influenza viruses and severe acute respiratory syndrome coronavirus 2. Sci. Rep. 2021, 11, 821.
  60. Morokutti-Kurz, M.; Fröba, M.; Graf, P.; Große, M.; Grassauer, A.; Auth, J.; Schubert, U.; Prieschl-Grassauer, E. Iota-carrageenan neutralizes SARS-CoV-2 and inhibits viral replication in vitro. PLoS ONE 2021, 16, e0237480.
  61. Song, S.; Peng, H.; Wang, Q.; Liu, Z.; Dong, X.; Wen, C.; Ai, C.; Zhang, Y.; Wang, Z.; Zhu, B. Inhibitory activities of marine sulfated polysaccharides against SARS-CoV-2. Food Funct. 2020, 11, 7415–7420.
  62. Davidson, I.W.; Sutherland, I.W.; Lawson, C.J. Purification and properties of an alginate lysase from a marine bacterium. Biochem. J. 1976, 159, 703–707.
  63. Meena, R.; Chhatbar, M.; Prasad, K.; Siddhanta, A.K. Development of a robust hydrogel system based on agar and sodium alginate blend. Poly. Int. 2008, 57, 329–336.
  64. Smidsrød, O.; Skja, G. Alginate as immobilization matrix for cells. Trends Biotechnol. 1990, 8, 71–78.
  65. Ratner, B.D.; Hoffman, A.S.; Schoen, F.J.; Lemons, J.E. Biomaterials Science: An Introduction to Materials in Medicine; Academic Press: Toronto, ON, Canada, 2012.
  66. Mastromarino, P.; Petruzziello, R.; Macchia, S.; Rieti, S.; Nicoletti, R.; Orsi, N. Antiviral activity of natural and semisynthetic polysaccharides on the early steps of rubella virus infection. J. Antimicrob. Chemother. 1997, 39, 339–345.
  67. Bandyopadhyay, S.S.; Navid, M.H.; Ghosh, T.; Schnitzler, P.; Ray, B. Structural features and in vitro antiviral activities of sulfated polysaccharides from Sphacelaria indica. Phytochemistry 2011, 72, 276–283.
  68. Sinha, S.; Astani, A.; Ghosh, T.; Schnitzler, P.; Ray, B. Polysaccharides from Sargassum tenerrimum: Structural features, chemical modification and anti-viral activity. Phytochemistry 2010, 71, 235–242.
  69. Fabra, M.J.; Falcó, I.; Randazzo, W.; Sánchez, G.; López-Rubio, A. Antiviral and antioxidant properties of active alginate edible films containing phenolic extracts. Food Hydrocoll. 2018, 81, 96–103.
  70. Falcó, I.; Flores-Meraz, P.L.; Randazzo, W.; Sánchez, G.; López-Rubio, A.; Fabra, M.J. Antiviral activity of alginate-oleic acid based coatings incorporating green tea extract on strawberries and raspberries. Food Hydrocoll. 2019, 87, 611–618.
More
Information
Subjects: Virology
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: 358
Entry Collection: COVID-19
Revisions: 2 times (View History)
Update Date: 17 Aug 2023
1000/1000
Video Production Service