Next Article in Journal
Marine Fungal Cerebroside Flavuside B Protects HaCaT Keratinocytes against Staphylococcus aureus Induced Damage
Next Article in Special Issue
Nutritional Value and Biofunctionalities of Two Edible Green Seaweeds (Ulva lactuca and Caulerpa racemosa) from Indonesia by Subcritical Water Hydrolysis
Previous Article in Journal
Chitosan-Based Scaffold for Mineralized Tissues Regeneration
Previous Article in Special Issue
Seaweed Protein Hydrolysates and Bioactive Peptides: Extraction, Purification, and Applications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Applying Seaweed Compounds in Cosmetics, Cosmeceuticals and Nutricosmetics

1
Centro de Investigaciones Biomédicas (CINBIO), Departamento de Enxeñería Química, Universidade de Vigo (Campus Ourense), Edificio Politécnico, As Lagoas, 32004 Ourense, Spain
2
Departamento de Química Analítica, Universidade de Vigo (Campus Ourense), Edificio Politécnico, As Lagoas, 32004 Ourense, Spain
*
Author to whom correspondence should be addressed.
Submission received: 31 August 2021 / Revised: 24 September 2021 / Accepted: 27 September 2021 / Published: 29 September 2021
(This article belongs to the Special Issue Nutra-Cosmeceuticals from Algae for Health and Wellness)

Abstract

:
The interest in seaweeds for cosmetic, cosmeceutics, and nutricosmetics is increasing based on the demand for natural ingredients. Seaweeds offer advantages in relation to their renewable character, wide distribution, and the richness and versatility of their valuable bioactive compounds, which can be used as ingredients, as additives, and as active agents in the formulation of skin care products. Bioactive compounds, such as polyphenols, polysaccharides, proteins, peptides, amino acids, lipids, vitamins, and minerals, are responsible for the biological properties associated with seaweeds. Seaweed fractions can also offer technical features, such as thickening, gelling, emulsifying, texturizing, or moistening to develop cohesive matrices. Furthermore, the possibility of valorizing industrial waste streams and algal blooms makes them an attractive, low cost, raw and renewable material. This review presents an updated summary of the activities of different seaweed compounds and fractions based on scientific and patent literature.

Graphical Abstract

1. Introduction

Consumer preferences towards green and eco-friendly products have increased in the last few years [1,2]. This trend is also found in cosmetics, which represent a competitive and rapidly changing global market demanding natural, safe, and efficient ingredients for the development of novel skin care products [3,4,5]. Other relatively new products are cosmeceuticals and nutricosmetics. The term cosmeceutical is used to define active and safe products developed and tested by the cosmetics industry to provide benefits to skin appearance and are effective for preventing and treating different dermatologic conditions [6] by offering a variety of functions [6,7,8]. A number of active ingredients, including vitamins, phytochemicals, enzymes, antioxidants, and essential oils, are also considered [9], and can be used for the formulation of creams, lotions, ointments, or masks. The use of cosmeceuticals has drastically risen in the last few years [10], in a market that also incorporates other less-traditional population segments, such as men and children [11,12,13]. Both cosmetics and cosmeceuticals have to be safe, efficient, and have good sensorial quality features [6,14]; nutricosmetics also require optimal characteristics. For the optimal development of these products, cooperation in areas such as biotechnology, chemistry, food technology, pharmaceutical technology, and toxicology is needed [15].
Marine resources represent a widely available and promising source of unique and active compounds with the potential to produce cosmetics, cosmeceuticals, and nutricosmetics. Among them, seaweeds represent a sustainable and renewable resource, gaining increasing attention for these applications [16,17]. Furthermore, valorization of waste seaweeds, such as beach-casts, which are disposed of in landfills without commercial value, could represent an attractive low-cost source for cosmeceutical industries [18]. Similarly, the valorization of invasive species could contribute to the creation of natural and eco-friendly ingredients for the cosmetic industry [19] while also contributing to the restoration of affected environments. Regardless the origin and type of seaweed, the development of environmentally-friendly sustainable extraction methods, allowing a low extraction time, minimum usage of solvents, higher extraction yields, and quality, are increasingly demanded [5,19,20,21,22].
The use of seaweed-derived ingredients in cosmetic products has increased in recent years as a result of the many scientific studies that have proved the potential skincare properties of seaweed bioactives [23,24]. Among those biologically active molecules, carotenoids, fatty acids, polysaccharides, phlorotannins, vitamins, sterols, tocopherol, phycobilins, and phycocyanins have attracted attention [9,25,26,27,28,29]. Such rich compositions have converted seaweed into potential ingredients in classical cosmetics, such as solid soaps, to replace sodium lauryl sulfate/sodium laureth sulfate [22], but many algal extracts have also been used in nutritional supplements, cosmetics, and alternative medicines recommended for skin-related diseases [30]. In this latter case, they are added as the active ingredient, because they can provide a variety of activities, including photoprotective, moisturizing, antioxidant, anti-melanogenic, anti-allergic, anti-inflammatory, anti-acne, anti-wrinkling, antimicrobial, antiaging, whitening, etc. [16,31,32,33]. Furthermore, they exhibit low cytotoxicity and low allergen contents [34].
Excellent comprehensive reviews on the subject have recently been published, including on the chemical diversity and unique properties of algal bioactive molecules or extracts for cosmetic uses [5,16,28,33,35,36,37] and the progress made in the application of bioactives from marine organisms as cosmeceuticals [27,38,39]. Most of these have emphasized the importance and scientific evidence of algae-derived compounds and their benefits, as well as current application in the cosmetic industry and their challenges and limitations in the development of cosmeceuticals [3,23,24,29,34,40]. Others have reported on particular components, such as carbohydrates [41,42], or the specific beneficial actions on hyperpigmentation, photoaging, and acne [2,23,24], as well as on perspectives for the development of greener extraction methods [35], particularly those using safe solvents [3].
The present review tries to update the advances in this field, presenting an initial section summarizing the activities of algal components of particular relevance for cosmetic and cosmeceutical formulations and then by trying to offer the multiple and faceted benefits and functions that these seaweed components can provide to products where they can be incorporated as ingredients and additives, conferring other textural, functional, and sensorial properties. The potential applications are presented based on information in the scientific literature, but also using patents claiming the use of algae and algal components.

2. Seaweed Components and Bioactivity

2.1. Polysaccharides

Seaweeds contain an important carbohydrate fraction forming part of their cell walls and these polysaccharides are specific to each type of algae: in brown alginate, laminaran and fucoidan; in green ulvan and in red agar, carrageenan is the most important. Polysaccharides are receiving increasing attention for their biofunctional and physicochemical characteristics [43]. Sulfated polysaccharides are highly interesting due to their health benefits and biological activities [32,44,45,46,47,48,49,50]. A key aspect of these polysaccharides is the close relationship between the activity and their composition and structure, particularly, their molecular weight. Therefore, depolymerization is usually proposed to enhance the activity [51], but other structural modifications can also be performed. Simple hydrophobization reactions, such as esterification, acylation, alkylation, amidation, or cross-linking reactions on native hydroxyl-, amine, or carboxylic acid functions can also enhance bioactivity [52]. Examples of these activities are summarized in Table 1.
Alginates composed of chains of d-mannuronic acid and l-guluronic acid are found in brown seaweeds. These compounds show other properties in relation to cosmetics and well-being products, particularly anti-allergic properties [32,71], an action that is also observed in formulations of hydrogels with alginate [72], and can also prevent obesity [73,74]. Laminarin does not form viscous solutions and has prebiotic [75,76], antioxidant [77,78,79], and anti-photoaging and regenerative [69] properties. Based on the wound healing [80] properties of laminarin sulfate, novel hydrogel systems have been developed [81,82,83]. In addition, promising outcomes have been exhibited in several biomedical applications, such as tissue engineering, cancer therapies, antioxidant, and anti-inflammatory properties [84]. Degradation by irradiation can enhance the radical scavenging capacity and inhibitory activity against melanin synthesis in melanoma cells [2,59].
Fucoidans are heteropolysaccharides with fucose and other monosaccharides, such as xylose, galactose, mannose, and glucuronic acid, as well as other components, mainly sulfate, uronic acids, and acetyl groups. Fucoidans offer promising potential as cosmetic ingredient [34,85,86] since they are non-toxic, biodegradable, and biocompatible [87,88], and they present a wide variety of biological properties [23,24,49,61,89,90,91]; they also reduce antioxidant and antiradical properties [23,24,34,67,92], depending on the molecular weight and sulfate content [93,94]. Fucoidans have shown confirmed benefits for preventing and treating skin photoaging and have in vitro inhibition of UVB-induced collagenase and gelatinase activities, ex vivo inhibition on elastase activity in human skin [2,64,66,95], inhibition of wrinkle-related enzymes and enhanced collagen synthesis in human dermal fibroblasts [67], and anti-inflammatory action in relation to extracellular matrix degradation by matrix metalloproteinases [27,32,42,65,96].
Sulfated polysaccharides from green algae (rhamnans, arabinogalactans, galactans and mannans) present variable compositions and structures and some properties are highly influenced by the molecular weight in terms of antiradical and chelating properties [97,98,99,100]. Ulvans are highly complex and variable sulfated polysaccharides from ulvales, composed mainly of rhamnose, xylose, glucose, glucuronic acid, iduronic acid, and sulfate [34,89,101,102]. Ulvans exhibit a variety of activities, including gelling [101,103], anti-aging [51], anti-hyperlipidemic and antiherpetic properties [71,104].
Agar is mainly composed of β-d-galactopyranose and 3,6-anhydro-α-l-galactopyranose units with variable amounts of sulfate, pyruvate, and uronate substituents. Agar has pharmaceutical and industrial cosmetic applications, including its use as a thickener and as an ingredient for tablets or capsules to carry and release drugs [105,106]. Carrageenans are generally recognized as safe (GRAS) and are approved for food applications, and are high-molecular-weight sulfated linear polysaccharides with a backbone of alternating 3- α-d-galactopyranose and 4-β-d-galactopyranose with anhydrogalactose residues [54,55,107,108]. Porphyran is a complex sulfated galactan found in Porphyra sp. with interesting therapeutic properties. These polysaccharides have uses as gelling agent, nutritional supplement, with antioxidants [109,110,111,112], and are antiallergic [32,113], show tyrosinase inhibitory activity [62], protection against ultraviolet B radiation [59], anti-inflammatory and antitumoral activity, and can promote the growth of beneficial bacteria in intestinal microbiota [76,113] without toxicity in mice models [114,115]. Agaro-oligosaccharides (AOS) and carrageenan-oligosaccharides (COS) present enhanced biological properties compared to native ones, in relation to prebiotic, antitumoral, and antioxidant actions, related to their chemical structure, molecular weight, degree of polymerization, and the flexibility of the glycosidic linkages [116].

2.2. Proteins, Peptides and Aminoacids

Some seaweeds are a rich source of proteins, their cultivation offers a higher protein yield per unit area (2.5–7.5 tons/Ha/year) compared to terrestrial crops, but their successful extraction is largely influenced by the presence of polysaccharides, such as alginates in brown seaweed or carrageenans in red seaweed [117]. Seasonal variations and habitat affect the proteins, peptides, and amino acids contents in seaweed; generally, red algae (Rhodophyceae) have higher contents (up to 47%) than green (Chlorophyceae) (between 9–26%), whereas brown (Phaeophyceae) have a lower concentration (3–15%) [73,118,119,120]. The proteins in the three groups of macroalgae contain all essential amino acids, and non-essential amino acids are also present [25,121,122,123]. Protein and bioactive peptides from seaweed show many health benefits and have high antioxidant properties, mainly in molecules with low molecular weights, which are also considered safer than synthetic molecules and have reduced side effects [3,124,125,126,127].
Bioactive peptides usually contain 3–20 amino acid residues and both their amino acid composition and the sequence influences their activities, such as antioxidant and antimicrobial activities, among others of pharmacological interest [128,129,130,131]. Carnosine, glutathione, and taurine are peptides with antioxidant and chelating properties [132]. Due to the lack of a carboxyl group, taurine is not a “true” amino acid but has a number of health-promoting properties, being accumulated in the thalli of several red algae, such as Ahnfeltia plicata, Euthora cristata, and Ceramium virgatum [133]. The peptide, PPY1, is composed of five amino acids and is obtained by enzymatic hydrolysis from Pyropia yezoensis, and it shows anti-inflammatory effects through the suppression of inflammatory cytokines [134]. The peptides, PYP1-5 and Porphyra 334, extracted from Porphyra yezoensis f. coreana Ueda showed an increase in elastin and collagen production and a decrease in the expression of matrix metalloproteinases (MMP) [135]. Ultrasound-assisted enzymatic hydrolysis has also been proposed for the successful extraction of iodinated amino acids from Palmaria palmata and Porphyra umbilicalis (red seaweeds) [136].
Mycosporine-like amino acids (MAAs) are secondary metabolites synthesized for protection against solar radiation [28,137,138]. They consist of cyclohexenone or cyclohexenimine chromophore with various amino acids, mainly glycine or iminoalcohol groups, as substituents and show antioxidant and photoprotective properties [3,137,139,140,141,142,143,144]. Among the most abundant compounds, mainly in Rhodophyceae shinorine, porphyra-334, palythine, asterina-330, mycosporine-glycine, palythinol, and palythene have been described [145,146], and their contents are dependent on the geographic, seasonal and bathymetric conditions, increasing during summer and decreasing with water depth [147]. A multifunctional cosmetic liposome formulation containing UV filters, vitamins (A, C, and E), Ginkgo biloba extract (rich on quercetin), and Phorphyra umbilicalis extract (rich in proteins, vitamins, minerals and mainly in MAA’s porphyra-334 and shinorine) was efficient against signs of aging [148] by increasing hydration and reducing wrinkles and skin roughness. Leandro et al. [149] incorporated an extract of Asparagopsis armata (ASPAR’AGE™) containing MAA molecules in lotions with anti-aging properties, a hydrolyzed extract Aosaine® (three-quarters of aosaine consists of amino acids that are very similar those responsible for skin elasticity) extracted from Ulva lactuca, which present anti-aging, anti-wrinkle and stimulation of collagen properties. An extract (rich in minerals, trace elements and amino acids) from Gelidium corneum improves skin softness and restores elasticity. Therefore, MAAs have different properties, such as serving as natural sunscreens, possess antioxidants, anti-inflammatory, and anti-aging, and are stimulators of skin renewal, activators of cells proliferation, etc., making them a promising and safe option for pharmaceutical and cosmetic industries [150] (Table 2).
Due to the toxic effect of several synthetic dyes and the high consumer demand for natural colors in food, pharmaceuticals, cosmetics, and textile industries there has been increasing interest in the use of phycobiliproteins in the food (C-phycocyanin) and cosmetic fields (C-phycocyanin and R-phycoerythrin). Phycobiliproteins are a class of water-soluble compounds composed of proteins that are covalently bound to linear tetrapyrroles, known as phycobilins, with fluorescent properties and high molecular weights and can be used for reddish colorings [28,118,160,161,162,163]. B-phycoerythrin resists changes in pH, possesses antioxidant properties [164], and can be used as a pink or purple dye in cosmetics [165]. Phycobilins can be red (phycoerythrins) or blue (phycocyanins and allophycocyanins) and phycocyanin is usually the major pigment microalgae (Spirulina spp.), whereas the characteristic red color of Rhodophyta phyla is due to both the phycoerythrin and phycocyanin pigments. Phycobiliproteins (concretely, R-phycoerythrin, phycocyanin, and allophycocyanin) extracted from Gracilaria gracilis presented high antioxidant and radical scavenging activities, primarily when harvested in winter [157], and the extraction can yield up to 46.5% of R-phycoerythtin using an aqueous solution of ionic liquids (cholinium chloride) to remove it from fresh algal biomass [166]. Saluri et al. [167] studied Furcellaria lumbricalis and Coccotylus truncatus and found an exponential correlation between R-phycoerythrin and allophycocyanin concentrations and collection depth. The contents of phycoerythrin and phycocyanin were slightly higher and lower, respectively, for dried commercial Porphyra spp. extracts in comparison to Spirulina spp. [168].

2.3. Phenolics and Terpenoids

Phenolic compounds are secondary plant metabolites with a basic structure with one or more aromatic rings, presenting one or more attached -OH groups. They are synthesized as part of the defense mechanisms in plants. Phlorotannins are secondary metabolites of phloroglucinol (1,3,5-trihydroxybenzene), are structurally less complex than terrestrial tannins, and are found in polymerized structures with ether, phenyl or 1,4-dibenzodioxin linkages [169,170].
Phlorotanins are increasingly considered for cosmeceutical applications, based on their antioxidative [171,172,173,174,175], anti-allergic [27,176,177,178,179], anti-inflammatory [27,180,181], tyrosinase inhibitory [182,183,184,185,186], and antidiabetic [175] activities. Skin protection against UV irradiation was confirmed in mouse skin models [187,188]. Phlorotannins also attenuated the expression of MMP-1 (an interstitial collagenase mainly responsible for the degradation of dermal collagen in human skin aging process) [27,28,189]. Dioxinodehydroeckol from Ecklonia cava proved to be an effective repair agent for skin damage against UVB [190]. On the other hand, fucofuroeckol-A derived from the brown seaweed Ecklonia stolonifera Okamura, exhibited protective activity against UVB radiation [191]; other studies also exhibited similar results for eckol and dieckol [192,193]. A correlation between the antioxidant activity and the hyaluronidase inhibitory capacity with higher molecular weight phlorotannins was observed [172], a behavior that was also observed in other works [194,195,196]. Some properties of brown algal phlorotannins are summarized in Table 3.
Meroterpenoids exhibited antioxidant properties and can prevent skin photoaging without the risk of cytotoxicity [205]. Other meroterpenoid derivatives have also shown interesting properties in relation to protection from cell damage caused by UVA irradiation [206] and photodamage attenuation on irradiated cells [207]. In addition, the hypopigmenting effect of meroterpenoids has been associated with brown algae [208].

2.4. Lipids

Seaweed present a low lipidic content (usually under 5%), but they are highly unsaturated and the ω3:ω-6 fatty acids ratio is highly favorable [73,209,210]. Among the most abundant fatty acids are γ-linolenic acid, arachidonic acid, eicosapentanoic acid, and docosahexanoic acid, but other lipid types, such as sterols and phospholipids, are also found [211,212]. The main sterols found are fucosterol, isofucosterol, and clionasterol [213,214]. Several biological properties have been associated with lipids [211,215]. Polyunsaturated fatty acids (PUFA) can benefit skin barrier protection and other biological functions can be enhanced; nutricosmetics could contribute an anti-obesity effect [211,216,217] and the regulation of inflammatory responses [25,218]. Being structural components of cell membranes, sterols regulate membrane fluidity and permeability and other properties, such as antioxidant, antiproliferative, and anti-photodamage, and anti-inflammatory effects have been reported for fucosterol [28,188,219,220,221]. An effect against the malarial parasite Plasmodium falciparum has been exhibited [222]. The viability of human keratinocytes irradiated with UVB was not affected when cells were incubated with fucosterol, and a marked decrease in UV-irradiated MMPs and increased type-I procollagen production were observed [28,206]; other authors obtained results consistent with these observations [49,61]. Phospholipids, mainly made up of fatty acids containing a phosphate group and a simple organic molecule, have been reported to help with carotenoid absorption [223]; in other work, authors showed a reduction of body weight and fat mass in mice drinking water with lipid capsules prepared using phospholipids [224]. In addition, seaweed essential oil has been evaluated, and Rexliene and Sridhar reported the antimicrobial and anti-dandruff properties of red seaweed Portieria hornemannii essential oil [225]. Subsequently, an antibacterial film was created with a carrageenan biopolymer blended with extracted seaweed essential oil, showing adequate bio-physical, mechanical, and anti-microbial properties. Table 4 summarizes the biological activities associated with lipids.

2.5. Vitamins

Vitamins obtained from diet and through topical application are essential for many functions of human skin. Supplementation is considered for protection against dehydration and premature aging of the skin, cosmetic prevention of damage by sun exposure, regulation of the secretory activity of the sebaceous glands, and the preservation of the anatomical integrity of adnexial structures [232]. Vitamins are popular ingredients in many cosmeceuticals and skin care products. Vitamins A, C, E, K and vitamin complex B are the most important and clinically validated for skin photoaging prevention and treatment [233] and the common vitamins in algae are vitamins A, B, C, and E [3,16,25,234].
Vitamin A or the retinol form shows antioxidant and antiwrinkle capacity [37,235,236] and is topically used in cosmetics to reduce facial hyperpigmentation and fine wrinkles [237]. The concentration of vitamin complex B (B1, B2, B3 or niacine, B6, B9 or folic acid, B12) is generally higher in green and red seaweeds [3,238]. Vitamin B3 active forms added to skin care products include: niacinamide, nicotinic acid nicotinate esters. Niacinamide is an antioxidant, reduces hyperpigmentation (also due to blue light-induced), and improves aspects of the epidermis by reducing the trans-epidermal water loss [7,239]. Red algae and other species are good sources of vitamin B12 for vegetarians; this vitamin shows anti-aging properties and is essential for hair and nail growth and health [25,240,241,242].
Vitamin C is used in the cosmeceutical industry as it is an l-ascorbic acid of which the biologically active form is most known [236]. The red algae Ceramium rubrum and Porphyra leucosticta show high vitamin C or ascorbate content. This vitamin, topically applied, has antioxidant, detoxifying, antiviral, anti-inflammatory, antimicrobial, and anti-stress effects, and can be used for enhancing tissue cell growth, repairing blood vessels, teeth, and bones [7,243]. Many studies reported skin improvements in fine lines and reduction of pigmentation and inflammation if present in an appropriate concentration in a cosmetic formula [7,244]. Several works confer tyrosinase inhibition to vitamin C due to it interacting with copper ions, which reduces melanogenesis [236].
Vitamin E (α, ã, ä tocopherol), the most abundant fat-soluble vitamin of non-saponifiable lipids in many algae, can be extracted from different green, red, or/and brown seaweeds [245], and is effective against UV damage, photoaging, and skin cancer when is in a high concentration and in a non-esterified form [209,246]. Cosmetic formulations usually include vitamin C since it regenerates oxidized vitamin E [7]. Vitamin K, found in high concentrations in some seaweeds, has well-known blood clotting properties (wound, bruises, marks, and scar healing) [247,248,249].

