POTENTIAL APPLICATIONS OF BIOACTIVE COMPONENTS FROM BROWN ALGAE

Main Article Content

Thuy Ngoc Trang Nguyen
Men Thanh Tran
Huy Phuc Nguyen
Hang Diem Le
Diem Thi Ngoc Vo
Thien Minh Nguyen
Xuyen Ngoc Le

Abstract

Recently, macroalgae has found extensive utilization within the domain of biotechnology. Based on their variety of bioactive components, species of class Phaeophyceae are involved in the food, cosmetic, and pharmaceutical industries. Studies have proved that these unique compounds show beneficial activities for human health. With peculiar properties, including antioxidant, antimicrobial, antiviral activities, and related functionalities, each compound holds potential value for human health. This review discusses the bioactive compounds of brown algae and the recent multiple applications of those components that make up algae’s potential in industrial companies. The review was conducted by searching and selecting references related to the content of interest with popular web search engines such as Google Scholar and PubMed.

Downloads

Download data is not yet available.

Article Details

How to Cite
1.
Nguyen T, Tran M, Nguyen H, Le H, Vo D, Nguyen T, Le X. POTENTIAL APPLICATIONS OF BIOACTIVE COMPONENTS FROM BROWN ALGAE. journal [Internet]. 29Sep.2023 [cited 17May2024];13(3). Available from: https://journal.tvu.edu.vn/index.php/journal/article/view/2434
Section
Articles

