Algae Metabolites in Cosmeceutical: Comparison
Please note this is a comparison between Version 2 by Peter Tang and Version 1 by Yoon-Yen Yow.

Cosmeceuticals are topical cosmetic-pharmaceutical hybrids which refer to a cosmetic product with active ingredients claiming to have medicinal or drug-like benefits to skin health. Marine algae are rich in bioactive substances that have shown to exhibit strong benefits to the skin, particularly in overcoming rashes, pigmentation, aging, and cancer. 

  • marine algae
  • cosmeceuticals
  • UV-radiation
  • anti-aging
  • anticancer
  • skin whitening

1. Introduction

1.1. Synthetic Versus Natural Ingredients in Cosmetic Industry

Cosmeceuticals are topical cosmetic-pharmaceutical hybrids which refer to a cosmetic product with active ingredients claiming to have medicinal or drug-like benefits to skin health [1,2][1][2]. Globally, the cosmeceutical industry is growing each year due to the trend of modern lifestyle. More recently, the cosmeceutical industry is progressively shifting to natural bioactive ingredients because of the ineffectiveness of synthetic cosmetics [3].
Ineffectiveness of synthetic cosmetics includes their side effects and low absorption rate. The low absorption rate of cosmetics could be due to the big size of the molecular compounds. A study by Bos and Marcus [4] asserted that only compounds with the molecular weight lesser than 500 Dalton (Da) could penetrate through the skin. Cyclosporin (MW 1202 Da), a topical immunosuppressant, was not effective against psoriasis and allergic contact dermatitis as a higher molecular weight of the compounds inhibits skin penetration. Still, it was effective in psoriasis treatment when directly injected into the skin. Some of the side effects include irritation and allergic reaction to the users. According to a case study, hydroxybenzoic acid esters (parabens), which are widely used in cosmetic products, has been reported to mimic oestrogen; hence, increasing the incidence of breast cancer and causing the development of malignant melanoma [5].
In addition, a study on a population conducted by the Centers for Disease Control and Prevention reported that 97% of 2540 individuals were exposed to phthalates (a component of plastic that appears in cosmetic products; for instance, dibutyl phthalate in nail polish), which could result in DNA damage in human sperm [6]. In 2004, the Environment California, Environmental Working Group, and Friends of the Earth issued reports on cosmetic products containing chemical ingredients that lacked safety data. Some of these chemicals caused adverse effects in animal studies such as male genitalia congenital disabilities, altered pregnancy outcomes, and decreased in sperm counts [6]. As a result, consumers have changed their preference and opted for natural cosmetic products. The global market value for natural cosmetics was about $34.5 billion in 2018, and it is estimated to reach approximately $54.5 billion in 2027 [7]. The ever-expanding market for skincare products and continual search for innovative ingredients has led to the development of a multitude of cosmeceutical products based on natural bioactive ingredients, which include plants, herbs, and even marine algae [8].
Macroalgae are classified into three major classes, namely Phaeophyceae (brown algae), Rhodophyceae (red algae), and Chlorophyceae (green algae). Based on the total culture production, it is estimated that about 59% of brown algae, 40% of red algae, and less than 1% of green algae are produced worldwide [9]. Marine algae are rich sources of structurally diverse bioactive compounds, which are absent in other taxonomic groups. Algae contain 10 times greater diversity of compounds than terrestrial plants [10] and they have a totally different flavonoid composition from vegetables and fruits. Macroalgae are a rich source of catechins and flavonols [11]. Furthermore, algae-derived phlorotannin possesses a unique structure, which is not found in terrestrial plants and this compound may constitute up to 25% of the dry weight of brown algae [11]. Algae produce a wide array of primary metabolites, such as unsaturated fatty acids, polysaccharides, vitamins, and essential amino acids [12,13][12][13]. Additionally, many research findings reported that secondary metabolites derived from algae such as fucoidan, fucoxanthin, sulphated polysaccharide, polyphenol and fucosterol were shown to possess anti-inflammation, antioxidant, anticancer, antibacterial and anti-aging effects [14,15,16,17,18][14][15][16][17][18]. The demand for algae bioactive compounds in cosmeceuticals is rapidly increasing as they contain natural extracts which are considered safe; thus, rendering fewer side effects on humans. In ancient times, marine algae were used as medicine to treat skin-related diseases, such as atopic dermatitis and matrix metalloproteinase (MMP) related disease [12]. In a nutshell, marine algae are a promising resource for the development of cosmeceuticals.
Marine algae can survive in harsh conditions (i.e., withstand heat, cold, ultra-violet radiation, salinity, and desiccation) [8,9,19][8][9][19] due to their ability to adapt to physiological changes by producing stress tolerant substances. For example, algae produce organic osmolytes during stress conditions, which also act as antioxidants and heat protectants. Algae grow under desiccation by producing specialized spores which remain dormant during stress conditions and revive once the conditions return to normal. The presence of thick cell walls with protective layers of chemical substances and mucilage sheath helps to delay the process of desiccation. Algae that grow in cold desserts can endure the subzero temperature and protect the cells from UV irradiation by producing spores that have thick cell walls and reserve food as lipid and sugars [20]. In addition, marine algae uptake inorganic ions to balance extracellular ion concentration and produce organic osmolytes which protects them from desiccation and UV lights. A study reported that Dunaliella salina has 55 novel membrane-associated proteins that showed changes in the composition and structure of the membranes associated with algae adaptation to salinity [21]. Algae are rich in a wide variety of secondary metabolites to help them adapt and survive in harsh conditions. Algae could also adapt to desiccation stress by producing specialized spores such as aplanspores, which are rich in astaxanthin. Astaxanthin is a carotenoid that protects the cells from photo-oxidation. Algae exposed to UV radiations will produce UV screening compounds such as mycosporine-like amino acids (MAA), which acted as antioxidants and involved in osmotic regulations. Furthermore, algae exposed to high solar radiation and low nitrogen concentration produce more β-carotene, such as Dunaliella [20]. Thus, algae that are naturally exposed to oxidative stress develop defense systems that protect them against reactive oxygen species (ROS) and free radicals. These compounds could be used in cosmetics to protect the cells against the adverse effects of UV radiation. Some of the environmental benefits of algae include fixation of carbon dioxide. Studies have reported that large cultivation of microalgae capable of uptaking carbon from the atmosphere; for instance, Spirulina platensis with carbon fixation rate of 318 mg/L−1d−1 and Chlorella vulgaris with carbon fixation rate of 251 mg/L−1d−1 [22,23][22][23].

