Next Article in Journal
Individualized Management of Osteoarthritis: The Role of Pharmacogenomics to Optimize Pain Therapy
Previous Article in Journal
On the Molecular Origin of the Toxicity of Erophaca baetica (L.) Boiss.
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Pharmacological Potential of Algal Polysaccharides in Food Applications and Chronic Disease Management

1
College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
2
Fujian Province Key Laboratory for the Development of Bioactive Material from Marine Algae, Quanzhou 362000, China
3
College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou 362000, China
4
College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
5
State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China
*
Authors to whom correspondence should be addressed.
Future Pharmacol. 2025, 5(2), 29; https://doi.org/10.3390/futurepharmacol5020029
Submission received: 17 April 2025 / Revised: 11 June 2025 / Accepted: 12 June 2025 / Published: 13 June 2025

Abstract

:
Algal polysaccharides are a kind of bioactive compound with diverse pharmacological applications, yet their structure–activity relationships and therapeutic potential in chronic disease management remain systematically underexplored. This review comprehensively analyzes the structural characteristics of brown, red, and green algal polysaccharides, revealing how specific structural features—such as glycosidic linkage patterns and sulfate group positioning—dictate their biological activities. We also demonstrated their multifaceted roles in diabetes, cancer, and cardiovascular diseases through distinct mechanisms, including gut microbiota modulation via short-chain fatty acid production, antioxidant enzyme activation, and targeted inhibition of pathological signaling pathways like mTOR and JAK-STAT3. The work further evaluates extraction methodologies, highlighting the advantages of emerging techniques such as enzyme-assisted and ultrasonic extraction for preserving bioactive integrity. By integrating fundamental research with practical applications in functional foods, this synthesis provides critical insights for harnessing algal polysaccharides in precision nutrition and sustainable biomedicine, while identifying key challenges in standardization and environmental safety that warrant future investigation.

Graphical Abstract

1. Introduction

As primary producers of marine ecosystems, seaweed is the basis of matter and energy [1,2], the value of which far exceeds that of land crops and has become a key factor in industrial development. Owing to their unique composition, seaweed is valuable in many fields.
Seaweeds are taxonomically classified into three primary phyla based on pigmentation: brown (Phaeophyceae), red (Rhodophyta), and green (Chlorophyta) algae [3]. As one of the most abundant marine resources, seaweeds contain a diverse array of bioactive constituents, such as polysaccharides, proteins, peptides, lipids, amino acids, dietary fibers, and vital minerals [4,5,6]. In addition, algae-derived polysaccharides including alginate, agar, and carrageenan have unique structural characteristics that make them valuable functional ingredients in the food industry, where they can be used as thickeners, gelling agents, and stabilizers. Therefore, information about algae polysaccharides is essential.
To comprehensively explore the publication volume of the literature related to seaweed, we adopted a systematic and rigorous search strategy using the Web of Science Core Collection (WOSCC) database [7]. Our search string incorporated the following keywords and synonyms within the WOSCC topic field: “algal polysaccharides”, “seaweed bioactive compounds”, “fucoidan”, “carrageenan”, “alginate”, “Brown algal polysaccharides”, “Red algal polysaccharides”, “Green algal polysaccharides”, combined with “extraction methods”, “pharmacological activity”, “food applications,” and “chronic disease management.” This generated a dataset of approximately 5251 articles. Subsequently, we refined our dataset by excluding non-English publications and non-original research articles, thereby removing a portion of articles from our initial collection. To ensure our review encompassed the latest research, we focused on titles, abstracts, and full texts published from the inception of the WOSCC database through October 2024. For data analysis convenience, we extracted metadata from the WOSCC database in text file format (.txt), including the complete record and cited references for each paper. We then utilized CiteSpace, a specialized software tool for scientometric analysis, to generate visual representations of structural relationships within the scientific literature [8]. Using CiteSpace Advanced version 6.2.4 (for 64-bit computers), we created collaborative networks of authors, institutions, and countries, as well as conducted keyword and co-citation analyses of the references (Figure 1). It turns out that people are becoming more and more interested in algae research. This review aims to critically synthesize current knowledge on (1) structure–activity relationships of algal polysaccharides across brown, red, and green algal classes; (2) their pharmacological mechanisms in chronic disease management; and (3) emerging extraction technologies and food applications. By evaluating these interconnected domains, we seek to establish a comprehensive framework for advancing algae-based functional materials in precision nutrition and sustainable biomedicine.

2. Classification and Structural Characteristics of Algal Polysaccharides

A range of techniques can be utilized to isolate polysaccharides from marine algae (Table 1), including aqueous thermal extraction, acid-assisted isolation, alkaline extraction, enzymatic extraction, and microwave extraction [9,10,11]. The methodologies and parameters used for extraction, including duration, temperature, pH, bias amount, solvent ratio, and type of elution solvent, can significantly alter the composition, extraction efficiency, and molecular weight of polysaccharides.
In the realm of food science, conventional extraction methodologies are often characterized by their inefficiency and time-consuming nature, which can result in the degradation or loss of certain bioactive compounds. On the other hand, innovative extraction approaches, such as ultrasonic-assisted extraction, have been shown to enhance the efficiency of the extraction process while preserving a higher proportion of these valuable constituents. When juxtaposed with traditional techniques, these contemporary methods also exhibit a reduced environmental impact. Ultrasonic-assisted extraction of fucoidan from Arctic brown algae was shown to enhance both antioxidant capacity and anticancer effects compared to conventional dynamic maceration, while better preserving the structural integrity of sulfated polysaccharides [23]. These findings emphasize that the extraction protocol must be carefully selected based on both yield and intended biological applications.

2.1. Brown Algal Polysaccharides

Brown algae polysaccharides (BSPs) are a class of bioactive substances that mainly exist in the cell–matrix and interstitial regions of brown algae and are the main natural functional components of brown algae [24].
Recent studies highlight significant variations in fucoidan’s biochemical composition and bioactivity across species and environments [25]. Arctic Fucus vesiculosus shows seasonal fluctuations, with galactose content peaking during reproduction while maintaining fucose dominance, and winter harvests exhibiting elevated sulfation correlating with enhanced DPPH radical scavenging [26]. Arctic-adapted F. spiralis and F. distichus contain higher xylose and mannose, demonstrating 2.1-fold stronger ORAC antioxidant activity than temperate species, alongside potent ACE inhibition for hypertension management [27,28]. Temperate Ascophyllum nodosum maintains stable fucose ratios and no cytotoxicity in human cells; these structural differences directly influence bioactivities and diabetes management [29]. Fucoidan has two main chain structures (labeled I and II in Figure 2a). Structure 1 is composed of (1 → 3)-α-L-fucopyranose as the main chain, while structure 2 contains α-L-fucopyranose residues linked via alternating (1 → 3) and (1 → 4) glycosidic bonds. These structural variations may influence bioactivity; for example, higher galactose content correlates with enhanced antiviral effects in Arctic isolates [30]. Due to its distinctive origin and chemical composition, fucoidan exhibits a range of exceptional biofunctional characteristics. For instance, it maintains immune balance in the intestine by regulating the Th1/Th2 ratio of intestinal helper T cells and the expression of immunoglobulin A, and an antiviral effect, not directly through the inactivation of virus particles, may act by inhibiting virus adsorption [31]. In the treatment of diabetes complications, fucoidan has been shown to reduce diabetes damage to the liver and reproductive system, reduce inflammation, enhance antioxidant capacity, and restore liver function [32].
Alginates are major structural polysaccharides in the cytoskeleton and extracellular compartments of brown algae. Alginate’s molecular structure (Figure 2b) consists of β-D-mannuronic acid (M-residues) and α-L-guluronic acid (G-residues), which are epimers [33]. Alginate has good gelling and thickening properties and is therefore widely used as a stabilizer and emulsifier for food products [34,35]. In 2018, the US Food and Drug Administration (FDA) added alginate to its dietary fiber list, requiring it to be counted when labeling total dietary fiber [36].
In brown algae, laminarin serves as an important reserve carbohydrate. This β-glucan, composed of 25–50 glucose monomers connected by 1,3-glycosidic bonds, primarily accumulates in the fibrillar cell walls and intercellular matrix [37]. Based on terminal group composition, laminarin chains are categorized into two types: M-chains contain D-mannitol at their reducing ends, while G-chains lack this mannitol moiety (Figure 2d). The primary sources of laminarin are brown algae belonging to the Laminaria and Alaria genera, which account for about 22–49% of algae dry matter. Enhancing dietary fiber consumption is just one of the benefits of laminarin and its enzymatic breakdown products. Research indicates that these substances can also impede the development of melanoma and colon cancer cells, and they exhibit properties that may prevent the spread of cancer [38,39]. These attributes suggest their potential utility in cancer treatment strategies.
These discoveries prompt scientific inquiry into the functional attributes of brown algae polysaccharides, spurring efforts to integrate these compounds into the development of health-focused food products in the future.