References

[1] Lyu M, Wang YF, Fan GW, Wang XY, Xu
SY, Zhu Y. Balancing herbal medicine and functional food for prevention and treatment of cardiometabolic diseases through modulating gut microbiota. Frontiers In Microbiology. 2017;8: 2146.
https://doi.org/10.3389/fmicb.2017.02146.
[2] Agregán R, Munekata PE, Domínguez R, Carballo
J, Franco D, Lorenzo JM. Proximate composition,
phenolic content and in vitro antioxidant activity
of aqueous extracts of the seaweeds Ascophyllum
nodosum, Bifurcaria bifurcata and Fucus vesiculosus.
Effect of addition of the extracts on the oxidative
stability of canola oil under accelerated storage conditions. Food Research International. 2017;99: 986–
994. https://doi.org/10.1016/j.foodres.2016.11.009.
[3] Charoensiddhi S, Conlon MA, Franco CM, Zhang
W. The development of seaweed-derived bioactive compounds for use as prebiotics and nutraceuticals using enzyme technologies. Trends in
Food Science & Technology. 2017;70: 20–23.
https://doi.org/10.1016/j.tifs.2017.10.002.
[4] Cao J, Wang J, Wang S, Xu X. Porphyra species:
A mini-review of its pharmacological and nutritional
properties. Journal of Medicinal Food. 2016;19(2):
111–119.
[5] Swamy MA. Marine algal sources for
treating bacterial diseases. Advances in Food
and Nutrition Research. 2011;64: 71–84.
https://doi.org/10.1016/B978-0-12-387669-0.00006-
5.
[6] Gheda SF, El-Adawi HI, El-Deeb NM. Antiviral profile of brown and red seaweed polysaccharides against
hepatitis C virus. Iranian Journal of Pharmaceutical
Research: IJPR. 2016;15(3): 483–491.
[7] Kalenik TK, Dobrynina EV, Ostapenko VM, Torii
Y, Hiromi J. Research of pigments of blue-green
algae Spirulina platensis for practical use in confectionery technology. Proceedings of the Voronezh State
University of Engineering Technologies. 2019;81(2):
170–176. https://doi.org/10.20914/2310-1202-2019-
2-170-176.
[8] Ambati RR, Phang SM, Ravi S, Aswathanarayana
RG. Astaxanthin: Sources, extraction, stability, biological activities and its commercial applications
– A review. Marine Drugs. 2014;12(1): 128–152.
https://doi.org/10.3390/md12010128.
[9] Montero L, del Pilar Sánchez-Camargo A, Ibánez ˜
E, Gilbert-López B. Phenolic compounds from
edible algae: Bioactivity and health benefits. Current
Medicinal Chemistry. 2018;25(37): 4808–4826.
https://doi.org/10.2174/0929867324666170523120101.
[10] Gheda S, Naby MA, Mohamed T, Pereira L,
Khamis A. Antidiabetic and antioxidant activity
of phlorotannins extracted from the brown
seaweed Cystoseira compressa in streptozotocininduced diabetic rats. Environmental Science
and Pollution Research. 2021;28: 22886–22901.
https://doi.org/10.1007/s11356-021-12347-5.
[11] Alghazeer R, Elmansori A, Sidati M, Gammoudi
F, Azwai S, Naas H, et al. In vitro antibacterial activity of flavonoid extracts of two selected libyan algae against multi-drug resistant
bacteria isolated from food products. Journal
of Biosciences and Medicines. 2017;5(1): 26–48.
https://doi.org/10.4236/jbm.2017.51003.
[12] Kim JH, Lee JE, Kim KH, Kang NJ. Beneficial effects of marine algae-derived carbohydrates
for skin health. Marine Drugs. 2018;16(11): 459.
https://doi.org/10.3390/md16110459.
[13] Mannino AM, Micheli C. Ecological function of
phenolic compounds from mediterranean fucoid algae and seagrasses: An overview on the genus Cystoseira
sensu lato and Posidonia oceanica (L.) Delile. Journal
of Marine Science and Engineering. 2020;8(1): 19.
https://doi.org/10.3390/jmse8010019.
[14] Cotas J, Leandro A, Monteiro P, Pacheco
D, Figueirinha A, Gonc¸alves AM , et
al. Seaweed phenolics: From extraction to
applications. Marine Drugs. 2020;18(8): 384.
https://doi.org/10.3390/md18080384.
[15] Palanisam SK, Arumugam V, Rajendran S,
Ramadoss A, Nachimuthu, S, Peter DM,
Sundaresan U. Chemical diversity and antiproliferative activity of marine algae. Natural
