Next Article in Journal
Selective Utilization of Polyguluronate by the Human Gut Bacteroides Species
Previous Article in Journal
Haemostatic and Biocompatibility Evaluation of Alginate-Functionalized Polylactide Composite Containing Zinc Sulphide and Hardystonite
Previous Article in Special Issue
Therapeutic Potential of Sea Cucumber-Derived Bioactives in the Prevention and Management of Brain-Related Disorders: A Comprehensive Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anti-Inflammatory and Neuroprotective Effects of Undaria pinnatifida Fucoidan

1
Faculty of Health, School of Pharmacy and Pharmacology, University of Tasmania, Hobart, TAS 7005, Australia
2
Marinova Pty Ltd., 249 Kennedy Drive, Cambridge, TAS 7170, Australia
3
Central Science Laboratory, College of Science and Engineering, University of Tasmania, Hobart, TAS 7001, Australia
4
School of Psychological Sciences, Psychology, University of Tasmania, Launceston, TAS 7248, Australia
*
Author to whom correspondence should be addressed.
Mar. Drugs 2025, 23(9), 350; https://doi.org/10.3390/md23090350
Submission received: 30 July 2025 / Revised: 25 August 2025 / Accepted: 28 August 2025 / Published: 29 August 2025

Abstract

Undaria pinnatifida fucoidan (UPF), a sulphated polysaccharide derived from brown seaweed, has attracted scientific and clinical interest for its wide-ranging anti-inflammatory and neurodegenerative properties. A growing body of research shows that UPF inhibits NF-κB and MAPK signalling pathways, reduces pro-inflammatory cytokines (TNF-α, IL-1β, IL-6), decreases ROS production, and suppresses iNOS and COX-2 activity, thereby mitigating oxidative and inflammatory damage in vitro. In vivo studies confirm these actions, demonstrating reduced systemic inflammation, promoted antioxidant defence, modulated gut microbiota composition, and improved production of beneficial microbial metabolites. In parallel, emerging evidence highlights UPF’s neuroprotective potential, characterised by protection against neuroinflammation and oxidative stress, the attenuation of amyloid-beta deposition, and improvement in neuronal function. Importantly, low- to medium-molecular-weight and highly sulphated UPF fractions consistently exhibit stronger bioactivities, suggesting a structural basis for its therapeutic potential. This review integrates mechanistic evidence from cellular, preclinical, and emerging clinical studies, highlighting UPF as a versatile marine-derived agent with therapeutic relevance for inflammatory and neurodegenerative diseases, and outlines future research directions toward clinical translation.

1. Introduction

Fucoidans are a class of highly sulphated polysaccharides that are abundantly present in the cell walls of brown algae, including Undaria pinnatifida [1]. These polysaccharides have attracted considerable scientific attention due to their diverse and potent biological activities in the treatment of inflammatory-related diseases [2], metabolic disorders [3], cardiovascular conditions [4], and several cancers [5,6,7].
The structure of Undaria pinnatifida fucoidan (UPF) is characterised by its sulphated galactofucan architecture, where a fucose-galactose backbone with α-(1→3)-linked L-fucose is interspersed with α-(1→4)/α-(1→3)/α-(1→6) galactosyl residues that introduce branch points, as well as sulphate esters that are characteristically placed at C2/C4 of fucose and at C3/C4 of galactose [8,9]. In contrast, Fucus spp. (typical “type II” fucoidans) predominantly feature an all-fucose backbone with alternating α-(1→3)/α-(1→4) linkages and sulphate mainly at C2/C4, with far less galactose incorporation [10,11]. Across extracts, UPF commonly presents as moderately to highly sulphated material spanning a broad molecular-weight distribution, from low-molecular-weight (LMW) fractions (<10 kDa) to very high-molecular-weight (HMW) material (>300 kDa), with many food/biomedical fractions falling in the range of 10 to 500 kDa, largely dictated by harvest and processing [12,13]. Its co-monomer composition typically includes abundant galactose alongside fucose, with minor xylose, glucose, mannose, and occasional uronic acids, supporting the galactofucan designation and differentiating UPF from fucoidans of Laminaria or Fucus that are often more fucose-dominant [14,15,16,17]. Collectively, UPF’s C2/C4-focused sulphation, galactose-enriched backbone with branching via α-(1→6) galactose, and broad yet tuneable molecular-weight profile provide a clear structural context for interpreting its distinct bioactivity relative to other brown-algal fucoidans [8]. The structural diversity of fucoidans, shaped by factors such as seaweed species, degree and position of sulphation, molecular weight, and extraction methods, plays a crucial role in determining their bioactivity [18,19,20,21].
Among the various sources of fucoidan, recent research has increasingly focused on UPF, which has shown notable anti-inflammatory [22], antioxidant [23], immunomodulatory [24], antiviral [25], and neuroprotective effects [26], suggesting its significant potential in biomedical applications.
This review aims to provide a comprehensive overview of the anti-inflammatory and neuroprotective activities of UPF. Specifically, it synthesises current evidence on the molecular mechanisms responsible for these biological effects and evaluates the therapeutic potential of UPF in the management of chronic inflammatory conditions and neurodegenerative diseases, identifying directions for future research and clinical applications.

2. Materials and Methods

This literature review synthesises findings from peer-reviewed articles obtained through systematic searches of PubMed, Scopus, Web of Science, and ScienceDirect covering the period from 2000 to 2025. Keywords used included “Undaria pinnatifida”, “fucoidan,” “sulphated polysaccharide”, “structure,” “biological activity,” “anti-inflammation,” “neuroprotection,” and “mechanisms”. Only peer-reviewed original research articles and review papers published in English were considered. Inclusion criteria were studies that investigated the biological activities of UPF or its fractions in in vitro, in vivo, or clinical trials; provided mechanistic insights related to inflammation or neuroprotection; and reported clear experimental results. Exclusion criteria included conference abstracts, non-peer-reviewed reports, studies not specific to UPF, and those lacking structural, mechanistic, or biological relevance. Reference lists of selected articles were also screened to identify additional eligible publications. In total, 129 studies were included in this review.
For each included study, we extracted experimental settings and key findings of UPF used, including model/system, dose/regimen, main action mechanisms, and references in the main text. We also recorded physicochemical and sourcing details of UPF used, including algal part/tissue, origin (including supplier, if stated), extraction and purification, molecular weight, degree of sulphation, content of uronic acid, purity, monosaccharide composition (ratios of fucose, galactose, xylose, glucose, mannose, glucuronic acid, arabinose, rhamnose, fructose, and galacturonic acid), and references in the Supplementary Table S1. When a parameter was not reported in the source article, it was coded as NR (not reported).

3. Anti-Inflammatory Activity of UPF

3.1. In Vitro Studies

Several in vitro studies have indicated that UPF effectively inhibits the nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signalling pathways, suppresses oxidative stress, and reduces the expression of pro-inflammatory mediators, thereby mitigating diverse inflammation-related responses (Figure 1).
A recent investigation has shown that a 4-h pre-treatment with UPF (10, 50, and 100 µg/mL) significantly suppressed the lipopolysaccharide (LPS)-induced upregulation of pro-inflammatory cytokines, including tumour necrosis factor alpha (TNF-α), interleukin-1 beta (IL-1β), and IL-6, in human macrophages and peripheral blood mononuclear cells (PBMCs) [27]. A low-molecular-weight UPF (100 µg/mL) also exhibited significant anti-inflammation effects by downregulating the gene expression of these pro-inflammatory cytokines in LPS-induced RAW264.7 cells [28]. Similarly, in a viral challenge model, UPF (200 µg/mL) reduced pro-inflammatory cytokines, including IL-6, IFN-α, interferon gamma (IFN-γ), and TNF-α, in SARS-CoV-2 infected Caco-2-Nint cells [25]. These effects are primarily mediated through inhibition of key inflammatory signalling pathways, such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinase (MAPK), which are known to regulate cytokine gene transcription [29]. For example, treatment with UPF suppresses the nuclear translocation of NF-κB p65 and reduces the phosphorylation of p38 and extracellular signal-regulated kinases (ERK)1/2 MAPKs, leading to a marked decrease in pro-inflammatory cytokine expression [1,28,29]. Vaamonde-García et al. (2021) reported that UPF (5 µg/mL) treatment attenuated IL-1β-induced inflammation in osteoarthritic chondrocytes by blocking nuclear translocation of NF-κB and inhibiting its activation [30]. In addition, UPF inhibited NF-κB signalling and downregulated IL-6 and TNF-α in human colon carcinoma cell line (Caco-2) cells [31].
The suppression of oxidative stress represents a fundamental mechanism by which UPF alleviates inflammation. Oxidative stress, driven by excess reactive oxygen species (ROS), is a key initiator of inflammatory signalling cascades in various cell types [32]. In intestinal epithelial cell line (IEC-6) cells, UPF (100 µg/mL) significantly reduced oxidative damage induced by hydrogen peroxide (H2O2), as evidenced by decreased levels of malondialdehyde (MDA) and increased activity of antioxidant enzymes, including catalase (CAT), total superoxide dismutase (T-SOD), and glutathione (GSH) [33]. These changes were associated with protection against apoptosis and inhibition of pro-inflammatory responses, indicating that UPF enhances cellular antioxidant defences to maintain redox balance. Moreover, in RAW264.7 macrophages, low molecular weight UPF (100 µg/mL) inhibited LPS-induced ROS production and suppressed the phosphorylation of key MAPK signalling proteins (p38, ERK1/2, and JNK), leading to a significant reduction in the expression of inflammatory markers such as TNF-α, IL-6, and IL-1β [28]. Phull et al. (2017) found that UPF (15.52–500 µg/mL) exerted significant antioxidant activity in a dose-dependent manner in various in vitro antioxidant assays, including iron chelating, hydroxyl, nitric oxide, and DPPH activity, along with a reduction in inflammation responses in rabbit articular chondrocytes [34]. Another in vitro study also indicated that Undaria pinnatifida water extract (UPE) obtained by ultrasonication (200 and 400 µg/mL) significantly suppressed ROS production and restored H2O2-induced viability reduction in monkey kidney (Vero) cells in a dose-dependent manner. [35]. The cell-protective activity of the extract in this study was attributed to its capability to decrease pro-apoptotic protein (Bax) and increase anti-apoptotic protein (Bcl-2) [35].
Interestingly, there are an increasing number of studies correlating the antioxidant properties of UPF with its sulphate content and molecular weight [9,12,14,35,36,37]. For instance, UPF fractions with higher sulphation levels have been shown to exhibit significantly greater antioxidant activity compared to their lower-sulphated counterparts [9]. Moreover, fractionation studies revealed that low molecular weight UPF components possess enhanced antioxidant effects relative to high molecular weight forms, particularly in assays such as DPPH radical scavenging and ferric-reducing antioxidant power (FRAP) tests [12].
UPF inhibits major inflammatory mediators, including inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), nitric oxide (NO), and prostaglandin E2 (PGE2), whose overproduction exacerbates inflammatory responses and tissue damage [38,39]. For instance, low molecular weight UPF (100 µg/mL) alleviated LPS-induced inflammation in RAW264.7 cells by suppressing iNOS and COX2 activities [28]. Similarly, Song et al. in 2015 demonstrated that UPF (50 µg/mL) significantly inhibited iNOS and COX-2 expression, as well as attenuated the production of NO and PGE2 in LPS-stimulated RAW264.7 macrophages [40]. Lim et al. (2022) also reported that high molecular weight UPF (500 µg/mL) mitigated MG-H1-induced inflammation in Caco-2 cells by suppressing protein expression of COX-2 and iNOS and inhibiting NF-κB activation [31]. Additionally, UPF exerted potent anti-inflammatory effects in rabbit articular chondrocytes, where UPF significantly reduced COX-2 expression in a dose-dependent (0–100 µg/mL, 24 h) and time-dependent (30 µg/mL, 0–48 h) manner [34]. Moreover, Vaamonde-García et al. found that UPF (5 µg/mL) significantly inhibited IL-1β-induced production of NO, PGE2, and IL-6 in osteoarthritic chondrocytes, suggesting an immunomodulatory role of UPF in inflammatory conditions [30].
Recent in vitro investigations have suggested that UPF is capable of suppressing chemokine secretion in various inflammatory cells. Chemokines, also known as chemotactic cytokines, are a family of small signalling proteins that significantly contribute to regulating the migration and activation of immune cells during inflammatory responses [1,41]. According to the structure of N-terminal cysteine residues, chemokines are classified into four major subfamilies, including CXXXC (fractalkine), C-X-C (IL-8), C-C (monocyte chemoattractant protein [MCP-1], or monocyte inflammatory protein [MIP-1α], and MIP-1β), and C chemokines (lymphotactin) [41]. Chen et al. (2025) demonstrated that sulphated Undaria pinnatifida polysaccharides (50 and 200 µg/mL) significantly reduced MCP-1 production in vitro during oxalate crystal-induced inflammation in renal cells [42]. This reduction was linked to decreased cellular inflammation and oxidative stress, indicating the potential role of UPF in modulating chemokine-driven immune cell recruitment [42]. Kim et al. also reported that UPF treatment (100 µg/mL) significantly suppressed MCP-1 expression in 3T3-L1 adipocytes, indicating that UPF inhibits inflammation-associated chemokine signalling during adipocyte differentiation [43]. In addition, Vaamonde-García et al. (2022) indicated that UPF (5 µg/mL) significantly downregulated IL-6 and IL-8 (CXCL8) in IL-1β-induced human chondrocyte cells [44]. Similarly, Wimmer et al. (2025) demonstrated that UPF significantly reduced the secretion of pro-inflammatory chemokines (IL-8 and MCP-1) and increased production of anti-inflammatory cytokines (IL-6 and IL-10) in the Caco-2/THP-1 co-culture system after microbial stimulation [45], indicating that UPF can suppress immune cell recruitment and inflammatory signalling at the gut mucosal level. Moreover, a study on atopic dermatitis found that UPF significantly inhibited the mRNA expression of several key chemokines, including thymus- and activation-regulated chemokine (TARC), macrophage-derived chemokine (MDC), and RANTES (also known as CCL5), in TNF-α or IFN-γ-induced human epidermal keratinocytes [46].
The major outcomes derived from in vitro investigations into the anti-inflammatory effects of UPF are listed in Table 1.