2.2. Red Algal Polysaccharides

Red algae are common macroalgae that can be classified into carrageenan and agar varieties on the basis of their carbohydrate composition. The polysaccharide content varies with growth and environmental conditions and is reported to be at least 40% to 50% of its dry weight [40].
Carrageenan is derived mainly from red algae of the families Gracilariaceae, Gelidiaceae, and Pterocladiaceae. It mainly consists of three types: kappa, lambda, and iota. All these types have D-galactose monomers as the basic structural backbone, which are linked with each other through alternating α-(1 → 3) and β-(1 → 4) glycosidic bonds (Figure 2e). Carrageenan exhibits unique thermal reversibility along with excellent gelation and film-forming properties comparable to gelatin. These characteristics make it particularly suitable as a gelling and film-forming agent in food processing applications such as jelly confections, fruit gels, fruit juices, and jams [41,42,43]. Its thermal properties further enhance these functional capabilities. These unique properties make carrageenan particularly suitable for creating edible gels and films in food products.
Agarose (Figure 2c) contains alternating D-galactose and 3,6-anhydro-L-galactose units, linked by β-(1 → 3) and α-(1 → 4) bonds [44]. The U.S. Food and Drug Administration initially approved agar as a food-grade additive derived from seaweed, authorizing its use for stabilization, thickening, and texture enhancement applications. Because agar is indigestible, it can be used to prepare dietary formulas and foods for individuals with diabetes [45]. Tyeb’s research team engineered an innovative agar-based wound patch incorporating iodo-potassium iodide and glycerin (AKIG), which showed remarkable efficacy in managing infected diabetic wounds in Wistar rats, achieving both infection eradication and full tissue regeneration [46]. Agar is particularly interesting in food packaging applications. For example, antibacterial nanocomposite films can be designed by incorporating AgNPs into biopolymers. These composite films produce strong antibacterial activity with high stability [47].
Red algae polysaccharides, as a versatile natural resource, have shown significant potential in food applications and health benefits. In the future, industry experts should develop more health-friendly and environmentally friendly food processing solutions, bringing more innovative possibilities to the industry.

2.3. Green Algal Polysaccharides

The polysaccharides in green algae are predominantly water-soluble sulfated polysaccharides found in the cell interstitium, and they also exist in the cell walls where the microfibrils are mainly composed of cellulose or mannose. Ulva and Enteromorpha are the main genera of green algae [48]. Among the polysaccharides from green algae, the structural characteristics of Ulva lactuca polysaccharide have been the most studied so far. Ulvan’s molecular architecture primarily consists of l-iduronic acid (IdoA), d-xylose (Xyl), d-glucuronic acid (GlcA), and sulfated l-rhamnose (Rha3S), with the sulfate group specifically esterified at the third position of the rhamnose moiety [49,50]. The fundamental repeating units in ulvan predominantly occur as three distinct disaccharide forms: Type A ulvanobiuronic acid (A3s), Type B ulvanobiuronic acid (B3s), and ulvanobioses type U (U3s) (Figure 2f). These three types constitute the major ulvanobioses found in ulvan. Ulvanobioses can be formed through various degradation pathways, including the β-elimination mechanism, which is catalyzed by ulvan lyases (such as those from the PL24, PL25, and PL28 families) [51]. These enzymes specifically cleave the β-glycosidic bonds in ulvan. Consequently, the extraction and purification steps of these ulvanobioses include protease hydrolysis, dialysis, ion exchange chromatography (IEC), and gel permeation chromatography (GPC) to ensure the integrity of their structure and function [52]. Ulvan enhances glucose uptake through the selective inhibition of key carbohydrate-metabolizing enzymes—AST, ALT, and GGT, which normally facilitate starch breakdown and absorption in both systemic circulation and intestinal lumen [53]. Ulvan demonstrates dual regulatory effects on glucose metabolism and physical function by upregulating INSR and AMPK expressions while simultaneously suppressing JNK, JAK-STAT3 signaling, and cellular senescence markers (p16/p38) [54].
Polysaccharides in green algae, especially Ulva polysaccharide, are rich in bioactive compounds, which have aroused widespread concern in the food science community. These substances are being eyed for their potential as functional food ingredients, owing to their beneficial biological activities, minimal toxicity, and environmentally friendly nature.

3. Application of Algal Polysaccharides in Food

3.1. As Food Additives

In recent years, constant development has taken place concerning the origin, category, function, and impact of food additives, and the discovery of nontoxic environmentally friendly safe food additives has been emphasized. Therefore, algal polysaccharides are considered important sources for the development of food additives owing to their unique physicochemical characteristics, nontoxicity, and environmentally friendly nature [55,56]. While algal polysaccharides offer significant benefits as food additives, it is important to consider that seaweeds can accumulate both essential and toxic elements from their marine environment. Certain species may contain elevated levels of heavy metals e.g., arsenic, cadmium, and lead, with concentrations varying by geographic location, species, and harvest season [57,58]. Brown algae from industrial coastal areas have been reported to contain Sr levels up to 2838.86 mg/kg in some cases [59]. An analysis of the metal element content in F. spiralis collected from Arctic regions (the Norwegian Sea, Irminger Sea, and Barents Sea) was conducted, along with an assessment of the health risks posed by the identified elements to humans, taking into account body weight and daily seaweed intake. The results indicate that this seaweed does not accumulate hazardous concentrations of toxic elements. Furthermore, it can serve as a source of dietary elements to meet daily nutritional requirements [60]. Fucus distichus exhibits a metal pollution index (MPI) ranging from 38.7 to 77.2 across different geographical regions, suggesting that its heavy metal content is significantly influenced by environmental conditions. However, health risk assessments of all samples showed hazard index (HI) values below 1, indicating no significant health risks [25]. Balina et al. evaluated the chemical composition of the brown seaweed Fucus vesiculosus collected from the Gulf of Riga. It was found to contain higher levels of both macroelements (490—21,500 ppm; P, K, Ca, Mg, Na, Fe, Mn) and trace elements (0.11—930 ppm; Zn, Cu, Cr, Pb, Sr, As, Cd, Se) compared to terrestrial plants [61]. Therefore, it is not recommended to use F. vesiculosus from the Gulf of Riga as food. Instead, its best potential application lies in utilizing it as biomass feedstock for biogas production. Regulatory measures have established maximum limits for these contaminants in seaweed products. Proper processing methods, including polysaccharide purification steps described in Section 2, can significantly reduce these risks while preserving bioactive properties [62].

3.1.1. As a Food Thickener

During hydration, algal polysaccharides develop extensive intermolecular hydrogen bonds that stabilize a three-dimensional architecture, resulting in simultaneous water retention and increased system viscosity. This property endows algal polysaccharides with remarkable viscosity and the capacity to form gels, making them capable of forming gel-like aqueous systems; therefore, they are suitable for application as thickeners. Three common major classes of seaweed thickeners are agar, carrageenan, and sodium alginate [63,64]. Since carrageenan has a linear macromolecular structure and possesses polyelectrolyte properties, it can form high-viscosity solutions [34]. When added to condiments such as soy sauce, fish sauce, and shrimp paste as thickening agents, it enhances the viscosity of the product and adjusts the flavor.

3.1.2. As Food Stabilizers

The incorporation of alginate into cream-based baked goods results in a stable gel network that imparts a smoother and more resilient texture, even when subjected to the rigors of freezing and thawing [34]. Propylene glycol alginate (PGA), in contrast to alginate, displays exceptional tolerance to salts and remains stable in the presence of high concentrations of electrolytes, avoiding the typical salting-out phenomenon observed with alginate [65]. Crucially, PGA also prevents the precipitation that is commonly triggered by calcium and other divalent metal ions, thus bolstering the stability of alginate in food formulations. These attributes have made PGA a popular ingredient in a variety of foods, including yogurt, milk drinks, and fruit juices, where its ability to increase product stability and texture is particularly beneficial [66]. PGA is added to yogurt to provide a natural texture, hence preventing the development of these unsightly rough surfaces, resulting in smooth and glossy products. When agar is used with glucose syrup or granulated sugar to make soft sweets, the resulting clarity and texture exceed those of other soft sweets [67]. It is worth noting that beyond the food industry, alginate and its derivatives are also widely used in personal care products such as hair care and skincare formulations. For example, due to its excellent moisturizing and film-forming properties, alginate is used in hair masks to enhance hair shine and smoothness. At the same time, its biocompatibility and antibacterial properties make it an ideal ingredient for skincare products, effectively protecting the skin barrier and reducing inflammation [68,69]. Additionally, alginate is also used in facial masks, where its water-absorbing and film-forming characteristics help lock in moisture, improving skin firmness and elasticity [70].
In general, algal polysaccharides present a variety of advantages as food additives that might provide a series of benefits: improved food texture, prolonged preservation time, and increased sensory properties. Moreover, there are some controversies over food safety, such as with respect to carrageenan. Thus, the exploration of risks in the research and application of algal polysaccharides is highly important for guaranteeing food safety.

3.2. As Functional Food Ingredients

Studies have shown that algal polysaccharides exhibit many physiological functions, such as antibacterial [71], antitumor [72,73], antioxidant, and immunomodulatory activities [74,75]. Therefore, functional foods with seaweed polysaccharides as the main component are gradually being developed and utilized [76].

3.2.1. Regulating the Intestinal Microbiota

Some polysaccharides found in seaweeds are low-fermentable sugars [77], which can be fermented by intestinal probiotics, altering the composition of the gut microbiota and producing beneficial metabolic byproducts that promote gut health, indicating their potential as prebiotics [24,78,79]. Algal polysaccharides function as prebiotics, reducing the intestinal pH [80], inhibiting the growth of pathogens [81], and increasing the expression of tight junction proteins [82]. The secretion of anti-inflammatory cytokines and antibodies from monocytes cells (DCs), T cells, and B cells is stimulated, thereby enhancing intestinal immunity (Figure 3) [24,83]. Furthermore, short-chain fatty acids (SCFAs) critically maintain gut homeostasis through their ligand–receptor interactions with intestinal epithelial cells, primarily via the activation of GPR41 and GPR43, two key G protein-coupled receptors involved in microbial–host crosstalk. These receptors play crucial roles in managing blood glucose levels, lipid profiles, obesity, and insulin signaling pathways [84,85,86]. Fucoidan is extracted from Pearsonothuria graeffei, and its identified structure strongly eliminates gut microbiota dysbiosis and metabolic disorders in high-fat diet (HFD) murine models [87].
In addition, there was an alteration in the level of fermentative bacteria in the contents of the rat cecum following the ingestion of the diet with algal polysaccharides, which further provided evidence that its gut microbiota composition could be drastically influenced by dietary intervention. Consuming algal polysaccharides and treating them as prebiotics is a promising strategy for regulating the gut microbiota and deserves further exploration in clinical settings.