2.6. Minerals

Seaweeds have a high mineral content, about 8–40% [250,251,252], and this wide range is dependent on seaweed phylum and species, seaweed oceanic residence time, geographical locations, wave exposure, and seasonal and annual environmental factors [234]. Seaweeds possess most of the mineral elements from the sea, and their content depends on the pH, temperature, and the concentration of the minerals in seawater. Seaweeds have been described as an ideal safe natural source of minerals. Inorganic ions play important roles in different functions of the skin, whereas others can be considered dangerous as a consequence of dermal exposure [253]. Table 5 shows the average mineral content in different type of seaweeds.
Seaweeds contain a variety of mineral elements, macro-elements, and trace elements, which are an excellent mineral source for cosmeceutical benefits for humans. Several minerals (e.g., Ca, Fe, Mg, P, Na, Zn, Cu, and Se) are recognized as necessary for health and well-being. All seaweeds contain high amounts of both macro minerals (Ca, Mg, Na, K, and P) and trace elements (Fe, Zn, I, Cu, Se, and Mn) [234,250,255]. High potassium contents were reported in red macroalgal Gracilaria species and the brown macroalgal species Laminaria digitata; nevertheless, seaweeds have low Na/K ratios (<1.5) [250]. Caulerpa veravelensis, Ulva lactuca, and Sargassum polycystum contain higher amounts of calcium. Seaweeds have been described as a good source of iodine, which is present in several chemical forms, and brown algae contains greater amounts, up to over 1% wet weight; its accumulation in seaweed tissues could be 30,000 times its concentration in sea water [254,255]. According to Peñalver and coauthors, seaweeds are a primary source of iodine, allowing to achieve daily iodine requirements [234], as it is an essential element in order to maintain thyroid function and health [234,251].
Polefka et al. summarized the scientific evidence available on the benefits and risks of topical application of mineral salts [256]. Seaweeds are, in general, a better source of minerals than sea salts, because the proportion of minerals are closer to those in human skin and body’s plasm and the penetration of nutrients is better [39]. Due to this high affinity to human skin, mineral sea salts used in cosmetics are rapidly absorbed, and refresh and replenish or hydrate the skin [39,257]. Several skin care and cosmetic products contain various nutrients and minerals from seaweed, seawater, or sea mud, especially for their therapeutic properties fir psoriasis and other skin-related disorders, and for their beneficial effects on skin (they help to retain water for a longer time, restores skin pH, help in blood circulation, acne repair, and prevention, and have anti-aging effects) [39,257,258]. Alves et al. reported that high concentrated forms of marine minerals and trace elements provide a protective effect against UV radiation [259].

2.7. Pigments

Regarding the concentration of pigments, seaweeds are classified into three groups: green (chlorophylls a, b and c), brown (carotenoids), and red (phycobilins as phycoerythrin). In addition, free radical scavenging, inhibiting melanogenesis, and photoprotection are some of the properties of these compounds that make them suitable for skin care [260]. Carotenoids are isoprenoid molecules produced by photosynthetic plants, fungi, and algae. These lipophilic compounds can be chemically classified as carotenes, such as α-carotene, β-carotene, and lycopene, and xanthophylls. Carotenoids are used as colors in foods and as natural color enhancers, in the food, pharmaceutical, and cosmetic industries. Some act as provitamin A, and recently they have attracted considerable interest due to their antioxidant and anti-inflammatory properties [261]. β-Carotene helps to counteract free radicals involved in various diseases and premature aging [28]. In this context, the extracts obtained from three brown seaweeds were assessed to study antioxidant capacities, where fucoxanthin, violaxanthin, â-carotene, cyanidin-3-O-glucoside, and other carotenoid and chlorophyll derivatives were also characterized. The results suggest that these compounds are responsible for antioxidant properties [262].
Fucoxanthin is the main carotenoid in brown algae, this xanthophyll can counteract oxidative stress caused by UV radiation [171] and suppresses tyrosinase activity in UVB-irradiated guinea pig and melanogenesis in UVB-irradiated mice [27]; anti-melanogenic, anti-aging and antioxidant activities were also associated with this compound [40]. Fucoxanthin enhanced the fat burning rate of fat cells in adipose tissue and might be used to treat obesity and reduce the risk of certain disorders, such as type 2 diabetes [26,28,261,263,264]. Some reported actions are summarized in Table 6.

3. Technological Functions

According to their functions, cosmetic ingredients are classified as (i) additives; (ii) stabilizing or excipient agents; and (iii) bioactive compounds, with real cosmetic functions [28,35]. Algal components can be used as technical ingredients to improve texture, color or stability of cosmetics, but also as bioactive agents, since they can confer a variety of biological desirable actions, which are applicable in the manufacturing of cosmeceuticals and skin care products [38,273]. Macroalgal components can be included in cosmetics as thickening or gelling agents, antioxidant, and colorants, or as active ingredients in hydrating, antiaging, skin-whitening, and pigmentation reduction products. These dual potentialities are summarized in Figure 1.
The incorporation of seaweed components was successful in different physical forms, and are commercially available in soaps, shampoos, sprays, hydrogels, or creams [274,275]. Their efficiency and stability can be enhanced with suitable carrier systems or vesicles, such as liposomes, nano/microparticles, emulsions, hydrogels, etc., designed to carry active agents in commercial products to achieve promoted effects [276,277,278].

3.1. Antimicrobial Agents

The antimicrobial properties of seaweed components are well known, in particular for food and pharmaceutical industries [279,280,281]. Extracts from macroalga have shown antibacterial and antifungal activities, the most active components being terpenoids and phlorotannins [27,281], which can avoid the side effects and allergic reactions associated with synthetic drugs [282]. Extracts from brown and green seaweeds proved effective against acne vulgaris [201,283], brown algal extracts against common skin pathogenic bacteria, such as methicillin-resistant Staphylococcus aureus, Staphylococcus aureus and Staphylococcus epidermidis [284,285,286], green algal extracts showed activity against oral bacteria [78,206], and red algae are active against Staphylococcus and Candida sp. [28,287,288,289,290]. In order to have products with a wider spectrum of protection, mixtures could be a valid approach. Widowati et al. formulated a moisturizer cream with adequate color and odor, using an antibacterial extract obtained from mixtures of Sargassum duplicatum, Sargassum echinocarpum, and S. polycystum extracts, which inhibited the development of bacteria for a longer period of time [291]. All seaweeds contained steroids, quinones, flavonoids, and alkaloids, and saponins were only found in S. duplicatum.

3.2. Antioxidants

Since many cosmetic and cosmeceutical formulations contain a lipidic component, they are highly susceptible to lipid peroxidation. The addition of antioxidants is needed to protect from oxidative deterioration, which also maintains the sensorial properties of the cosmetic products, in the context of appearance and odor. The contradictory data on the safety of synthetic chemical antioxidants have incentivized the search and use of natural compounds with antioxidant properties. Seaweeds represent an abundant and widespread source of compounds with confirmed antiradical and reducing properties [173,284,285,286,292]. Furthermore, they showed the potential to protect and/or retard oxidation of cosmetic products [20] and have a wide range of biological properties. The most efficient algal compounds are phlorotannin-derived fractions, but peptides and polysaccharide fractions also display reducing properties and antiradical capacity [97,293]; the phenolic compounds found in red seaweed can also scavenge free radicals and also show other properties, such as the inhibition of tyrosinase [294].

3.3. Sensorial Properties

The incorporation of different seaweed ingredients has to be evaluated in relation to organoleptic, spreadability, and hedonic tests [295,296]. Seaweed can provide different compounds with coloring compounds as an alternative to synthetic, mineral, and plant dyes, and show lower allergenic properties. Among the major compounds with this property are phycobilins and carotenoids, which cover a wide range of blue, yellow, orange, and red colors [29], as well as other biologically interesting properties [264].
Aroma is a key feature in cosmetics and cosmeceuticals, and the potential of seaweeds to produce terpenoids, carotenoids, fatty acid derivatives, and sulfur compounds is well known [29,297].

3.4. Texturizing

Thickening, gelling, and texturizing agents are used to control viscoelasticity and to form a cohesive internal structure in cosmetic products. Alginate, has been traditionally used in the cosmetic industry as a stabilizer for emulsions and suspensions due to its high stability, and for its thickening and gelling properties [56,57,171,298]. Later, authors indicated that it could be used as a hydrogel for the encapsulation of bioactives, drug delivery systems, and tissue engineering. The use of extreme pH values is not recommended and the concentration of polyvalent metal ions must be controlled in cosmetic formulations using alginates by means of adding sequestrants to avoid altering the viscosity and alginic acid precipitation [28,299]. Agar, a polysaccharide from red algae, can be applied to control both the viscosity and emollience of cosmetic products. Dita et al. confirmed that agar from Gracilaria sp. has a gelling agent capable of having a thickening effect on certain products, such as liquid bath soap, as a cocamide DEA (diethanolamine) substitute [300]. Carrageenans are commonly used in cosmetics as stabilizing, thickening, and gelling agents due to their excellent properties, such as gel-forming ability and chemical stability [27,30,58,301]. The rheological behavior of carrageenan and hybrid carrageenans is temperature sensitive and also depends on the structure, sulfate content, or molecular weight [28,302,303]. Carrageenans can be degraded by carrageenases to produce a number of even-numbered carrageenan oligosaccharides, which exhibit different attractive functions, such as anti-inflammation, anti-tumor, anticoagulation, or antithrombosis effects [304].
Bagal-Kestwal et al. summarized the use of carrageenans (κ, λ, and ι with sodium) as binder and emulsion stabilizers, preventing constituent separation in toothpastes in a recent comprehensive review [274]. In a previous work, these carrageenans were proposed as bodying, emulsion stabilizer, thickeners, dispersion media for shampoos, body lotion, and other cosmetic creams, and as an ingredient binder for personal lubricants [305]. Some comprehensive works [278,306] discussed the most recent breakthroughs in the field of skin care and rejuvenation using cosmeceutical facial masks developed using biopolymer-based hydrogels, which are commonly used for sensitive skins with cooling and soothing effects. Tiwari and co-workers explained the potential of biopolymers in the development of topical matrices (cream, ointment, and gel) employed as dosage forms for burn treatments [298]. Later, authors detailed the potential of carrageenans for drugs delivery or alginates for wound dressings due to their hemostatic potential. Wasupalli et al. pointed out the ability of carrageenans to form unique thermoreversible gels that are very useful to encapsulate active compounds in the cosmetic field [302]. Graham et al. corroborated the potential of thermoresponsive polymers, including agarose or carrageenan, to be used in cosmetics [307]. Hu et al. proposed a simple method to prepare hydrophilic−hydrophobic core−shell microparticles using seaweed polymers (alginates, κ-carrageenan, or agarose) with great prospective applications in the protection of unstable compounds and delivery and controlled release of drugs or bioactives in cosmetics [301].

4. Bioactive Functions

4.1. Moisturization

Moisturizer agents help to maintain skin appearance and elasticity, improving its barrier role against harmful environmental factors [28]. Polysaccharides in cosmetics are efficient at maintaining hydration, and algal extracts that are rich in polysaccharides would be an alternative to hydroxy acids [38] and are also promising for their various properties that are beneficial to skin, including antioxidant, anti-melanogenic, and skin anti-aging properties [23,24,41]. Water:propylene glycol (1:1) extracts of Laminaria japonica showed skin moisturizing properties in in vivo tests with human skin [28,308]. Wang et al. reported that polysaccharides from this seaweed absorbed and retained more moisture than polysaccharides from both the red algae Chondrus cripus, which provides hydrating, moisturizing, and therapeutic effects, and from the green algae Codium tomentosum, which can regulate water distribution in skin [28]. Agar is used as a moisturizer for skin and hair [46]. Mineral-rich seaweed extracts may be found in skin moisturizing agents, facial cleansing products, masks, make-up removers, bath additives, and in products to prevent cellulites. Fatty acids, either in the diet or topically applied, are efficient at preventing trans-epidermal water loss [37].

4.2. Skin Whitening

Skin whitening, particularly demanded in Asia, is also desired to achieve fair and flawless skin. Tyrosinase catalyzes two distinct significant reactions in melanin synthesis: the hydroxylation of l-tyrosine to 3,4-dihydroxy-l-phenylalanine, which is oxidized to dopaquinone, and further converted to melanin. Sun exposure increases the synthesis of both tyrosinase and melanosomes. Different seaweed components can be active tyrosinase inhibitors and are commonly proposed for skin whitening [27,192,193,309]; and brown algal extracts are as effective as kojic acid [284,285,286,310,311]. Similarly, Park et al. reported that P. yezoensis extracts could be proposed as a safe and effective agent to enhance skin whitening and prevention or alleviation of skin wrinkle formation. The extracts exhibited a significant decrease in tyrosinase activity, but was less marked than arbutin. However, arbutin could have secondary undesirable effects, whereas these aqueous seaweed extracts promoted collagen production and, in a study with 23 volunteers, they also enhanced skin brightness [312].
Due to the variety of activities, different fractions of seaweeds have been combined to achieve complementary actions, i.e., between phenolics and polysaccharides [313]. In addition, seaweed mixtures can be explored for their dermo-cosmetic potential [195,291,314,315], i.e., a cream mask from a mixture of seaweeds showing antibacterial, cell proliferation, moisture retention, and tyrosinase inhibitory activities, and also high spread and adhesive abilities, being a nonirritant and safe [314]. In addition, combination with other marine ingredients, such as nanomelanin from Halomonas venusta, isolated from a marine sponge Callyspongia sp., incorporated in a cream fortified with concentrates of seaweed Gelidium spinosum showed antioxidant, antimicrobial, and wound healing activity in addition to improved texture [316].

4.3. UV Protection, Antioxidant and Antiaging

Skin aging, causing thinning, dryness, laxity, fragility, enlarged pores, fine lines, and wrinkles, is a complex process of intrinsic and extrinsic aging. Intrinsic aging refers to the natural degradation of the skin, whereas extrinsic aging results from reactive oxygen species (ROS) generated during exposure to UV radiation [28]. Although the human body possesses an endogenous antioxidant system able to block reactive oxygen species, under conditions of oxidative stress, these defenses can be insufficient and may lead to free radical cell damage to proteins, lipids, and DNA. ROS accumulation may be responsible for photoaging complications, such as cutaneous inflammation, erythema, premature aging, melanoma, and skin cancer [317]. UVB-induced decreased cell viability could be restored by eckstolonol treatment through the enzymatic activities of catalase and superoxide dismutase [200]. Ultraviolet B irradiation induces the production of matrix metalloproteinases, and is structurally and functionally related to zinc endopeptidases, capable of digesting extracellular matrix components, such as collagens, proteoglycans, fibronectin, and laminin [64,90]. Sun-damaged skin shows significantly elevated levels of active gelatinases than intrinsically aged skin, since prolonged exposure to UV radiation causes the enzymatic breakage of collagen and elastin fibers, which are responsible for maintaining the elasticity and integrity of skin [6]. Bioactive compounds derived from marine sources [29] and from algae, especially phlorotannins, have potential anti-photoaging agents, preventing UV-induced oxidative stress, and also inhibit the expressions of MMPs in human dermal fibroblasts [27,176,318]. Riani et al. reported antioxidant and anti-collagenase activity of a Sargassum plagyophyllum extract as active pharmaceutical ingredient for anti-wrinkle cosmetics [319]. The potential of fucoxanthin was also confirmed, and its incorporation was compatible with other components in homogeneous water creams [295].
Since these compounds are preferentially extracted in organic solvents, different examples of macerated extracts with potential photoprotective action can be found [4,204,320,321]. Since other compounds, such as mycosporine-like amino acids, sulfated polysaccharides, carotenoids, and polyphenols, exhibit photoprotective action though a wide range of biological activities, including ultraviolet absorbing, antioxidant, matrix-metalloproteinase inhibitors and anti-aging activities, crude extracts with complex composition can be promising [322]. Gager et al. reported that the phlorotannin-enriched fractions, extracted by maceration and further purified by a liquid–liquid extraction showed antioxidant and photoprotective activities comparable to those of commercial molecules and the anti-aging activity of the obtained fraction was higher than that of epigallocatechin gallate [204]. The efficiency of mixtures of components has been described. Hameury et al. [323] confirmed that an association of ingredients from marine origin revealed activity on the epidermis and the dermis, by regulation of proteins involved in gene expression, cell survival and metabolism, inflammatory processes, dermal extracellular matrix synthesis, melanogenesis and keratinocyte proliferation, migration, and differentiation, thus helping to prevent the visible signs of skin aging.

5. Patents

Seaweeds and their components have been claimed as functional, sensorial, and biological agents in the formulation of cosmetics, cosmeceuticals, and nutricosmetics. Some representative examples on their utilization in the formulation of products with different claimed actions are summarized in Table 7.
Seaweeds can be used either fresh or fermented [349,363,399], and are usually incorporated as extracts, but also a pure single compounds, such as P-334 and DP-334 from Porphyra dentata [378], can be found. Both single species and seaweed mixtures have been found [359,386,387,400]. In addition, seaweed extracts can be combined with extracts from terrestrial plants, medicinal herbs, mushroom, microalgae, and fish [401,402,403,404,405,406], as well as with conventional ingredients [332,350,351,407,408,409,410] or even gold [372]. These mixtures of species and combination with other raw materials during manufacturing of cosmetics can be adopted to generate synergistic effects [335,359,375,411].
A variety of formulations has been found, including liposomes [335] and nano-liposome emulsions for improving the stability of extracts and its compatibility in the cosmetic system, reaching a deep layer of skin and minimizing sensitization responses by direct contact with epidemic cells [334]. Not only have creams been the object of patents, but other specific products, such as masks [412], disposable glove-shaped hand films [357], or mist compositions with fine particles that are widely dispersed when sprayed [413].
Seaweed fractions can confer functional properties or technical properties, such as thickening [346] and emulsifying properties of the polysaccharides, alginate, agar, and carrageenan [324], which also can impart water retention ability, conferring a smooth or moist humectant feeling without imparting stickiness [325,329]. Particularly, in hair cosmetics, they can provide a glossy and elastic feeling for hair and a moist feeling for the scalp [333,350]. Sensorial properties, such as suppressing the stickiness or stiffness of hair, facilitating hairdressing, the extensibility and spread on hair and producing a good feeling in its use, which are desirable for these types of products [330]. Seaweed components can also replace different additives, such as antimicrobials [342] or conventional ultraviolet ray blockers [382].
Seaweed components are interesting in the formulation of different hygienic products, including deodorants, shampoos [330,349,414,415], and cleaning supplies [328,416], especially water washing-free cleaning agents without surfactants. Other proposed formulations of cosmetics were aimed at skin condition [417] and moisturizing [403,410] improvements. Cosmeceuticals containing seaweeds are non-irritants [376], and can perform different functions, such as improving psoriasis and preventing skin problems, especially atopic dermatitis [393], hyperpigmentation [375,401,402,408], acne [410], wrinkles [406], and hair loss [351,401,402]. Many products formulated claims of a plurality of skin care effects, i.e., moisturizing, repair, and anti-aging [338,339,418] or melanin-formation inhibitory action, alleviation of skin stains and freckles, amelioration of roughened and dry skins, and conferring skins gloss and tension [387].
A number of nutricosmetics have been designed to be used in common foods and beverages to improve skin appearance and firmness [386,387], but also claim to improve immunity, strengthening the body’s constitution and improving skin antioxidant capacity [368], losing weight, and beautifying skin [419].
Other patents have claimed pollution-free, safe, sanitary, ecological, environmentally friendly and energy-saving preparation processes [338,339,374,377,419] and also purification and deodorization stages [380,419,420,421,422,423].

6. Conclusions and Future Trends

Cosmetics, cosmeceuticals, and nutricosmetics are daily-use products that are gaining increasing commercial importance for improving the appearance of skin and for treating various dermatologic conditions. Seaweeds are a source of valuable components for the formulation of products due to the variety of functional, sensorial and biological properties they can confer. A diverse group of biologically active compounds, including vitamins, minerals, amino acids, carbohydrates, and lipids, can be extracted from seaweeds to develop conventional and novel cosmeceutical products. The possibility of offering a vast array of activities makes seaweeds a highly attractive renewable and versatile resource, and the importance of extraction and purification processes should also be considered. Other important aspects requiring study are in relation to greener extraction of bioactives, their chemical and biological characterization, as well as stabilization and delivery into novel products. As with other ingredients and applications, quality control and standardization are required for the commercial use of seaweed bioactives.

Author Contributions

Conceptualization, E.F. and H.D.; resources, M.D.T., E.F. and H.D.; writing—original draft preparation, M.P.C., E.M.B., L.L.-H., T.F.-A., N.F.-F., M.D.T., E.F. and H.D.; writing—review and editing, N.F.-F., M.D.T., E.F. and H.D.; project administration, H.D.; funding acquisition, M.D.T. and H.D. All authors have read and agreed to the published version of the manuscript.