3.2. In Vivo Studies

A substantial body of in vivo studies further supports the notion that UPF exerts its anti-inflammatory effects by suppressing pro-inflammatory cytokines such as TNF-α, IL-1β, and IL-6 [22,47,48,49]. Herath et al. (2020) reported that a 7-day oral administration of UPF (400 mg/kg/day) significantly attenuated particulate matter (PM) and ovalbumin (OVA)-induced IL-4, IL-17a, and IL-33 increase in lungs of a murine model of allergic airway inflammation [48]. In a later study, 27 days of UPF supplementation (400 mg/kg/day) significantly reduced TNF-α, IL-6, and IL-1β, and mitigated inflammation responses in the colon of dietary fibre deficiency (FD)-induced mice [33]. Similar effects of UPF were reported by Shi et al. (2024) in a Syrian hamster model of virus infection, where 6 days oral administration of UPF (200 mg/kg/day) alleviated SARS-CoV-2-induced lung and gastrointestinal tract injury by suppressing gene expression of TNF-α and IL-6 [25]. Lim et al. (2022) also indicated that 4 weeks oral administration of high molecular weight UPF (25 mg/kg/day) significantly inhibited MG-H1-caused TNF-α increase in mouse colon tissues [31]. Similarly, a 10-week oral administration of UPF (400 mg/kg/day) suppressed systemic inflammation in a high-fat diet (HFD)-induced obese mouse model [22]. The results of the study showed that UPF significantly reduced the expression of pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6) in skeletal muscle, small intestine, and hypothalamus [22].
The antioxidant properties of UPF have been well demonstrated in animal models of inflammatory-related disorders. In HFD-fed mice, a 10-week oral administration of sulphated polysaccharides from Undaria pinnatifida significantly (100, 300, and 500 mg/kg/day) reduced markers of oxidative stress, including MDA and SOD in liver tissues, alongside an inhibition of triglycerides (TG), low-density lipoprotein cholesterol (LDL-c), and TNF-α production, suggesting that the suppression of oxidative stress contributed to hepatic lipid metabolism improvements, and mitigated HFD-induced inflammatory conditions [50]. Similarly, 7 days of UPF oral gavage (100 and 400 mg/kg/day) attenuated MDA in the serum and lungs of PM-induced allergic airway inflammatory mice [48]. Phull et al. (2017) also reported that 25 days UPF administration (150 mg/kg/day) significantly decreased arthritis-induced endogenous antioxidant enzymes such as CAT, peroxidase (POD), and SOD [34]. This reduction was mainly due to UPF capability to scavenge the free radicals, abrogate ROS-induced oxidative stress, and maintain the oxidative flux [34]. In contrast, Kang et al. found that 14 days UPF intraperitoneal administration (100 mg/kg/day) markedly prevented oxidative stress in carbon tetrachloride (CCL4)-induced rats by increasing antioxidant enzymes (CAT, SOD, and glutathione peroxidase [GPx]), and decreasing markers of oxidative damage (MDA) in liver [51]. These results are in line with the findings of Zheng et al. (2023), where 27 days of UPF treatment (400 mg/kg/day) significantly elevated the levels of CAT and T-SOD, and attenuated myeloperoxidase (MPO) and MDA production in the colon tissues of FD-induced mice [33], suggesting that UPF exerts a protective effect against inflammation-associated oxidative damage by enhancing endogenous antioxidant defences and reducing lipid peroxidation, thereby contributing to the amelioration of oxidative stress in various inflammatory disease models.
Recent studies have also demonstrated that UPF exhibits strong antioxidant activities in an in vivo zebrafish model, a vertebrate species with notable biochemical and physiological similarities to mammals [37,52]. The findings suggest that UPF effectively mitigates oxidative stress induced by 2,2′-azobis (2-amidinopropane) dihydrochloride (AAPH) and H2O2, as evidenced by increased survival rates, reduced cellular apoptosis, decreased heart rate, and lower levels of ROS and lipid peroxidation [37,52].
UPF has been shown to ameliorate inflammatory responses by modulating gut microbiota composition. A bidirectional relationship exists between gut dysbiosis and host inflammation, whereby microbial imbalance promotes inflammatory processes, which in turn further disrupt the gut microbial ecosystem [53,54,55,56]. Recent studies have indicated that UPF treatment significantly attenuated intestinal inflammation by restoring microbial balance, notably decreasing Firmicutes and increasing Bacteroidetes in the gut of HFD-induced obese mice [50,57,58,59,60]. Comparable outcomes were observed in FD-induced inflammatory mouse models, where 27 days of oral UPF administration (300 and 400 mg/kg/day) led to a marked increase in Bacteroidetes and a reduction in Firmicutes within colon tissues [33,61]. As the dominant phyla in the gut, Firmicutes and Bacteroidetes play key roles in maintaining intestinal homeostasis [62], and imbalances in their ratio have been associated with various inflammatory disorders [63,64,65,66]. The anti-inflammatory effects of UPF may also derive from its prebiotic properties, as evidenced by reductions in pathogenic taxa (Faecalibaculum, Desulfovibrionales, Proteobacteria, and Clostridia) and enrichment of beneficial bacteria (Akkermania muciniphila, Bacteroides, Bifidobacterium spp., and Lactobacillus) [58,59,60,67]. Additionally, Park et al. (2024) reported that 4 weeks of UPF supplementation (50, 100, and 200 mg/kg/day) significantly increased the abundance of Papillibacter cinnamivorans, a butyrate-producing bacterium, in immunosuppressed rats [68]. Butyrate, one of the short-chain fatty acids (SCFAs), mitigates inflammation by interacting with immune cells, promoting anti-inflammatory cytokines, and suppressing pro-inflammatory mediators through G-protein coupled receptors (GPR41/43) and inhibition of histone deacetylases (HDACs) [69,70]. Similarly, Zheng et al. (2023) have suggested that 27 days of UPF supplementation (400 mg/kg/day) significantly restored HFD-induced reduction in colonic SCFAs, including acetate, propionate, and butyrate [33], suggesting that UPF may exert its anti-inflammatory effects, at least in part, by restoring SCFA levels and modulating immune responses through established SCFA-mediated pathways.
UPF has been reported to attenuate immune cell infiltration, including macrophages and T cells, and to ameliorate inflammatory responses in allergic conditions. Herath et al. (2020) indicated that 7 days of UPF oral gavage (400 mg/kg/day) significantly reduced PM-exacerbated infiltration of inflammatory cells, such as F4/80+ macrophages, CD4+ T lymphocytes, Gr-1+ granulocytes, and eosinophils, in the trachea and lungs of OVA-sensitised mice [48]. The results also showed that UPF decreased serum level of immunoglobulin E (IgE) and suppressed inflammatory provocation-induced increase in goblet cell hyperplasia and mucus secretion [48], suggesting potent therapeutic effects of UPF in allergic airway inflammation. Similarly, Yu et al. (2024) demonstrated that 16 days administration of ethanol-extracted UPE (50, 100, and 200 mg/kg/day) mitigated combined allergic rhinitis and asthma syndrome by inhibiting the accumulation of inflammatory cells, including epithelial cells, eosinophils, neutrophils, lymphocytes, and macrophages, in both nasal and bronchoalveolar lavage fluid, as well as a reduction in Th2 cytokines expression (IL-4, IL-5, and IL-13) [71].
The capability of UPF to re-establish immune homeostasis also plays a significant role in mitigating inflammatory conditions. Several in vivo studies have demonstrated that UPF exerts immunomodulatory effects by upregulating the expression of the anti-inflammatory cytokine IL-10 while concurrently downregulating the production of pro-inflammatory cytokines in various animal models of inflammatory intestinal diseases [50,59,61,72]. The main results of in vivo evaluations of UPF effects are listed in Table 2.
To aid interpretation, the study-level characteristics of the UPF used in each experiment (source, extraction/purification, molecular weight, sulphation, uronic acid, purity, monosaccharides) are summarised in Supplementary Table S1. For transparency, non-reported items are indicated as NR.

3.3. Clinical Trials

A growing body of clinical evidence supports the therapeutic potential of UPF in modulating metabolic, immune, and inflammatory responses in humans. Various human studies have demonstrated that incorporating Undaria pinnatifida (4–6 g/day) into diets help improve metabolic parameters, including suppressed postprandial glycaemia, modulated appetite sensations, reduced waist circumference and blood pressure, as well as a decrease in total cholesterol, LDL-cholesterol, and resistin levels [73,74,75,76]. Undaria pinnatifida also exerts potent immunostimulatory properties to manage Herpes infections by promoting healing and preventing reactivation [77]. Moreover, a clinical trial showed that a single dose of UPF (1 g) modulated microRNA expression related to immune response and inflammation, highlighting its systemic regulatory potential [78]. Additionally, a combination of UPF and Fucus vesiculosus fucoidan significantly increased faecal lysozyme levels, a protein known for its antimicrobial and anti-inflammatory functions, suggesting that UPF may promote mucosal barrier integrity and reduce mucosal inflammation [79]. Cox et al. (2023) reported that 3 weeks of UPF supplementation (1 g/day) significantly increased salivary immunoglobulin (Ig) A contents after an intensified training, indicating UPF properties to enhance mucosal immunity and provide protective anti-inflammatory benefits [80,81]. In a double-blind randomised placebo-controlled clinical trial, 2 weeks of UPF administration (1 g/day) significantly suppressed the upregulation of inflammatory cytokines induced by high-intensity exercise [82]. Similarly, UPF combined with green-lipped mussel mitigated joint pain and prediabetes in a randomised, double-blinded clinical setting, demonstrating that UPF elicits antioxidant and anti-inflammatory effects [83].

4. Neuroprotective Effect of UPF

The scientific literature has reported an increasing number of studies highlighting the neuroprotective effects of UPF in promoting brain health and mitigating the progression of neurodegenerative diseases such as Alzheimer’s (AD). These effects arise from multifactorial interplay involving anti-inflammatory [22], anti-apoptotic [84], antiviral [26], and antioxidant activities [85] of UPF. Findings from both in vitro and in vivo investigations demonstrate that UPF can attenuate neuronal apoptosis, inhibit amyloid-β (Aβ) aggregation, and suppress the activation of microglia and astrocytes by reducing oxidative stress and neuroinflammation across various experimental models of neurodegenerative conditions [22,26].

