Next Article in Journal
Editorial for the Special Issue “Current Research on Cancer Biology and Therapeutics: 2nd Edition”
Next Article in Special Issue
Edible Alginate–Lecithin Films Enriched with Different Coffee Bean Extracts: Formulation, Non-Cytotoxic, Anti-Inflammatory and Antimicrobial Properties
Previous Article in Journal
Designing Analogs of SAAP-148 with Enhanced Antimicrobial and Anti-LPS Activities
Previous Article in Special Issue
Ions-Induced Alginate Gelation According to Elemental Analysis and a Combinatorial Approach
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Systematic Characteristics of Fucoidan: Intriguing Features for New Pharmacological Interventions

1
Department of Food Science Nutrition, Pukyong National University, Busan 48513, Republic of Korea
2
Department of Smart Green Technology Engineering, Pukyong National University, Busan 48513, Republic of Korea
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(21), 11771; https://doi.org/10.3390/ijms252111771
Submission received: 4 October 2024 / Revised: 26 October 2024 / Accepted: 28 October 2024 / Published: 1 November 2024

Abstract

:
Fucoidan, a sulfated polysaccharide found primarily in brown algae, is known for exhibiting various biological activities, many of which have been attributed to its sulfate content. However, recent advancements in techniques for analyzing polysaccharide structures have highlighted that not only the sulfate groups but also the composition, molecular weight, and structures of the polysaccharides and their monomers play a crucial role in modulating biological effects. This review comprehensively provides the monosaccharide composition, degree of sulfation, molecular weight distribution, and linkage of glycosidic bonds of fucoidan, focusing on the diversity of its biological activities based on various characteristics. The implications of these findings for future applications and potential therapeutic uses of fucoidan are also discussed.

1. Introduction

Polysaccharides are complex carbohydrates composed of long chains of monosaccharide units linked together by glycosidic bonds. The structural complexity of these carbohydrates renders polysaccharides such as carrageenan, lignin, and chitosan therapeutically valuable. Among the polysaccharides with therapeutic potential, fucoidan—a marine polysaccharide derived from algae and containing fucose—exhibits a broad range of biological activities [1,2,3,4,5]. It is well known that the sulfate group content is important in determining the biological activity of algae-derived fucoidans [6,7,8]. However, recent technical advancements in the analysis of structural features such as its monosaccharide composition, degree of sulfation, molecular weight distribution, and glycosidic bond linkages have provided a more nuanced understanding of how these characteristics influence fucoidan’s biological functions [9,10,11].
With improvements in analytical techniques, it has become possible to more accurately characterize the structural properties of fucoidan, a complex marine sulfated biopolysaccharides with heterogeneous chemical structures, which were previously difficult to elucidate [12,13]. As a result, there has been a surge in studies investigating the relationship between fucoidan’s physicochemical characteristics and bioactivity [14]. While early research focused primarily on the correlation between the degree of sulfation and its bioactivity, more recent studies have examined the impact of other structural features, including monosaccharide composition, molecular weight distribution, and glycosidic bond linkages [15,16,17]. This review presents the activities of fucoidan based on its characteristics, focusing on its biological activities, including its anticoagulant, antitumor, anticancer, anti-inflammatory, antioxidant, immunomodulatory, hypocholesterolemic, antibacterial, and anti-SARS-CoV-2 effects.
Emerging evidence suggests that the monosaccharide ratio and types of monosaccharides, such as fucose, galactose, and mannose, play a crucial role in modulating both the efficacy and mechanisms of action of fucoidan’s biological effects [18].
This review aims to synthesize recent findings on how the structural composition of fucoidan influences its biological activities, providing a comprehensive overview of how these functional properties are determined (Table S1). Additionally, it explores how these insights can inform future research directions and guide the development of potential therapeutic applications of fucoidan in various fields.

2. The Role of Monosaccharides of Fucoidan in Bioactivity

Fucoidan, a polysaccharide derived from algae, is notable for its high fucose content, a monosaccharide that significantly contributes to its bioactivity [19,20]. The relationship between fucose content and the biological activities of fucoidan—such as its antioxidant, anticoagulant, antiviral, and anti-inflammatory effects—has been well established [21,22,23,24,25,26]. Research indicates that fucoidan with a higher fucose content often exhibits enhanced biological efficacy [22,27]. For instance, fucoidans with elevated fucose concentrations tend to exhibit stronger anticoagulant and anti-inflammatory properties, likely due to the unique sulfation patterns associated with fucose residues [28,29,30]. However, while fucose is a key component, its content alone does not solely determine the bioactivity of the polysaccharide.
Additionally, other monosaccharides present in fucoidan, such as galactose, mannose, glucose, and xylose, play significant roles in modulating its overall bioactivity [31,32,33]. These sugars influence the molecular weight, structural configuration, and functional properties of fucoidan, thereby affecting its interactions with biological targets [34,35,36]. For instance, mannose and galactose have been shown to enhance the antiviral and immunomodulatory effects of fucoidan, while glucose and xylose can impact its antioxidant capacity [37,38,39]. Thus, while fucose is crucial for fucoidan’s activity, the specific composition and ratio of other monosaccharides are equally vital for optimizing its therapeutic potential (Table 1).
Research has demonstrated that the presence of monosaccharides in fucoidan can significantly influence its bioactivity. For example, Rashed et al. (2021) identified a fucoidan fraction containing fucose, glucose, galactose, and mannose that exhibited high antioxidant capacity and antisteatotic action [40]. Ptak et al. (2021) suggested that glucose content may affect bioactivity, likely due to the presence of laminarin in the extract [41]. Furthermore, Dobrinčić et al. (2021) noted that extraction conditions could alter the content of various monosaccharides and sulfate groups, thereby influencing fucoidan’s properties [42]. The arrangement of these monosaccharides, together with the degree of sulfation, plays a crucial role in determining fucoidan’s overall bioactivity, emphasizing the importance of both sugar composition and sulfation patterns in its therapeutic potential.
The biological effects of fucoidan are closely tied to its specific monosaccharide composition, which varies significantly across different algae species. For instance, Bilan et al. (2017) extracted fucoidan from Sargassum aquifolium (Phaeophyceae, Fucales), predominantly composed of fucose, galactose, mannose, glucuronic acid, and xylose, which have demonstrated robust anticoagulant and anticancer effects in vitro, as evidenced by their activity against HePG2, A549, and HBL-100 cell lines [43]. Similarly, polysaccharides from Undaria pinnatifida (Phaeophyceae, Laminariales), containing fucose, glucuronic acid, galactose, and mannose, exhibit potent antioxidant properties in zebrafish models at concentrations starting from 0.625 mM, while also showing efficacy in attenuating SARS-CoV-2 infection in hamster models at dosages of 100 and 200 mg/kg body weight [44,45]. The polysaccharides from Fucus vesiculosus (Phaeophyceae, Fucales), which consist of fucose, xylose, galactose, and uronic acids, have been shown to suppress inflammatory responses in RAW 264.7 macrophage cells and demonstrate significant antioxidant activity in zebrafish models at a concentration of 0.625 mM [45,46]. Additionally, Sargassum fusiforme (Phaeophyceae, Fucales) polysaccharides, rich in fucose, galactose, and mannose, have been reported to exhibit anti-inflammatory effects in both RAW 264.7 macrophages and zebrafish embryos at concentrations of 25, 50, and 100 μg/mL [47].
Moreover, the polysaccharides of Fucus serratus (Phaeophyceae, Fucales), containing fucose, galactose, glucuronic acid, and xylose, and Fucus distichus subsp. evanescens (formerly Fucus evanescens), containing fucose, galactose, and xylose, have also shown remarkable potential in promoting bone formation and vascularization, as demonstrated in human outgrowth endothelial cells (OECs) and mesenchymal stem cells (MSCs) at a concentration of 100 μg/mL [48]. In terms of immunomodulatory and antiviral activity, Saccharina latissima (Phaeophyceae, Laminariales) polysaccharides, composed of fucose, xylose, galactose, mannose, glucuronic acid, and uronic acids, have been found to stimulate lymphocyte proliferation in spleen cells derived from BALB/c mice at concentrations ranging from 25 to 250 μg/mL [49,50]. Furthermore, Stoechospermum polypodioides (formerly Stoechospermum marginatum) (Phaeophyceae, Dictyotales) polysaccharides, composed of fucose, xylose, mannose, galactose, glucose, and galacturonic acid, have shown potent antiviral activity against a range of viruses in Vero cells, with an effective concentration (EC50) of 3.5 ± 0.63 μg/mL [51]. Polysaccharides from Cladosiphon okamuranus (Phaeophyceae, Ectocarpales), characterized by a high content of fucose and uronic acid, have demonstrated effectiveness in mitigating atopic dermatitis symptoms through immunomodulation in BALB/c mice models [52].
Furthermore, Saccharina japonica (formerly Laminaria japonica) (Phaeophyceae, Laminariales) polysaccharides, comprising fucose, mannose, and glucose, have been reported to possess antibacterial properties and anti-SARS-CoV-2 activity in studies involving various bacterial strains and viral infection models, with effective concentrations ranging from 6.25 to 50 μg/mL [53]. Collectively, the diversity in monosaccharide composition among these polysaccharides underpins their wide-ranging bioactivities, offering a compelling basis for their application in therapeutic interventions. However, to fully exploit the bioactive potential of these algae-derived polysaccharides, further research is needed to refine the methods for extraction, purification, and characterization, thereby enabling more precise control over their composition and enhancing their therapeutic efficacy across various biomedical domains.
The bioactivity of fucoidan is not solely dependent on its fucose content; it is the intricate interplay between fucose and other monosaccharides that determines its full range of biological effects. Understanding these relationships is crucial for optimizing the use of fucoidan in various biomedical applications. While fucose is an essential component of fucoidan, its content alone does not dictate bioactivity. The sulfate content, the presence of other monosaccharides, and the overall structural characteristics of the polysaccharide all contribute to its biological activities. This complex interplay highlights the importance of considering multiple factors when evaluating the potential therapeutic applications of fucoidan.
Table 1. Bioactivity according to monosaccharide composition of brown algae fucoidan.
Table 1. Bioactivity according to monosaccharide composition of brown algae fucoidan.
Algae SourceMonosaccharide CompositionBioactivityExperimental MethodologiesExperiment ModelConcentration Reference
Sargassum aquifoliumFuccose, galactose, mannose, glucuronic acid, xyloseAnticoagulant and antitumor activitiesIn vitroHuman cancer cell lines HepG2 (hepatocellular carcinoma), LU-1 (lung adenocarcinoma), and RD (rhabdomiosarcoma) ND[43]
Sargassum plagiophyllumFucose, galactose, xylose, mannoseAnticancer activityIn vitroHepG2, A549, and HBL-100ND[54]
Sargassum horneriFucose, galactose, mannose, xylose, rhamnoseAnti-inflammatory activityIn vitroRAW 264.7 cellsEC50 (μg/mL): 87.12[22]
Fucose, galactose, mannose, glucuronic acidAntitumorIn vitroThe DLD-1 (ATCC # CCL-221™) human colon carcinoma cell line200 μg/mL[55]
Undaria pinnatifidaFucose, glucuronic acid, galactose, mannose Antioxidant activitiesIn vivoZebrafish0.625 mM[45]
Fucose, glucose, galactoseAnticancer activityIn vivoSprague Dawley rats100, 200, and 300 mg/kg of body weight[56]
Fucose, glucuronic acid, galactose, mannoseAttenuation of SARS-CoV-2 infectionIn vitro
In vivo
Caco-2-Nint cells, a producer cell line expressing the SARS-CoV-2 N protein via lentiviral transduction 7.8, 15.6, 31.3, 62.5, 125, 500, and 1000 μg/mL[44]
Ascophyllum nodosumFucose, galactose, mannose, glucuronic acid, uronic acidFour-week-old female specific-pathogen-free (SPF) Syrian hamstersOrally gavaged with high dose (Hd; 200 mg/day/kg body weight) or low dose (Ld; 100 mg/day/kg body weight)
Dictyopteris divaricataFucose, xylose, mannose, glucose, galactoseAntioxidant and immunomodulatory activitiesIn vivoRAW 264.7 murine macrophagesND[57]
Sargassum crassifoliumFucose, galactoseImmunomodulatory activityIn vitroBone marrow cells from C3H/HeJ female mice3 μg/mL[58]
Stoechospermum polypodioidesFucose, xylose, mannose, galactose, glucose, galacturonic acidAntiviral activityIn vitroVero cells by a virus plaque reduction assayEC50 (μg/mL): 3.55 ± 0.63[51]
Sargassum polycystumFucose, xylose, mannose, galactose, glucose, rhamnoseAntioxidant activity, anticancer activityIn vitroMCF-7 cells25, 50, 75, 100, 125, and 150 μg/mL[59]
Sargassum siliquosumFucose, xylose, mannose, galactose, glucose, rhamnoseAntioxidant activity, anti-inflammatory activityIn vitroRAW 264.7 cell0.25–1 μg/mL[15]
Fucus serratusFucose, galactose, glucuronic acid, xyloseBone formation and vascularizationIn vitroHuman outgrowth endothelial cells (OECs), mesenchymal stem cells (MSCs)100 μg/mL[48]
Fucus distichus subsp. evanescensFucose, xylose, galactose
Fucose, xylose, mannose, galactose, glucose, glucuronic acidAnticancer activityIn vitroThe SK-MEL-5 (ATCC # HTB-70), SK-MEL-28 (ATCC # HTB-72) human malignant melanoma cell lines100–400 μg/mL[60]
Saccharina latissimaFucose, xylose, galactose, mannose, glucuronic acid, uronic acidImmunostimulatory, hypocholesterolemic activities In vitroLymphocyte Stimulatory Activity (spleen cell of BALB/c mice)
Assessment of Hypocholesterolemic Effect (in vitro intestinal model)
25, 100, and 250 μg/mL[49,50]
Cladosiphon okamuranusFucose, uronic acidAlleviates atopic dermatitis symptoms through immunomodulation In vitro
In vivo
RAW 264.7 cells;
atopic dermatitis (AD) model_Male BALB/c mice aged 6 weeks old
31.25, 62.5, 125, 250, 500, and 1000 μg/mL
100 mg of the cream
[52]
Sargassum fusiformeFucose, galactose, mannoseAnti-inflammatoryIn vitro
In vivo
RAW 264.7 macrophages;
zebrafish embryo
25, 50, and 100 μg/mL[47]
Macrocystis pyriferaFucose, Xylose, Glucuronic acidAntioxidant activitiesIn vivoZebrafish0.625 mM[45]
Padina boergeseniiFucose, galactose, glucose, xyloseAntioxidant and anticancerIn vitroHuman cervical carcinoma cells (HeLa cell line)20, 40, and 60 μg/mL[61]
Fucus vesiculosusFucose, xylose, galactose, mannoseAntioxidant activitiesIn vivoZebrafish0.625 mM[45]
Fucose, xylose, galactose, uronic acidsInhibition of inflammatory responseIn vitroRAW 264.7 macrophages 0.1 μg/mL[46]
Saccharina japonicaFucose, mannose, glucoseAntibacterial activity and anti SARS-CoV-2In vitroBacteria including Staphylococcus aureus ATCC6538, Listeria monocytogenes ATCC19115, Escherichia coli ATCC25922, Shigella flexneri CMCC51574, Salmonella typhimurium ATCC14028, and Vibrio parahaemolyticus CGMCC1.16140, 6.25, 12.5, 25, and 50 μg/mL[53]
Ishige okamuraeFucose, galactose, glucose, xyloseEffect on recovery from immunosuppressionIn vivoBALB/c mice induced CTX (cyclooxygenase-thromboxane A2 synthetase) immunomodulatory models20, 40, and 80 mg/kg[62]
ND: no data.