Product Research. 2019;33(14): 2120–2124.
https://doi.org/10.1080/14786419.2018.1488701.
[16] Pereira L. Therapeutic and nutritional uses of algae.
Boca Raton, Florida: CRC Press, Taylor & Francis
Group; 2018.
[17] Zheng LX, Chen XQ, Cheong KL. Current
trends in marine algae polysaccharides: The
digestive tract, microbial catabolism, and
prebiotic potential. International Journal of
Biological Macromolecules. 2020;151: 344–354.
https://doi.org/10.1016/j.ijbiomac.2020.02.168.
[18] Carpena M, Caleja C, García-Oliveira P, Pereira C,
Sokovic M, Ferreira IC, et al. Red algae as source of
nutrients with antioxidant and antimicrobial potential.
In: Christopher John Smith (Ed.) Proceedings of
the 1st International Electronic Conference on Food
Science and Functional Foods, 10-25 November 2020.
2020;70(1): 5. https://doi.org/10.3390/foods_2020-
07646.
[19] Kirpenko NI, Usenko OM, Musiy TO. Content
of proteins, carbohydrates, and lipids in the cells
of green algae at short-term temperature fluctuations. Hydrobiological Journal. 2017;53(1): 50–59.
https://doi.org/10.1615/hydrobj.v53.i1.50.
[20] Holdt SL, Kraan S. Bioactive compounds in seaweed: functional food applications and legislation.
Journal of Applied Phycology. 2011;23: 543–597.
https://doi.org/10.1007/s10811-010-9632-5.
[21] Xie X, Lu X, Wang L, He L, Wang G.
High light intensity increases the concentrations
of β-carotene and zeaxanthin in marine red
macroalgae. Algal Research. 2020;47: 101852.
https://doi.org/10.1016/j.algal.2020.101852.
[22] Dolganyuk V, Andreeva A, Budenkova E,
Sukhikh S, Babich O, Ivanova S, et al. Study
of morphological features and determination of
the fatty acid composition of the microalgae
lipid complex. Biomolecules. 2020;10(11): 1–15.
https://doi.org/10.3390/biom10111571.
[23] Anis M, Ahmed S, Hasan MM. Algae as nutrition,
medicine and cosmetic: The forgotten history, present
status and future trends. World Journal of Pharmacy
and Pharmaceutical Sciences. 2017;6(6): 1934–1959.
https://doi.org/10.20959/WJPPS20176-9447.
[24] Praiboon J, Palakas S, Noiraksa T, Miyashita K. Seasonal variation in nutritional composition and antiproliferative activity of brown seaweed, Sargassum
oligocystum. Journal of Applied Phycology. 2018;30:
101–111. https://doi.org/10.1007/s10811-017-1248-6.
[25] Del Mondo A, Smerilli A, Sané E, Sansone C,
Brunet C. Challenging microalgal vitamins for human
health. Microbial Cell Factories. 2020;19(1): 1–23.
https://doi.org/10.1186/s12934-020-01459-1.
[26] bdel-Latif HH, Shams El-Din NG, Ibrahim HA.
Antimicrobial activity of the newly recorded red
alga Grateloupia doryphora collected from the Eastern Harbor, Alexandria, Egypt. Journal of Applied Microbiology. 2018;125(5): 1321–1332. doi:
10.1111/jam.14050.
[27] Hwang PA, Phan NN, Lu WJ, Hieu BTN,
Lin YC. Low-molecular-weight fucoidan and highstability fucoxanthin from brown seaweed exert prebiotics and anti-inflammatory activities in Caco-2
cells. Food & Nutrition Research. 2016;60(1): 1–9.
https://doi.org/10.3402/fnr.v60.32033.
[28] Rodríguez-Luna A, Ávila-Román J, Oliveira H,
Motilva V, Talero E. Fucoxanthin and rosmarinic acid
combination has anti-inflammatory effects through
regulation of NLRP3 inflammasome in UVB-exposed
HaCaT keratinocytes. Marine Drugs. 2019;17(8): 1–
14. https://doi.org/10.3390/md17080451.
[29] Ganesan P, Matsubara K, Sugawara T, Hirata
T. Marine algal carotenoids inhibit angiogenesis by down-regulating FGF-2-mediated intracellular signals in vascular endothelial cells. Molecular and Cellular Biochemistry. 2013;380: 1–9.
https://doi.org/10.1007/s11010-013-1651-5.
[30] Mei C, Zhou S, Zhu L, Ming J, Zeng F, Xu R.
Antitumor effects of Laminaria extract fucoxanthin
on lung cancer. Marine Drugs. 2017;15(2): 1–12.
https://doi.org/10.3390/md15020039.
[31] Kim KN, Ahn G, Heo SJ, Kang SM, Kang
MC, Yang HM, et al. Inhibition of tumor
