A Review on Fucoidan Structure, Extraction Techniques, and Its Role as an Immunomodulatory Agent
Abstract
:1. Introduction
2. Structure of Fucoidan
3. Extraction Techniques and Potential Impurities
4. Immunomodulatory Effects
Study | Cell Line | Concentration | Algal Species and Fucoidan Purified | Cell Signaling Activity | Reference |
---|---|---|---|---|---|
Inflammatory related cytokine modulation and expressing anti-obesity effects | 3T3-L1 | 1–100 µg/mL | Undaria pinnatifida Polysaccharides (≈60%) and sulfate (≈25%) with less protein content | Significantly decrease the expression of inflammation-related genes during adipogenesis in 3T3-L1 adipocytes. Adipogenesis major markers (c/EBPα, PPARγ) were down regulated via fucoidan. Inactivation of aP2 led to the weakening of TNF-α, MCP-1, PA-1 levels. Lipid accumulation and ROS content in adipocytes were attenuated by fucoidan. | Kim et al. (2012) [52] |
Fine dust (FD) induced inflammatory responses in HaCaT keratinocytes are ameliorated by fucoidan | Human skin keratinocytes (HaCaT) | 12.5–100 µg/mL | Sargassum horneri purified fucoidan fraction SHC4-6 was reported as a highly sulfated mannofucan (≈45 kDa) | SHC4-6 dose-dependently lowered ROS levels in Fine Dust-induced HaCaT keratinocytes, also downregulated inflammatory cytokines, tumour necrosis factor-α, interleukin (IL)-1β, -5, -6, -8, -13, interferon-γ, and chemokines, macrophage-derived chemokine, eotoxin, and thymus and activation regulated chemokine. Molecular mediators of MAPK and NF-κB pathway were downregulated by SHC4-6. This could successfully recover the impact of FD on skin barrier molecular mediators. | Fernando et al. (2021) [2] |
LPS induced inflammation in macrophages is attenuated by fucoidan from Sargassum swartzii | RAW 264.7 macrophage cells | 25–200 µg/mL | Fucoidan fraction F4 composed of Polysaccharide (approximately 60%), Sulfate (approximately 33.99%) with a low amount of Protein (0.41%) and Polyphenols (0.32 %) | Significantly decrease the NO production stimulated by LPS and also downregulate the expression of inflammatory mediators such as iNOS and COX-2 including pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β), with a dose-dependent manner. The anti-inflammatory effect was exhibited via the suppression of TLR mediated MyD88, IKK complex ultimately blocking NF-κB and MAPK activation. | Jayawardena et al. (2020) [5] |
Inflammatory responses stimulated via LPS in macrophages are inhibited via Padina commersonii purified fucoidan | RAW 264.7 macrophage cells | 25–100 µg/mL | Purified fucoidan was rich in fucose and sulfate. Composed of 76.57 ± 2.54% polysaccharides and 11.20 ± 0.10% sulfates. FTIR results demonstrated structural similarity with commercial fucoidan. | Significantly down-regulated LPS-activated mRNA and protein expression levels of TLR2, TLR4, and MyD88 which are the inducers of NF-κB transcriptional factors via blocking TLR/MyD88/NF-κB signal transduction. | Sanjeewa et al. (2019) [39] |
Anti-inflammatory effects of Sargassum horneri were evaluated in RAW 264.7 macrophages and zebrafish model | RAW 264.7 macrophage cells | 12.5–50 µg/mL | A fucoidan (SHCF2) was purified via enzyme assisted extraction and FPLC system. Composed of polysaccharides (approximately 65%) and sulfate (approximately 12.5%) with protein (approximately 14%) | Inhibited the LPS-stimulated NO production in RAW 264.7 cells (IC50 = 40 μg/mL) via the nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signal pathways. Specifically, SHCF2 down-regulated the heart-beating rate, cell death, ROS, and NO levels in LPS-exposed zebrafish embryo. | Sanjeewa et al. (2019) [62] |
Potential molecular mechanisms of fucoidan from Saccharina japonica is evaluated against LPS induced macrophages | RAW 264.7 macrophage cells | 50–200 µg/mL | Fraction 6 (SF6) Composed of polysaccharides approximately 58%) and sulfate (approximately 36%) with low amount of protein (approximately 1%) | SF6 remarkably inhibited LPS-induced production of various inflammatory mediators and pro-inflammation cytokines, including nitric oxide (NO), NO synthase (iNOS), cyclooxygenase-2 (COX-2), tumor necrosis factor-α (TNF-α), interleukin-β (IL-β), and interleukin-6 (IL-6). A mechanism study showed that SF6 could effectively inhibit inflammatory responses through blocking LPS-induced inflammation pathways, including nuclear factor-κB (NF-κB), mitogen-activated protein kinase (MAPK), and Janus kinase (JAK)-2 and signal transducer and activator of transcription (STAT)-1/3 pathways. | Ye et al. (2020) [70] |