3. The Role of Sulfate of Fucoidan in Bioactivity

The correlation between the degree of sulfation in fucoidan and its bioactivity is significant and well documented across multiple studies [63,64,65,66,67,68]. The degree of sulfation varies among different algae species, and numerous studies have shown that sulfate content plays a pivotal role in modulating fucoidan’s biological efficacy (Table 2). Research indicates that sulfate content is a crucial factor in determining fucoidan’s biological activities, particularly its antioxidant and anticoagulant properties. These effects, however, are influenced by the structural characteristics of fucoidan. The sulfate groups in fucoidan are primarily attached to fucose residues, often at the C-2 and C-4 positions [69]. The sulfation pattern directly impacts its bioactivity, with higher sulfate content imparting a stronger negative charge, which enhances its binding interactions with specific proteins.
The impact of sulfation on fucoidan’s bioactivity also extends to its anticoagulant properties. A minimum charge density of 0.5 sulfate groups per sugar unit is required for fucoidan to exhibit effective procoagulant activity in factor VIII/factor IX-deficient plasma [70]. For instance, polysaccharides extracted from Sargassum aquifolium (Phaeophyceae, Fucales), though with an unspecified sulfate content, have shown anticoagulant and antitumor activities in vitro, specifically against human epithelial carcinoma cell lines (HepG2, A549, and HBL-100) [43]. In contrast, polysaccharides derived from Sargassum horneri (Phaeophyceae, Fucales), with a defined sulfate content of 18.47%, have demonstrated significant anti-inflammatory properties, inhibiting inflammatory responses in RAW 264.7 cells with an IC50 value of 87.12 μg/mL, suggesting that higher sulfation enhances their anti-inflammatory potential [22].
Other algae species such as Undaria pinnatifida (Phaeophyceae, Laminariales) and Fucus distichus subsp. evanescens exhibit notable anticancer properties, with sulfate contents of 29.14% and 28%, respectively [56,60]. Polysaccharides from Undaria pinnatifida have shown anticancer efficacy in vivo in Sprague Dawley rats, with concentrations of fucoidans ranging from 100 to 300 mg/kg body weight. Similarly, polysaccharides from Fucus distichus subsp. evanescens demonstrate anticancer activity in human malignant melanoma cell lines at fucoidan concentrations of 100–400 μg/mL. These findings underscore the importance of sulfate groups in enhancing the anticancer effects of algae-derived polysaccharides. The degree of sulfation and the position of sulfate groups on the fucoidan backbone are directly related to its potential activities [71]. For instance, 4O-sulfation has been shown to significantly contribute to the anticancer activity of fucoidans, and when 4O-sulfation was removed using an endo-sulfatase, the resulting desulfated fucoidans exhibited reduced inhibition of colony formation in cancer cells [72].
Polysaccharides from Fucus serratus and Fucus distichus subsp. evanescens have been shown to promote bone formation and vascularization, with sulfate contents of 21.54% and 46.88%, respectively. The bioactivity of these polysaccharides has been observed in human outgrowth endothelial cells (OECs) and mesenchymal stem cells (MSCs) at concentrations of 100 μg/mL. The higher sulfate content in Fucus distichus subsp. evanescens appears to enhance its effectiveness in promoting bone formation and vascularization, suggesting a correlation between sulfate content and biological activity in tissue regeneration [48].
Polysaccharides from Saccharina latissima, with a sulfate content of 14.3%, have exhibited immunostimulatory and hypocholesterolemic activities in vitro. The presence of sulfate groups is believed to play a significant role in modulating the immune response [49,50]. Similarly, Cladosiphon okamuranus polysaccharides, containing 17.6% sulfate, have been effective in alleviating atopic dermatitis symptoms through immunomodulation, as demonstrated in RAW 264.7 cell models and BALB/c mice. These studies further support the link between sulfation and immunomodulatory activities in algae-derived polysaccharides [52].
Antioxidant and antiviral properties are also closely associated with sulfate content. Polysaccharides from Macrocystis pyrifera, containing 26.0 ± 0.6% sulfate, have demonstrated antioxidant activity in zebrafish models, providing protective effects against oxidative stress [45]. The ratio of sulfate content to fucose has been proposed as an effective indicator of antioxidant activity in fucoidan samples. Higher sulfate content enhances fucoidan’s effectiveness as an antioxidant, likely due to its increased capacity to scavenge free radicals. On the antiviral front, Stoechospermum polypodioides, with a sulfate content of 13%, exhibits potent antiviral activity in vitro, with studies on Vero cells showing an effective concentration (EC50) of 3.5 ± 0.63 μg/mL, highlighting the role of sulfate groups in boosting antiviral efficacy [51].
Polysaccharides from Saccharina japonica, with sulfate contents of 28.7 ± 2.6%, 24.7 ± 0.9%, and 28.7 ± 2.6% in different fucoidan fractions, have shown both antibacterial and anti-SARS-CoV-2 activities. These findings suggest that varying levels of sulfation influence their antiviral and antibacterial properties [53]. Additionally, Ascophyllum nodosum polysaccharides, with a sulfate content of 22.6 ± 0.8%, have demonstrated promising results in attenuating SARS-CoV-2 infection in Syrian hamster models, further supporting the potential of sulfated polysaccharides as therapeutic agents against viral infections [44].
Interestingly, the relationship between sulfation and bioactivity is not always straightforward. In some cases, increasing sulfate content enhances certain activities while diminishing others. For instance, when the sulfate content of low-molecular-mass fucoidan was increased from 18.7% to 32.1%, its antioxidant and anti-inflammatory activities improved, but its anti-lipogenesis activity decreased [73].
The degree of sulfation in algae-derived polysaccharides is a key determinant of their bioactivity, with higher sulfate content often correlating with increased biological efficacy. These findings highlight the need for further research to optimize sulfation processes and elucidate the underlying mechanisms by which sulfate groups enhance the bioactive properties of these compounds. Such research could pave the way for the development of novel therapeutic agents based on sulfated polysaccharides from algae.
Table 2. Influence of sulfate group content and monosaccharide distribution in brown algae fucoidan on its bioactivity.
Table 2. Influence of sulfate group content and monosaccharide distribution in brown algae fucoidan on its bioactivity.
Algae SourceSulfate (%)Fuc (%)Glc (%)Xyl (%)Man (%)GlcA (%)Rha
(%)
Gal (%)UAs (%)BioactivityExperimental MethodologiesExperiment ModelConcentration Reference
Sargassum aquifoliumND9.2ND2.222.2ND8.512.6Anticoagulant and antitumor activitiesIn vitroHuman cancer cell lines HepG2 (hepatocellular carcinoma), LU-1 (lung adenocarcinoma), and RD (rhabdomiosarcoma) ND[43]
Sargassum plagiophyllumF1: 9.8
F2: 21.9
F3: 15.1
F1: 55.5
F2: 71.1
F3: 69.1
NDF1: 4.5
F2: 2.5
F3: 1.9
F1: 15.7
F2: 11.2
F3: 9.9
NDNDF1: 22.9
F2: 13.5
F3: 12.2
F1: 22.9
F2: 12.6
F3: 16.3
Anticancer activityIn vitroHepG2, A549, and HBL-100IC50
F1: 800 μg/mL
F2: 600 μg/mL
F3: 700 μg/mL
[54]
Sargassum horneri18.4736.86ND7.3811.27ND5.2330.09NDAnti-inflammatory activityIn vitroRAW 264.7 cellsIC50 = 87.12 μg/mL[22]
418510NDNDNDND5NDAntitumorIn vitroThe DLD-1 (ATCC # CCL-221™) human colon carcinoma cell line200 ug/mL[55]
Sargassum crassifolium27.554.36ND1.490.6NDND43.557.6Immunomodulatory activityIn vitroBone marrow cells from C3H/HeJ female mice3 μg/mL[74]
Stoechospermum polypodioides1396ND2NDNDND2NDAntiviral activityIn vitroVero cells by a virus plaque reduction assayEC50 (μg/mL): 3.55 ± 0.63[51]
Sargassum polycystum22.35 ± 0.2346.811.513.25.6ND8.614.3NDAntioxidant activity, anticancer activityIn vitroMCF-7 cells25, 50, 75, 100, 125, and 150 μg/mL[59]
Sargassum siliquosum6.01 ± 0.5347.13 ± 0.478.53 ± 4.139.07 ± 0.386.97 ± 2.93ND3.47 ± 0.1224.83 ± 0.74NDAntioxidant activity, anti-inflammatory activityIn vitroRAW 264.7 cell 0.25–1 μg/mL[15]
Fucus serratus21.5476.2ND6.5ND11.2ND3.3NDBone formation and vascularizationIn vitroHuman outgrowth endothelial cells (OECs), mesenchymal stem cells (MSCs)100 μg/mL[48]
Fucus distichus subsp. evanescens46.8876.7ND9.8NDNDND5.7ND
2887.11.31.84.42ND1.6NDAnticancer activityIn vitroThe SK-MEL-5 (ATCC # HTB-70), SK-MEL-28 (ATCC # HTB-72) human malignant melanoma cell lines100–400 μg/mL[60]
Saccharina latissima14.359.1 ± 2.73.2 ± 1.63.0 ± 1.12.0 ± 0.6NDND20.8 ± 4.212.0 ± 2.1Immunostimulatory, hypocholesterolemic activities In vitroLymphocyte Stimulatory Activity (spleen cell of BALB/c mice)
Assessment of Hypocholesterolemic Effect (in vitro intestinal model)
25, 100, 250 μg/mL[49,50]
Cladosiphon okamuranus17.652.7NDNDNDNDNDND18Alleviates atopic dermatitis symptoms through immunomodulation In vitro
In vivo
RAW 264.7 cells;
Atopic dermatitis (AD) model_Male BALB/c mice aged 6 weeks old
31.25, 62.5, 125, 250, 500, and 1000 μg/mL[52]
Sargassum fusiforme17.6 ± 0.3646.32NDND24ND1.1727.36NDAnti-inflammatoryIn vitro
In vivo
RAW 264.7 macrophages;
zebrafish embryo
25, 50, and 100 μg/mL[47]
Macrocystis pyrifera26.0 ± 0.6NDNDNDNDNDNDNDNDAntioxidant activitiesIn vivo Zebrafish0.625 mM[45]
Padina boergesenii17.72 ± 0.2543.1 ± 0.2311.6 ± 0.1014.2 ± 0.12NDNDND17.3 ± 0.179.43 ± 0.17Antioxidant and anticancerIn vitroHuman cervical carcinoma cells (HeLa cell line)20, 40, and 60 μg/mL[61]
Fucus vesiculosus30.8 ± 4.2NDNDNDNDNDNDNDNDAntioxidant activitiesIn vivo Zebrafish0.625 mM[45]
9.9 ± 2.990.4 ± 2.0ND2.4 ± 0.7NDNDND3.3 ± 0.73.8 ± 0.7Inhibition of inflammatory responseIn vitroRAW 264.7 macrophages 0.1 μg/mL[46]
Saccharina japonicaFucoidan: 28.7 ± 2.6
Dfuc1: 24.7 ± 0.9
Dfuc2: 23.3 ± 1.0
Fucoidan: 69.14
Dfuc1: 69.84
Dfuc2: 58.55
NDNDFucoidan: 12.86
Dfuc1: 13.69
Dfuc2: 20.73
NDNDFucoidan: 18.00
Dfuc1: 16.43
Dfuc2: 20.73
Fucoidan: 16.2 ± 0.4
Dfuc1: 15.0 ± 0.6
Dfuc2: 15.5 ± 1.0
Antibacterial activity and anti SARS-CoV-2 In vitroBacteria including Staphylococcus aureus ATCC6538, Listeria monocytogenes ATCC19115, Escherichia coli ATCC25922, Shigella flexneri CMCC51574, Salmonella typhimurium ATCC14028, and Vibrio parahaemolyticus CGMCC1.161450, 25, 12.5, 6.25, and 0 μg/mL[53]
Undaria pinnatifida29.1427.1519.34NDNDNDND53.513.21Anticancer activityIn vivoSprague Dawley rats100, 200, and 300 mg/kg of body weight[56]
25.1 ± 1.4NDNDNDNDNDNDNDNDAntioxidant activitiesIn vivo Zebrafish0.625 mM[45]
29.9 ± 0.592.7NDND1.51ND33.1NDAttenuation of SARS-CoV-2 infection In vitro
In vivo
Caco-2-Nint cells, a producer cell line expressing the SARS-CoV-2 N protein via lentiviral transduction 7.8, 15.6, 31.3, 62.5, 125, 500, and 1000 μg/mL [44]
Ascophyllum nodosum22.6 ± 0.856.9NDND1.51ND7.45.3 ± 0.1Four-week-old female specific-pathogen-free (SPF) Syrian hamstersOrally gavaged with high dose (Hd; 200 mg/day/kg body weight) or low dose (Ld; 100 mg/day/kg body weight
Ishige okamurae27.659ND109ND811NDEffect on recovery from immunosuppressionIn vivoBALB/c mice induced CTX (cyclooxygenase-thromboxane A2 synthetase) immunomodulatory models20, 40, and 80 mg/kg[62]
ND, no data; Fuc, fucose; Glc, glucose; Xyl, xylose; Man, mannose; GlcA, glucuronic acid; Rha, rhamnose; Gal, galactose; UAs: uronic acids.