Funding

This work has received financial support from the Ministry of Science, Innovation, and Universities of Spain (RTI2018-096376-B-I00) and the Xunta de Galicia (Centro singular de investigación de Galicia accreditation 2019–2022) and the European Union (European Regional Development Fund-ERDF)—Ref. ED431G2019/06.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are available in the original papers cited.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Soulioti, I.; Diomidous, M.; Theodosopoulou, H.; Violaki, N.; Plessa, H.; Charalambidou, M.; Pistolis, J.; Plessas, S.T. Cosmetics: History, products, industry, legislation, regulations and implications in public health. Rev. Clin. Pharmacol. Pharmacokinet. 2013, 27, 5–15. [Google Scholar]
  2. Pangestuti, R.; Shin, K.-H.; Kim, S.-K. Anti-photoaging and potential skin health benefits of seaweeds. Mar. Drugs 2021, 19, 172. [Google Scholar] [CrossRef] [PubMed]
  3. Pimentel, F.B.; Alves, R.C.; Rodrigues, F.; Oliveira, M.B.P.P. Macroalgae-Derived Ingredients for Cosmetic Industry—An Update. Cosmetics 2018, 5, 2. [Google Scholar] [CrossRef] [Green Version]
  4. Resende, D.I.S.P.; Ferreira, M.; Magalhães, C. Trends in the use of marine ingredients in anti-aging cosmetics. Algal Res. 2021, 55, 102273. [Google Scholar] [CrossRef]
  5. Aslam, A.; Bahadar, A.; Liaquat, R.; Saleem, M.; Waqas, A.; Zwawi, M. Algae as an attractive source for cosmetics to counter environmental stress. Sci. Total Environ. 2021, 772, 144905. [Google Scholar] [CrossRef]
  6. Preetha, J.P.; Karthika, K. Cosmeceuticals—An evolution. Int. J. Chemtech Res. 2009, 1, 1217–1223. [Google Scholar]
  7. Chaudhari, P.M.; Kawade, P.V.; Funne, S.M. Cosmeceuticals-a review. Int. J. Pharm. Technol. 2011, 3, 774–798. [Google Scholar]
  8. Draelos, Z.D. Cosmeceuticals: Efficacy and influence on skin tone. Dermatol. Clin. 2014, 32, 137–143. [Google Scholar] [CrossRef]
  9. Querellou, J.; Børresen, T.; Boyen, C.; Dobson, A.; Höfle, M.G.; Ianora, A.; Jaspars, M.; Kijjoa, A.; Olafsen, J.; Rigos, G.; et al. Marine Biotechnology: Realising the Full Potential of Europe. In EurOCEAN—Challenges for Marine Research in the Next Decade; McDonough, N., Ed.; VLIZ Special Publication: Oostende, Belgium, 2010; Volume 47, p. 21. [Google Scholar]
  10. Brandt, F.S.; Cazzaniga, A.; Hann, M. Cosmeceuticals: Current trends and market analysis. Semin. Cutan. Med. Surg. 2011, 30, 141–143. [Google Scholar] [CrossRef]
  11. Pham, A.K.; Dinulos, J.G. Cosmeceuticals for children: Should you care? Curr. Opin. Pediatr. 2014, 26, 446–451. [Google Scholar] [CrossRef]
  12. De Lacerda, D.; Thioly-Bensoussan, D.; Burke, K. Cosmeceuticals for Men. Available online: https://pubmed.ncbi.nlm.nih.gov/24308151/ (accessed on 28 September 2021).
  13. Draelos, Z.D. Cosmeceuticals for Male Skin. Dermatol. Clin. 2018, 36, 17–20. [Google Scholar] [CrossRef]
  14. Lin, T.J. Evolution of cosmetics: Increased need for experimental clinical medicine. J. Exp. Clin. Med. 2010, 2, 49–52. [Google Scholar] [CrossRef]
  15. Faria-Silva, C.; Ascenso, A.; Costa, A.M.; Marto, J.; Carvalheiro, M.; Ribeiro, H.M.; Simões, S. Feeding the skin: A new trend in food and cosmetics convergence. Trends Food Sci. Technol. 2020, 95, 21–32. [Google Scholar] [CrossRef]
  16. Pereira, L. Seaweeds as Source of Bioactive Substances and Skin Care Therapy—Cosmeceuticals, Algotheraphy, and Thalassotherapy. Cosmetics 2018, 5, 68. [Google Scholar] [CrossRef] [Green Version]
  17. Sotelo, C.G.; Blanco, M.; Ramos, P.; Vazquez, J.A.; Perez-Martin, R.I. Sustainable sources from aquatic organisms for cosmeceuticals ingredients. Cosmetics 2021, 8, 48. [Google Scholar] [CrossRef]
  18. Zárate, R.; Portillo, E.; Teixidó, S.; de Carvalho, M.A.A.P.; Nunes, N.; Ferraz, S.; Seca, A.M.L.; Rosa, G.P.; Barreto, M.C. Pharmacological and cosmeceutical potential of Seaweed Beach-casts of Macaronesia. Appl. Sci. 2020, 10, 5831. [Google Scholar] [CrossRef]
  19. Félix, R.; Carmona, A.M.; Félix, C.; Novais, S.C.; Lemos, M.F.L. Industry-friendly hydroethanolic extraction protocols for Grateloupia turuturu UV-shielding and antioxidant compounds. Appl. Sci. 2020, 10, 5304. [Google Scholar] [CrossRef]
  20. Balboa, E.M.; Soto, M.L.; Nogueira, D.R.; González-López, N.; Conde, E.; Moure, A.; Vinardell, M.P.; Mitjans, M.; Domínguez, H. Potential of antioxidant extracts produced by aqueous processing of renewable resources for the formulation of cosmetics. Ind. Crop. Prod. 2014, 58, 104–110. [Google Scholar] [CrossRef] [Green Version]
  21. Dolorosa, M.T.; Nurjanah; Purwaningsih, S.; Anwar, E. Utilization of Kappaphycus alvarezii and Sargassum plagyophyllum from Banten as cosmetic creams. IOP Conf. Ser. Earth Environ. Sci. 2019, 404, 012008. [Google Scholar] [CrossRef]
  22. Wahyuni, T. The Potential and Application of Eucheuma sp. For Solid Soap: A Review. IOP Conf. Ser. Earth Environ. Sci. 2021, 750, 012048. [Google Scholar] [CrossRef]
  23. Jesumani, V.; Du, H.; Aslam, M.; Pei, P.; Huang, N. Potential use of seaweed bioactive compounds in skincare—A review. Mar. Drugs 2019, 17, 688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Jesumani, V.; Du, H.; Pei, P.; Zheng, C.; Cheong, K.-L.; Huang, N. Unravelling property of polysaccharides from Sargassum sp. as an anti-wrinkle and skin whitening property. Int. J. Biol. Macromol. 2019, 140, 216–224. [Google Scholar] [CrossRef] [PubMed]
  25. Lordan, S.; Ross, R.P.; Stanton, C. Marine bioactives as functional food ingredients: Potential to reduce the incidence of chronic diseases. Mar. Drugs 2011, 9, 1056–1100. [Google Scholar] [CrossRef] [Green Version]
  26. Gellenbeck, K.W. Utilization of algal materials for nutraceutical and cosmeceutical applications—What do manufacturers need to know? J. Appl. Phycol. 2012, 24, 309–313. [Google Scholar] [CrossRef]
  27. Thomas, N.V.; Kim, S. Beneficial effects of marine algal compounds in cosmeceuticals. Mar. Drugs 2013, 11, 146–164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Wang, H.M.D.; Chen, C.C.; Huynh, P.; Chang, J.S. Exploring the potential of using algae in cosmetics. Biores. Technol. 2015, 184, 355–362. [Google Scholar] [CrossRef]
  29. Couteau, C.; Coiffard, L. Phycocosmetics and other marine cosmetics, specific cosmetics formulated using marine resources. Mar. Drugs 2020, 18, 322. [Google Scholar] [CrossRef]
  30. Kharkwal, H.; Joshi, D.; Panthari, P.; Pant, M.K.; Kharkwal, A.C. Algae as future drugs. Asian J. Pharm. Clin. Res. 2012, 5, 1–4. [Google Scholar]
  31. Senevirathne, W.S.M.; Kim, S.K. Cosmeceuticals from Algae. In Functional Ingredients from Algae for Foods and Nutraceuticals, 1st ed.; Domínguez, H., Ed.; Woodhead Publishing: Cambridge, UK, 2013; pp. 694–713. [Google Scholar]
  32. Vo, T.; Ngo, D.; Kang, K.; Jung, W.; Kim, S. The beneficial properties of marine polysaccharides in alleviation of allergic responses. Mol. Nutr. Food Res. 2015, 59, 129–138. [Google Scholar] [CrossRef] [PubMed]
  33. Salehi, B.; Sharifi-Rad, J.; Seca, A.M.L.; Pinto, D.C.G.A.; Michalak, I.; Trincone, A.; Mishra, A.P.; Nigam, M.; Zam, W.; Martins, N. Current Trends on Seaweeds: Looking at Chemical Composition, Phytopharmacology, and Cosmetic Applications. Molecules 2019, 24, 4182. [Google Scholar] [CrossRef] [Green Version]
  34. Morais, T.; Cotas, J.; Pacheco, D.; Pereira, L. Seaweeds compounds: An ecosustainable source of cosmetic ingredients? Cosmetics 2021, 8, 8. [Google Scholar] [CrossRef]
  35. Bedoux, G.; Hardouin, K.; Burlot, A.S.; Bourgougnon, N. Bioactive components from seaweeds: Cosmetic applications and future development. Adv. Bot. Res. 2014, 71, 345–378. [Google Scholar]
  36. Agatonovic-Kustrin, S.; Morton, D.W. Cosmeceuticals derived from bioactive substances found in marine algae. J. Oceanogr. Mar. Res. 2013, 1, 106. [Google Scholar]
  37. Couteau, C.; Coiffard, L. Seaweed Application in Cosmetics in Seaweed. In Health and Disease Prevention; Fleurence., J., Levine, I., Eds.; Nikki Levy. Elsevier Inc.: Amsterdam, The Netherlands, 2016; pp. 423–441. [Google Scholar]
  38. Kim, S.K.; Ravichandran, Y.D.; Khan, S.B.; Kim, Y.T. Prospective of the cosmeceuticals derived from marine organisms. Biotechnol. Bioproc. Engineer. 2008, 13, 511–523. [Google Scholar] [CrossRef]
  39. Kim, S.K. Review: Marine cosmeceuticals. J. Cosmet. Dermatol. 2014, 13, 56–67. [Google Scholar] [CrossRef] [PubMed]
  40. Thiyagarasaiyar, K.; Goh, B.-H.; Jeon, Y.-J.; Yow, Y.-Y. Algae Metabolites in Cosmeceutical: An Overview of Current Applications and Challenges. Mar. Drugs 2020, 18, 323. [Google Scholar] [CrossRef] [PubMed]
  41. Ahmed, A.B.A.; Adel, M.; Karimi, P.; Peidayesh, M. Pharmaceutical, cosmeceutical, and traditional applications of marine carbohydrates. Adv. Food Nutr. Res. 2014, 73, 197–220. [Google Scholar]
  42. Kim, J.H.; Lee, J.-E.; Kim, K.H.; Kang, N.J. Beneficial effects of marine algae-derived carbohydrates for skin health. Mar. Drugs 2018, 16, 459. [Google Scholar] [CrossRef] [Green Version]
  43. Shanura Fernando, I.P.; Asanka Sanjeewa, K.K.; Samarakoon, K.W.; Kim, H.S.; Gunasekara, U.K.D.S.S.; Park, Y.J.; Abeytungaa, D.T.U.; Lee, W.W.; Jeon, Y.-J. The potential of fucoidans from Chnoospora minima and Sargassum polycystum in cosmetics: Antioxidant, anti-inflammatory, skin-whitening, and antiwrinkle activities. J. Appl. Phycol. 2018, 30, 3223–3232. [Google Scholar] [CrossRef]
  44. Morya, V.; Kim, J.; Kim, E. Algal fucoidan: Structural and size-dependent bioactivities and their perspectives. Appl. Microbiol. Biotechnol. 2012, 93, 71–82. [Google Scholar] [CrossRef]
  45. Casas, M.P.; Rodríguez-Hermida, V.; Pérez-Larrán, P.; Conde, E.; Liveri, M.T.; Ribeiro, D.; Fernandes, E.; Domínguez, H. In vitro bioactive properties of phlorotannins recovered from hydrothermal treatment of Sargassum muticum. Sep. Purif. Technol. 2016, 167, 117–126. [Google Scholar] [CrossRef]
  46. Ruocco, N.; Costantini, S.; Guariniello, S.; Costantini, M. Polysaccharides from the marine environment with pharmacological, cosmeceutical and nutraceutical potential. Molecules 2016, 21, 551. [Google Scholar] [CrossRef]
  47. Lee, Y.-E.; Kim, H.; Seo, C.; Park, T.; Lee, K.B.; Yoo, S.-Y.; Hong, S.-C.; Kim, J.T.; Lee, J. Marine polysaccharides: Therapeutic efficacy and biomedical applications. Arch. Pharm. Res. 2017, 40, 1006–1020. [Google Scholar] [CrossRef]
  48. Sanjeewa, K.K.A.; Kang, N.; Ahn, G.; Jee, Y.; Kim, Y.T.; Jeon, Y.J. Bioactive potentials of sulfated polysaccharides isolated from brown seaweed Sargassum spp in related to human health applications: A review. Food Hydrocoll. 2018, 81, 200–208. [Google Scholar] [CrossRef]
  49. Fernando, I.P.S.; Kim, K.N.; Kim, D.; Jeon, Y.J. Algal polysaccharides: Potential bioactive substances for cosmeceutical applications. Crit. Rev. Biotechnol. 2019, 39, 99–113. [Google Scholar] [CrossRef] [PubMed]
  50. Cikoš, A.M.; Jerković, I.; Molnar, M.; Šubarić, D.; Jokić, S. New trends for macroalgal natural products applications. Nat. Prod. Res. 2021, 35, 1180–1191. [Google Scholar] [CrossRef] [PubMed]
  51. Fournière, M.; Bedoux, G.; Lebonvallet, N.; Lescchiera, R.; Goff-Pain, C.L.; Bourgougnon, N.; Latire, T. Poly-and oligosaccharide ulva sp. Fractions from enzyme-assisted extraction modulate the metabolism of extracellular matrix in human skin fibroblasts: Potential in anti-aging dermo-cosmetic applications. Mar. Drugs 2021, 19, 156. [Google Scholar] [CrossRef] [PubMed]
  52. Wong, T.; Brault, L.; Gasparotto, E.; Vallée, R.; Morvan, P.Y.; Ferrières, V.; Nugier-Chauvin, C. Formation of Amphiphilic Molecules from the Most Common Marine Polysaccharides, toward a Sustainable Alternative? Molecules 2021, 26, 4445. [Google Scholar] [CrossRef]
  53. Xu, S.-Y.; Kan, J.; Hu, Z.; Liu, Y.; Du, H.; Pang, G.-C.; Cheong, K.-L. Quantification of Neoagaro-Oligosaccharide Production through Enzymatic Hydrolysis and Its Anti-Oxidant Activities. Molecules 2018, 23, 1354. [Google Scholar] [CrossRef] [Green Version]
  54. Zhang, W.; Jin, W.; Duan, D.; Zhang, Q. Structural analysis and anti-complement activity of polysaccharides extracted from Grateloupia livida (Harv.) Yamada. J. Oceanol. Limnol. 2019, 37, 806–814. [Google Scholar] [CrossRef]
  55. Zhang, Y.H.; Song, X.N.; Lin, Y.; Xiao, Q.; Du, X.P.; Chen, Y.H.; Xiao, A.F. Antioxidant capacity and prebiotic effects of Gracilaria neoagaro oligosaccharides prepared by agarase hydrolysis. Int. J. Biol. Macromol. 2019, 137, 177–186. [Google Scholar] [CrossRef]
  56. Sachan, N.K.; Pushkar, S.; Jha, A.; Bhattcharya, A. Sodium alginate: The wonder polymer for controlled drug delivery. J. Pharm. Res. 2009, 2, 1191–1199. [Google Scholar]
  57. Priyadarshani, I.; Rath, B. Commercial and industrial applications of micro algae—A review. J. Algal Biomass Util. 2012, 3, 89–100. [Google Scholar]
  58. Xue, C.; Yu, G.; Hirata, T.; Terao, J.; Lin, H. Antioxidative activities of several marine polysaccharides evaluated in a phosphatidylcholine-liposomal suspension and organic solvents. Biosci. Biotechnol. Biochem. 1998, 62, 206–209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Thevanayagam, H.; Mohamed, S.M.; Chu, W.-L. Assessment of UVB-photoprotective and antioxidative activities of carrageenan in keratinocytes. J. Appl. Phycol. 2014, 26, 1813–1821. [Google Scholar] [CrossRef]
  60. Sun, Z.; Mohamed, M.A.A.; Park, S.Y.; Yi, T.H. Fucosterol protects cobalt chloride induced inflammation by the inhibition of hypoxia-inducible factor through PI3K/Akt pathway. Int. Immunopharmacol. 2015, 29, 642–647. [Google Scholar] [CrossRef] [PubMed]
  61. Fernando, I.P.S.; Jayawardena, T.U.; Kim, H.S.; Vaas, A.; De Silva, H.I.C.; Nanayakkara, C.M.; Abeytunga, D.T.U.; Lee, W.; Ahn, G.; Lee, D.S.; et al. A keratinocyte and integrated fibroblast culture model for studying particulate matter-induced skin lesions and therapeutic intervention of fucosterol. Life Sci. 2019, 233, 116714. [Google Scholar] [CrossRef] [PubMed]
  62. Xie, X.-T.; Zhang, X.; Liu, Y.; Chen, X.-Q.; Cheong, K.-L. Quantification of 3,6-anhydro-galactose in red seaweed polysaccharides and their potential skin-whitening activity. 3 Biotech 2020, 10, 189. [Google Scholar] [CrossRef]
  63. Senni, K.; Gueniche, F.; Foucault-Bertaud, A.; Igondjo-Tchen, S.; Fioretti, F.; Colliec-Jouault, S.; Durand, P.; Guezennec, J.; Godeau, G.; Letourneur, D. Fucoidan a sulfated polysaccharide from brown algae is a potent modulator of connective tissue proteolysis. Arch. Biochem. Biophys. 2006, 445, 56–64. [Google Scholar] [CrossRef] [Green Version]
  64. Moon, H.J.; Lee, S.H.; Ku, M.J.; Yu, B.C.; Jeon, M.J.; Jeong, S.H.; Stonik, V.A.; Zvyagintseca, T.N.; Ermakova, S.P.; Lee, Y.P. Fucoidan inhibits UVB-induced MMP-1 promoter expression and down regulation of type I procollagen synthesis in human skin fibroblasts. Eur. J. Dermatol. 2009, 19, 129–134. [Google Scholar] [CrossRef] [Green Version]
  65. Wang, L.; Oh, J.-Y.; Lee, W.; Jeon, Y.-J. Fucoidan isolated from Hizikia fusiforme suppresses ultraviolet B-induced photodamage by down-regulating the expressions of matrix metalloproteinases and pro-inflammatory cytokines via inhibiting NF-κB, AP-1, and MAPK signaling pathways. Int. J. Biol. Macromol. 2021, 166, 751–759. [Google Scholar] [CrossRef]
  66. Su, W.; Wang, L.; Fu, X.; Ni, L.; Duan, D.; Xu, J.; Gao, X. Protective effect of a fucose-rich fucoidan isolated from Saccharina japonica against ultraviolet B-induced photodamage in vitro in human keratinocytes and in vivo in Zebrafish. Mar. Drugs 2020, 18, 316. [Google Scholar] [CrossRef] [PubMed]
  67. Wang, L.; Jayawardena, T.U.; Yang, H.-W.; Lee, H.-G.; Jeon, Y.-J. The potential of sulfated polysaccharides isolated from the brown seaweed Ecklonia maxima in cosmetics: Antioxidant, anti-melanogenesis, and photoprotective activities. Antioxidants 2020, 9, 724. [Google Scholar] [CrossRef] [PubMed]
  68. Ayoub, A.; Pereira, J.M.; Rioux, L.; Turgeon, S.L.; Beaulieu, M.; Moulin, V.J. Role of seaweed Laminaran from Saccharina longicruris on matrix deposition during dermal tissue-engineered production. Int. J. Biol. Macromol. 2015, 75, 13–20. [Google Scholar] [CrossRef]
  69. Ozanne, H.; Toumi, H.; Roubinet, B.; Landemarre, L.; Lespessailles, E.; Daniellou, R.; Cesaro, A. Laminarin Effects, a β-(1,3)-Glucan, on Skin Cell Inflammation and Oxidation. Cosmetics 2020, 7, 66. [Google Scholar] [CrossRef]
  70. Jiang, N.; Li, B.; Wang, X.; Xu, X.; Liu, X.; Li, W.; Chang, X.; Li, H.; Qi, H. The antioxidant and antihyperlipidemic activities of phosphorylated polysaccharide from Ulva pertusa. Int. J. Biol. Macromol. 2020, 145, 1059–1065. [Google Scholar] [CrossRef]
  71. Yu, B.; Bi, D.; Yao, L.; Li, T.; Gu, L.; Xu, H.; Li, X.; Li, H.; Hu, Z.; Xu, X. The inhibitory activity of alginate against allergic reactions in an ovalbumin-induced mouse model. Food Funct. 2020, 11, 2704–2713. [Google Scholar] [CrossRef]
  72. Szekalska, M.; Sosnowska, K.; Tomczykowa, M.; Winnicka, K.; Kasacka, I.; Tomczyk, M. In vivo anti-inflammatory and anti-allergic activities of cynaroside evaluated by using hydrogel formulations. Biomed. Pharmacother. 2020, 121, 109681. [Google Scholar] [CrossRef]
  73. Holdt, S.L.; Kraan, S. Bioactive compounds in seaweed: Functional food applications and legislation. J. Appl. Phycol. 2011, 23, 543–597. [Google Scholar] [CrossRef]
  74. Xing, M.; Cao, Q.; Wang, Y.; Xiao, H.; Zhao, J.; Zhang, Q.; Ji, A.; Song, S. Advances in Research on the Bioactivity of Alginate Oligosaccharides. Mar. Drugs 2020, 18, 144. [Google Scholar] [CrossRef] [Green Version]
  75. Deville, C.; Gharbi, M.; Dandrifosse, G.; Peulen, O. Study on the effects of laminarin, a polysaccharide from seaweed, on gut characteristics. J. Sci. Food Agric. 2007, 87, 1717–1725. [Google Scholar] [CrossRef]
  76. Shannon, E.; Conlon, M.; Hayes, M. Seaweed Components as Potential Modulators of the Gut Microbiota. Mar. Drugs 2021, 19, 358. [Google Scholar] [CrossRef]
  77. Choi, J.; Ha, Y.; Joo, C.; Cho, K.K.; Kim, S.; Choi, I.S. Inhibition of oral pathogens and collagenase activity by seaweed extracts. J. Environ. Biol. 2012, 33, 115–121. [Google Scholar]
  78. Gómez-Mascaraque, L.G.; Martínez-Sanz, M.; Martínez-López, R.; Martínez-Abad, A.; Panikuttira, B.; López-Rubio, A.; Tuohy, M.G.; Hogan, S.A.; Brodkorb, A. Characterization and gelling properties of a bioactive extract from Ascophyllum nodosum obtained using a chemical-free approach. Curr. Res. Food Sci. 2021, 4, 354–364. [Google Scholar] [CrossRef] [PubMed]
  79. Huang, Y.; Jiang, H.; Mao, X.; Ci, F. Laminarin and Laminarin Oligosaccharides Originating from Brown Algae: Preparation, Biological Activities, and Potential Applications. J. Ocean Univ. China 2021, 20, 641–653. [Google Scholar] [CrossRef]
  80. Kadam, S.U.; Tiwari, B.K.; O’Donnell, C.P. Extraction, structure and biofunctional activities of laminarin from brown algae. Int. J. Food Sci. Technol. 2015, 50, 24–31. [Google Scholar] [CrossRef]
  81. Costa, A.M.S.; Rodrigues, J.M.M.; Pérez-Madrigal, M.M.; Dove, A.P.; Mano, J.F. Modular Functionalization of Laminarin to Create Value-Added Naturally Derived Macromolecules. J. Am. Chem. Soc. 2020, 142, 19689–19697. [Google Scholar] [CrossRef] [PubMed]
  82. Li, J.; Cai, C.; Yang, C.; Li, J.; Sun, T.; Yu, G. Recent advances in pharmaceutical potential of brown algal polysaccharides and their derivatives. Curr. Pharm. Des. 2019, 25, 1290–1311. [Google Scholar] [CrossRef]
  83. Tümen Erden, S.; Ekentok Atici, C.; Cömez, B.; Sezer, A.D. Preparation and in vitro characterization of laminarin based hydrogels. J. Res. Pharm. 2021, 25, 164–172. [Google Scholar]
  84. Zargarzadeh, M.; Amaral, A.J.R.; Custódio, C.A.; Mano, J.F. Biomedical applications of laminarin. Carbohydr. Polym. 2020, 232, 115774. [Google Scholar] [CrossRef]
  85. Narayanaswamy, R.; Jo, B.W.; Choi, S.K.; Ismail, I.S. Fucoidan: Versatile cosmetic ingredient. An overview. J. Appl. Cosmetol. 2013, 31, 131–138. [Google Scholar]
  86. Baweja, P.; Kumar, S.; Sahoo, D.; Levine, I. Biology of Seaweeds in Seaweed. In Health and Disease Prevention; Fleurence, J., Levine, I., Eds.; Nikki Levy Elsevier Inc.: Amsterdam, The Netherlands, 2016; pp. 41–106. [Google Scholar]
  87. Li, N.; Zhang, Q.; Song, J. Toxicological evaluation of fucoidan extracted from Laminaria japonica in Wistar rats. Food Chem. Toxicol. 2005, 43, 421–426. [Google Scholar] [CrossRef]
  88. Citkowska, A.; Szekalska, M.; Winnicka, K. Possibilities of fucoidan utilization in the development of pharmaceutical dosage forms. Mar. Drugs 2019, 17, 458. [Google Scholar] [CrossRef] [Green Version]