Study | Cell Line | Concentration | Algal Species and Fucoidan Purified | Cell Signaling Activity | Reference |
---|---|---|---|---|---|
Fucoidan altered the immunomodulatory markers and DCs phenotype | Human monocyte-derived dendritic cells (DCs) | 100 μg/mL fucoidan | Standard fucoidan-Fucoidan purified from Fucus vesiculosus was purchased | Fucoidan elevated the expression of HLA-DR and co-stimulatory molecules of DCs, induces their Th1-promoting tumor necrosis factor α (TNF-α) and interleukin-12 (IL-12) secretion. This fucoidan is suggested to be used in DC-based vaccines for cancer immunotherapy. | Yang et al. (2008) [72] |
Fucoidan express immunostimulating and DC maturing potential | Bone marrow-derived dendritic cells (DCs) | 50 μg/mL | Standard fucoidan-Fucoidan purified from Fucus vesiculosus was purchased | The production of IL-12, TNF-α, major histocompatibility complex class I, II, CD54, and CD86 were promoted by fucoidan. Further fucoidan treated DCs expressed p65 (NF-κB) nuclear translocation. | Kim et al. (2008) [73] |
Effect of fucoidan on spleen DCs and in vivo | Spleen dendritic cells (DCs) | C57BL/6 mice were treated with 10 mg/kg fucoidan for 24 h | Standard fucoidan-Fucoidan purified from Fucus vesiculosus was purchased | Systemic administration of fucoidan induced up-regulation of CD40, CD80 and CD86 expression and production of IL-6, IL-12 and TNF-a in spleen cDCs. Fucoidan also promoted the generation of IFN-c-producing Th1 and Tc1 cells in an IL-12-dependent manner. Moreover, fucoidan enhanced OVA-induced up-regulation of MHC class I and II on spleen cDCs and strongly prompted the proliferation of OVA-specific CD4 and CD8 T cells. The study reveals the potential of fucoidan to function as an adjuvant to induce Th1 immune response. Further, fucoidan promote CTL activation. Suggested to be useful in tumor vaccine development. | Jin et al. (2014) [74] |
Immunomodulating potential of fucoidans on murine macrophages and splenocytes | RAW 264.7 cells, peritoneal macrophages and normal splenocytes | 50–300 μg/mL | Anion exchange column purified fucoidan from Undaria pinnatifida | Fucoidan induced TNF-α expression from both types of macrophages. The TNF-α-inducing activity of UP-F was higher than that of FV-F. The chemokine expression (RANTES and MIP-1α) was also promoted in RAW 264.7 macrophages. The IL-6 including chemokines were significantly improved in UPF treated splenocytes. | Yoo et al. (2007) [75] |
Macrophages and glial cells were examined for immune related properties against IFN-γ stimulation and fucoidan treatment | Glia (C6, BV-2) and macrophages (RAW 264.7, peritoneal primary cells) | 50 μg/mL | Standard fucoidan-Fucoidan purified from Fucus vesiculosus was purchased | In glial cells IFN-γ induced inflammation was suppressed by fucoidan via JAK/STAT/IRF-1 and p-p38. The signaling positively regulated IFN-g-induced iNOS, which were inhibited by fucoidan. Contrastingly, in RAW macrophages, fucoidan promoted immune responses induced via IFN-γ. Confirmed the dual regulation of p38 in BV-2 microglia and primary peritoneal macrophages. | Do et al. (2010) [76] |
In vivo viral replication and host immune defense system were assessed against fucoidan treatment | Macrophages were collected from BALB/c mice | 10 μg/mL | Fucoidan prepared from Undaria pinnatifida | Fucoidan oral administration protected mice from infection with HSV-1. CTL activity of HSV-1 mice was enhanced by fucoidan. Phagocytic activity of macrophages and B cell blastogenesis in vitro was significantly stimulated by the fucoidan, while no significant change in the release of NO2− by macrophages was observed. | Hayashi et al. (2008) [3] |
Immune boosting properties of fucoidan from Nizamuddinia zanardinii | RAW 264.7 macrophage cell | 10, 25 and 50 μg/mL | Anion exchange column purified fucoidan. | The active fraction (F3) promoted the secretion of NO, TNF-α, IL-1β, IL-6 in RAW 264.7 macrophages. Further NK cells were activated to release TNF-α, IFN-γ, granzyme-B, perforin, NKG2D and FasL. The activity was mediated through NF-κB and MAPK pathways. | Tabarsa et al. (2020) [100] |
5. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Wijesinghe, W.A.J.P.; Jeon, Y.-J. Biological activities and potential industrial applications of fucose rich sulfated polysaccharides and fucoidans isolated from brown seaweeds: A review. Carbohyd. Polym. 2012, 88, 13–20. [Google Scholar] [CrossRef]