4. The Role of Molecular Weight of Fucoidan in Bioactivity

Fucoidan and its molecular weight play a pivotal role in determining various bioactivities [75,76,77,78]. Generally, higher-molecular-weight fucoidan tends to exhibit greater bioactivity; however, the degree of sulfation can influence this effect. The relationship between molecular weight and bioactivity is not always consistent. As molecular weight increases, the number of potential sulfation sites also rises, which can lead to stronger biological activities in vivo, such as antiviral effects by binding to pathogens or viruses and blocking their activity. Additionally, lower-molecular-weight fucoidans likely enhance absorption and interaction with cellular targets, contributing to significant biological effects, including modulation of cellular signaling pathways involved in cell proliferation and immune regulation, as well as direct anticancer and antioxidant activities. Research has shown that the molecular weight of algae-derived fucoidan significantly affects its solubility, absorption, distribution, and interactions with biological targets, ultimately modulating its therapeutic potential (Table 3). Undaria pinnatifida contains fucoidan with a molecular weight of 97.9 kDa, demonstrating anticancer activity in vitro, particularly in studies using Sprague Dawley rats at dosages of 100, 200, and 300 mg/kg of body weight [56]. This molecular weight may reflect an optimal balance between bioavailability and biological efficacy. Fucoidans from Fucus distichus subsp. evanescens, with a molecular weight of 60 kDa, have shown strong anticancer activity in human malignant melanoma cell lines, and their relatively low molecular weight likely facilitates enhanced cellular uptake and more effective interaction with cancer cells, demonstrating higher cytotoxicity and greater inhibitory activity of cell transformation, thus resulting in enhanced anticarcinogenicity [60]. In contrast, Sargassum horneri contains fucoidan with a low-to-high molecular weight range of 20–140 kDa, exhibiting significant antitumor activity in human colon carcinoma cells [55].
Conversely, fucoidans with higher molecular weights, such as those derived from Fucus serratus, which have a molecular weight of 272 kDa, have been shown to promote bone formation and vascularization, as demonstrated in human outgrowth endothelial cells (OECs) and mesenchymal stem cells (MSCs) [48]. The high molecular weight of these fucoidans may provide structural advantages that are crucial for scaffolding in bone and vascular applications, enhancing binding capabilities and prolonging retention in biological systems, leading to sustained therapeutic effects. In addition, fucoidans from Fucus distichus subsp. evanescens, with a molecular weight of 84 kDa, have similarly been found to promote bone formation and vascularization, with a molecular weight offering a balance between bioactivity and tissue penetration [48].
In terms of immune modulation, fucoidans from Saccharina latissima, with a molecular weight of 137 kDa, have shown immunostimulatory activity in assays featuring spleen cell derived from BALB/c mice, suggesting that this molecular weight may be optimal for triggering immune responses [49,50]. On the other hand, Cladosiphon okamuranus contains fucoidans with a molecular weight of 49.8 kDa, which have been effective in alleviating atopic dermatitis symptoms through immunomodulation in both BALB/c mice and RAW 264.7 cells [52]. It has been found that the lower molecular weight of these fucoidans may allow for more efficient absorption and interaction with immune cells, thereby enhancing their immunomodulatory effects.
Fucoidans from Macrocystis pyrifera, with a low molecular weight of 70.4 kDa, have demonstrated antioxidant activity in zebrafish models [45]. The molecular weight of these fucoidans likely optimizes solubility and cellular uptake, contributing to their antioxidant properties. Similarly, fucoidans from Stoechospermum polypodioides, with a molecular weight of 40 kDa, have exhibited potent antiviral activity [51]. Fucoidans with lower molecular weights are often more effective in antiviral applications, as their size facilitates better penetration of viral envelopes and interaction with viral proteins. In the case of Saccharina japonica, fucoidans with varying molecular weights, including 90.8 kDa, have demonstrated antibacterial and anti-SARS-CoV-2 activity [53].
Similarly, fucoidans from Ascophyllum nodosum, with a molecular weight of 124.3 kDa, have shown promise in attenuating SARS-CoV-2 infection in Syrian hamsters, suggesting that higher molecular weights may contribute to the stability and duration of antiviral effects [44]. The relationship between molecular weight and bioactivity is evident across various algae species, with relatively low-molecular-weight fucoidans generally showing better absorption, solubility, and bioavailability, which enhances their biological effects, such as their anti-inflammatory and antiviral activities. Conversely, higher-molecular-weight fucoidans may offer structural advantages and prolonged therapeutic effects, making them suitable for applications such as bone formation and immune modulation. Based on these studies, future research should emphasize that the specific effects of fucoidan can vary depending on the particular bioactivity being investigated, highlighting the importance of adjusting fucoidan’s molecular weight to suit the desired applications [79].
Table 3. Relationship between molecular weight of brown algae source and bioactivity.
Table 3. Relationship between molecular weight of brown algae source and bioactivity.
Algae SourceMolecular Weight (kDa)BioactivityExperimental MethodologiesExperiment ModelConcentration Reference
Sargassum aquifoliumNDAnticoagulant and antitumor activitiesIn vitroHuman cancer cell lines HepG2 (hepatocellular carcinoma), LU-1 (lung adenocarcinoma), and RD (rhabdomiosarcoma) ND[43]
Sargassum plagiophyllumF1: 20
F2: 35
F3: 30
Anticancer activityIn vitroHepG2, A549, and HBL-100ND[54]
Sargassum horneri20–140AntitumorIn vitroThe DLD-1 (ATCC # CCL-221™) human colon carcinoma cell line200 μg/mL[55]
30Anti-inflammatory activityIn vitroRAW 264.7 cellsIC50 = 87.12 μg/mL[22]
Dictyopteris divaricata58.05Antioxidant and immunomodulatory activitiesIn vivoRAW 264.7 murine macrophagesND[57]
Sargassum crassifolium230Immunomodulatory activityIn vitroBone marrow cells from C3H/HeJ female mice3 μg/mL[74]
Stoechospermum polypodioides40Antiviral activityIn vitroVero cells by a virus plaque reduction assayEC50 (μg/mL): 3.55 ± 0.63[51]
Sargassum polycystumNDAntioxidant activity, anticancer activityIn vitroMCF-7 cells25, 50, 75, 100, 125, and 150 μg/mL [59]
Sargassum siliquosumNDAntioxidant activity, anti-inflammatory activityIn vitroRAW 264.7 cell 0.25–1 μg/mL[15]
Fucus serratus272Bone formation and vascularizationIn vitroHuman outgrowth endothelial cells (OECs), mesenchymal stem cells (MSCs)100 μg/mL[48]
Fucus evanescens84
60Anticancer activityIn vitroThe SK-MEL-5 (ATCC # HTB-70), SK-MEL-28 (ATCC # HTB-72) human malignant melanoma cell lines100–400 μg/mL [60]
Saccharina latissima137Immunostimulatory, hypocholesterolemic activities In vitroLymphocyte Stimulatory Activity (spleen cell of BALB/c mice)
Assessment of Hypocholesterolemic Effect (in vitro intestinal model)
25, 100, and 250 μg/mL[49,50]
Cladosiphon okamuranus49.8Alleviates atopic dermatitis symptoms through immunomodulation In vitro
In vivo
RAW 264.7 cells;
atopic dermatitis (AD) model_Male BALB/c mice aged 6 weeks old
31.25, 62.5, 125, 250, 500, and 1000 μg/mL[52]
Sargassum fusiforme60–150Anti-inflammatoryIn vitro
In vivo
RAW 264.7 macrophages;
zebrafish embryo
25, 50, and 100 μg/mL [47]
Macrocystis pyrifera70.4Antioxidant activitiesIn vivo Zebrafish0.625 mM[45]
Padina boergesenii224Antioxidant and anticancerIn vitroHuman cervical carcinoma cells (HeLa cell line)20, 40, and 60 μg/mL[61]
Fucus vesiculosus97.7Antioxidant activitiesIn vivo Zebrafish0.625 mM[45]
70Inhibition of inflammatory responseIn vitroRAW 264.7 macrophages 0.1 μg/mL[46]
Saccharina japonicaFucoidan: 90.8
Dfuc1: 19.2
Dfuc2: 5.5
Antibacterial activity and anti SARS-CoV-2In vitroBacteria including Staphylococcus aureus ATCC6538, Listeria monocytogenes ATCC19115, Escherichia coli ATCC25922, Shigella flexneri CMCC51574, Salmonella typhimurium ATCC14028, and Vibrio parahaemolyticus CGMCC1.161450, 25, 12.5, 6.25, and 0 μg/mL[53]
Undaria pinnatifida97.9Anticancer activityIn vivoSprague Dawley rats100, 200, and 300 mg/kg of body weight[56]
168.5Antioxidant activitiesIn vivoZebrafish0.625 mM[45]
141.7Attenuation of SARS-CoV-2 infection In vitro
In vivo
Caco-2-Nint cells, a producer cell line expressing the SARS-CoV-2 N protein via lentiviral transduction 7.8, 15.6, 31.3, 62.5, 125, 500, and 1000 μg/mL[44]
Ascophyllum nodosum124.3Four-week-old female specific-pathogen-free (SPF) Syrian hamstersOrally gavaged with high dose (Hd; 200 mg/day/kg body weight) or low dose (Ld; 100 mg/day/kg body weight)
1.4–40Antioxidant activityIn vitroScavenging activity on DPPH radical [80]
Ishige okamurae12.9Effect on recovery from immunosuppressionIn vivoBALB/c mice induced CTX (cyclooxygenase-thromboxane A2 synthetase) immunomodulatory models20, 40, and 80 mg/kg[62]
ND: no data.