growth in vitro and in vivo by fucoxanthin
against melanoma B16F10 cells. Environmental
Toxicology and Pharmacology. 2013;35(1): 39–46.
https://doi.org/10.1016/j.etap.2012.10.002.
[32] Chung TW, Choi HJ, Lee JY, Jeong HS, Kim CH,
Joo M, et al. Marine algal fucoxanthin inhibits the
metastatic potential of cancer cells. Biochemical and
biophysical research communications. 2013;439(4):
580–585. https://doi.org/10.1016/j.bbrc.2013.09.019.
[33] Atya ME, El-Hawiet A, Alyeldeen MA, Ghareeb DA,
Abdel-Daim MM, El-Sadek MM. In vitro biological activities and in vivo hepatoprotective role of
brown algae-isolated fucoidans. Environmental Science and Pollution Research. 2021;28: 19664–19676.
https://doi.org/10.1007/s11356-020-11892-9.
[34] Wang SK, Li Y, White WL, Lu J. Extracts from New Zealand Undaria pinnatifida containing fucoxanthin
as potential functional biomaterials against cancer in
vitro. Journal of Functional Biomaterials. 2014;5(2):
29–42. https://doi.org/10.3390/jfb5020029.
[35] Airanthi MWA, Hosokawa M, Miyashita K.
Comparative antioxidant activity of edible Japanese
brown seaweeds. Journal of Food Science.
2011;76(1): 104–111. https://doi.org/10.1111/j.1750-
3841.2010.01915.x.
[36] Boo HJ, Hyun JH, Kim SC, Kang JI, Kim MK,
Kim SY, et al. Fucoidan from Undaria pinnatifida induces apoptosis in A549 human lung carcinoma cells. Phytotherapy Research. 2011;25(7):
1082–1086. https://doi.org/10.1002/ptr.3489.
[37] Lomartire S, Gonc¸alves AM. An overview of potential seaweed-derived bioactive compounds for pharmaceutical applications. Marine Drugs. 2022;20(2):
141. https://doi.org/10.3390/md20020141.
[38] Palanisamy S, Vinosha M, Marudhupandi
T, Rajasekar P, Prabhu NM. In vitro
antioxidant and antibacterial activity of sulfated
polysaccharides isolated from Spatoglossum
asperum. Carbohydrate Polymers. 2017;170: 296–
304. https://doi.org/10.1016/j.carbpol.2017.04.085.
[39] Liu M, Liu Y, Cao MJ, Liu GM, Chen Q, Sun
L, Chen H. Antibacterial activity and mechanisms
of depolymerized fucoidans isolated from Laminaria
japonica. Carbohydrate Polymers. 2017;172: 294–
305. https://doi.org/10.1016/j.carbpol.2017.05.060.
[40] Krylova NV, Ermakova SP, Lavrov VF, Leneva I A,
Kompanets GG, Iunikhina OV, et al. The comparative
analysis of antiviral activity of native and modified
fucoidans from brown algae Fucus evanescens in
vitro and in vivo. Marine Drugs. 2020;18(4): 224.
https://doi.org/10.3390/md18040224.
[41] Zhu W, Chiu LCM, Ooi VEC, Chan PKS, Ang Jr
PO. Antiviral property and mode of action of a sulphated polysaccharide from Sargassum patens against
herpes simplex virus type 2. International Journal of Antimicrobial Agents. 2004;24(3): 279–283.
https://doi.org/10.1016/j.ijantimicag.2004.02.022.
[42] 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(2-3): 235–242.
https://doi.org/10.1016/j.phytochem.2009.10.014.
[43] Pozharitskaya ON, Obluchinskaya ED, Shikov AN.
Mechanisms of bioactivities of fucoidan from
the brown seaweed Fucus vesiculosus L. of the
Barents Sea. Marine Drugs. 2020;18(5): 275.
https://doi.org/10.3390/md18050275.
[44] Irhimeh MR, Fitton JH, Lowenthal RM. Pilot
clinical study to evaluate the anticoagulant
activity of fucoidan. Blood Coagulation
& Fibrinolysis. 2009;20(7): 607–610.
https://doi.org/10.1097/MBC.0b013e32833135fe.
[45] Koh HSA, Lu J, Zhou W. Structural dependence
of sulfated polysaccharide for diabetes management:
fucoidan from Undaria pinnatifida inhibiting α-
glucosidase more strongly than α-amylase and amyloglucosidase. Frontiers in Pharmacology. 2020;11:
831. https://doi.org/10.3389/fphar.2020.00831.
[46] Jia RB, Wu J, Li ZR, Ou ZR, Lin L, Sun B,
Zhao M. Structural characterization of polysaccharides from three seaweed species and their hypoglycemic and hypolipidemic activities in type
2 diabetic rats. International Journal of Biological Macromolecules. 2020;155: 1040–1049.