- Fernando, I.P.S.; Dias, M.; Madusanka, D.M.D.; Han, E.J.; Kim, M.J.; Heo, S.J.; Lee, K.; Cheong, S.H.; Ahn, G. Low molecular weight fucoidan fraction ameliorates inflammation and deterioration of skin barrier in fine-dust stimulated keratinocytes. Int. J. Biol. Macromol. 2021, 168, 620–630. [Google Scholar] [CrossRef] [PubMed]
- Hayashi, K.; Nakano, T.; Hashimoto, M.; Kanekiyo, K.; Hayashi, T. Defensive effects of a fucoidan from brown alga Undaria pinnatifida against herpes simplex virus infection. Int. Immunopharmacol. 2008, 8, 109–116. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Zhang, Q.; Zhang, Z.; Song, H.; Li, P. Potential antioxidant and anticoagulant capacity of low molecular weight fucoidan fractions extracted from Laminaria japonica. Int. J. Biol. Macromol. 2010, 46, 6–12. [Google Scholar] [CrossRef]
- Jayawardena, T.U.; Sanjeewa, K.K.A.; Nagahawatta, D.P.; Lee, H.G.; Lu, Y.A.; Vaas, A.; Abeytunga, D.T.U.; Nanayakkara, C.M.; Lee, D.S.; Jeon, Y.J. Anti-Inflammatory Effects of Sulfated Polysaccharide from Sargassum Swartzii in Macrophages via Blocking TLR/NF-Kappab Signal Transduction. Mar. Drugs 2020, 18, 601. [Google Scholar] [CrossRef] [PubMed]
- Hahn, T.; Lang, S.; Ulber, R.; Muffler, K. Novel procedures for the extraction of fucoidan from brown algae. Process. Biochem 2012, 47, 1691–1698. [Google Scholar] [CrossRef]
- Fernando, I.P.S.; Sanjeewa, K.K.A.; Samarakoon, K.W.; Lee, W.W.; Kim, H.S.; Kang, N.; Ranasinghe, P.; Lee, H.S.; Jeon, Y.J. A fucoidan fraction purified from Chnoospora minima; a potential inhibitor of LPS-induced inflammatory responses. Int. J. Biol. Macromol. 2017, 104, 1185–1193. [Google Scholar] [CrossRef]
- Nagaoka, M.; Shibata, H.; Kimura-Takagi, I.; Hashimoto, S.; Kimura, K.; Makino, T.; Aiyama, R.; Ueyama, S.; Yokokura, T. Structural study of fucoidan from Cladosiphon okamuranus TOKIDA. Glycoconj. J. 1999, 16, 19–26. [Google Scholar] [CrossRef]
- Khovidhunkit, W.; Kim, M.S.; Memon, R.A.; Shigenaga, J.K.; Moser, A.H.; Feingold, K.R.; Grunfeld, C. Effects of infection and inflammation on lipid and lipoprotein metabolism: Mechanisms and consequences to the host. J. Lipid Res. 2004, 45, 1169–1196. [Google Scholar] [CrossRef] [Green Version]
- Koeberle, A.; Werz, O. Multi-target approach for natural products in inflammation. Drug Discov. Today 2014, 19, 1871–1882. [Google Scholar] [CrossRef]
- Asanka Sanjeewa, K.K.; Lee, W.W.; Kim, J.-I.; Jeon, Y.-J. Exploiting biological activities of brown seaweed Ishige okamurae Yendo for potential industrial applications: A review. J. Appl. Phycol. 2017, 29, 3109–3119. [Google Scholar] [CrossRef]
- Huttemann, M.; Helling, S.; Sanderson, T.H.; Sinkler, C.; Samavati, L.; Mahapatra, G.; Varughese, A.; Lu, G.; Liu, J.; Ramzan, R.; et al. Regulation of mitochondrial respiration and apoptosis through cell signaling: Cytochrome c oxidase and cytochrome c in ischemia/reperfusion injury and inflammation. Biochim. Biophys. Acta 2012, 1817, 598–609. [Google Scholar] [CrossRef] [PubMed]
- Jayawardena, T.U.; Fernando, I.P.S.; Lee, W.W.; Sanjeewa, K.K.A.; Kim, H.S.; Lee, D.S.; Jeon, Y.J. Isolation and purification of fucoidan fraction in Turbinaria ornata from the Maldives; Inflammation inhibitory potential under LPS stimulated conditions in in-vitro and in-vivo models. Int. J. Biol. Macromol. 2019, 131, 614–623. [Google Scholar] [CrossRef] [PubMed]
- Zvyagintseva, T.N.; Shevchenko, N.M.; Popivnich, I.B.; Isakov, V.V.; Scobun, A.S.; Sundukova, E.V.; Elyakova, L.A. A new procedure for the separation of water-soluble polysaccharides from brown seaweeds. Carbohyd. Res. 1999, 322, 32–39. [Google Scholar] [CrossRef]
- Ale, M.T.; Mikkelsen, J.D.; Meyer, A.S. Important determinants for fucoidan bioactivity: A critical review of structure-function relations and extraction methods for fucose-containing sulfated polysaccharides from brown seaweeds. Mar. Drugs 2011, 9, 2106–2130. [Google Scholar] [CrossRef] [Green Version]
- Zhang, H.; Row, K.H. Extraction and separation of polysaccharides from Laminaria japonica by size-exclusion chromatography. J. Chromatogr Sci. 2015, 53, 498–502. [Google Scholar] [CrossRef] [Green Version]
- Patankar, M.S.; Oehninger, S.; Barnett, T.; Williams, R.L.; Clark, G.F. A revised structure for fucoidan may explain some of its biological activities. J. Biol. Chem. 1993, 268, 21770–21776. [Google Scholar] [CrossRef] [PubMed]