5. The Role of Glycosidic Linkage of Fucoidan in Bioactivity

Fucoidan’s bioactivity is significantly influenced by its diverse glycosidic bonds, which are crucial for determining its biological functions (Table 4). Typically, fucoidan’s backbone is composed of α-L-fucopyranose residues linked mainly through (1→3) and (1→4) glycosidic linkages [81,82]. These linkages not only determine the structural conformation of fucoidan but also affect its solubility, interactions with biological targets, and overall bioactivity. The configuration, position, and type of these glycosidic bonds greatly impact fucoidan’s antioxidant, anti-inflammatory, anticancer, antiviral, and immunomodulatory properties.
In particular, the ratio of (1→3) to (1→4) linkages influences how fucoidan interacts with different enzymes and cell surface receptors. For example, fucoidans with a higher proportion of (1→3) linkages tend to exhibit enhanced antioxidant and antiviral activities, likely due to their more flexible and accessible structure. Meanwhile, (1→4)-linked fucoidans are associated with stronger anti-inflammatory and anticancer properties, possibly because of their more rigid conformation, which may enhance interactions with immune cells and tumor microenvironments [83].
Regarding fucoidans from Sargassum aquifolium, they contain 2-linked α-d-Manp and 4-linked β-d-GlcAp glycosidic bonds, which have been associated with anticoagulant and antitumor activities in vitro [43]. These effects were observed in cells derived from human cancer cell lines such as the HePG2, human cervical carcinoma, and RD cell lines. The presence of these specific glycosidic linkages likely enhances the fucoidans’ interactions with key cell surface receptors involved in coagulation and tumor suppression, contributing to their bioactivity. Undaria pinnatifida contains fucoidans with β-d-Galp, α-type glycosidic linkages, which have demonstrated notable anticancer effects in vitro, particularly in studies with Sprague Dawley rats [56]. The specific glycosidic bonds, in this case, contribute to structural conformation that facilitates binding to cancer cell receptors such as selectins, thereby triggering apoptotic and anti-proliferative pathways, which are key to fucoidan’s anticancer properties [84,85,86].
Fucoidans from Padina borgeseni contain (1→4)-l-fucose, (1→6)-β-d-galactose, and β-d-mannuronic acid linkages, which contribute to their antioxidant and anticancer activities [61]. The arrangement of these glycosidic bonds plays a critical role in free radical scavenging, reducing oxidative stress in cells and enhancing the fucoidans’ therapeutic potential. Additionally, fucoidan from Stoechospermum polypodioides, with [(1→4) and (1→3)-linked α-l-fucopyranosyl] glycosidic bonds, has exhibited strong antiviral activity in vitro against Vero cells. These linkages, combined with sulfate groups, likely enhance binding affinity to viral components, effectively inhibiting viral replication at an EC50 of 3.5 ± 0.63 μg/mL [51,87,88].
Moreover, a specific structure from Sargassum horneri was identified as a branched polysaccharide, with a backbone consisting of repeating units of →3-α-L-Fucp(2SO3)-1→4-α-L-Fucp(2,3SO3)-1→ and side chains including α-L-Fucp-1→2-α-L-Fucp-1→ or α-L-Fucp-1→3-α-L-Fucp(4SO3)-1→ attached at C4 [55]. The high molecular weight and abundance of α-l-Fucp-1→3-α-l-Fucp(4SO3)-1→ side chains exhibited the most potent anticancer and radiosensitizing effects on the colony formation of DLD-1 cells.
Fucoidans from Saccharinajaponica contain a variety of glycosidic bonds, including α-(1→3)-linked fucose residues, which have been linked to both antibacterial and anti-SARS-CoV-2 activity [53]. The complexity and diversity of these bonds provide multiple interaction sites with bacterial and viral proteins, amplifying the bioactivity of the fucoidan [14,89]. Furthermore, Undaria pinnatifida fucoidan has (α1→4)-linked l-fucopyranose glycosidic bonds, which play a crucial role in attenuating SARS-CoV-2 infection [44]. The structural conformation driven by these bonds likely interferes with viral entry or replication, making these fucoidans promising antiviral candidates [90,91].
Moreover, the position of glycosidic linkages can influence the conformational entropy of oligosaccharides, which, in turn, affects their recognition, processing, and overall properties [92]. This suggests that specific linkage patterns in fucoidan may contribute to its diverse bioactivities, including its anticoagulant, antioxidant, and anti-inflammatory effects [81]. For instance, fucoidan with alternating (1→3) and (1→4) linkages has shown stronger inhibitory effects on α-D-glucosidase, which is relevant for its anti-diabetic activity [82].
Another study on fucoidan from Sargassum binderi suggested a main structure of →3)fuc-2-OSO3 (1→3)fuc-2-OSO3 (1→ [93], which indicates its safety for food product applications. Although experiments on biological activity were not conducted alongside structural elucidation, some fucoidans, such as those from the sea cucumber Pearsonothuria graeffei, exhibit a tetrasaccharide repeating unit with a backbone of [→3Fuc (2S, 4S) α1→3Fucα1→3Fuc (4S) α1→3Fuc]n [94]. In the case of fucoidans from Cladosiphon okamuranus, the backbone consists of α-1,3-linked fucopyranoside units with fucose branching at C-2 [95].
Interestingly, glycosidic linkages can vary within the same fucoidan molecule. For instance, fucoidan from Ascophyllum nodosum features a highly branched core region with predominantly α-(1→3)-linked fucosyl residues and some α-(1→4) linkages, with branch points at position 2 of the →3-linked internal residues [96]. This structural complexity contributes to the diverse biological activities of fucoidans, suggesting that variations in the structural features may be significant for their various biological properties.
The glycosidic bond connections in algae-derived fucoidan play a crucial role in determining its biological activity. Linkages such as (1→3) and (1→4)-linked α-fucopyranose or β-D-Galp-(1→4) significantly influence fucoidan’s structural conformation and its ability to interact with biological targets, thereby enhancing its bioactivity. A deeper understanding of glycosidic bonds in fucoidan can provide essential insights for developing novel therapeutic agents. Future research should focus on further elucidating the precise structures of these linkages and their mechanisms of action to maximize the therapeutic potential of algae-derived fucoidan in clinical applications [82,97].

6. Advancements in Analytical Techniques: Exploring the Diverse Structure of Fucoidan

Initially, fucoidan was believed to consist solely of sulfated fucose. This understanding persisted until the discovery of fucoidan in Macrocystis pyrifera in the early 1980s, which revealed the presence of not only fucose but also other monosaccharides such as galactose and xylose, challenging the earlier, simplified view of fucoidan’s composition [98]. Subsequent studies confirmed that fucoidan is much more structurally diverse than previously thought, with its composition varying depending on the source and species. Research demonstrated that the monosaccharide composition of fucoidan could include a range of polysaccharides and that this variability contributed to its functional diversity [99]. To analyze this complexity, researchers employed various analytical techniques, such as acetylation analysis, high-performance liquid chromatography (HPLC), and gas–liquid chromatography (GLC), which allowed for precise identification and quantification of the monosaccharides present in fucoidan, enhancing our understanding of fucoidan’s intricate structure [98,100].
The significant advances in analytical technologies over recent decades have greatly enhanced the ability to elucidate the complex structure of fucoidan. (Figure 1) In the past, fucoidan’s heterogeneous and highly branched structure made detailed structural analysis challenging. However, the development of high-resolution mass spectrometry (HR-MS) has provided researchers with the ability to accurately determine the molecular weights of fucoidan’s polysaccharide residues and map the bonding patterns between them, offering a clearer view of its structural complexity [101]. In addition, nuclear magnetic resonance (NMR) spectroscopy has significantly complemented the elucidation of fucoidan’s complex structure [58,102]. It has played a crucial role in determining the primary structure of fucoidan, including its repeating units and sulfation patterns, enabling researchers to construct a more comprehensive three-dimensional model of fucoidan. For example, enzymatic degradation, methylation analysis, and NMR spectroscopy have been used together to clarify the primary structure of fucoidan from Holothuria tubulosa [58]. Additionally, high-performance size-exclusion chromatography combined with multiple-angle laser light scattering and viscometry can provide information on chain conformation and molecular weight [58].
Chromatographic techniques such as HPLC and GC have further aided in separating and analyzing fucoidan’s components, particularly when coupled with advanced methods like mass spectrometry or methylation analysis. These techniques, along with enzymatic degradation methods, have made it possible to break down fucoidan into smaller oligosaccharides, simplifying the study of its structural motifs and facilitating a deeper understanding of its diverse monosaccharide makeup.
Hydrophilic interaction liquid chromatography (HILIC) coupled with electrospray mass spectrometry (ESI-FTMS) and high-energy collision-induced dissociation (HCD-MS/MS), along with 2D NMR spectroscopy analysis, has been successfully employed to map fucoidan oligosaccharides [103]. These techniques provide detailed information on the sequence and sulfation patterns of oligosaccharides. ESI-MS has been used to determine the chemical structure of fucoidan, while small-angle X-ray scattering (SAXS) has also been used to confirm bonding arrangements and spatial architecture in crystallized segments of fucoidan, further contributing to the understanding of its structure [104]. ESI-MS can reveal the backbone composition and branching patterns, while SAXS provides insights into the molecular-level conformation. Spectroscopic methods such as Fourier-transform infrared (FT-IR) spectroscopy and Raman spectroscopy, combined with chemometrics, have been developed for determining the purity of extracted fucoidan [102,105,106]. FT-IR spectroscopy is useful for confirming the presence of fucoidan and identifying its structural characteristics. This technique can detect changes in functional groups and is particularly helpful in analyzing the effects of various treatments on fucoidan structure.
Additionally, advances in bioinformatics have allowed for more efficient processing and interpretation of the complex data generated by these techniques, enabling researchers to visualize and comprehend the full structural diversity of fucoidan in unprecedented detail. X-ray crystallography, though traditionally difficult to apply to large polysaccharides, has also been used to confirm bonding arrangements and spatial architecture in crystallized segments of fucoidan, further contributing to the understanding of its structure.
As these technologies have advanced, it has become clear that the chemical composition of fucoidan is not only more complex but also more variable than initially assumed. Fucoidan’s structure differs significantly depending on its source, particularly among different species of brown algae. Broadly, fucoidans have been classified into two structural types: Type I, which consists of 1,3-α-L-Fucp units with sulfate groups at the O-2 and O-4 positions, and Type II, which features alternating 1,3- and 1,4-α-L-Fucp units with sulfate groups at the O-2, O-3, and O-4 positions [107]. Recent studies have uncovered additional structural variations, including different sulfate substitution patterns, molecular weights, branching configurations, and polysaccharide residues, although they generally retain a backbone like Type I and Type II fucoidans [108]. These variations in structure are significant because they lead to different biological effects, even though these compounds are all categorized under the comprehensive term “fucoidan” [109]. Thus, the advancement of analytical technologies has not only made it easier to analyze fucoidan’s structure but has also revealed its remarkable structural diversity, providing new opportunities to explore its biological and pharmacological potentials.