3.2.2. Antioxidant

Recently, increasing amounts of research has focused on antioxidant compounds originating from dietary sources, including algal polysaccharides [88,89,90]. This trend reflects not only the increased interest in food safety but also the possible role of natural products in the antioxidant area.
Recent studies have elucidated the structure–activity relationships of fucoidan’s antioxidant effects. As previously demonstrated, the total antioxidant capacity exhibits a strong positive correlation with polyphenol content, while showing only a weak association with xylose content [91]. Importantly, these antioxidant properties are mediated through multiple mechanisms, particularly through the concentration-dependent inhibition of protein denaturation, which shows a strong correlation with fucose content and a moderate correlation with sulfate groups [91]. Among the tested compounds, purified fucoidan demonstrated the most potent activity against diclofenac sodium. Furthermore, fucoidans have been shown to effectively stabilize the membrane integrity of human red blood cells (HRBCs). The ability of fucoidan to eliminate hydroxyl radicals (OH) and counteract hemolysis induced by hydrogen peroxide (H2O2) increases with increasing concentration, while a certain degree of scavenging activity was also observed for superoxide radicals (O2−) [92]. Sulfated polysaccharides from algae significantly inhibit the respiratory burst of polymorphonuclear leukocytes (PMNs) and effectively eliminate free radicals produced during the respiratory burst [93]. Polysaccharides derived from brown seaweed exhibit potent free radical scavenging capacity against both DPPH and hydroxyl radicals (OH). Experimental evidence demonstrates their ability to inhibit hepatic lipid peroxidation in murine models while providing cytoprotective effects against Fe2+/H2O2 induced oxidative damage, ultimately attenuating mitochondrial swelling and preventing erythrocyte hemolysis [94]. Research in animal models demonstrates laminarin’s capacity to upregulate antioxidant defenses, notably increasing SOD and CAT activities, while significantly reducing MDA levels associated with oxidative lipid damage [95]. For red algae, sulfated polysaccharides such as those from Porphyra spp. have shown comparable antioxidant efficacy through hydroxyl radical scavenging and GPx activation [96]. Similarly, ulvan from green algae (Ulva lactuca) effectively suppresses lipid peroxidation and enhances antioxidant enzyme activity [97]. It is clear that polysaccharides from algae—regardless of if they are brown, red, or green—possess antioxidant capabilities. These antioxidants could enhance other health benefits of algal polysaccharides, including their anti-inflammatory and anticancer properties.

3.2.3. Cancer Fighting

Algal polysaccharides have a variety of biological activities and play a key role in cancer prevention and immune modulation due to their diverse biological activities. Most cases of breast cancer are associated with a family history, and polysaccharides extracted from red algae have been shown to influence the expression levels of apoptotic proteins (such as the Bax gene), P53, and caspase 8 in MCF7 and CoCa2 cells, thereby exerting anti-breast cancer and anti-colon cancer effects [98]. Fucoidans, carrageenan, and sulfated polysaccharides extracted from Laurencia papillosa also demonstrate therapeutic potential in breast cancer treatment [99,100]. In the treatment of hepatocellular carcinoma, chemotherapy and immunotherapy are current mainstays, yet they can lead to severe side effects. The use of natural compounds to treat cancer is becoming increasingly popular. For example, Ulva lactuca polysaccharide (ULP) was found to increase the expression of P53 and inhibit the mammalian target of rapamycin (mTOR) pathway, thereby inhibiting the growth of hepatocellular carcinoma cells [101]. Laminarin promotes the expression of the senescence marker protein-30, which significantly reduces the viability of Bel-7404 and HepG2 cells. Intratumoral injection of laminarin in Hepa 1–6 tumor-bearing mice can reduce tumor volume and weight [39]. Fucoidan suppresses the malignant characteristics of A549 lung adenocarcinoma cells through the dual inhibition of both PI3K/Akt and Erk-mediated signaling cascades [102]. Fucoidan can enhance the efficacy of conventional chemotherapy drugs through synergistic interactions. Notably, the co-administration of fucoidan with paclitaxel was shown to induce cell cycle arrest at the G2/M phase and produce significant synergistic effects in cancer treatment [103]. This finding highlights the potential of algal polysaccharides as adjuvants to improve existing cancer therapies while potentially reducing side effects. In fact, the antitumor effects of algal polysaccharides themselves are not mediated by one pathway but rather involve multiple mechanisms that may involve tumor cell apoptosis, the inhibition of stimulated immune responses, tumor cell proliferation, and the stimulation of immune responses. Fucoidan’s anti-diabetic potential through multiple mechanisms. Notably, fucoidan from Fucus vesiculosus demonstrates potent inhibition of dipeptidyl peptidase-IV (DPP-IV), a key enzyme in glucose metabolism, with an IC50 of 1.11 μg mL−1 [104]. This DPP-IV inhibition enhances incretin activity, thereby improving glucose-dependent insulin secretion and representing a promising pathway for managing hyperglycemia in diabetes.
While algal polysaccharides demonstrate diverse bioactivities, their pharmacological potential is intrinsically linked to pharmacokinetic properties that remain partially characterized. Critically, the pharmacological potential of algal polysaccharides is closely linked to their pharmacokinetic properties. Fucoidan, a representative sulfated polysaccharide, exhibits molecular weight-dependent absorption: low-molecular-weight fucoidan (LMWF, <10 kDa) achieves faster oral absorption and higher bioavailability than high-molecular-weight forms (>100 kDa) due to enhanced intestinal permeability [105,106]. Pharmacokinetic studies in rats reveal that orally administered fucoidan is primarily distributed to the kidneys, spleen, and liver; this organ-specific biodistribution profile aligns well with its known immunomodulatory and potential anticancer activities [107]. The compound exhibits a plasma half-life consistent with a dosing regimen requiring multiple daily administrations. Formulation strategies such as nano-encapsulation significantly improve fucoidan’s penetration and cellular uptake, enhancing its cytotoxicity against tumor cells [106]. Importantly, clinical studies confirm that the co-administration of fucoidan with chemotherapeutics (e.g., letrozole or tamoxifen) does not alter the plasma pharmacokinetics of these drugs, supporting its safety as an adjuvant [108].

3.3. Application in Food Preservation

Algal polysaccharides stand out as natural food preservatives that are both safe and eco-friendly. Alginate, in particular, is an affordable option that offers a suite of advantages, such as antimicrobial action and film-forming ability [109,110]. These attributes have made it a popular choice for preserving an array of food products, from fruits and meats to seafood, as noted in recent studies [111].
Algal polysaccharides can be utilized as food preservatives, primarily because of their antibacterial, moisturizing, and film-forming properties. Currently, various algal polysaccharides, including sodium alginate, ULP, laminarin, laver polysaccharide, and Sargassum polysaccharide, have been shown to be effective at preserving these compounds [112,113]. Sodium alginate is the most extensively studied alginate polysaccharide coating preservative, distinguished by its extensive availability, ease of acquisition, low cost, and effective preservation capabilities. However, there are several limitations in the use of single sodium alginate films, such as functional instability and poor mechanical and barrier properties, which drawbacks inevitably limit their further application and industrial scalability. Consequently, these considerations necessitate chemical derivatization of sodium alginate to achieve tailored properties. The polysaccharide backbone possesses four reactive centers amenable to molecular modification. Multiple chemical modification approaches can be employed to functionalize sodium alginate, such as oxidation reactions, copolymerization, grafting reactions, esterification, amidation, and Ugi modification, to introduce other polymer or nonpolymer molecules to form sodium alginate derivatives [114,115]. Through this treatment, sodium alginate acquires enhanced performance attributes, including greater water resistance, microbial inhibition capability, and emulsion stabilization effects, thereby increasing its applications in food preservation and meeting diverse market demands. Researchers have treated fruits such as cherries, plums, and tomatoes with sodium alginate composite-coating agents [116,117], and the results indicate that these fruits exhibit significantly reduced browning and spoilage rates during storage. The composite coatings effectively inhibited the respiration of fruits and reduced water evaporation, thereby extending their shelf-life.

3.4. Applications in Food Packaging

3.4.1. Edible Films

Alginate has excellent membrane properties and is suitable for edible films (Figure 4). The addition of antibacterial agents and antioxidants can improve the performance of edible films produced by algal polysaccharides [118,119,120], thereby enhancing their strong antibacterial action against foodborne bacteria and DPPH free radical scavenging ability. This improvement helps to suppress the growth of microorganisms and fat oxidation in packaged foods [121,122], which ultimately extends shelf-life.
An alginate-edible coating can prolong the shelf-life of pepper by forming a protective film that inhibits microbial growth A coating material (KGM/KC-CO) consisting of κ-carrageenan (KC) with other ingredients was developed to extend the shelf-life of chickens [118]. Edible coatings and films of a wide variety have been studied for their role in preserving the quality of meat products. Alginate–agar coating significantly extends the shelf-life of beef and enhances its sensory attributes [123]. For products from fish, the development of new coating technologies with antioxidant properties allows packaging material functionalities to be performed well enough and extends storage stability and freshness of fish products very well [124].

3.4.2. Microencapsulation in Food Products

Algal polysaccharides have excellent emulsifying and gelling properties [125], as well as film-forming properties [126]; hence, they are essential wall materials for microcapsules (Figure 4). The encapsulation of substances in algal polysaccharide capsules increases their ability to resist various environmental factors and decreases the toxic side effects of the preparation without reducing its efficacy [127].
This ability of alginate to form a stable gel upon mixing with calcium ions has been utilized by scientists in the development of microcapsules for small molecule encapsulation since this gel remains stable in acidic gastric juices but dissolves in the alkaline environment of the small intestine [128]. Recent investigations demonstrate that red algal polysaccharides serve as effective encapsulating agents for BSG-derived bioactive peptides, exhibiting exceptional encapsulation efficiency during simulated gastrointestinal conditions [129]. Using a co-precipitation method, chitosan can be combined with carrageenan in this process to effectively microencapsulate essential oils, such as chili oil [130]. In this way, this not only improves the stability and bioavailability of spices but also improves their release characteristics in foods, enhance flavor, and prolong their shelf life. Probiotics encapsulated with microcapsules prepared from algal polysaccharides as raw materials retain extremely high activity both in simulated gastric and intestinal environments and thus improve the survival rate of Lactobacillus plantarum [131]. Polysaccharides extracted from Agaricus bisporus mushrooms are encapsulated by κ-carrageenan. It has a high swelling capacity at alkaline pH and a slower release rate in acidic media, allowing the controlled release of the encapsulated polysaccharide in target environmental conditions [132].