4.1. In Vitro Studies

Several in vitro studies demonstrated that UPF is capable of suppressing inflammation in neurodegenerative conditions, primarily through inhibition of the NF-κB signalling pathway, mitigation of oxidative stress, and modulation of adenosine monophosphate-activated protein kinase (AMPK) and mechanistic target of rapamycin (mTOR) pathways (Figure 2).
Notably, Giuliani et al. (2025) reported that UPF (100 µg/mL) significantly attenuated herpes simplex virus type I (HSV-1)-induced AD-like pathology [26]. This included a reduction in amyloid precursor protein (APP) production and Aβ synthesis, alongside inhibiting NF-κB pathway activation and reducing IL-6 expression [26]. Ethanol-extracted UPE (5 µg/mL) also has been suggested to reduce endoplasmic reticulum (ER) stress and increase cell viability in hypothalamic neurons via Akt/mTOR signalling, highlighting its anti-inflammatory and neuroprotective potential [86]. ER stress is closely linked to the activation of inflammatory responses and is increasingly recognised as a contributing factor in the pathogenesis of various neurodegenerative diseases [87]. Additionally, Chen et al. (2025) suggested that low molecular weight UPF (0.125 mg/mL) significantly ameliorated LPS-induced macrophage inflammatory state by promoting its polarisation from pro-inflammatory M1 phenotype to anti-inflammatory M2 phenotype through the AMPK/mTOR pathway [88]. Literature has indicated that modulating the AMPK/mTOR pathway regulates microglia polarisation and reduces neuroinflammation [89].
UPF has also been shown to exert neuroprotective effects in various in vitro cell models of neurodegeneration by enhancing cell viability and attenuating cytotoxicity, particularly in response to neurotoxic insults such as Aβ and oxidative stress [26,84,85,90,91,92]. For instance, Wei et al. (2017) demonstrated that pre-treatment with UPF (100, 200, 400 µg/mL) for 24 h protected PC12 cells from apoptosis induced by Aβ25–35 and d-galactose (D-Gal), alongside elevated levels of SOD and GSH [85]. Similar effects were observed in a rat cholinergic basal forebrain neuron model of AD conditions, where treatment of a commercial UPF (1 µM) inhibited cellular and neurotoxic effects of Aβ1−42 and suppressed ROS production [90]. In addition, UPF demonstrated strong free radical-scavenging activity, effectively inhibiting DPPH and hydroxyl radicals, and reducing ROS production as well as Aβ synthesis in HSV-1-infected retinal pigment epithelium (RPE) cells [26]. HSV-1 infection and Aβ synthesis have been associated with the development of AD [93]. Mohibbullah et al. (2018) also reported that ethanol-extracted UPE (15 µg/mL) enhanced cell viability and reduced cytotoxicity in hippocampal neurons by decreasing ROS generation, membrane phosphatidylserine exposure, genomic DNA degradation, and restoring hypoxia-induced mitochondrial depolarization [92]. Notably, although both fucoidans reduced Aβ1–42-induced oxidative stress and apoptosis levels, UPF exhibited stronger neuroprotective effects than Fucus vesiculosus fucoidan, likely due to its distinct structural features such as higher sulphate content and specific molecular weight distribution [84].
The key in vitro outcomes regarding the neuroprotective activity of UPF are comprehensively outlined in Table 3.

4.2. In Vivo Studies

UPF has emerged as a promising neuroprotective agent due to its ability to attenuate neuroinflammation. For example, 10 weeks of UPF oral administration (400 mg/kg/day) significantly attenuated HFD-induced neuroinflammation in obese mice by downregulating the expression of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6, and IFN-γ) in hypothalamic tissues and suppressing the production of inflammation-related proteins (leucine-rich repeat serine/threonine-protein kinase 2 [Lrrk2], wolframin [Wfs1], and neuroglobin [Ngb]) in the nucleus accumbens [22]. Hu et al. (2014) also reported that a 10-day intrathecal injection of commercial UPF (15, 50, and 100 mg/kg/day) mitigated rat neuropathic pain induced by L5 spinal nerve ligation (SNL). The findings suggested that UPF inhibited microglia and astrocyte activation in the lumbar spinal cord and reduced TNF-α, IL-1β, and IL-6 expression in the spinal dorsal horn [94]. Similarly, Che et al. (2017) demonstrated that intraperitoneal injection of commercial UPF (80 and 160 mg/kg/day) for 7 days significantly ameliorated cerebral ischemia–reperfusion injury (IRI)-caused neurological impairment in rats and significantly decreased the levels of pro-inflammatory cytokines, including IL-1β, IL-6, MPO, and TNF-α [95].
Several in vivo studies have highlighted the effects of UPF in neuroprotection and rehabilitation, demonstrating its beneficial properties in inhibiting oxidative stress and attenuating neurotoxic protein aggregation. Specifically, 21 days of oral administration of UPF (50, 100, and 200 mg/kg/day) improved learning and memory impairments in AD-model mice induced by D-Gal, where UPF exhibited potent antioxidant effects, enhancing SOD and GSH activity [85]. The ability of UPF to promote learning and memory in this study is largely attributed to its enhancement of acetylcholine (ACh) content and choline acetyl transferase (ChAT) enzyme activity, along with the inhibition of acetylcholine esterase (AChE) enzyme activity, which are key factors involved in the cognitive dysfunction characteristic of AD [85]. UPF also has been reported to reduce oxidative stress-related proteins (SOD and MDA), suppress pro-apoptotic proteins (p-p53 and Bax), and elevate anti-apoptotic protein (Bcl-2) in IRI-induced rats by inhibiting MAPK pathway [95]. Similarly, Wang et al. (2016) illustrated that intraperitoneal pre-treatment of low molecular weight commercial UPF (50 mg/kg) significantly suppressed neuronal damage and neurological deficits in aged mice after traumatic brain injury (TBI), where UPF exerted these protective effects by inhibiting oxidative stress (reduced MDA, 4-hydroxynonenal [4-HNE], ROS and increased CAT, SOD, GPx) and mitochondrial dysfunction (suppressed cytochrome c release) [96]. In addition, the neuroprotective effects of UPF have been found in an invertebrate model of AD, where UPF (500 ng/mL) alleviated Aβ-induced paralysis by decreasing Aβ deposition and ROS production in transgenic Caenorhabditis elegans [97]. Taken together, these findings suggest that UPF confers neuroprotection across diverse experimental models by modulating oxidative stress, mitochondrial integrity, and apoptosis-related pathways, supporting its potential as a therapeutic agent in the prevention and treatment of neurodegenerative disorders (Table 4).
Key physicochemical information for the UPF used in the cited studies is provided in Supplementary Table S1 (source, extraction/purification, molecular weight, sulphation, uronic acid, purity, monosaccharides), with unreported parameters marked as NR.

5. Discussion

5.1. Toxicity Study of UPF

UPF demonstrates a favourable safety profile across preclinical and clinical studies. The U.S. Food and Drug Administration (FDA) issued “no questions” responses to GRAS notices for fucoidans derived from Undaria pinnatifida and Fucus vesiculosus, supporting their safety for use in food applications with estimated daily intakes of up to 250 mg/day [98,99]. In animal models, repeated oral administration of UPF for four weeks at doses up to 1350 mg/kg/day in SD rats produced no treatment-related adverse effects on clinical signs, body weight, haematology, or histopathology [100]. Similarly, sub-chronic assessment of fucoidan from Laminaria japonica administered to Wistar rats for six months demonstrated no significant toxicological effects at 300 mg/kg/day [101]. Additionally, standard genotoxicity tests, including the bacterial reverse-mutation assay, in vitro chromosomal aberration, and in vivo micronucleus test, demonstrated no mutagenic or clastogenic potential [102]. Human data further support the safety of UPF use. A pilot study administering 3 g/day of UPF for 12 days reported mild, subclinical prolongation of activated partial thromboplastin time, without clinically significant anticoagulant activity [103]. Other trials with oral doses of 1–3 g/day for several weeks to months reported no adverse effects or alterations in liver and kidney function [98,99]. Given fucoidan’s structural similarity to heparin, theoretical anticoagulant interactions remain a consideration, particularly in patients on antithrombotic therapy [104,105]. Importantly, safety is influenced by extraction and purification. Commercial UPF preparations, such as those produced using proprietary aqueous extraction processes, are highly purified and standardised, ensuring consistency and minimising the risk of contaminants [45,49]. Collectively, UPF appears non-toxic, non-genotoxic, and well-tolerated in both preclinical and clinical settings, supporting its safe use as a functional ingredient and potential therapeutic agent, with caution advised for individuals with bleeding disorders or on anticoagulant therapy.

5.2. Pharmacokinetics of UPF

The pharmacokinetics of UPF have not yet been comprehensively characterised, representing an important gap in linking systemic exposure with its biological effects. Limited in vitro work has shown that UPF interacts minimally with hepatic CYP450 enzymes but may influence catechol-O-methyltransferase metabolism [106]. However, studies of fucoidans from other brown algae suggest limited gastrointestinal absorption, influenced by molecular weight, sulfation degree, and structural composition [107,108,109]. For instance, orally administered fucoidan from Fucus vesiculosus in rats showed prolonged blood circulation (mean residence time = 6.79 h) and preferential accumulation in the kidneys, spleen, and liver, with relatively low absorption due to high molecular weight and sulphation [109]. Similarly, in mice, low-molecular-weight fucoidan (9.5 kDa) from Saccharina japonica exhibited more rapid absorption (reached maximum concentration at 1.5 h) and higher bioavailability (28.3 %) compared to its high-molecular-weight counterpart [110]. Mechanistic studies using fluorescein-isothiocyanate-labelled fucoidan indicate clathrin-mediated intestinal uptake and tissue distribution, notably to the kidney and liver [111]. Additionally, a Heparin Red fluorescence assay has been shown to enable direct quantification of UPF and other fucoidans in human plasma (0.5–20 µg/mL) [112]. Other human trials have also detected fucoidans (from Undaria pinnatifida and Cladosiphon okamuranus) can cross the intestinal wall and enter circulation, with absorption efficiency estimated at up to 1% of the oral dose absorbed [113,114]. Taken together, these findings suggest that while systemic uptake of oral fucoidan is limited, it is measurable and may be sufficient to exert biological effects directly or indirectly through gastrointestinal interactions and the generation of bioactive oligosaccharides.
By contrast, the high molecular weight and polyanionic characteristics of UPF favour skin-surface and epidermal retention with limited systemic exposure after topical use [115]. For example, in a mouse barrier-disruption model, UPF (171 kDa) accelerated transepidermal water loss recovery and normalised keratinocyte differentiation markers, consistent with effective local bioavailability in the viable epidermis [115]. However, although topical pharmacokinetics of UPF remain largely unexplored, a closely related fucoidan ointment (735 kDa, source Fucus vesiculosus) showed linear pharmacokinetics after single topical doses (50–150 mg/kg) in rats, with sustained plasma profiles, an apparent elimination half-life of 20.75 ± 9.43 h, and low absolute bioavailability (17.7 ± 7.7% of the applied dose) [116]. Tissue measurements demonstrated a pronounced skin “reservoir” with measurable permeation to underlying muscle but no plasma accumulation after 5 once-daily doses (100 mg/kg), supporting predominantly local disposition with limited systemic exposure [116]. Although derived from Fucus vesiculosus, these findings are mechanistically applicable to UPF given the shared high molecular weight and sulphate density that govern dermal transport. Topical fucoidan (Cladosiphon okamuranus, 49.8 kDa) has also demonstrated anti-inflammatory activity in the skin, as seen in atopic dermatitis and wound models, including reductions in mast cells, epidermal thickness, and serum IgE in mice, as well as broader dermatologic benefits in human cosmetic testing [117,118,119]. Although systemic exposure to topically applied fucoidan is low, local dermal exposure yields significant pharmacological effects [116,120]. For instance, a Fucus vesiculosus fucoidan (735 kDa) cream dose-dependently inhibited carrageenan-induced oedema and mechanical allodynia in rats, with high-dose efficacy comparable to diclofenac gel [120]. These efficacy data align with pharmacokinetics findings of predominant local tissue disposition after topical dosing [120]. Importantly, the lack of comprehensive pharmacokinetic studies on UPF represents a key knowledge gap, and future work is needed to establish dose-exposure-response relationships that can better explain its anti-inflammatory and neuroprotective activities.

5.3. Structure–Activity Relationships of UPF

Synthesis of the available data reveals recurring trends across the reviewed studies. In terms of molecular weight, low- to medium-molecular-weight UPF often shows stronger anti-inflammatory readouts, such as reduced NF-κB/MAPK signalling, iNOS/COX-2, and pro-inflammatory cytokines [27,28,30,42,44,47,71,88]. Fractionation studies also report superior primary antioxidant capacity for <10 kDa compared to >300 kDa fractions, consistent with better diffusion/receptor access [12]. By contrast, very high-molecular-weight UPF can excel in viscosity/barrier-related endpoints, restoring epithelial integrity in MG-H1 injury models in Caco-2 cells and mice [31]. Regarding to the degree of sulphation, UPF is typically a sulphated galactofucan with sulphate esters at fucose C2/C4 and galactose C3/C4 [8,9], and higher sulphate density in fucoidans generally strengthens protein–polysaccharide interactions [121,122], such as P-selectin binding, and downstream anti-inflammatory effects, though activity remains chemotype- and context-dependent [123]. Additionally, for co-monomer composition/branching, Undaria pinnatifida commonly bears substantial galactose (galactofucan) with branching (including 1,6-linked galactose) [124,125], and this architecture recurs in UPF studies reporting mucosal and immunomodulatory benefits, for instance, butyrogenic shifts and cytokine dampening in Simulator of the Human Intestinal Microbial Ecosystem (SHIME) and mouse gut models [33,45,81,126], and improved mucosal immune markers in athletes [79], whereas more fucose-dominant chemotypes from other algae, such as Fucus vesiculosus, can differ in neuroprotective potencies/selectivity [84]. A current literature also reported, compared to other species, a purified Fucus vesiculosus fucoidan (high fucose, reduced phenolics) inhibited protein denaturation with IC50 of 0.20 mg/mL, outperforming diclofenac (0.37 mg/mL), and stabilised human red blood corpuscles (HRBC) membranes to 88% at 0.5 mg/mL underscoring strong species- and structure-dependence of these surrogate anti-inflammatory readouts [127].