7. Conclusions

Fucoidan has long been studied for its diverse range of biological activities, traditionally attributed to the presence and distribution of sulfate groups. However, accumulating evidence now suggests that the monosaccharide composition, specifically the types, ratios, and distribution of monosaccharides such as fucose, galactose, and mannose, plays an equally critical, if not more significant, role in modulating its bioactivity.
The factor of a lower molecular weight enhances bioavailability by enabling easier absorption and cellular uptake, while the factor of a higher molecular weight prolongs therapeutic effects due to facilitation of slower degradation and extended circulation in the body. The sulfate content and specific binding positions of sulfate groups are critical in modulating fucoidan’s bioactivity, as they influence its ability to interact with proteins and cell receptors. The factor of a higher sulfate content increases the negative charge of fucoidan, enhancing electrostatic interactions with positively charged biological molecules, which is crucial for its anticoagulant, antiviral, and anti-inflammatory effects. Similarly, the composition of polysaccharides, including the types and ratios of monosaccharides, affects the structural conformation of fucoidan, impacting its binding affinity to receptors and its ability to trigger specific biological responses. These combined factors, including polysaccharide composition, sulfate content, the binding positions of sulfate groups, and molecular weight, work synergistically to determine fucoidan’s therapeutic potential by influencing its stability, receptor binding, and overall bioactivity. Additionally, external factors such as extraction methods can lead to variations in fucoidan’s chemical characteristics, even within the same species of algae. This variability may result in differences in biological activity, further emphasizing the need for more systematic research. This highlights the need for a comprehensive understanding of fucoidan’s characteristic–function relationship, rather than focusing solely on sulfate groups, to explain the reasons behind fucoidan’s bioactivity.
Future research should prioritize the in-depth structural characterization of fucoidan by focusing on optimizing methods for extraction, purification, and analysis to better control polysaccharide composition, molecular weight distribution, and sulfation binding patterns. Additionally, investigating how molecular weight and structural configuration influence cleavage, absorption, receptor binding, the production of secondary metabolites, and subsequent biological interactions is crucial. By advancing the understanding of these factors, including receptor interactions and bioavailability, fucoidan’s full therapeutic potential can be realized. These insights are essential for developing precise fucoidan-based therapies, leading to the comprehensive application of fucoidan.
There are not many sulfated biopolymers like fucoidan, but some sulfated polysaccharides have been found in marine organisms. With the recent advancements in the structural analysis of marine-derived polysaccharides, research on the diverse properties of these sulfated polysaccharides is expected to progress rapidly. While studies on the correlation between the characteristics and biological activities of sulfated polysaccharides other than fucoidan are still in the early stages, it is anticipated that more comprehensive data on the relationships between the monosaccharide composition, abundance, and biological activity of marine-derived sulfated polysaccharides will be published and utilized in the future. This will likely lead to a clearer understanding of the biological mechanisms and structure–function relationships of not only fucoidan but also other sulfated polysaccharides.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms252111771/s1.

Author Contributions

Conceptualization: S.J. and B.R.; resources: S.L. and G.L.; data curation: S.J. and J.H.; writing—original draft preparation: S.J. and B.R.; writing—review and editing: S.J., J.H., and B.R.; visualization: S.J. and S.L.; supervision: B.R. and J.H.; project administration: B.R.; funding acquisition: B.R. All authors have read and agreed to the published version of the manuscript.

Funding

This work received funding from the Emerging Researcher Challenge Support Program at Pukyong National University (RS-2023-01410001). The authors extend their appreciation to the Ministry of Oceans and Fisheries, Korea (RS-2023-00255684), and the Korea Institute of Marine Science & Technology Promotion (KIMST), Ministry of Oceans and Fisheries (20220488). This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (RS-2023-00208025).