  89. Jiao, G.L.; Yu, G.L.; Zhang, J.Z.; Ewart, H.S. Chemical structures and bioactivities of sulfated polysaccharides from marine algae. Mar. Drugs 2011, 9, 196–223. [Google Scholar] [CrossRef] [Green Version]
  90. Wijesinghe, W.; Jeon, Y. Biological activities and potential industrial applications of fucose rich sulfated polysaccharides and fucoidans isolated from brown seaweeds: A review. Carbohydr. Polym. 2012, 88, 13–20. [Google Scholar] [CrossRef]
  91. Kartik, A.; Akhil, D.; Lakshmi, D.; Panchamoorthy Gopinath, K.; Arun, J.; Sivaramakrishnan, R.; Pugazhendhi, A. A critical review on production of biopolymers from algae biomass and their applications. Bioresour. Technol. 2021, 329, 124868. [Google Scholar] [CrossRef] [PubMed]
  92. Xue, C.-H.; Fang, Y.; Lin, H.; Chen, L.; Li, Z.-J.; Deng, D.; Lu, C.-X. Chemical characters and antioxidative properties of sulfated polysaccharides from Laminaria japonica. J. Appl. Phycol. 2001, 13, 67–70. [Google Scholar] [CrossRef]
  93. Kim, S.M.; Kang, K.; Jeon, J.S.; Jho, E.H.; Kim, C.Y.; Nho, C.W.; Um, B.H. Isolation of phlorotannins from Eisenia bicyclis and their hepatoprotective effect against oxidative stress induced by tert-butyl hyperoxide. Appl. Biochem. Biotechnol. 2011, 165, 1296–1307. [Google Scholar] [CrossRef] [PubMed]
  94. Li, Y.; Wang, X.; Jiang, Y.; Wang, J.; Hwang, H.; Yang, X.; Wang, P. Structure characterization of low molecular weight sulfate Ulva polysaccharide and the effect of its derivative on iron deficiency anemia. Int. J. Biol. Macromol. 2019, 126, 747–754. [Google Scholar] [CrossRef]
  95. Senni, K.; Pereira, J.; Gueniche, F.; Delbarre-Ladrat, C.; Sinquin, C.; Ratiskol, J.; Godeau, G.; Fischer, A.-M.; Helley, D.; Colliec-Jouault, S. Marine polysaccharides: A source of bioactive molecules for cell therapy and tissue engineering. Mar. Drugs 2011, 9, 1664–1681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Ali Karami, M.; Sharif Makhmalzadeh, B.; Pooranian, M.; Rezai, A. Preparation and optimization of silibinin-loaded chitosan–fucoidan hydrogel: An in vivo evaluation of skin protection against UVB. Pharm. Dev. Technol. 2021, 26, 209–219. [Google Scholar] [CrossRef]
  97. Costa, L.S.; Fidelis, G.P.; Telles, C.B.S.; Dantas-Santos, N.; Camara, R.B.G.; Cordeiro, S.L.; Costa, M.S.; Almetida-Lima, J.; Oliveira, R.M.; Alburquerque, I.R.; et al. Antioxidant and antiproliferative activities of heterofucans from the seaweed Sargassum filipendula. Mar. Drugs 2011, 9, 952–966. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Qiao, L.; Li, Y.; Chi, Y.; Ji, Y.; Gao, Y.; Hwang, H.; Aker, W.G.; Wang, P. Rheological properties, gelling behavior and texture characteristics of polysaccharide from Enteromorpha prolifera. Carbohydr. Polym. 2016, 136, 1307–1314. [Google Scholar] [CrossRef] [PubMed]
  99. Lin, G.-P.; Wu, D.-S.; Xiao, X.-W.; Huang, Q.-Y.; Chen, H.-B.; Liu, D.; Fu, H.; Chen, X.-H.; Zhao, C. Structural characterization and antioxidant effect of green alga Enteromorpha prolifera polysaccharide in Caenorhabditis elegans via modulation of microRNAs. Int. J. Biol. Macromol. 2020, 150, 1084–1092. [Google Scholar] [CrossRef]
  100. Adrien, A.; Bonnet, A.; Dufour, D.; Badouin, S.; Maugard, T.; Bridiau, N. Pilot production of ulvans from Ulva sp. and their effects on hyaluronan and collagen production in cultured dermal fibroblasts. Carbohydr. Polym. 2017, 157, 1306–1314. [Google Scholar] [CrossRef]
  101. Lahaye, M.; Robic, A. Structure and function properties of Ulvan, a polysaccharide from green seaweeds. Biomacromol 2007, 8, 1765–1774. [Google Scholar] [CrossRef]
  102. Mo’o, F.R.C.; Wilar, G.; Devkota, H.P.; Wathoni, N. Ulvan, a polysaccharide from Macroalga Ulva sp.: A review of chemistry, biological activities and potential for food and biomedical applications. Appl. Sci. 2020, 10, 5488. [Google Scholar] [CrossRef]
  103. Kidgell, J.T.; Carnachan, S.M.; Magnusson, M.; Lawton, R.J.; Sims, I.M.; Hinkley, S.F.R.; de Nys, R.; Glasson, C.R.K. Are all ulvans equal? A comparative assessment of the chemical and gelling properties of ulvan from blade and filamentous Ulva. Carbohyd. Polym. 2021, 264, 118010. [Google Scholar] [CrossRef] [PubMed]
  104. Li, B.; Xu, H.; Wang, X.; Wang, Y.; Jiang, N.; Qi, H.; Liu, X. Antioxidant and antihyperlipidemic activities of high sulfate content purified polysaccharide from Ulva pertusa. Int. J. Biol. Macromol. 2020, 146, 756–762. [Google Scholar] [CrossRef]
  105. Chen, X.; Fu, X.; Huang, L.; Xu, J.; Gao, X. Agar oligosaccharides: A review of preparation, structures, bioactivities and application. Carbohydr. Polym. 2021, 265, 118076. [Google Scholar]
  106. Aziz, E.; Batool, R.; Khan, M.U.; Rauf, A.; Akhtar, W.; Heydary, M.; Rehman, S.; Shahzad, T.; Malik, A.; Mosavat, S.-H.; et al. An overview on red algae bioactive compounds and their pharmaceutical applications. J. Altern. Complement. Med. 2021, 17, 20190203. [Google Scholar]
  107. Laurienzo, P. Marine polysaccharides in pharmaceutical applications: An overview. Mar. Drugs 2010, 8, 2435–2465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Ghanbarzadeh, M.; Golmoradizadeh, A.; Homaei, A. Carrageenans and carrageenases: Versatile polysaccharides and promising marine enzymes. Phytochem. Rev. 2018, 17, 535–571. [Google Scholar] [CrossRef]
  109. Isaka, S.; Cho, K.; Nakazono, S.; Abu, R.; Ueno, M.; Kim, D.; Oda, T. Antioxidant and anti-inflammatory activities of porphyrin isolated from discolored nori (Porphyra yezoensis). Int. J. Biol. Macromol. 2015, 74, 68–75. [Google Scholar] [CrossRef]
  110. Beaumont, M.; Tran, R.; Vera, G.; Niedrist, D.; Rousset, A.; Pierre, R.; Shastri, V.P.; Forget, A. Hydrogel-Forming Algae Polysaccharides: From Seaweed to Biomedical Applications. Biomacromolecules 2021, 22, 1027–1052. [Google Scholar] [CrossRef]
  111. Kwon, M.; Nam, T. Porphyran induces apoptosis related signal pathway in AGS gastric cancer cell lines. Life Sci. 2006, 79, 1956–1962. [Google Scholar] [CrossRef]
  112. Pacheco-Quito, E.-M.; Ruiz-Caro, R.; Veiga, M.-D. Carrageenan: Drug Delivery Systems and Other Biomedical Applications. Mar. Drugs 2020, 18, 583. [Google Scholar] [CrossRef] [PubMed]
  113. Qiu, Y.; Jiang, H.; Fu, L.; Ci, F.; Mao, X. Porphyran and oligo-porphyran originating from red algae Porphyra: Preparation, biological activities, and potential applications. Food Chem. 2021, 349, 129209. [Google Scholar] [CrossRef]
  114. Araujo, I.W.F.; Vanderlei, E.S.O.; Rodrigues, J.A.G.; Coura, C.O.; Quindere, A.L.G.; Fontes, B.P.; Queiroz, I.N.L.; Jorge, R.J.B.; Bezerra, M.M.; Silva, A.A.R.; et al. Effects of a sulfated polysaccharide isolated from the red seaweed Solieria filiformis on models of nociception and inflammation. Carbohydr. Polym. 2011, 86, 1207–1215. [Google Scholar] [CrossRef] [Green Version]
  115. Zhang, Y.; Tian, R.; Wu, H.; Li, X.; Li, S.; Bian, L. Evaluation of acute and sub-chronic toxicity of Lithothamnion sp. in mice and rats. Toxicol. Rep. 2020, 7, 852–858. [Google Scholar] [CrossRef]
  116. Cheong, K.-L.; Qiu, H.-M.; Du, H.; Liu, Y.; Khan, B.M. Oligosaccharides derived from red seaweed: Production, properties, and potential health and cosmetic application. Molecules 2018, 23, 245. [Google Scholar] [CrossRef] [Green Version]
  117. Bleakley, S.; Hayes, M. Algal proteins: Extraction, application, and challenges concerning production. Foods 2017, 6, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Fleurence, J. Seaweed Proteins. In Proteins in Food Processing; Yada, R.Y., Ed.; Woodhead Publishing Limited: Cambridge, UK, 2004; pp. 197–213. [Google Scholar]
  119. Gressler, V.; Fujii, M.T.; Martins, A.P.; Colepicolo, P.; Mancini-Filho, J.; Pinto, E. Biochemical composition of two red seaweed species grown on the Brazilian coast. J. Sci. Food Agric. 2011, 91, 1687–1692. [Google Scholar] [CrossRef]
  120. Cheung, R.C.F.; Ng, T.B.; Wong, J.H. Marine peptides: Bioactivities and applications. Mar. Drugs 2015, 13, 4006–4043. [Google Scholar] [CrossRef] [PubMed]
  121. Qasim, R. Amino acids composition of some common seaweeds. Pak. J. Pharmac. Sci. 1991, 4, 49–54. [Google Scholar]
  122. Wong, K.H.; Cheung, P.C.K. Nutritional evaluation of some subtropical red and green seaweeds. Part II. In vitro protein digestibility and amino acid profiles of protein concentrates. Food Chem. 2001, 72, 11–17. [Google Scholar] [CrossRef]
  123. Matanjun, P.; Mohamed, S.; Mustapha, N.M.; Muhammad, K. Nutrient content of tropical edible seaweeds, Eucheuma cottoni, Caulerpa lentillifera and Sargassum polycysum. J. Appl. Phycol. 2009, 21, 75–80. [Google Scholar] [CrossRef]
  124. Admassu, H.; Abdalbasit, M.; Gasmalla, A.; Yang, R.; Zhao, W. Bioactive peptides derived from seaweed protein and their health benefits: Antihypertensive, antioxidant, and antidiabetic properties. J. Food Sci. 2018, 83, 6–16. [Google Scholar] [CrossRef] [Green Version]
  125. Meisel, H. Multifunctional peptides encrypted in milk proteins. Biofactors 2004, 21, 55–61. [Google Scholar] [CrossRef]
  126. Murray, B.A.; FitzGerald, R.J. Angiotensin converting enzyme inhibitory peptides derived from food proteins: Biochemistry, bioactivity and production. Curr. Pharmac. Des. 2007, 13, 773–791. [Google Scholar] [CrossRef]
  127. Harnedy, P.A.; FitzGerald, R.J. Bioactive proteins, peptides, and amino acids from macroalgae. J. Phycol. 2011, 47, 218–232. [Google Scholar] [CrossRef]
  128. Indumathi, P.; Mehta, A. A novel anticoagulant peptide from the nori hydrolysate. J. Funct. Foods 2016, 20, 606–617. [Google Scholar] [CrossRef]
  129. Jo, C.; Khan, F.F.; Khan, M.I.; Iqbal, J. Marine bioactive peptides: Types, structures, and physiological functions. Food Rev. Int. 2016, 33, 44–61. [Google Scholar] [CrossRef]
  130. 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]
  131. Sridhar, K.; Inbaraj, B.S.; Chen, B.-H. Recent developments on production, purification and biological activity of marine peptides. Food Res. Int. 2021, 47, 110468. [Google Scholar] [CrossRef]
  132. Pangestuti, R.; Kim, S.K. Seaweed Proteins, Peptides, and Amino Acids. In Seaweed Sustainability: Food and Non-Food Applications; Tiwari, B.K., Toy, D.J., Eds.; Academic Press: San Diego, CA, USA, 2015; pp. 125–140. [Google Scholar]
  133. Yanshin, N.; Kushnareva, A.; Lemesheva, V.; Birkemeyer, C.; Tarakhovskaya, E. Chemical composition and potential practical application of 15 red algal species from the White Sea Coast (the Arctic Ocean). Molecules 2021, 26, 2489. [Google Scholar] [CrossRef]
  134. Lee, H.-A.; Kim, I.-H.; Nam, T.-J. Bioactive peptide from Pyropia yezoensis and its anti-inflammatory activities. Int. J. Molec. Med. 2015, 36, 1701–1706. [Google Scholar] [CrossRef] [Green Version]
  135. Ryu, J.; Park, S.J.; Kim, I.H.; Choi, Y.H.; Nam, T.J. Protective effect of porphyra-334 on UVA-induced photoaging in human skin fibroblasts. Int. J. Mol. Medic. 2014, 34, 796–803. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Romarís-Hortas, V.; Bermejo-Barrera, P.; Moreda-Piñeiro, A. Ultrasound-assisted enzymatic hydrolysis for iodinated amino acid extraction from edible seaweed before reversed-phase high performance liquid chromatography-inductively coupled plasma-mass spectrometry. J. Chromatogr. A 2013, 1309, 33–40. [Google Scholar] [CrossRef]
  137. Shick, J.M.; Dunlap, W.C.; Buettner, G.R. Ultraviolet (UV) Protection in Marine Organisms II, Biosynthesis, Accumulation, and Sunscreening Function of Mycosporine-Like Amino Acids. In Free Radicals in Chemistry, Biology and Medicine; Yoshikawa, S., Toyokuni, S., Yamamoto, Y., Naito, Y., Eds.; OICA International: London, UK, 2000; pp. 215–228. [Google Scholar]
  138. Bedoux, G.; Hardouin, K.; Marty, C.; Taupin, L.; Vandanjon, L.; Bourgougnon, N. Chemical characterization and photoprotective activity measurement of extracts from the red macroalga Solieria chordalis. Bot. Mar. 2014, 57, 291–301. [Google Scholar] [CrossRef] [Green Version]
  139. Dunlap, W.C.; Yamamoto, Y. Small-molecule antioxidants in marine organisms: Antioxidant activity of mycosporine-glycine. Compar. Biochem. Physiol. 1995, 112, 105–114. [Google Scholar] [CrossRef]
  140. Bandaranayake, W.M. Mycosporines: Are they nature’s sunscreens? Nat. Prod. Rep. 1998, 15, 159–172. [Google Scholar] [CrossRef]
  141. Conde, F.R.; Churio, M.S.; Previtali, C.M. The photoprotector mechanism of mycosporine-like amino acids. Excited-state properties and photostability of porphyra-334 in aqueous solution. J. Photochem. Photobiol. B Biol. 2000, 56, 139–144. [Google Scholar] [CrossRef]
  142. Gröniger, A.; Sinha, R.P.; Klisch, M.; Häder, D.P. Photoprotective compounds in cyanobacteria, phytoplankton and macroalgae—A database. J. Photochem. Photobiol. B Biol. 2000, 58, 115–122. [Google Scholar] [CrossRef]
  143. Adams, N.L.; Shick, J.M. Mycosporine-like amino acids prevent UVB-induced abnormalities durin early development of the green sea urchin Strongylocentrotus droebachiensis. Mar. Biol. 2001, 138, 267–280. [Google Scholar] [CrossRef]
  144. Singh, S.P.; Kumari, S.; Rastogi, R.P.; Singh, K.L.; Sinha, R.P. Mycosporine-like amino acids (MAAs): Chemical structure, biosynthesis and significance as UV-absorbing/screening compounds. Indian J. Experim. Biol. 2008, 46, 7–17. [Google Scholar]
  145. Karsten, U.; Sawall, T.; Wiencke, C. A survey of the distribution of UV-absorbing substances in tropical macroalgae. Phycol. Res. 2006, 46, 271–279. [Google Scholar]
  146. Rastogi, R.P.; Sonani, R.R.; Madamwar, D. UV Photoprotectants from Algae—Synthesis and Bio-Functionalities. In Algal Green Chemistry; Rastogi, R.P., Madamwar, D., Pandey, A., Eds.; Elsevier: Amsterdam, The Netherlands, 2017; pp. 17–38. [Google Scholar]
  147. Orfanoudaki, M.; Hartmann, A. Chemical profiling of mycosporine-like amino acids in twenty-three red algal species. J. Phycol. 2019, 55, 393–403. [Google Scholar] [CrossRef]
  148. Gianeti, M.D.; Maia Campos, P.M.B.G. Efficacy evaluation of a multifunctional cosmetic formulation: The benefits of a combination of active antioxidant substances. Molecules 2014, 19, 18268–18282. [Google Scholar] [CrossRef]
  149. Leandro, A.; Pereira, L.; Gonçalves, A.M.M. Diverse applications of marine macroalgae. Mar. Drugs 2020, 18, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Chrapusta, E.; Kaminski, A.; Duchnik, K.; Bober, B.; Adamski, M.; Bialczyk, J. Mycosporine-like amino acids: Potential health and beauty ingredients. Mar. Drugs 2017, 15, 326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Schmid, D.; Schürch, C.; Zülli, F. UV-A sunscreen from red algae for protection against premature skin aging. Cosmetics 2004, 2004, 139–143. [Google Scholar]
  152. Rangel, K.C.; Villela, L.Z.; Pereira, K.D.C.; Debonsi, H.M.; Gaspar, L.R. Assessment of the photoprotective potential and toxicity of Antarctic red macroalgae extracts from Curdiea racovitzae and Iridaea cordata for cosmetic use. Algal Res. 2020, 50, 101984. [Google Scholar] [CrossRef]
  153. Barceló-Villalobos, M.; Figueroa, F.L.; Korbee, N.; Álvarez-Gómez, F.; Abreu, M.H. Production of Mycosporine-Like amino acids from Gracilaria vermiculophylla (Rhodophyta) cultured through one year in an integrated multi-trophic aquaculture (IMTA) system. Mar. Biotechnol. 2017, 19, 246–254. [Google Scholar] [CrossRef] [PubMed]
  154. Athukorala, Y.; Trang, S.; Kwok, C.; Yuan, Y.V. Antiproliferative and antioxidant activities and mycosporine-Like amino acid profiles of wild-Harvested and cultivated edible Canadian marine red macroalgae. Molecules 2016, 21, 119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Pliego-Cortés, H.; Bedoux, G.; Boulho, R.; Taupin, L.; Freile-Pelegrin, Y.; Bourgougnon, N.; Robledo, D. Stress tolerance and photoadaptation to solar radiation in Rhodymenia pseudopalmata (Rhodophyta) through mycosporine-like amino acids, phenolic compounds, and pigments in an Integrated Multi-Trophic Aquaculture System. Algal Res. 2019, 41, 101542. [Google Scholar] [CrossRef]
  156. Leelapornpisid, P.; Mungmai, L.; Sirithunyalug, B.; Jiranusornkul, S.; Peerapornpisal, Y. A novel moisturizer extracted from freshwater macroalga [Rhizoclonium hieroglyphicum (C. Agardh) Kützing] for skin care cosmetic. Chiang Mai J. Sci. 2014, 41, 1195–1207. [Google Scholar]
  157. Francavilla, M.; Franchi, M.; Monteleone, M.; Caroppo, C. The red seaweed Gracilaria gracilis as a multi products source. Mar. Drugs 2013, 11, 3754–3776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Fan, X.; Bai, L.; Mao, X.; Zhang, X. Novel peptides with anti-proliferation activity from the Porphyra haitanesis hydrolysate. Process Biochem. 2017, 60, 98–107. [Google Scholar] [CrossRef]
  159. Verdy, C.; Branka, J.E.; Mekideche, N. Quantitative assessment of lactate and progerin production in normal human cutaneous cells during normal ageing: Effect of an Alaria esculenta extract. Int. J. Cosmet. Sci. 2011, 33, 462–466. [Google Scholar] [CrossRef]
  160. Arad, S.; Yaron, A. Natural pigments from red microalgae for use in foods and cosmetics. Trends Food Sci. Technol. 1992, 3, 92–97. [Google Scholar] [CrossRef]
  161. Bermejo, R.; Talavera, E.M.; del Valle, C.; Alvarez-Pez, J.M. C-phycocyanin incorporated into reverse micelles: A fluorescence study. Colloids Surf. B Biointerfaces 2000, 18, 51–59. [Google Scholar] [CrossRef]
  162. Chronakis, I.S. Biosolar proteins from aquatic algae. Dev. Food Sci. 2000, 41, 39–75. [Google Scholar] [CrossRef]
  163. Rossano, R.; Ungaro, N.; D’Ambrosio, A.; Liuzzi, G.M.; Riccio, P. Extracting and purifying R-phycoeythrin from Mediterranean red algae Corallina elongata Ellis and Solander. J. Biotecnol. 2003, 101, 289–296. [Google Scholar] [CrossRef]
  164. Sekar, S.; Chandramohan, M. Phycobiliproteins as a commodity: Trends in applied research, patents and commercialization. J. Appl. Phycol. 2008, 20, 113–136. [Google Scholar] [CrossRef]
  165. Viskari, P.J.; Colyer, C.L. Rapid extraction of phycobiliproteins from cultures cyanobacteria samples. Anal. Biochem. 2003, 319, 263–271. [Google Scholar] [CrossRef]
  166. Martins, M.; Vieira, F.A.; Correia, I.; Ferreira, R.A.S.; Abreu, H.; Coutinho, J.A.P.; Ventura, S.P.M. Recovery of phycobiliproteins from the red macroalga Gracilaria sp. using ionic liquid aqueous solutions. Green Chem. 2016, 18, 4287–4296. [Google Scholar] [CrossRef]
  167. Saluri, M.; Kaldmäe, M.; Rospu, M.; Sirkel, H.; Paalme, T.; Landreh, M.; Tuvikene, R. Spatial variation and structural characteristics of phycobiliproteins from the red algae Furcellaria lumbricalis and Coccotylus truncatus. Algal Res. 2020, 52, 102058. [Google Scholar] [CrossRef]
  168. Osório, C.; Machado, S.; Peixoto, J.; Bessada, S.; Pimentel, F.B.; Alves, R.C.; Oliveira, M.B.P.P. Pigments content (chlorophylls, fucoxanthin and phycobiliproteins) of different commercial dried algae. Separations 2020, 7, 33. [Google Scholar] [CrossRef]
  169. Fernando, I.P.S.; Lee, W.; Ahn, G. Marine algal flavonoids and phlorotannins; an intriguing frontier of biofunctional secondary metabolites. Crit. Rev. Biotechnol. 2021, 1–23. [Google Scholar] [CrossRef] [PubMed]
  170. Shrestha, A.; Pradhan, R.; Ghotekar, S.; Dahikar, S.; Marasini, B.P. Phytochemical Analysis and Anti-Microbial Activity of Desmostachya Bipinnata: A review. J. Med. Chem. Sci. 2021, 4, 36–41. [Google Scholar]
  171. Wijesinghe, W.; Jeon, Y. Biological activities and potential cosmeceutical applications of bioactive components from brown seaweeds: A review. Phytochem. Rev. 2011, 10, 431–443. [Google Scholar] [CrossRef]
  172. Ferreres, F.; Lopes, G.; Gil-Izquierdo, A.; Andrade, P.B.; Sousa, C.; Mouga, T.; Valentão, P. Phlorotannin extracts from fucales characterized by HPLC-DAD-ESI-MSn: Approaches to hyaluronidase inhibitory capacity and antioxidant properties. Mar. Drugs 2012, 10, 2766–2781. [Google Scholar] [CrossRef] [Green Version]
  173. Balboa, E.M.; Conde, E.; Moure, A.; Falqué, E.; Domínguez, H. In vitro antioxidant properties of crude extracts and compounds from brown algae. Food Chem. 2013, 138, 1764–1785. [Google Scholar] [CrossRef] [PubMed]
  174. Phasanasophon, K.; Kim, S. Antioxidant and Cosmeceutical Activities of Agarum cribrosum Phlorotannin Extracted by Ultrasound Treatment. Nat. Prod. Commun. 2018, 13, 565–570. [Google Scholar] [CrossRef]
  175. Gheda, S.; Naby, M.A.; Mohamed, T.; Pereira, L.; Khamis, A. Antidiabetic and antioxidant activity of phlorotannins extracted from the brown seaweed Cystoseira compressa in streptozotocin-induced diabetic rats. Environ. Sci. Pollut. Res. 2021, 28, 22886–22901. [Google Scholar] [CrossRef] [PubMed]