5.4. Limitation

Several clinical studies (Section 3.3 Clinical trials) have reported promising anti-inflammatory and immunomodulatory effects of UPF, including improvements in metabolic parameters, mucosal immunity, and exercise-induced inflammation. However, its neuroprotective potential, although strongly supported by preclinical studies (Section 4. Neuroprotective Effects of UPF), has not yet been evaluated in human trials, suggesting the urgent need for well-designed, large-scale clinical studies to justify its therapeutic relevance in clinical practice. Additionally, several studies did not report one or more critical UPF characteristics, such as molecular weight, sulphate content, or monosaccharide composition. This suggests that cross-study comparisons should be interpreted cautiously. We therefore indicated non-reported items as NR in Supplementary Table S1 in this review. Standardised reporting of UPF chemotype (source/tissue, extraction/purification, molecular weight, sulfation, and monosaccharides) would materially strengthen future syntheses and help relate structure to function.

6. Conclusions

Undaria pinnatifida fucoidan (UPF) represents a structurally unique sulphated galactofucan with anti-inflammatory and neuroprotective activities demonstrated in vitro, in vivo, and in ongoing clinical studies. Its anti-inflammatory effects are consistently mediated through inhibition of NF-κB and MAPK pathways, downregulation of pro-inflammatory cytokines and mediators, reduction in oxidative stress, and modulation of immune and gut microbiota responses. Neuroprotective actions observed in preclinical studies include attenuation of neuroinflammation, reduction in oxidative damage and amyloid burden, and enhancement of neuronal survival and antioxidant defences. Notably, low- to medium-molecular-weight and highly sulphated fractions exhibit stronger bioactivities, underscoring clear structure-activity relationships. Despite these promising findings, the molecular mechanisms underlying UPF’s actions remain incompletely understood, and its therapeutic effects in humans have yet to be fully confirmed. Future research should focus on clarifying molecular mechanisms, standardising UPF extraction and structural characterisation, and developing delivery strategies to optimise bioavailability. Most critically, rigorously designed clinical trials are required to confirm efficacy and safety, thereby supporting the integration of UPF into evidence-based therapeutic applications.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/md23090350/s1, Table S1. Study-level characteristics of Undaria pinnatifida fucoidan (UPF) used in Section 3 and Section 4.

Author Contributions

Conceptualisation, C.Y. and V.C.; methodology, C.Y. and V.C.; validation, C.Y., C.D., B.C.W., S.K.A., F.J., J.T., R.W., L.J. and V.C.; resources, C.D., B.C.W. and V.C.; data curation, C.Y., C.D., B.C.W. and V.C.; writing—original draft preparation, C.Y. and V.C.; writing—review and editing, C.Y., C.D., B.C.W., S.K.A., F.J., J.T., R.W., L.J. and V.C.; visualisation, C.Y. and V.C.; supervision, R.W., L.J. and V.C.; project administration, C.D., B.C.W. and V.C.; funding acquisition, V.C., C.D. and B.C.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by The Marine Bioproducts Cooperative Research Centre (MBCRC)-Project 301.

Conflicts of Interest

The current study was sponsored by Marinova Pty. Ltd. (Cambridge, TAS, Australia). C.D. and B.C.W. are employees of Marinova Pty. Ltd. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as potential conflicts of interest.