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

The authors would like to express their gratitude to Zhong-ji Qian from the School of Chemistry and Environment, Guangdong Ocean University, Zhanjiang 524088, China, for his invaluable contribution to the mechanism-related aspects of this research.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Karagoz, P.; Khiawjan, S.; Marques, M.P.; Santzouk, S.; Bugg, T.D.; Lye, G.J. Pharmaceutical applications of lignin-derived chemicals and lignin-based materials: Linking lignin source and processing with clinical indication. Biomass Convers. Biorefinery 2023, 14, 26553–26574. [Google Scholar] [CrossRef]
  2. Pei, W.; Deng, J.; Wang, P.; Wang, X.; Zheng, L.; Zhang, Y.; Huang, C. Sustainable lignin and lignin-derived compounds as potential therapeutic agents for degenerative orthopaedic diseases: A systemic review. Int. J. Biol. Macromol. 2022, 212, 547–560. [Google Scholar] [CrossRef] [PubMed]
  3. Oliyaei, N.; Moosavi-Nasab, M.; Mazloomi, S.M. Therapeutic activity of fucoidan and carrageenan as marine algal polysaccharides against viruses. 3 Biotech 2022, 12, 154. [Google Scholar] [CrossRef] [PubMed]
  4. Gopinath, V.; Saravanan, S.; Al-Maleki, A.; Ramesh, M.; Vadivelu, J. A review of natural polysaccharides for drug delivery applications: Special focus on cellulose, starch and glycogen. Biomed. Pharmacother. 2018, 107, 96–108. [Google Scholar] [CrossRef] [PubMed]
  5. Lee, Y.-E.; Kim, H.; Seo, C.; Park, T.; Lee, K.B.; Yoo, S.-Y.; Hong, S.-C.; Kim, J.T.; Lee, J. Marine polysaccharides: Therapeutic efficacy and biomedical applications. Arch. Pharmacal Res. 2017, 40, 1006–1020. [Google Scholar] [CrossRef]
  6. Berteau, O.; Mulloy, B. Sulfated fucans, fresh perspectives: Structures, functions, and biological properties of sulfated fucans and an overview of enzymes active toward this class of polysaccharide. Glycobiology 2003, 13, 29R–40R. [Google Scholar] [CrossRef]
  7. Cumashi, A.; Ushakova, N.A.; Preobrazhenskaya, M.E.; D’Incecco, A.; Piccoli, A.; Totani, L.; Tinari, N.; Morozevich, G.E.; Berman, A.E.; Bilan, M.I. A comparative study of the anti-inflammatory, anticoagulant, antiangiogenic, and antiadhesive activities of nine different fucoidans from brown seaweeds. Glycobiology 2007, 17, 541–552. [Google Scholar] [CrossRef]
  8. Bilan, M.I.; Usov, A.I. Structural analysis of fucoidans. Nat. Prod. Commun. 2008, 3, 1934578X0800301011. [Google Scholar] [CrossRef]
  9. Anisha, G.S.; Padmakumari, S.; Patel, A.K.; Pandey, A.; Singhania, R.R. Fucoidan from marine macroalgae: Biological actions and applications in regenerative medicine, drug delivery systems and food industry. Bioengineering 2022, 9, 472. [Google Scholar] [CrossRef]
  10. Venkatesan, J.; Murugan, S.S.; Seong, G.H. Fucoidan-based nanoparticles: Preparations and applications. Int. J. Biol. Macromol. 2022, 217, 652–667. [Google Scholar] [CrossRef]
  11. Wei, Q.; Fu, G.; Wang, K.; Yang, Q.; Zhao, J.; Wang, Y.; Ji, K.; Song, S. Advances in research on antiviral activities of sulfated polysaccharides from seaweeds. Pharmaceuticals 2022, 15, 581. [Google Scholar] [CrossRef] [PubMed]
  12. 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]
  13. Ale, M.T.; Meyer, A.S. Fucoidans from brown seaweeds: An update on structures, extraction techniques and use of enzymes as tools for structural elucidation. Rsc Adv. 2013, 3, 8131–8141. [Google Scholar] [CrossRef]
  14. Wang, Y.; Xing, M.; Cao, Q.; Ji, A.; Liang, H.; Song, S. Biological activities of fucoidan and the factors mediating its therapeutic effects: A review of recent studies. Mar. Drugs 2019, 17, 183. [Google Scholar] [CrossRef] [PubMed]
  15. Wang, S.-H.; Huang, C.-Y.; Chen, C.-Y.; Chang, C.-C.; Huang, C.-Y.; Dong, C.-D.; Chang, J.-S. Structure and biological activity analysis of fucoidan isolated from Sargassum siliquosum. ACS Omega 2020, 5, 32447–32455. [Google Scholar] [CrossRef] [PubMed]
  16. Lu, W.; Yang, Z.; Chen, J.; Wang, D.; Zhang, Y. Recent advances in antiviral activities and potential mechanisms of sulfated polysaccharides. Carbohydr. Polym. 2021, 272, 118526. [Google Scholar] [CrossRef]
  17. Yang, Y.; Liu, D.; Wu, J.; Chen, Y.; Wang, S. In vitro antioxidant activities of sulfated polysaccharide fractions extracted from Corallina officinalis. Int. J. Biol. Macromol. 2011, 49, 1031–1037. [Google Scholar] [CrossRef]
  18. Pradhan, B.; Patra, S.; Nayak, R.; Behera, C.; Dash, S.R.; Nayak, S.; Sahu, B.B.; Bhutia, S.K.; Jena, M. Multifunctional role of fucoidan, sulfated polysaccharides in human health and disease: A journey under the sea in pursuit of potent therapeutic agents. Int. J. Biol. Macromol. 2020, 164, 4263–4278. [Google Scholar] [CrossRef]
  19. Ale, M.T.; Maruyama, H.; Tamauchi, H.; Mikkelsen, J.D.; Meyer, A.S. Fucose-containing sulfated polysaccharides from brown seaweeds inhibit proliferation of melanoma cells and induce apoptosis by activation of caspase-3 in vitro. Mar. Drugs 2011, 9, 2605–2621. [Google Scholar] [CrossRef]
  20. Costa, L.S.; Fidelis, G.P.; Cordeiro, S.L.; Oliveira, R.M.; Sabry, D.A.; Câmara, R.B.G.; Nobre, L.T.D.B.; Costa, M.S.S.P.; Almeida-Lima, J.; Farias, E. Biological activities of sulfated polysaccharides from tropical seaweeds. Biomed. Pharmacother. 2010, 64, 21–28. [Google Scholar] [CrossRef]
  21. Vo, T.-S.; Kim, S.-K. Fucoidans as a natural bioactive ingredient for functional foods. J. Funct. Foods 2013, 5, 16–27. [Google Scholar] [CrossRef]
  22. Sanjeewa, K.A.; Fernando, I.; 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. Int. J. Biol. Macromol. 2018, 107, 803–807. [Google Scholar] [CrossRef] [PubMed]
  23. Kang, M.-C.; Lee, H.; Choi, H.-D.; Jeon, Y.-J. Antioxidant properties of a sulfated polysaccharide isolated from an enzymatic digest of Sargassum thunbergii. Int. J. Biol. Macromol. 2019, 132, 142–149. [Google Scholar] [CrossRef] [PubMed]
  24. Achmad, H.; Huldani, H.; Feby Ramadhany, Y. Antimicrobial activity and sulfated polysaccharides antibiofilms in marine algae against dental plaque bacteria: A literature review. Syst. Rev. Pharm. 2020, 11, 459–465. [Google Scholar]
  25. Hans, N.; Malik, A.; Naik, S. Antiviral activity of sulfated polysaccharides from marine algae and its application in combating COVID-19: Mini review. Bioresour. Technol. Rep. 2021, 13, 100623. [Google Scholar] [CrossRef]
  26. Ushakova, N.; Morozevich, G.; Ustyuzhanina, N.; Bilan, M.; Usov, A.; Nifantiev, N.; Preobrazhenskaya, M. Anticoagulant activity of fucoidans from brown algae. Biochem. (Mosc.) Suppl. Ser. B Biomed. Chem. 2009, 3, 77–83. [Google Scholar] [CrossRef]
  27. Yoo, H.J.; You, D.-J.; Lee, K.-W. Characterization and immunomodulatory effects of high molecular weight fucoidan fraction from the sporophyll of Undaria pinnatifida in cyclophosphamide-induced immunosuppressed mice. Mar. Drugs 2019, 17, 447. [Google Scholar] [CrossRef]
  28. 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]
  29. Ale, M.T.; Mikkelsen, J.D.; Meyer, A.S. Designed optimization of a single-step extraction of fucose-containing sulfated polysaccharides from Sargassum sp. J. Appl. Phycol. 2012, 24, 715–723. [Google Scholar] [CrossRef]
  30. Wijesinghe, W.; Jeon, Y.-J. Biological activities and potential industrial applications of fucose rich sulfated polysaccharides and fucoidans isolated from brown seaweeds: A review. Carbohydr. Polym. 2012, 88, 13–20. [Google Scholar] [CrossRef]
  31. Mensah, E.O.; Kanwugu, O.N.; Panda, P.K.; Adadi, P. Marine fucoidans: Structural, extraction, biological activities and their applications in the food industry. Food Hydrocoll. 2023, 142, 108784. [Google Scholar] [CrossRef]
  32. Huang, L.; Shen, M.; Morris, G.A.; Xie, J. Sulfated polysaccharides: Immunomodulation and signaling mechanisms. Trends Food Sci. Technol. 2019, 92, 1–11. [Google Scholar] [CrossRef]
  33. Apostolova, E.; Lukova, P.; Baldzhieva, A.; Katsarov, P.; Nikolova, M.; Iliev, I.; Peychev, L.; Trica, B.; Oancea, F.; Delattre, C. Immunomodulatory and anti-inflammatory effects of fucoidan: A review. Polymers 2020, 12, 2338. [Google Scholar] [CrossRef] [PubMed]
  34. 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]
  35. Kusaykin, M.; Bakunina, I.; Sova, V.; Ermakova, S.; Kuznetsova, T.; Besednova, N.; Zaporozhets, T.; Zvyagintseva, T. Structure, biological activity, and enzymatic transformation of fucoidans from the brown seaweeds. Biotechnol. J. Healthc. Nutr. Technol. 2008, 3, 904–915. [Google Scholar] [CrossRef]
  36. Li, B.; Lu, F.; Wei, X.; Zhao, R. Fucoidan: Structure and bioactivity. Molecules 2008, 13, 1671–1695. [Google Scholar] [CrossRef]
  37. Karnjanapratum, S.; You, S. Molecular characteristics of sulfated polysaccharides from Monostroma nitidum and their in vitro anticancer and immunomodulatory activities. Int. J. Biol. Macromol. 2011, 48, 311–318. [Google Scholar] [CrossRef]
  38. Shao, P.; Chen, M.; Pei, Y.; Sun, P. In intro antioxidant activities of different sulfated polysaccharides from chlorophytan seaweeds Ulva fasciata. Int. J. Biol. Macromol. 2013, 59, 295–300. [Google Scholar] [CrossRef]
  39. Abdel-Tawwab, M.; Harikrishnan, R.; Devi, G.; Bhat, E.A.; Paray, B.A. Stimulatory effects of seaweed Laminaria digitata polysaccharides additives on growth, immune-antioxidant potency and related genes induction in Rohu carp (Labeo rohita) during Flavobacterium columnare infection. Aquaculture 2024, 579, 740253. [Google Scholar] [CrossRef]
  40. El Rashed, Z.; Lupidi, G.; Kanaan, H.; Grasselli, E.; Canesi, L.; Khalifeh, H.; Demori, I. Antioxidant and Antisteatotic activities of a new Fucoidan extracted from Ferula hermonis roots harvested on Lebanese mountains. Molecules 2021, 26, 1161. [Google Scholar] [CrossRef]
  41. Ptak, S.H.; Errico, M.; Christensen, K.V. Extracting activity patterns: Exploratory data analysis on a fucoidan extract data set with mixed variables. Algal Res. 2021, 54, 102220. [Google Scholar] [CrossRef]
  42. Dobrinčić, A.; Dobroslavić, E.; Pedisić, S.; Balbino, S.; Garofulić, I.E.; Čož-Rakovac, R.; Dragović-Uzelac, V. The effectiveness of the Fucus virsoides and Cystoseira barbata fucoidan isolation as a function of applied pre-treatment and extraction conditions. Algal Res. 2021, 56, 102286. [Google Scholar] [CrossRef]
  43. Bilan, M.I.; Ustyuzhanina, N.E.; Shashkov, A.S.; Thanh, T.T.T.; Bui, M.L.; Van Tran, T.T.; Usov, A.I. Sulfated polysaccharides of the Vietnamese brown alga Sargassum aquifolium (Fucales, Sargassaceae). Carbohydr. Res. 2017, 449, 23–31. [Google Scholar] [CrossRef] [PubMed]
  44. Shi, F.-S.; Xie, Y.-H.; Yang, Y.-L.; Xu, L.-D.; Li, J.-J.; Wang, X.; Zhu, L.-Y.; Wang, W.-W.; Shen, P.-L.; Huang, Y.-W. Fucoidan from Ascophyllum nodosum and Undaria pinnatifida attenuate SARS-CoV-2 infection in vitro and in vivo by suppressing ACE2 and alleviating inflammation. Carbohydr. Polym. 2024, 332, 121884. [Google Scholar] [CrossRef] [PubMed]
  45. Silva, M.M.C.L.; dos Santos Lisboa, L.; Paiva, W.S.; Batista, L.A.N.C.; Luchiari, A.C.; Rocha, H.A.O.; Camara, R.B.G. Comparison of in vitro and in vivo antioxidant activities of commercial fucoidans from Macrocystis pyrifera, Undaria pinnatifida, and Fucus vesiculosus. Int. J. Biol. Macromol. 2022, 216, 757–767. [Google Scholar] [CrossRef] [PubMed]
  46. Wang, L.; Oliveira, C.; Li, Q.; Ferreira, A.S.; Nunes, C.; Coimbra, M.A.; Reis, R.L.; Martins, A.; Wang, C.; Silva, T.H. Fucoidan from Fucus vesiculosus inhibits inflammatory response, both in vitro and in vivo. Mar. Drugs 2023, 21, 302. [Google Scholar] [CrossRef]