  176. Joe, M.; Kim, S.; Choi, H.; Shin, W.; Park, G.; Kang, D.; Kim, Y.K. The inhibitory effects of eckol and dieckol from Ecklonia stolonifera on the expression of matrix metalloproteinase-1 in human dermal fibroblasts. Biol. Pharm. Bull. 2006, 29, 1735–1739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Le, Q.; Li, Y.; Qian, Z.; Kim, M.; Kim, S. Inhibitory effects of polyphenols isolated from marine alga Ecklonia cava on histamine release. Process Biochem. 2009, 44, 168–176. [Google Scholar] [CrossRef]
  178. Barbosa, M.; Lopes, G.; Valentão, P.; Ferreres, F.; Gil-Izquierdo, A.; Pereira, D.M.; Andrade, P.B. Edible seaweeds’ phlorotannins in allergy: A natural multi-target approach. Food Chem. 2018, 265, 233–241. [Google Scholar] [CrossRef]
  179. Sugiura, Y.; Kinoshita, Y.; Misumi, S.; Yamatani, H.; Katsuzaki, H.; Hayashi, Y.; Murase, N. Correlation between the seasonal variations in phlorotannin content and the antiallergic effects of the brown alga Ecklonia cava subsp. stolonifera. Algal Res. 2021, 58, 102398. [Google Scholar] [CrossRef]
  180. Barbosa, M.; Lopes, G.; Andrade, P.-B.; Valentão, P. Bioprospecting of brown seaweeds for biotechnological applications: Phlorotannin actions in inflammation and allergy network. Trends Food Sci. Technol. 2019, 86, 153–171. [Google Scholar] [CrossRef]
  181. Catarino, M.D.; Amarante, S.J.; Mateus, N.; Silva, A.M.S.; Cardoso, S.M. Brown algae phlorotannins: A marine alternative to break the oxidative stress, inflammation and cancer network. Foods 2021, 10, 1478. [Google Scholar] [CrossRef] [PubMed]
  182. Kang, H.S.; Kim, H.R.; Byun, D.S.; Son, B.W.; Nam, T.J.; Choi, J.S. Tyrosinase inhibitors isolated from the edible brown alga Ecklonia stolonifera. Arch. Pharm. Res. 2004, 27, 1226–1232. [Google Scholar] [CrossRef] [PubMed]
  183. Heo, S.J.; Ko, S.; Cha, S.H.; Kang, D.H.; Park, H.S.; Choi, Y.U.; Kim, D.; Jung, W.K.; Yeon, Y.J. Effect of phlorotannins isolated from Ecklonia cava on melanogenesis and their protective effect against photo-oxidative stress induced by UV-B radiation. Toxicol. Vitr. 2009, 23, 1123–1130. [Google Scholar] [CrossRef] [PubMed]
  184. Kang, S.M.; Heo, S.J.; Kim, K.N.; Lee, S.H.; Yang, H.M.; Kim, A.D.; Jeon, Y.J. Molecular docking studies of a phlorotannin, dieckol isolated from Ecklonia cava with tyrosinase inhibitory activity. Bioorg. Med. Chem. 2012, 20, 311–316. [Google Scholar] [CrossRef] [PubMed]
  185. Kim, J.H.; Lee, S.; Park, S.; Park, J.S.; Kim, Y.H.; Yang, S.Y. Slow-Binding Inhibition of Tyrosinase by Ecklonia cava Phlorotannins. Mar. Drugs 2019, 17, 359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Susano, P.; Silva, J.; Alves, C.; Martins, A.; Gaspar, H.; Pinteus, S.; Mouga, T.; Goettert, M.I.; Petrovski, Ž.; Branco, L.B.; et al. Unravelling the Dermatological Potential of the Brown Seaweed Carpomitra costata. Mar. Drugs 2021, 19, 135. [Google Scholar] [CrossRef]
  187. Hwang, H.; Chen, T.; Nines, R.G.; Shin, H.; Stoner, G.D. Photochemoprevention of UVB-induced skin carcinogenesis in SKH-1 mice by brown algae polyphenols. Int. J. Cancer 2006, 119, 2742–2749. [Google Scholar] [CrossRef]
  188. Hwang, E.; Park, S.-Y.; Sun, Z.-W.; Shin, H.-S.; Lee, D.-G.; Yi, T.H. The protective effects of fucosterol against skin damage in UVB-Irradiated human dermal fibroblasts. Mar. Biotechnol. 2014, 16, 361–370. [Google Scholar] [CrossRef]
  189. Handajani, F.; Prabowo, S. Sargassum duplicatum extract reduced artritis severity score and periarticular tissue matrix metalloproteinase- 1 (MMP-1) expression in ajuvan artritis exposed to cold stressor. Sys. Rev. Pharm. 2020, 11, 302–307. [Google Scholar]
  190. Ryu, B.; Ahn, B.-N.; Kang, K.-H.; Kim, Y.-S.; Li, Y.-X.; Kong, C.-S.; Kim, S.-K.; Kim, D.G. Dioxinodehydroeckol protects human keratinocyte cells from UVB-induced apoptosis modulated by related genes Bax/Bcl-2 and caspase pathway. J. Photochem. Photobiol. B 2015, 153, 352–357. [Google Scholar] [CrossRef]
  191. Vo, T.S.; Kim, S.-K.; Ryu, B.; Ngo, D.H.; Yoon, N.-Y.; Bach, L.G.; Hang, N.T.N.; Ngo, D.N. The suppressive activity of fucofuroeckol-a derived from brown algal Ecklonia stolonifera okamura on UVB-induced mast cell degranulation. Mar. Drugs 2018, 16, 1. [Google Scholar] [CrossRef] [Green Version]
  192. Manandhar, B.; Paudel, P.; Seong, S.H.; Jung, H.A.; Choi, J.S. Characterizing eckol as a therapeutic aid: A systematic review. Mar. Drugs 2019, 17, 361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Manandhar, B.; Wagle, A.; Seong, S.H.; Paudel, P.; Kim, H.-R.; Jung, H.A.; Choi, J.S. Phlorotannins with potential anti-tyrosinase and antioxidant activity isolated from the marine seaweed Ecklonia stolonifera. Antioxidants 2019, 8, 240. [Google Scholar] [CrossRef] [Green Version]
  194. Sanjeewa, K.K.A.; Kim, E.-A.; Son, K.-T.; Jeon, Y.-J. Bioactive properties and potentials cosmeceutical applications of phlorotannins isolated from brown seaweeds: A review. J. Photochem. Photobiol. B 2016, 162, 100–105. [Google Scholar] [CrossRef] [PubMed]
  195. Freitas, R.; Martins, A.; Silva, J.; Alves, C.; Pinteus, S.; Alves, J.; Teodoro, F.; Ribeiro, H.M.; Gonçalves, L.; Petrovski, Ž.; et al. Highlighting the Biological Potential of the Brown Seaweed Fucus spiralis for Skin Applications. Antioxidants 2020, 9, 611. [Google Scholar]
  196. Arunkumar, K.; Raj, R.; Raja, R.; Carvalho, I.S. Brown seaweeds as a source of anti-hyaluronidase compounds. S. Afr. J. Bot. 2021, 139, 470–477. [Google Scholar] [CrossRef]
  197. Ko, S.; Lee, M.; Lee, J.; Lee, S.; Lim, Y.; Jeon, Y. Dieckol, a phlorotannin isolated from a brown seaweed, Ecklonia cava, inhibits adipogenesis through AMP-activated protein kinase (AMPK) activation in 3T3-L1 preadipocytes. Environ. Toxicol. Pharmacol. 2013, 36, 1253–1260. [Google Scholar] [CrossRef]
  198. Eom, S.H.; Lee, E.H.; Park, K.; Kwon, J.Y.; Kim, P.H.; Jung, W.K.; Kim, Y.M. Eckol from Eisenia bicyclis inhibits inflammation through the Akt/NF-κB signaling in Propionibacterium acnes-induced human keratinocyte Hacat cells. J. Food Biochem. 2017, 41, 12312. [Google Scholar] [CrossRef]
  199. Hermund, D.B.; Plaza, M.; Turner, C.; Jónsdóttir, R.; Kristinsson, H.G.; Jacobsen, C.; Nielsen, K.F. Structure dependent antioxidant capacity of phlorotannins from Icelandic Fucus vesiculosus by UHPLC-DAD-ECD-QTOFMS. Food Chem. 2018, 240, 904–909. [Google Scholar]
  200. Jang, J.; Ye, B.-R.; Heo, S.-J.; Oh, C.; Kang, D.-H.; Kim, J.H.; Affan, A.; Yoon, K.-T.; Choi, Y.-U.; Park, S.C.; et al. Photo-oxidative stress by ultraviolet-B radiation and antioxidative defense of eckstolonol in human keratinocytes. Environ. Toxicol. Pharmacol. 2012, 34, 926–934. [Google Scholar] [CrossRef]
  201. Lee, J.-H.; Eom, S.-H.; Lee, E.-H.; Jung, Y.-J.; Kim, H.-J.; Jo, M.-R.; Son, K.-T.; Lee, H.-J.; Kim, J.H.; Lee, M.-S.; et al. In vitro antibacterial and synergistic effect of phlorotannins isolated from edible brown seaweed Eisenia bicyclis against acne-related bacteria. Algae 2014, 29, 47–55. [Google Scholar] [CrossRef]
  202. Sugiura, Y.; Matsuda, K.; Yamada, Y.; Nishikawa, M.; Shioya, K.; Katsuzaki, H.; Imai, K.; Amano, H. Isolation of a new anti-allergic phlorotannin, phlorofucofuroeckol-B, from an edible brown alga, Eisenia arborea. Biosci. Biotechnol. Biochem. 2006, 70, 2807–2811. [Google Scholar] [CrossRef] [Green Version]
  203. Le Lann, K.; Surget, G.; Couteau, C.; Coiffard, L.; Cerantola, S.; Gaillard, F.; Larnicol, M.; Zubia, M.; Gerard, F.; Poupart, N.; et al. Sunscreen, antioxidant, and bactericide capacities of phlorotannins from the brown macroalga Halidrys siliquosa. J. Appl. Phycol. 2016, 28, 3547–3559. [Google Scholar] [CrossRef] [Green Version]
  204. Gager, L.; Connan, S.; Molla, M.; Couteau, C.; Arbona, J.F.; Coiffard, L.; Cérantola, S.; Stiger-Pouvreau, V. Active phlorotannins from seven brown seaweeds commercially harvested in Brittany (France) detected by 1H NMR and in vitro assays: Temporal variation and potential valorization in cosmetic applications. J. Appl. Phycol. 2020, 32, 2375–2386. [Google Scholar] [CrossRef]
  205. Lee, J.H.; Ko, J.-Y.; Samarakoon, K.; Oh, J.-Y.; Heo, S.-J.; Kim, C.-Y.; Nah, J.-W.; Jang, M.-K.; Lee, J.-S.; Jeon, Y.-J. Preparative isolation of sargachromanol E from Sargassum siliquastrum by centrifugal partition chromatography and its anti-inflammatory activity. Food Chem. Toxicol. 2013, 62, 54–60. [Google Scholar] [CrossRef] [PubMed]
  206. Kim, J.; Ahn, B.; Kong, C.; Kim, S. The chromenesargachromanol E inhibits ultraviolet A-induced ageing of skin in human dermal fibroblasts. Br. J. Dermatol. 2013, 168, 968–976. [Google Scholar] [CrossRef] [PubMed]
  207. Balboa, E.M.; Li, Y.; Ahn, B.; Eom, S.; Domínguez, H.; Jiménez, C.; Rodríguez, J. Photodamage attenuation effect by a tetraprenyltoluquinol chromane meroterpenoid isolated from Sargassum muticum. J. Photochem. Photobiol. B 2015, 148, 51–58. [Google Scholar] [CrossRef]
  208. Azam, M.S.; Choi, J.; Lee, M.-S.; Kim, H.-R. Hypopigmenting Effects of Brown Algae-Derived Phytochemicals: A Review on Molecular Mechanisms. Mar. Drugs 2017, 15, 297. [Google Scholar] [CrossRef] [Green Version]
  209. Hamid, N.; Ma, Q.; Boulom, S.; Liu, T.; Zheng, Z.; Balbas, J.; Robertson, J. Seaweed Minor Constituents. In Seaweed Sustainability: Food and Non-Food Applications; Tiwari, B.K., Troy, D., Eds.; Academic Press: London, UK, 2015; pp. 193–242. [Google Scholar]
  210. Stiger-Pouvreau, V.; Zubia, M. Macroalgal diversity for sustainable biotechnological development in French tropical overseas territories. Bot. Mar. 2020, 63, 17–41. [Google Scholar] [CrossRef] [Green Version]
  211. 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]
  212. Biris-Dorhoi, E.-S.; Michiu, D.; Pop, C.R.; Rotar, A.M.; Tofana, M.; Pop, O.L.; Socaci, S.A.; Farcas, A.C. Macroalgae—A Sustainable Source of Chemical Compounds with Biological Activities. Nutrients 2020, 12, 3085. [Google Scholar] [CrossRef] [PubMed]
  213. Kumari, P.; Kumar, M.; Reddy, C.R.K.; Jha, B. Algal Lipids, Fatty Acids and Sterols. In Functional Ingredients from Algae for Foods and Nutraceuticals; Domínguez, H., Ed.; Woodhead Publishing: Cambridge, UK, 2013; pp. 87–134. [Google Scholar]
  214. Lever, J.; Brkljača, R.; Kraft, G.; Urban, S. Natural Products of Marine Macroalgae from South Eastern Australia, with Emphasis on the Port Phillip Bay and Heads Regions of Victoria. Mar. Drugs 2020, 18, 142. [Google Scholar] [CrossRef] [Green Version]
  215. Saadaoui, I.; Rasheed, R.; Abdulrahman, N.; Bounnit, T.; Cherif, M.; Al Jabri, H.; Mraiche, F. Algae-Derived Bioactive Compounds with Anti-Lung Cancer Potential. Mar. Drugs 2020, 18, 197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Plaza, M.; Cifuentes, A.; Ibáñez, E. In the search of new functional food ingredients from algae. Trends Food Sci. Technol. 2008, 19, 31–39. [Google Scholar] [CrossRef] [Green Version]
  217. Gómez-Zorita, S.; González-Arceo, M.; Trepiana, J.; Eseberri, I.; Fernández-Quintela, A.; Milton-Laskibar, I.; Aguirre, L.; González, M.; Portillo, M.P. Anti-Obesity Effects of Macroalgae. Nutrients 2020, 12, 2378. [Google Scholar] [CrossRef] [PubMed]
  218. Lange, K.W.; Hauser, J.; Nakamura, Y.; Kanaya, S. Dietary seaweeds and obesity. Food Sci. Hum. Wellness 2015, 4, 87–96. [Google Scholar] [CrossRef] [Green Version]
  219. Lee, S.; Lee, Y.S.; Jung, S.H.; Kang, S.S.; Shin, K.H. Anti-oxidant activities of fucosterol from the marine algae Pelvetia siliquosa. Arch. Pharm. Res. 2003, 26, 719–722. [Google Scholar] [CrossRef] [PubMed]
  220. Abdul, Q.A.; Choi, R.J.; Jung, H.A.; Choi, J.S. Health benefit of fucosterol from marine algae: A review. J. Sci. Food Agric. 2016, 96, 1856–1866. [Google Scholar] [CrossRef]
  221. 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]
  222. Perumal, P.; Sowmiya, R.; Prasanna Kumar, S.; Ravikumar, S.; Deepak, P.; Balasubramani, G. Isolation, structural elucidation and antiplasmodial activity of fucosterol compound from brown seaweed, Sargassum linearifolium against malarial parasite Plasmodium falciparum. Nat. Prod. Res. 2017, 32, 1316–1319. [Google Scholar] [CrossRef]
  223. Sugawara, T.; Kushiro, M.; Zhang, H.; Nara, E.; Ono, H.; Nagao, A. Lysophosphatidylcholine enhances carotenoid uptake from mixed micelles by Caco-2 human intestinal cells. J. Nutr. 2001, 131, 2921–2927. [Google Scholar] [CrossRef] [Green Version]
  224. Okada, T.; Mizuno, Y.; Sibayama, S.; Hosokawa, M.; Miyashita, H. Antiobesity effects of Undaria lipid capsules prepared with scallop phospholipids. J. Food Sci. 2011, 76, 2–6. [Google Scholar] [CrossRef]
  225. Rexliene, J.; Sridhar, J. Extraction and characterization of essential oil from Portieria hornemannii with applications in antibacterial edible films. Res. J. Biotechnol. 2021, 16, 81–89. [Google Scholar]
  226. Lee, H.J.; Dang, H.T.; Kang, G.J.; Yang, E.J.; Park, S.S.; Yoon, W.J.; Jung, J.H.; Kang, H.K.; Yoo, E.S. Two enone fatty acids isolated from Gracilaria verrucosa suppress the production of inflammatory mediators by down-regulating NF-êB and STAT1 activity in lipopolysaccharide-stimulated raw 264.7 cells. Arch. Pharm. Res. 2009, 32, 453–462. [Google Scholar] [CrossRef] [PubMed]
  227. Patra, J.K.; Das, G.; Baek, K. Chemical composition and antioxidant and antibacterial activities of an essential oil extracted from an edible seaweed, Laminaria japonica L. Molecules 2015, 20, 12093–12113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Lee, Y.; Shin, K.; Jung, S.; Lee, S. Effects of the extracts from the marine algae Pelvetia siliquosa on hyperlipidemia in rats. Korean J. Pharmacogn. 2004, 35, 143–146. [Google Scholar]
  229. Zhangfan, M.; Xiaoling, S.; Ping, D.; Gaoli, L.; Shize, P.; Xiangran, S.; Haifeng, H.; Li, P.; Jie, H. Fucosterol exerts antiproliferative effects on human lung cancer cells by inducing apoptosis, cel cycle arrest and targeting of Raf/MEK/ERK signaling pathway. Phytomedicine 2019, 61, 152809. [Google Scholar]
  230. Richard, D.; Kefi, K.; Barbe, U.; Bausero, P.; Visioli, F. Polyunsaturated fatty acids as antioxidants. Pharmacol. Res. 2008, 57, 451–455. [Google Scholar] [CrossRef]
  231. Mohy El-Din, S.M. Fatty acid profiling as bioindicator of chemical stress in marine Pterocladia capillacea, Sargassum hornschuchii and Ulva lactuca. Int. J. Environ. Sci. Technol. 2018, 15, 791–800. [Google Scholar] [CrossRef]
  232. Calandra, I.P.; Simonetti, S. Nutritional supplements in dermocosmetology. Ann. Ital. Dermatol. Clin. Sper. 1995, 49, 49–56. [Google Scholar]
  233. Jacob, L.; Baker, C.; Farris, P. Vitamin-based cosmeceuticals. Cosmet. Dermatol. 2012, 25, 405. [Google Scholar]
  234. 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]
  235. Kafi, R.; Kwak, H.S.R.; Schumacher, W.E.; Cho, S.; Hanft, V.N.; Hamilton, T.A.; King, A.L.; Neal, J.D.; Varani, J.; Fisher, G.J.; et al. Improvement of naturally aged skin with vitamin a (retinol). Arch. Dermatol. 2007, 143, 606–612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Searle, T.; Al-Niaimi, F.; Ali, F.R. The top 10 cosmeceuticals for facial hyperpigmentation. Dermatol. Ther. 2020, 33, 14095. [Google Scholar] [CrossRef] [PubMed]
  237. Bissett, D.L.; Oblong, J.E.; Goodman, L.J. Topical Vitamins. In Cosmetic Dermatology: Products and Procedures; Draelos, Z.D., Ed.; Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2015; pp. 336–345. [Google Scholar]
  238. Kilinç, B.; Semra, C.; Gamze, T.; Hatice, T.; Koru, E. Seaweeds for Food and Industrial Applications. In Food Industry; Muzzalupo, I., Ed.; InTech: Rijeka, Croatia, 2013; pp. 735–748. [Google Scholar]
  239. Campiche, R.; Curpen, S.J.; Lutchmanen-Kolanthan, V.; Gougeon, S.; Cherel, M.; Laurent, G.; Gempeler, M.; Schuetz, R. Pigmentation effects of blue light irradiation on skin and how to protect against them. Int. J. Cosmet. Sci. 2020, 42, 399–406. [Google Scholar] [CrossRef] [PubMed]
  240. Kim, S.K.; Bhatnagar, I. Physical, chemical, and biological properties of wonder kelp-Laminaria. Adv. Food Nutr. Res. 2011, 64, 85–96. [Google Scholar] [PubMed]
  241. Watanabe, F.; Yabuta, Y.; Bito, T.; Teng, F. Vitamin B12-containing plant food sources for vegetarians. Nutrients 2014, 6, 1861–1873. [Google Scholar] [CrossRef] [Green Version]
  242. Bito, T.; Tanioka, Y.; Watanabe, F. Characterization of vitamin B12 compounds from marine foods. Fish. Sci. 2018, 84, 747–755. [Google Scholar] [CrossRef]
  243. Manela-Azulay, M.; Bagatin, E. Cosmeceuticals vitamins. Clin. Dermatol. 2009, 27, 469–474. [Google Scholar] [CrossRef]
  244. Lorencini, M.; Brohem, C.A.; Dieamant, G.C.; Zanchin, N.I.; Maibach, H.I. Active ingredients against human epidermal aging. Ageing Res. Rev. 2014, 15, 100–115. [Google Scholar] [CrossRef]
  245. Paiva, L.; Lima, E.; Neto, A.I.; Marcone, M.; Baptista, J. Health-promoting ingredients from four selected Azorean macroalgae. Food Res. Int. 2016, 89, 432–438. [Google Scholar] [CrossRef] [PubMed]
  246. Mercurio, D.G.; Wagemaker, T.A.L.; Alves, V.M.; Benevenuto, C.G.; Gaspar, L.R.; Maia Campos, P.M.B.G. In vivo photoprotective effects of cosmetic formulations containing UV filters, vitamins, Ginkgo biloba and red algae extracts. J. Photochem. Photobiol. B 2015, 153, 121–126. [Google Scholar] [CrossRef]
  247. Sumi, H.; Osada, K.; Yatagai, C.; Naito, S.; Yanagisawa, Y. High concentration of vitamin K1 proved in the seaweeds and sweet potato leaves. J.-Jpn. Soc. Food Sci. Technol. 2003, 50, 63–66. [Google Scholar] [CrossRef] [Green Version]
  248. Kamao, M.; Suhara, Y.; Tsugawa, N.; Uwano, M.; Yamaguchi, N.; Uenishi, K.; Ishida, H.; Sasaki, S.; Okano, T. Vitamin K content of foods and dietary vitamin K intake in Japanese young women. J. Nutr. Sci. Vitaminol. 2007, 53, 464–470. [Google Scholar] [CrossRef] [Green Version]
  249. Del Mondo, A.; Smerilli, A.; Sané, E.; Sansone, C.; Brunet, C. Challenging microalgal vitamins for human health. Microb. Cell Factories 2020, 19, 201. [Google Scholar] [CrossRef] [PubMed]
  250. Rupérez, P. Mineral content of edible marine seaweeds. Food Chem. 2002, 79, 23–26. [Google Scholar] [CrossRef]
  251. Circuncisão, A.R.; Catarino, M.D.; Cardoso, S.M.; Silva, A.M.S. Minerals from Macroalgae Origin: Health Benefits and Risks for Consumers. Mar. Drugs 2018, 16, 400. [Google Scholar] [CrossRef] [Green Version]
  252. Lozano Muñoz, I.; Díaz, N.F. Minerals in edible seaweed: Health benefits and food safety issues. Crit. Rev. Food Sci. Nutr. 2020, 18, 1–16. [Google Scholar] [CrossRef] [PubMed]
  253. Tarnowska, M.; Briancon, S.; Resende de Azevedo, J.; Chevalier, Y.; Bolzinger, M.-A. Inorganic ions in the skin: Allies or enemies? Int. J. Pharm. 2020, 591, 119991. [Google Scholar] [CrossRef]
  254. Kraan, S. Pigments and Minor Compounds in Algae. In Functional Ingredients from Algae for Foods and Nutraceuticals; Domínguez, H., Ed.; Woodhead Publishing: Cambridge, UK, 2013; pp. 205–251. [Google Scholar]
  255. Mohamed, S.; Hashim, S.N.; Rahman, H.A. Seaweeds: A sustainable functional food for complementary and alternative therapy. Trends Food Sci. Technol. 2012, 23, 83–96. [Google Scholar] [CrossRef]
  256. Polefka, T.G.; Bianchini, R.J.; Shapiro, S. Interaction of mineral salts with the skin: A literature survey. Int. J. Cosmet. Sci. 2012, 34, 416–423. [Google Scholar] [CrossRef]
  257. Martins, A.; Vieira, H.; Gaspar, H.; Santos, S. Review: Marketed marine natural products in the pharmaceutical and cosmeceutical industries: Tips for success. Mar. Drugs 2014, 12, 1066–1101. [Google Scholar] [CrossRef] [Green Version]
  258. Guillerme, J.-B.; Couteau, C.; Coiffard, L. Applications for Marine Resources in Cosmetics. Cosmetics 2017, 4, 35. [Google Scholar] [CrossRef] [Green Version]
  259. Alves, A.; Sousa, E.; Sousa, E.; Kijjoa, A.; Pinto, M. Marine-derived compounds with potential use as cosmeceuticals and nutricosmetics. Molecules 2020, 25, 2536. [Google Scholar] [CrossRef] [PubMed]
  260. Chakdar, H.; Pabbi, S. Algal Pigments for Human Health and Cosmeceuticals. In Algal Green Chemistry; Rastogi, R.P., Madamwar, D., Pandey, A., Eds.; Elsevier: Amsterdam, The Netherlands, 2017; pp. 171–188. [Google Scholar]
  261. Christaki, E.; Bonos, E.; Giannenasa, I.; Florou-Paneria, P. Functional properties of carotenoids originating from algae. J. Sci. Food Agric. 2013, 93, 5–11. [Google Scholar] [CrossRef]
  262. Rajauria, G. In-Vitro Antioxidant Properties of Lipophilic Antioxidant Compounds from 3 Brown Seaweed. Antioxidants 2019, 8, 596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  263. Maeda, H.; Hosokawa, M.; Sashima, T.; Funayama, K.; Miyashita, K. Fucoxanthin from edible seaweed, Undaria pinnatifida, shows antiobesity effect through UCP1 expression in white adipose tissues. Biochem. Biophys. Res. Commun. 2005, 332, 392–397. [Google Scholar] [CrossRef] [PubMed]