References

  1. Sanjeewa, K.K.A.; Herath, K.H.I.N.M.; Yang, H.-W.; Choi, C.S.; Jeon, Y.-J. Anti-Inflammatory Mechanisms of Fucoidans to Treat Inflammatory Diseases: A Review. Mar. Drugs 2021, 19, 678. [Google Scholar] [CrossRef] [PubMed]
  2. Apostolova, E.; Lukova, P.; Baldzhieva, A.; Katsarov, P.; Nikolova, M.; Iliev, I.; Peychev, L.; Trica, B.; Oancea, F.; Delattre, C.; et al. Immunomodulatory and Anti-Inflammatory Effects of Fucoidan: A Review. Polymers 2020, 12, 2338. [Google Scholar] [CrossRef] [PubMed]
  3. Yang, Z.; Wang, H.; Liu, N.; Zhao, K.; Sheng, Y.; Pang, H.; Shao, K.; Zhang, M.; Li, S.; He, N. Algal polysaccharides and derivatives as potential therapeutics for obesity and related metabolic diseases. Food Funct. 2022, 13, 11387–11409. [Google Scholar] [CrossRef]
  4. Yao, Y.; Yim, E.K.F. Fucoidan for cardiovascular application and the factors mediating its activities. Carbohydr. Polym. 2021, 270, 118347. [Google Scholar] [CrossRef] [PubMed]
  5. Fernandes, A.S.; Oliveira, C.; Reis, R.L.; Martins, A.; Silva, T.H. Marine-Inspired Drugs and Biomaterials in the Perspective of Pancreatic Cancer Therapies. Mar. Drugs 2022, 20, 689. [Google Scholar] [CrossRef]
  6. Miyata, Y.; Matsuo, T.; Ohba, K.; Mitsunari, K.; Mukae, Y.; Otsubo, A.; Harada, J.; Matsuda, T.; Kondo, T.; Sakai, H. Present Status, Limitations and Future Directions of Treatment Strategies Using Fucoidan-Based Therapies in Bladder Cancer. Cancers 2020, 12, 3776. [Google Scholar] [CrossRef]
  7. 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]
  8. Ke, S.; Zhang, B.; Yu, Y.; Wang, S.; Jin, W.; Wu, J.; Chen, J.; Zhang, H.; Wei, B.; Wang, H. Structural characterization of sulfated galactofucan from Undaria pinnatifida and its effect on type 2 diabetic mice. J. Oceanol. Limnol. 2022, 41, 300–313. [Google Scholar] [CrossRef]
  9. Koh, H.S.A.; Lu, J.; Zhou, W. Structure characterization and antioxidant activity of fucoidan isolated from Undaria pinnatifida grown in New Zealand. Carbohydr. Polym. 2019, 212, 178–185. [Google Scholar] [CrossRef]
  10. 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]
  11. Gasilova, E.R.; Lapina, I.M.; Kulminskaya, A.A.; Skorik, Y.A. Branched architecture of fucoidan characterized by dynamic and static light scattering. Colloid Polym. Sci. 2020, 298, 1349–1359. [Google Scholar] [CrossRef]
  12. Yu, J.; Li, Q.; Wu, J.; Yang, X.; Yang, S.; Zhu, W.; Liu, Y.; Tang, W.; Nie, S.; Hassouna, A.; et al. Fucoidan Extracted From Sporophyll of Undaria pinnatifida Grown in Weihai, China – Chemical Composition and Comparison of Antioxidant Activity of Different Molecular Weight Fractions. Front. Nutr. 2021, 8, 636930. [Google Scholar] [CrossRef]
  13. Cheong, K.-L.; Chen, W.; Wang, M.; Zhong, S.; Veeraperumal, S. Therapeutic Prospects of Undaria pinnatifida Polysaccharides: Extraction, Purification, and Functional Activity. Mar. Drugs 2025, 23, 163. [Google Scholar] [CrossRef]
  14. Mak, W.; Hamid, N.; Liu, T.; Lu, J.; White, W.L. Fucoidan from New Zealand Undaria pinnatifida: Monthly variations and determination of antioxidant activities. Carbohydr. Polym. 2013, 95, 606–614. [Google Scholar] [CrossRef] [PubMed]
  15. Wang, L.; Oliveira, C.; Li, Q.; Ferreira, A.S.; Nunes, C.; Coimbra, M.A.; Reis, R.L.; Martins, A.; Wang, C.; Silva, T.H.; et al. Fucoidan from Fucus vesiculosus Inhibits Inflammatory Response, Both In Vitro and In Vivo. Mar. Drugs 2023, 21, 302. [Google Scholar] [CrossRef]
  16. 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] [PubMed]
  17. Zhang, T.; Wu, S.; Ai, C.; Wen, C.; Liu, Z.; Wang, L.; Jiang, L.; Shen, P.; Zhang, G.; Song, S. Galactofucan from Laminaria japonica is not degraded by the human digestive system but inhibits pancreatic lipase and modifies the intestinal microbiota. Int. J. Biol. Macromol. 2021, 166, 611–620. [Google Scholar] [CrossRef] [PubMed]
  18. Chadwick, M.; Carvalho, L.G.; Vanegas, C.; Dimartino, S. A Comparative Review of Alternative Fucoidan Extraction Techniques from Seaweed. Mar. Drugs 2025, 23, 27. [Google Scholar] [CrossRef] [PubMed]
  19. Saetan, U.; Nontasak, P.; Palasin, K.; Saelim, H.; Wonglapsuwan, M.; Mayakun, J.; Pongparadon, S.; Chotigeat, W. Potential health benefits of fucoidan from the brown seaweeds Sargassum plagiophyllum and Sargassum polycystum. J. Appl. Phycol. 2021, 33, 3357–3364. [Google Scholar] [CrossRef]
  20. Zayed, A.; Cao, H.T.T.; Trang, V.T.D.; Ulber, R. Structural tailoring of fucoidan backbones for maximizing their benefits: Enzymatic, chemical, and physical strategies. J. Appl. Phycol. 2023, 35, 2445–2462. [Google Scholar] [CrossRef]
  21. Hu, S.; Chen, S.; Zhu, H.; Du, M.; Jiang, W.; Liu, Y.; Gao, X.; Su, L.; Xu, Y. Low Molecular Weight, 4-O-Sulfation, and Sulfation at Meta-Fucose Positively Promote the Activities of Sea Cucumber Fucoidans on Improving Insulin Resistance in HFD-Fed Mice. Mar. Drugs 2021, 20, 37. [Google Scholar] [CrossRef] [PubMed]
  22. Yang, C.; Dwan, C.; Wimmer, B.C.; Ronci, M.; Wilson, R.; Johnson, L.; Caruso, V. Neuroprotective and Anti-Inflammatory Activity of Undaria pinnatifida Fucoidan In Vivo—A Proteomic Investigation. Mar. Drugs 2025, 23, 189. [Google Scholar] [CrossRef] [PubMed]
  23. Ebrahimi, B.; Lessa, R.C.; Baroutian, S.; Zhou, Q.; Chen, X.; Lu, J. Antioxidant and immunomodulatory activity of Perna canaliculus oil extract in combination with low molecular weight fucoidan extracted from Undaria pinnatifida. Futur. Foods 2025, 11, 100652. [Google Scholar] [CrossRef]
  24. Li, Q.; Wang, X.; Wan, Y.; Hu, X.; Liu, J.; Wang, J. In vivo immunomodulatory activity of fucoidan from brown alga Undaria pinnatifida in sarcoma 180-bearing mice. J. Funct. Foods 2023, 103, 105486. [Google Scholar] [CrossRef]
  25. 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.; et al. 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]
  26. Giuliani, M.; Uboldi, C.; Dellatorre, F.G.; Latour, E.; Ponce, N.M.A.; Stortz, C.A.; Lassalle, V.L.; Ayala-Peña, V.B. Undaria pinnatifida fucoidan extract inhibits activation of the NF-κB signaling pathway by herpes simplex virus type 1 and prevents amyloid-β peptide synthesis in retinal pigment epithelium cells. Arch. Virol. 2025, 170, 27. [Google Scholar] [CrossRef] [PubMed]
  27. Ahmad, T.; Eapen, M.S.; Ishaq, M.; Park, A.Y.; Karpiniec, S.S.; Stringer, D.N.; Sohal, S.S.; Fitton, J.H.; Guven, N.; Caruso, V.; et al. Anti-Inflammatory Activity of Fucoidan Extracts In Vitro. Mar. Drugs 2021, 19, 702. [Google Scholar] [CrossRef]
  28. Kim, K.J.; Yoon, K.Y.; Lee, B.Y. Low molecular weight fucoidan from the sporophyll of Undaria pinnatifida suppresses inflammation by promoting the inhibition of mitogen-activated protein kinases and oxidative stress in RAW264.7 cells. Fitoterapia 2012, 83, 1628–1635. [Google Scholar] [CrossRef]
  29. Flórez-Fernández, N.; Vaamonde-García, C.; Torres, M.D.; Buján, M.; Muíños, A.; Muiños, A.; Lamas-Vázquez, M.J.; Meijide-Faílde, R.; Blanco, F.J.; Domínguez, H. Relevance of the Extraction Stage on the Anti-Inflammatory Action of Fucoidans. Pharmaceutics 2023, 15, 808. [Google Scholar] [CrossRef]
  30. Vaamonde-García, C.; Flórez-Fernández, N.; Torres, M.D.; Lamas-Vázquez, M.J.; Blanco, F.J.; Domínguez, H.; Meijide-Faílde, R. Study of fucoidans as natural biomolecules for therapeutical applications in osteoarthritis. Carbohydr. Polym. 2021, 258, 117692. [Google Scholar] [CrossRef]
  31. Lim, J.M.; Yoo, H.J.; Lee, K.W. High Molecular Weight Fucoidan Restores Intestinal Integrity by Regulating Inflammation and Tight Junction Loss Induced by Methylglyoxal-Derived Hydroimidazolone-1. Mar. Drugs 2022, 20, 580. [Google Scholar] [CrossRef] [PubMed]
  32. Li, P.; Chang, M. Roles of PRR-Mediated Signaling Pathways in the Regulation of Oxidative Stress and Inflammatory Diseases. Int. J. Mol. Sci. 2021, 22, 7688. [Google Scholar] [CrossRef] [PubMed]
  33. Zheng, W.; Jia, J.; Zhang, C.; Zhang, P.; Song, S.; Ai, C. Undaria pinnatifida fucoidan ameliorates dietary fiber deficiency-induced inflammation and lipid abnormality by modulating mucosal microbiota and protecting intestinal barrier integrity. Int. J. Biol. Macromol. 2023, 247, 125724. [Google Scholar] [CrossRef]
  34. Phull, A.-R.; Majid, M.; Haq, I.-U.; Khan, M.R.; Kim, S.J. In vitro and in vivo evaluation of anti-arthritic, antioxidant efficacy of fucoidan from Undaria pinnatifida (Harvey) Suringar. Int. J. Biol. Macromol. 2017, 97, 468–480. [Google Scholar] [CrossRef] [PubMed]
  35. Lee, J.-H.; Kim, J.-H.; Kim, S.-M.; Kim, J.-Y.; Kim, J.-H.; Eom, S.-J.; Kang, M.-C.; Song, K.-M. The Antioxidant Activity of Undaria pinnatifida Sporophyll Extract Obtained Using Ultrasonication: A Focus on Crude Polysaccharide Extraction Using Ethanol Precipitation. Antioxidants 2023, 12, 1904. [Google Scholar] [CrossRef]
  36. Zhao, Y.; Zheng, Y.; Wang, J.; Ma, S.; Yu, Y.; White, W.L.; Yang, S.; Yang, F.; Lu, J. Fucoidan Extracted from Undaria pinnatifida: Source for Nutraceuticals/Functional Foods. Mar. Drugs 2018, 16, 321. [Google Scholar] [CrossRef]
  37. Silva, M.M.C.L.; Lisboa, L.d.S.; 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]
  38. Kim, M.E.; Lee, J.S. Advances in the Regulation of Inflammatory Mediators in Nitric Oxide Synthase: Implications for Disease Modulation and Therapeutic Approaches. Int. J. Mol. Sci. 2025, 26, 1204. [Google Scholar] [CrossRef]
  39. Yang, H.; Xuefeng, Y.; Shandong, W.; Jianhua, X. COX-2 in liver fibrosis. Clin. Chim. Acta 2020, 506, 196–203. [Google Scholar] [CrossRef]
  40. Song, K.-M.; Ha, S.J.; Lee, J.-E.; Kim, S.-H.; Kim, Y.H.; Kim, Y.; Hong, S.P.; Jung, S.K.; Lee, N.H. High yield ultrasonication extraction method for Undaria pinnatifida sporophyll and its anti-inflammatory properties associated with AP-1 pathway suppression. LWT 2015, 64, 1315–1322. [Google Scholar] [CrossRef]
  41. Zhang, J.M.; An, J. Cytokines, inflammation, and pain. Int. Anesthesiol. Clin. 2007, 45, 27–37. [Google Scholar] [CrossRef]
  42. Chen, X.-W.; Long, J.; Zhang, Q.; Huang, L.-H.; Sun, X.-Y. Sulfated Undaria pinnatifida polysaccharides inhibit kidney stone formation through crystalline modulation and relieving cellular oxidative damage and inflammation. Biomater. Sci. 2025, 13, 1512–1528. [Google Scholar] [CrossRef]
  43. 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]
  44. Vaamonde-García, C.; Capelo-Mera, E.; Flórez-Fernández, N.; Torres, M.D.; Rivas-Murias, B.; Mejide-Faílde, R.; Blanco, F.J.; Domínguez, H. In Vitro Study of the Therapeutic Potential of Brown Crude Fucoidans in Osteoarthritis Treatment. Int. J. Mol. Sci. 2022, 23, 14236. [Google Scholar] [CrossRef]
  45. Wimmer, B.C.; Dwan, C.; De Medts, J.; Duysburgh, C.; Rotsaert, C.; Marzorati, M. Undaria pinnatifida Fucoidan Enhances Gut Microbiome, Butyrate Production, and Exerts Anti-Inflammatory Effects in an In Vitro Short-Term SHIME® Coupled to a Caco-2/THP-1 Co-Culture Model. Mar. Drugs 2025, 23, 242. [Google Scholar] [CrossRef]
  46. Yang, J.-H. Topical Application of Fucoidan Improves Atopic Dermatitis Symptoms in NC/Nga Mice. Phytother. Res. 2012, 26, 1898–1903. [Google Scholar] [CrossRef]
  47. Li, X.; Li, J.; Li, Z.; Sang, Y.; Niu, Y.; Zhang, Q.; Ding, H.; Yin, S. Fucoidan from Undaria pinnatifida prevents vascular dysfunction through PI3K/Akt/eNOS-dependent mechanisms in the l-NAME-induced hypertensive rat model. Food Funct. 2016, 7, 2398–2408. [Google Scholar] [CrossRef]
  48. Herath, K.H.I.N.M.; Kim, H.J.; Kim, A.; Sook, C.E.; Lee, B.-Y.; Jee, Y. The Role of Fucoidans Isolated from the Sporophylls of Undaria pinnatifida against Particulate-Matter-Induced Allergic Airway Inflammation: Evidence of the Attenuation of Oxidative Stress and Inflammatory Responses. Molecules 2020, 25, 2869. [Google Scholar] [CrossRef] [PubMed]
  49. Shanmugasundaram, D.; Dwan, C.; Wimmer, B.C.; Srivastava, S. Fucoidan Ameliorates Testosterone-Induced Benign Prostatic Hyperplasia (BPH) in Rats. Res. Rep. Urol. 2024, 16, 283–297. [Google Scholar] [CrossRef] [PubMed]
  50. Jiang, P.; Zheng, W.; Sun, X.; Jiang, G.; Wu, S.; Xu, Y.; Song, S.; Ai, C. Sulfated polysaccharides from Undaria pinnatifida improved high fat diet-induced metabolic syndrome, gut microbiota dysbiosis and inflammation in BALB/c mice. Int. J. Biol. Macromol. 2021, 167, 1587–1597. [Google Scholar] [CrossRef] [PubMed]
  51. Kang, K.S.; Kim, I.D.; Kwon, R.H.; Ha, B.J. Undaria pinnatifida fucoidan extract protects against CCl4-induced oxidative stress. Biotechnol. Bioprocess Eng. 2008, 13, 168–173. [Google Scholar] [CrossRef]
  52. Oh, J.-Y.; Kim, E.-A.; Kang, S.I.; Yang, H.-W.; Ryu, B.; Wang, L.; Lee, J.-S.; Jeon, Y.-J. Protective Effects of Fucoidan Isolated from Celluclast-Assisted Extract of Undaria pinnatifida Sporophylls against AAPH-Induced Oxidative Stress In Vitro and In Vivo Zebrafish Model. Molecules 2020, 25, 2361. [Google Scholar] [CrossRef]
  53. Buttó, L.F.; Haller, D. Dysbiosis in intestinal inflammation: Cause or consequence. Int. J. Med. Microbiol. 2016, 306, 302–309. [Google Scholar] [CrossRef]
  54. Zeng, M.Y.; Inohara, N.; Nuñez, G. Mechanisms of inflammation-driven bacterial dysbiosis in the gut. Mucosal Immunol. 2017, 10, 18–26. [Google Scholar] [CrossRef] [PubMed]
  55. Nagao-Kitamoto, H.; Kamada, N. Host-microbial Cross-talk in Inflammatory Bowel Disease. Immune Netw. 2017, 17, 1–12. [Google Scholar] [CrossRef]
  56. Liyanage, N.M.; Nagahawatta, D.P.; Jayawardena, T.U.; Jeon, Y.-J. The Role of Seaweed Polysaccharides in Gastrointestinal Health: Protective Effect against Inflammatory Bowel Disease. Life 2023, 13, 1026. [Google Scholar] [CrossRef]
  57. Liu, M.; Ma, L.; Chen, Q.; Zhang, P.; Chen, C.; Jia, L.; Li, H. Fucoidan alleviates dyslipidemia and modulates gut microbiota in high-fat diet-induced mice. J. Funct. Foods 2018, 48, 220–227. [Google Scholar] [CrossRef]
  58. Li, L.; Wang, Y.; Yuan, J.; Liu, Z.; Ye, C.; Qin, S. Undaria pinnatifida improves obesity-related outcomes in association with gut microbiota and metabolomics modulation in high-fat diet-fed mice. Appl. Microbiol. Biotechnol. 2020, 104, 10217–10231. [Google Scholar] [CrossRef]
  59. Zhang, P.; Jia, J.; Jiang, P.; Zheng, W.; Li, X.; Song, S.; Ai, C. Polysaccharides from edible brown seaweed Undaria pinnatifida are effective against high-fat diet-induced obesity in mice through the modulation of intestinal microecology. Food Funct. 2022, 13, 2581–2593. [Google Scholar] [CrossRef] [PubMed]
  60. Yang, C.; Dwan, C.; Wimmer, B.C.; Wilson, R.; Johnson, L.; Caruso, V. Fucoidan from Undaria pinnatifida Enhances Exercise Performance and Increases the Abundance of Beneficial Gut Bacteria in Mice. Mar. Drugs 2024, 22, 485. [Google Scholar] [CrossRef]
  61. Zheng, W.; Jia, J.; Tang, S.; Song, S.; Ai, C. Undaria pinnatifida fucoidan contributes to anti-inflammation activity of Bacteroides in fiber-deficient mice via modulation of gut microbiota and protection of intestinal barrier integrity. Int. J. Biol. Macromol. 2023, 252, 126256. [Google Scholar] [CrossRef]