https://doi.org/10.1016/j.ijbiomac.2019.11.068.
[47] Chkhikvishvili ID, Ramazanov ZM. Phenolic substances of brown algae and their antioxidant activity.
Applied Biochemistry and Microbiology. 2000;36:
289–291. https://doi.org/10.1007/BF02742582.
[48] Nagayama K, Iwamura Y, Shibata T, Hirayama I,
Nakamura T. Bactericidal activity of phlorotannins
from the brown alga Ecklonia kurome. Journal of
Antimicrobial Chemotherapy. 2002;50(6): 889–893.
https://doi.org/10.1093/jac/dkf222.
[49] Lee DS, Eom SH, Jeong SY, Shin HJ, Je JY, Lee
EW, et al. Anti-methicillin-resistant Staphylococcus
aureus (MRSA) substance from the marine bacterium Pseudomonas sp. UJ-6. Environmental Toxicology and Pharmacology. 2013;35(2): 171–177.
https://doi.org/10.1016/j.etap.2012.11.011.
[50] Na HJ, Moon PD, Ko SG, Lee HJ, Jung
HA, Hong SH, et al. Sargassum hemiphyllum
inhibits atopic allergic reaction via the regulation of inflammatory mediators. Journal of
Pharmacological Sciences. 2005;97(2): 219–226.
https://doi.org/10.1254/jphs.fp0040326.
[51] Sugiura Y, Matsuda K, Okamoto T, Kakinuma
M, Amano H. Anti-allergic effects of the
brown alga Eisenia arborea on Brown Norway
rats. Fisheries Science. 2008;74: 180–186.
https://doi.org/10.1111/j.1444-2906.2007.01508.x.
[52] Pangestuti R, Kim SK. Neuroprotective effects of
marine algae. Marine Drugs. 2011;9(5): 803–818.
https://doi.org/10.3390/md9050803.
[53] Custodio L, Silvestre L, Rocha MI, Rodrigues MJ,
Vizetto-Duarte C, Pereira H, et al. Methanol extracts
from Cystoseira tamariscifolia and Cystoseira
nodicaulis are able to inhibit cholinesterases
and protect a human dopaminergic cell line
from hydrogen peroxide-induced cytotoxicity.
Pharmaceutical Biology. 2016;54(9): 1687–1696.
https://doi.org/10.3109/13880209.2015.1123278.
[54] Andrade PB, Barbosa M, Matos RP, Lopes
G, Vinholes J, Mouga T, Valentão P.
Valuable compounds in macroalgae extracts.
Food Chemistry. 2013;138(2-3): 1819–1828.
https://doi.org/10.1016/j.foodchem.2012.11.081.
[55] Kannan RR, Aderogba MA, Ndhlala AR, Stirk WA, Van Staden J. Acetylcholinesterase inhibitory
activity of phlorotannins isolated from the brown
alga, Ecklonia maxima (Osbeck) Papenfuss. Food
Research International. 2013;54(1): 1250–1254.
https://doi.org/10.1016/j.foodres.2012.11.017.
[56] Abdelhamid A, Jouini M, Bel Haj Amor H, Mzoughi
Z, Dridi M, Ben Said R, Bouraoui A. Phytochemical analysis and evaluation of the antioxidant,
anti-inflammatory, and antinociceptive potential of
phlorotannin-rich fractions from three Mediterranean
brown seaweeds. Marine Biotechnology. 2018;20: 60–
74. https://doi.org/10.1007/s10126-017-9787-z.
[57] Kim MM, Kim SK. Effect of phloroglucinol
on oxidative stress and inflammation. Food and
Chemical Toxicology. 2010;48(10): 2925–2933.
https://doi.org/10.1016/j.fct.2010.07.029.
[58] Kong CS, Kim JA, Yoon NY, Kim SK. Induction of
apoptosis by phloroglucinol derivative from Ecklonia
cava in MCF-7 human breast cancer cells. Food
and Chemical Toxicology. 2009;47(7): 1653–1658.
https://doi.org/10.1016/j.fct.2009.04.013.
[59] Ferreres F, Lopes G, Gil-Izquierdo A, Andrade PB,
Sousa C, Mouga T, Valentão P. Phlorotannin extracts
from fucales characterized by HPLC-DAD-ESI-MS n:
Approaches to hyaluronidase inhibitory capacity and
antioxidant properties. Marine Drugs. 2012;10(12):
2766–2781. https://doi.org/10.3390/md10122766.
[60] Heo SJ, Ko SC, Kang SM, Cha SH, Lee SH, Kang
DH, et al. Inhibitory effect of diphlorethohydroxycarmalol on melanogenesis and its protective effect
against UV-B radiation-induced cell damage. Food
and Chemical Toxicology. 2010;48(5): 1355–1361. https://doi.org/10.1016/j.fct.2010.03.001.
[61] Shiino M, Watanabe Y, Umezawa K. Synthesis of N-substituted N-nitrosohydroxylamines as
inhibitors of mushroom tyrosinase. Bioorganic
& Medicinal Chemistry. 2001;9(5): 1233–1240.
https://doi.org/10.1016/S0968-0896(01)00003-7.