  264. Gammone, M.A.; D’Orazio, N. Anti-obesity activity of the marine carotenoid fucoxanthin. Mar. Drugs 2015, 13, 2196–2214. [Google Scholar] [CrossRef] [PubMed]
  265. Rajauria, G.; Foley, B.; Abu-Ghannam, N. Characterization of dietary fucoxanthin from Himanthalia elongata brown seaweed. Food Res. Int. 2017, 99, 995–1001. [Google Scholar] [CrossRef]
  266. Shimoda, H.; Tanaka, J.; Shan, S.; Maoka, T. Anti-pigmentary activity of fucoxanthin and its influence on skin mRNA expression of melanogenic molecules. J. Pharm. Pharmacol. 2010, 62, 1137–1145. [Google Scholar] [CrossRef]
  267. Peng, J.; Yuan, J.P.; Wu, C.F.; Wang, J.H. Fucoxanthin, a marine carotenoid present in brown seaweeds and diatoms: Metabolism and bioactivities relevant to human health. Mar. Drugs 2011, 9, 1806–1828. [Google Scholar] [CrossRef] [PubMed]
  268. Heo, S.; Jeon, Y. Protective effect of fucoxanthin isolated from Sargassum siliquastrum on UV-B induced cell damage. J. Photochem. Photobiol. B 2009, 95, 101–107. [Google Scholar] [CrossRef] [PubMed]
  269. Heo, S.-J.; Yoon, W.-J.; Kim, K.-N.; Ahn, G.-N.; Kang, S.-M.; Kang, D.-H.; Affan, A.; Oh, C.; Jung, W.-K.; Jeon, Y.-J. Evaluation of anti-inflammatory effect of fucoxanthin isolated from brown algae in lipopolysaccharide-stimulated RAW 264.7 macrophages. Food Chem. Toxicol. 2010, 48, 2045–2051. [Google Scholar] [CrossRef]
  270. Matsui, M.; Tanaka, K.; Higashiguchi, N.; Okawa, H.; Yamada, Y.; Tanaka, K.; Taira, S.; Aoyama, T.; Takanishi, M.; Natsume, C.; et al. Protective and therapeutic effects of fucoxanthin against sunburn caused by UV irradiation. J. Pharmacol. Sci. 2016, 132, 55–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  271. Nie, J.; Chen, D.; Lu, Y.; Dai, Z. Effects of various blanching methods on fucoxanthin degradation kinetics, antioxidant activity, pigment composition, and sensory quality of Sargassum fusiforme. LWT-Food Sci. Tecnol. 2021, 143, 111179. [Google Scholar] [CrossRef]
  272. Jia, Y.P.; Sun, L.; Yu, H.S.; Liang, L.P.; Li, W.; Ding, H.; Song, X.B.; Zhang, L.J. The Pharmacological Effects of Lutein and Zeaxanthin on Visual Disorders and Cognition Diseases. Molecules 2017, 22, 610. [Google Scholar] [CrossRef] [PubMed]
  273. Naser, W. The cosmetic effects of various natural biofunctional ingredients against skin aging: A review. Int. J. Appl. Pharm. 2021, 13, 10–18. [Google Scholar] [CrossRef]
  274. Bagal-Kestwal, D.R.; Pan, M.H.; Chiang, B.-H. Properties and Applications of Gelatin, Pectin, and Carrageenan Gels. In Bio Monomers for Green Polymeric Composite Materials; Visakh, P.M., Bayraktar, O., Menon, G., Eds.; John Wiley & Sons Ltd.: Hoboken, NJ, USA, 2019; pp. 117–140. [Google Scholar]
  275. Tarman, K.; Ain, N.H.; Sulistiawati, S.; Hardjito, L.; Sadi, U. Biological process to valorise marine algae. IOP Conf. Ser. Earth Environ. Sci. 2020, 414, 012026. [Google Scholar] [CrossRef]
  276. Li, D.; Wu, Z.; Martini, N.; Wen, J. Advanced carrier systems in cosmetics and cosmeceuticals: A review. J. Cosmet. Sci. 2011, 62, 549–563. [Google Scholar]
  277. Peng, J.; Xu, W.; Ni, D.; Zhang, W.; Zhang, T.; Guang, C.; Mu, W. Preparation of a novel water-soluble gel from Erwinia amylovora levan. Int. J. Biol. Macromol. 2019, 122, 469–478. [Google Scholar] [CrossRef]
  278. Prima, N.R.; Andriyono, S. Techniques of additional Kappaphycus alvarezii on seaweed face mask production. IOP Conf. Ser. Earth Environ. Sci. 2021, 679, 012021. [Google Scholar] [CrossRef]
  279. Eom, S.; Kim, Y.; Kim, S. Antimicrobial effect of phlorotannins from marine brown algae. Food Chem. Toxicol. 2012, 50, 3251–3255. [Google Scholar] [CrossRef]
  280. Abu-Ghannam, N.; Rajauria, G. Antimicrobial Activity of Compounds Isolated from Algae. In Functional Ingredients from Algae for Foods and Nutraceuticals; Domínguez, H., Ed.; Woodhead Publishing: Cambridge, UK, 2013; pp. 287–306. [Google Scholar]
  281. Pérez, M.J.; Falqué, E.; Domínguez, H. Antimicrobial action of compounds from marine seaweed. Mar. Drugs 2016, 14, 52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  282. Lopes, G.; Sousa, C.; Silva, L.R.; Pinto, E.; Andrade, P.B.; Bernardo, J.; Mouga, T.; Valentão, P. Can phlorotannins purified extracts constitute a novel pharmacological alternative for microbial infections with associated inflammatory conditions? PLoS ONE 2012, 7, 31145. [Google Scholar] [CrossRef]
  283. Amiguet, V.T.; Jewell, L.E.; Mao, H.; Sharma, M.; Hudson, J.B.; Durst, T.; Allard, M.; Rochefort, G.; Arnason, J.T. Antibacterial properties of a glycolipid-rich extract and active principle from Nunavik collections of the macroalgae Fucus evanescens C. Agardh (Fucaceae). Can. J. Microbiol. 2011, 57, 745–749. [Google Scholar] [CrossRef] [PubMed]
  284. Arguelles, E.D.L.R.; Sapin, A.B. Bioprospecting of Turbinaria ornata (Fucales, phaeophyceae) for cosmetic application: Antioxidant, tyrosinase inhibition and antibacterial activities. J. Int. Soc. Southeast Asian Agric. Sci. 2020, 26, 30–41. [Google Scholar]
  285. Arguelles, E.R.; Sapin, A.B. Bioactive properties of Sargassum siliquosum J. Agardh (Fucales, Ochrophyta) and its potential as source of skin-lightening active ingredient for cosmetic application. J. Appl. Pharm. Sci. 2020, 10, 51–58. [Google Scholar]
  286. Arguelles, E.L.R. Evaluation of antioxidant capacity, tyrosinase inhibition, and antibacterial activities of brown seaweed, Sargassum ilicifolium (Turner) c. agardh 1820 for cosmeceutical application. J. Fish. Environ. 2021, 45, 64–77. [Google Scholar]
  287. Kim, I.H.; Lee, D.G.; Lee, S.H.; Ha, J.M.; Ha, B.J.; Kim, S.K.; Le, J.H. Antibacterial activity of Ulva lactuca against methicillin-resistant Staphylococcus aureus (MRSA). Biotechnol. Bioprocess Eng. 2007, 12, 579–582. [Google Scholar] [CrossRef]
  288. Pierre, G.; Sopena, V.; Juin, C.; Mastouri, A.; Graber, M.; Maugard, T. Antibacterial activity of a sulfated galactan extracted from the marine alga Chaetomorpha aerea against Staphylococcus aureus. Biotechnol. Bioprocess Eng. 2011, 16, 937–945. [Google Scholar] [CrossRef]
  289. Ha, Y.; Choi, J.; Lee, B.; Moon, H.E.; Cho, K.K.; Choi, I.S. Inhibitory effects of seaweed extracts on the growth of the vaginal bacterium Gardnerella vaginalis. J. Environ. Biol. 2014, 35, 537–542. [Google Scholar] [PubMed]
  290. Wei, Y.; Liu, Q.; Yu, J.; Feng, Q.; Zhao, L.; Song, H.; Wang, W. Antibacterial mode of action of 1, 8-dihydroxy-anthraquinone from porphyrahaitanensis against Staphylococcus aureus. Nat. Prod. Res. 2015, 29, 976–979. [Google Scholar] [CrossRef]
  291. Widowati, I.; Suprijanto, J.; Trianto, A.; Puspita, M.; Bedoux, G.; Bourgougnon, N. Antibacterial activity and proximate analysis of Sargassum extracts as cosmetic additives in a moisturizer cream. AACL Bioflux 2019, 12, 1961–1969. [Google Scholar]
  292. Poyato, C.; Thomsen, B.R.; Hermund, D.B.; Ansorena, D.; Astiasarán, I.; Jónsdóttir, R.; Kristinsson, H.G.; Jacobsen, C. Antioxidant effect of water and acetone extracts of Fucus vesiculosus on oxidative stability of skin care emulsions. Eur. J. Lipid Sci. Technol. 2017, 119, 1600072. [Google Scholar] [CrossRef] [Green Version]
  293. 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] [Green Version]
  294. Nursid, M.; Khatulistiani, T.S.; Noviendri, D.; Hapsari, F.; Hardiyati, T. Total phenolic content, antioxidant activity and tyrosinase inhibitor from marine red algae extract collected from Kupang, East Nusa Tenggara. IOP Conf. Ser. Earth Environ. Sci. 2020, 493, 012013. [Google Scholar] [CrossRef]
  295. Sari, D.S.P.; Saputra, E.; Alamsjah, M.A. Potential of fucoxanthin content in Sargassum sp. On sunscreen cream preparation. Int. J. Recent Technol. Eng. 2019, 7, 448–451. [Google Scholar]
  296. Fransiska, D.; Darmawan, M.; Sinurat, E.; Sedayu, B.B.; WArdana, Y.W.; Herdiana, Y.; Setiana, G.P. Characteristics of Oil in Water (o/w) Type Lotions Incorporated with Kappa/Iota Carrageenan. IOP Conf. Ser. Earth Environ. Sci. 2021, 715, 012050. [Google Scholar] [CrossRef]
  297. Cunha, S.C.; Fernandes, J.O.; Vallecillos, L.; Cano-Sancho, G.; Domingo, J.L.; Pocurull, E.; Borrull, F.; Mauvault, A.L.; Ferrari, E.; Fernández-Tejedor, M.; et al. Co-occurrence of musk fragrances and UV-filters in seafood and macroalgae collected in European hotspots. Environ. Res. 2015, 143, 65–71. [Google Scholar] [CrossRef]
  298. Tiwari, R.; Tiwari, G.; Lahiri, A.; Vadivelan, R.; Rai, A.K. Localized delivery of drugs through medical textiles for treatment of burns: A perspective approach. Adv. Pharm. Bull. 2021, 11, 248–260. [Google Scholar]
  299. Mohan, R.; Singh, S.; Kumar, G.; Srivastava, M. Evaluation of gelling behavior of natural gums and their formulation prospects. Indian J. Pharm. Educ. Res. 2020, 54, 1016–1023. [Google Scholar] [CrossRef]
  300. Dita, L.R.; Sudarno, S.; Triastuti, J. Utilization of agar Gracilaria sp. as a natural thickener on liquid bath soap formulation. IOP Conf. Ser. Earth Environ. Sci. 2020, 441, 012021. [Google Scholar] [CrossRef]
  301. Hu, B.; Han, L.; Ma, R.; Phillips, G.O.; Nishinari, K.; Fang, Y. All-natural food-grade hydrophilic–hydrophobic core–shell microparticles: Facile fabrication based on gel-network-restricted antisolvent method. ACS Appl. Mater. Interfaces 2019, 11, 11936–11946. [Google Scholar] [CrossRef] [PubMed]
  302. Wasupalli, G.K.; Verma, D. Polysaccharides as Biomaterials. In Fundamental Biomaterials: Polymers; Thomas, S., Balakrishnan, P., Sreekala, M.R., Eds.; Woodhead Publishing: Cambridge, UK, 2018; pp. 37–70. [Google Scholar]
  303. Tafuro, G.; Costantini, A.; Baratto, G.; Francescato, S.; Busata, L.; Semenzato, A. Characterization of polysaccharidic associations for cosmetic use: Rheology and texture analysis. Cosmetics 2021, 8, 62. [Google Scholar] [CrossRef]
  304. Zhu, B.; Ni, F.; Sun, Y.; Zhu, X.; Yin, H.; Yao, Z.; Du, Y. Insight into carrageenases: Major review of sources, category, property, purification method, structure, and applications. Crit. Rev. Biotechnol. 2018, 38, 1261–1276. [Google Scholar] [CrossRef]
  305. Vilela, A.; Cosme, F.; Pinto, T. Emulsions, Foams, and Suspensions: The Microscience of the Beverage Industry. Beverages 2018, 4, 25. [Google Scholar] [CrossRef] [Green Version]
  306. Nilforoushzadeh, M.A.; Amirkhani, M.A.; Zarrintaj, P.; Salehi Moghaddam, A.; Mehrabi, T.; Alavi, S.; Mollapour Sisakht, M. Skin care and rejuvenation by cosmeceutical facial mask. J. Cosmet. Dermatol. 2018, 17, 693–702. [Google Scholar] [CrossRef]
  307. Graham, S.; Marina, P.F.; Blencowe, A. Thermoresponsive polysaccharides and their thermoreversible physical hydrogel networks. Carbohydr. Polym. 2019, 207, 143–159. [Google Scholar] [CrossRef] [PubMed]
  308. Choi, J.; Moon, W.S.; Choi, J.N.; Do, K.H.; Moon, S.H.; Cho, K.K.; Han, C.J.; Choi, I.S. Effects of seaweed Laminaria japonica extracts on skin moisturizing activity in vivo. Int. J. Cosmet. Sci. 2013, 64, 193–205. [Google Scholar]
  309. Dolorosa, M.T.; Nurjanah; Purwaningsih, S.; Anwar, E.; Hidayat, T. Tyrosinase inhibitory activity of Sargassum plagyophyllum and Eucheuma cottonii methanol extracts. IOP Conf. Ser. Earth Environ. Sci. 2019, 278, 012020. [Google Scholar] [CrossRef]
  310. Cha, S.H.; Ko, S.C.; Kim, D.; Jeon, Y.J. Screening of marine algae for potential tyrosinase inhibitor: Those inhibitors reduced tyrosinase activity and melanin synthesis in zebrafish. J. Dermatol. 2011, 38, 354–363. [Google Scholar] [CrossRef] [PubMed]
  311. Chan, Y.Y.; Kim, K.H.; Cheah, S.H. Inhibitory effects of Sargassum polycystum on tyrosinase activity and melanin formation in B16F10 murine melanoma cells. J. Ethnopharmacol. 2011, 137, 1183–1188. [Google Scholar] [CrossRef]
  312. Park, J.; Lee, H.; Choi, S.; Pandey, L.K.; Depuydt, S.; De Saeger, J.; Park, J.-T.; Han, T. Extracts of red seaweed, Pyropia yezoensis, inhibit melanogenesis but stimulate collagen synthesis. J. Appl. Phycol. 2021, 33, 653–662. [Google Scholar] [CrossRef]
  313. Jesumani, V.; Du, H.; Pei, P.; Aslam, M.; Huang, N. Comparative study on skin protection activity of polyphenol-rich extract and polysaccharide-rich extract from Sargassum vachellianum. PLoS ONE 2020, 15, e0227308. [Google Scholar]
  314. Thu, N.T.H.; Anh, H.T.L.; Hien, H.T.M.; Ha, N.C.; Tam, L.T.; Khoi, T.X.; Duc, T.M.; Hong, D.D. Preparation and evaluation of cream mask from Vietnamese seaweeds. J. Cosmet. Sci. 2018, 69, 447–462. [Google Scholar] [PubMed]
  315. Pratama, G.; Yanuarti, R.; Ilhamdy, A.F.; Suhana, M.P. Formulation of sunscreen cream from Eucheuma cottonii and Kaempferia galanga (zingiberaceae). IOP Conf. Ser. Earth Environ. Sci. 2019, 278, 012062. [Google Scholar] [CrossRef]
  316. Poulose, N.; Sajayan, A.; Ravindran, A.; Sreechithra, T.V.; Vardhan, V.; Selvin, J.; Kiran, G.S. Photoprotective effect of nanomelanin-seaweed concentrate in formulated cosmetic cream: With improved antioxidant and wound healing properties. J. Photochem. Photobiol. B 2020, 205, 111816. [Google Scholar] [CrossRef] [PubMed]
  317. Pallela, R. Antioxidants from Marine Organisms and Skin Care. In Systems Biology of Free Radicals and Antioxidants; Laher, I., Ed.; Springer: Berlin/Heidelberg, Germany, 2014; pp. 3771–3783. [Google Scholar]
  318. Raikou, V.; Protopapa, E.; Kefala, V. Photo-protection from marine organisms. Rev. Clin. Pharmacol. Pharmacokinet. 2011, 25, 131–136. [Google Scholar]
  319. Riani Mansauda, K.L.; Anwar, E.; Nurhayati, T. Antioxidant and anti-collagenase activity of sargassum plagyophyllum extract as an anti-wrinkle cosmetic ingredient. Pharmacogn. Mag. 2018, 10, 932–936. [Google Scholar]
  320. Kasitowati, R.D.; Wahyudi, A.; Asmara, R.; Aliviyanti, D.; Iranawati, F.; Panjaitan, M.A.P.; Pratiwi, D.C.; Arsad, S. Identification photoprotective activity of marine seaweed: Eucheuma sp. IOP Conf. Ser. Earth Environ. Sci. 2021, 679, 012014. [Google Scholar] [CrossRef]
  321. Sami, F.J.; Soekamto, N.H.; Firdaus; Latip, J. Bioactivity profile of three types of seaweed as an antioxidant, uv-protection as sunscreen and their correlation activity. Food Res. 2021, 5, 441–447. [Google Scholar] [CrossRef]
  322. Pangestuti, R.; Siahaan, E.A.; Kim, S.-K. Photoprotective substances derived from marine algae. Mar. Drugs 2018, 16, 399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  323. Hameury, S.; Borderie, L.; Monneuse, J.-M.; Skorski, G.; Pradines, D. Prediction of skin anti-aging clinical benefits of an association of ingredients from marine and maritime origins: Ex vivo evaluation using a label-free quantitative proteomic and customized data processing approach. J. Cosmet. Dermatol. 2019, 18, 355–370. [Google Scholar] [CrossRef] [Green Version]
  324. Uji, Y.; Shibayama, J.; Shirakawa, Y. Oil-In-Water Type Emulsified Composition for External Skin Use. JPH0366281B2, 16 October 1991. [Google Scholar]
  325. Ando, H.; Ando, Y. Cosmetic Containing Agar Oligosaccharide and/or Its Esterified Substance. JP3223038B2, 29 October 2001. [Google Scholar]
  326. Kang, N.G.; Park, B.G. Solid-Type Cosmetic Composition for Moisturizing. KR20120019409A, 6 March 2012. [Google Scholar]
  327. Luo, Q.; Peng, Y. Powder-Containing Oil-in-Water Type Solid-State Cosmetic Composition and Preparation Method and Application Thereof. CN110664628A, 10 January 2020. [Google Scholar]
  328. Kawagishi, F.; Yamada, K.; Yokota, S. Pack Cosmetic. JPH11302124A, 2 November 1999. [Google Scholar]
  329. Hasunuma, K.; Saito, M. Powdery Pack Cosmetic. JPH08217631A, 27 August 1996. [Google Scholar]
  330. Nakagaki, E.; Suzuki, T. Hair Cosmetic. JP2779926B2, 23 July 1998. [Google Scholar]
  331. Sawaki, S.; Yamada, K. Water-Soluble Powdery Cosmetic. JP2889922B2, 10 May 1999. [Google Scholar]
  332. Pedroso De Oliveira, A.P. A Base Composition for Preparing Multi-Functional Formulations for Skin Care and Process for the Preparation Thereof. MXPA04009861A, 18 April 2005. [Google Scholar]
  333. Igarashi, Y.; Kobayashi, M.; Oka, S.; Takagaki, K. Scalp and Hair Cosmetic. JP2015030670A, 16 February 2015. [Google Scholar]
  334. Bi, L.; Pan, S.; Shao, X.; Sui, H.; Zhao, L.; Zou, P. Nano-Liposome Emulsion and Preparation Method Thereof. CN103876982A, 25 June 2014. [Google Scholar]
  335. Billiotte, J.-C.; Dampeirou, C. Cosmetic Skin Firming Compsn. For Combating Effects of Stress and Ageing. CH686997A5, 30 August 1996. [Google Scholar]
  336. Kim, K.B.; Ko, H.J.; Lee, D.H.; Lee, G.S.; Pyo, H.B. Ulva Spp Seaweed Hydrolysates That Have High Glucuronic Acid Cotent, Preparation Method Thereof and Antiaging Cosmetic Composition Containing the Same. KR101356535B1, 29 January 2014. [Google Scholar]
  337. Kong, Q.; Zhou, L. Anti-Aging Cosmetic Containing Seaweed Extract and Preparation Method Thereof. CN105030587A, 11 November 2015. [Google Scholar]
  338. Yao, Z. Marine Bioactive Cosmetic. CN112426381A, 2 March 2021. [Google Scholar]
  339. Yao, Z. Marine Bioactive Cosmetic. CN112451429A, 9 March 2021. [Google Scholar]
  340. Yang, H. Seaweed-Containing Cosmetic Additive and Preparation Method and Application Thereof. CN107669588A, 9 February 2018. [Google Scholar]
  341. Min, G.H. Functional Cosmetic Composition for Anti-Dust and Anti-Inflammatory Containing Natural Extract as an Effective Ingredient and Functional Cosmetic Including the Same. KR101988489B1, 12 June 2019. [Google Scholar]
  342. Kawashima, Y.; Uchibori, T. Antimicrobial Agent. JP2879590B2, 5 April 1999. [Google Scholar]
  343. Andre, G.; Pellegrini, L.; Pellegrini, M. Use of Undaria Pinnatifida Seaweed Extract in Cosmetic or Dermatological Compositions for Protecting the Skin and Visible Organs from the Harmful Effects of Oxygen Radicals and Atmospheric Pollution. FR2837383A1, 26 September 2003. [Google Scholar]
  344. Gwon, S.B.; Jo, J.H.; Joo, W.H.; Kim, E.H.; Lee, J.H.; Lee, J.H.; Park, S.H.; Ryu, G.Y. Antioxidant and Whitening Functional Cosmetics. KR101351187B1, 14 January 2014. [Google Scholar]
  345. Imada, K.; Mitsui, Y. Anti-Perspiration and Deodorization Cosmetic, and Method for Producing the Same. JP2008184395A, 14 August 2008. [Google Scholar]
  346. Park, D.I. Cosmetic Composition that Is Effective against Wrinkles. KR20120119797A, 31 October 2012. [Google Scholar]
  347. Hagino, H.; Saito, M. Cosmetics. US2004131580A1, 8 July 2004. [Google Scholar]
  348. Subuchi, H. Hair Shampoo, Hair Treatment, Hair-Growing Agent and Cosmetic Cream. JP2006008599A, 12 January 2006. [Google Scholar]
  349. Holtkoetter, O.; Scheunemann, V.; Schulze Zur Wiesche, E. Hair Treatment Agent. EP2457556A2, 23 November 2011. [Google Scholar]
  350. Aono, M.; Yamaoka, Y. Scalp and Hair Cosmetic. JP2012184220A, 27 September 2012. [Google Scholar]
  351. Cha, Y.J.; Hong, Y.C.; Hong, Y.K.; Kim, Y.J. Cosmetic Manufacturing Method Using Natural Originated Material and Cosmetic Thereof. KR20150063336A, 9 June 2015. [Google Scholar]
  352. Cheng, G.; Cheng, J. Nano-Element Shampoo. CN104739747A, 1 July 2015. [Google Scholar]
  353. Kim, K.E. Kelp, Deep Cleansing, Shampoo, Cosmetics. KR20110013849A, 10 February 2011. [Google Scholar]
  354. Egawa, M.; Nakamura, R. Skin Care Method. JP2015043973A, 12 March 2015. [Google Scholar]
  355. Courtin, O. Cosmetic Composition for Combating the Adverse Effects of Agents in the Atmosphere. FR2688137A1, 10 September 1993. [Google Scholar]
  356. Ishii, K. Combined Cosmetic. JP2011032200A, 17 February 2011. [Google Scholar]
  357. Wu, X. Cosmetic Hand Film. CN202682386U, 23 January 2013. [Google Scholar]
  358. Han, J.; Zhao, Y. Moisturizing and Oil-Controlling Cosmetic Composition and Preparation Method of Composition. CN111407684A, 14 July 2020. [Google Scholar]
  359. Bi, L.; Mou, W.; Sui, H.; Yang, J.; Yu, J.; Zhao, L.; Zou, P. Seaweed Composition with Oil-Control Function and Cosmetic Thereof. CN108904340A, 30 November 2018. [Google Scholar]
  360. Li, Y. Liquid Cosmetic Formula and Preparation Method Thereof. CN112022768A, 4 December 2020. [Google Scholar]
  361. Qian, F. Oil-Control Acne-Preventive Cosmetic with Pure Herbal Essence and Preparation Method Thereof. CN111888287A, 6 November 2020. [Google Scholar]
  362. Chen, J.; Jin, Z.; Ouyang, Z.; Quan, C.; Yu, G.; Zhang, F. Preparing Method of Traditional Chinese Medicine Anti-Acne Cosmetic. CN108434039A, 24 August 2018. [Google Scholar]
  363. Kim, D.S. Cosmetic Composition Comprising the Ozonized Oil and Femented Extract of Seaweed. KR20180013659A, 7 February 2018. [Google Scholar]