  62. Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. What Is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14. [Google Scholar] [CrossRef]
  63. Spychala, M.S.; Venna, V.R.; Jandzinski, M.; Doran, S.J.; Durgan, D.J.; Ganesh, B.P.; Ajami, N.J.; Putluri, N.; Graf, J.; Bryan, R.M.; et al. Age-related changes in the gut microbiota influence systemic inflammation and stroke outcome. Ann. Neurol. 2018, 84, 23–36. [Google Scholar] [CrossRef]
  64. Stojanov, S.; Berlec, A.; Štrukelj, B. The Influence of Probiotics on the Firmicutes/Bacteroidetes Ratio in the Treatment of Obesity and Inflammatory Bowel disease. Microorganisms 2020, 8, 1715. [Google Scholar] [CrossRef] [PubMed]
  65. An, J.; Kwon, H.; Kim, Y.J. The Firmicutes/Bacteroidetes Ratio as a Risk Factor of Breast Cancer. J. Clin. Med. 2023, 12, 2216. [Google Scholar] [CrossRef]
  66. Yin, Y.; Yang, T.; Tian, Z.; Shi, C.; Yan, C.; Li, H.; Du, Y.; Li, G. Progress in the investigation of the Firmicutes/Bacteroidetes ratio as a potential pathogenic factor in ulcerative colitis. J. Med Microbiol. 2025, 74, 001966. [Google Scholar] [CrossRef] [PubMed]
  67. Ren, P.; Liu, M.; Wei, B.; Tang, Q.; Wang, Y.; Xue, C. Fucoidan exerts antitumor effects by regulating gut microbiota and tryptophan metabolism. Int. J. Biol. Macromol. 2025, 300, 140334. [Google Scholar] [CrossRef]
  68. Park, E.-J.; Kim, J.-Y.; Jaiswal, V.; Park, H.-S.; Ki, D.-B.; Lee, Y.-S.; Lee, H.-J. High-molecular-weight Fucoidan exerts an immune-enhancing effect in RAW 264.7 cells and cyclophosphamide-induced immunosuppression rat by altering the gut microbiome. Int. Immunopharmacol. 2024, 139, 112677. [Google Scholar] [CrossRef]
  69. He, J.; Zhang, P.; Shen, L.; Niu, L.; Tan, Y.; Chen, L.; Zhao, Y.; Bai, L.; Hao, X.; Li, X.; et al. Short-Chain Fatty Acids and Their Association with Signalling Pathways in Inflammation, Glucose and Lipid Metabolism. Int. J. Mol. Sci. 2020, 21, 6356. [Google Scholar] [CrossRef]
  70. Vinolo, M.A.; Rodrigues, H.G.; Nachbar, R.T.; Curi, R. Regulation of Inflammation by Short Chain Fatty Acids. Nutrients 2011, 3, 858–876. [Google Scholar] [CrossRef] [PubMed]
  71. Yu, Z.N.; Fan, Y.J.; Van Nguyen, T.; Piao, C.H.; Lee, B.-H.; Lee, S.-Y.; Shin, H.S.; Kim, T.-G.; Song, C.H.; Chai, O.H. Undaria pinnatifida extract attenuates combined allergic rhinitis and asthma syndrome by the modulation of epithelial cell dysfunction and oxidative stress. Acta Biochim. Biophys. Sin. 2024, 57, 792–804. [Google Scholar] [CrossRef]
  72. Men, Q.; Zhang, P.; Zheng, W.; Song, S.; Ai, C. Fucoidan alleviates Salmonella-induced inflammation and mortality by modulating gut microbiota and metabolites, protecting intestinal barrier, and inhibiting NF-κB pathway. Food Biosci. 2023, 56. [Google Scholar] [CrossRef]
  73. Zaharudin, N.; Tullin, M.; Pekmez, C.T.; Sloth, J.J.; Rasmussen, R.R.; Dragsted, L.O. Effects of brown seaweeds on postprandial glucose, insulin and appetite in humans – A randomized, 3-way, blinded, cross-over meal study. Clin. Nutr. 2021, 40, 830–838. [Google Scholar] [CrossRef]
  74. Yoshinaga, K.; Mitamura, R. Effects of Undaria pinnatifida (Wakame) on Postprandial Glycemia and Insulin Levels in Humans: A Randomized Crossover Trial. Plant Foods Hum. Nutr. 2019, 74, 461–467. [Google Scholar] [CrossRef] [PubMed]
  75. Teas, J.; Baldeón, M.E.; Chiriboga, D.E.; Davis, J.R.; Sarriés, A.J.; Braverman, L.E. Could dietary seaweed reverse the metabolic syndrome? Asia Pac. J. Clin. Nutr. 2009, 18, 145–154. [Google Scholar]
  76. Izaola, O.; Primo, D.; Rico Bargues, D.; Belen Martin-Diana, A.; Martinez Villaluenga, C.; Miranda, J.; de Luis, D.A. Effects of a snack enriched with carob and Undaria pinnatifida (wakame) on metabolic parameters in a double blind, randomized clinical trial in obese patients. Nutr. Hosp. 2020, 37, 465–473. [Google Scholar]
  77. Cooper, R.; Dragar, C.; Elliot, K.; Fitton, J.; Godwin, J.; Thompson, K. GFS, a preparation of Tasmanian Undaria pinnatifida is associated with healing and inhibition of reactivation of Herpes. BMC Complement. Altern. Med. 2002, 2, 11. [Google Scholar] [CrossRef]
  78. Gueven, N.; Spring, K.J.; Holmes, S.; Ahuja, K.; Eri, R.; Park, A.Y.; Fitton, J.H. Micro RNA Expression after Ingestion of Fucoidan; A Clinical Study. Mar. Drugs 2020, 18, 143. [Google Scholar] [CrossRef]
  79. Cox, A.J.; Cripps, A.W.; Taylor, P.A.; Fitton, J.H.; West, N.P. Fucoidan Supplementation Restores Fecal Lysozyme Concentrations in High-Performance Athletes: A Pilot Study. Mar. Drugs 2020, 18, 412. [Google Scholar] [CrossRef]
  80. Olas, K.; Butterweck, H.; Teschner, W.; Schwarz, H.P.; Reipert, B. Immunomodulatory properties of human serum immunoglobulin A: Anti-inflammatory and pro-inflammatory activities in human monocytes and peripheral blood mononuclear cells. Clin. Exp. Immunol. 2005, 140, 478–490. [Google Scholar] [CrossRef] [PubMed]
  81. Cox, A.J.; Speer, H.; Radcliffe, C.R.; Masocha, K.; Ramsey, R.; West, N.P.; Pyne, D.B. Immunomodulatory effects of fucoidan in recreationally active adult males undertaking 3-weeks of intensified training. J. Sports Sci. 2023, 41, 1875–1882. [Google Scholar] [CrossRef]
  82. McFadden, B.A.; Vincenty, C.S.; Chandler, A.J.; Cintineo, H.P.; Lints, B.S.; Mastrofini, G.F.; Arent, S.M. Effects of fucoidan supplementation on inflammatory and immune response after high-intensity exercise. J. Int. Soc. Sports Nutr. 2023, 20, 2224751. [Google Scholar] [CrossRef]
  83. Tay, A.; Jiang, Y.; Signal, N.; O’BRien, D.; Chen, J.; Murphy, R.; Lu, J. Combining mussel with fucoidan as a supplement for joint pain and prediabetes: Study protocol for a randomized, double-blinded, placebo-controlled trial. Front. Nutr. 2022, 9, 1000510. [Google Scholar] [CrossRef]
  84. Alghazwi, M.; Smid, S.; Karpiniec, S.; Zhang, W. Comparative study on neuroprotective activities of fucoidans from Fucus vesiculosus and Undaria pinnatifida. Int. J. Biol. Macromol. 2019, 122, 255–264. [Google Scholar] [CrossRef] [PubMed]
  85. Wei, H.; Gao, Z.; Zheng, L.; Zhang, C.; Liu, Z.; Yang, Y.; Teng, H.; Hou, L.; Yin, Y.; Zou, X. Protective Effects of Fucoidan on Aβ25–35 and d-Gal-Induced Neurotoxicity in PC12 Cells and d-Gal-Induced Cognitive Dysfunction in Mice. Mar. Drugs 2017, 15, 77. [Google Scholar] [CrossRef] [PubMed]
  86. Kim, J.; Moon, I.S.; Goo, T.-W.; Moon, S.-S.; Seo, M. Algae Undaria pinnatifida Protects Hypothalamic Neurons against Endoplasmic Reticulum Stress through Akt/mTOR Signaling. Molecules 2015, 20, 20998–21009. [Google Scholar] [CrossRef] [PubMed]
  87. Singh, R.; Kaur, N.; Choubey, V.; Dhingra, N.; Kaur, T. Endoplasmic reticulum stress and its role in various neurodegenerative diseases. Brain Res. 2023, 1826, 148742. [Google Scholar] [CrossRef] [PubMed]
  88. Chen, M.; Wang, J.; Zhang, P.; Jiang, Z.; Chen, S.; Liang, S.; Ma, T.; Liao, H.; Tan, W.; Niu, C.; et al. Low molecular weight fucoidan induces M2 macrophage polarization to attenuate inflammation through activation of the AMPK/mTOR autophagy pathway. Eur. J. Pharmacol. 2024, 986, 177134. [Google Scholar] [CrossRef]
  89. Wu, Y.-Q.; Xiong, J.; He, Z.-L.; Yuan, Y.; Wang, B.-N.; Xu, J.-Y.; Wu, M.; Zhang, S.-S.; Cai, S.-F.; Zhao, J.-X.; et al. Metformin promotes microglial cells to facilitate myelin debris clearance and accelerate nerve repairment after spinal cord injury. Acta Pharmacol. Sin. 2021, 43, 1360–1371. [Google Scholar] [CrossRef]
  90. Jhamandas, J.H.; Wie, M.B.; Harris, K.; MacTavish, D.; Kar, S. Fucoidan inhibits cellular and neurotoxic effects of β-amyloid (Aβ) in rat cholinergic basal forebrain neurons. Eur. J. Neurosci. 2005, 21, 2649–2659. [Google Scholar] [CrossRef]
  91. Mohibbullah, M.; Hannan, M.A.; Choi, J.-Y.; Bhuiyan, M.M.H.; Hong, Y.-K.; Choi, J.-S.; Choi, I.S.; Moon, I.S. The Edible Marine Alga Gracilariopsis chorda Alleviates Hypoxia/Reoxygenation-Induced Oxidative Stress in Cultured Hippocampal Neurons. J. Med. Food 2015, 18, 960–971. [Google Scholar] [CrossRef] [PubMed]
  92. Mohibbullah, M.; Haque, M.N.; Khan, M.N.A.; Park, I.-S.; Moon, I.S.; Hong, Y.-K. Neuroprotective effects of fucoxanthin and its derivative fucoxanthinol from the phaeophyte Undaria pinnatifida attenuate oxidative stress in hippocampal neurons. J. Appl. Phycol. 2018, 30, 3243–3252. [Google Scholar] [CrossRef]
  93. Phuna, Z.X.; Madhavan, P. A reappraisal on amyloid cascade hypothesis: The role of chronic infection in Alzheimer’s disease. Int. J. Neurosci. 2023, 133, 1071–1089. [Google Scholar] [CrossRef] [PubMed]
  94. Hu, C.; Zhang, G.; Zhao, Y.T. Fucoidan attenuates the existing allodynia and hyperalgesia in a rat model of neuropathic pain. Neurosci. Lett. 2014, 571, 66–71. [Google Scholar] [CrossRef]
  95. Che, N.; Ma, Y.; Xin, Y. Protective Role of Fucoidan in Cerebral Ischemia-Reperfusion Injury through Inhibition of MAPK Signaling Pathway. Biomol. Ther. 2017, 25, 272–278. [Google Scholar] [CrossRef]
  96. Wang, T.; Zhu, M.; He, Z.Z. Low-Molecular-Weight Fucoidan Attenuates Mitochondrial Dysfunction and Improves Neurological Outcome After Traumatic Brain Injury in Aged Mice: Involvement of Sirt3. Cell. Mol. Neurobiol. 2016, 36, 1257–1268. [Google Scholar] [CrossRef]
  97. Wang, X.; Yi, K.; Zhao, Y. Fucoidan inhibits amyloid-β-induced toxicity in transgenic Caenorhabditis elegans by reducing the accumulation of amyloid-β and decreasing the production of reactive oxygen species. Food Funct. 2018, 9, 552–560. [Google Scholar] [CrossRef]
  98. U.S. Food and Drug Administration. GRAS Notice No. 565: Fucoidan from Undaria pinnatifida; U.S. Food and Drug Administration: Silver Spring, MD, USA, 2015.
  99. U.S. Food and Drug Administration. GRAS Notice No. 661: Fucoidan from Fucus vesiculosus; U.S. Food and Drug Administration: Silver Spring, MD, USA, 2017.
  100. Kim, K.-J.; Lee, O.-H.; Lee, H.-H.; Lee, B.-Y. A 4-week repeated oral dose toxicity study of fucoidan from the Sporophyll of Undaria pinnatifida in Sprague–Dawley rats. Toxicology 2010, 267, 154–158. [Google Scholar] [CrossRef]
  101. Li, N.; Zhang, Q.; Song, J. Toxicological evaluation of fucoidan extracted from Laminaria japonica in Wistar rats. Food Chem. Toxicol. 2005, 43, 421–426. [Google Scholar] [CrossRef]
  102. Kim, K.-J.; Lee, O.-H.; Lee, B.-Y. Genotoxicity studies on fucoidan from Sporophyll of Undaria pinnatifida. Food Chem. Toxicol. 2010, 48, 1101–1104. [Google Scholar] [CrossRef] [PubMed]
  103. Irhimeh, M.R.; Fitton, J.H.; Lowenthal, R.M. Pilot clinical study to evaluate the anticoagulant activity of fucoidan. Blood Coagul. Fibrinolysis 2009, 20, 607–610. [Google Scholar] [CrossRef]
  104. 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]
  105. Dockal, M.; Till, S.; Knappe, S.; Ehrlich, H.J.; Scheiflinger, F. Anticoagulant Activity and Mechanism of Non-Anticoagulant Sulfated Polysaccharides. Blood 2011, 118, 1208. [Google Scholar] [CrossRef]
  106. Mathew, L.; Burney, M.; Gaikwad, A.; Nyshadham, P.; Nugent, E.K.; Gonzalez, A.; Smith, J.A. Preclinical Evaluation of Safety of Fucoidan Extracts From Undaria pinnatifida and Fucus vesiculosus for Use in Cancer Treatment. Integr. Cancer Ther. 2016, 16, 572–584. [Google Scholar] [CrossRef]
  107. Nagamine, T.; Nakazato, K.; Tomioka, S.; Iha, M.; Nakajima, K. Intestinal Absorption of Fucoidan Extracted from the Brown Seaweed, Cladosiphon okamuranus. Mar. Drugs 2014, 13, 48–64. [Google Scholar] [CrossRef] [PubMed]
  108. Zhang, E.; Chu, F.; Xu, L.; Liang, H.; Song, S.; Ji, A. Use of fluorescein isothiocyanate isomer I to study the mechanism of intestinal absorption of fucoidan sulfate in vivo and in vitro. Biopharm. Drug Dispos. 2018, 39, 298–307. [Google Scholar] [CrossRef] [PubMed]
  109. Pozharitskaya, O.N.; Shikov, A.N.; Faustova, N.M.; Obluchinskaya, E.D.; Kosman, V.M.; Vuorela, H.; Makarov, V.G. Pharmacokinetic and Tissue Distribution of Fucoidan from Fucus vesiculosus after Oral Administration to Rats. Mar. Drugs 2018, 16, 132. [Google Scholar] [CrossRef]
  110. Tan, J.; Song, Y.; Wang, J.; Wu, N.; Yue, Y.; Zhang, Q. Pharmacokinetics of fucoidan and low molecular weight fucoidan from Saccharina japonica after oral administration to mice. J. Oceanol. Limnol. 2023, 41, 1900–1909. [Google Scholar] [CrossRef]
  111. Bai, X.; Zhang, E.; Hu, B.; Liang, H.; Song, S.; Ji, A. Study on Absorption Mechanism and Tissue Distribution of Fucoidan. Molecules 2020, 25, 1087. [Google Scholar] [CrossRef]
  112. Warttinger, U.; Giese, C.; Harenberg, J.; Krämer, R. Direct quantification of brown algae-derived fucoidans in human plasma by a fluorescent probe assay. arXiv 2016, arXiv:1608.00108. [Google Scholar] [CrossRef]
  113. Irhimeh, M.R.; Fitton, J.H.; Lowenthal, R.M.; 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] [PubMed]
  114. Tokita, Y.; Nakajima, K.; Mochida, H.; Iha, M.; Nagamine, T. Development of a Fucoidan-Specific Antibody and Measurement of Fucoidan in Serum and Urine by Sandwich ELISA. Biosci. Biotechnol. Biochem. 2010, 74, 350–357. [Google Scholar] [CrossRef]
  115. Chen, Y.; Li, X.; Gan, X.; Qi, J.; Che, B.; Tai, M.; Gao, S.; Zhao, W.; Xu, N.; Hu, Z. Fucoidan from Undaria pinnatifida Ameliorates Epidermal Barrier Disruption via Keratinocyte Differentiation and CaSR Level Regulation. Mar. Drugs 2019, 17, 660. [Google Scholar] [CrossRef]
  116. 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]
  117. 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]
  118. Fitton, J.H.; Dell’Acqua, G.; Gardiner, V.-A.; Karpiniec, S.S.; Stringer, D.N.; Davis, E. Topical Benefits of Two Fucoidan-Rich Extracts from Marine Macroalgae. Cosmetics 2015, 2, 66–81. [Google Scholar] [CrossRef]
  119. Chen, B.-R.; Hsu, K.-T.; Hsu, W.-H.; Lee, B.-H.; Li, T.-L.; Chan, Y.-L.; Wu, C.-J. Immunomodulation and mechanisms of fucoidan from Cladosiphon okamuranus ameliorates atopic dermatitis symptoms. Int. J. Biol. Macromol. 2021, 189, 537–543. [Google Scholar] [CrossRef] [PubMed]
  120. Obluchinskaya, E.D.; Pozharitskaya, O.N.; Flisyuk, E.V.; Shikov, A.N. Formulation, Optimization and In Vivo Evaluation of Fucoidan-Based Cream with Anti-Inflammatory Properties. Mar. Drugs 2021, 19, 643. [Google Scholar] [CrossRef] [PubMed]
  121. Bachelet, L.; Bertholon, I.; Lavigne, D.; Vassy, R.; Jandrot-Perrus, M.; Chaubet, F.; Letourneur, D. Affinity of low molecular weight fucoidan for P-selectin triggers its binding to activated human platelets. Biochim. Biophys. Acta Gen. Subj. 2009, 1790, 141–146. [Google Scholar] [CrossRef]
  122. Zayed, A.; Al-Saedi, D.A.; Mensah, E.O.; Kanwugu, O.N.; Adadi, P.; Ulber, R. Fucoidan’s Molecular Targets: A Comprehensive Review of Its Unique and Multiple Targets Accounting for Promising Bioactivities Supported by In Silico Studies. Mar. Drugs 2023, 22, 29. [Google Scholar] [CrossRef]
  123. 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.; et al. 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] [PubMed]
  124. Hemmingson, J.A.; Falshaw, R.; Furneaux, R.H.; Thompson, K. Structure and Antiviral Activity of the Galactofucan Sulfates Extracted from Undaria Pinnatifida (Phaeophyta). J. Appl. Phycol. 2006, 18, 185–193. [Google Scholar] [CrossRef]
  125. Lee, J.; Lee, S.; Synytsya, A.; Capek, P.; Lee, C.W.; Choi, J.W.; Cho, S.; Kim, W.J.; Park, Y.I. Low Molecular Weight Mannogalactofucans Derived from Undaria pinnatifida Induce Apoptotic Death of Human Prostate Cancer Cells In Vitro and In Vivo. Mar. Biotechnol. 2018, 20, 813–828. [Google Scholar] [CrossRef]
  126. Zheng, W.; Tang, S.; Ren, X.; Song, S.; Ai, C. Fucoidan alleviated colitis aggravated by fiber deficiency through protecting the gut barrier, suppressing the MAPK/NF-κB pathway, and modulating gut microbiota and metabolites. Front. Nutr. 2025, 11, 1462584. [Google Scholar] [CrossRef] [PubMed]
  127. 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] [PubMed]
Figure 1. Anti-inflammatory activities of UPF in vitro. NF-κB: nuclear factor kappa B; MAPK: mitogen-activated protein kinase; TNF-α: tumour necrosis factor alpha; IL-6: interleukin-6; IL-1β: interleukin-1 beta; ROS: reactive oxygen species; MDA: malondialdehyde; SOD: superoxide dismutase; GSH: glutathione; IL-8: interleukin-8; MCP-1: monocyte chemoattractant protein-1; iNOS: inducible nitric oxide synthase; COX2: cyclooxygenase-2; UPF: Undaria pinnatifida fucoidan.
Figure 1. Anti-inflammatory activities of UPF in vitro. NF-κB: nuclear factor kappa B; MAPK: mitogen-activated protein kinase; TNF-α: tumour necrosis factor alpha; IL-6: interleukin-6; IL-1β: interleukin-1 beta; ROS: reactive oxygen species; MDA: malondialdehyde; SOD: superoxide dismutase; GSH: glutathione; IL-8: interleukin-8; MCP-1: monocyte chemoattractant protein-1; iNOS: inducible nitric oxide synthase; COX2: cyclooxygenase-2; UPF: Undaria pinnatifida fucoidan.
Marinedrugs 23 00350 g001
Figure 2. Neuroprotective effects of UPF in vitro (with the use of Biovisart.com.cn, accessed on 19 August 2025). NF-κB: nuclear factor kappa B; TNF-α: tumour necrosis factor alpha; IL-6: interleukin-6; IL-1β: interleukin-1 beta; ROS: reactive oxygen species; SOD: superoxide dismutase; GSH: glutathione; Aβ: amyloid-beta peptides; AMPK: adenosine monophosphate-activated protein kinase; mTOR: mechanistic target of rapamycin; UPF: Undaria pinnatifida fucoidan.
Figure 2. Neuroprotective effects of UPF in vitro (with the use of Biovisart.com.cn, accessed on 19 August 2025). NF-κB: nuclear factor kappa B; TNF-α: tumour necrosis factor alpha; IL-6: interleukin-6; IL-1β: interleukin-1 beta; ROS: reactive oxygen species; SOD: superoxide dismutase; GSH: glutathione; Aβ: amyloid-beta peptides; AMPK: adenosine monophosphate-activated protein kinase; mTOR: mechanistic target of rapamycin; UPF: Undaria pinnatifida fucoidan.
Marinedrugs 23 00350 g002
Table 1. In vitro anti-inflammatory activity of UPF.
Table 1. In vitro anti-inflammatory activity of UPF.
Cell LineModelCompoundTested
Concentration
Effective
Concentration
ActivityReference
Murine RAW264.7 cellsLPS
stimulation
Low molecular weight UPF1, 10 and 100 µg/mL1, 10 and 100 µg/mLReduced ROS, COX-2 and iNOS; suppressed phosphorylation of p38, ERK1/2, and JNK; and downregulated TNF-α, IL-6, and IL-1β [28]
UPF12.5, 25, and 50 µg/mL50 µg/mLInhibited COX-2 and iNOS, and attenuated production of NO and PGE2 [40]
Human osteoarthritic chondrocytesIL-1β-
induced
inflammation
UPF5, 30, and 100 µg/mL5 µg/mLInhibited NF-κB activation; and reduced production of NO, PGE2, and IL-6 [30]
UPF1, 5, and 30 µg/mL5 µg/mLDownregulated IL-6 and IL-8 (CXCL8); upregulated Nrf-2, HO-1, and SOD-2. [44]
THP-1 cells and PBMCsLPS-induced inflammationUPF10, 50, 100, and 200 µg/mL10, 50, and 100 µg/mLReduced expression of TNF-α, IL-1β, and IL-6 [27]
Caco-2-Nint cellsSARS-CoV-2 infectionUPF0–1000 µg/mL200 µg/mLReduced IL-6, IFN-α, IFN-γ, and TNF-α [25]
Caco-2 cellsMG-H1-induced inflammationHigh molecular weight UPF0–1000 µg/mL500 µg/mLInhibited NF-κB signalling; downregulated IL-6 and TNF-α; and suppressed COX-2 and iNOS expression [31]
IEC-6 cellsH2O2-induced oxidative stressUPF10, 20, 50, and 100 µg/mL100 µg/mLDecreased levels of MDA, and increased CAT, T-SOD, and GSH [33]
Rabbit articular chondrocytesAntioxidant assaysUPF0–500 µg/mL2.5–100 µg/mLReduced COX-2; scavenged DPPH, nitric oxide and hydroxyl radicals; and exhibited iron chelating activity [34]
Vero cellsH2O2-induced viability reductionWater-ultrasonicated UPE50, 100, 200, and 400 µg/mL200 and 400 µg/mLSuppressed ROS production; decreased Bax; and increased Bcl-2 [35]
Human renal cellsOxalate crystal-induced inflammationsulphated Undaria pinnatifida polysaccharides50, 100, 150, 200, and 250 µg/mL200 µg/mLReduced ROS and MCP-1 production; increased SOD content; and decreased secretion of TNF-α and IL-1β [42]
3T3-L1 adipocytesAdipogenesisUPF1, 10, and 100 µg/mL100 µg/mLReduced production of ROS, SOD, and GPx; and downregulated expression of TNF-α, MCP-1 and PAI-1 [43]
Caco-2/THP-1 cocultureMicrobial stimulationUPF2.5 g/L2.5 g/LReduced secretion of IL-8 and MCP-1; decreased TNF-α; and increased IL-6 and IL-10 [45]
Human epidermal keratinocyte cell lineTNF-α or IFN-γ-induced inflammationUPF400 µg/mL400 µg/mLInhibited expression of TARC, MDC, and RANTES (CCL5) [46]
Table 2. In vivo anti-inflammatory activity of UPF.
Table 2. In vivo anti-inflammatory activity of UPF.
ModelAnimalCompoundDoseTreatmentTissueResultReference
HFD-induced obesityMale and female C57BL/6J miceUPF400 mg/kg/dayOral supplementation for 10 weeksSkeletal MuscleReduced TNF-α, IL-1β, and IL-6[22]
Small
Intestine
Reduced TNF-α, IL-1β, IL-6, NF-κB, Tjp1, GPR41, and GPR43
PlasmaReduced IL-1α and IL-6
Male and female C57BL/6J miceUPF400 mg/kg/dayOral supplementation for 10 weeksFaecesIncreased abundance of Bacteroidetes, Bacteroides/Prevotella, Akkermansia muciniphila, and Lactobacillus; and reduced F/B ratio[60]
Male BALB/c miceSulphated polysaccharides from Undaria pinnatifida150 and 300 mg/kg/dayOral gavage
for 10 weeks
SerumReduced levels of TC, TG, and LDL-c; increased HDL-c; suppressed FITC and LPS[59]
LiverIncreased expression of ABCG8, PPAR-γ, PGC-1α and CAT; reduced content of TC, TG, and MDA; and inhibited LPS production
ColonIncreased IL-10 expression; and reduced IL-6
FaecesIncreased abundance of Bacteroidetes, Bacteroidaceae, and Prevotellaceae; decreased Firmicutes, and Proteobacteria; increased levels acetate, propionate, and butyrate; and reduced F/B ratio
HFD-induced obesityMale BALB/c miceSulphated polysaccharides from Undaria pinnatifida100, 300, and 500 mg/kg/dayOral gavage
for 10 weeks
SerumReduced levels of TC, TG, LDL-c, LPS, and FITC; and increased HDL-c[50]
LiverSuppressed levels of LDL-c and MDA; and increased SOD
ColonDecreased TNF-α; and increased IL-10
FaecesIncreased Bacteroidetes abundance; reduced Firmicutes, Desulfovibrionales, and Clostridia; and increased levels acetate, propionate, and butyrate
Male C57BL/6J miceUndaria pinnatifida powder10% (w/w)Oral supplementation for 10 weeksFaecesIncreased acetic acid, propionic acid, and butyric acid; increased Bacteroidetes, Bacteroidaceae, and Bacteroides; and reduced Firmicutes, Lachnospiraceae, Streptococcaceae, Marinifilaceae[58]
HFD-induced dyslipidaemiaMale BALB/c miceUPF50 and 100 mg/kg/dayOral gavage
for 8 weeks
SerumSuppressed levels of TC and LDL-c[57]
LiverAttenuated levels of TG and CHO
FaecesIncreased Bacteroidetes; and reduced Firmicutes
l-NAME-induced hypertensionMale SD ratsUPF20 and 100 mg/kg/dayOral gavage
for 4 weeks
Thoracic aortaIncreased phosphorylation of eNOS and Akt; and decreased levels of iNOS and NO[47]
SerumDecreased levels of TNF-α and IL-1β
Particulate-matter-induced allergic airway inflammationFemale BALB/c miceUPF100 and 400 mg/kg/dayOral gavage
for 7 days
LungSuppressed MDA level; attenuated eosinophils, Gr-1+ cells, F4/80+ macrophage, and CD4+ T cell infiltration; and reduced IL-4, IL-17a, and IL-33[48]
TracheaAttenuated eosinophils, Gr-1+ cells, F4/80+ macrophage, and CD4+ T cell infiltration
SerumInhibited MDA level; attenuated total IgE; and reduced IL-4
Testosterone-induced BPHMale SD ratsUPF40 and 400 mg/kg/dayOral administration for 4 weeksProstateReduced levels of testosterone and DHT; increased Bax; and reduced Bcl-2 expression[49]
SerumDecreased levels of IL-1β, TNF-α, testosterone, DHT, and PSA
Fibre deficiency-induced intestinal inflammationMale BALB/c miceUPF100 and 400 mg/kg/dayOral supplementation for 4 weeksColonIncreased levels of occludin, ZO-1, and claudin-3; reduced expression of TNF-α, IL-6, and IL-1β; increased IL-10; suppressed MDA, MPO, and LPS; promoted CAT and T-SOD; and increased production of acetate, propionate, and butyrate[33]
Male BALB/c miceUPF300 mg/kg/dayOral gavage
for 4 weeks
ColonReduced expression of TNF-α and IL-1β; elevated occludin and IL-10; increased levels of T-SOD and CAT; and decreased COX-2, iNOS, and LPS[61]
FaecesIncreased abundance of Bacteroidetes and Bacteroidales; and decreased Firmicutes, Clostridiales, and Ruminococcaceae
SARS-CoV-2 infectionFemale Syrian hamstersUPF100 and 200 mg/kg/dayOral administration for 6 daysLungDownregulated ACE2, IL-6, and TNF-α[25]
ColonReduced levels of ACE2, IL-6, and TNF-α
FaecesDecreased Firmicutes, Limosillactobacter; increased Bacteroidota, Patescibacteria, Allobaculum, Candidatus Saccharimonas, and Ileibacteria; and increased levels of acetate and propionate
MG-H1-induced intestinal inflammationMale ICR miceHigh molecular weight UPF25 and 75 mg/kg/dayOral administration for 4 weeks ColonInhibited MPO activity; and decreased expression of ZO-1, RAGE, and TNF-α[31]
Carrageenan induced inflammationMale SD ratsUPF50 and 150 mg/kg/dayOral gavage
for 25 days
SerumDecreased production of CAT, POD, and SOD[34]
CCL4-induced oxidative stressFemale SD ratsUPF100 mg/kg/dayIntraperitoneal
injection for 2 weeks
SerumReduced levels of GOT, GPT, ALP, and LDH[51]
LiverDecreased MDA production; and increased SOD, CAT, and GPx
Broad-spectrum antibiotics (ABX)-induced tumour modelMale C57BL/6 miceUPF400 mg/kg/dayOral gavage
for 3 weeks
Tumour tissueReduced levels of CD31+, Bcl2; increased Bax level and CD8+ cells; and decreased CD4+ cells and IDO1 expression[67]
FaecesIncreased abundance of Akkermansia, Bifidobacterium, and Lactobacillus
Cyclophosphamide-induced immunosuppressionMale SD ratsHigh molecular weight UPF50, 100, and 200 mg/kg/dayOral administration for 4 weeksFaecesIncreased abundance of Papillibacter cinnamivorans and Desulfomicrobium orale; and reduced Marvinbryantia formatexigens[68]
Ovalbumin-induced CARASMale BALB/c miceEthanol-extracted UPE50, 100, and 200 mg/kg/dayOral administration for 16 daysSerumAttenuated IgE and IgG1 levels; and increased IgG2a[71]
Nasal lavage fluidIncreased expression of IFN-γ, SOD, and HO-1; reduced IL-4, IL-5, IL-13, and MDA; and enhanced ZO-1 and occludin
Bronchoalveolar lavage fluidDecreased levels of IL-4, IL-5, IL-13, and MDA; and increased HO-1 and occludin production
LungIncreased expression of occludin and ZO-1
Salmonella typhimurium-induced inflammationMale BALB/c miceUPF200 and 500 mg/kg/dayOral administration for 21 daysColonIncreased expression of occludin and claudin-1; reduced TNF-α, IKBα, p-IKBα, p65, and p-p65; elevated levels of CAT and SOD; and decreased MDA and iNOS[72]
FaecesReduced abundance of Proteobacteria, Colidextribacter, and Oscillibacter; increased Parabacteroides, Lactobacillus, Akkermansia, Lachnospiraceae_NK4A136 group and Muribaculum; and enhanced levels of acetate and butyrate
Table 3. In vitro neuroprotective activity of UPF.
Table 3. In vitro neuroprotective activity of UPF.
Cell LineModelCompoundTested
Concentration
Effective
Concentration
ActivityReference
Human RPE cell lineHSV-1-induced Aβ productionHCl-
extracted UPF
100 µg/mL100 µg/mLInhibited NF-κB phosphorylation, IL-6 expression, and Aβ42 synthesis; and reduced DPPH scavenging and ROS production [26]
Rat PC-12 cellsAβ-induced
neurotoxicity
UPF3.125–100 µg/mL3.125–100 µg/mLIncreased cell viability; reduced Aβ1–42 aggregation and cell apoptosis; and enhanced neurite outgrowth [84]
PC12 cells25–35 and d-Gal-induced
neurotoxicity
Water-extracted UPF100, 200, and 400 µg/mL100, 200, and 400 µg/mLImproved cell viability; prevented cell apoptosis; reduced levels of cleaved caspase-3, caspase-8, caspase-9, and cytochrome c; increased livin and X-linked apoptosis inhibitor protein expression; and elevated levels of SOD and GSH [85]
Hypothalamic neurons
(GT1-7 cells)
Tunicamycin-
induced ER stress
Ethanol-extracted UPE5–40 µg/mL5 µg/mLIncreased cell viability; reduced expression of CHOP and ATF-6; decreased levels of cleaved-PARP and cleaved-caspase-3; and modulated AKT/mTOR signalling [86]
BMDMsLPS-induced
macrophage
inflammation
Low molecular weight UPF0.0625, 0.125, 0.25, 0.5 mg/mL0.125 mg/mLReduced CD86+ proportion; increased CD206+ proportion; regulated AMPK/mTOR pathway [88]
Rat basal forebrain cholinergic
neurons
Aβ-induced
neurotoxicity
UPF50 nM–1 µM1 µMImproved neuronal survival; inhibited ROS generation and PKC phosphorylation; and blocked cleavage of caspases 9 and 3 [90]
Rat hippocampal neuronsHypoxia-mediated oxidative injuryEthanol-
extracted UPE
5, 15, 30 µg/mL15 µg/mLReduced ROS formation; increased cell viability; and decreased cytotoxicity [92]
Table 4. In vivo neuroprotective activity of UPF.
Table 4. In vivo neuroprotective activity of UPF.
ModelAnimalCompoundDoseTreatmentTissueResultReference
HFD-
induced
obesity
Male and
female C57BL/6J mice
UPF400 mg/kg/dayOral supplementation for 10 weeksHypothalamusReduced TNF-α, IL-1β, IL-6, and IFN-γ [22]
Nucleus
accumbens
Suppressed Lrrk2, Wfs1, and Ngb
SNL-induced neuropathic painMale SPF SD ratsUPF15, 50, and 100 mg/kg/dayIntrathecal injection for 10 daysLumbar spinal cordInhibited microglia and astrocyte activation; and reduced expression of GFAP and mac-1 [94]
Spinal dorsal hornDownregulated expression of TNF-α, IL-1β, and IL-6; and attenuated phosphorylation of ERK
IRI-caused neurological impairmentMale SD ratsUPF80 and 160 mg/kg/dayIntraperitoneal injection for 7 daysIschemic brainReduced levels of TNF-α, IL-1β, IL-6, MPO, SOD, MDA, p-p53, p-p38, p-ERK, p-JNK, and Bax; and increased Bcl-2 [95]
D-Gal-induced AD modelMale ICR miceUPF50, 100, and 200 mg/kg/dayOral administration for 21 daysBrainIncreased levels of Ach, ChAT, and GSH; reduced AChE activity; and decreased Aβ deposition [85]
SerumIncreased levels of SOD and GSH
Controlled cortical impact-induced TBIMale C57BL/6 miceLow molecular weight UPF10 and 50 mg/kgIntraperitoneal
injection
BrainDecreased brain oedema and cell apoptosis; reduced generation of MDA, 4-HNE, and ROS; increased levels of CAT, SOD, and GPx; suppressed cytochrome c release; and upregulated Sirt3 expression [96]
Aβ-induced AD modelCaenorhabditis elegansUPF50–500 ng/mLBath immersion methodEntire organismDecreased Aβ deposition, aggregation, and fibrillization; increased expression of pbs-2 and pbs-5; and reduced ROS production [97]
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

Yang, C.; Dwan, C.; Wimmer, B.C.; Ahamed, S.K.; James, F.; Thinley, J.; Wilson, R.; Johnson, L.; Caruso, V. Anti-Inflammatory and Neuroprotective Effects of Undaria pinnatifida Fucoidan. Mar. Drugs 2025, 23, 350. https://doi.org/10.3390/md23090350

AMA Style

Yang C, Dwan C, Wimmer BC, Ahamed SK, James F, Thinley J, Wilson R, Johnson L, Caruso V. Anti-Inflammatory and Neuroprotective Effects of Undaria pinnatifida Fucoidan. Marine Drugs. 2025; 23(9):350. https://doi.org/10.3390/md23090350

Chicago/Turabian Style

Yang, Cheng, Corinna Dwan, Barbara C. Wimmer, Sayed Koushik Ahamed, Fionnghuala James, Jigme Thinley, Richard Wilson, Luke Johnson, and Vanni Caruso. 2025. "Anti-Inflammatory and Neuroprotective Effects of Undaria pinnatifida Fucoidan" Marine Drugs 23, no. 9: 350. https://doi.org/10.3390/md23090350

APA Style

Yang, C., Dwan, C., Wimmer, B. C., Ahamed, S. K., James, F., Thinley, J., Wilson, R., Johnson, L., & Caruso, V. (2025). Anti-Inflammatory and Neuroprotective Effects of Undaria pinnatifida Fucoidan. Marine Drugs, 23(9), 350. https://doi.org/10.3390/md23090350

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