- Chevolot, L.; Foucault, A.; Chaubet, F.; Kervarec, N.; Sinquin, C.; Fisher, A.-M.; Boisson-Vidal, C. Further data on the structure of brown seaweed fucans: Relationships with anticoagulant activity. Carbohyd. Res. 1999, 319, 154–165. [Google Scholar] [CrossRef]
- Mabeau, S.; Kloareg, B.; Joseleau, J.-P. Fractionation and analysis of fucans from brown algae. Phytochemistry 1990, 29, 2441–2445. [Google Scholar] [CrossRef]
- Colliec, S.; Fischer, A.M.; Tapon-Bretaudiere, J.; Boisson, C.; Durand, P.; Jozefonvicz, J. Anticoagulant properties of a fucoïdan fraction. Thromb. Res. 1991, 64, 143–154. [Google Scholar] [CrossRef]
- Bilan, M.I.; Grachev, A.A.; Shashkov, A.S.; Nifantiev, N.E.; Usov, A.I. Structure of a fucoidan from the brown seaweed Fucus serratus L. Carbohydr Res. 2006, 341, 238–245. [Google Scholar] [CrossRef]
- Dubois, M.; Gilles, K.; Hamilton, J.K.; Rebers, P.A.; Smith, F. A colorimetric method for the determination of sugars. Nature 1951, 168, 167. [Google Scholar] [CrossRef] [PubMed]
- Duarte, M.E.; Cardoso, M.A.; Noseda, M.D.; Cerezo, A.S. Structural studies on fucoidans from the brown seaweed Sargassum stenophyllum. Carbohydr. Res. 2001, 333, 281–293. [Google Scholar] [CrossRef] [PubMed]
- Chizhov, A.O.; Dell, A.; Morris, H.R.; Haslam, S.M.; McDowell, R.A.; Shashkov, A.S.; Nifant’ev, N.E.; Khatuntseva, E.A.; Usov, A.I. A study of fucoidan from the brown seaweed Chorda filum. Carbohydr Res. 1999, 320, 108–119. [Google Scholar] [CrossRef]
- Wang, J.; Zhang, Q.; Zhang, Z.; Zhang, H.; Niu, X. Structural studies on a novel fucogalactan sulfate extracted from the brown seaweed Laminaria japonica. Int. J. Biol. Macromol. 2010, 47, 126–131. [Google Scholar] [CrossRef] [PubMed]
- Usoltseva, R.V.; Shevchenko, N.M.; Malyarenko, O.S.; Anastyuk, S.D.; Kasprik, A.E.; Zvyagintsev, N.V.; Ermakova, S.P. Fucoidans from brown algae Laminaria longipes and Saccharina cichorioides: Structural characteristics, anticancer and radiosensitizing activity in vitro. Carbohydr. Polym. 2019, 221, 157–165. [Google Scholar] [CrossRef] [PubMed]
- Vishchuk, O.S.; Ermakova, S.P.; Zvyagintseva, T.N. The Effect of Sulfated (1→3)-α-l-Fucan from the Brown Alga Saccharina cichorioides Miyabe on Resveratrol-Induced Apoptosis in Colon Carcinoma Cells. Mar. Drugs 2013, 11, 194–212. [Google Scholar] [CrossRef] [Green Version]
- Obluchinskaya, E.D.; Pozharitskaya, O.N.; Zakharov, D.V.; Flisyuk, E.V.; Terninko, I.I.; Generalova, Y.E.; Smekhova, I.E.; Shikov, A.N. The Biochemical Composition and Antioxidant Properties of Fucus vesiculosus from the Arctic Region. Mar. Drugs 2022, 20, 193. [Google Scholar] [CrossRef] [PubMed]
- Skriptsova, A.V.; Shevchenko, N.M.; Tarbeeva, D.V.; Zvyagintseva, T.N. Comparative Study of Polysaccharides from Reproductive and Sterile Tissues of Five Brown Seaweeds. Mar. Biotechnol. 2012, 14, 304–311. [Google Scholar] [CrossRef]
- Honya, M.; Mori, H.; Anzai, M.; Araki, Y.; Nisizawa, K. Monthly changes in the content of fucans, their constituent sugars and sulphate in cultured Laminaria japonica. In Proceedings of the Sixteenth International Seaweed Symposium, Cebu City, Philippines, 12–17 April 1998; Springer: Dordrecht, The Netherland, 1999; pp. 411–416. [Google Scholar]
- Chirinos, R.; Rogez, H.; Campos, D.; Pedreschi, R.; Larondelle, Y. Optimization of extraction conditions of antioxidant phenolic compounds from mashua (Tropaeolum tuberosum Ruíz & Pavón) tubers. Sep. Purif. Technol. 2007, 55, 217–225. [Google Scholar] [CrossRef]
- Black, W.A.P.; Dewar, E.T.; Woodward, F.N. Manufacture of algal chemicals. IV—Laboratory-scale isolation of fucoidin from brown marine algae. J. Sci. Food Agric. 1952, 3, 122–129. [Google Scholar] [CrossRef]
- Mian, A.J.; Percival, E. Carbohydrates of the brown seaweeds himanthalia lorea, bifurcaria bifurcata, and Padina pavonia. Carbohyd Res. 1973, 26, 133–146. [Google Scholar] [CrossRef]
- Rioux, L.E.; Turgeon, S.L.; Beaulieu, M. Characterization of polysaccharides extracted from brown seaweeds. Carbohyd Polym 2007, 69, 530–537. [Google Scholar] [CrossRef]
- Kylin, H.J. Biochemistry of seaweeds. Z. Physiol. Chem. 1918, 101, 236–247. [Google Scholar] [CrossRef]
- Nelson, W.L.; Cretcher, L.H. The carbohydrate acid sulfate of Macrocystis pyrifera. J. Biol. Chem. 1931, 94, 147–154. [Google Scholar]
- Ponce, N.M.; Pujol, C.A.; Damonte, E.B.; Flores, M.L.; Stortz, C.A. Fucoidans from the brown seaweed Adenocystis utricularis: Extraction methods, antiviral activity and structural studies. Carbohydr. Res. 2003, 338, 153–165. [Google Scholar] [CrossRef]