  364. Hua, C.; Huili, S.; Xin, C.; Zhigang, C. Marine Biological Function Cosmetic for Minimizing Pores. CN102178636A, 14 September 2011. [Google Scholar]
  365. Chen, Y.; Yang, S.; Zhou, Z. Production Process of Moisturizing Lotion with Pore Cleaning Function. CN111991324A, 28 August 2020. [Google Scholar]
  366. Nakamura, R. Pore-Shrinking Agent. JP2015030675A, 16 February 2015. [Google Scholar]
  367. Chun, J.U. Composition Having Anti-Wrinkle Effects Using the Natural Plant Extract and Cosmetic Composition Comprising the Same. KR101952695B1, 27 February 2019. [Google Scholar]
  368. Wang, J. Making Method of Anti-Wrinkle Beautifying Seaweed-Containing Flour. CN105685784A, 22 June 2016. [Google Scholar]
  369. Yu, K. Anti-Aging Composition and Cosmetic Product Containing Same. CN109431888A, 8 March 2019. [Google Scholar]
  370. Chen, H.; Chen, L.; Ge, X.; Wang, Y.; Yue, K. Composition Capable of Resisting Aging and Fading Spots and Cosmetic Containing Composition. CN108670927A, 19 October 2018. [Google Scholar]
  371. Osawa, Y.; Sawaki, S.; Tamaoki, S. Elastin-Like Agent and Cosmetic Containing the Same. JP2003342150A, 3 December 2003. [Google Scholar]
  372. Deng, Y. Vitamin B (VB) Face Cream Cosmetic Containing 24K Gold. CN102824285A, 19 December 2012. [Google Scholar]
  373. Gerčikovs, I.; Lando, O. Cosmetic Preparation for Taking Care of the Feet Skin. LV14321A, 20 April 2011. [Google Scholar]
  374. Gedouin, A.; Vallee, R. Production of Cosmetic Composition for Protecting Skin from Effects of Atmospheric Pollution. FR2779953A1, 24 December 1999. [Google Scholar]
  375. Hori, M.; Nishibe, Y.; Tanaka, K. Cosmetic Composition. JP2003104835A, 9 April 2003. [Google Scholar]
  376. Yang, Z. Seaweed Extraction Liquid, Preparation Method of Seaweed Extraction Liquid, Whitening Composition and Preparation Method of Whitening Composition. CN108324601A, 27 July 2018. [Google Scholar]
  377. Li, Y.; Pei, Q.; Shi, C.; Shi, H.; Shi, Z. Pearl Cosmetic Cellular Liquor and Preparation Method Thereof. CN106265463A, 4 January 2017. [Google Scholar]
  378. Choi, C.Y.; Choi, S.J.; Im, S.J.; Jeong, C.S.; Jeong, J.C.; Jo, A.; Kang, H.W.; Kim, H.G.; Kim, J.Y.; Kim, J.Y.; et al. Pharmaceutical Composition and Cosmetic Composition for Skin Regeneration Comprising Active Ingredient Extracted and Isolated from Porphyra Dentata. WO2020175754A1, 3 September 2020. [Google Scholar]
  379. Yang, G.; Zhao, J.; Zhao, Y. Facial Beautifying Composition and Preparation Method Thereof. CN111544369A, 18 August 2020. [Google Scholar]
  380. Takabayashi, M. Anti-Suntan Cosmetic and its Production. JP3568979B2, 22 September 2004. [Google Scholar]
  381. Paik, H.K. Cosmetic Compositions Containing Natural Ultraviolet Intercepting Agent Based Seaweed from Jeju Island. KR20140089997A, 16 July 2014. [Google Scholar]
  382. Han, J.S.; Lee, E.J.; Lee, J.W.; Oh, J.Y.; Park, S.G. Sunscreen Cosmetic Composition. KR20170004842A, 11 January 2017. [Google Scholar]
  383. Murakami, M.; Ota, K.; Saito, M.; Sumida, Y. Weight-Reducing Composition and Method for Reducing Weight. JPH08104618A, 23 April 1996. [Google Scholar]
  384. Azuma, T.; Hayashi, Y.; Ishihata, S.; Kuroda, A.; Sakai, K.; Sato, S. Sheetlike Pack Cosmetic for Body. JP2001019615A, 23 January 2001. [Google Scholar]
  385. Takabayashi, M. Beautifying and Whitening Cosmetic. JP2970767B2, 2 November 1999. [Google Scholar]
  386. Kawai, N.; Tanaka, K.; Wakamatsu, K. Cosmetic Composition. JP2001139419A, 22 May 2001. [Google Scholar]
  387. Kawai, N.; Tanaka, K.; Wakamatsu, K. Cosmetic Composition. JP2001302491A, 31 October 2001. [Google Scholar]
  388. Adachi, K.; Kotake, Y.; Suzuki, Y. Preparation for External Use for Skin. JP3460904B2, 27 October 2003. [Google Scholar]
  389. Kang, S.W.; Kim, E.J.; Kim, S.J. Skin Moisturizing and Funtional Composition Containing Fermented Seeweed. KR20190029897A, 21 March 2019. [Google Scholar]
  390. Han, S.H.; Hong, Y.J.; Kim, H.C.; Kim, Y.J. Cosmetic Composition Containing Gulfweed Extract, Sea Staghorn Extract, and Brown Seaweed Extract. WO2012070835A2, 31 May 2012. [Google Scholar]
  391. Ahn, D.H.; Kim, J.Y.; Moon, J.N.; Moon, W.S. Cosmetic Composition Containing Seaweed Extract. WO2015099280A1, 2 July 2015. [Google Scholar]
  392. Zhou, Y. Cosmetic Preparation with Anti-Allergic and Repairing Effects and Preparation Method Thereof. CN108578353A, 28 September 2018. [Google Scholar]
  393. Kang, D.H.; Kim, M.G.; Lee, B.H. Beauty Expenses Composite Containing Seaweed Extract. KR20150022365A, 4 March 2015. [Google Scholar]
  394. Cong, L.; Gao, H.; Li, C.; Lin, S.; Liu, D.; Liu, P.; Mao, Y.; Zhang, C.; Zhang, W. Sunscreen Cosmetic Composition and Method for Preparing Seaweed Sunscreen Components of Composition. CN104644511A, 27 May 2015. [Google Scholar]
  395. Cai, C.; Li, Y.; Wang, H. Facial Mask for Removing Freckles, Whitening and Relieving Sunburn and Preparation Method of Facial Mask. CN106619272A, 10 May 2017. [Google Scholar]
  396. Kawakubo, A.; Kouno, S.; Matsuka, S.; Motoyoshi, K.; Ninomiya, M.; Ota, Y. Photo-Aging Resister and Skin Cosmetic Containing the Resister. JP3432033B2, 28 July 2003. [Google Scholar]
  397. Gan, X.; Zhang, K. Eye-Beautifying Firming Eye Cream Cosmetic and Preparation Method Thereof. CN109875933A, 14 June 2019. [Google Scholar]
  398. Choi, J.B.; Jo, J.H.; Joo, W.H.; Kim, E.H.; Lee, J.H.; Lee, J.H.; Ryu, G.Y. Antioxidant And Whitening Functional Cosmetics. KR101413328B1, 1 July 2014. [Google Scholar]
  399. Li, K.; Liang, Y.; Wang, T.; Yue, Q.; Zhao, L. Method for Preparing Seaweed Fermentation Solution by Virtue of Probiotics Fermentation and Application of Seaweed Fermentation Solution in Cosmetics. CN108653059A, 16 October 2018. [Google Scholar]
  400. Choi, B.S.; Choi, J.H.; Chun, H.S.; Kim, S.; Kim, S.J.; Lee, H.J.; Park, E.T.; Park, S.E. Food Composition Comprising Algae Extracts and the Use Thereof. KR20130054518A, 27 May 2013. [Google Scholar]
  401. Hirose, K.; Hirose, Y. Composition Having Bleaching Activity and Cosmetic Containing the Same. JP2006036680A, 9 February 2006. [Google Scholar]
  402. Hirose, K.; Hirose, Y. Composition Having Hair Loss Preventing Action and Hair Loss Preventing Cosmetic Containing the Same. JP2006052151A, 23 February 2006. [Google Scholar]
  403. Zhang, R. Pure Natural Moisturizing Cosmetic. CN103690407A, 2 April 2014. [Google Scholar]
  404. Park, J.W. Cosmetic Composition for Spa Comprising Natural Mixture as Effective Component. KR20180024141A, 8 March 2018. [Google Scholar]
  405. Ku, W.L.; Lee, H.J. Cosmetic Composition Using Pine Tree and Seaweeds and Manufacturing Method of it. KR20200122677A, 28 October 2020. [Google Scholar]
  406. Park, S.L. Cosmetic Composition for Anti-Wrinkle Activity Comprising Fermented Soybean Paste and Seaweed Extracts as Active Ingredient. KR20200080500A, 7 July 2020. [Google Scholar]
  407. Oouma, T.; Takekoshi, Y.; Takahashi, T. Cosmetics. US2002009472A1, 24 January 2002. [Google Scholar]
  408. Choi, K.J. Cosmetics Composition. KR20090126670A, 9 December 2009. [Google Scholar]
  409. Guo, Z. Cosmetic Containing Active Peptide. CN105982831A, 5 October 2016. [Google Scholar]
  410. Qiu, H.Y.; BI, H.Y.; Chen, X.; Chen, W.Y.; Du, L.N.; Jin, Y.G. Curcumin Hydrogel Combined with Photodynamics to Treat Acne. J. Int. Pharm. Res. 2017, 44, 1125–1130. [Google Scholar]
  411. Cho, J.H. Cosmetic Composition Contained Seaweeds Extract with Silver Nano-Particles Colloid. KR20080035090A, 23 April 2008. [Google Scholar]
  412. Chen, J.; Chen, J. Anti-Inflammatory Sedation Patch Mask. CN112402339A, 26 February 2021. [Google Scholar]
  413. Kim, J.E.; Yang, Y.J. Mist Containing Seaweed Extract. KR20180080058A, 11 July 2018. [Google Scholar]
  414. Fukuhara, K.; Kamiyama, K. Hair Cosmetic. JP2005272396A, 6 October 2005. [Google Scholar]
  415. Kim, K.E. Kelp Shampco, Pock, Cosmetics. KR20110006337A, 20 January 2011. [Google Scholar]
  416. Chen, F.; Li, Y.; Wang, X. Washing Cosmetic Containing Seaweed Extract-Carrageen. CN104434696A, 25 March 2015. [Google Scholar]
  417. Sato, Y.; Sigihara, Y. Cosmetic Composition, Screening Method, and Cosmetic Method. WO2019098352A1, 23 May 2019. [Google Scholar]
  418. Huang, J.; Li, C.; Lin, S.; Liu, D.; Ma, J.; Qian, J.; Xiang, Q. Water-Free Washing-Free Gold Seaweed Cleaning Oil. CN109431878A, 8 March 2019. [Google Scholar]
  419. Chen, H.C. Diet Cosmetic Food and Its Production. CN101028075A, 5 September 2007. [Google Scholar]
  420. Takahashi, N. Production of Cosmetic or Bathing Agent. JPS62286907A, 12 December 1987. [Google Scholar]
  421. Hashimoto, H.; Hiraki, Y.; Ichioka, M.; Inami, M.; Kamiyama, S.; Kono, H.; Nagaoka, M.; Yoshikawa, S. Highly Pure Fucoidan and Preparation Thereof. JP2000351801A, 10 October 2000. [Google Scholar]
  422. Kusuoku, H.; Nishizawa, Y.; Shibuya, Y. Purification of Seaweed Extract. JP2000109407A, 18 April 2000. [Google Scholar]
  423. Kusuoku, H.; Nishizawa, Y.; Shibuya, Y. Method for Purifying Seaweed Extract. JP2005145983A, 9 June 2005. [Google Scholar]
Figure 1. Cosmeceutical potential of algae components.
Figure 1. Cosmeceutical potential of algae components.
Marinedrugs 19 00552 g001
Table 1. Some activities and properties of seaweeds polysaccharides of interest in cosmeceutical formulations.
Table 1. Some activities and properties of seaweeds polysaccharides of interest in cosmeceutical formulations.
ComponentProperties/ActivitiesSeaweedReferences
AgarThickener; antioxidantPterocladia, Pterocladiella, Gelidium amansii, Gracilaria[28,46,53,54,55]
AlginateHigh stability, thickening agent, gelling agentBrown seaweeds[34,56,57]
CarrageenansAntioxidant, antitumor, antiaging, thickeners properties, radiation protectionRed seaweeds, Porphyra haitanensis, Gracilaria chouae, Gracilaria blodgettii[16,49,58,59,60,61,62]
FucoidansPhotoaging inhibition; minimized elastase activity; antioxidant, anti-inflammatory collagenase and elastase inhibition, skin-whiteningFucoidan (Sigma), Ascophyllum nodosum, Chnoospora minima, Ecklonia maxima, Hizikia fusiforme, Saccharina japonica, Sargassum hemiphyllum, Sargassum horneri, Sargassum polycystum, Sargassum vachellianum[2,23,24,43,44,46,48,63,64,65,66,67]
LaminaranReconstructed dermis; skin cell anti-inflammation; antioxidantSaccharina longicruris, Laminarin (Sigma)[68,69]
PolysaccharidesHydrationSaccharina japonica, Chondrus crispus, Codium tomentosum[28]
UlvanAntiaging, antiherpeticUlva pertusa, Ulva sp.[51,70]
Table 2. Some activities and properties of seaweeds protein, peptides, and amino acids of interest in the cosmeceutical formulations.
Table 2. Some activities and properties of seaweeds protein, peptides, and amino acids of interest in the cosmeceutical formulations.
Extract/CompoundActivitySeaweedReference
Eleven mycosporine-like amino acidsUV-protective effect, antioxidantAgarophyton chilense, Pyropia plicata and Champia novae-zelandiae[147]
Mycosporine-like amino acids extract (with porphyra-334 and shinorine in a ratio of 2:1)Anti-agingPhorphyra umbilicalis[151]
Mycosporine-like amino acids extract (mainly palythine and asterina-330)Antioxidant, UV-protective effect, anti-agingCurdieara covitzae, Iridaea cordata[152]
Mycosporine-like amino acids extract (mainly porphyra-334, shinorine, palythine and asterina-330)Antioxidant; UV-protective effectGracilaria vermiculophylla[153]
Mycosporine-like amino acids extract (mainly palythine, asterina-330, shinorine, palythinol, porphyra-334 and usujirene)Antioxidant, antiproliferativeChondrus crispus, Mastocarpus stellatus, Palmaria palmata[154]
Mycosporine-like amino acids extract (mainly deoxygadusol, palythene and usujirene)AntioxidantRhodymenia pseudopalmata[155]
Aqueous extract from freshwater macroalga (mainly polysaccharides and amino acids)Skin moisturizing effectRhizoclonium hieroglyphicum[156]
Peptide PPY1Anti-inflammatoryPyropia yezoensis[134]
Peptides PYP1-5 and porphyra 334 Increase production of elastin and collagenPorphyra yezoensis f. coreana Ueda[135]
Methanol extract rich in proteins, vitamins, minerals, porphyra-334 and shinorineHydration, skin protective, anti-wrinkle, anti-roughnessPhorphyra umbilicalis[148]
Phycobiliproteins (R-phycoerythrin allophycocyanin and phycocyanin)AntioxidantGracilaria gracilis[157]
Hydrolyzed extractAntitumorPorphyra haitanesis[158]
Algae extractDecrease of progerin production, anti-elastase, anti-collagenaseAlaria esculenta[159]
Table 3. Examples of recent studies confirming the phlorotannin activities of interest for cosmeceutical products.
Table 3. Examples of recent studies confirming the phlorotannin activities of interest for cosmeceutical products.
CompoundActivitySeaweedReferences
Dioxinodehydroeckol Preventive activity against UVB-induced apoptosisEcklonia cava[190]
DieckolAdipogenesis inhibitory effectEcklonia cava[197]
EckolAnti-inflammatory, anti-tyrosinaseEisenia bicyclis, Ecklonia stolonifera[192,193,198]
Eckol, 6,6′-bieckol, 8,8′-bieckol, dieckol, and phlorofucofuroeckol-AAntiallergicEcklonia cava, E. stolonifera[179]
Fucofuroeckol-AProtective against UVBEcklonia stolonifera Okamura[191]
FuhalolAntioxidantCystoseira compressa[175]
Fucophloroethol (isomer)AntioxidantFucus vesiculosus[199]
EckstolonolAntioxidant enzymatic activities of catalase and superoxide dismutase Ecklonia cava[200]
Octaphlorethol AAntioxidative effectsIshige foliacea[201]
Phlorofucofuroeckol AHepatoprotective effect against oxidative stressEisenia bicyclis[93]
Tyrosinase inhibitory activityEcklonia stolonifera[182]
2-phloroeckol and 2-O-(2,4,6-trihydroxyphenyl)-6,60-bieckolTyrosinase inhibitory activityEcklonia cava[185]
Phlorofucofuroeckol BAntiallergicEisenia arborea[202]
PhlorotanninsAntioxidant, anticoagulant, antiinflammatory, antibacterial, antiviral, antitumor; antidiabetic, photoprotectiveBrown algae, Ascophyllum nodosum, Fucus serratus, Himanthalia elongata, Halidrys siliquosa[45,172,175,198,203,204]
Table 4. Activities of seaweed lipids of interest in the formulation of cosmeceuticals.
Table 4. Activities of seaweed lipids of interest in the formulation of cosmeceuticals.
CompoundActivitySeaweedReferences
E-10-oxooctadec-8-enoic acid, E-9-oxooctadec-10- enoic acidAnti-inflammatoryGracilaria verrucosa[226]
Essential oil (tetradeconoic acid, hexadecanoic acid, (9Z, 12Z)-9,12-octadecadienoic acid, (9Z)-hexadec-9-enoic acid)Antibacterial activity against Staphylococcus aureus and Bacillus cereus
Antioxidant: radical scavenging (DPPH, superoxide, ABTS)
Laminaria japonica[227]
FucosterolAntioxidant: increased antioxidative enzymes (superoxide dismutase, catalase, glutathione peroxidase)Pelvetia siliquosa[219,228]
FucosterolAnti-photodamage: decreased UVB-induced MMPs and increased procollagen
Anti-inflammatory
Hizikia fusiformis[60,188]
PhytosterolAntitumoralCommercial (Sigma)[229]
Lipidic profileAntioxidant, enzyme inhibitionUlva rigida, Gracilaria sp., Fucus vesiculosus, Saccharina latissima[211]
Unsaturated fatty acidsAntioxidantBrown algae[230]
Fatty acid profilingBioindicator of chemical stressPterocladia capillacea, Sargassum hornschuchii, Ulva lactuca[231]
Table 5. Minimum and maximum values (g/100 g or mg/kg dry weight) for macro and micro elements found in edible European macroalgae [234,254].
Table 5. Minimum and maximum values (g/100 g or mg/kg dry weight) for macro and micro elements found in edible European macroalgae [234,254].
Element (Concentration)Brown AlgaeGreen AlgaeRed Algae
Ca (%)0.89–1.320.21–1.870.39–45.0
Mg (%)0.22–1.20.12–2.80.20–167
P (%)0.15–0.980.21–5000.10–1.40
K (%)3.8–11.51.1–8.10.33–10.2
Na (%)1.3–7.10.52–8.91.1–4.3
S (%)1.33–1.50.23–8.51.5–4.0
Cu (ppm)1.1–11.01.6–12.1<0.4–35.0
I (ppm)0.20–50020–10000.24–1200
Fe (ppm)15.8–27017.7–289016–1820
Mn (ppm)<1–52.7<2–347<1–748
Zn (ppm)2.5–52.31.98–847.2–714.4
Table 6. Biological activities of algal pigments of interest in cosmeceuticals.
Table 6. Biological activities of algal pigments of interest in cosmeceuticals.
Extract/CompoundActivitySeaweedReferences
97% fucoxanthin extractAntioxidant (DPPH scavenging capacity, reducing power)Himanthalia elongata[265]
FucoxanthinAntioxidant, anti-melanogenesisBrown seaweeds[266,267]
AntiobesityUndaria pinnatifida[263]
Skin protective (antiphotodamage, anti-pigmentary, antiphotoaging, anti-wrinklingSargassum siliquastrum[268]
Anti-inflammatoryMyagropsis myagroides[269]
Tyrosinase activityLaminaria japonica[266]
PhotoprotectiveUndaria pinnatifida[270]
AntioxidantSargassum fusiforme,[271]
LuteinWhitening; visual disorders and cognition diseasesRhodophyta spp.[28,272]
Table 7. Examples of patents claiming the use of seaweed and seaweed components in cosmetic, cosmeceutical, and nutricosmetics formulations to confer different properties.
Table 7. Examples of patents claiming the use of seaweed and seaweed components in cosmetic, cosmeceutical, and nutricosmetics formulations to confer different properties.
ActivityApplicant CompanyReferences
Functional and sensorial
Emulsifying, water retention, gellingAsahi Denka Kogyo Kk; Health Care Ltd.; Ichimaru Pharcos Inc; Iwasekenjiro Shoten Kk; Lg Household & Amp Lvxinyan Guangdong Bio Tech Co. Ltd.[324,325,326,327]
Film formingKowa Techno Search Kk[328]
Improved water solubility and imparting excellent feeling of useArtnature Co. Ltd.; Kanebo Ltd.; Koosee Kk; Kyoei Kagaku Kogyo Kk; Natura Cosmeticos Sa; Toyo Shinyaku Co. Ltd.;[329,330,331,332,333]
Stabilization systemYantai New Era Health Industry Daily Chemical Co. Ltd.[334]
Biological
Anti-aging and antistressGivenchy Parfums; Hanbul Cosmetics Co. Ltd.; Hainan Hairun Biolog Technology Co. Ltd.
Shengfeng Yantai Agricultural Tech Co. Ltd.
[335,336,337,338,339]
Anti-inflammatoryExplzn Inc; Yantai Yucheng Enterprise Man Consulting Co. Ltd.[340,341]
AntimicrobialNippon Enu Yuu Esu Kk[342]
AntioxidantGelyma; Jeollanamdo[343,344]
Antiperspirant, desodorantJapan Natural Lab Co. Ltd.[345]
Anti-wrinkleMamachi Co. Ltd.[346]
Bood circulationKowa Techno Search Kk[328]
Hair and scalp care and treatment, hair growthClean Sea Co. Ltd.; Henkel Ag & Co Kgaa; Kose Corp; Nantong Snakebite Therapy Res Inst; Pinebio Co. Ltd.; Sako Kk; Shirako Co. Ltd.; Lion Corp[347,348,349,350,351,352,353,354]
MoisturizingAmazonebio Co. Ltd.; Clarins; Jingmen Nuoweiying New Material Tech Co. Ltd.; Kracie Home Products Ltd.; Qingdao Better Biolog Science & Technology Co. Ltd.[308,355,356,357,358]
Oil control, acne prevention and removal of acne marksYantai New Era Health Ind Daily Chemical Co. Ltd.; Guangzhou Yuanmeisheng Cosmetic Co. Ltd.; Shanghai Bonaquan Cosmetics Co. Ltd.; Suzhou Cosmetic Materials Co. Ltd.; Tubio; Yantai New Era Health Ind Daily Chemical Co. Ltd.[359,360,361,362,363]
Pore shrinking, cleaning and minimizingFoshan Aai Cosmetic Health Care Product Co. Ltd.; South China Sea Inst Oceanology; Rongding Guangdong Biotechnology Co. Ltd.; Kose Corp[364,365,366]
Prevention and amelioration of aged and rough skinAnhui Shuanglu Flour Co. Ltd.; Dzintars As; Explzn Inc; Guangzhou Saliai Stemcell Science & Technology Co. Ltd.; Kyoei Chemical Ind; Nox Bellcow Zs Nonwoven Chemical Ltd.; Shanghai Bonaquan Cosmetics Co. Ltd.; Wuhu Chuanshi Information Tech Co. Ltd.[360,367,368,369,370,371,372,373]
Protecting from pollutionCodif International Sa[374]
Safe melanin production and whitenningIchimaru Pharcos Inc; Shenzhen Sanda Cosmetics Co. Ltd.[375,376]
Skin regeneration and epidermal cell repairBeihai Yuanlong Pearl Company Ltd.; Jeonnam Bioindustry Found; Hexie Tech Co. Ltd.; Mokpo Marin Food Industry Res Center; Yantai New Era Health Industry Daily Chemical Co. Ltd.[334,377,378,379]
Sunscreen and anti-sun tanLg Household & Health Care Ltd.; Mikimoto Seiyaku Kk; Miin[380,381,382]
Weight-reduction and slimmingKanebo Ltd.; Sekisui Plastics[383,384]
WhiteningIchimaru Pharcos Inc; Lion Corp; Mikimoto Seiyaku Kk; World Costec Co. Ltd.[385,386,387,388,389]
Mixed Effects, More Than One Of The Following Actions
Antiaging, anti-allergic, anti-inflammatory, antioxidant, anti-wrinkle; cleaning, moisturizing, repairing, sunscreen, whitenningAmorepacific Corp; Baiyun Lianjia Fine Chemical Factory; Ecomine Co. Ltd.; Foshan Chancheng Relakongjian Biotechnology Co. Ltd.; Guangdong Danz Group Co. Ltd.; Guangzhou Baiyun Lianjia Fine Chemical Factory; Guangzhou Keneng Cosmetic Res Co. Ltd.; Guangzhou Xibao Daily Chemical Co. Ltd.; Hainan Shiboli Biotechnology Co. Ltd.; I2b Co. Ltd.; Jeollanamdo; Kaiso Shigen Kenkyusho Kk; Pola Chem Ind Inc[344,390,391,392,393,394,395,396,397,398]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