4. Future Prospects and Conclusions

Marine algal polysaccharides represent a promising class of bioactive compounds with diverse applications in nutrition, food technology, and biomedicine. Their unique structural properties, including sulfation patterns and glycosidic linkages, underlie their broad biological activities, ranging from antioxidant and immunomodulatory effects to gut microbiota regulation. These functional properties have driven their increasing utilization as natural ingredients in functional foods, dietary supplements, and pharmaceutical formulations.
Despite this potential, several critical challenges must be addressed to ensure sustainable development of algal polysaccharide-based products. Environmental concerns, particularly regarding the accumulation of marine pollutants and heavy metals in seaweed biomass, highlight the need for rigorous quality control measures and standardized safety assessments [133,134]. Furthermore, the variability in polysaccharide composition due to species, geographical origin, and seasonal factors necessitates the establishment of comprehensive characterization protocols.
Looking ahead, advancements in extraction and purification technologies will be crucial for improving the consistency and bioactivity of algal polysaccharides. Collaborative efforts between researchers, industry stakeholders, and regulatory agencies are essential to establish safety guidelines and promote sustainable harvesting practices. By addressing these challenges, algal polysaccharides can be further developed into safe and effective functional ingredients, supporting their expanded use in health-promoting applications while ensuring environmental sustainability.

Author Contributions

X.W.: Formal analysis, Investigation, Resources, Writing—original draft, Writing—review and editing, visualization. Y.G.: Writing—review and editing. C.Z. and C.D.: Conceptualization, Resources, Writing—review and editing, Visualization, Supervision, Funding acquisition. All authors have read and agreed to the published version of the manuscript.

Funding

The project was funded by FuXiaQuan National Independent Innovation Demonstration Zone Collaborative Innovation Platform Project (2023FX0001) and Fujian Province Key Laboratory for the Development of Bioactive Material from Marine Algae (2022KF11).

Acknowledgments

The authors would like to thank the reviewers and Journal Editor for their thoughtful reading of the manuscript and constructive comments.

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. And there is any conflict of interest for you and the co-authors.