  47. Zhuang, S.-J.; Xu, H.-K.; Hu, X.; Wu, T.-C.; Li, J.-N.; Lee, H.-G.; Yu, P.; Dai, Y.-L.; Jeon, Y.-J. In vitro and in vivo anti-inflammatory properties of an active fucoidan fraction from Sargassum fusiforme and a fraction-based hydrogel. Int. J. Biol. Macromol. 2024, 265, 130866. [Google Scholar] [CrossRef]
  48. Wang, F.; Xiao, Y.; Neupane, S.; Ptak, S.H.; Römer, R.; Xiong, J.; Ohmes, J.; Seekamp, A.; Fretté, X.; Alban, S. Influence of fucoidan extracts from different fucus species on adult stem cells and molecular mediators in in vitro models for bone formation and vascularization. Mar. Drugs 2021, 19, 194. [Google Scholar] [CrossRef]
  49. Michalak, L.; Morales-Lange, B.; Montero, R.; Horn, S.J.; Mydland, L.T.; Øverland, M. Impact of biorefinery processing conditions on the bioactive properties of fucoidan extracts from Saccharina latissima on SHK-1 cells. Algal Res. 2023, 75, 103221. [Google Scholar] [CrossRef]
  50. Moreira, A.S.; Gaspar, D.; Ferreira, S.S.; Correia, A.; Vilanova, M.; Perrineau, M.-M.; Kerrison, P.D.; Gachon, C.M.; Domingues, M.R.; Coimbra, M.A. Water-soluble Saccharina latissima polysaccharides and relation of their structural characteristics with in vitro immunostimulatory and hypocholesterolemic activities. Mar. Drugs 2023, 21, 183. [Google Scholar] [CrossRef]
  51. Adhikari, U.; Mateu, C.G.; Chattopadhyay, K.; Pujol, C.A.; Damonte, E.B.; Ray, B. Structure and antiviral activity of sulfated fucans from Stoechospermum marginatum. Phytochemistry 2006, 67, 2474–2482. [Google Scholar] [CrossRef] [PubMed]
  52. Chen, B.-R.; Hsu, K.-T.; Li, T.-L.; Chan, Y.-L.; Wu, C.-J. Topical application of fucoidan derived from Cladosiphon okamuranus alleviates atopic dermatitis symptoms through immunomodulation. Int. Immunopharmacol. 2021, 101, 108362. [Google Scholar] [CrossRef]
  53. Sun, X.; Ai, C.; Wen, C.; Peng, H.; Yang, J.; Cui, Y.; Song, S. Inhibitory effects of fucoidan from Laminaria japonica against some pathogenic bacteria and SARS-CoV-2 depend on its large molecular weight. Int. J. Biol. Macromol. 2023, 229, 413–421. [Google Scholar] [CrossRef]
  54. Suresh, V.; Senthilkumar, N.; Thangam, R.; Rajkumar, M.; Anbazhagan, C.; Rengasamy, R.; Gunasekaran, P.; Kannan, S.; Palani, P. Separation, purification and preliminary characterization of sulfated polysaccharides from Sargassum plagiophyllum and its in vitro anticancer and antioxidant activity. Process Biochem. 2013, 48, 364–373. [Google Scholar] [CrossRef]
  55. Rasin, A.B.; Silchenko, A.S.; Kusaykin, M.I.; Malyarenko, O.S.; Zueva, A.O.; Kalinovsky, A.I.; Airong, J.; Surits, V.V.; Ermakova, S.P. Enzymatic transformation and anti-tumor activity of Sargassum horneri fucoidan. Carbohydr. Polym. 2020, 246, 116635. [Google Scholar] [CrossRef]
  56. Han, Y.; Wu, J.; Liu, T.; Hu, Y.; Zheng, Q.; Wang, B.; Lin, H.; Li, X. Separation, characterization and anticancer activities of a sulfated polysaccharide from Undaria pinnatifida. Int. J. Biol. Macromol. 2016, 83, 42–49. [Google Scholar] [CrossRef]
  57. Cui, Y.; Liu, X.; Li, S.; Hao, L.; Du, J.; Gao, D.; Kang, Q.; Lu, J. Extraction, characterization and biological activity of sulfated polysaccharides from seaweed Dictyopteris divaricata. Int. J. Biol. Macromol. 2018, 117, 256–263. [Google Scholar] [CrossRef]
  58. Chang, Y.; Hu, Y.; Yu, L.; McClements, D.J.; Xu, X.; Liu, G.; Xue, C. Primary structure and chain conformation of fucoidan extracted from sea cucumber Holothuria tubulosa. Carbohydr. Polym. 2016, 136, 1091–1097. [Google Scholar] [CrossRef]
  59. Palanisamy, S.; Vinosha, M.; Marudhupandi, T.; Rajasekar, P.; Prabhu, N.M. Isolation of fucoidan from Sargassum polycystum brown algae: Structural characterization, in vitro antioxidant and anticancer activity. Int. J. Biol. Macromol. 2017, 102, 405–412. [Google Scholar] [CrossRef]
  60. Anastyuk, S.D.; Shevchenko, N.M.; Ermakova, S.P.; Vishchuk, O.S.; Nazarenko, E.L.; Dmitrenok, P.S.; Zvyagintseva, T.N. Anticancer activity in vitro of a fucoidan from the brown alga Fucus evanescens and its low-molecular fragments, structurally characterized by tandem mass-spectrometry. Carbohydr. Polym. 2012, 87, 186–194. [Google Scholar] [CrossRef]
  61. Cholaraj, R.; Venkatachalam, R. Investigation of antioxidant and anticancer potential of fucoidan (in-vitro & in-silico) from brown seaweed Padina boergesenii. Algal Res. 2024, 79, 103442. [Google Scholar]
  62. Qin, L.; Cao, J.; Xu, H.; Li, N.; Wang, K.; Zhang, L.; Qu, C.; Miao, J. Structural characterization of a sulfated polysaccharide from Ishige okamurae and its effect on recovery from immunosuppression. Int. J. Biol. Macromol. 2023, 236, 123948. [Google Scholar] [CrossRef]
  63. Veena, C.K.; Josephine, A.; Preetha, S.P.; Varalakshmi, P. Beneficial role of sulfated polysaccharides from edible seaweed Fucus vesiculosus in experimental hyperoxaluria. Food Chem. 2007, 100, 1552–1559. [Google Scholar] [CrossRef]
  64. Wang, J.; Guo, H.; Zhang, J.; Wang, X.; Zhao, B.; Yao, J.; Wang, Y. Sulfated modification, characterization and structure–antioxidant relationships of Artemisia sphaerocephala polysaccharides. Carbohydr. Polym. 2010, 81, 897–905. [Google Scholar] [CrossRef]
  65. 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]
  66. Schneider, T.; Ehrig, K.; Liewert, I.; Alban, S. Interference with the CXCL12/CXCR4 axis as potential antitumor strategy: Superiority of a sulfated galactofucan from the brown alga Saccharina latissima and fucoidan over heparins. Glycobiology 2015, 25, 812–824. [Google Scholar] [CrossRef]
  67. Alencar, P.O.C.; Lima, G.C.; Barros, F.C.N.; Costa, L.E.; Ribeiro, C.V.P.; Sousa, W.M.; Sombra, V.G.; Abreu, C.M.W.; Abreu, E.S.; Pontes, E.O. A novel antioxidant sulfated polysaccharide from the algae Gracilaria caudata: In vitro and in vivo activities. Food Hydrocoll. 2019, 90, 28–34. [Google Scholar] [CrossRef]
  68. Jayawardena, T.U.; Sanjeewa, K.A.; Nagahawatta, D.; Lee, H.-G.; Lu, Y.-A.; Vaas, A.; Abeytunga, D.; Nanayakkara, C.; Lee, D.-S.; Jeon, Y.-J. Anti-inflammatory effects of sulfated polysaccharide from Sargassum swartzii in macrophages via blocking TLR/NF-Κb signal transduction. Mar. Drugs 2020, 18, 601. [Google Scholar] [CrossRef]
  69. Lim, S.J.; Aida, W.M.W. Extraction of sulfated polysaccharides (fucoidan) from brown seaweed. In Seaweed Polysaccharides; Elsevier: Amsterdam, The Netherlands, 2017; pp. 27–46. [Google Scholar]
  70. Zhang, Z.; Till, S.; Jiang, C.; Knappe, S.; Reutterer, S.; Scheiflinger, F.; Szabo, C.M.; Dockal, M. Structure-activity relationship of the pro-and anticoagulant effects of Fucus vesiculosus fucoidan. Thromb. Haemost. 2014, 112, 429–437. [Google Scholar] [CrossRef]
  71. Zayed, A.; El-Aasr, M.; Ibrahim, A.-R.S.; Ulber, R. Fucoidan characterization: Determination of purity and physicochemical and chemical properties. Mar. Drugs 2020, 18, 571. [Google Scholar] [CrossRef] [PubMed]
  72. Silchenko, A.; Rasin, A.; Zueva, A.; Kusaykin, M.; Zvyagintseva, T.; Rubtsov, N.; Ermakova, S. Discovery of a fucoidan endo-4O-sulfatase: Regioselective 4O-desulfation of fucoidans and its effect on anticancer activity in vitro. Carbohydr. Polym. 2021, 271, 118449. [Google Scholar] [CrossRef]
  73. Chen, C.-Y.; Wang, S.-H.; Huang, C.-Y.; Dong, C.-D.; Huang, C.-Y.; Chang, C.-C.; Chang, J.-S. Effect of molecular mass and sulfate content of fucoidan from Sargassum siliquosum on antioxidant, anti-lipogenesis, and anti-inflammatory activity. J. Biosci. Bioeng. 2021, 132, 359–364. [Google Scholar] [CrossRef]
  74. Yuguchi, Y.; Bui, L.M.; Takebe, S.; Suzuki, S.; Nakajima, N.; Kitamura, S.; Thanh, T.T.T. Primary structure, conformation in aqueous solution, and intestinal immunomodulating activity of fucoidan from two brown seaweed species Sargassum crassifolium and Padina australis. Carbohydr. Polym. 2016, 147, 69–78. [Google Scholar] [CrossRef]
  75. Yang, C.; Chung, D.; Shin, I.-S.; Lee, H.; Kim, J.; Lee, Y.; You, S. Effects of molecular weight and hydrolysis conditions on anticancer activity of fucoidans from sporophyll of Undaria pinnatifida. Int. J. Biol. Macromol. 2008, 43, 433–437. [Google Scholar] [CrossRef]
  76. Zhao, X.; Guo, F.; Hu, J.; Zhang, L.; Xue, C.; Zhang, Z.; Li, B. Antithrombotic activity of oral administered low molecular weight fucoidan from Laminaria japonica. Thromb. Res. 2016, 144, 46–52. [Google Scholar] [CrossRef]
  77. Sun, T.; Zhang, X.; Miao, Y.; Zhou, Y.; Shi, J.; Yan, M.; Chen, A. Studies on antiviral and immuno-regulation activity of low molecular weight fucoidan from Laminaria japonica. J. Ocean Univ. China 2018, 17, 705–711. [Google Scholar] [CrossRef]
  78. Suprunchuk, V.E. Low-molecular-weight fucoidan: Chemical modification, synthesis of its oligomeric fragments and mimetics. Carbohydr. Res. 2019, 485, 107806. [Google Scholar] [CrossRef]
  79. Morya, V.; Kim, J.; Kim, E.-K. Algal fucoidan: Structural and size-dependent bioactivities and their perspectives. Appl. Microbiol. Biotechnol. 2012, 93, 71–82. [Google Scholar] [CrossRef]
  80. Yuan, Y.; Macquarrie, D. Microwave assisted extraction of sulfated polysaccharides (fucoidan) from Ascophyllum nodosum and its antioxidant activity. Carbohydr. Polym. 2015, 129, 101–107. [Google Scholar] [CrossRef]
  81. Kopplin, G.; Rokstad, A.M.; Mélida, H.; Bulone, V.; Skjåk-Bræk, G.; Aachmann, F.L. Structural characterization of fucoidan from Laminaria hyperborea: Assessment of coagulation and inflammatory properties and their structure–function relationship. ACS Appl. Bio Mater. 2018, 1, 1880–1892. [Google Scholar] [CrossRef]
  82. Wen, Y.; Gao, L.; Zhou, H.; Ai, C.; Huang, X.; Wang, M.; Zhang, Y.; Zhao, C. Opportunities and challenges of algal fucoidan for diabetes management. Trends Food Sci. Technol. 2021, 111, 628–641. [Google Scholar] [CrossRef]
  83. Wang, M.; Veeraperumal, S.; Zhong, S.; Cheong, K.-L. Fucoidan-derived functional oligosaccharides: Recent developments, preparation, and potential applications. Foods 2023, 12, 878. [Google Scholar] [CrossRef] [PubMed]
  84. Lin, Y.; Qi, X.; Liu, H.; Xue, K.; Xu, S.; Tian, Z. The anti-cancer effects of fucoidan: A review of both in vivo and in vitro investigations. Cancer Cell Int. 2020, 20, 154. [Google Scholar] [CrossRef]
  85. Atashrazm, F.; Lowenthal, R.M.; Woods, G.M.; Holloway, A.F.; Dickinson, J.L. Fucoidan and cancer: A multifunctional molecule with anti-tumor potential. Mar. Drugs 2015, 13, 2327–2346. [Google Scholar] [CrossRef] [PubMed]
  86. Jin, J.-O.; Yadav, D.; Madhwani, K.; Puranik, N.; Chavda, V.; Song, M. Seaweeds in the oncology arena: Anti-cancer potential of fucoidan as a drug—A review. Molecules 2022, 27, 6032. [Google Scholar] [CrossRef]
  87. Lee, J.-B.; Hayashi, K.; Hashimoto, M.; Nakano, T.; Hayashi, T. Novel antiviral fucoidan from sporophyll of Undaria pinnatifida (Mekabu). Chem. Pharm. Bull. 2004, 52, 1091–1094. [Google Scholar] [CrossRef]
  88. Yang, C.-W.; Hsu, H.-Y.; Lee, Y.-Z.; Jan, J.-T.; Chang, S.-Y.; Lin, Y.-L.; Yang, R.-B.; Chao, T.-L.; Liang, J.-J.; Lin, S.-J. Natural fucoidans inhibit coronaviruses by targeting viral spike protein and host cell furin. Biochem. Pharmacol. 2023, 215, 115688. [Google Scholar] [CrossRef]
  89. Chaisuwan, W.; Phimolsiripol, Y.; Chaiyaso, T.; Techapun, C.; Leksawasdi, N.; Jantanasakulwong, K.; Rachtanapun, P.; Wangtueai, S.; Sommano, S.R.; You, S. The antiviral activity of bacterial, fungal, and algal polysaccharides as bioactive ingredients: Potential uses for enhancing immune systems and preventing viruses. Front. Nutr. 2021, 8, 772033. [Google Scholar] [CrossRef]