López-Hortas, L.; Flórez-Fernández, N.; Torres, M.D.; Ferreira-Anta, T.; Casas, M.P.; Balboa, E.M.; Falqué, E.; Domínguez, H. Applying Seaweed Compounds in Cosmetics, Cosmeceuticals and Nutricosmetics. Mar. Drugs 2021, 19, 552. https://0-doi-org.brum.beds.ac.uk/10.3390/md19100552

AMA Style

López-Hortas L, Flórez-Fernández N, Torres MD, Ferreira-Anta T, Casas MP, Balboa EM, Falqué E, Domínguez H. Applying Seaweed Compounds in Cosmetics, Cosmeceuticals and Nutricosmetics. Marine Drugs. 2021; 19(10):552. https://0-doi-org.brum.beds.ac.uk/10.3390/md19100552

Chicago/Turabian Style

López-Hortas, Lucía, Noelia Flórez-Fernández, Maria D. Torres, Tania Ferreira-Anta, María P. Casas, Elena M. Balboa, Elena Falqué, and Herminia Domínguez. 2021. "Applying Seaweed Compounds in Cosmetics, Cosmeceuticals and Nutricosmetics" Marine Drugs 19, no. 10: 552. https://0-doi-org.brum.beds.ac.uk/10.3390/md19100552

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