References

  1. Cotas, J.; Gomes, L.; Pacheco, D.; Pereira, L. Ecosystem services provided by seaweeds. Hydrobiology 2023, 2, 75–96. [Google Scholar] [CrossRef]
  2. Anbuchezhian, R.; Karuppiah, V.; Li, Z. Prospect of marine algae for production of industrially important chemicals. In Algal Biorefinery: An Integrated Approach; Springer: Berlin/Heidelberg, Germany, 2015; pp. 195–217. [Google Scholar] [CrossRef]
  3. Mišurcová, L. Chemical composition of seaweeds. In Handbook of Marine Macroalgae: Biotechnology and Applied Phycology; Wiley: Hoboken, NJ, USA, 2011; pp. 171–192. [Google Scholar]
  4. Peñalver, R.; Lorenzo, J.M.; Ros, G.; Amarowicz, R.; Pateiro, M.; Nieto, G. Seaweeds as a functional ingredient for a healthy diet. Mar. Drugs 2020, 18, 301. [Google Scholar] [CrossRef]
  5. Figueroa, V.; Farfán, M.; Aguilera, J.M. Seaweeds as novel foods and source of culinary flavors. Food Rev. Int. 2023, 39, 1–26. [Google Scholar] [CrossRef]
  6. Lozano Muñoz, I.; Díaz, N.F. Minerals in edible seaweed: Health benefits and food safety issues. Crit. Rev. Food Sci. Nutr. 2020, 62, 1592–1607. [Google Scholar] [CrossRef]
  7. Segaran, T.C.; Azra, M.N.; Noor, M.I.M.; Danish-Daniel, M.; Burlakovs, J.; Lananan, F.; Xu, J.; Kari, Z.A.; Wei, L.S. Knowledge map analysis of the global seaweed research using CiteSpace. Heliyon 2024, 10, e28418. [Google Scholar] [CrossRef]
  8. Segaran, T.C.; Azra, M.N.; Handayani, K.S.; Lananan, F.; Xu, J. Seaweed and climate change: A map review. Mar. Environ. Res. 2023, 192, 106216. [Google Scholar] [CrossRef]
  9. Yuan, Y.; Macquarrie, D. Microwave assisted extraction of sulfated polysaccharides (fucoidan) from Ascophyllum nodosum and its antioxidant activity. Carbohydr. Polym. 2015, 129, 101–107. [Google Scholar] [CrossRef]
  10. Lomartire, S.; Gonçalves, A.M. Novel technologies for seaweed polysaccharides extraction and their use in food with therapeutically applications—A review. Foods 2022, 11, 2654. [Google Scholar] [CrossRef]
  11. Otero, P.; Carpena, M.; Garcia-Oliveira, P.; Echave, J.; Soria-Lopez, A.; García-Pérez, P.; Prieto, M.A. Seaweed polysaccharides: Emerging extraction technologies, chemical modifications and bioactive properties. Crit. Rev. Food Sci. Nutr. 2023, 63, 1901–1929. [Google Scholar] [CrossRef]
  12. Kadam, S.U.; Alvarez, C.; Tiwari, B.K.; O’Donnell, C.P. Extraction of biomolecules from seaweeds. In Seaweed Sustainability; Tiwari, B.K., Troy, D.J., Eds.; Academic Press: Cambridge, MA, USA, 2015; pp. 243–269. [Google Scholar] [CrossRef]
  13. Lin, J.; Jiao, G.; Kermanshahi-Pour, A. Algal polysaccharides-based hydrogels: Extraction, synthesis, characterization, and applications. Mar. Drugs 2022, 20, 306. [Google Scholar] [CrossRef]
  14. Ojima, T.; Rahman, M.M.; Kumagai, Y.; Nishiyama, R.; Narciso, J.; Inoue, A. Polysaccharide-degrading enzymes from marine gastropods. Methods Enzymol. 2018, 605, 457–497. [Google Scholar] [CrossRef] [PubMed]
  15. Rajauria, G.; Ravindran, R.; Garcia-Vaquero, M.; Rai, D.K.; Sweeney, T.; O’Doherty, J. Molecular characteristics and antioxidant activity of laminarin extracted from the seaweed species Laminaria hyperborea, using hydrothermal-assisted extraction and a multi-step purification procedure. Food Hydrocoll. 2021, 112, 106332. [Google Scholar] [CrossRef]
  16. Fernando, I.P.S.; Dias, M.K.H.M.; Madusanka, D.M.D.; Han, E.J.; Kim, M.J.; Jeon, Y.J.; Ahn, G. Step gradient alcohol precipitation for the purification of low molecular weight fucoidan from Sargassum siliquastrum and its UVB protective effects. Int. J. Biol. Macromol. 2020, 163, 26–35. [Google Scholar] [CrossRef] [PubMed]
  17. Alboofetileh, M.; Rezaei, M.; Tabarsa, M.; Rittà, M.; Donalisio, M.; Mariatti, F.; Cravotto, G. Effect of different non-conventional extraction methods on the antibacterial and antiviral activity of fucoidans extracted from Nizamuddinia zanardinii. Int. J. Biol. Macromol. 2019, 124, 131–137. [Google Scholar] [CrossRef]
  18. Magnusson, M.; Yuen, A.K.; Zhang, R.; Wright, J.T.; Taylor, R.B.; Maschmeyer, T.; de Nys, R. A comparative assessment of microwave assisted (MAE) and conventional solid-liquid (SLE) techniques for the extraction of phloroglucinol from brown seaweed. Algal Res. 2017, 23, 28–36. [Google Scholar] [CrossRef]
  19. Carreira-Casais, A.; Otero, P.; Garcia-Perez, P.; Garcia-Oliveira, P.; Pereira, A.G.; Carpena, M.; Soria-Lopez, A.; Simal-Gandara, J.; Prieto, M.A. Benefits and drawbacks of ultrasound-assisted extraction for the recovery of bioactive compounds from marine algae. Int. J. Environ. Res. Public Health 2021, 18, 9153. [Google Scholar] [CrossRef]
  20. Kadam, S.; O’Donnell, C.; Rai, D.; Hossain, M.; Burgess, C.; Walsh, D.; Tiwari, B. Laminarin from Irish brown seaweeds Ascophyllum nodosum and Laminaria hyperborea: Ultrasound assisted extraction, characterization and bioactivity. Mar. Drugs 2015, 13, 4270–4280. [Google Scholar] [CrossRef]
  21. Wu, S.C. Antioxidant activity of sulfated seaweeds polysaccharides by novel assisted extraction. In Solubility of Polysaccharides; Xu, Z., Ed.; IntechOpen: London, UK, 2017; pp. 89–108. [Google Scholar] [CrossRef]
  22. Saravana, P.S.; Tilahun, A.; Gerenew, C.; Tri, V.D.; Kim, N.H.; Kim, G.D.; Chun, B.S. Subcritical water extraction of fucoidan from Saccharina japonica: Optimization, characterization and biological studies. J. Appl. Phycol. 2018, 30, 579–590. [Google Scholar] [CrossRef]
  23. Obluchinskaya, E.D.; Pozharitskaya, O.N.; Lapina, I.M.; Kulminskaya, A.A.; Zhurishkina, E.V.; Shikov, A.N. Comparative evaluation of dynamic maceration and ultrasonic assisted extraction of fucoidan from four arctic brown algae on its antioxidant and anticancer properties. Mar. Drugs 2025, 23, 230. [Google Scholar] [CrossRef]
  24. Okolie, C.L.; Rajendran, S.R.C.K.; Udenigwe, C.C.; Aryee, A.N.; Mason, B. Prospects of brown seaweed polysaccharides (BSP) as prebiotics and potential immunomodulators. J. Food Biochem. 2017, 41, e12392. [Google Scholar] [CrossRef]
  25. Obluchinskaya, E.D.; Pozharitskaya, O.N.; Gorshenina, E.V.; Zakharov, D.V.; Flisyuk, E.V.; Terninko, I.I.; Generalova, Y.E.; Shikov, A.N. Arctic edible brown alga Fucus distichus L.: Biochemical composition, antiradical potential and human health risk. Plants 2023, 12, 2380. [Google Scholar] [CrossRef] [PubMed]
  26. Obluchinskaya, E.D.; Pozharitskaya, O.N.; Zakharov, D.V.; Flisyuk, E.V.; Terninko, I.I.; Generalova, Y.E.; Smekhova, I.E.; Shikov, A.N. The Biochemical composition and antioxidant properties of Fucus vesiculosus from the Arctic region. Mar. Drugs 2022, 20, 193. [Google Scholar] [CrossRef] [PubMed]
  27. Francisco, J.; Horta, A.; Pedrosa, R.; Afonso, C.; Cardoso, C.; Bandarra, N.M.; Gil, M.M. Bioaccessibility of antioxidants and fatty acids from Fucus spiralis. Foods 2020, 9, 440. [Google Scholar] [CrossRef] [PubMed]
  28. Flores-Contreras, E.A.; Araújo, R.G.; Rodríguez-Aguayo, A.A.; Guzmán-Román, M.; García-Venegas, J.C.; Nájera-Martínez, E.F.; Sosa-Hernández, J.E.; Iqbal, H.M.N.; Melchor-Martínez, E.M.; Parra-Saldivar, R. Polysaccharides from the Sargassum and brown algae genus: Extraction, purification, and their potential therapeutic applications. Plants 2023, 12, 2445. [Google Scholar] [CrossRef]
  29. Keleszade, E.; Patterson, M.; Trangmar, S.; Guinan, K.J.; Costabile, A. Clinical efficacy of brown seaweeds Ascophyllum nodosum and Fucus vesiculosus in the prevention or delay progression of the metabolic syndrome: A review of clinical trials. Molecules 2021, 26, 714. [Google Scholar] [CrossRef]
  30. Grice, I.D.; Mariottini, G.L. Glycans with antiviral activity from marine organisms. In Marine Organisms as Model Systems in Biology and Medicine; Springer: Berlin/Heidelberg, Germany, 2018; pp. 439–475. [Google Scholar]
  31. Yang, C.H.; Tian, J.J.; Ko, W.S.; Shih, C.J.; Chiou, Y.L. Oligo-fucoidan improved unbalance the Th1/Th2 and Treg/Th17 ratios in asthmatic patients: An ex vivo study. Exp. Ther. Med. 2019, 17, 3–10. [Google Scholar] [CrossRef]
  32. Ma, Z.G.; Kong, C.Y.; Song, P.; Zhang, X.; Yuan, Y.P.; Tang, Q.Z. Geniposide protects against obesity-related cardiac injury through AMPKα-and Sirt1-dependent mechanisms. Oxidative Med. Cell. Longev. 2018, 2018, 6053727. [Google Scholar] [CrossRef]
  33. Rashedy, S.H.; Abd El Hafez, M.S.; Dar, M.A.; Cotas, J.; Pereira, L. Evaluation and characterization of alginate extracted from brown seaweed collected in the Red Sea. Appl. Sci. 2021, 11, 6290. [Google Scholar] [CrossRef]
  34. Qin, Y. Seaweed hydrocolloids as thickening, gelling, and emulsifying agents in functional food products. In Bioactive Seaweeds for Food Applications; Academic Press: Cambridge, MA, USA, 2018; pp. 135–152. [Google Scholar] [CrossRef]
  35. Terescenco, D.; Hadj Benali, L.; Canivet, F.; Benoit le Gelebart, M.; Hucher, N.; Gore, E.; Picard, C. Biosourced polymers in cosmetic emulsions: A hidden potential of the alginates as thickeners and gelling agents. Int. J. Cosmet. Sci. 2021, 43, 573–587. [Google Scholar] [CrossRef]
  36. Younes, M.; Aggett, P.; Aguilar, F.; Crebelli, R.; Filipič, M.; Dusemund, B. Re-evaluation of propane 1, 2 diol alginate (E 405) as a food additive. EFSA J. 2018, 16, e05371. [Google Scholar] [CrossRef]
  37. Labourel, A.; Jam, M.; Jeudy, A.; Hehemann, J.H.; Czjzek, M.; Michel, G. The β-glucanase ZgLamA from Zobellia galactanivorans evolved a bent active site adapted for efficient degradation of algal laminarin. J. Biol. Chem. 2014, 289, 2027–2042. [Google Scholar] [CrossRef] [PubMed]
  38. Menshova, R.V.; Ermakova, S.P.; Anastyuk, S.D.; Isakov, V.V.; Dubrovskaya, Y.V.; Kusaykin, M.I.; Zvyagintseva, T.N. Structure, enzymatic transformation and anticancer activity of branched high molecular weight laminaran from brown alga Eisenia bicyclis. Carbohydr. Polym. 2014, 99, 101–109. [Google Scholar] [CrossRef] [PubMed]
  39. Tian, L.; Li, C.M.; Li, Y.F.; Huang, T.M.; Chao, N.X.; Luo, G.R.; Mo, F.R. Laminarin from seaweed (Laminaria japonica) inhibits hepatocellular carcinoma through upregulating senescence marker protein-30. Cancer Biother. Radiopharm. 2020, 35, 277–283. [Google Scholar] [CrossRef]
  40. Jiang, J.L.; Zhang, W.Z.; Ni, W.X.; Shao, J.W. Insight on structure-property relationships of carrageenan from marine red algal: A review. Carbohydr. Polym. 2021, 257, 117642. [Google Scholar] [CrossRef]
  41. Blakemore, W.R.; Harpell, A.R. Carrageenan. In Food Stabilisers, Thickeners and Gelling Agents; Wiley: Hoboken, NJ, USA, 2009; pp. 73–94. [Google Scholar]
  42. Bagal-Kestwal, D.R.; Pan, M.H.; Chiang, B.H. Properties and applications of gelatin, pectin, and carrageenan gels. In Bio Monomers for Green Polymeric Composite Materials; Wiley: Hoboken, NJ, USA, 2019; pp. 117–140. [Google Scholar] [CrossRef]
  43. Agoda-Tandjawa, G.; Le Garnec, C.; Boulenguer, P.; Gilles, M.; Langendorff, V.J.F.H. Rheological behavior of starch/carrageenan/milk proteins mixed systems: Role of each biopolymer type and chemical characteristics. Food Hydrocoll. 2017, 73, 300–312. [Google Scholar] [CrossRef]
  44. Lee, S.B.; Cho, S.J.; Kim, J.A.; Lee, S.Y.; Kim, S.M.; Lim, H.S. Metabolic pathway of 3,6-anhydro-L-galactose in agar-degrading microorganisms. Biotechnol. Bioprocess Eng. 2014, 19, 866–878. [Google Scholar] [CrossRef]
  45. Armisen, R.; Gaiatas, F. Agar. In Handbook of Hydrocolloids; Woodhead Publishing: Cambridge, UK, 2009; pp. 82–107. [Google Scholar]
  46. Tyeb, S.; Kumar, N.; Kumar, A.; Verma, V.J.A.A.B.M. Agar–Iodine Transdermal Patches for Infected Diabetic Wounds. ACS Appl. Bio Mater. 2020, 3, 7515–7530. [Google Scholar] [CrossRef]
  47. Rhim, J.W.; Wang, L.F.; Hong, S.I. Preparation and characterization of agar/silver nanoparticles composite films with antimicrobial activity. Food Hydrocoll. 2013, 33, 327–335. [Google Scholar] [CrossRef]
  48. Wang, J.; Li, N.; Jiang, P.; Boo, S.M.; Lee, W.J.; Cui, Y.; Qin, S. Ulva and Enteromorpha (Ulvaceae, Chlorophyta) from two sides of the Yellow Sea: Analysis of nuclear rDNA ITS and plastid rbc L sequence data. Chin. J. Oceanol. Limnol. 2010, 28, 762–768. [Google Scholar] [CrossRef]
  49. Tian, H.; Yin, X.; Zeng, Q.; Zhu, L.; Chen, J. Isolation, structure, and surfactant properties of polysaccharides from Ulva lactuca L. from South China Sea. Int. J. Biol. Macromol. 2015, 79, 577–582. [Google Scholar] [CrossRef]
  50. Tziveleka, L.-A.; Ioannou, E.; Roussis, V. Ulvan, a bioactive marine sulphated polysaccharide as a key constituent of hybrid biomaterials: A review. Carbohydr. Polym. 2019, 218, 355–370. [Google Scholar] [CrossRef] [PubMed]
  51. Tang, T.; Zhu, B.; Yao, Z. Biochemical characterization and elucidation the action mode of a new PL25 family ulvan lyase from marine bacterium Alteromonas sp. TK-45 (2). Algal Res. 2022, 67, 102854. [Google Scholar] [CrossRef]
  52. Li, C.; Wang, H.; Zhu, B.; Yao, Z.; Ning, L. Polysaccharides and oligosaccharides originated from green algae: Structure, extraction, purification, activity and applications. Bioresour. Bioprocess. 2024, 11, 85. [Google Scholar] [CrossRef]
  53. Rizk, M.Z.; Aly, H.F.; Matloub, A.A.; Fouad, G.I. The anti-hypercholesterolemic effect of ulvan polysaccharide extracted from the green alga Ulva fasciata on aged hypercholesterolemic rats. Asian J. Pharm. Clin. Res. 2016, 9, 165–176. [Google Scholar]
  54. Ruan, Q.; Chen, Y.; Wen, J.; Qiu, Y.; Huang, Y.; Zhang, Y.; Zhao, C. Regulatory mechanisms of the edible alga Ulva lactuca polysaccharide via modulation of gut microbiota in diabetic mice. Food Chem. 2022, 409, 135287. [Google Scholar] [CrossRef]
  55. Usman, A.; Khalid, S.; Usman, A.; Hussain, Z.; Wang, Y. Algal polysaccharides, novel application, and outlook. In Algae Based Polymers, Blends, and Composites; Elsevier: Amsterdam, The Netherlands, 2017; pp. 115–153. [Google Scholar] [CrossRef]
  56. Ścieszka, S.; Klewicka, E. Algae in food: A general review. Crit. Rev. Food Sci. Nutr. 2019, 59, 3538–3547. [Google Scholar] [CrossRef]
  57. Karthikeyan, P.; Marigoudar, S.R.; Mohan, D.; Sharma, K.V.; Murthy, M.R. Prescribing sea water quality criteria for arsenic, cadmium and lead through species sensitivity distribution. Ecotoxicol. Environ. Saf. 2021, 208, 111612. [Google Scholar] [CrossRef]
  58. Hussain, M.M.; Wang, J.; Bibi, I.; Shahid, M.; Niazi, N.K.; Iqbal, J.; Mian, I.A.; Shaheen, S.M.; Bashir, S.; Shah, N.S.; et al. Arsenic speciation and biotransformation pathways in the aquatic ecosystem: The significance of algae. J. Hazard. Mater. 2021, 403, 124027. [Google Scholar] [CrossRef]
  59. Siddique, M.A.M.; Hossain, M.S.; Islam, M.M.; Rahman, M.; Kibria, G. Heavy metals and metalloids in edible seaweeds of Saint Martin’s Island, Bay of Bengal, and their potential health risks. Mar. Pollut. Bull. 2022, 181, 113866. [Google Scholar] [CrossRef]
  60. Obluchinskaya, E.D.; Pozharitskaya, O.N.; Zakharov, D.V.; Flisyuk, E.V.; Terninko, I.I.; Generalova, Y.E.; Shikov, A.N. Biochemical composition, antiradical potential and human health risk of the Arctic edible brown seaweed Fucus spiralis L. J. Appl. Phycol. 2023, 35, 365–380. [Google Scholar] [CrossRef]
  61. Balina, K.; Romagnoli, F.; Blumberga, D. Chemical composition and potential use of Fucus vesiculosus from Gulf of Riga. Energy Procedia 2016, 95, 43–49. [Google Scholar] [CrossRef]
  62. Wassie, T.; Niu, K.; Xie, C.; Wang, H.; Xin, W. Extraction techniques, biological activities and health benefits of marine algae Enteromorpha prolifera polysaccharide. Front. Nutr. 2021, 8, 747928. [Google Scholar] [CrossRef] [PubMed]
  63. Bose, I.; Nousheen; Roy, S.; Yaduvanshi, P.; Sharma, S.; Chandel, V.; Biswas, D. Unveiling the potential of marine biopolymers: Sources, classification, and diverse food applications. Materials 2023, 16, 4840. [Google Scholar] [CrossRef]
  64. Thiviya, P.; Gamage, A.; Liyanapathiranage, A.; Makehelwala, M.; Dassanayake, R.S.; Manamperi, A.; Madhujith, T. Algal polysaccharides: Structure, preparation and applications in food packaging. Food Chem. 2023, 405, 134903. [Google Scholar] [CrossRef]
  65. Nilsen-Nygaard, J.; Hattrem, M.N.; Draget, K.I. Propylene glycol alginate (PGA) gelled foams: A systematic study of surface activity and gelling properties as a function of degree of esterification. Food Hydrocoll. 2016, 57, 80–91. [Google Scholar] [CrossRef]
  66. Chen, L.; Xu, W.; Chen, D.; Chen, G.; Liu, J.; Zeng, X.; Zhu, H. Digestibility of sulfated polysaccharide from the brown seaweed Ascophyllum nodosum and its effect on the human gut microbiota in vitro. Int. J. Biol. Macromol. 2018, 112, 1055–1061. [Google Scholar] [CrossRef]
  67. Yu, H.; Liu, H.; Wang, L.; Zhang, Y.; Tian, H.; Ma, X. Effect of poly-γ-glutamic acid on the stability of set yoghurts. J. Food Sci. Technol. 2018, 55, 4634–4641. [Google Scholar] [CrossRef]
  68. Abka-Khajouei, R.; Tounsi, L.; Shahabi, N.; Patel, A.K.; Abdelkafi, S.; Michaud, P. Structures, properties and applications of alginates. Mar. Drugs 2022, 20, 364. [Google Scholar] [CrossRef]
  69. Pournaki, S.K.; Aleman, R.S.; Hasani-Azhdari, M.; Marcia, J.; Yadav, A.; Moncada, M. Current Review: Alginate in the Food Applications. Multidiscip. J. 2024, 7, 281–301. [Google Scholar] [CrossRef]
  70. Liao, Y.; Chang, C.; Nagarajan, D.; Chen, C.; Chang, J. Algae-Derived Hydrocolloids in Foods: Applications and Health-Related Issues. Bioengineered 2021, 12, 3787–3801. [Google Scholar] [CrossRef]
  71. Braicu, C.; Buse, M.; Busuioc, C.; Drula, R.; Gulei, D.; Raduly, L.; Berindan-Neagoe, I. A Comprehensive Review on MAPK: A Promising Therapeutic Target in Cancer. Cancers 2019, 11, 1618. [Google Scholar] [CrossRef] [PubMed]
  72. Yao, W.; Qiu, H.M.; Cheong, K.L.; Zhong, S. Advances in anti-cancer effects and underlying mechanisms of marine algae polysaccharides. Int. J. Biol. Macromol. 2022, 221, 472–485. [Google Scholar] [CrossRef] [PubMed]
  73. Xie, P.; Horio, F.; Fujii, I.; Zhao, J.; Shinohara, M.; Matsukura, M. A novel polysaccharide derived from algae extract inhibits cancer progression via JNK, not via the p38 MAPK signaling pathway. Int. J. Oncol. 2018, 52, 1380–1390. [Google Scholar] [CrossRef]
  74. Zhong, Q.; Wei, B.; Wang, S.; Ke, S.; Chen, J.; Zhang, H.; Wang, H. The Antioxidant activity of polysaccharides derived from marine organisms: An overview. Mar. Drugs 2019, 17, 674. [Google Scholar] [CrossRef]
  75. Rocha de Souza, M.C.; Marques, C.T.; Guerra Dore, C.M.; Ferreira da Silva, F.R.; Oliveira Rocha, H.A.; Leite, E.L. Antioxidant activities of sulfated poly-saccharides from brown and red seaweeds. J. Appl. Phycol. 2007, 19, 153–160. [Google Scholar] [CrossRef]
  76. Gomez-Zavaglia, A.; Prieto Lage, M.A.; Jimenez-Lopez, C.; Mejuto, J.C.; Simal-Gandara, J. The potential of seaweeds as a source of functional ingre-dients of prebiotic and antioxidant value. Antioxidants 2019, 8, 406. [Google Scholar] [CrossRef]
  77. Nie, J.; Fu, X.; Wang, L.; Xu, J.; Gao, X. A systematic review of fermented Saccharina japonica: Fermentation conditions, metabolites, potential health benefits and mechanisms. Trends Food Sci. Technol. 2022, 123, 15–27. [Google Scholar] [CrossRef]
  78. Zaporozhets, T.S.; Besednova, N.N.; Kuznetsova, T.A.; Zvyagintseva, T.N.; Makarenkova, I.D.; Kryzhanovsky, S.P.; Melnikov, V.G. The prebiotic potential of polysaccharides and extracts of seaweeds. Russ. J. Mar. Biol. 2014, 40, 1–9. [Google Scholar] [CrossRef]
  79. Lopez-Santamarina, A.; Miranda, J.M.; Mondragon, A.D.C.; Lamas, A.; Cardelle-Cobas, A.; Franco, C.M.; Cepeda, A. Potential Use of Marine Sea-weeds as Prebiotics: A Review. Molecules 2020, 25, 1004. [Google Scholar] [CrossRef]
  80. Yang, C.; Huang, S.; Lin, Z.; Chen, H.; Xu, C.; Lin, Y.; Sun, H.; Huang, F.; Lin, D.; Guo, F. Polysaccharides from Enteromorpha prolifera alleviate hypercholes-terolemia via modulating the gut microbiota and bile acid metabolism. Food Funct. 2022, 13, 12194–12207. [Google Scholar] [CrossRef]
  81. Hajji, M.; Hamdi, M.; Sellimi, S.; Ksouda, G.; Laouer, H.; Li, S.; Nasri, M. Structural characterization, antioxidant and antibacterial activities of a novel polysaccharide from Periploca laevigata root barks. Carbohydr. Polym. 2019, 206, 380–388. [Google Scholar] [CrossRef] [PubMed]
  82. Guo, H.; Zhou, Y.; Xie, Q.; Chen, H.; Zhang, M.; Yu, L.; Yan, G.; Chen, Y.; Lin, X.; Zhang, Y.; et al. Protective effects of Laminaria japonica polysaccharide composite microcapsules on the survival of Lactobacillus plantarum during simulated gastrointestinal digestion and heat treatment. Mar. Drugs 2024, 22, 308. [Google Scholar] [CrossRef]
  83. Raposo, M.F.; De Morais, A.M.M.B.; De Morais, R.M.S.C. Emergent sources of prebiotics: Seaweeds and microalgae. Mar. Drugs 2016, 14, 27. [Google Scholar] [CrossRef]
  84. Pi, Y.; Fang, M.; Li, Y.; Cai, L.; Han, R.; Sun, W.; Li, X. Gut microbiota in regulating natural bioactive polysaccharides on metabolic diseases: A review. Nutrients 2024, 16, 2838. [Google Scholar] [CrossRef] [PubMed]
  85. Bocanegra, A.; Macho-González, A.; Garcimartín, A.; Benedí, J.; Sánchez-Muniz, F.J. Whole alga, algal extracts, and compounds as ingredients of functional foods: Composition and action mechanism relationships in the prevention and treatment of type-2 diabetes mellitus. Int. J. Mol. Sci. 2021, 22, 3816. [Google Scholar] [CrossRef]
  86. Garg, S.S.; Kushwaha, K.; Dubey, R.; Gupta, J. Association between obesity, inflammation and insulin resistance: Insights into signaling pathways and therapeutic interventions. Diabetes Res. Clin. Pract. 2023, 200, 110691. [Google Scholar] [CrossRef]
  87. Li, Y.; Liu, N.; Wang, X.; Tang, X.; Zhang, L.; Meinita, M.D.N.; Zhang, J. Comparative genomics and systematics of Betaphycus, Eucheuma, and Kappaphycus (Solieriaceae: Rhodophyta) based on mitochondrial genome. J. Appl. Phycol. 2018, 30, 3435–3443. [Google Scholar] [CrossRef]
  88. Zhang, Z.; Wang, F.; Wang, X.; Liu, X.; Hou, Y.; Zhang, Q. Extraction of the polysaccharides from five algae and their potential antioxidant activity in vitro. Carbohydr. Polym. 2010, 82, 118–121. [Google Scholar] [CrossRef]
  89. Fleita, D.; El-Sayed, M.; Rifaat, D. Evaluation of the antioxidant activity of enzymatically-hydrolyzed sulfated polysaccharides extracted from red algae; Pterocladia capillacea. LWT-Food Sci. Technol. 2015, 63, 1236–1244. [Google Scholar] [CrossRef]
  90. Sokolova, E.V.; Barabanova, A.O.; Bogdanovich, R.N.; Khomenko, V.A.; Solov’eva, T.F.; Yermak, I.M. In vitro antioxidant properties of red algal polysaccharides. Biomed. Prev. Nutr. 2011, 1, 161–167. [Google Scholar] [CrossRef]
  91. 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]
  92. Wang, L.; Li, B.; Dionysiou, D.D.; Chen, B.; Yang, J.; Li, J. Overlooked formation of H2O2 during the hydroxyl radical-scavenging process when using alcohols as scavengers. Environ. Sci. Technol. 2022, 56, 3386–3396. [Google Scholar] [CrossRef] [PubMed]
  93. Matsui, M.S.; Muizzuddin, N.; Arad, S.; Marenus, K. Sulfated polysaccharides from red microalgae have antiinflammatory properties in vitro and in vivo. Appl. Biochem. Biotechnol. 2003, 104, 13–22. [Google Scholar] [CrossRef] [PubMed]
  94. Airanthi, M.W.A.; Sasaki, N.; Iwasaki, S.; Baba, N.; Abe, M.; Hosokawa, M.; Miyashita, K. Effect of brown seaweed lipids on fatty acid composition and lipid hydroperoxide levels of mouse liver. J. Agric. Food Chem. 2011, 59, 4156–4163. [Google Scholar] [CrossRef]
  95. Cheng, D.; Liang, B.; Li, M.; Jin, M. Influence of laminarin polysaccahrides on oxidative damage. Int. J. Biol. Macromol. 2011, 48, 63–66. [Google Scholar] [CrossRef]
  96. Xue, S.; Zang, Y.; Chen, J.; Shang, S.; Gao, L.; Tang, X. Ultraviolet-B radiation stress triggers reactive oxygen species and regulates the antioxidant defense and photosynthesis systems of intertidal red algae Neoporphyra haitanensis. Front. Mar. Sci. 2022, 9, 1043462. [Google Scholar] [CrossRef]
  97. BelHadj, S.; Hentati, O.; Elfeki, A.; Hamden, K. Inhibitory activities of Ulva lactuca polysaccharides on digestive enzymes related to diabetes and obesity. Arch. Physiol. Biochem. 2013, 119, 81–87. [Google Scholar] [CrossRef]
  98. Ghannam, A.; Murad, H.; Jazzara, M.; Odeh, A.; Allaf, A.W. Isolation, Structural characterization, and antiproliferative activity of phycocolloids from the red seaweed Laurencia papillosa on MCF-7 human breast cancer cells. Int. J. Biol. Macromol. 2018, 108, 916–926. [Google Scholar] [CrossRef]
  99. Murad, H.; Hawat, M.; Ekhtiar, A.; AlJapawe, A.; Abbas, A.; Darwish, H.; Sbenati, O.; Ghannam, A. Induction of G1-phase cell cycle arrest and apoptosis pathway in MDA-MB-231 human breast cancer cells by sulfated polysaccharide extracted from Laurencia papillosa. Cancer Cell Int. 2016, 16, 39. [Google Scholar] [CrossRef]
  100. Jazzara, M.; Ghannam, A.; Soukkarieh, C.; Murad, H. Anti-proliferative activity of λ-carrageenan through the induction of apoptosis in human breast cancer cells. Iran. J. Cancer Prev. 2016, 9, e3836. [Google Scholar] [CrossRef]
  101. Zhao, C.; Lin, G.; Wu, D.; Liu, D.; You, L.; Högger, P.; Xiao, J. The algal polysaccharide ulvan suppresses growth of hepatoma cells. Food Front. 2020, 1, 83–101. [Google Scholar] [CrossRef]
  102. Alwarsamy, M.; Gooneratne, R.; Ravichandran, R. Effect of fucoidan from Turbinaria conoides on human lung adenocarcinoma epithelial (A549) cells. Carbohydr. Polym. 2016, 152, 207–213. [Google Scholar] [CrossRef] [PubMed]
  103. Chen, P.H.; Chiang, P.C.; Lo, W.C.; Su, C.W.; Wu, C.Y.; Chan, C.H.; Wu, Y.C.; Cheng, H.C.; Deng, W.P.; Lin, H.K.; et al. A novel fucoidan complex-based functional beverage attenuates oral cancer through inducing apoptosis, G2/M cell cycle arrest and retarding cell migration/invasion. J. Funct. Foods 2021, 85, 104665. [Google Scholar] [CrossRef]
  104. Pozharitskaya, O.N.; Obluchinskaya, E.D.; Shikov, A.N. Mechanisms of bioactivities of fucoidan from the brown seaweed Fucus vesiculosus L. of the Barents Sea. Mar. Drugs 2020, 18, 275. [Google Scholar] [CrossRef]
  105. Li, Y.; McGowan, E.; Chen, S.; Santos, J.; Yin, H.; Lin, Y. Immunopotentiating activity of fucoidans and relevance to cancer immunotherapy. Mar. Drugs 2023, 21, 128. [Google Scholar] [CrossRef]
  106. 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]
  107. Xu, H.; Yang, X.; Zhao, Z.; Wang, Y. Research progress and thinking on the metabolism of polysaccharide drugs in vivo and the key technology of PK/PD. Chin. J. Clin. Pharmacol. Ther. 2021, 26, 851. [Google Scholar]
  108. Hsu, H.Y.; Hwang, P.A. Clinical applications of fucoidan in translational medicine for adjuvant cancer therapy. Clin. Transl. Med. 2019, 8, e15. [Google Scholar] [CrossRef]
  109. Kajla, P.; Chaudhary, V.; Dewan, A.; Bangar, S.P.; Ramniwas, S.; Rustagi, S.; Pandiselvam, R. Seaweed-based biopolymers for food packaging: A sustainable approach for a cleaner tomorrow. Int. J. Biol. Macromol. 2024, 274, 133166. [Google Scholar] [CrossRef]
  110. Roy, S.; Chawla, R.; Santhosh, R.; Thakur, R.; Sarkar, P.; Zhang, W. Agar-based edible films and food packaging application: A comprehensive review. Trends Food Sci. Technol. 2023, 141, 104198. [Google Scholar] [CrossRef]
  111. Cheng, C.; Chen, S.; Su, J.; Zhu, M.; Zhou, M.; Chen, T.; Han, Y. Recent advances in carrageenan-based films for food packaging applications. Front. Nutr. 2022, 9, 1004588. [Google Scholar] [CrossRef] [PubMed]
  112. Teixeira, D.C.; Kokojka, F.; Kolukuluri, R.S.; Marks, R.S.; Naik, A.S. Deep dive into seaweed-based preservatives for sustainable production and consumption of meat products. Food Rev. Int. 2024, 41, 1223–1258. [Google Scholar] [CrossRef]
  113. Cho, T.J.; Rhee, M.S. Health functionality and quality control of laver (Porphyra, Pyropia): Current issues and future perspectives as an edible seaweed. Mar. Drugs 2019, 18, 14. [Google Scholar] [CrossRef]
  114. Zongrui, T.; Yu, C.; Wei, L. Grafting derivate from alginate. In Biopolymer Grafting; Elsevier: Amsterdam, The Netherlands, 2018; pp. 115–173. [Google Scholar] [CrossRef]
  115. Yang, J.S.; Xie, Y.J.; He, W. Research progress on chemical modification of alginate: A review. Carbohydr. Polym. 2011, 84, 33–39. [Google Scholar] [CrossRef]
  116. Nair, M.S.; Tomar, M.; Punia, S.; Kukula-Koch, W.; Kumar, M. Enhancing the functionality of chitosan-and alginate-based active edible coatings/films for the preservation of fruits and vegetables: A review. Int. J. Biol. Macromol. 2020, 164, 304–320. [Google Scholar] [CrossRef]
  117. Senturk Parreidt, T.; Müller, K.; Schmid, M. Alginate-based edible films and coatings for food packaging applications. Foods 2018, 7, 170. [Google Scholar] [CrossRef]
  118. Perera, K.Y.; Sharma, S.; Pradhan, D.; Jaiswal, A.K.; Jaiswal, S. Seaweed polysaccharide in food contact materials (active packaging, intelligent packaging, edible films, and coatings). Foods 2021, 10, 2088. [Google Scholar] [CrossRef]
  119. Mohamed, S.A.; El-Sakhawy, M.; El-Sakhawy, M.A.M. Polysaccharides, protein and lipid-based natural edible films in food packaging: A review. Carbohydr. Polym. 2020, 238, 116178. [Google Scholar] [CrossRef]
  120. Surendhiran, D.; Li, C.; Cui, H.; Lin, L. Marine algae as efficacious bioresources housing antimicrobial compounds for preserving foods-A review. Int. J. Food Microbiol. 2021, 358, 109416. [Google Scholar] [CrossRef]
  121. Deshmukh, R.K.; Gaikwad, K.K. Natural antimicrobial and antioxidant compounds for active food packaging applications. Biomass Convers. Biorefinery 2024, 14, 4419–4440. [Google Scholar] [CrossRef]
  122. Chang, B.P.; Trinh, B.M.; Tadele, D.T.; Bandara, N.; Mekonnen, T.H. Natural antioxidant and antimicrobial agents and processing technologies for the design of active food packaging polymers. Polym. Rev. 2023, 63, 961–1013. [Google Scholar] [CrossRef]
  123. Zhang, S.; Qamar, S.A.; Junaid, M.; Munir, B.; Badar, Q.; Bilal, M. Algal polysaccharides based nanoparticles for targeted drug delivery applications. Starch Stärke 2022, 74, 2200014. [Google Scholar] [CrossRef]
  124. Kilincceker, O.; Dogan, İ.S.; Kucukoner, E. Effect of edible coatings on the quality of frozen fish fillets. LWT-Food Sci. Technol. 2009, 42, 868–873. [Google Scholar] [CrossRef]
  125. Wang, S.; Qi, H.; Xiang, Z. Research progress in applications of amphiphilic algal polysaccharides in emulsification and dispersion. Biomass Chem. Eng. 2022, 56, 37–46. [Google Scholar] [CrossRef]
  126. Carpintero, M.; Marcet, I.; Rendueles, M.; Díaz, M. Algae as an additive to improve the functional and mechanical properties of protein and polysaccharide-based films and coatings. A review of recent studies. Food Packag. Shelf Life 2023, 38, 101128. [Google Scholar] [CrossRef]
  127. Zhang, B.; Liu, Y.; Wang, H.; Liu, W.; Cheong, K.L.; Teng, B. Effect of sodium alginate-agar coating containing ginger essential oil on the shelf life and quality of beef. Food Control 2021, 130, 108216. [Google Scholar] [CrossRef]
  128. Frent, O.D.; Vicas, L.G.; Duteanu, N.; Morgovan, C.M.; Jurca, T.; Pallag, A.; Marian, E. Sodium alginate—Natural microencapsulation material of polymeric microparticles. Int. J. Mol. Sci. 2022, 23, 12108. [Google Scholar] [CrossRef]
  129. Garzón, A.G.; Cian, R.E.; Drago, S.R. Effects of agar-carrageenan wall materials and core-to-wall material ratio on physicochemical properties and in vitro bioaccessibility of microencapsulated bioactive peptides. Food Hydrocoll. 2023, 139, 108570. [Google Scholar] [CrossRef]
  130. Djihad, N.; Oukil, N.F.; Hamid, S.; Attia, A.; Petronilho, S. Microencapsulation of a green microalga (Chlorella vulgaris) by complex coacervation for its valuation as a feasible ingredient in pear snacks. Algal Res. 2024, 83, 103727. [Google Scholar] [CrossRef]
  131. Lee, I.C.; Caggianiello, G.; van Swam, I.I.; Taverne, N.; Meijerink, M.; Bron, P.A.; Spano, G.; Kleerebezem, M. Strain-specific features of extracellular polysaccharides and their impact on Lactobacillus plantarum-host interactions. Appl. Environ. Microbiol. 2016, 82, 3959–3970. [Google Scholar] [CrossRef]
  132. El-Deeb, N.M.; Ibrahim, O.M.; Mohamed, M.A.; Farag, M.M.; Farrag, A.A.; El-Aassar, M.R. Alginate/κ-carrageenan oral microcapsules loaded with Agaricus bisporus polysaccharides MH751906 for natural killer cells mediated colon cancer immunotherapy. Int. J. Biol. Macromol. 2022, 205, 385–395. [Google Scholar] [CrossRef] [PubMed]
  133. Huang, L.; Lee, J.Y.; Park, Y.K.; Lee, J. Heavy metals in seaweed: Implications for health benefits, risks, and safety regulations. J. Agric. Food Res. 2025, 21, 101830. [Google Scholar] [CrossRef]
  134. Picardo, M.; Filatova, D.; Nunez, O.; Farré, M. Recent advances in the detection of natural toxins in freshwater environments. TrAC Trends Anal. Chem. 2019, 112, 75–86. [Google Scholar] [CrossRef]
Figure 1. Global distribution of publications on seaweed. The number of the literature is based on the ISI Web of Science search engine search using the seaweed topics.
Figure 1. Global distribution of publications on seaweed. The number of the literature is based on the ISI Web of Science search engine search using the seaweed topics.
Futurepharmacol 05 00029 g001
Figure 2. The common structural formulas of algal polysaccharides. (a) Structures of different backbones of fucoidans; (b) structure of alginate; (c) structure of agarose; (d) structure of laminarin; (e) structure of the main repeating disaccharides found in ulvan, ulvanobiuronic acids A3S and B3S, and ulvanobioses U3S and U2′S, 3S; (f) structure of three typical carrageenens, including λ-carrageenan, κ-carrageenan, and ι-carrageenan.
Figure 2. The common structural formulas of algal polysaccharides. (a) Structures of different backbones of fucoidans; (b) structure of alginate; (c) structure of agarose; (d) structure of laminarin; (e) structure of the main repeating disaccharides found in ulvan, ulvanobiuronic acids A3S and B3S, and ulvanobioses U3S and U2′S, 3S; (f) structure of three typical carrageenens, including λ-carrageenan, κ-carrageenan, and ι-carrageenan.
Futurepharmacol 05 00029 g002
Figure 3. Algal polysaccharides regulate the intestinal flora. Figure created with Figdraw (https://www.figdraw.com). Solid arrows pointing upwards (↑) denote increase, upregulation, activation, or promotion. Solid arrows pointing downwards (↓) denote decrease, downregulation, inhibition, or suppression. Dashed Lines indicate indirect effects, potential pathways, signaling to distal sites, or modulation.
Figure 3. Algal polysaccharides regulate the intestinal flora. Figure created with Figdraw (https://www.figdraw.com). Solid arrows pointing upwards (↑) denote increase, upregulation, activation, or promotion. Solid arrows pointing downwards (↓) denote decrease, downregulation, inhibition, or suppression. Dashed Lines indicate indirect effects, potential pathways, signaling to distal sites, or modulation.
Futurepharmacol 05 00029 g003
Figure 4. Applications of algal polysaccharides in food packaging. Figure created with Biorender (https://biorender.com/).
Figure 4. Applications of algal polysaccharides in food packaging. Figure created with Biorender (https://biorender.com/).
Futurepharmacol 05 00029 g004
Table 1. Extraction methods for algal polysaccharides.
Table 1. Extraction methods for algal polysaccharides.
Extraction MethodAdvantagesDisadvantagesReferences
Acid extractionHigh polysaccharide yield with short processing time.Breaking glycosidic bonds[12]
Calcium chloride extractionPolysaccharides dissolve completely, high yield, short amount of time.With more impurities, polysaccharides may degrade, loss of activity.[13]
Enzyme extractionMild, the time is short, environmentally friendly with no pollution.High requirements for technology, equipment, small-scale production.[14]
Hot water extractionStable, economically convenient, easy to operate, maintain molecular structure well.High temperature and low yield.[15]
Alcohol extractionShort time consumption and low loss of activityHigh cost, complex operation, and low yield.[16]
Microwave-assisted extractionHigh efficiency and rapidity, less solvent use.More impurities, inconsistent results.[17,18]
Ultrasound-assisted extractionSimple, faster extraction, higher yields, and shorter costs and processing timesHigher ultrasonic power may lead to chemical decomposition of polysaccharides[19,20]
Pressurized liquid extractionShort time and use less solvent. Sample dissolution more fully, extraction rate is high.May affect its functional properties.[21,22]
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

Wu, X.; Guo, Y.; Dai, C.; Zhao, C. The Pharmacological Potential of Algal Polysaccharides in Food Applications and Chronic Disease Management. Future Pharmacol. 2025, 5, 29. https://doi.org/10.3390/futurepharmacol5020029

AMA Style

Wu X, Guo Y, Dai C, Zhao C. The Pharmacological Potential of Algal Polysaccharides in Food Applications and Chronic Disease Management. Future Pharmacology. 2025; 5(2):29. https://doi.org/10.3390/futurepharmacol5020029

Chicago/Turabian Style

Wu, Xue, Yuxin Guo, Congjie Dai, and Chao Zhao. 2025. "The Pharmacological Potential of Algal Polysaccharides in Food Applications and Chronic Disease Management" Future Pharmacology 5, no. 2: 29. https://doi.org/10.3390/futurepharmacol5020029

APA Style

Wu, X., Guo, Y., Dai, C., & Zhao, C. (2025). The Pharmacological Potential of Algal Polysaccharides in Food Applications and Chronic Disease Management. Future Pharmacology, 5(2), 29. https://doi.org/10.3390/futurepharmacol5020029

Article Metrics

Back to TopTop