- Asanka Sanjeewa, K.K.; Jayawardena, T.U.; Kim, H.S.; Kim, S.Y.; Shanura Fernando, I.P.; Wang, L.; Abetunga, D.T.U.; Kim, W.S.; Lee, D.S.; Jeon, Y.J. Fucoidan isolated from Padina commersonii inhibit LPS-induced inflammation in macrophages blocking TLR/NF-kappaB signal pathway. Carbohydr. Polym. 2019, 224, 115195. [Google Scholar] [CrossRef]
- Wang, J.; Liu, L.; Zhang, Q.B.; Zhang, Z.S.; Qi, H.M.; Li, P.C. Synthesized oversulphated, acetylated and benzoylated derivatives of fucoidan extracted from Laminaria japonica and their potential antioxidant activity in vitro. Food Chem. 2009, 114, 1285–1290. [Google Scholar] [CrossRef]
- Rodriguez-Jasso, R.M.; Mussatto, S.I.; Pastrana, L.; Aguilar, C.N.; Teixeira, J.A. Microwave-assisted extraction of sulfated polysaccharides (fucoidan) from brown seaweed. Carbohyd. Polym. 2011, 86, 1137–1144. [Google Scholar] [CrossRef] [Green Version]
- Sparr Eskilsson, C.; Björklund, E. Analytical-scale microwave-assisted extraction. J. Chromatogr. A 2000, 902, 227–250. [Google Scholar] [CrossRef]
- Belanger, J.M.; Pare, J.R. Applications of microwave-assisted processes (MAP) to environmental analysis. Anal. Bioanal. Chem. 2006, 386, 1049–1058. [Google Scholar] [CrossRef]
- Flórez-Fernández, N.; López-García, M.; González-Muñoz, M.J.; Vilariño, J.M.L.; Domínguez, H. Ultrasound-assisted extraction of fucoidan from Sargassum muticum. J. Appl. Phycol. 2017, 29, 1553–1561. [Google Scholar] [CrossRef]
- Hmelkov, A.B.; Zvyagintseva, T.N.; Shevchenko, N.M.; Rasin, A.B.; Ermakova, S.P. Ultrasound-assisted extraction of polysaccharides from brown alga Fucus evanescens. Structure and biological activity of the new fucoidan fractions. J. Appl. Phycol. 2017, 30, 2039–2046. [Google Scholar] [CrossRef]
- Okolie, C.L.; CK Rajendran, S.R.; Udenigwe, C.C.; Aryee, A.N.A.; Mason, B. Prospects of brown seaweed polysaccharides (BSP) as prebiotics and potential immunomodulators. J. Food Biochem. 2017, 41, e12392. [Google Scholar] [CrossRef]
- Sanjeewa, K.K.A.; Fernando, I.P.S.; Kim, E.-A.; Ahn, G.; Jee, Y.; Jeon, Y.-J. Anti-inflammatory activity of a sulfated polysaccharide isolated from an enzymatic digest of brown seaweed Sargassum horneri in RAW 264.7 cells. Nutr. Res. Practice 2016, 11, 3–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Obluchinskaya, E.D.; Pozharitskaya, O.N.; Shikov, A.N. In Vitro Anti-Inflammatory Activities of Fucoidans from Five Species of Brown Seaweeds. Mar. Drugs 2022, 20, 606. [Google Scholar] [CrossRef]
- Usov, A.I.; Bilan, M.I.; Ustyuzhanina, N.E.; Nifantiev, N.E. Fucoidans of Brown Algae: Comparison of Sulfated Polysaccharides from Fucus vesiculosus and Ascophyllum nodosum. Mar. Drugs 2022, 20, 638. [Google Scholar] [CrossRef]
- Khanra, S.; Mondal, M.; Halder, G.; Tiwari, O.N.; Gayen, K.; Bhowmick, T.K. Downstream processing of microalgae for pigments, protein and carbohydrate in industrial application: A review. Food Bioprod. Process. 2018, 110, 60–84. [Google Scholar] [CrossRef]
- Nathan, C. Points of control in inflammation. Nature 2002, 420, 846–852. [Google Scholar] [CrossRef]
- Morales, A. Intravesical therapy of bladder cancer: An immunotherapy success story. Int. J. Urol. 1996, 3, 329–333. [Google Scholar] [CrossRef]
- Kim, K.J.; Lee, B.Y. Fucoidan from the sporophyll of Undaria pinnatifida suppresses adipocyte differentiation by inhibition of inflammation-related cytokines in 3T3-L1 cells. Nutr. Res. 2012, 32, 439–447. [Google Scholar] [CrossRef]
- Samad, F.; Uysal, K.T.; Wiesbrock, S.M.; Pandey, M.; Hotamisligil, G.S.; Loskutoff, D.J. Tumor necrosis factor α is a key component in the obesity-linked elevation of plasminogen activator inhibitor 1. Proc. Natl. Acad. Sci. USA 1999, 96, 6902–6907. [Google Scholar] [CrossRef] [Green Version]
- Du, B.; Lin, C.; Bian, Z.; Xu, B. An insight into anti-inflammatory effects of fungal beta-glucans. Trends Food Sci. Technol. 2015, 41, 49–59. [Google Scholar] [CrossRef]
- Nagahawatta, D.P.; Liyanage, N.M.; Jayawardhana, H.H.A.C.; Lee, H.-G.; Jayawardena, T.U.; Jeon, Y.-J. Anti-Fine Dust Effect of Fucoidan Extracted from Ecklonia maxima Laves in Macrophages via Inhibiting Inflammatory Signaling Pathways. Mar. Drugs 2022, 20, 413. [Google Scholar] [CrossRef]
- Bhatt, D.; Ghosh, S. Regulation of the NF-kappaB-Mediated Transcription of Inflammatory Genes. Front. Immunol. 2014, 5, 71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hayden, M.S.; Ghosh, S. Shared principles in NF-kappaB signaling. Cell 2008, 132, 344–362. [Google Scholar] [CrossRef] [Green Version]
- Sanjeewa, K.K.A.; Fernando, I.P.S.; Kim, S.-Y.; Kim, H.-S.; Ahn, G.; Jee, Y.; Jeon, Y.-J. In vitro and in vivo anti-inflammatory activities of high molecular weight sulfated polysaccharide; containing fucose separated from Sargassum horneri: Short communication. Int. J. Biol. Macromol. 2018, 107, 803–807. [Google Scholar] [CrossRef] [PubMed]
- Pozharitskaya, O.N.; Obluchinskaya, E.D.; Shikov, A.N. Mechanisms of Bioactivities of Fucoidan from the Brown Seaweed Fucus vesiculosus L. of the Barents Sea. Mar. Drugs 2020, 18, 275. [Google Scholar] [CrossRef]
- Huang, P.; Han, J.; Hui, L. MAPK signaling in inflammation-associated cancer development. Protein. Cell 2010, 1, 218–226. [Google Scholar] [CrossRef] [Green Version]
- Koedel, U.; Merbt, U.M.; Schmidt, C.; Angele, B.; Popp, B.; Wagner, H.; Pfister, H.W.; Kirschning, C.J. Acute brain injury triggers MyD88-dependent, TLR2/4-independent inflammatory responses. Am. J. Pathol. 2007, 171, 200–213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sanjeewa, K.K.A.; Jayawardena, T.U.; Kim, S.-Y.; Kim, H.-S.; Ahn, G.; Kim, J.; Jeon, Y.-J. Fucoidan isolated from invasive Sargassum horneri inhibit LPS-induced inflammation via blocking NF-κB and MAPK pathways. Algal. Res. 2019, 41, 101561. [Google Scholar] [CrossRef]
- Paunovic, V.; Harnett, M.M. Mitogen-Activated Protein Kinases as Therapeutic Targets for Rheumatoid Arthritis. Drugs 2013, 73, 101–115. [Google Scholar] [CrossRef] [PubMed]
- Wu, G.-J.; Shiu, S.-M.; Hsieh, M.-C.; Tsai, G.-J. Anti-inflammatory activity of a sulfated polysaccharide from the brown alga Sargassum cristaefolium. Food Hydrocoll. 2016, 53, 16–23. [Google Scholar] [CrossRef]
- Zhang, W.; Zhang, X.; Zou, K.; Xie, J.; Zhao, S.; Liu, J.; Liu, H.; Wang, J.; Wang, Y. Seabuckthorn berry polysaccharide protects against carbon tetrachloride-induced hepatotoxicity in mice via anti-oxidative and anti-inflammatory activities. Food Funct. 2017, 8, 3130–3138. [Google Scholar] [CrossRef] [PubMed]
- Moncada, S.; Higgs, A. The L-arginine-nitric oxide pathway. N. Engl. J. Med. 1993, 329, 2002–2012. [Google Scholar] [CrossRef]
- Park, J.Y.; Pillinger, M.H.; Abramson, S.B. Prostaglandin E2 synthesis and secretion: The role of PGE2 synthases. Clin. Immunol. 2006, 119, 229–240. [Google Scholar] [CrossRef]
- Nathan, C.F. Secretory products of macrophages. J. Clin. Investig. 1987, 79, 319–326. [Google Scholar] [CrossRef] [Green Version]
- Imada, K.; Leonard, W.J. The Jak-STAT pathway. Mol. Immunol. 2000, 37, 1–11. [Google Scholar] [CrossRef]
- Ye, J.; Chen, D.; Ye, Z.; Huang, Y.; Zhang, N.; Lui, E.M.K.; Xue, C.; Xiao, M. Fucoidan Isolated from Saccharina japonica Inhibits LPS-Induced Inflammation in Macrophages via Blocking NF-kappaB, MAPK and JAK-STAT Pathways. Mar. Drugs 2020, 18, 328. [Google Scholar] [CrossRef]
- Li, B.; Lu, F.; Wei, X.; Zhao, R. Fucoidan: Structure and bioactivity. Molecules 2008, 13, 1671–1695. [Google Scholar] [CrossRef] [Green Version]
- Yang, M.; Ma, C.; Sun, J.; Shao, Q.; Gao, W.; Zhang, Y.; Li, Z.; Xie, Q.; Dong, Z.; Qu, X. Fucoidan stimulation induces a functional maturation of human monocyte-derived dendritic cells. Int. Immunopharmacol. 2008, 8, 1754–1760. [Google Scholar] [CrossRef] [PubMed]
- Kim, M.H.; Joo, H.G. Immunostimulatory effects of fucoidan on bone marrow-derived dendritic cells. Immunol. Lett. 2008, 115, 138–143. [Google Scholar] [CrossRef] [PubMed]
- Jin, J.O.; Zhang, W.; Du, J.Y.; Wong, K.W.; Oda, T.; Yu, Q. Fucoidan can function as an adjuvant in vivo to enhance dendritic cell maturation and function and promote antigen-specific T cell immune responses. PLoS ONE 2014, 9, e99396. [Google Scholar] [CrossRef] [PubMed]
- Yoo, Y.-C.; Kim, W.-J.; Kim, S.-Y.; Kim, S.-M.; Chung, M.-K.; Park, J.-W.; Suh, H.-H.; Lee, K.-B.; Park, Y.-I. Immunomodulating Activity of a Fucoidan Isolated from Korean Undaria pinnatifida Sporophyll. Algae 2007, 22, 333–338. [Google Scholar] [CrossRef]
- Do, H.; Kang, N.S.; Pyo, S.; Billiar, T.R.; Sohn, E.H. Differential regulation by fucoidan of IFN-gamma-induced NO production in glial cells and macrophages. J. Cell Biochem. 2010, 111, 1337–1345. [Google Scholar] [CrossRef]
- Nagamine, T.; Nakazato, K.; Tomioka, S.; Iha, M.; Nakajima, K. Intestinal Absorption of Fucoidan Extracted from the Brown Seaweed, Cladosiphon okamuranus. Mar. Drugs 2015, 13, 48–64. [Google Scholar] [CrossRef]