  90. Pradhan, B.; Nayak, R.; Patra, S.; Bhuyan, P.P.; Behera, P.K.; Mandal, A.K.; Behera, C.; Ki, J.-S.; Adhikary, S.P.; MubarakAli, D. A state-of-the-art review on fucoidan as an antiviral agent to combat viral infections. Carbohydr. Polym. 2022, 291, 119551. [Google Scholar] [CrossRef]
  91. Krylova, N.V.; Ermakova, S.P.; Lavrov, V.F.; Leneva, I.A.; Kompanets, G.G.; Iunikhina, O.V.; Nosik, M.N.; Ebralidze, L.K.; Falynskova, I.N.; Silchenko, A.S. The comparative analysis of antiviral activity of native and modified fucoidans from brown algae Fucus evanescens in vitro and in vivo. Mar. Drugs 2020, 18, 224. [Google Scholar] [CrossRef]
  92. Striegel, A.M.; Boone, M.A. Influence of glycosidic linkage on solution conformational entropy of oligosaccharides: Malto-vs. isomalto-and cello-vs. laminarioligosaccharides. Biopolymers 2011, 95, 228–233. [Google Scholar] [CrossRef] [PubMed]
  93. Lim, S.J.; Mustapha, W.A.W.; Maskat, M.Y.; Latip, J.; Badri, K.H.; Hassan, O. Chemical properties and toxicology studies of fucoidan extracted from Malaysian Sargassum binderi. Food Sci. Biotechnol. 2016, 25, 23–29. [Google Scholar] [CrossRef] [PubMed]
  94. Hu, Y.; Li, S.; Li, J.; Ye, X.; Ding, T.; Liu, D.; Chen, J.; Ge, Z.; Chen, S. Identification of a highly sulfated fucoidan from sea cucumber Pearsonothuria graeffei with well-repeated tetrasaccharides units. Carbohydr. Polym. 2015, 134, 808–816. [Google Scholar] [CrossRef] [PubMed]
  95. Lim, S.J.; Aida, W.M.W.; Schiehser, S.; Rosenau, T.; Böhmdorfer, S. Structural elucidation of fucoidan from Cladosiphon okamuranus (Okinawa mozuku). Food Chem. 2019, 272, 222–226. [Google Scholar] [CrossRef] [PubMed]
  96. Marais, M.-F.; Joseleau, J.-P. A fucoidan fraction from Ascophyllum nodosum. Carbohydr. Res. 2001, 336, 155–159. [Google Scholar] [CrossRef]
  97. Crawford, C.; Schultz-Johansen, M.; Luong, P.; Vidal-Melgosa, S.; Hehemann, J.-H.; Seeberger, P. Automated synthesis of fucoidan enables molecular investigations in marine glycobiology. ChemRxiv 2024. [Google Scholar] [CrossRef]
  98. 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]
  99. Fernando, I.S.; Kim, D.; Nah, J.-W.; Jeon, Y.-J. Advances in functionalizing fucoidans and alginates (bio) polymers by structural modifications: A review. Chem. Eng. J. 2019, 355, 33–48. [Google Scholar] [CrossRef]
  100. Karmakar, P.; Pujol, C.A.; Damonte, E.B.; Ghosh, T.; Ray, B. Polysaccharides from Padina tetrastromatica: Structural features, chemical modification and antiviral activity. Carbohydr. Polym. 2010, 80, 513–520. [Google Scholar] [CrossRef]
  101. Deepak, P.; Perumal, P.; Balakrishnan, R.; Balasubramanian, B.; Velmurugan, P. Assessment of antioxidant and digestive enzyme inhibition by phyco-molecules isolated from marine brown alga Sargassum wightii. Food Humanit. 2024, 2, 100226. [Google Scholar] [CrossRef]
  102. Ganapathy, S.; Lingappa, S.; Naidu, K.; Selvaraj, U.; Ramachandiran, S.; Ponnusamy, S.; Somasundaram, S.T. Isolation and bioactive potential of fucoidan from marine macroalgae Turbinaria conoides. ChemistrySelect 2019, 4, 14114–14119. [Google Scholar] [CrossRef]
  103. An, Z.; Zhang, Z.; Zhang, X.; Yang, H.; Lu, H.; Liu, M.; Shao, Y.; Zhao, X.; Zhang, H. Oligosaccharide mapping analysis by HILIC-ESI-HCD-MS/MS for structural elucidation of fucoidan from sea cucumber Holothuria floridana. Carbohydr. Polym. 2022, 275, 118694. [Google Scholar] [CrossRef] [PubMed]
  104. Thanh, T.T.T.; Tran, V.T.T.; Yuguchi, Y.; Bui, L.M.; Nguyen, T.T. Structure of fucoidan from brown seaweed Turbinaria ornata as studied by electrospray ionization mass spectrometry (ESIMS) and small angle X-ray scattering (SAXS) techniques. Mar. Drugs 2013, 11, 2431–2443. [Google Scholar] [CrossRef] [PubMed]
  105. Choi, J.-I.; Lee, S.G.; Han, S.J.; Cho, M.; Lee, P.C. Effect of gamma irradiation on the structure of fucoidan. Radiat. Phys. Chem. 2014, 100, 54–58. [Google Scholar] [CrossRef]
  106. Pielesz, A.; Biniaś, W. Cellulose acetate membrane electrophoresis and FTIR spectroscopy as methods of identifying a fucoidan in Fucus vesiculosus Linnaeus. Carbohydr. Res. 2010, 345, 2676–2682. [Google Scholar] [CrossRef]
  107. Shang, Q. Revisit the effects of fucoidan on gut microbiota in health and disease: What do we know and what do we need to know? Bioact. Carbohydr. Diet. Fibre 2020, 23, 100221. [Google Scholar] [CrossRef]
  108. Etman, S.M.; Elnaggar, Y.S.; Abdallah, O.Y. Fucoidan, a natural biopolymer in cancer combating: From edible algae to nanocarrier tailoring. Int. J. Biol. Macromol. 2020, 147, 799–808. [Google Scholar] [CrossRef]
  109. Oliveira, C.; Neves, N.M.; Reis, R.L.; Martins, A.; Silva, T.H. A review on fucoidan antitumor strategies: From a biological active agent to a structural component of fucoidan-based systems. Carbohydr. Polym. 2020, 239, 116131. [Google Scholar] [CrossRef]
Figure 1. Analytical techniques used to explore the complex structure of the sulfated polysaccharide fucoidan, including the phenol–sulfuric acid method/thin-layer chromatography (TLC), Fourier transform infrared (FT-IR) spectroscopy, high-performance liquid chromatography (HPLC), ultra-performance liquid chromatography–tandem mass spectrometry (UPLC-MS/MS), and nuclear magnetic resonance (NMR) spectroscopy. Phenol–sulfuric acid and TLC methods are applied for the quantification of primary carbohydrates and analysis of sulfate groups. The increase in sulfate groups is positively correlated with the increase in antioxidant capacity, and the spots of different colors represent unique compounds or chemical components of the sample mixture. FT-IR spectroscopy and HPLC are used for functional group identification and monosaccharide composition analysis, while UPLC-MS/MS provides detailed structural insights through the mass-to-charge ratio (m/z) distribution, cluster analysis, and solid-phase extraction. Finally, NMR spectroscopy provides a comprehensive understanding of the backbone structure and branching pattern of the polysaccharide. Each technique provides unique information, allowing for a deeper understanding of the diverse and complex structural properties of fucoidan.
Figure 1. Analytical techniques used to explore the complex structure of the sulfated polysaccharide fucoidan, including the phenol–sulfuric acid method/thin-layer chromatography (TLC), Fourier transform infrared (FT-IR) spectroscopy, high-performance liquid chromatography (HPLC), ultra-performance liquid chromatography–tandem mass spectrometry (UPLC-MS/MS), and nuclear magnetic resonance (NMR) spectroscopy. Phenol–sulfuric acid and TLC methods are applied for the quantification of primary carbohydrates and analysis of sulfate groups. The increase in sulfate groups is positively correlated with the increase in antioxidant capacity, and the spots of different colors represent unique compounds or chemical components of the sample mixture. FT-IR spectroscopy and HPLC are used for functional group identification and monosaccharide composition analysis, while UPLC-MS/MS provides detailed structural insights through the mass-to-charge ratio (m/z) distribution, cluster analysis, and solid-phase extraction. Finally, NMR spectroscopy provides a comprehensive understanding of the backbone structure and branching pattern of the polysaccharide. Each technique provides unique information, allowing for a deeper understanding of the diverse and complex structural properties of fucoidan.
Ijms 25 11771 g001
Table 4. Role of glycosidic bond structures in brown algae in determining its bioactivity.
Table 4. Role of glycosidic bond structures in brown algae in determining its bioactivity.
Algae SourceGlycosidic Bond Connection BioactivityExperimental MethodologiesExperiment ModelConcentration Reference
Sargassum aquifolium[2-linked α-d-Manp and 4-linked β-d-GlcpA]Anticoagulant and antitumor activitiesIn vitroHuman cancer cell lines HepG2 (hepatocellular carcinoma), LU-1 (lung adenocarcinoma), and RD (rhabdomiosarcoma) ND[43]
Sargassum horneri[α-l-Fucp-1→3-α-l-Fucp(4SO3)-1→]AntitumorIn vitroThe DLD-1 (ATCC # CCL-221™) human colon carcinoma cell line200 μg/mL[55]
Fucus distichus subsp. evanescens[3)-α-L-Fucp-(2SO3)-(1→4)-α-L-Fucp-(2,3SO3)-(1→]Anticancer activityIn vitroThe SK-MEL-5 (ATCC # HTB-70), SK-MEL-28 (ATCC # HTB-72) human malignant melanoma cell lines100–400 μg/mL[60]
Sargassum crassifolium[3)-α-L-Fucp-(1→3)-α-L-Fucp (SO3)-(1→4)-α-L-Fucp-(SO3)-(1→]Immunomodulatory activityIn vitroBone marrow cells from C3H/HeJ female mice3 μg/mL[74]
Sargassum polycystum[3)-α-L-Fucp-(1→3)-α-L-Fucp-(1→]Antioxidant activity, anticancer activityIn vitroMCF-7 cells25, 50, 75, 100, 125, and 150 μg/ml[59]
Sargassum siliquosum[3)-α-L-Fucp-(2SO3)-(1→4)-α-L-Fucp-(1→]Antioxidant activity, anti-inflammatory activityIn vitroRAW 264.7 cell0.25–1 μg/mL[15]
Padina boergesenii(1–4)-L fucose, (1–6) β-D galactose, α and β-D Manncronic acid Antioxidant and anticancerIn vitroHuman cervical carcinoma cells (HeLa cell line)20, 40, and 60 μg/mL[61]
SaccharinajaponicaA:→3)-α-l-Fucp(2,4S)-(1→
B:→3)-α-l-Fucp(2S)-(
C:→3)-α-l-Fucp-(1→
D:→4)-β-d-Manp(1→
E:→6)-β-d-Galp(1→
Antibacterial activity and anti SARS-CoV-2 In vitroBacteria including Staphylococcus aureus ATCC6538, Listeria monocytogenes ATCC19115, Escherichia coli ATCC25922, Shigella flexneri CMCC51574, Salmonella typhimurium ATCC14028, and Vibrio parahaemolyticus CGMCC1.161450, 25, 12.5, 6.25, and 0 μg/mL[53]
Undaria pinnatifida[β-D-Galp, α-type glycosidic linkages]Anticancer activityIn vivoSprague Dawley rats100, 200, and 300 mg/kg of body weight[56]
α(1,4)-linked L-fucopyranose Attenuation of SARS-CoV-2 infection In vitro
In vivo
Caco-2-Nint cells, a producer cell line expressing the SARS-CoV-2 N protein via lentiviral transduction 7.8, 15.6, 31.3, 62.5, 125, 500, 1000 μg/mL [44]
Ascophyllum nodosum[(1→3) and (1→4) linked α-l-fucopyranose]Four-week-old female specific-pathogen-free (SPF) Syrian hamstersOrally gavaged with high dose (Hd; 200 mg/day/kg body weight) or low dose (Ld; 100 mg/day/kg body weight)
Ishige okamurae[→3)-α-l-Fucp-(1→, →4)-α-l-Fucp-(1→, →6)-β-d-Galp-(1→ and →3)-β-d-Galp-(1 → residues with sulfate groups at C-2/C-4 the of (1→3)-α-l-Fucp and C-6 the of (1→3)-β-d-Galp]Effect on recovery from immunosuppressionIn vivoBALB/c mice induced CTX (cyclooxygenase-thromboxane A2 synthetase) immunomodulatory models20, 40, and 80 mg/kg[62]
Stoechospermum polypodioides[(1→4)- and (1→3)-linked-α-l-fucopyranosyl]Antiviral activityIn vitroVero cells by a virus plaque reduction assayEC50 (μg/mL): 3.55 ± 0.63[51]
ND: no data.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jeong, S.; Lee, S.; Lee, G.; Hyun, J.; Ryu, B. Systematic Characteristics of Fucoidan: Intriguing Features for New Pharmacological Interventions. Int. J. Mol. Sci. 2024, 25, 11771. https://doi.org/10.3390/ijms252111771

AMA Style

Jeong S, Lee S, Lee G, Hyun J, Ryu B. Systematic Characteristics of Fucoidan: Intriguing Features for New Pharmacological Interventions. International Journal of Molecular Sciences. 2024; 25(21):11771. https://doi.org/10.3390/ijms252111771

Chicago/Turabian Style

Jeong, Seungjin, Seokmin Lee, Geumbin Lee, Jimin Hyun, and Bomi Ryu. 2024. "Systematic Characteristics of Fucoidan: Intriguing Features for New Pharmacological Interventions" International Journal of Molecular Sciences 25, no. 21: 11771. https://doi.org/10.3390/ijms252111771

APA Style

Jeong, S., Lee, S., Lee, G., Hyun, J., & Ryu, B. (2024). Systematic Characteristics of Fucoidan: Intriguing Features for New Pharmacological Interventions. International Journal of Molecular Sciences, 25(21), 11771. https://doi.org/10.3390/ijms252111771

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop