Next Article in Journal
Influence of Super Absorbent Polymer on Root Characteristics and Anchorage of Amorpha fruticosa on Rocky Slope
Previous Article in Journal
Style Recognition of Door God Paintings by Hypothesis Testing for Texture Features of Painting Patterns
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Seaweed Exhibits Therapeutic Properties against Chronic Diseases: An Overview

by
Maria Dyah Nur Meinita
1,2,3,*,
Dicky Harwanto
1,4 and
Jae-Suk Choi
1,5,*
1
Seafood Research Center, Industry Academy Cooperation Foundation (IACF), Silla University, 606, Advanced Seafood Processing Complex, Wonyang-ro, Amnam-dong, Seo-gu, Busan 49277, Korea
2
Faculty of Fisheries and Marine Science, Jenderal Soedirman University, Purwokerto 53123, Indonesia
3
Center for Maritime Bioscience Studies, Jenderal Soedirman University, Purwokerto 53123, Indonesia
4
Faculty of Fisheries and Marine Science, Diponegoro University, Semarang 50275, Indonesia
5
Department of Food Biotechnology, College of Medical and Life Sciences, Silla University, 140, Baegyang-Daero 700 Beon-Gil, Sasang-gu, Busan 46958, Korea
*
Authors to whom correspondence should be addressed.
Appl. Sci. 2022, 12(5), 2638; https://doi.org/10.3390/app12052638
Submission received: 6 December 2021 / Revised: 24 February 2022 / Accepted: 1 March 2022 / Published: 3 March 2022
(This article belongs to the Section Food Science and Technology)

Abstract

:
Seaweeds or marine macroalgae are known for producing potentially bioactive substances that exhibit a wide range of nutritional, therapeutic, and nutraceutical properties. These compounds can be applied to treat chronic diseases, such as cancer, cardiovascular disease, osteoporosis, neurodegenerative diseases, and diabetes mellitus. Several studies have shown that consumption of seaweeds in Asian countries, such as Japan and Korea, has been correlated with a lower incidence of chronic diseases. In this study, we conducted a review of published papers on seaweed consumption and chronic diseases. We used the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) method for this study. We identified and screened research articles published between 2000 and 2021. We used PubMed and ScienceDirect databases and identified 107 articles. This systematic review discusses the potential use of bioactive compounds of seaweed to treat chronic diseases and identifies gaps where further research in this field is needed. In this review, the therapeutic and nutraceutical properties of seaweed for the treatment of chronic diseases such as neurodegenerative diseases, obesity, diabetes, cancer, liver disease, cardiovascular disease, osteoporosis, and arthritis were discussed. We concluded that further study on the identification of bioactive compounds of seaweed, and further study at a clinical level, are needed.

1. Introduction

Numerous scientific articles have been published on functional foods with putative beneficial health properties. The term “functional food” was coined as a result of widespread interest in specific foods that may promote health [1]. These properties are due to the ability of a food item to lower the risk of chronic diseases and help manage such diseases, thereby improving quality of life [1,2]. According to a global analysis of new functional food categories, products focused on digestive health capture the interest of a broader audience than products with a narrow focus, such as products targeting specific illnesses [3].
Seaweeds or macroalgae are regarded as one of the non-animal foods of the future based on their ability to grow without the need for arable land or freshwater resources, thereby avoiding competition with traditional crops [4,5,6]. Currently, seaweeds are particularly appealing because of their high nutrient and bioactive phytochemical content [7]. The active components of seaweed, such as sulfated polysaccharides [8], polyphenols [9], fucosterol [10], fucoxanthin [11], fucoidan [12], phlorotannin [13], and flavonoids [14], may lead to the development of novel drugs and functional foods, or nutraceuticals that may be used as a natural alternative to commercial synthetic drugs for certain chronic diseases [15].
In recent years, the food industry has increased the development and marketing of a wide range of functional food products using diverse food sources [16]. Functional food is defined as food containing one or more ingredients that provide health benefits in addition to energy and nutrition [17]. Conventional foods containing bioactive components can also be promoted as having positive health benefits. Some may be fortified or enhanced foods, created specifically to reduce disease risk in a specific group of people [3]. Seaweed has been consumed as a food in Asian countries since ancient times, particularly in China, Japan, and Korea [15]. In China, since 600 BC, seaweed has been served to honored guests, even to the King himself [18]. In Japan, Sargassum fusiforme (Hijiki), Eisenia bicyclis (Arame), Saccharina japonica (Kombu or Haidai), Undaria pinnatifida (Wakame or Quandai-cai), Ulva pertusa (ao-nori) and Porphyra sp. (nori) have been employed in ordinary cooking since the 8th century. As people from these countries have migrated around the world, this custom has moved with them, so that today there are more countries where seaweed is commonly consumed [19]. Coastal residents in South Asian countries, such as Indonesia, Malaysia, Philippines, Vietnam, and Thailand also eat fresh Gracilaria or Caulerpa seaweed, especially as an ingredient in salads. Some kelp species, such as Alaria esculenta and Himantalia elongate, are also consumed in European countries.
Seaweed has also been used in traditional Eastern medicine owing to its beneficial health effects [20]. In Western countries, seaweed has mostly been used in the food, cosmetics, and pharmaceutical industries as a source of functional polysaccharides (e.g., agar, carrageenans, and alginates) [21,22]. However, edible seaweeds are increasingly being consumed in Europe, especially in France; in the United States and South American countries, they are considered to be novel functional foods [15,21]. Several studies have revealed that dietary habits and traditional culture in consuming seaweed in Asian countries, especially in Japan and Korea, are correlated with a lower incidence of chronic diseases such as cancer, cardiovascular disease, hypertension, osteoporosis, and obesity [6,23,24,25,26,27]. Several studies have also revealed that consumption of seaweed can reduce the occurrence of chronic pathologies, including neurodegenerative diseases [28], cardiovascular disease [29], obesity [30], diabetes [14], cancer [31], liver disease [32], osteoporosis [33], and arthritis [34]. Furthermore, collaborative cohort studies have been conducted to investigate the effect of the seaweed diet in Japan and Korea, and the results of these studies showed that seaweed-containing diets are related to protection against cancer (Iso and Kubota 2007; Kim et al. 2020c). Thus, seaweed consumption appears to have an important impact on the occurrence of chronic diseases. The aim of this study is to review the potential of seaweed and seaweed-derived bioactive compounds to prevent and treat chronic diseases. This comprehensive review collected the available data from published papers to identify gaps where further research is required.

2. Materials and Methods

This study followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) method [35]. The literature search was conducted using PubMed and ScienceDirect as online databases for collecting and screening the research articles that met the criteria. The criteria included article type, year of publication, language used, and topic. We screened the research articles that used English, were published between 2000 and 2021, and where the topic met our scope. To screen the topic, we used specific keywords which were found in the titles, abstracts, or text content. The keywords “seaweed” and “macroalgae” were combined with other keywords such as nutritional, bioactive compound, chronic disease, neurodegenerative disease, cardiovascular disease, obesity, diabetes, cancer, liver disease, osteoporosis, and arthritis. Articles that met the requirements were analyzed, extracted, and reviewed.

3. Results

A systematic review of the published studies yielded 213 articles. Then, through identification, screening, eligibility, and inclusion, 107 articles that fulfilled the review criteria were selected. Previous studies have observed that the use of seaweed modulates several common chronic diseases, including diabetes, neurodegenerative diseases, obesity, cancer, liver disease, cardiovascular disease, osteoporosis, and arthritis (Figure 1). Most of the studies were conducted using in vitro (39.4%) and in vivo (36.4%) models, and only 2.3% of the studies that we found were clinical studies.
We summarized seaweed research on chronic disease based on three groups of seaweeds: brown (Phaeophyceae), red (Rhodophyceae), and green (Chlorophyceae) (Figure 2). Studies of chronic disease were conducted on brown seaweed (68%), red seaweed (18%), and green seaweed (14%). Among the brown seaweeds, the most studied species were Ecklonia (21.3%), Sargassum (20.2%), and Fucus (9%). In red seaweed, Gracilaria (20.8%) and Gelidium (16.7%) were the top two most studied species for potential use in the treatment of chronic diseases. The most studied green seaweed species were Ulva (47.4%), Codium (26.3%), and Caulerpa (15.8%). The phylum of Phaeophyta was the most studied species on cancer (40%), diabetes (85%), arthritis (67%), neurodegenerative diseases (71%), obesity (59%), osteoporosis (46%), liver disease (80%) and cardiovascular disease (84%). The studies of seaweed in treating chronic diseases were conducted in vivo, in vitro, in silico and as clinical studies. There was a lack of clinical studies conducted; however, a clinical study was conducted to test a Maritech® seaweed extract formulation containing extract of Fucus vesiculosis, Macrocystis pyrifera, and Laminaria japonica on osteoarthritis patients [36]. The study showed that seaweed extract can reduce the symptoms of osteoarthritis in a dose-dependent manner.

4. Nutritional Value of Seaweed

Seaweed contains a variety of important macro- and micronutrients, including proteins, carbohydrates, phenols, vitamins, and minerals [37,38]. However, the biochemical composition of seaweeds is affected by the species, time of collection, and habitat, as well as by external factors such as temperature, light intensity, and nutrient concentration in water. For example, a study by Garcia-Vaquero et al. [37] reported that the proximate composition of dry matter of Laminaria digitata, Laminaria hyperborea, and Ascophyllum nodosum was highest in the autumn period. While the study conducted by Khairy and El-Shafay [39] showed that the protein, carbohydrate, lipid, ash, fatty acid and amino acid content of Ulva lactuca, Jania rubens, and Pterocladia capillacea varied throghout different seasons. Physcio–chemical parameters also influence the biochemical composition of red seaweed Catenella repens [40].

4.1. Carbohydrates

In seaweed, the carbohydrate content is high, the majority of which is dietary fiber and is not absorbed by the human body [1]. The carbohydrate of seaweed is different from the carbohydrate of land-plants. In seaweed, the storage of carbohydrates has an important function in photosynthesis and osmoregulation. The type and amount of carbohydrate content in seaweed also vary based on many factors such as the type of seaweed, and physical, chemical, and biological factors. Most of the red seaweeds contain sulfated galactan (agar and carrageenan), and some of the brown seaweeds contain sulfated fucans and alginates, while cellulose has commonly been found in large amounts in green seaweed [41].
Carbohydrates in seaweed are found in large amounts, including monosaccharides, disaccharides, and polysaccharides; however, their composition depends on many factors. The components of monosaccharides, such as galactose, glucose, mannose, xylose, fructose, fucose, and arabinose, were found in the total sugars of seaweed [42,43]. The most representative polysaccharides in seaweed are agar, alginates, ulvan, and carrageenans [44,45]. Total polysaccharide content ranges from 4% to 76% of dry weight (dw), with the highest content found in the genera Ascophyllum, Porphyra, and Palmaria. In general, green seaweed genera such as Ulva also have a high content, which can be up to 65% of dw [1]. For example, the carbohydrate contents of Ulva rigida and Ulva pertusa are 58% dw and 56% dw, respectively [4,46]. Other studies reported high carbohydrate content in Gracilaria fisheri and Gracilaria tenuistipitata, of 63% and 59% dw, respectively [47]. For brown seaweed, Hizikia fusiforme exhibited a very high amount of carbohydrates, with a value of 72% dw [48].

4.2. Proteins and Amino Acids

Among seaweeds, red seaweed contains the highest protein content, which ranges from 0.67% to 45.0% dw, followed by green seaweed (3.42–29.80% dw) and brown seaweed (5.02–19.66% dw) [49]. Specifically, the protein content of red seaweed varies, with values of 9.32% dw for Gelidium latifolium, 15.58% dw for Gracilaria verrucosa [50], and 26.69% dw for Plocamium telfairiae [51]. Green seaweeds such as Caulerpa lentillifera, Ulva rigida, and Ulva pertusa contain protein contents of 7%, 9.3%, and 21.5% dw, respectively [4,46]. The protein contents of brown seaweeds Hizikia fusiforme and Fucus vesiculosus were reported to be 12.2% [52] and 15.1% dw [4], respectively. The protein content of seaweed also depends on the season. For example, Palmaria palmata (Dulse) collected on the French Atlantic coast during winter and spring, exhibited a higher protein content (21.9% dw) than dusring the summer and autumn months (11.9% dw), with essential amino acids constituting 26% to 50% dw of the protein [53]. Several studies have examined the amino acid composition of seaweeds, and aspartic acid, glutamic acid, taurine, threonine, arginine, and alanine have been reported to be high in concentration. To compare, the concentrations of aspartic acid and threonine in Ulva armoricana, Ulva Pertusa, Palmaria palmata, and Porphyra tenra were found to be much higher than in leguminous plants and ovalbumin [54]. Finally, glutamic and aspartic acids seem to be the most abundant amino acids in most seaweeds [55]. The reference nutrient intake (RNI) for protein is a 5-g portion equal to a maximum of 1.97%, 4.5%, and 2.98% dw from brown, red, and green seaweed, respectively. The digestibility of protein in edible seaweed species ranged from 14.7% to 86.2% dw, with Porphyra tenera showing the highest value [49].

4.3. Lipids and Fatty Acids

Generally, total lipids found in seaweed range between 0.3% and 7.0% dw [15]. A previous study reported that Undaria pinnatifida contains lipids comprising 3.71% dw [56]. In contrast, the lipid content of Ulva rigida, Gracilaria sp., Fucus vesiculosus, and Saccharina latissima ranged from 0.4% to 2.8% dw [4].
Polyunsaturated fatty acids (PUFAs) are common lipids found in seaweed, and are of great interest due to their biological activity. In seaweeds, PUFAs contain a substantial amount of ω-3 fatty acids for almost half of the total lipid content [57]. Seaweed also contains sterol, an important lipid of various types. Brown seaweeds primarily contain fucosterol, while cholesterol is the predominant sterol type in red seaweeds [3,58,59]. Taken together, many essential fatty acids found in seaweeds could be combined to increase their efficacy as a dietary supplement or as part of a well-balanced diet.

4.4. Ash

The ash content of seaweed is quite high compared to common land plants, ranging from 8.0% to 48.68% dw [15,50]. However, this amount depends on several factors. The ash content of Saccharina latissima, Laminaria spp., and Alaria esculenta was lowest in September, October, and November, and highest in spring (February to June); however, it only varied slightly throughout the year. For these species, total dry weight was lowest from January to March and highest from July to September [1]. Peñalver and colleagues [60] reviewed natural products from many seaweed species and demonstrated that brown seaweed contains more ash in general. Moreover, proximate analysis on red seaweed found an average value for ash content of 22.9 ± 10.99 g/100 g, which is much higher than that in terrestrial plants [61]. These results were confirmed in red seaweed Gelidium elegans, containing 24.1% ash dw [62], and the total ash of Gracilaria verrucosa reached as high as 48.68% dw [50].

4.5. Moisture

The moisture of seaweed is influenced by many factors, especially postharvest treatment. Drying method is the main factor that might influence the moisture of seaweed. Some fresh seaweeds have a moisture content of 80–90%, whereas seaweeds that are dried by air have a moisture level of 10–20% [21,63]. A nutritional analysis of selected Azorean macroalgae (Ulva compressa, Ulva rigida, Gelidium microdon, and Pterocladiella capillacea) reported that moisture content varied between 83.2% and 90.0% [15]. Holdt and Kraan [1] reported that water content was lower in some species, but not less than 68.0% dw.

4.6. Dietary Fiber

Non-digestible carbohydrates and lignin are considered dietary fibers [64]. Unlike sugar or starch, dietary fiber cannot be directly digested by digestive enzymes. In seaweed, dietary fiber consists mostly of carrageenan and agar (red seaweed), alginate (brown seaweed), and ulvans (green seaweed), which represent 25% to 75% dw [65]. According to their solubility in water, fibers are classified as soluble dietary fiber (SDF) or insoluble dietary fiber (IDF). Seaweed dietary fiber content is similar to or higher than that of terrestrial plants. The SDF content is typically higher in red seaweeds, such as Chondrus and Porphyra (15–22% dw). The brown seaweeds Fucus and Laminaria/Saccharina have higher IDF contents (27–40% dw) [60]. Research by Neto et al. [4] demonstrated that among four seaweeds, the brown seaweed Fucus vesiculosus contains the highest total fiber, with a value of 45% dw. Dietary fiber intake for Asians is recommended to be met by consuming 8 g of seaweed, which can meet up to 12.5% of daily requirements [66]. In addition, dietary fiber can generally retain oil and water, which is beneficial for promoting digestive health.

4.7. Minerals and Vitamins

Seaweeds are rich sources of important minerals and vitamins. However, amounts vary according to seaweed species, phylum, season, and environmental, geographical, and physiological factors. In particular, seaweeds contain substantial amounts of calcium (Ca), magnesium (Mg), sodium (Na), phosphorus (P), iodine (I), zinc (Zn), and iron (Fe), at much higher levels than in terrestrial plants. Ca and Mg were found to be the major minerals in seaweed. A study of several seaweeds from the Kenyan Coast demonstrated that brown seaweed (Hypnea musciformis and Sargassum oligocystum) and red seaweed (Laurencia intermedia) contain high amounts of Ca and Mg [67]. Calcium phosphate is more bioavailable in seaweeds than calcium carbonate, which is found in milk [57]. MacArtain and colleagues [66] demonstrated that 8 g of Ulva lactuca (sea lettuce), which provides 260 mg of Ca, provides approximately 37% of the RNI of Ca for an adult male, while cheddar cheese only provides 5% of the RNI in the same portion. Furthermore, seaweed also contains one of the best natural sources of P, which is beneficial in the human diet. According to the World Health Organization (WHO) and Food and Agriculture Organization (FAO), the dietary reference value of P for adults (>13 years) is 700 µg/day [7]. Brown algae have been identified as important sources of I for the prevention and treatment of iodine deficiency goiters. The daily intake of I for adults is 150 µg/day, and excessive consumption should be avoided [7]. Ca and P are essential for heart and smooth muscle contraction, as well as for the skeleton. Mg is an important cofactor of many enzymes, including those involved in cellular respiration, while Na is responsible for maintaining the body’s water and electrolyte balance. A high intake of Ca, Na, and K is linked to a lower mean systolic pressure and a lower risk of hypertension [7].
Edible seaweeds are also a valuable source of vitamin content, particularly water-soluble vitamin C and B complexes and the fat-soluble vitamins A and E [21]. A study on several red seaweeds showed that, in general, the concentration of vitamin C was higher than that of vitamin A and vitamin E [61]. Research by Rajapakse and Kim [57] reported that red and brown algae are rich in vitamin C and A. Undaria pinnatifida and Porphyra umbilicalis are rich in B vitamins [7]. Vitamins A, B, C, and E are important antioxidants produced by seaweeds. Vitamin A is found in large amounts in Fucus spiralis (1.41 mg/100 g of dry weight), Osmundea pinnatifida (1.20 mg/100 g of dry weight), and Porphyra/Pyropia spp. (1.27 mg/100 g of dry weight). Vitamin C is present in large amounts in Ulva lactuca (10 mg/8 g of dry weight) and Undaria pinnatifida (14 mg/100 g of dry weight). Vitamin B12 is abundant in species such as Ulva spp. and Porphyra/Pyropia spp., with a recommended dietary allowance (RDA) of 2.5 mg/day for adults [66]. These findings indicate that seaweed minerals and vitamins have a greater potential to be exploited in high-demand functional food categories.

5. Bioactive Compounds in Seaweed

Seaweed are potential source of bioactive compounds including fucoidan, agars, kappa carrageenans, ulvans, alginates, laminarin, fucosterol, phlorotannins, lectins, alkaloids, diterpenes, and fucoxanthin (Figure 3).

5.1. Fucoidan

Fucoidan is a high-molecular-weight polysaccharide found in brown seaweeds, such as Fucus serratus, Ascophyllum nodosum, and Undaria pinnatifida [68]. Fucoidan is made up of two chains: one with (1→3)- α-L-fucopyranose as the main chain and one with α-L-fucopyranose linked by (1→3) and (1→4) as the main chain. Single and double substitutions in the sulfate groups of both skeletons can occur at the C-2 or C-4 positions. Some fucoidans have substituted branches at positions C-2 and C-3 [69]. Fucoidans are the most abundant polysaccharides in brown seaweeds, followed by alginates and laminarans; however, their contents vary depending on the species, geographical area, harvest season, and environmental factors such as salinity and nutrients [70]. Fucoidan is a large part of the cell wall comprised of carbohydrate and sulfate content. For example, Hizikia fusiformis contains 99% dw fucoidan [48], whereas Ascophyllum nodosum contains 84.6% dw fucoidan [71]. Several studies have reported numerous pharmacological properties of fucoidans such as antioxidant, anti-inflammatory, anticoagulant, antimicrobial, anticancer, immunomodulatory, and hepatoprotective activities [72,73,74,75,76,77].

5.2. Agars

Agar is a linear polysaccharide derived from the cell walls of red seaweeds Gelidium and Gracilaria [78]. Agar is composed of alternating 3,6-anhydro-L-galactose and D-galactose units linked by α-(1,3) and β-(1,4) glycosidic bonds [79], and is popular as a phycocolloid consisting mainly of agarose and agaropectin units [78]. Removal of the agaropectin component from agar yields agarose [80]. The quality of the agar is determined by the type, pattern, and degree of substitution, as well as molecular weight, chemical composition (pyruvate, methoxyl, and sulfate), and physical properties (gel strength, gel syneresis, viscosity, gel temperature, and melting temperature), which determine its market value [81]. Agar is generally used as an additive in the food industry because of its gelling ability, and agar and its derivatives have received increasing attention for therapeutic purposes, apart from their rheological properties [80].

5.3. Carrageenans

Carrageenan is a hydrocolloid consisting of sulfated galactans with alternating (1-4)-anhydro-D-galactose and (1-3)-D-galactose backbones isolated from red seaweeds (Rhodophyceae) [82]. Carrageenan is generally considered safe for routine use as a gelling agent and thickener in foods [83]. Based on their chemical structure and properties, carrageenans are divided into kappa (κ), iota (ι), and lambda (λ), which have one, two, and three sulfate groups per disaccharide unit, respectively [83,84]. Kappa carrageenans form the strongest gels of any carrageenan, which makes them useful in the food, dairy, and pharmaceutical industries as thickeners, gelling agents, and stabilizers [85]. Kappaphycus alvarezii and Eucheuma denticulatum [84] are the most important commercially cultivated warm-water carrageenan species, producing kappa- and iota-carrageenan, respectively. These seaweeds are mainly grown commercially in Indonesia, the Philippines, Malaysia, Brazil, and Tanzania [86].

5.4. Ulvans

Ulvans are highly contentious sulphated polyelectrolytes with gelling properties that can be extracted from green seaweeds, which are mainly composed of rhamnose (5.0–92.2 mol%), glucuronic acid (2.6–52.0 mol%), iduronic acid (0.6–15.3 mol%), and xylose (0.0–38.0 mol%) as the main monomer sugars [87]. Ulvans also contain a common constituting disaccharide, such as aldobiuronic acid, (1→4)-D-glucuronic acid-(1→4)- L-rhamnose-3-sulfate- (1→], and iduronic acid [45]. The average molecular weight of ulvans ranges from 189 to 8.200 kDa [88]. Ulva cell-wall polysaccharides account for 38% to 54% of dry algal matter [88]. Ulvans are mainly found in Ulva sp. with high amounts of water-soluble ulvan and insoluble cellulose, as well as a minor amount of peculiar alkali-soluble linear xyloglucan and glucuronan [42]. In this regard, ulvan exhibits potent applications in biomaterial science (wound dressings, biofilm prevention, excipients, and tissue engineering), pharmaceuticals (antiviral, antioxidant, antihyperlipidemic, anticancer, anticoagulant, and immunostimulatory), functional foods, and agriculture [65,87].

5.5. Alginates

Alginates, which are primarily found in brown algae, are linear polysaccharides with varying mannuronic and glucuronic acid ratios [65]. This ratio varies between brown algae species, and can be determined using proton nuclear magnetic resonance (1H NMR). The monomers in alginate, b-d-mannuronic acid (M) and its C-5 epimer α-l-guluronic acid (G), can be arranged in varying proportions to form a chain bound by 1→4 linkages [42]. The M and G residues are organized into blocks of consecutive M (M-blocks), consecutive G (G-blocks), or alternating M and G residues (MG-blocks). Alginate is a phycocolloid that can be dissolved in water to produce a gel with specific rheological properties. Ascophyllum, Laminaria, and Mycrocystis are the most common commercial sources of phaeophytes for alginates, and Sargassum, Durvillea, Eklonia, Lessonia, and Turbinaria as minor sources [89]. Alginate has also been extensively researched as a biomaterial in biomedical science because of its biocompatibility, low toxicity, and easy availability [80]. The pharmacological properties of alginate as immunomodulatory, antioxidant, and anticoagulant agents have been developed in recent years [70].

5.6. Laminaran

Laminaran is a glucan, built up from a homopolymer of β-d-glucose (β-glucan) linked by a 1→3 glycosidic linkage (in some cases, having 1→6 linkages that form a branch and ramifications in the O-6 position), which may contain a mannitol unit or a few uronic acid residues at their reducing end [70]. The average molecular weight of laminaran is 5 kDa [42]. Laminaran can be found in the fronds of Laminaria/Saccharina and, to a lesser extent, Ascophyllum, Fucus, and Undaria. The content varies seasonally and by habitat, but it can reach up to 32% of the dry weight [45]. Laminaran does not gel or form viscous solutions, and its main application may be in medical and pharmaceutical applications.

5.7. Phytosterols

Seaweeds contain large amounts of phytosterols, such as fucosterol, which is the main sterol in brown algae and cholesterol in red algae; however, the sterol composition of green algae is relatively heterogeneous, with a complex mixture of 28-isofucosterol, ergosterol, β-sitosterol, poriferasterol, cholesterol, and others [90,91]. These compounds may have particular biological activities, including antioxidant, antidiabetic, anti-inflammatory, anticancer, hepatoprotective, and anti-Alzheimer’s disease activity [92]. However, accurate determination of the biological activity of individual phytosterols is currently difficult because of the high cost and scarcity of pure phytosterols [93]. Fucosterol and 24-methylenecholesterol are sterols found in brown algae such as Sargassum fusiforme and Undaria pinnatifida, which have been linked to a variety of health benefits in humans [94]. A feasible, economical, and efficient technique for the rapid extraction of phytosterol may be conducted using microwave-assisted extraction coupled with high-speed counter-current chromatography [95].

5.8. Phlorotannins

Marine algae contain bioactive polyphenolic molecules that can modulate biological properties, such as phlorotannins. Phlorotannins are tannin derivatives composed of phloroglucinol-based phenolics (1,3,5-trihydroxybenzene) that are synthesized through the acetate-malonate pathway [94]. They are thought to be the defense compounds in brown seaweeds with high concentrations (up to 25%), and are stored in special vesicles (physodes) [4]. A variety of phlorotannins has been discovered, including eckol, phlorofucofuroeckol A, dieckol, 6,6-bieckol, 8,8-bieckol, 7-phloroeckol, fucodiphloroethol G, phloroglucinol, and bifuhalol [96,97,98,99]. These compounds exhibit a variety of biological activities, including high antioxidant activity [100]; inhibition of α-amylase, α-glucosidase, and lipase, which are key enzymes in obesity and diabetes control [96,101,102]; and neuroprotective activities [97,98]. Phlorotannins were found to be non-toxic in cell lines, invertebrates, microalgae, seaweeds, plants, animals (fish, mice, rats, and dogs), and humans [99].

5.9. Carotenoids

Carotenoids are fat-soluble, highly unsaturated red, orange, or yellow pigments composed of isoprenoids, and their basic structure consists of eight isoprene units with a C 40 backbone [103]. Carotenoids are naturally present in plants, fungi, bacteria, and algae. Several types of carotenoids produced by seaweeds are β-carotene and zeaxanthin (red seaweed), fucoxanthin (brown seaweed), and siphonaxanthin (green seaweed). Fucoxanthin is one of the most abundant marine carotenoids with different health benefits, including anti-oxidative activity [104], and significantly reduces animal weight gain [105]. Furthermore, fucoxanthin can protect neuronal cells from oxidative-stress-induced neurotoxicity [106,107]. Siphonaxanthin is a marine carotenoid and a derivative of lutein found in green algae, such as Caulerpa lentillifera, Codium cylindricum, and Codium fragile [108]. The characteristic structure of siphonaxanthin is a keto group located at C-8 and an extra hydroxyl group at C-19 [109].

5.10. Lectins

Lectins are sugar-binding proteins, useful for deciphering the glycocode [10]. They are found in most organisms, from viruses and bacteria to plants and animals [110]. In general, marine algal lectins have low molecular weights compared to land-plant lectins, and they appear to induce negligible immunogenicity due to their small size. Furthermore, due to the presence of disulfide bonds and high specificity for complex carbohydrates over monosaccharides, marine algae lectins have greater molecular stability than plant lectins [111]. Lectins have biotechnological significance in a variety of fields, such as biochemistry, agriculture, and pharmacology, including nociception and inflammation [112].

5.11. Alkaloids

Bisindole alkaloids are a large group of structurally diverse secondary metabolites produced by a diverse range of organisms from both terrestrial and marine environments. Numerous unique bisindole alkaloids, including meridianins, topsentins, nortopsentins, dragmacidins, variolins, and rhopaladins, have been reported from sponges, actinomycetes, tunicates, and green algae [113]. Furthermore, some of these marine bisindole compounds have been discovered to have potent and diverse bioactive properties, such as antifungal, antibacterial, antiviral, cytotoxic, anti-inflammatory, and notable antitumor properties [114].

5.12. Halogenated Compounds

Terpenes are compounds found in seaweeds, consisting of two, three, four, or six isoprene units. Sesquiterpenes are terpenes with antibacterial, antifungal, and anti-inflammatory properties [115]. Eleganolone and eleganonal belong to the family of diterpenes, and are usually found in Bifurcaria bifurcata [116]. Diterpenes are non-volatile halogenated compounds with xenicane, dolabellane, and prenylated guaiane skeletons with different structures. Brown algae from the genus Dictyota are highly abundant in diterpenes. Dictyodial, dictyol C, and dictyol H, which are algal terpenes, have previously been isolated from various Dictyota species. These secondary metabolites deter feeding by marine herbivores [94].

6. Pharmacological Properties of Seaweed to Overcome Chronic Disease

Numerous studies have reported the pharmacological properties of seaweed for the treatment of chronic diseases, such as neurodegenerative diseases, cardiovascular diseases, obesity, diabetes, cancer, liver disease, osteoporosis, and arthritis. The bioactive compounds contained in seaweed are suggested to be beneficial for the management of chronic diseases. The mechanisms of each of these chronic diseases are shown in Figure 4.

6.1. Cancer

The mechanism of cancer cell formation and proliferation is shown in Figure 4a. Numerous studies on the effects of seaweed extracts against breast cancer have been reported. Sargassum hemiphyllum fucoidan inhibited the viability of MCF-7 and MDA-MB-231 cell lines by modulating the miR-29c/ADAM12 and miR-17-5p/PTEN axes [72]. Laurencia papillosa [117] and Sargassum sp. [118] showed significant inhibition of viability of the MCF-7 cell line. A high level of inhibition of the MDA-MB-231 cell line was also observed following treatment with Bifurcaria bifurcata [119] and Ulva fasciata by downregulating the EGFR/PI3K/Akt pathway [120]. Eucheuma cottonii suppressed the growth of tumor cells in rats inoculated with breast-cancer tumors in in vivo experiment [118]. Other in vivo studies reported that Wistar rats administered with 50 mg/kg body weight orally, daily for 10 weeks, suppressed breast carcinogenesis [121]. Studies of the effects of seaweed-derived compounds on cancer which were conducted in vitro and in vivo are summarized in Table 1.
Carrageenan extract from Gigartina pistillata inhibits cell growth in colorectal cancer stem cells with an IC50 value of 1 µg/mL [125]. Moreover, Hizikia fusiforme inhibited EJ tumor growth both in vivo and in vitro through G1-phase cell cycle arrest by downregulating cyclins and cyclin-dependent kinases (CDKs), as well as by inhibiting the expression of MMP-9 by downregulating NF-κB, AP-1, and Sp-1 [122]. In addition, seaweed extract also exhibited potent anticancer activity against hepatocellular carcinoma (HCC) cell lines [123], the U87MG cell line [129], and sarcoma 180 ascites tumor cells both in vitro and in vivo [126]. Furthermore, the inhibition of HT-29 cell proliferation by fucoidan may be due to the downregulation of IGF-IR signaling by the main IRS-1/PI3K/AKT pathway.

6.2. Neurodegenerative Disease

Neurodegenerative diseases are age-related chronic and progressive loss of neurons, such as Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis, Huntington’s disease, multiple sclerosis, cerebral ischemia, and traumatic brain injury [130,131]. The major cellular and molecular events that cause neurodegeneration are oxidative stress, misfolded proteins, impaired mitochondrial function, apoptosis induction, proteostasis impairment, and neuroinflammation [132]. The mechanism of neurodegenerative disease is shown in Figure 4b. Studies of the effects of seaweed-derived compounds on neurodegenerative diseases are summarized in Table 2.
The activation of microglia—a macrophage cell production in the central nervous system (CNS) due to the excessive production of inflammatory mediators such as nitric oxide (NO), prostaglandin E2 (PGE2), and pro-inflammatory cytokines, e.g., tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-6, and reactive oxygen species (ROS)—can cause chronic neurodegeneration [130,143]. Myagropsis myagroides sargachromenol reduced inflammation in lipopolysaccharide (LPS)-stimulated microglia by downregulating the IκB-α/NF-κB and ERK/JNK pathways [130]. One of the factors causing misfolded proteins that is associated with neurodegenerative diseases is increased endoplasmic reticulum (ER) stress. Ethanol extract from Undaria pinnatifida was reported to decrease endoplasmic reticulum (ER) voltage through the Akt/mTOR signaling pathway [133]. Fucoxanthin extract of Sargassum horneri showed neurodegenerative effects by decreasing H2O2-induced neurotoxicity via upregulating of the PI3K/Akt cascade and inhibition of the ERK pathway [107]. Bisindole alkaloids, namely racemocin A and racemocin B from Caulerpa racemose, reduced the Aβ25–35-induced SH-SY5Y cell (neuroblast from neural tissue) damage by increasing cell viability of 5.5% and 14.6% for racemocin A and racemocin B, respectively [114]. Fucoxanthin may rescue cerebral ischemic/reperfusion injury from nerve inflammation and oxidative stress by promoting the Nrf2/HO-1 signaling pathway in rats [21]. Reducing Ca2+-mediated neurotoxicity could protect rats with cerebral ischemic/reperfusion injury from nerve inflammation and oxidative stress [136].
Alzheimer’s disease is a type of dementia that causes a global, progressive, and irreversible deterioration of various cognitive functions (memory, attention, concentration, language, and thinking, among others) [144]. The main cause of Alzheimer’s disease is the dysregulation of β-amyloid (Aβ) levels, which induces neuronal death via multiple mechanisms, including oxidative stress, excitotoxicity, apoptosis, and inflammation [145]. According to Kim et al. [135], Ishige foliacea may decrease Aβ accumulation, thus inhibiting AChE and BACE1 [134], and suppress memory deficits by enhancing the BDNF–TrkB–ERK signaling pathway in the hippocampus. Butanol extracts from Ecklonia cava have anti-Aβ effects on Aβ-related pathogenesis, including amyloidogenic processing, Aβ oligomerization, Aβ fibrillization, and Aβ-induced neuronal death [137]. By downregulating the ERK, p38 MAPK, and JNK pathways, compounds isolated from Ishige okamurae reduced Aβ25–35-induced phosphorylation both in vitro and in vivo [139]. Phenolic compounds from Gracilaria beckeri, Ecklonia maxima, Gelidium pristoides, and Ulva rigida, such as phloroglucinol, catechin, and epicatechin 3-glucoside, significantly reduced anti-acetylcholinesterase and butyrylcholinesterase activities [140].
Parkinson’s disease is a prevalent neurological disease affecting the movement of the elderly. It is characterized by the formation of Lewy bodies, the death of dopaminergic neurons in the substantia nigra pars compacta (SNpc), and dopamine depletion (DA) [116,146]. During aging, the concentration of iron in the brain can change, resulting in metabolic stress [147,148]. The neuroprotective activity of Bifurcaria bifurcata fractions suggested that two major diterpenes (eleganolone and eleganonal) have potential for Parkinson’s disease management because of their iron-reducing activities [116]. Silva et al. [149] reported that among the three macroalgae tested, Saccorhiza polyschides exhibited the highest potential as a therapeutic agent against AD-induced toxicity, exhibiting anti-apoptotic effects associated with mitochondrial protection and caspase-3 inhibition in a model of Parkinson’s disease. Two extracts, Undaria pinnatifida and fucoxanthin, showed moderate inhibition and were inactive against hMAO-A and hMAO-B [135]. These results suggest that fucoxanthin may be beneficial for Parkinson’s disease due to its ability as a potent dopamine D3/D4 agonist agent. An in vivo study reported that rats fed sulfated agaran exhibited great neuroprotection due to its ability to reduce oxidative stress, and showed recovery of behavioral activity and improved weight gain [141].
Based on previous research, extracts of phlorotannins and their derivatives from brown seaweed demonstrate neuroprotective activity. Ecklonia cava phlorotannin downregulates the expression and activity of alpha- and gamma-secretase [138], while its derivatives dieckol and phlorofucofuroeckol increase acetylcholine and reduce anticholinesterase activities [98], which lead to a reduction in Aβ production. In addition, dieckol and eckol from Eisenia bicyclis showed a higher binding affinity for hMAOs by hydrogen bonding and hydrophobic interactions compared to hMAO-A and hMAO-B [97].

6.3. Liver Disease

The liver is the main metabolic organ for the detoxification of drugs and xenobiotics, crucial to combatting oxidative-stress-inducing agents that circulate in the blood [150]. However, excessive alcohol intake can cause liver diseases, including alcoholic liver disease (ALD) and nonalcoholic fatty liver disease (NAFLD). The mechanism of alcocholic liver disease is shown in Figure 4c. Several studies on bioactive compounds for liver disease management have been conducted (Table 3).
    Recent studies have reported that     Sargassum ilicifolium showed better inhibition of nephroprotective and hepatoprotective effects than Halymenia porphyroides in the liver and kidney of rats after damage by administration of acetaminophen or cisplatin [151]. Hepatoprotection has been reported with the administration of Turbinaria decurrens fucoidan, which ameliorates antioxidant status and decreases lipid peroxidation marker levels [73]. Administration of 50 mg/kg Sargassum fluitans ethanol extract reduced the levels of APAP and CCl4 in liver damage models through the inhibition of inflammation and fibrosis in liver tissue [32]. This result is in accordance with the research on the hepatoprotective effects of Myagropsis myagroides, Sargassum henslowianum, and Sargassum siliquastrum [152]. Fucus vesiculosus fucoidan may exert thioacetamide (TAA)-induced liver injury by downregulating pro-inflammatory cytokines [153]. Moreover, fucoidan from Cladosiphon okamuranus showed hepatoprotective effects by improving the antioxidant defense system and reducing ROS [155].
The ALD symptoms, such as fatty liver disease and hepatitis, can progress to steatohepatitis, liver fibrosis, cirrhosis, and the most severe form of liver cancer [158]. 7-phloro-eckol isolated from Ecklonia cava showed an inhibitory effect on ALD by reducing alcohol-induced oxidative stress injury in HepG2/CYP2E1 cells [100]. Meanwhile, other seaweeds such as Ulva lactuca can reduce D-Gal-induced DNA damage and necrosis levels in rats [8]. NAFLD refers to a group of liver disorders that range from fat accumulation in the liver (steatosis) to nonalcoholic steatohepatitis (necrosis and inflammation), with some cases progressing to fibrosis, cirrhosis, and liver failure [159]. One of the progressive stages of NAFLD is called nonalcoholic steatohepatitis (NASH), which is associated with hepatocyte injury, excessive oxidative stress, and chronic inflammation in the fatty liver, and can progress to more serious liver diseases such as cirrhosis and hepatocellular carcinoma [160,161]. The phytocomplex of Fucus vesiculosus and Ascophyllum nodosum suggests a reduction in the postprandial glycemic peak and the blood glucose curve (AUC), and an inhibitory effect on carbohydrate digestion [154].

6.4. Diabetes

In recent years, diabetes has become a major health problem worldwide, particularly among youth [162]. Diabetes is a metabolic disorder characterized by insufficient insulin secretion and improper insulin utilization [163]. It is divided into two types: insulin-dependent diabetes mellitus (type 1 diabetes) and non-insulin-dependent diabetes mellitus (type 2 diabetes) [101]. Previous studies have shown that seaweed, which is high in bioactive compounds, has anti-diabetic properties (Table 4).
Type 2 diabetes is the most common type of diabetes, and its prevalence is increasing significantly worldwide. The mechanism of diabetes is shown in Figure 4d. Hyperglycemia plays an important role in the development of type 2 diabetes and complications associated with the disease, such as microvascular and macrovascular diseases [176]. Several studies have been conducted that suggest extracts from seaweeds could combat diabetes. For instance, flavonoids present in Enteromorpha prolifera exhibited hypoglycemic effects by upregulating IRS1/PI3K/AKT and downregulating the JNK1/2 insulin pathway in the liver [14]. This study also reported that treatment in type 2 diabetic mice enriched the abundance of gut microbiota, such as Turicibacter and Alisties [9]. Meanwhile, phlorotannin (dieckol) of Ecklonia cava, fucoidan, and fucoxanthin of Sargassum hemiphyllum reduced total glucose, lipid, serum insulin levels, and body weight in vivo [101,102,165]. Furthermore, polyphenols extracted from Ecklonia cava also inhibited the activation of high glucose-induced hepatic stellate cells (HSCs) by downregulating ROS and/or GSH and inhibiting TGF-β secretion [164]. In addition, Padina arborescens may ameliorate hyperglycemia and dyslipidemia in in C57BL/KsJ-db/db mice. A novel phenolic compound from Ishige foliacea, octaphlorethol A, was tested in vitro, and may increase glucose transporter 4 (Glut4) translocation, which is mediated by PI3K/Akt and AMPK activation [170] Additionally, octaphlorethol A was found to downregulate hepatic gluconeogenesis in vivo by inhibiting G6Pase and PEPCK activity [171]. Polysaccharide fractions from Scagassum, Sargassum fusiforme, and Macrocystis pyrifera may regulate glucose, triglyceride (TG), and total cholesterol, subsequently ameliorating liver and kidney damage [173].
Many seaweeds also exhibit inhibitory effects on α-glucosidase and α-amylase, such as Ascophyllum nodosum, Fucus vesiculosus, and Undaria pinnatifida [168,169]. Bioactive compounds from Ecklonia stolonifera and Eisenia bicyclis, including phlorotannin, showed inhibitory effects on protein tyrosine phosphatase 1 B (PTP1B) and α-glucosidase [174], whereas phlorotannin showed moderate inhibitory effects on rat lens aldose reductase (RLAR), human recombinant aldose reductase (HRAR), and PTP1B [10]. Other studies have demonstrated that lectin isolated from Bryothamnion seaforthii exerts hypoglycemic and hypolipidemic effects, reduces insulin resistance, and improves pancreatic β-cell function in rats with streptozotocin (STZ)-induced diabetes [111]. In the same animal model, Ulva rigida showed inhibition of carbohydrate metabolism, hyperlipidemia, and oxidative stress [175].

6.5. Obesity

Recently, the anti-obesity activity of seaweed has received considerable attention (Table 5). Obesity is generally considered a risk factor for a number of chronic metabolic diseases, dyslipidemia, hypertension, and hyperglycemia [177]. Obesity can also lead to musculoskeletal problems and an increased risk of cancers, such as colorectal, breast, and endometrial cancers [178].
    The mechanism of obesity is shown in     Figure 4e. Obesity upregulates proinflammatory cytokines such as tumor necrosis factor (TNF)- α, interleukin (IL)-1β, IL-6, and leptin; TNF-α and IL-1β subsequently mediate the regulation of matrix metalloproteinases (MMPs) such as MMP-1, MMP-3, MMP-10, MMP-12, and MMP-1310-12 by stimulating the nuclear factor-kappa B (NF-κB) pathway. MMP levels are also influenced by mitogen-activated protein kinases (MAPKs), such as ERK1/2, c-Jun NH2-terminal kinase, and p38 subfamilies [190]. A high-fat diet increases the levels of gut inflammatory cytokines such as TNF-α, IL-1β, and IL-12, which are linked to weight gain, adiposity, and enhanced plasma insulin and glucose levels [191]. Obesity-related diseases such as metabolic syndrome may be reduced with an intervention that decreases gut and systemic inflammation [192]. Modulating the gut microbiota seems to be an option for reducing obesity [30]. Moreover, siphonaxanthin from Codium cylindricum showed anti-obesity activity since it accumulates in the stomach and small intestine, while its metabolites are absorbed and accumulated in the white adipose tissue (WAT) [109].
In addition, elevated aP2, ACC, and PPARγ gene expression levels cause excessive fat accumulation [181]. Sargassum miyabei and Saccorhiza polyschides exhibited anti-obesity effects by downregulating adipogenic and lipogenic gene expression, and intestinal and systemic glucose metabolism, thus inhibiting α-amylase activity [179,180]. Furthermore, Plocamium telfairiae [188] and Gelidium amansii [187] also inhibited adipogenic and lipogenic gene expression in 3T3-L1 preadipocytes. In vivo, seaweed reduced body weight, triglycerides, total glucose, fatty liver, and white adipose tissue in the tested animals [43,188]. Frequently bioactive compounds that are found in seaweed for anti-obesity activity are fucosterol [183,184] and phlototannin [96,185]. Previous studies have reported that siphonaxanthin, a carotenoid from Codium cylindricum, can restore antioxidative capacity and reduce somatic stress in obese mice [189].

6.6. Cardiovascular Disease

Seaweeds are increasingly in demand because they contain dietary fiber, peptides, and carotenoids that have the potential to prevent cardiovascular disease (CVD) [193]. Cardiovascular disease refers to a group of heart- and blood-vessel disorders, including coronary heart disease, cerebrovascular disease, peripheral artery disease, congenital heart disease, rheumatic heart disease, deep vein thrombosis, and pulmonary embolism [194]. The effects of seaweed-derived compounds on cardiovascular disease are shown in Table 6.
    The mechanism of cardiovascular disease is shown in     Figure 4f. The inhibition of angiotensin I-converting enzyme (ACE-I) is a well-established approach for the treatment of hypertension [194]. Paiva et al. [199] suggested that phenolic content and amino acid profiles, such as peptides and phlorotannins, possessed a higher inhibitory effect on ACE-I yield [199]. Furthermore, Sargachromenol D from Sargassum siliquastrum could have antihypertensive activity, as it can function as an antagonist of the L-type Ca2+ channel and endothelin A/B2 receptors.
Dyslipidemia is a major risk factor for the development of atherosclerosis and related CVDs and is a major cause of death in many countries. Dyslipidemia is commonly characterized by elevated levels of total cholesterol (TC), triglyceride (TG), and low-density lipoprotein cholesterol (LDL-C), and decreased levels of high-density lipoprotein cholesterol (HDL-C) in the blood [202]. A previous study suggested that fucoidan of Saccharina sculpera may inhibit cholesterol synthesis and reverse transport by downregulating HMG-CoA-R; upregulating LCAT; modulating fatty acid synthesis by downregulating SREBP-1c; and accelerating mitochondrial β-oxidation by upregulation of PPARα, PPARγ, and LPL [195]. Several seaweeds from the Karachi coast showed hypolipidemic effects by reducing the lipid profile. Solieria robusta was found to be the most effective in reducing the lipid profile in diet-induced hyperlipidemic rats among Iyengaria stellata, Colpomenia sinuosa, Spatoglossum asperum, and Caulerpa racemosa [196]. In addition, both fucoidan A2 and A3 from Ascophyllum nodosum reduced the lipid profile, including plasma total cholesterol, triglyceride, and fat pad index, by regulating reverse cholesterol transport (RCT) [29,197]. Cardioprotection was also shown in a study by Thomes et al., who found that Cladosiphon okamuranus fucoidan reduced the lipid profile—including total cholesterol, triglycerides, and low-density lipoprotein (LDL)—via reverse transport isoproterenol-induced myocardial infarction in rats [155], and phlorotannin from Ecklonia cava in a DOX-induced rat cardiotoxicity model [198]. Other studies have reported that polyphenol extract and dieckol Ecklonia cava may reduce lipid profiles in the serum of mice fed a high fat diet (HFD), and reduce lipid accumulation in 3T3-L1 cells [201].

6.7. Arthritis

Arthritis is the most common chronic inflammatory disease; it is characterized by structural and biochemical changes in major joint tissues, including cartilage matrix degradation and insufficient extracellular matrix synthesis (ECM), resulting in pain, stiffness, and joint failure [203,204]. Arthritis consists of several types, such as juvenile idiopathic arthritis (JIA) [205], psoriatic arthritis [206], gouty arthritis [207], rheumatoid arthritis (the most common form) [208], and osteoarthritis (OA) [209]. Previous studies on the potential of seaweed compounds for arthritis treatment are reported in Table 7.
Among several types of arthritis, seaweed bioactivity against OA is the most widely reported. OA is a common type of arthritis that primarily affects the knee [104]. A previous study suggested that OA joint degeneration results from a combination of mechanical stresses and biochemical factors, such as ROS and MMPs, which act as precursors to pro-inflammatory cytokines, including IL-1α, IL-1β, and TNF-α [204,214]. As a result, cartilage degradation was attenuated in the model tested [190]. Previous studies have reported that phlorotannin-rich extracts isolated from Ecklonia cava reduce inflammation by downregulating pro-inflammatory cytokines [13]. Moreover, Lobophora variegate fucan reduced articular inflammation by downregulating paw edema and serum TNF-α [210]. Meanwhile, Laurencia glandulifera and Codium fragile exhibited anti-OA effects by inhibiting NF-κB activity and COX-2 expression in RAW264.7 cells in vitro, and reduced carrageenan-induced rat edema in vivo [212,213]. A study by Rivanor et al. [112] reported that Caulerpa cupressoides lectin may reduce leukocyte influx and the expression levels of pro-inflammatory cytokines, including IL-1β and TNF-α, in the temporomandibular joint. The mechanism of OA is shown in Figure 4g.
Research on the benefits of seaweed for the treatment of OA was conducted in a clinical study by combining Phase I and II trials [12]. The formulation used contained Maritech® fucoidan-rich extracts of Fucus vesiculosis, Macrocystis pyrifera, and Laminaria japonica, with additional vitamin B6, zinc, and manganese taken daily. The study was conducted for 12 weeks in 11 participants, and for 10 weeks in one participant. A multilevel linear model revealed a decrease of 18% for the 100 mg treatment and of 52% for the 1000 mg dose in the average COAT score at the end of the study. This result suggests that treatment should be conducted for more than 12 weeks in a dose-dependent manner to reduce the symptoms of OA. A phase III randomized controlled trial (RCT) must be conducted to ensure its safety.
Rheumatoid arthritis is a chronic joint disorder affecting the cartilage and subchondral bone, affecting approximately 1% of the world’s population. The fucoidan derivative of Undaria pinnatifida showed potential as a rheumatoid arthritis agent both in vitro and in vivo by downregulating pro-inflammatory cytokines such as COX-2 expression [75]. Other studies have reported that alginic acid from Sargassum wightii may downregulate pro-inflammatory cytokines such as cycloxygenase-2 (COX-2), lipoxygenase (5-LOX), xanthine oxidase (XO), and myeloperoxidase (MPO) in adjuvant-induced arthritis [211] and type II collagen-induced arthritis rat models [34].

6.8. Osteoporosis

Osteoporosis is a major public health issue affecting the aging population, characterized by low bone mass. Osteoporosis is linked to decreased bone formation by osteoblasts and increased bone resorption by osteoclasts; these lead to microarchitectural deterioration of bone tissue, excessive bone fragility, and increased bone fracture risk [11,215,216]. In recent years, natural products from seaweed have been explored and investigated more specifically as sources for osteoporosis treatment. Studies on the osteoprotective activity of seaweeds are summarized in Table 8.
Some marine algae have been investigated for their beneficial effects as osteogenic agents because of their important minerals, such as manganese, zinc, calcium, and amino acids, which can promote bone metabolism [223]. Bone-forming cells include osteoblasts, osteocytes, and bone-lining cells, whereas osteoclasts participate in bone resorption [218]. Red seaweeds have been reported to have osteoprotective effects on osteogenic differentiation. Dichotomaria obtusata [215], Ceramium secundatum, and Plocamium lyngbyanum [221] methanolic extracts significantly upregulated osteogenic activity and increased mineralization of bone cells tested in vitro. Furthermore, Ceramium secundatum and Plocamium lyngbyanum increased the opercular bone area of zebrafish larvae in vivo [221]. Moreover, two powder-residue-derived extracts of Ceramium pallidum caused significant osteogenic differentiation and produced mineralogenic effects in vitro, and increased opercular bone and bone density in zebrafish larvae in vivo [216].
Antioxidant fucoidans from Dictyota mertensii protect pro-osteoblastic cells under oxidative stress by deregulating osteoblast activity [217]. Furthermore, Padina pavonica promotes pro-osteogenic effects by enhancing osteoblast differentiation and subsequent mineralization, as well as by regulating the expression of earlier osteoblast-specific markers [218]. In contrast, fucoxanthin from Laminaria digitata and Ascophyllum nodosum extract showed low pro-osteogenic activity in the two cell types tested [11]. Phenolic compounds from Cladophora rupestris and Codium fragile promoted osteoblast-like cells in vitro and the mineralized area in zebrafish larvae in vivo [7]. Diphlorethohydroxycamalol-derived phlorotannin isolated from Ishige okamurae showed anti-osteoporosis effects by upregulating osteoblast differentiation against H2O2-induced oxidative damage and promoting bone resorption, by decreasing the expression level of the receptor activator of NF-κB ligand [220].
The mechanism of osteoporosis is shown in Figure 4h. Osteoclasts are large multinucleated myeloid cells that can break down the bone matrix by proteolytic degradation and decalcification [224]. The receptor activator of nuclear factor-κB (RANKL) has been identified as an important transcription factor for osteoclast differentiation from the monocyte/macrophage lineage. Gracilaria verrucose downregulated RANKL-induced osteoclast differentiation by inhibiting the c-Fos-NFATc1 signaling pathway in vitro, and inhibited bone loss in an OVX mouse model when tested in vivo [33]. Meanwhile, fucoidan extract from Sargassum hemiphyllum prevents the differentiation of osteoclasts and inflammatory bone loss by regulating the Akt/GSK3β/PTEN/NFATc1 signaling pathway and calcineurin activity [219].

7. Conclusions

The use of seaweeds as food and medicine has a long history, and the benefit of seaweeds for health have been revealed. Many published studies have demonstrated that extracts from seaweeds can contribute to the reduction and modulation of several chronic diseases. However, most of the studies have been conducted in vitro and in vivo, and only three seaweed studies have been conducted at the clinical stage. Hence, further clinical studies involving human subjects are required to confirm these therapeutic effects. Results from clinical studies will provide useful information for understanding the mechanisms underlying the effects of seaweed in modulating chronic diseases. Furthermore, the bioactive compounds in seaweed need to be purified and identified to better understand the mechanisms and pathways that they affect and, therefore, to develop them as functional food and nutraceutical products. Future research on the identification, isolation, and purification of seaweed’s bioactive compounds and its mechanism to treat chronic diseases might provide a significant contribution to overcoming increasing chronic disease in the world.

Author Contributions

Conceptualization, M.D.N.M. and J.-S.C.; methodology, M.D.N.M.; software, D.H.; validation M.D.N.M., D.H. and J.-S.C.; formal analysis, M.D.N.M.; investigation, M.D.N.M.; resources, D.H.; data curation, D.H.; writing—original draft preparation, M.D.N.M. and J.-S.C.; writing—review and editing, M.D.N.M., D.H. and J.-S.C.; visualization, M.D.N.M. and D.H.; supervision, J.-S.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Ministry of Oceans and Fisheries and Busan Metropoli tan City through the Capacity Building Project for the Development of the Busan Seafood Industry- Based Complex.

Data Availability Statement

Data supporting reported results are available upon request.

Acknowledgments

We thank to Silla University, Jenderal Soedirman University and Diponegoro University for the reseach collaboration.

Conflicts of Interest

The authors declare that they have no conflicts of interest to declare.

References

  1. Holdt, S.L.; Kraan, S. Bioactive compounds in seaweed: Functional food applications and legislation. J. Appl. Phycol. 2011, 23, 543–597. [Google Scholar] [CrossRef]
  2. Plasek, B.; Lakner, Z.; Kasza, G.; Temesi, Á. Consumer evaluation of the role of functional food products in disease prevention and the characteristics of target groups. Nutrients 2020, 12, 69. [Google Scholar] [CrossRef] [Green Version]
  3. Mendis, E.; Kim, S.K. Present and future prospects of seaweeds in developing functional foods. Adv. Food Nutr. Res. 2011, 64, 1–15. [Google Scholar] [CrossRef]
  4. Neto, R.T.; Marçal, C.; Queirós, A.S.; Abreu, H.; Silva, A.M.S.; Cardoso, S.M. Screening of Ulva rigida, Gracilaria sp., Fucus vesiculosus and Saccharina latissima as functional ingredients. Int. J. Mol. Sci. 2018, 19, 2987. [Google Scholar] [CrossRef] [Green Version]
  5. Pandey, A.; Chauhan, O.; Semwal, A. Pandey 2020. Seaweeds—A Potential Source for Functional Foods.pdf. Def. Life Sci. J. 2020, 5, 315–322. [Google Scholar] [CrossRef]
  6. Brown, E.M.; Allsopp, P.J.; Magee, P.J.; Gill, C.I.R.; Nitecki, S.; Strain, C.R. Seaweed and human health. Nutr. Rev. 2014, 72, 205–216. [Google Scholar] [CrossRef]
  7. Leandro, A.; Pacheco, D.; Cotas, J.; Marques, J.C.; Pereira, L.; Gonçalves, A.M.M. Seaweed’s Bioactive Candidate Compounds to Food Industry and Global Food Security. Life 2020, 10, 140. [Google Scholar] [CrossRef]
  8. Sathivel, A.; Balavinayagamani; Rao, B.R.H.; Devaki, T. Sulfated polysaccharide isolated from Ulva lactuca attenuates D-galactosamine induced DNA fragmentation and necrosis during liver damage in rats. Pharm. Biol. 2014, 52, 498–505. [Google Scholar] [CrossRef]
  9. Lin, G.; Liu, X.; Yan, X.; Liu, D.; Yang, C.; Liu, B.; Huang, Y.; Zhao, C. Role of green macroalgae Enteromorpha prolifera polyphenols in the modulation of gene expression and intestinal microflora profiles in type 2 diabetic mice. Int. J. Mol. Sci. 2019, 20, 25. [Google Scholar] [CrossRef] [Green Version]
  10. Jung, H.A.; Islam, M.N.; Lee, C.M.; Oh, S.H.; Lee, S.; Jung, J.H.; Choi, J.S. Kinetics and molecular docking studies of an anti-diabetic complication inhibitor fucosterol from edible brown algae Eisenia bicyclis and Ecklonia stolonifera. Chem. Biol. Interact. 2013, 206, 55–62. [Google Scholar] [CrossRef]
  11. Walsh, P.J.; McGrath, S.; McKelvey, S.; Ford, L.; Sheldrake, G.; Clarke, S.A. The osteogenic potential of brown seaweed extracts. Mar. Drugs 2019, 17, 141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Myers, S.P.; O’Connor, J.; Fitton, J.H.; Brooks, L.; Rolfe, M.; Connellan, P.; Wohlmuth, H.; Cheras, P.A.; Morris, C. A combined Phase I and II open-label study on the Immunomodulatory effects of seaweed extract nutrient complex. Biol. Targets Ther. 2011, 5, 45–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Shin, H.; Hwang, H.J.; Kang, K.J.; Lee, B.H. An Antioxidative and Antiinflammatory Agent for Potential Treatment of Osteoarthritis from Ecklonia cava. Arch. Pharm. Res. 2006, 29, 165–171. [Google Scholar] [CrossRef] [PubMed]
  14. Yan, X.; Yang, C.; Lin, G.; Chen, Y.; Miao, S.; Liu, B.; Zhao, C. Antidiabetic Potential of Green Seaweed Enteromorpha prolifera Flavonoids Regulating Insulin Signaling Pathway and Gut Microbiota in Type 2 Diabetic Mice. J. Food Sci. 2019, 84, 165–173. [Google Scholar] [CrossRef] [Green Version]
  15. Paiva, L.; Lima, E.; Neto, A.I.; Marcone, M.; Baptista, J. Nutritional and Functional Bioactivity Value of Selected Azorean Macroalgae: Ulva compressa, Ulva rigida, Gelidium microdon, and Pterocladiella capillacea. J. Food Sci. 2017, 82, 1757–1764. [Google Scholar] [CrossRef]
  16. Gray, J.; Armstrong, G.; Farley, H. Opportunities and constraints in the functional food market. Nutr. Food Sci. 2003, 33, 213–218. [Google Scholar] [CrossRef]
  17. Plaza, M.; Santoyo, S.; Jaime, L.; Reina, G.G.-B.; Herrero, M.; Señoráns, F.J.; Ibáñez, E. Screening for bioactive compounds from algae. J. Pharm. Biomed. Anal. 2010, 51, 450–455. [Google Scholar] [CrossRef]
  18. Pereira, L. A Review of The Nutrient Composition of Selected Edible Seaweed. In Seaweed: Ecology, Nutrient Composition and Medicinal Uses; Pomin, V.H., Ed.; Nova Science Publishers, Inc.: Coimbra, Portugal, 2011; ISBN 9781614708780. [Google Scholar]
  19. McHugh, D.J. A Guide to the Seaweed Industry; Food and Agriculture Organization: Rome, Italy, 2003; ISBN 92-5-104958-0. [Google Scholar]
  20. Kolanjinathan, K.; Ganesh, P.; Saranraj, P. Pharmacological Importance of Seaweeds: A Review. World J. Fish Mar. Sci. 2014, 6, 1–15. [Google Scholar] [CrossRef]
  21. Bocanegra, A.; Bastida, S.; Benedí, J.; Ródenas, S.; Sánchez-muniz, F.J. Characteristics and Nutritional and Cardiovascular-Health Properties of Seaweeds. J. Med. Food 2009, 12, 236–258. [Google Scholar] [CrossRef]
  22. Faggio, C.; Pagano, M.; Dottore, A.; Genovese, G.; Morabito, M. Evaluation of anticoagulant activity of two algal polysaccharides. Nat. Prod. Res. 2016, 30, 1934–1937. [Google Scholar] [CrossRef]
  23. Iso, H.; Kubota, Y. Nutrition and Disease in the Japan Collaborative Cohort Study for Evaluation of Cancer (JACC). Asian Pac. J. Cancer Prev. 2007, 8, 35–80. [Google Scholar] [PubMed]
  24. Kim, J.; Shin, A.; Lee, J.-S.; Youn, S.; Yoo, K.-Y. Dietary Factors and Breast Cancer in Korea: An Ecological Study. Breast J. 2020, 15, 683–686. [Google Scholar] [CrossRef] [PubMed]
  25. Park, J.-K.; Woo, H.W.; Kim, M.K.; Shin, J.; Lee, Y.-H.; Shin, D.H.; Shin, M.-H.; Choi, B.Y. Dietary iodine, seaweed consumption, and incidence risk of metabolic syndrome among postmenopausal women: A prospective analysis of the Korean Multi—Rural Communities Cohort Study (MRCohort). Eur. J. Nutr. 2020, 60, 135–146. [Google Scholar] [CrossRef] [PubMed]
  26. Iso, H. Lifestyle and Cardiovascular Disease in Japan. J. Atheroscler. Thromb. 2010, 18, 83–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Michikawa, T.; Inoue, M.; Shimazu, T.; Sawada, N. Seaweed consumption and the risk of thyroid cancer in women: The Japan Public Health Center-based Prospective Study and for the Japan Public Health Center-based Prospective Study Group. Eur. J. Cancer Prev. 2008, 21, 254–260. [Google Scholar] [CrossRef]
  28. Hu, P.; Li, Z.; Chen, M.; Sun, Z.; Ling, Y.; Jiang, J.; Huang, C. Structural elucidation and protective role of a polysaccharide from Sargassum fusiforme on ameliorating learning and memory deficiencies in mice. Carbohydr. Polym. 2016, 139, 150–158. [Google Scholar] [CrossRef]
  29. Yang, Z.; Liu, G.; Wang, Y.; Yin, J.; Wang, J.; Xia, B.; Li, T.; Yang, X.; Hou, P.; Hu, S.; et al. Fucoidan A2 from the Brown Seaweed Ascophyllum nodosum Lowers Lipid by Improving Reverse Cholesterol Transport in C57BL/6J Mice Fed a High-Fat Diet. J. Agric. Food Chem. 2019, 67, 5782–5791. [Google Scholar] [CrossRef]
  30. Kim, J.; Choi, J.H.; Oh, T.; Ahn, B.; Unno, T. Codium fragile Ameliorates High-Fat Diet-Induced Metabolism by Modulating the Gut Microbiota in Mice. Nutrients 2020, 12, 1848. [Google Scholar] [CrossRef]
  31. Mary, J.S.; Vinotha, P.; Pradeep, A.M. Screening for in vitro cytotoxic activity of seaweed, Sargassum sp. against hep-2 and MCF-7 cancer cell lines. Asian Pac. J. Cancer Prev. 2012, 13, 6073–6076. [Google Scholar] [CrossRef]
  32. Quintal-novelo, C.; Rangel-méndez, J.; Ortiz-tello, Á.; Graniel-sabido, M.; De Vaca, R.P.; Moo-puc, R. A Sargassum fluitans Borgesen Ethanol Extract Exhibits a Hepatoprotective Effect In Vivo in Acute and Chronic Liver Damage Models. BioMed Res. Int. 2018, 2018, 6921845. [Google Scholar] [CrossRef] [Green Version]
  33. Kim, K.J.; Lee, Y.J.; Hwang, Y.H.; Kang, K.Y.; Yee, S.T.; Son, Y.J. In vitro and in vivo effects of Gracilaria verrucosa extracts on osteoclast differentiation. J. Clin. Med. 2017, 6, 32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Sarithakumari, C.H.; Kurup, G.M. International Immunopharmacology Alginic acid isolated from Sargassum wightii exhibits anti-inflammatory potential on type II collagen induced arthritis in experimental animals. Int. Immunopharmacol. 2013, 17, 1108–1115. [Google Scholar] [CrossRef] [PubMed]
  35. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G.; Altman, D.; Antes, G.; Atkins, D.; Barbour, V.; Barrowman, N.; Berlin, J.A.; et al. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. PLoS Med. 2009, 6, e1000097. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Myers, S.P.; Connor, J.O.; Fitton, J.H.; Brooks, L.; Rolfe, M.; Connellan, P.; Wohlmuth, H.; Cheras, P.A.; Morris, C. A combined phase I and II open label study on the effects of a seaweed extract nutrient complex on osteoarthritis. Biologics 2010, 4, 33–44. [Google Scholar] [CrossRef] [Green Version]
  37. Garcia-Vaquero, M.; Rajauria, G.; Miranda, M.; Sweeney, T.; Lopez-Alonso, M.; O’Doherty, J. Seasonal Variation of the Proximate Composition, Mineral Content, Fatty Acid Profiles and Other Phytochemical Constituents of Selected Brown Macroalgae. Mar. Drugs 2021, 19, 204. [Google Scholar] [CrossRef]
  38. Rajauria, G. Optimization and validation of reverse phase HPLC method for qualitative and quantitative assessment of polyphenols in seaweed. J. Pharm. Biomed. Anal. 2018, 148, 230–237. [Google Scholar] [CrossRef]
  39. Khairy, H.M.; El-Shafay, S.M. Seasonal variations in the biochemical composition of some common seaweed species from the coast of Abu Qir Bay, Alexandria, Egypt. Oceanologia 2013, 55, 435–452. [Google Scholar] [CrossRef] [Green Version]
  40. Banerjee, K.; Ghosh, R.; Homechaudhuri, S.; Mitra, A. Seasonal variation in the biochemical composition of red seaweed (Catenella repens) from Gangetic delta, northeast coast of India. J. Earth Syst. Sci. 2009, 118, 497–505. [Google Scholar] [CrossRef] [Green Version]
  41. Rioux, L.E.; Turgeon, S.L. Seaweed Carbohydrates; Elsevier Inc.: Amsterdam, The Netherlands, 2015; ISBN 9780124199583. [Google Scholar]
  42. Stiger-Pouvreau, V.; Bourgougnon, N.; Deslandes, E. Carbohydrates from Seaweeds; Elsevier Inc.: Amsterdam, The Netherlands, 2016; ISBN 9780128027936. [Google Scholar]
  43. Yang, T.H.; Chiu, C.Y.; Lu, T.J.; Liu, S.H.; Chiang, M.T. The Anti-Obesity Effect of Polysaccharide-Rich Red Algae (Gelidium amansii) Hot-Water Extracts in High-Fat Diet-Induced Obese Hamsters. Mar. Drugs 2019, 17, 532. [Google Scholar] [CrossRef] [Green Version]
  44. Bauer, S.; Jin, W.; Zhang, F.; Linhardt, R.J. The Application of Seaweed Polysaccharides and Their Derived Products with Potential for the Treatment of Alzheimer’s Disease. Mar. Drugs 2021, 19, 89. [Google Scholar] [CrossRef]
  45. Usman, A.; Khalid, S.; Usman, A.; Hussain, Z.; Wang, Y. Algal Polysaccharides, Novel Application, and Outlook. In Algae Based Polymers, Blends, and Composites: Chemistry, Biotechnology and Materials Science; Elsevier: Amsterdam, The Netherlands, 2017; pp. 115–153. [Google Scholar] [CrossRef]
  46. Lee, S.Y.; Chang, J.H.; Lee, S.B. Chemical composition, saccharification yield, and the potential of the green seaweed Ulva pertusa. Biotechnol. Bioprocess Eng. 2014, 19, 1022–1033. [Google Scholar] [CrossRef]
  47. Nunraksa, N.; Rattanasansri, S.; Praiboon, J.; Chirapart, A. Proximate composition and the production of fermentable sugars, levulinic acid, and HMF from Gracilaria fisheri and Gracilaria tenuistipitata cultivated in earthen ponds. J. Appl. Phycol. 2019, 31, 683–690. [Google Scholar] [CrossRef]
  48. Wang, L.; Jayawardena, T.U.; Yang, H.W.; Lee, H.G.; Kang, M.C.; Sanjeewa, K.K.A.; Oh, J.Y.; Jeon, Y.J. Isolation, characterization, and antioxidant activity evaluation of a fucoidan from an enzymatic digest of the edible seaweed, Hizikia fusiforme. Antioxidants 2020, 9, 363. [Google Scholar] [CrossRef] [PubMed]
  49. Cherry, P.; O’hara, C.; Magee, P.J.; Mcsorley, E.M.; Allsopp, P.J. Risks and benefits of consuming edible seaweeds. Nutr. Rev. 2019, 77, 307–329. [Google Scholar] [CrossRef] [Green Version]
  50. Kawaroe, M.; Sari, D.W.; Hwangbo, J.; Santoso, J. Optimum fermentation process for red macroalgae Gelidium latifolium and Gracillaria verrucosa. J. Eng. Technol. Sci. 2015, 47, 674–687. [Google Scholar] [CrossRef] [Green Version]
  51. Lu, Y.A.; Lee, H.G.; Li, X.; Hyun, J.M.; Kim, H.S.; Kim, T.H.; Kim, H.M.; Lee, J.J.; Kang, M.C.; Jeon, Y.J. Anti-obesity effects of red seaweed, Plocamium telfairiae, in C57BL/6 mice fed a high-fat diet. Food Funct. 2020, 11, 2299–2308. [Google Scholar] [CrossRef]
  52. Zheng, T.; Liu, C.; Yang, J.; Liu, Q.; Li, J. Hijiki seaweed (Hizikia fusiformis): Nutritional value, safety concern and arsenic removal method. Adv. Mater. Res. 2013, 634–638, 1247–1252. [Google Scholar] [CrossRef]
  53. Galland-Irmouli, A.V.; Fleurence, J.; Lamghari, R.; Luçon, M.; Rouxel, C.; Barbaroux, O.; Bronowicki, J.P.; Villaume, C.; Guéant, J.L. Nutritional value of proteins from edible seaweed Palmaria palmata (Dulse). J. Nutr. Biochem. 1999, 10, 353–359. [Google Scholar] [CrossRef]
  54. Fleurence, J. Seaweed Proteins; Woodhead Publishing Limited: Sawston, UK, 2004; ISBN 9781855737235. [Google Scholar]
  55. Lafarga, T.; Acién-fernández, F.G.; Garcia-vaquero, M. Bioactive peptides and carbohydrates from seaweed for food applications: Natural occurrence, isolation, purification, and identification. Algal Res. 2020, 48, 101909. [Google Scholar] [CrossRef]
  56. Prabhasankar, P.; Ganesan, P.; Bhaskar, N.; Hirose, A.; Stephen, N.; Gowda, L.R.; Hosokawa, M.; Miyashita, K. Edible Japanese seaweed, wakame (Undaria pinnatifida) as an ingredient in pasta: Chemical, functional and structural evaluation. Food Chem. 2009, 115, 501–508. [Google Scholar] [CrossRef]
  57. Rajapakse, N.; Kim, S.K. Nutritional and Digestive Health Benefits of Seaweed. Adv. Food Nutr. Res. 2011, 64, 17–28. [Google Scholar]
  58. Meinita, M.D.N.; Harwanto, D.; Sohn, J.-H.; Kim, J.-S.; Choi, J.-S. Hizikia fusiformis: Pharmacological and Nutritional Properties. Foods 2021, 10, 1660. [Google Scholar] [CrossRef] [PubMed]
  59. Meinita, M.D.N.; Harwanto, D.; Tirtawijaya, G.; Negara, B.F.S.P.; Sohn, J.H.; Kim, J.S.; Choi, J.S. Fucosterol of marine macroalgae: Bioactivity, safety and toxicity on organism. Mar. Drugs 2021, 19, 545. [Google Scholar] [CrossRef] [PubMed]
  60. 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]
  61. Gamero-Vega, G.; Palacios-Palacios, M.; Quitral, V. Nutritional Composition and Bioactive Compounds of Red Seaweed: A Mini-Review. J. Food Nutr. Res. 2020, 8, 431–440. [Google Scholar] [CrossRef]
  62. Choi, J.; Kim, K.J.; Koh, E.J.; Lee, B.Y. Gelidium elegans extract ameliorates type 2 diabetes via regulation of MAPK and PI3K/Akt signaling. Nutrients 2018, 10, 51. [Google Scholar] [CrossRef] [Green Version]
  63. Nisizawa, K.; Noda, H.; Kikuchi, R.; Watanabe, T. The main seaweed foods in Japan. Hydrobiologia 1987, 151–152, 5–29. [Google Scholar] [CrossRef]
  64. European Food Safety Authority. Scientific Opinion on Dietary Reference Values for carbohydrates and dietary fibre. EFSA J. 2010, 8, 1462. [Google Scholar] [CrossRef]
  65. Xu, S.Y.; Huang, X.; Cheong, K.L. Recent advances in marine algae polysaccharides: Isolation, structure, and activities. Mar. Drugs 2017, 15, 388. [Google Scholar] [CrossRef] [Green Version]
  66. MacArtain, P.; Gill, C.I.R.; Brooks, M.; Campbell, R.; Rowland, I.R. Nutritional value of edible seaweeds. Nutr. Rev. 2007, 65, 535–543. [Google Scholar] [CrossRef]
  67. Muraguri, E.N.; Wakibia, J.G.; Kinyuru, J.N. Chemical Composition and Functional Properties of Selected Seaweeds from the Kenya Coast. J. Food Res. 2016, 5, 114. [Google Scholar] [CrossRef]
  68. 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] [Green Version]
  69. Wang, Y.; Xing, M.; Cao, Q.; Ji, A.; Liang, H.; Song, S. Biological activities of fucoidan and the factors mediating its therapeutic effects: A review of recent studies. Mar. Drugs 2019, 17, 183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Salehi, B.; Sharifi-rad, J.; Seca, A.M.L.; Pinto, D.C.G.A. Current Trends on Seaweeds: Looking at Chemical. Molecules 2019, 24, 4182. [Google Scholar] [CrossRef] [Green Version]
  71. Yin, J.; Wang, J.; Li, F.; Yang, Z.; Yang, X.; Sun, W.; Xia, B.; Li, T.; Song, W.; Guo, S. The fucoidan from the brown seaweed: Ascophyllum nodosum ameliorates atherosclerosis in apolipoprotein E-deficient mice. Food Funct. 2019, 10, 5124–5139. [Google Scholar] [CrossRef]
  72. Wu, S.Y.; Yan, M.D.; Wu, A.T.H.; Yuan, K.S.P.; Liu, S.H. Brown Seaweed Fucoidan Inhibits Cancer Progression by Dual Regulation of mir-29c/ADAM12 and miR-17-5p/PTEN Axes in Human Breast Cancer Cells. J. Cancer 2016, 7, 2408–2419. [Google Scholar] [CrossRef] [Green Version]
  73. Meenakshi, S.; Umayaparvathi, S.; Saravanan, R.; Manivasagam, T.; Balasubramanian, T. International Journal of Biological Macromolecules Hepatoprotective effect of fucoidan isolated from the seaweed Turbinaria decurrens in ethanol intoxicated rats. Int. J. Biol. Macromol. 2014, 67, 367–372. [Google Scholar] [CrossRef]
  74. Rupérez, P.; Ahrazem, O.; Leal, J.A. Potential antioxidant capacity of sulfated polysaccharides from the edible marine brown seaweed Fucus vesiculosus. J. Agric. Food Chem. 2002, 50, 840–845. [Google Scholar] [CrossRef]
  75. 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]
  76. Patel, S. Therapeutic importance of sulfated polysaccharides from seaweeds: Updating the recent findings. 3 Biotech 2012, 2, 171–185. [Google Scholar] [CrossRef] [Green Version]
  77. Okolie, C.L.; Subin, S.R.; Udenigwe, C.C.; Aryee, A.N.A.; Mason, B. Prospects of brown seaweed polysaccharides (BSP) as prebiotics and potential immunomodulators. J. Food Biochem. 2017, 41, e12392. [Google Scholar] [CrossRef]
  78. Cotas, J.; Leandro, A.; Pacheco, D.; Gonçalves, A.M.M.; Pereira, L. A comprehensive review of the nutraceutical and therapeutic applications of red seaweeds (Rhodophyta). Life 2020, 10, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Hemmingson, J.A.; Furneaux, R.H.; Murray-Brown, V.H. Biosynthesis of agar polysaccharides in Gracilaria chilensis bird, McLachlan et Oliveira. Carbohydr. Res. 1996, 287, 101–115. [Google Scholar] [CrossRef]
  80. Patil, N.P.; Le, V.; Sligar, A.D.; Mei, L.; Chavarria, D.; Yang, E.Y.; Baker, A.B. Algal Polysaccharides as Therapeutic Agents for Atherosclerosis. Front. Cardiovasc. Med. 2018, 5, 153. [Google Scholar] [CrossRef] [Green Version]
  81. Torres, M.D.; Flórez-Fernández, N.; Domínguez, H. Integral utilization of red seaweed for bioactive production. Mar. Drugs 2019, 17, 314. [Google Scholar] [CrossRef] [Green Version]
  82. Jol, C.N.; Neiss, T.G.; Penninkhof, B.; Rudolph, B.; De Ruiter, G.A. A novel high-performance anion-exchange chromatographic method for the analysis of carrageenans and agars containing 3,6-anhydrogalactose. Anal. Biochem. 1999, 268, 213–222. [Google Scholar] [CrossRef]
  83. Necas, J.; Bartosikova, L. Carrageenan: A review. Vet. Med. (Praha) 2013, 58, 187–205. [Google Scholar] [CrossRef] [Green Version]
  84. Preez, R.; Paul, N.; Mouatt, P.; Majzoub, M.E.; Thomas, T.; Panchal, S.K.; Brown, L. Carrageenans from the Red Seaweed Sarconema filiforme Attenuate Symptoms of Diet-Induced Metabolic Syndrome in Rats. Mar. Drugs 2020, 18, 97. [Google Scholar] [CrossRef] [Green Version]
  85. Meinita, M.D.N.; Marhaeni, B.; Jeong, G.T.; Hong, Y.K. Sequential acid and enzymatic hydrolysis of carrageenan solid waste for bioethanol production: A biorefinery approach. J. Appl. Phycol. 2019, 31, 2507–2515. [Google Scholar] [CrossRef]
  86. Van De Velde, F.; Lourenço, N.D.; Pinheiro, H.M.; Bakker, M. Carrageenan: A Food-Grade and Biocompatible Support for Immobilisation Techniques. Adv. Synth. Catal. 2002, 344, 815–835. [Google Scholar] [CrossRef]
  87. Kidgell, J.T.; Magnusson, M.; de Nys, R.; Glasson, C.R.K. Ulvan: A systematic review of extraction, composition and function. Algal Res. 2019, 39, 101422. [Google Scholar] [CrossRef]
  88. Lahaye, M. NMR spectroscopic characterisation of oligosaccharides from two Ulva rigida ulvan samples (Ulvales, Chlorophyta) degraded by a lyase. Carbohydr. Res. 1998, 314, 1–12. [Google Scholar] [CrossRef]
  89. Bixler, H.J.; Porse, H. A decade of change in the seaweed hydrocolloids industry. J. Appl. Phycol. 2011, 23, 321–335. [Google Scholar] [CrossRef]
  90. Patterson, G.W. The distribution of sterols in algae. Lipids 1971, 6, 120–127. [Google Scholar] [CrossRef]
  91. Bogie, J.; Hoeks, C.; Schepers, M.; Tiane, A.; Cuypers, A.; Leijten, F.; Chintapakorn, Y.; Suttiyut, T.; Pornpakakul, S.; Struik, D.; et al. Dietary Sargassum fusiforme improves memory and reduces amyloid plaque load in an Alzheimer’s disease mouse model. Sci. Rep. 2019, 9, 4908. [Google Scholar] [CrossRef] [Green Version]
  92. Hannan, M.A.; Sohag, A.A.M.; Dash, R.; Haque, M.N.; Mohibbullah, M.; Oktaviani, D.F.; Hossain, M.T.; Choi, H.J.; Moon, I.S. Phytosterols of marine algae: Insights into the potential health benefits and molecular pharmacology. Phytomedicine 2020, 69, 153201. [Google Scholar] [CrossRef] [PubMed]
  93. Fernandes, P.; Cabral, J.M.S. Phytosterols: Applications and recovery methods. Bioresour. Technol. 2007, 98, 2335–2350. [Google Scholar] [CrossRef]
  94. Gupta, S.; Abu-ghannam, N. Bioactive potential and possible health effects of edible brown seaweeds. Trends Food Sci. Technol. 2011, 22, 315–326. [Google Scholar] [CrossRef] [Green Version]
  95. Xiao, X.H.; Yuan, Z.Q.; Li, G.K. Preparation of phytosterols and phytol from edible marine algae by microwave-assisted extraction and high-speed counter-current chromatography. Sep. Purif. Technol. 2013, 104, 284–289. [Google Scholar] [CrossRef]
  96. Kwon, T.H.; Wu, Y.X.; Kim, J.S.; Woo, J.H.; Park, K.T.; Kwon, O.J.; Seo, H.J.; Kim, T.; Park, N.H. 6,6′-Bieckol inhibits adipocyte differentiation through downregulation of adipogenesis and lipogenesis in 3T3-L1 cells. J. Sci. Food Agric. 2015, 95, 1830–1837. [Google Scholar] [CrossRef]
  97. Jung, H.A.; Roy, A.; Jung, J.H.; Choi, J.S. Evaluation of the inhibitory effects of eckol and dieckol isolated from edible brown alga Eisenia bicyclis on human monoamine oxidases A and B. Arch. Pharm. Res. 2017, 40, 480–491. [Google Scholar] [CrossRef]
  98. Myung, C.; Shin, H.; Bao, H.Y.; Yeo, S.J.; Lee, B.H.; Kang, J.S. Improvement of Memory by Dieckol and Phlorofucofuroeckol in Ethanol-Treated Mice: Possible Involvement of the Inhibition of Acetylcholinesterase. Arch. Pharm. Res. 2005, 28, 691–698. [Google Scholar] [CrossRef]
  99. Negara, B.F.S.P.; Sohn, J.H.; Kim, J.-S.; Choi, J.-S. Effects of Phlorotannins on Organisms: Focus on the Safety, Toxicity, and Availability of Phlorotannins. Foods 2021, 10, 452. [Google Scholar] [CrossRef]
  100. Lin, L.; Yang, S.; Xiao, Z.; Hong, P.; Sun, S.; Zhou, C.; Qian, Z.J. The Inhibition Effect of the Seaweed Polyphenol, 7-Phloro-Eckol from Ecklonia cava on Alcohol-Induced Oxidative Stress in HepG2/CYP2E1 Cells. Mar. Drugs 2021, 19, 158. [Google Scholar] [CrossRef]
  101. Lee, S.H.; Min, K.H.; Han, J.S.; Lee, D.H.; Park, D.B.; Jung, W.K.; Park, P.J.; Jeon, B.T.; Kim, S.K.; Jeon, Y.J. Effects of brown alga, Ecklonia cava on glucose and lipid metabolism in C57BL/KsJ-db/db mice, a model of type 2 diabetes mellitus. Food Chem. Toxicol. 2012, 50, 575–582. [Google Scholar] [CrossRef]
  102. Kang, M.C.; Wijesinghe, W.A.J.P.; Lee, S.H.; Kang, S.M.; Ko, S.C.; Yang, X.; Kang, N.; Jeon, B.T.; Kim, J.; Lee, D.H.; et al. Dieckol isolated from brown seaweed Ecklonia cava attenuates type II diabetes in db/db mouse model. Food Chem. Toxicol. 2013, 53, 294–298. [Google Scholar] [CrossRef]
  103. Bhatt, T.; Patel, K. Carotenoids: Potent to Prevent Diseases Review. Nat. Prod. Bioprospect. 2020, 10, 109–117. [Google Scholar] [CrossRef]
  104. Kong, Z.L.; Sudirman, S.; Hsu, Y.C.; Su, C.Y.; Kuo, H.P. Fucoxanthin-rich brown algae extract improves male reproductive function on streptozotocin- nicotinamide-induced diabetic rat model. Int. J. Mol. Sci. 2019, 20, 4485. [Google Scholar] [CrossRef] [Green Version]
  105. Grasa-López, A.; Miliar-García, Á.; Quevedo-Corona, L.; Paniagua-Castro, N.; Escalona-Cardoso, G.; Reyes-Maldonado, E.; Jaramillo-Flores, M.E. Undaria pinnatifida and fucoxanthin ameliorate lipogenesis and markers of both inflammation and cardiovascular dysfunction in an animal model of diet-induced obesity. Mar. Drugs 2016, 14, 148. [Google Scholar] [CrossRef] [Green Version]
  106. Paudel, P.; Seong, S.H.; Jung, H.A.; Choi, J.S. Characterizing fucoxanthin as a selective dopamine D3/D4 receptor agonist: Relevance to Parkinson’s disease. Chem. Biol. Interact. 2019, 310, 108757. [Google Scholar] [CrossRef]
  107. Yu, J.; Lin, J.J.; Yu, R.; He, S.; Wang, Q.W.; Cui, W.; Zhang, J.R. Fucoxanthin prevents H2O2-induced neuronal apoptosis via concurrently activating the PI3-K/Akt cascade and inhibiting the ERK pathway. Food Nutr. Res. 2017, 61, 1304678. [Google Scholar] [CrossRef] [Green Version]
  108. Takaichi, S. Carotenoids in Algae: Distributions, Biosyntheses and Functions. Mar. Drugs 2011, 9, 1101–1118. [Google Scholar] [CrossRef]
  109. Li, Z.; Zheng, J.; Luo, X.; Manabe, Y.; Hirata, T. Absorption and Tissue Distribution of Siphonaxanthin from Green Algae. Mar Drugs 2020, 18, 291. [Google Scholar] [CrossRef]
  110. Ziółkowska, N.E.; Wlodawer, A. Structural studies of algal lectins with anti-HIV activity. Acta Biochim. Pol. 2006, 53, 617–626. [Google Scholar] [CrossRef] [Green Version]
  111. Freire, M.; Alves, D.A.; Kalline, F.; Barreto, D.A.; De Vasconcelos, M.A.; Gonzaga, L.; Carneiro, R.F.; Torquato, L.; Nagano, C.S.; Sampaio, A.H.; et al. Antihyperglycemic and antioxidant activities of a lectin from the marine red algae, Bryothamnion seaforthii, in rats with streptozotocin-induced diabetes. Int. J. Biol. Macromol. 2020, 158, 773–780. [Google Scholar] [CrossRef]
  112. Line, R.; Vasconcelos, H.; Rocha, D.; Ramos, A.; Freitas, D.; Cavalcante, J.; Ariévilo, J.; Rodrigues, G.; Maria, K.; Pereira, A.; et al. International Immunopharmacology A lectin from the green seaweed Caulerpa cupressoides reduces mechanical hyper-nociception and in fl ammation in the rat temporomandibular joint during zymosan-induced arthritis. Int. Immunopharmacol. 2014, 21, 34–43. [Google Scholar] [CrossRef] [Green Version]
  113. De Souza, É.T.; De Lira, D.P.; De Queiroz, A.C.; Da Silva, D.J.C.; De Aquino, A.B.; Mella, E.A.C.; Lorenzo, V.P.; De Miranda, G.E.C.; De Araújo-Júnior, J.X.; Chaves, M.C.D.O.; et al. The antinociceptive and anti-inflammatory activities of caulerpin, a bisindole alkaloid isolated from seaweeds of the genus Caulerpa. Mar. Drugs 2009, 7, 689–704. [Google Scholar] [CrossRef] [Green Version]
  114. Liu, D.Q.; Mao, S.C.; Zhang, H.Y.; Yu, X.Q.; Feng, M.T.; Wang, B.; Feng, L.H.; Guo, Y.W. Racemosins A and B, two novel bisindole alkaloids from the green alga Caulerpa racemosa. Fitoterapia 2013, 91, 15–20. [Google Scholar] [CrossRef]
  115. Mickymaray, S.; Alturaiki, W. Antifungal efficacy of marine macroalgae against fungal isolates from bronchial asthmatic cases. Molecules 2018, 23, 3032. [Google Scholar] [CrossRef] [Green Version]
  116. Silva, J.; Alves, C.; Freitas, R.; Martins, A.; Pinteus, S.; Ribeiro, J.; Gaspar, H.; Alfonso, A.; Pedrosa, R. Antioxidant and Neuroprotective Potential of the Brown Seaweed Bifurcaria bifurcata in an in vitro Parkinson’s Disease Model. Mar. Drugs 2019, 17, 85. [Google Scholar] [CrossRef] [Green Version]
  117. 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]
  118. Shamsabadi, F.T.; Khoddami, A.; Fard, S.G.; Abdullah, R.; Othman, H.H.; Mohamed, S. Comparison of tamoxifen with edible seaweed (Eucheuma cottonii L.) Extract in suppressing breast tumor. Nutr. Cancer 2013, 65, 255–262. [Google Scholar] [CrossRef]
  119. Smyrniotopoulos, V.; Merten, C.; Firsova, D.; Fearnhead, H.; Tasdemir, D. Oxygenated Acyclic Diterpenes with Anticancer Activity from the Irish Brown Seaweed Bifurcaria bifurcata. Mar. Drugs 2020, 18, 581. [Google Scholar] [CrossRef]
  120. Lakshmi, T.P.; Vajravijayan, S.; Moumita, M.; Sakthivel, N.; Gunasekaran, K.; Krishna, R. A novel guaiane sesquiterpene derivative, guai-2-en-10α-ol, from Ulva fasciata Delile inhibits EGFR/PI3K/Akt signaling and induces cytotoxicity in triple-negative breast cancer cells. Mol. Cell. Biochem. 2018, 438, 123–139. [Google Scholar] [CrossRef]
  121. Abd-Ellatef, G.E.F.; Ahmed, O.M.; Abdel-Reheim, E.S.; Abdel-Hamid, A.H.Z. Ulva lactuca polysaccharides prevent Wistar rat breast carcinogenesis through the augmentation of apoptosis, enhancement of antioxidant defense system, and suppression of inflammation. Breast Cancer Targets Ther. 2017, 9, 67–83. [Google Scholar] [CrossRef] [Green Version]
  122. Song, J.; Won, S.Y.; Hwang, B.; Jung, S.; Choi, C. In Vitro and In Vivo Antitumor Efficacy of Hizikia fusiforme Celluclast Extract against. Nutrients 2020, 12, 2159. [Google Scholar] [CrossRef]
  123. 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] [Green Version]
  124. Kim, I.H.; Nam, T.J. Fucoidan downregulates insulin-like growth factor-I receptor levels in HT-29 human colon cancer cells. Oncol. Rep. 2018, 39, 1516–1522. [Google Scholar] [CrossRef] [Green Version]
  125. Cotas, J.; Marques, V.; Afonso, M.B.; Rodrigues, C.M.P.; Pereira, L. Antitumour potential of Gigartina pistillata carrageenans against colorectal cancer stem cell-enriched tumourspheres. Mar. Drugs 2020, 18, 50. [Google Scholar] [CrossRef] [Green Version]
  126. Lins, K.O.A.L.; Bezerra, D.P.; Alves, A.P.N.N.; Alencar, N.M.N.; Lima, M.W.; Torres, V.M.; Farias, W.R.L.; Pessoa, C.; De Moraes, M.O.; Costa-Lotufo, L.V. Antitumor properties of a sulfated polysaccharide from the red seaweed Champia feldmannii (Diaz-Pifferer). J. Appl. Toxicol. 2009, 29, 20–26. [Google Scholar] [CrossRef]
  127. Sae-lao, T.; Luplertlop, N.; Janvilisri, T.; Tohtong, R.; Bates, D.O.; Wongprasert, K. Sulfated galactans from the red seaweed Gracilaria fisheri exerts anti-migration effect on cholangiocarcinoma cells. Phytomedicine 2017, 36, 59–67. [Google Scholar] [CrossRef] [PubMed]
  128. Delgado-Roche, L.; Rodeiro, I.; Riera, M.; Herrera, J.A.; Venturi, I.; Hernández, Y.; Fernández, G.; Pérez, C.L.; Rodriguez, J.C.; Fernández, M.D.; et al. Chemoprotective effects of Ulva lactuca (green seaweed) aqueous-ethanolic extract against subchronic exposure to benzo(a)pyrene by CYP1A1 inhibition in mice. Phytother. Res. 2019, 33, 958–967. [Google Scholar] [CrossRef] [PubMed]
  129. Ropellato, J.; Carvalho, M.M.; Ferreira, L.G.; Noseda, M.D.; Zuconelli, C.R.; Gonçalves, A.G.; Ducatti, D.R.B.; Kenski, J.C.N.; Nasato, P.L.; Winnischofer, S.M.B.; et al. Sulfated heterorhamnans from the green seaweed Gayralia oxysperma: Partial depolymerization, chemical structure and antitumor activity. Carbohydr. Polym. 2015, 117, 476–485. [Google Scholar] [CrossRef] [PubMed]
  130. Kim, S.; Lee, M.S.; Lee, B.; Gwon, W.G.; Joung, E.J.; Yoon, N.Y.; Kim, H.R. Anti-inflammatory effects of sargachromenol-rich ethanolic extract of Myagropsis myagroides on lipopolysaccharide-stimulated BV-2 cells. BMC Complement. Altern. Med. 2014, 14, 231. [Google Scholar] [CrossRef] [Green Version]
  131. Tarozzi, A.; Angeloni, C.; Malaguti, M.; Morroni, F.; Hrelia, S.; Hrelia, P. Sulforaphane as a Potential Protective Phytochemical against Neurodegenerative Diseases. Oxid. Med. Cell. Longev. 2013, 2013, 415078. [Google Scholar] [CrossRef]
  132. Barbalace, M.C.; Malaguti, M.; Giusti, L.; Lucacchini, A.; Hrelia, S.; Angeloni, C. Anti-inflammatory activities of marine algae in neurodegenerative diseases. Int. J. Mol. Sci. 2019, 20, 3061. [Google Scholar] [CrossRef] [Green Version]
  133. Kim, J.; Moon, I.S.; Goo, T.; Moon, 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] [Green Version]
  134. Jung, H.A.; Ali, M.Y.; Choi, R.J.; Jeong, H.O.; Chung, H.Y.; Choi, J.S. Kinetics and molecular docking studies of fucosterol and fucoxanthin, BACE1 inhibitors from brown algae Undaria pinnatifida and Ecklonia stolonifera. Food Chem. Toxicol. 2016, 89, 104–111. [Google Scholar] [CrossRef]
  135. Kim, T.E.; Son, H.J.; Lim, D.W.; Yoon, M.; Lee, J.; Kim, Y.T.; Han, D.; Lee, C.; Um, M.Y. Memory-enhancing effects of Ishige foliacea extract: In vitro and in vivo study. J. Food Biochem. 2020, 44, e13162. [Google Scholar] [CrossRef]
  136. Kim, J.H.; Lee, N.S.; Jeong, Y.G.; Lee, J.-H.; Kim, E.J.; Han, S.Y. Protective efficacy of an Ecklonia cava extract used to treat transient focal ischemia of the rat brain. Anat. Cell Biol. 2012, 45, 103. [Google Scholar] [CrossRef] [Green Version]
  137. Kang, I.J.; Jeon, Y.E.; Yin, X.F.; Nam, J.S.; You, S.G.; Hong, M.S.; Jang, B.G.; Kim, M.J. Butanol extract of Ecklonia cava prevents production and aggregation of beta-amyloid, and reduces beta-amyloid mediated neuronal death. Food Chem. Toxicol. 2011, 49, 2252–2259. [Google Scholar] [CrossRef] [PubMed]
  138. Kang, I.J.; Jang, B.G.; In, S.; Choi, B.; Kim, M.; Kim, M.J. Phlorotannin-rich Ecklonia cava reduces the production of beta-amyloid by modulating alpha- and gamma-secretase expression and activity. Neurotoxicology 2013, 34, 16–24. [Google Scholar] [CrossRef]
  139. Kwon, O.Y.; Lee, S.H. Ameliorating Activity of Ishige okamurae on the Amyloid Beta-Induced Cognitive Deficits and Neurotoxicity through Regulating ERK, p38 MAPK, and JNK Signaling in Alzheimer′s Disease-Like Mice Model. Mol. Nutr. Food Res. 2020, 64, e1901220. [Google Scholar] [CrossRef]
  140. Olasehinde, T.A.; Olaniran, A.O.; Okoh, A.I. Phenolic composition, antioxidant activity, anticholinesterase potential and modulatory effects of aqueous extracts of some seaweeds on β-amyloid aggregation and disaggregation. Pharm. Biol. 2019, 57, 460–469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Souza, R.B.; Frota, A.F.; Sousa, R.S.; Cezario, N.A.; Santos, T.B.; Souza, L.M.F.; Coura, C.O.; Monteiro, V.S.; Filho, G.C.; Vasconcelos, S.M.M.; et al. Neuroprotective Effects of Sulphated Agaran from Marine Alga Gracilaria cornea in Rat 6-Hydroxydopamine Parkinson’s Disease Model: Behavioural, Neurochemical and Transcriptional Alterations. Basic Clin. Pharmacol. Toxicol. 2017, 120, 159–170. [Google Scholar] [CrossRef] [PubMed]
  142. Hannan, M.A.; Kang, J.Y.; Hong, Y.K.; Lee, H.; Choi, J.S.; Choi, I.S.; Moon, I.S. The marine alga Gelidium amansii promotes the development and complexity of neuronal cytoarchitecture. Phyther. Res. 2013, 27, 21–29. [Google Scholar] [CrossRef]
  143. Kim, M.J.; Li, Y.X.; Dewapriya, P.; Ryu, B.; Kim, S.K. Floridoside suppresses pro-inflammatory responses by blocking MAPK signaling in activated microglia. BMB Rep. 2013, 46, 398–403. [Google Scholar] [CrossRef]
  144. Pereira, L.; Valado, A. The Seaweed Diet in Prevention and Treatment of the. Mar. Drugs 2021, 19, 128. [Google Scholar] [CrossRef]
  145. Niizuma, K.; Endo, H.; Chan, P.H. Oxidative stress and mitochondrial dysfunction as determinants of ischemic neuronal death and survival. J. Neurochem. 2009, 109, 133–138. [Google Scholar] [CrossRef] [Green Version]
  146. Ramkumar, M.; Rajasankar, S.; Gobi, V.V.; Dhanalakshmi, C.; Manivasagam, T.; Justin Thenmozhi, A.; Essa, M.M.; Kalandar, A.; Chidambaram, R. Neuroprotective effect of Demethoxycurcumin, a natural derivative of Curcumin on rotenone induced neurotoxicity in SH-SY 5Y Neuroblastoma cells. BMC Complement. Altern. Med. 2017, 17, 217. [Google Scholar] [CrossRef] [Green Version]
  147. Ward, R.J.; Zucca, F.A.; Duyn, J.H.; Crichton, R.R.; Zecca, L. The role of iron in brain ageing and neurodegenerative disorders. Lancet Neurol. 2014, 13, 1045–1060. [Google Scholar] [CrossRef] [Green Version]
  148. Li, K.; Reichmann, H. Role of iron in neurodegenerative diseases. J. Neural Transm. 2016, 123, 389–399. [Google Scholar] [CrossRef]
  149. Silva, J.; Alves, C.; Pinteus, S.; Mendes, S.; Pedrosa, R. Seaweeds’ neuroprotective potential set in vitro on a human cellular stress model. Mol. Cell. Biochem. 2020, 473, 229–238. [Google Scholar] [CrossRef] [PubMed]
  150. Waly, M.I.; Al Alawi, A.A.; Al Marhoobi, I.M.; Rahman, M.S. Red seaweed (Hypnea bryodies and Melanothamnus somalensis) extracts counteracting azoxymethane-induced hepatotoxicity in rats. Asian Pac. J. Cancer Prev. 2016, 17, 5071–5074. [Google Scholar] [CrossRef] [PubMed]
  151. Sohail, N.; Hira, K.; Tariq, A.; Sultana, V.; Ehteshamul-Haque, S. Marine macro-algae attenuates nephrotoxicity and hepatotoxicity induced by cisplatin and acetaminophen in rats. Environ. Sci. Pollut. Res. 2019, 26, 25301–25311. [Google Scholar] [CrossRef] [PubMed]
  152. Wong, C.K.; Ooi, V.E.C.; Ang, P.O. Protective effects of seaweeds against liver injury caused by carbon tetrachloride in rats. Chemosphere 2000, 41, 173–176. [Google Scholar] [CrossRef]
  153. Tsai, M.Y.; Yang, W.C.; Lin, C.F.; Wang, C.M.; Liu, H.Y.; Lin, C.S.; Lin, J.W.; Lin, W.L.; Lin, T.C.; Fan, P.S.; et al. The ameliorative effects of fucoidan in thioacetaide-induced liver injury in mice. Molecules 2021, 26, 1937. [Google Scholar] [CrossRef] [PubMed]
  154. Gabbia, D.; Dall’Acqua, S.; Di Gangi, I.M.; Bogialli, S.; Caputi, V.; Albertoni, L.; Marsilio, I.; Paccagnella, N.; Carrara, M.; Giron, M.C.; et al. The Phytocomplex from Fucus vesiculosus and ascophyllum nodosum controls postprandial plasma glucose levels: An in vitro and in Vivo study in a mouse model of NASH. Mar. Drugs 2017, 15, 41. [Google Scholar] [CrossRef]
  155. Thomes, P.; Rajendran, M.; Pasanban, B.; Rengasamy, R. Phytomedicine Cardioprotective activity of Cladosiphon okamuranus fucoidan against isoproterenol induced myocardial infarction in rats. Eur. J. Integr. Med. 2010, 18, 52–57. [Google Scholar] [CrossRef]
  156. Cha, S.H.; Hwang, Y.; Heo, S.J.; Jun, H.S. Indole-4-carboxaldehyde Isolated from Seaweed, Sargassum thunbergii, Attenuates Methylglyoxal-Induced Hepatic Inflammation. Mar. Drugs 2019, 17, 486. [Google Scholar] [CrossRef] [Green Version]
  157. Jin, M.; Liu, H.; Hou, Y.; Chan, Z.; Di, W.; Li, L.; Zeng, R. Preparation, characterization and alcoholic liver injury protective effects of algal oligosaccharides from Gracilaria lemaneiformis. Food Res. Int. 2017, 100, 186–195. [Google Scholar] [CrossRef] [PubMed]
  158. Lieber, S. Alcohol and the Liver: 1994 Update. Gastroenterology 1994, 106, 1085–1105. [Google Scholar] [CrossRef]
  159. Heeba, G.H.; Morsy, M.A. Fucoidan ameliorates steatohepatitis and insulin resistance by suppressing oxidative stress and inflammatory cytokines in experimental non-alcoholic fatty liver disease. Environ. Toxicol. Pharmacol. 2015, 40, 907–914. [Google Scholar] [CrossRef] [PubMed]
  160. Friedman, S.L.; Neuschwander-Tetri, B.A.; Rinella, M.; Sanyal, A.J. Mechanisms of NAFLD development and therapeutic strategies. Nat. Med. 2018, 24, 908–922. [Google Scholar] [CrossRef]
  161. Takatani, N.; Kono, Y.; Beppu, F.; Okamatsu-Ogura, Y.; Yamano, Y.; Miyashita, K.; Hosokawa, M. Fucoxanthin inhibits hepatic oxidative stress, inflammation, and fibrosis in diet-induced nonalcoholic steatohepatitis model mice. Biochem. Biophys. Res. Commun. 2020, 528, 305–310. [Google Scholar] [CrossRef]
  162. Jia, R.; Li, Z.; Wu, J.; Ou, Z.; Liao, B.; Sun, B.; Lin, L.; Zhao, M. Mitigation mechanisms of Hizikia fusifarme polysaccharide consumption on type 2 diabetes in rats. Int. J. Biol. Macromol. 2020, 164, 2659–2670. [Google Scholar] [CrossRef]
  163. Yang, T.H.; Yao, H.T.; Chiang, M.T. Red algae (Gelidium amansii) reduces adiposity via activation of lipolysis in rats with diabetes induced by streptozotocin-nicotinamide. J. Food Drug Anal. 2015, 23, 758–765. [Google Scholar] [CrossRef] [Green Version]
  164. Yokogawa, K.; Matsui-Yuasa, I.; Tamura, A.; Terada, M.; Kojima-Yuasa, A. Inhibitory effects of Ecklonia cava extract on high glucose-induced hepatic stellate cell activation. Mar. Drugs 2011, 9, 2793–2808. [Google Scholar] [CrossRef] [Green Version]
  165. Lin, H.T.V.; Tsou, Y.C.; Chen, Y.T.; Lu, W.J.; Hwang, P.A. Effects of low-molecular-weight fucoidan and high stability fucoxanthin on glucose homeostasis, lipid metabolism, and liver function in a mouse model of type II diabetes. Mar. Drugs 2017, 15, 113. [Google Scholar] [CrossRef] [Green Version]
  166. Yu, W.-C.; Huang, R.-Y.; Chou, T.-C. Oligo-Fucoidan Improves Diabetes-Induced Renal Fibrosis via Activation of Sirt-1, GLP-1R, and Nrf2/HO-1: An In Vitro and In Vivo Study. Nutrients 2020, 12, 3068. [Google Scholar] [CrossRef]
  167. Park, M.H.; Han, J.S. Padina arborescens Ameliorates Hyperglycemia and Dyslipidemia in C57BL/KsJ-db/db Mice, a Model of Type 2 Diabetes Mellitus. J. Med. Food 2015, 18, 1088–1094. [Google Scholar] [CrossRef]
  168. Attjioui, M.; Ryan, S.; Ristic, A.K.; Higgins, T.; Goñi, O.; Gibney, E.R.; Tierney, J.; O’Connell, S. Comparison of edible brown algae extracts for the inhibition of intestinal carbohydrate digestive enzymes involved in glucose release from the diet. J. Nutr. Sci. 2021, 10, e5. [Google Scholar] [CrossRef] [PubMed]
  169. Kim, K.T.; Rioux, L.E.; Turgeon, S.L. Alpha-amylase and alpha-glucosidase inhibition is differentially modulated by fucoidan obtained from Fucus vesiculosus and Ascophyllum nodosum. Phytochemistry 2014, 98, 27–33. [Google Scholar] [CrossRef] [PubMed]
  170. Lee, S.H.; Kang, S.M.; Ko, S.C.; Lee, D.H.; Jeon, Y.J. Octaphlorethol A, a novel phenolic compound isolated from a brown alga, Ishige foliacea, increases glucose transporter 4-mediated glucose uptake in skeletal muscle cells. Biochem. Biophys. Res. Commun. 2012, 420, 576–581. [Google Scholar] [CrossRef] [PubMed]
  171. Lee, S.H.; Ko, S.C.; Kang, M.C.; Lee, D.H.; Jeon, Y.J. Octaphlorethol A, a marine algae product, exhibits antidiabetic effects in type 2 diabetic mice by activating AMP-activated protein kinase and upregulating the expression of glucose transporter 4. Food Chem. Toxicol. 2016, 91, 58–64. [Google Scholar] [CrossRef] [PubMed]
  172. Motshakeri, M.; Ebrahimi, M.; Goh, Y.M.; Othman, H.H.; Hair-bejo, M.; Mohamed, S. Effects of Brown Seaweed (Sargassum polycystum) Extracts on Kidney, Liver, and Pancreas of Type 2 Diabetic Rat Model. Evid. Based Complement. Altern. Med. 2014, 2014, 379407. [Google Scholar] [CrossRef] [Green Version]
  173. Jia, R.B.; Wu, J.; Li, Z.R.; Ou, Z.R.; Lin, L.; Sun, B.; Zhao, M. Structural characterization of polysaccharides from three seaweed species and their hypoglycemic and hypolipidemic activities in type 2 diabetic rats. Int. J. Biol. Macromol. 2020, 155, 1040–1049. [Google Scholar] [CrossRef]
  174. Moon, H.E.; Islam, M.N.; Ahn, B.R.; Chowdhury, S.S.; Sohn, H.S.; Jung, H.A.; Choi, J.S. Protein tyrosine phosphatase 1B and α-glucosidase inhibitory phlorotannins from edible brown algae, Ecklonia stolonifera and Eisenia bicyclis. Biosci. Biotechnol. Biochem. 2011, 75, 1472–1480. [Google Scholar] [CrossRef] [Green Version]
  175. Tas, S.; Celikler, S.; Ziyanok-Ayvalik, S.; Sarandol, E.; Dirican, M. Ulva rigida improves carbohydrate metabolism, hyperlipidemia and oxidative stress in streptozotocin-induced diabetic rats. Cell Biochem. Funct. 2011, 29, 108–113. [Google Scholar] [CrossRef]
  176. Baron, A.D. Postprandial hyperglycaemia and α-glucosidase inhibitors. Diabetes Res. Clin. Pract. 1998, 40, 51–55. [Google Scholar] [CrossRef]
  177. Snijder, M.B.; Zimmet, P.Z.; Visser, M.; Dekker, J.M.; Seidell, J.C.; Shaw, J.E. Independent and opposite associations of waist and hip circumferences with diabetes, hypertension and dyslipidemia: The AusDiab Study. Int. J. Obes. 2004, 28, 402–409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Calle, E.E.; Thun, M.J. Obesity and cancer. Oncogene 2004, 23, 6365–6378. [Google Scholar] [CrossRef] [PubMed]
  179. Kim, D.S.; Lee, S.G.; Kim, M.; Hahn, D.; Jung, S.K.; Cho, T.O.; Nam, J.O. Sargassum miyabei yendo brown algae exert anti-oxidative and anti-adipogeniceffects on 3t3-l1 adipocytes by downregulating pparγ. Medicina 2020, 56, 634. [Google Scholar] [CrossRef] [PubMed]
  180. Huebbe, P.; Nikolai, S.; Schloesser, A.; Herebian, D.; Glüer, C.; Zeyner, A.; Demetrowitsch, T.; Metges, C.C.; Roeder, T.; Schultheiss, G.; et al. An extract from the Atlantic brown algae Saccorhiza polyschides counteracts diet-induced obesity in mice via a gut related multi-factorial mechanisms. Oncotarget 2017, 8, 73501–73515. [Google Scholar] [CrossRef] [Green Version]
  181. Park, M.K.; Jung, U.; Roh, C. Fucoidan from marine brown algae inhibits lipid accumulation. Mar. Drugs 2011, 9, 1359–1367. [Google Scholar] [CrossRef] [Green Version]
  182. Kang, S.I.; Kim, M.H.; Shin, H.S.; Kim, H.M.; Hong, Y.S.; Park, J.G.; Ko, H.C.; Lee, N.H.; Chung, W.S.; Kim, S.J. A water-soluble extract of Petalonia binghamiae inhibits the expression of adipogenic regulators in 3T3-L1 preadipocytes and reduces adiposity and weight gain in rats fed a high-fat diet. J. Nutr. Biochem. 2010, 21, 1251–1257. [Google Scholar] [CrossRef]
  183. Jung, H.A.; Jung, H.J.; Jeong, H.Y.; Kwon, H.J.; Kim, M.S.; Choi, J.S. Anti-adipogenic activity of the edible brown alga Ecklonia stolonifera and its constituent fucosterol in 3T3-L1 adipocytes. Arch. Pharm. Res. 2014, 37, 713–720. [Google Scholar] [CrossRef]
  184. Lee, J.H.; Jung, H.A.; Kang, M.J.; Choi, J.S.; Kim, G. Do Fucosterol, isolated from Ecklonia stolonifera, inhibits adipogenesis through modulation of FoxO1 pathway in 3T3-L1 adipocytes. J. Pharm. Pharmacol. 2017, 69, 325–333. [Google Scholar] [CrossRef]
  185. Karadeniz, F.; Ahn, B.N.; Kim, J.A.; Seo, Y.; Jang, M.S.; Nam, K.H.; Kim, M.; Lee, S.H.; Kong, C.S. Phlorotannins suppress adipogenesis in pre-adipocytes while enhancing osteoblastogenesis in pre-osteoblasts. Arch. Pharm. Res. 2015, 38, 2172–2182. [Google Scholar] [CrossRef]
  186. Eo, H.; Jeon, Y.J.; Lee, M.; Lim, Y. Brown alga Ecklonia cava polyphenol extract ameliorates hepatic lipogenesis, oxidative stress, and inflammation by activation of AMPK and SIRT1 in high-fat diet-induced obese mice. J. Agric. Food Chem. 2015, 63, 349–359. [Google Scholar] [CrossRef]
  187. Kang, J.H.; Lee, H.A.; Kim, H.J.; Han, J.S. Gelidium amansii extract ameliorates obesity by down-regulating adipogenic transcription factors in diet-induced obese mice. Nutr. Res. Pract. 2017, 11, 17–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Kang, M.C.; Kang, N.; Ko, S.C.; Kim, Y.B.; Jeon, Y.J. Anti-obesity effects of seaweeds of Jeju Island on the differentiation of 3T3-L1 preadipocytes and obese mice fed a high-fat diet. Food Chem. Toxicol. 2016, 90, 36–44. [Google Scholar] [CrossRef] [PubMed]
  189. Zheng, J.; Manabe, Y.; Sugawara, T. Siphonaxanthin, a carotenoid from green algae Codium cylindricum, protects Ob/Ob mice fed on a high-fat diet against lipotoxicity by ameliorating somatic stresses and restoring anti-oxidative capacity. Nutr. Res. 2020, 77, 29–42. [Google Scholar] [CrossRef] [PubMed]
  190. Sudirman, S.; Chang, H.W.; Chen, C.K.; Kong, Z.L. A dietary polysaccharide from: Eucheuma cottonii downregulates proinflammatory cytokines and ameliorates osteoarthritis-associated cartilage degradation in obese rats. Food Funct. 2019, 10, 5697–5706. [Google Scholar] [CrossRef]
  191. Du Preez, R.; Majzoub, M.E.; Thomas, T.; Panchal, S.K.; Brown, L. Caulerpa lentillifera (Sea grapes) improves cardiovascular and metabolic health of rats with diet-induced metabolic syndrome. Metabolites 2020, 10, 500. [Google Scholar] [CrossRef]
  192. Yuan, X.; Nakao, T.; Satone, H.; Ohara, K.; Kominami, Y.; Ito, M.; Aizawa, T.; Ueno, T.; Ushio, H. The effects of brown algae-derived monosaccharide l-fucose on lipid metabolism in c57bl/6j obese mice. Nutrients 2020, 12, 3798. [Google Scholar] [CrossRef]
  193. Kishida, R.; Yamagishi, K.; Muraki, I.; Sata, M.; Tamakoshi, A.; Iso, H. Frequency of Seaweed Intake and Its Association with Cardiovascular Disease Mortality: The JACC Study. J. Atheroscler. Thromb. 2020, 27, 1340–1347. [Google Scholar] [CrossRef] [Green Version]
  194. Cardoso, S.M.; Pereira, O.R.; Seca, A.M.L.; Pinto, D.C.G.A.; Silva, A.M.S. Seaweeds as preventive agents for cardiovascular diseases: From nutrients to functional foods. Mar. Drugs 2015, 13, 6838–6865. [Google Scholar] [CrossRef] [Green Version]
  195. Ren, D.; Wang, Q.; Yang, Y.; Hu, Y.; Song, Y.; He, Y.; Liu, S.; Wu, L. Hypolipidemic effects of fucoidan fractions from Saccharina sculpera (Laminariales, Phaeophyceae). Int. J. Biol. Macromol. 2019, 140, 188–195. [Google Scholar] [CrossRef]
  196. Ara, J.; Sultana, V.; Qasim, R.; Ahmad, V.U. Hypolipidaemic activity of seaweed from Karachi coast. Phytother. Res. 2002, 16, 479–483. [Google Scholar] [CrossRef]
  197. Yang, Z.; Yin, J.; Wang, Y.; Wang, J.; Xia, B.; Li, T.; Yang, X.; Hu, S.; Ji, C.; Guo, S. The fucoidan A3 from the seaweed Ascophyllum nodosum enhances RCT-related genes expression in hyperlipidemic C57BL/6J mice. Int. J. Biol. Macromol. 2019, 134, 759–769. [Google Scholar] [CrossRef] [PubMed]
  198. Ahn, H.S.; Lee, D.H.; Kim, T.J.; Shin, H.C.; Jeon, H.K. Cardioprotective Effects of a Phlorotannin Extract Against Doxorubicin-Induced Cardiotoxicity in a Rat Model. J. Med. Food 2017, 20, 944–950. [Google Scholar] [CrossRef] [PubMed]
  199. Paiva, L.; Lima, E.; Neto, A.I.; Baptista, J. Angiotensin I-converting enzyme (ACE) inhibitory activity, antioxidant properties, phenolic content and amino acid profiles of fucus spiralis L. Protein hydrolysate fractions. Mar. Drugs 2017, 15, 311. [Google Scholar] [CrossRef] [PubMed]
  200. Park, B.G.; Shin, W.S.; Oh, S.; Park, G.M.; Kim, N.I.; Lee, S. A novel antihypertension agent, sargachromenol D from marine brown algae, Sargassum siliquastrum, exerts dual action as an L-type Ca2+ channel blocker and endothelin A/B2 receptor antagonist. Bioorg. Med. Chem. 2017, 25, 4649–4655. [Google Scholar] [CrossRef]
  201. Yeo, A.R.; Lee, J.; Tae, I.H.; Park, S.R.; Cho, Y.H.; Lee, B.H.; Shin, H.C.; Kim, S.H.; Yoo, Y.C. Anti-hyperlipidemic effect of polyphenol extract (SeapolynolTM) and dieckol isolated from Ecklonia cava in in vivo and in vitro models. Prev. Nutr. Food Sci. 2012, 17, 1–7. [Google Scholar] [CrossRef]
  202. Zhang, Q.; Fan, X.Y.; Guo, W.L.; Cao, Y.J.; Lin, Y.C.; Cheng, W.J.; Chen, L.J.; Rao, P.F.; Ni, L.; Lv, X.C. The protective mechanisms of macroalgae: Laminaria japonica consumption against lipid metabolism disorders in high-fat diet-induced hyperlipidemic rats. Food Funct. 2020, 11, 3256–3270. [Google Scholar] [CrossRef]
  203. Ryu, B.M.; Li, Y.; Qian, Z.J.; Kim, M.M.; Kim, S.K. Differentiation of human osteosarcoma cells by isolated phlorotannins is subtly linked to COX-2, iNOS, MMPs, and MAPK signaling: Implication for chronic articular disease. Chem. Biol. Interact. 2009, 179, 192–201. [Google Scholar] [CrossRef]
  204. Lee, D.; Hwang, E.; Park, S.; Chung, W.; Park, J.; Mavlonov, G.T.; Kim, I.; Kim, K.; Yi, T. Fucoidan Prevents the Progression of Osteoarthritis in Rats. J. Med. Food 2015, 18, 1032–1041. [Google Scholar] [CrossRef]
  205. Stoustrup, P.; Lerman, M.A.; Twilt, M. The Temporomandibular Joint in Juvenile Idiopathic Arthritis. Rheum. Dis. Clin. N. Am. 2021, 47, 607–617. [Google Scholar] [CrossRef]
  206. Poddubnyy, D.; Jadon, D.R.; Bosch, F.V.D.; Mease, P.J.; Gladman, D.D. Axial involvement in psoriatic arthritis: An update for rheumatologists. Semin. Arthritis Rheum. 2021, 51, 880–887. [Google Scholar] [CrossRef]
  207. Patil, T.; Soni, A.; Acharya, S. A brief review on in vivo models for Gouty Arthritis. Metab. Open 2021, 11, 100100. [Google Scholar] [CrossRef] [PubMed]
  208. Cush, J.J. Rheumatoid Arthritis: Early Diagnosis and Treatment. Med. Clin. N. Am. 2021, 105, 355–365. [Google Scholar] [CrossRef]
  209. Sun, K.; Luo, J.; Guo, J.; Yao, X.; Jing, X.; Guo, F. The PI3K/AKT/mTOR signaling pathway in osteoarthritis: A narrative review. Osteoarthr. Cartil. 2020, 28, 400–409. [Google Scholar] [CrossRef] [PubMed]
  210. Paiva, A.A.D.O.; Castro, A.J.G.; Nascimento, M.S.; Will, L.S.E.P.; Santos, N.D.; Araújo, R.M.; Xavier, C.A.C.; Rocha, F.A.; Leite, E.L. Antioxidant and anti-inflammatory effect of polysaccharides from Lobophora variegata on zymosan-induced arthritis in rats. Int. Immunopharmacol. 2011, 11, 1241–1250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  211. Sarithakumari, C.H.; Renju, G.L.; Kurup, G.M. Anti-inflammatory and antioxidant potential of alginic acid isolated from the marine algae, Sargassum wightii on adjuvant-induced arthritic rats. Inflammopharmacology 2013, 21, 261–268. [Google Scholar] [CrossRef] [PubMed]
  212. Chatter, R.; Ben Othman, R.; Rabhi, S.; Kladi, M.; Tarhouni, S.; Vagias, C.; Roussis, V.; Guizani-Tabbane, L.; Kharrat, R. In vivo and in vitro anti-inflammatory activity of neorogioltriol, a new diterpene extracted from the red algae Laurencia glandulifera. Mar. Drugs 2011, 9, 1293–1306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Moon, S.M.; Lee, S.A.; Hong, J.H.; Kim, J.S.; Kim, D.K.; Kim, C.S. Oleamide suppresses inflammatory responses in LPS-induced RAW264.7 murine macrophages and alleviates paw edema in a carrageenan-induced inflammatory rat model. Int. Immunopharmacol. 2018, 56, 179–185. [Google Scholar] [CrossRef]
  214. Lacey, D.C.; Simmons, P.J.; Graves, S.E.; Hamilton, J.A. Proinflammatory cytokines inhibit osteogenic differentiation from stem cells: Implications for bone repair during inflammation. Osteoarthr. Cartil. 2009, 17, 735–742. [Google Scholar] [CrossRef] [Green Version]
  215. Nekooei, M.; Shafiee, S.M.; Zahiri, M.; Maryamabadi, A.; Nabipour, I. The methanol extract of red algae, Dichotomaria obtusata, from Persian Gulf promotes in vitro osteogenic differentiation of bone marrow mesenchymal stem cells; A biological and phytochemical study. J. Pharm. Pharmacol. 2021, 73, 347–356. [Google Scholar] [CrossRef]
  216. Carson, M.A.; Nelson, J.; Cancela, M.L.; Laizé, V.; Gavaia, P.J.; Rae, M.; Heesch, S.; Verzin, E.; Gilmore, B.F.; Clarke, S.A. Screening for osteogenic activity in extracts from Irish marine organisms: The potential of Ceramium pallidum. PLoS ONE 2018, 13, e0207303. [Google Scholar] [CrossRef]
  217. Fidelis, G.P.; Ferreira Silva, C.H.; Nobre, L.T.D.B.; Medeiros, V.P.; Rocha, H.A.O.; Costa, L.S. Antioxidant fucoidans obtained from tropical seaweed protect pre-osteoblastic cells from hydrogen peroxide-induced damage. Mar. Drugs 2019, 17, 506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Minetti, M.; Bernardini, G.; Biazzo, M.; Gutierrez, G.; Geminiani, M.; Petrucci, T.; Santucci, A. Padina pavonica Extract Promotes In Vitro Differentiation and Functionality of Human Primary Osteoblasts. Mar. Drugs 2019, 17, 473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Lu, S.H.; Hsia, Y.J.; Shih, K.C.; Chou, T.C. Fucoidan prevents RANKL-stimulated osteoclastogenesis and LPS-induced inflammatory bone loss via regulation of AKt/GSK3β/PTEN/NFATc1 signaling pathway and calcineurin activity. Mar. Drugs 2019, 17, 345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  220. Lee, S.H.; Kim, M.; Park, M.H. Diphlorethohydroxycamalol isolated from Ishige okamurae prevents H2O2-induced oxidative damage via BMP2/Runx2 signaling in osteoblastic MC3T3-E1 cells. Fitoterapia 2021, 152, 104921. [Google Scholar] [CrossRef] [PubMed]
  221. Carson, M.A.; Nelson, J.; Cancela, M.L.; Laizé, V.; Gavaia, P.J.; Rae, M.; Heesch, S.; Verzin, E.; Maggs, C.; Gilmore, B.F.; et al. Red algal extracts from Plocamium lyngbyanum and Ceramium secundatum stimulate osteogenic activities in vitro and bone growth in zebrafish larvae. Sci. Rep. 2018, 8, 7725. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Surget, G.; Roberto, V.P.; Le Lann, K.; Mira, S.; Guérard, F.; Laizé, V.; Poupart, N.; Cancela, M.L. Marine green macroalgae: A source of natural compounds with mineralogenic and antioxidant activities. J. Appl. Phycol. 2016, 29, 575–584. [Google Scholar] [CrossRef] [Green Version]
  223. Mironidou-Tzouveleki, Μ.; Dokos, C.H.; Dokou, K. Effects of extracts of marine algae on osteoporosis. Aristotle Univ. Med. J. 2008, 35, 7–12. [Google Scholar]
  224. Boyle, W.J.; Simonet, W.S.; Lacey, D.L. Osteoclast differentiation and activation. Nature 2003, 423, 337–342. [Google Scholar] [CrossRef]
Figure 1. Number of publications on functional seaweed and chronic disease based on (a) the type of chronic disease and (b) the experimental model.
Figure 1. Number of publications on functional seaweed and chronic disease based on (a) the type of chronic disease and (b) the experimental model.
Applsci 12 02638 g001
Figure 2. Percentage of published papers of seaweed research on chronic disease based on (a) the phyla Chlorophyta (green color), Rhodophyta (red color), and Phaeophyta (brown color); (b) chronic disease for each phylum; and (c) experimental model for each phylum.
Figure 2. Percentage of published papers of seaweed research on chronic disease based on (a) the phyla Chlorophyta (green color), Rhodophyta (red color), and Phaeophyta (brown color); (b) chronic disease for each phylum; and (c) experimental model for each phylum.
Applsci 12 02638 g002
Figure 3. The chemical structures of fucoidan (1), agars (2), kappa carrageenans (3), ulvans (4), alginates (5), laminarin (6), fucosterol (7), phlorotannins (8), lectins (9), alkaloids (10), diterpenes (11), and fucoxanthin (12).
Figure 3. The chemical structures of fucoidan (1), agars (2), kappa carrageenans (3), ulvans (4), alginates (5), laminarin (6), fucosterol (7), phlorotannins (8), lectins (9), alkaloids (10), diterpenes (11), and fucoxanthin (12).
Applsci 12 02638 g003
Figure 4. Mechanisms of chronic diseases, including cancer (a), neurodegenerative disease (b), alcoholic liver disease (c), diabetes (d), obesity (e), cardiovascular disease (f), osteoarthritis (g) and osteoporosis (h).
Figure 4. Mechanisms of chronic diseases, including cancer (a), neurodegenerative disease (b), alcoholic liver disease (c), diabetes (d), obesity (e), cardiovascular disease (f), osteoarthritis (g) and osteoporosis (h).
Applsci 12 02638 g004
Table 1. The effects of seaweed-derived compounds on cancer.
Table 1. The effects of seaweed-derived compounds on cancer.
Algal SourceConstituentStudy TypeBiological EffectsRef.
Brown seaweed
Sargassum hemiphyllumFucoidanIn vitroInhibited the progression of MCF-7 and MDA-MB-231 cell lines[72]
Bifurcaria bifurcataDiterpenesIn vitroInhibited the growth of MDA-MB-231 cell line (IC50 = 11.6 to 32.0 µg/mL)[119]
Sargassum sp.Ethanol extractIn vitroInhibited the growth of MCF-7 cell line (IC50 = 250 µg/mL)[31]
Hizikia fusiformeSulfated polysaccharideIn vitro and in vivoInhibited the growth of human bladder cancer EJ cell line[122]
Laminaria japonicaLaminarinIn vitro and in vivoInhibited hepatocellular carcinoma (HCC) cell proliferation[123]
Fucus vesiculosusFucoidanIn vitroInhibited HT-29 cell prolifeation[124]
Red Seaweed
Laurencia papillosaSulfated polysaccharidesIn vitroInhibited MCF-7 cell viability[117]
Eucheuma cottoniiEthanol extractIn vivoInhibited the growth of breast tumor[118]
Gigartina pistillataCarrageenansIn vitroInhibited colorectal cancer stem cell viability (IC50 = 1 µg/mL)[125]
Champia feldmanniiSulfated polysaccharidesIn vitro and in vivoInhibited sarcoma 180 ascites tumor cell growth[126]
Gracilaria fisherSulfated galactansIn vitroRestored migration of CCA cells[127]
Green Seaweed
Ulva fasciataGuai-2-en-10a-olIn vitroInhibited the growth of MDA-MB-231 cell line[120]
Ulva lactucaUlvanIn vitro and in vivoInhibited MCF-7 cell viability
Inhibited breast carcinogenesis
[121]
Ulva lactucaAqueous-ethanolic extractIn vivoInhibited benzo(a)pyrene-induced toxicity in mice[128]
Gayralia oxyspermaSulfated heterorhamnansIn vitroInhibited the growth of U87MG cell line[129]
Table 2. The effects of seaweed-derived compounds on neurogenerative diseases.
Table 2. The effects of seaweed-derived compounds on neurogenerative diseases.
Algal SourceCompound of Interest and FractionStudy TypeBiological EffectsRef.
Brown seaweed (Phaeophyta)
Bifurcaria bifurcataPhenolic fractionIn vitroPrevented mitochondrial potential changes, decreased H2O2 production, and inhibited Caspase-3 activity[116]
Undaria pinnatifidaEthanol extractIn vitroDecreased ER stress via upregulating Akt/mTOR signaling pathway[133]
Undaria pinnatifidaFucoxanthin and fucosterolIn vitro and in silicoModerate inhibition for fucoxanthin and inactive for fucosterol on two isoenzymes
Fucoxanthin as potential dopamine D3/D4 agonist
[106]
Undaria pinnatifidaFucosterol and fucoxanthinIn vitroInhibitory effects on BACE1[134]
Ishige foliaceaEthanol extractIn vitro and in vivoDecreased Aβ accumulation
Inhibited AChE, BACE1, and memory deficit
[135]
Sargassum horneriFucoxanthinIn vitroDecreased H2O2-induced neurotoxicity by upregulating the PI3K/Akt cascade and inhibiting the ERK pathway[107]
Ecklonia cavaPolyphenolIn vitro and in vivoReduced Ca2+—mediated neurotoxicity on ischemic rats[136]
Ecklonia cavaDieckol and phlorofucofuroeckolIn vivoIncreased acetylcholine and reduced anticholinesterase activities[98]
Ecklonia cavaButanolIn vitro and in vivoReduced Aβ secretion and cell death[137]
Ecklonia cavaPhlorotanninIn vitroRegulated the expression and activity of alpha- and gamma-secretase
Reduced Aβ production
[138]
Eisenia bicyclisEckol and dieckolIn silicoInhibited hMAOs by its higher binding affinity[97]
Ishige okamuraeFresh seaweed and ethanolic extractIn vitro and vivoReduced Aβ25–35-induced phosphorylation by downregulating ERK, p38 MAPK, and JNK pathway[139]
Ecklonia maximaPhenolic extractIn vitroInhibited acetylcholinesterase and butyrylcholinesterase activities[140]
Sargassum fusiformePolysaccharideIn vivoReduced memory deficiencies and improved cognitive abilities of rats[28]
Ecklonia stoloniferaFucosterol and fucoxanthinIn vitroInhibitory effects on BACE1[134]
Red seaweed (Rhodophyta)
Gracilaria corneaSulfated agaranIn vivoReduced oxidative stress
Recovered behavioral activity and weight gain of rats to normal
[141]
Gracilaria beckeriPhenolic extractIn vitroInhibited acetylcholinesterase and butyrylcholinesterase activities[140]
Gelidium pristoidesPhenolic extractIn vitroInhibited acetylcholinesterase and butyrylcholinesterase activities[140]
Gelidium amansii (formerly Gelidium elegans)Ethanol extractsIn vitroPromoted the initial neuronal differentiation[142]
Green Seaweed (Chlorophyta)
Caulerpa racemosaRacemocin A and racemocin BIn vitroIncreased cell viability of SH-SY5Y cells[114]
Ulva rigidaPhenolic extractIn vitroInhibited acetylcholinesterase and butyrylcholinesterase activities[140]
Table 3. The effects of seaweed-derived compounds on liver disease.
Table 3. The effects of seaweed-derived compounds on liver disease.
Algal SourceConstituentsStudy TypeBiological EffectsRef.
Brown seaweed (Phaeophyta)
Ecklonia cava7-phloro-eckolIn vitroReduced alcohol-induced oxidative stress injury[100]
Sargassum fluitansEthanol extractIn vivoReduced the level of APAP and CCl4 induced liver damage[32]
Sargassum ilicifoliumEthanol extractIn vivoShowed nephroprotective and hepatoprotective effects[151]
Turbinaria decurrensFucoidanIn vivoImproved antioxidant status and reduced liver injury[73]
Myagropsis myagroidesAqueous extractsIn vivoReduced the CCl4 induced acute elevation in the levels of GPT and GOT in rats[152]
Sargassum henslowianumAqueous extractsIn vivoReduced the CCl4 induced acute elevation in the levels of GPT and GOT in rats[152]
Sargassum siliquastrumAqueous extractsIn vivoReduced the CCl4 induced acute elevation in the levels of GPT and GOT in rats[152]
Fucus vesiculosusFucoidanIn vivoAmeliorated thioacetamide (TAA)-induced liver injury[153]
Fucus vesiculosusPhytocomplexIn vitro and in vivoReduced both the postprandial glycemic peak and the blood glucose curve (AUC)
Inhibited in carbohydrate digestion
[154]
Ascophyllum nodosumPhytocomplexIn vitro and in vivoReduced both the postprandial glycemic peak and the blood glucose curve (AUC)
Inhibited in carbohydrate digestion
[154]
Cladosiphon okamuranusFucoidanIn vivoShowed isoproterenol-induced myocardial infarction[155]
Sargassum thunbergiiIndole-4-carboxaldehydeIn vitroReduced pro-inflammatory mediator, i.e., methylglyoxal (MGO) and advanced glycation end-product (AGE) formation[156]
Red Seaweed (Rhodophyta)
Halymenia porphyroidesEthanol extractIn vivoShowed nephroprotective and hepatoprotective effects[151]
Gracilaria lemaneiformisOligosaccharidesIn vivoExerted antioxidant defense system[157]
Green Seaweed (Chlorophyta)
Ulva lactucaSulfated polysaccharideIn vivoReduced D-Gal-induced DNA damage and necrosis level in rats[8]
Table 4. The effects of seaweed-derived compounds on diabetes.
Table 4. The effects of seaweed-derived compounds on diabetes.
Algal SourceConstituentStudy TypeBiological EffectsRef.
Brown Seaweed (Phaeophyta)
Ecklonia cavaAG-dieckolIn vivoReduced total glucose and lipid[101]
Ecklonia cavaDieckolIn vivoReduced blood glucose level, serum insulin level and body weight[102]
Ecklonia cavaPolyphenolIn vivoInhibited the activation of high glucose-induced hepatic stellate cells (HSCs)[164]
Sargassum hemiphyllumFucoidan and fucoxanthinIn vivoReduced urinary sugar
Reduced total glucose and lipid
[165]
Sargassum hemiphyllumOligo-fucoidanIn vitro and in vivoInhibited pro-inflammatory mediators
Inhibitory effects on diabetes-evoked renal fibrosis
[166]
Padina arborescensMethanolic extractIn vivoAmeliorated hyperglycemia and dyslipidemia[167]
Ascophyllum nodosumCarbohydrate- and polyphenolic-enriched extractsIn vitroInhibited α-glucosidase
Inhibition of sucrase (IC50 = 0.83 mg/mL)
[168]
Ascophyllum nodosumFucoidanIn vitroInhibited α-glucosidase (IC50 = 0.013–0.047 mg/mL)
Inhibited α -amylase (IC50 = 0.12–4.64 mg/mL)
[169]
Fucus vesiculosusCarbohydrate- and polyphenolic-enriched extractsIn vitroInhibited α-glucosidase[168]
Fucus vesiculosusFucoidanIn vitroInhibited α-glucosidase (IC50 = 0.049 mg/mL)[169]
Undaria pinnatifidaCarbohydrate- and polyphenolic-enriched extractsIn vitroInhibited α-glucosidase[168]
Ishige foliaceaOctaphlorethol AIn vitroUpregulated transporter 4 (Glut4) translocation[170]
Ishige foliaceaOctaphlorethol AIn vivoDownregulated hepatic gluconeogenesis[171]
Sargassum polycystumEthanolic and aqueous extractsIn vivoAmeliorated kidney, liver, and pancreas damage[172]
ScagassumPolysaccharide fractionIn vivoRegulated glucose, triglyceride (TG), and total cholesterol
Ameliorated liver and kidney damage
[173]
Sargassum fusiformePolysaccharide fractionIn vivoRegulated glucose, triglyceride (TG), and total cholesterol[173]
Macrocystis
pyrifera
Polysaccharide fractionIn vivoRegulated glucose, triglyceride (TG), and total cholesterol
Ameliorated liver and kidney damage
[173]
Laminaria japonicaFucoxanthinIn vitro and in vivoReduced NO production and ROS level
Increased insulin resistance
Ameliorated improved spermatogenesis and male reproductive function
[104]
Ecklonia stoloniferaPhlorotanninIn vitroInhibitory effects on PTP1B and α-glucosidase[174]
Ecklonia stoloniferaFucosterolIn vitroModerate inhibitory effects on RLAR, HRAR, and PTP1B[10]
Eisenia bicyclisPhlorotanninIn vitroInhibitory effects on PTP1B and α-glucosidase[174]
Eisenia bicyclisFucosterolIn vitroModerate inhibitory effects on RLAR, HRAR, and PTP1B[10]
Red Seaweed (Rhodophyta)
Bryothamnion seaforthiiLectinIn vivoExerted hypoglycemic and hypolipidemic effects
Reduce insulin resistance and improved pancreatic β-cell function
[111]
Green Seaweed (Chlorophyta)
Enteromorpha proliferaFlavonoidsIn vitroReduced inflammation in liver and kidney
Regulated insulin signaling pathway
Enriched the abundance of gut microbiota
[14]
Enteromorpha proliferaPolyphenolsIn vivoRegulated gene expression
Enriched the abundance of gut microbiota
[9]
Ulva rigidaEthanolic extractIn vitroAmeliorated carbohydrate metabolism, hyperlipidemia, and oxidative stress[175]
Table 5. The effects of seaweed-derived compounds on obesity.
Table 5. The effects of seaweed-derived compounds on obesity.
Algal SourceConstituentsStudy TypeBiological EffectsRef.
Brown seaweed (Phaeophyta)
Sargassum miyabeiCrude extractIn vitroReduced lipid accumulation and differentiation
Inhibited adipogenic and lipogenic gene expression
[179]
Saccorhiza polyschidesPolysaccharideIn vivoRegulated intestinal and systemic glucose metabolism
Inhibition of α-amylase activity
[180]
Fucus vesisulosusFucoidanIn vitroInhibited lipid accumulation[181]
Petalonia binghamiaeWater-soluble extractIn vivoInhibited adipogenic and lipogenic genes expression
Reduced body weight
[182]
Ecklonia stoloniferaFucosterolIn vitroInhibited adipogenic and lipogenic gene expression[183]
Ecklonia stoloniferaFucosterolIn vitroInhibited adipogenesis via FoxO1 pathway modulation[184]
Ecklonia cavaPhlorotanninIn vitroInhibited adipogenic expression[185]
Ecklonia cavaPolyphenol ExtractIn vivoInhibited lipogenesis[186]
Undaria pinnatifidaFucoxanthinIn vivoInhibited lipogenesis[105]
Eisenia bicyclis6,6′-BieckolIn vitroInhibited adipogenesis[96]
Red seaweed (Rhodophyta)
Gelidium amansii (formerly G. elegans)Polysaccharide-rich extractIn vivoDecreased triglyceride and total cholesterol levels
Decreased body and adipose tissue weights
[43]
Gelidium amansii (formerly G. elegans)Ethanolic extractIn vivoInhibited adipogenesis[187]
Plocamium telfairiaeEthanolic extractIn vitro and in vivoInhibited adipogenic and lipogenic gene expression[188]
Sarconema filiformeCarrageenanIn vivoModulated gut microbiota
Reduced body weight and lipid accumulation
[84]
Green Seaweed (Chlorophyta)
Codium cylindricumSiphonaxanthinIn vitro and in vivoAccumulated in stomach, small intestine, liver, and adipose tissues[109]
Codium fragileCrude extractIn vivoModulated gut microbiota
Reduced body weight and accumulation of cholesterol and glucose
[30]
Codium cylindricumSiphonaxanthinIn vitro and in vivoReduced oxidative and somatic stress on obese mice[189]
Table 6. The effects of seaweed-derived compounds on cardiovascular disease.
Table 6. The effects of seaweed-derived compounds on cardiovascular disease.
Algal SourceCompound of Interest and FractionStudy TypeBiological EffectsRef.
Brown Seaweed (Phaeophyceae)
Saccharina sculperaFucoidanIn vitroInhibited cholesterol synthesis and reverse transport
Modulated fatty acid synthesis
Accelerated mitochondrial β-oxidation
[195]
Iyengaria stellataEthanol extractIn vivoGreat inhibition of the lipid profile in diet-induced hyperlipidemic rats[196]
Colpomenia sinuosaEthanol extractIn vivoReduced lipid profile[196]
Spatoglossum asperumEthanol extractIn vivoCombination with triton showed great inhibition of the lipid profile in triton-induced hyperlipidemic rats[196]
Ascophyllum nodosumFucoidan A2In vivoReduced lipid profile, including plasma total cholesterol, triglyceride, and fat pad index[29]
Ascophyllum nodosumFucoidan A3In vivoReduced lipid profile, including plasma total cholesterol, triglyceride, and fat pad index[197]
Cladosiphon okamuranusFucoidanIn vivoReduced lipid profile via reverse transport[155]
Ecklonia cavaPhlorotanninIn vivoReduced lipid profile via reverse transport[198]
Fucus spiralisPeptide and phlorotanninsIn vitroHighly inhibited ACE-I yield[199]
Sargassum siliquastrumSargachromenol DIn vitro and in vivoExerted antagonist of L-type Ca2+ channel and endothelin A/B2 receptors[200]
Ecklonia cavaPolyphenol extract and dieckolIn vitro and in vivoReduced lipid profile in the serum of HFD-fed mice
Reduced lipid accumulation in 3T3-L1 cells
[201]
Red Seaweed (Rhodophyta)
Solieria robustaEthanol extractIn vivoGreat inhibition of the lipid profile in diet-induced hyperlipidemic rats[196]
Green Seaweed (Chlorophyta)
Caulerpa racemosaEthanol extractIn vivoReduced lipid profile[196]
Table 7. The effect of seaweed-derived compounds on arthritis.
Table 7. The effect of seaweed-derived compounds on arthritis.
Algal SourceConstituentsStudy TypeBiological EffectsRef.
Brown Seaweed (Phaeophyceae)
Ecklonia cavaPhlorotanninsIn vitroDownregulated pro-inflammatory cytokines[13]
Undaria pinnatifidaFucoidanIn vitro and in vivoDownregulated pro-inflammatory cytokines[75]
Lobophora variegataFucanIn vitro and in vivoReduced articular inflammation[210]
Sargassum wightiiAlginic acidIn vivoDownregulated pro-inflammatory cytokines [211]
Sargassum wightiiAlginic acidIn vivoDownregulated pro-inflammatory cytokines[34]
Fucus vesiculosisFucoidanClinical studyReduced the symptoms of osteoarthritis in a dose-dependent manner[12]
Macrocystis pyrifera,FucoidanClinical studyReduced the symptoms of osteoarthritis in a dose-dependent manner[12]
Laminaria
japonica
FucoidanClinical studyReduced the symptoms of osteoarthritis in a dose-dependent manner[12]
Red Seaweed (Rhodophyta)
Eucheuma cottoniiPolysaccharide-richIn vitroAttenuated cartilage degradation[190]
Laurencia glanduliferaNeorogioltriolIn vitro and in vivoDownregulated pro-inflammatory cytokines[212]
Green Seaweed (Chlorophyta)
Codium fragileOleamideIn vitro and in vivoDownregulated pro-inflammatory cytokines[213]
Caulerpa cupressoidesLectinIn vivoReduced temporomandibular joint inflammation[112]
Table 8. The effect of seaweed-derived compounds on osteoporosis.
Table 8. The effect of seaweed-derived compounds on osteoporosis.
Algal SourceConstituentsStudy TypeBiological EffectsRef.
Brown Seaweed (Phaeophyceae)
Dictyota mertensiiFucoidanIn vitroProtected bone tissue against oxidative stress[217]
Laminaria digitataFucoxanthinIn vitroLow pro-osteogenic effects[11]
Ascophyllum nodosumFucoxanthinIn vitroLow pro-osteogenic effects[11]
Padina pavonicaAcetonic extractIn vitroPromoted osteoblast differentiation
Regulated osteoblast-specific markers
[218]
Sargassum hemiphyllumFucoidanIn vitroProtected osteoclast differentiation and inflammatory bone loss[219]
Ishige okamuraeDiphloretho-hydroxycarmalolIn vitroPromoted osteoblastic differentiation from oxidative stress[220]
Red Seaweed (Rhodophyta)
Plocamium lyngbyanumMethanolic extractIn vitro and in vivoPromoted osteogenic differentiation and mineralization
Increased opercular bone area
[221]
Ceramium secundatumMethanolic extractIn vitro and in vivoPromoted osteogenic differentiation and mineralization
Increased opercular bone area
[221]
Dichotomaria obtusataMethanolic extractIn vitroUpregulated osteogenic activity[215]
Gracilaria verrucosaCrude extractIn vitro and in vivoDownregulated RANKL-induced osteoclast differentiation
Inhibited bone loss
[33]
Ceramium pallidumDichloromethane and methanol extractIn vitro and in vivoPromoted osteogenic differentiation and mineralization
Increased opercular bone area
[216]
Green Seaweed (Chlorophyta)
Cladophora rupestrisPhenolic extractIn vitro and in vivoPromoted osteoblast-like cell mineralization
Increased opercular bone area
[222]
Codium fragilePhenolic extractIn vitro and in vivoPromoted osteoblast-like cell mineralization
Increased opercular bone area
[222]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Meinita, M.D.N.; Harwanto, D.; Choi, J.-S. Seaweed Exhibits Therapeutic Properties against Chronic Diseases: An Overview. Appl. Sci. 2022, 12, 2638. https://doi.org/10.3390/app12052638

AMA Style

Meinita MDN, Harwanto D, Choi J-S. Seaweed Exhibits Therapeutic Properties against Chronic Diseases: An Overview. Applied Sciences. 2022; 12(5):2638. https://doi.org/10.3390/app12052638

Chicago/Turabian Style

Meinita, Maria Dyah Nur, Dicky Harwanto, and Jae-Suk Choi. 2022. "Seaweed Exhibits Therapeutic Properties against Chronic Diseases: An Overview" Applied Sciences 12, no. 5: 2638. https://doi.org/10.3390/app12052638

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