- Irhimeh, M.; Fitton, J.; Lowenthal, R.; Kongtawelert, P. A quantitative method to detect fucoidan in human plasma using a novel antibody. Methods Find. Exp. Clin. Pharmacol. 2005, 27, 705–710. [Google Scholar] [CrossRef]
- Fitton, J.H.; Stringer, D.N.; Karpiniec, S.S. Therapies from Fucoidan: An Update. Mar. Drugs 2015, 13, 5920–5946. [Google Scholar] [CrossRef] [Green Version]
- Pozharitskaya, O.N.; Shikov, A.N.; Obluchinskaya, E.D.; Vuorela, H. The Pharmacokinetics of Fucoidan after Topical Application to Rats. Mar. Drugs 2019, 17, 687. [Google Scholar] [CrossRef] [Green Version]
- Aleissa, M.S.; Alkahtani, S.; Abd Eldaim, M.A.; Ahmed, A.M.; Bungău, S.G.; Almutairi, B.; Bin-Jumah, M.; AlKahtane, A.A.; Alyousif, M.S.; Abdel-Daim, M.M. Fucoidan Ameliorates Oxidative Stress, Inflammation, DNA Damage, and Hepatorenal Injuries in Diabetic Rats Intoxicated with Aflatoxin B1. Oxidative Med. Cell. Longev. 2020, 2020, 9316751. [Google Scholar] [CrossRef] [Green Version]
- Xu, Y.; Zhang, Q.; Luo, D.; Wang, J.; Duan, D. Low molecular weight fucoidan ameliorates the inflammation and glomerular filtration function of diabetic nephropathy. J. Appl. Phycol. 2017, 29, 531–542. [Google Scholar] [CrossRef]
- Lin, H.-T.V.; Tsou, Y.-C.; Chen, Y.-T.; Lu, W.-J.; Hwang, P.-A. Effects of Low-Molecular-Weight Fucoidan and High Stability Fucoxanthin on Glucose Homeostasis, Lipid Metabolism, and Liver Function in a Mouse Model of Type II Diabetes. Mar. Drugs 2017, 15, 113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Matsumoto, S.; Nagaoka, M.; Hara, T.; Kimura-Takagi, I.; Mistuyama, K.; Ueyama, S. Fucoidan derived from Cladosiphon okamuranus Tokida ameliorates murine chronic colitis through the down-regulation of interleukin-6 production on colonic epithelial cells. Clin. Exp. Immunol. 2004, 136, 432–439. [Google Scholar] [CrossRef] [PubMed]
- Lean, Q.Y.; Eri, R.D.; Fitton, J.H.; Patel, R.P.; Gueven, N. Fucoidan Extracts Ameliorate Acute Colitis. PLoS ONE 2015, 10, e0128453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, S.-H.; Ko, C.-I.; Jee, Y.; Jeong, Y.; Kim, M.; Kim, J.-S.; Jeon, Y.-J. Anti-inflammatory effect of fucoidan extracted from Ecklonia cava in zebrafish model. Carbohyd. Polym. 2013, 92, 84–89. [Google Scholar] [CrossRef]
- Park, S.-B.; Chun, K.-R.; Kim, J.-K.; Suk, K.; Jung, Y.-M.; Lee, W.-H. The differential effect of high and low molecular weight fucoidans on the severity of collagen-induced arthritis in mice. Phytother. Res. 2010, 24, 1384–1391. [Google Scholar] [CrossRef]
- Ananthi, S.; Gayathri, V.; Malarvizhi, R.; Bhardwaj, M.; Vasanthi, H.R. Anti-arthritic potential of marine macroalgae Turbinaria ornata in Complete Freund’s Adjuvant induced rats. Exp. Toxicol. Pathol. 2017, 69, 672–680. [Google Scholar] [CrossRef]
- Phull, A.R.; Kim, S.J. Fucoidan as bio-functional molecule: Insights into the anti-inflammatory potential and associated molecular mechanisms. J. Funct. Foods 2017, 38, 415–426. [Google Scholar] [CrossRef]
- AlKahtane, A.A.; Abushouk, A.I.; Mohammed, E.T.; Alnasser, M.; Alarifi, S.; Ali, D.; Alessia, M.S.; Almeer, R.S.; AlBasher, G.; Alkahtani, S.; et al. Fucoidan alleviates microcystin-LR-induced hepatic, renal, and cardiac oxidative stress and inflammatory injuries in mice. Environ. Sci. Pollut. Res. 2020, 27, 2935–2944. [Google Scholar] [CrossRef]
- Chale-Dzul, J.; Pérez-Cabeza de Vaca, R.; Quintal-Novelo, C.; Olivera-Castillo, L.; Moo-Puc, R. Hepatoprotective effect of a fucoidan extract from Sargassum fluitans Borgesen against CCl4-induced toxicity in rats. Int. J. Biol. Macromol. 2020, 145, 500–509. [Google Scholar] [CrossRef]
- Park, J.; Cha, J.-D.; Choi, K.-M.; Lee, K.-Y.; Han, K.M.; Jang, Y.-S. Fucoidan inhibits LPS-induced inflammation in vitro and during the acute response in vivo. Int. Immunopharmacol. 2017, 43, 91–98. [Google Scholar] [CrossRef] [PubMed]
- Hu, Y.; Ren, D.; Song, Y.; Wu, L.; He, Y.; Peng, Y.; Zhou, H.; Liu, S.; Cong, H.; Zhang, Z.; et al. Gastric protective activities of fucoidan from brown alga Kjellmaniella crassifolia through the NF-κB signaling pathway. Int. J. Biol. Macromol. 2020, 149, 893–900. [Google Scholar] [CrossRef] [PubMed]
- Hwang, P.-A.; Phan, N.N.; Lu, W.-J.; Hieu, B.T.N.; Lin, Y.-C. Low-molecular-weight fucoidan and high-stability fucoxanthin from brown seaweed exert prebiotics and anti-inflammatory activities in Caco-2 cells. Food Nutr. Res. 2016, 60, 32033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Makarenkova, I.D.; Logunov, D.Y.; Tukhvatulin, A.I.; Semenova, I.B.; Besednova, N.N.; Zvyagintseva, T.N. Interactions between Sulfated Polysaccharides from Sea Brown Algae and Toll-Like Receptors on HEK293 Eukaryotic Cells In Vitro. Bull. Exp. Biol. Med. 2012, 154, 241–244. [Google Scholar] [CrossRef] [PubMed]
- Jang, J.-Y.; Moon, S.-Y.; Joo, H.-G. Differential effects of fucoidans with low and high molecular weight on the viability and function of spleen cells. Food Chem. Toxicol. 2014, 68, 234–238. [Google Scholar] [CrossRef]
- Maruyama, H.; Tamauchi, H.; Kawakami, F.; Yoshinaga, K.; Nakano, T. Suppressive effect of dietary fucoidan on proinflammatory immune response and MMP-1 expression in UVB-irradiated mouse skin. Planta Med. 2015, 81, 1370–1374. [Google Scholar]
- Myers, S.P.; Mulder, A.M.; Baker, D.G.; Robinson, S.R.; Rolfe, M.I.; Brooks, L.; Fitton, J.H. Effects of fucoidan from Fucus vesiculosus in reducing symptoms of osteoarthritis: A randomized placebo-controlled trial. Biologics 2016, 10, 81–88. [Google Scholar] [CrossRef] [Green Version]
- Nagamine, T.; Kadena, K.; Tomori, M.; Nakajima, K.; Iha, M. Activation of NK cells in male cancer survivors by fucoidan extracted from Cladosiphon okamuranus. Mol. Clin. Oncol. 2020, 12, 81–88. [Google Scholar] [CrossRef] [Green Version]
- Tabarsa, M.; Dabaghian, E.H.; You, S.; Yelithao, K.; Cao, R.; Rezaei, M.; Alboofetileh, M.; Bita, S. The activation of NF-kappaB and MAPKs signaling pathways of RAW264.7 murine macrophages and natural killer cells by fucoidan from Nizamuddinia zanardinii. Int. J. Biol. Macromol. 2020, 148, 56–67. [Google Scholar] [CrossRef]
- Lähteenmäki-Uutela, A.; Rahikainen, M.; Camarena-Gómez, M.T.; Piiparinen, J.; Spilling, K.; Yang, B. European Union legislation on macroalgae products. Aquac. Int. 2021, 29, 487–509. [Google Scholar] [CrossRef]
- Fitton, J.H.; Stringer, D.N.; Park, A.Y.; Karpiniec, S.S. Therapies from Fucoidan: New Developments. Mar. Drugs 2019, 17, 571. [Google Scholar] [CrossRef] [Green Version]
- Hsu, H.-Y.; Lin, T.-Y.; Hu, C.-H.; Shu, D.T.F.; Lu, M.-K. Fucoidan upregulates TLR4/CHOP-mediated caspase-3 and PARP activation to enhance cisplatin-induced cytotoxicity in human lung cancer cells. Cancer Lett. 2018, 432, 112–120. [Google Scholar] [CrossRef] [PubMed]
- Tocaciu, S.; Oliver, L.J.; Lowenthal, R.M.; Peterson, G.M.; Patel, R.; Shastri, M.; McGuinness, G.; Olesen, I.; Fitton, J.H. The Effect of Undaria pinnatifida Fucoidan on the Pharmacokinetics of Letrozole and Tamoxifen in Patients With Breast Cancer. Integr. Cancer Ther. 2018, 17, 99–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tsai, H.-L.; Tai, C.-J.; Huang, C.-W.; Chang, F.-R.; Wang, J.-Y. Efficacy of Low-Molecular-Weight Fucoidan as a Supplemental Therapy in Metastatic Colorectal Cancer Patients: A Double-Blind Randomized Controlled Trial. Mar. Drugs 2017, 15, 122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chauvierre, C.; Aid-Launais, R.; Aerts, J.; Chaubet, F.; Maire, M.; Chollet, L.; Rolland, L.; Bonafé, R.; Rossi, S.; Bussi, S.; et al. Pharmaceutical Development and Safety Evaluation of a GMP-Grade Fucoidan for Molecular Diagnosis of Cardiovascular Diseases. Mar. Drugs 2019, 17, 699. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Jayawardena, T.U.; Nagahawatta, D.P.; Fernando, I.P.S.; Kim, Y.-T.; Kim, J.-S.; Kim, W.-S.; Lee, J.S.; Jeon, Y.-J. A Review on Fucoidan Structure, Extraction Techniques, and Its Role as an Immunomodulatory Agent. Mar. Drugs 2022, 20, 755. https://doi.org/10.3390/md20120755
Jayawardena TU, Nagahawatta DP, Fernando IPS, Kim Y-T, Kim J-S, Kim W-S, Lee JS, Jeon Y-J. A Review on Fucoidan Structure, Extraction Techniques, and Its Role as an Immunomodulatory Agent. Marine Drugs. 2022; 20(12):755. https://doi.org/10.3390/md20120755
Chicago/Turabian StyleJayawardena, Thilina U., D. P. Nagahawatta, I. P. S. Fernando, Yong-Tae Kim, Jin-Soo Kim, Won-Suk Kim, Jung Suck Lee, and You-Jin Jeon. 2022. "A Review on Fucoidan Structure, Extraction Techniques, and Its Role as an Immunomodulatory Agent" Marine Drugs 20, no. 12: 755. https://doi.org/10.3390/md20120755
APA StyleJayawardena, T. U., Nagahawatta, D. P., Fernando, I. P. S., Kim, Y. -T., Kim, J. -S., Kim, W. -S., Lee, J. S., & Jeon, Y. -J. (2022). A Review on Fucoidan Structure, Extraction Techniques, and Its Role as an Immunomodulatory Agent. Marine Drugs, 20(12), 755. https://doi.org/10.3390/md20120755