1.2. Current Applications of Algae-Derived Metabolites in Cosmeceutical Industrial

The transition from synthetic compounds to natural products such as marine algae have been attracting the attention of many researchers since algae possess a wide range of pharmacological activities with negligible cytotoxicity effects in human cells [24]. Marine algae are used for different purposes in food, pharmaceutical, biofuel, agriculture, and cosmetic industries. Industries, such as Cyanotech, Fuji Chemical Seambiotic, and Mera Pharmaceuticals are producers of microalgae biomass contributing to products in pharmaceuticals, cosmetics, and nutritious feed [25]. Interestingly, phycocyanin (usually found in red algae and cyanobacteria) is accepted as a natural color additive in food and cosmetics by the Food and Drug Administration (FDA) due to its non-toxic, natural, and biodegradable properties. Accordingly, it becomes the major target of the market in the United States [26].
Meanwhile, carotenoid such as astaxanthin plays a crucial role in scavenging free radicals in the human body and it is considered a strong antioxidant; hence, its popularity as a human dietary supplement. Leading cosmeceutical industries, such as Unilever, L’Oreal, Henkel, and Beiersdorf are expected to improve the growth of carotenoid market value in the European market [27]. The market value for carotenoids is expected to reach about $1.53 billion by 2021 [27,28][27][28].
Furthermore, red algae Gracilaria account for most of the raw material for the agar extraction. It is reported by the Food and Agriculture Organization (FAO) of the United Nations that more than 80% of the agar were produced from Gracilaria species, which are mainly produced by China and Indonesia [29]. Gracilaria species have been widely used in cosmetics due to their stabilizing, thickening, and gelling characters. Commercially available products from Gracilaria species include hydrogel soap by Sea Laria®, facial mask by Balinique®, and hydrating cream by Thalasso® [29].
A number of algae-based skin products have been marketed, such as Algenist (an anti-aging moisturizer containing microalgae oil and alguronic acid from algae) [30], Helionori® by Gelyma and Helioguard365® (a sunscreen product containing MAAs from red seaweed Porphyra umbilicalis) [31], Protulines® by Exsymol S.A.M., Monaco (an anti-aging agent from protein-rich extract of Arthrospira), and Dermochlorella by Codif, St. Malo, France (an anti-wrinkling agent from Chlorella vulgaris extract) [32]. Therefore, bioactive compounds derived from algae could be considered a potential cosmeceutical agent for skincare.

1.3. UV Radiation and Skin-Related Diseases

Skin is one of the most complex and largest organs that serves as a protective barrier against water losses and environmental stresses, such as ultraviolet radiation (UVR), pathogens, physical agents, and chemicals [33]. The skin comprises three layers—epidermis, dermis, and hypodermis. The presence of keratinocyte cells and melanocyte cells in the epidermis layer plays a vital role in repairing damaged skin and protecting the skin from UV light. The dermis consists of elastin, hyaluronic acid, and collagen which involves tissue repair and stability, whereas hypodermis consists of fats, which involved in body insulation [9]. Several skin-related diseases that have been reported include acne, eczema, dermatitis, hives, psoriasis, and pityriasis rosea which cause rashes [34]. Other skin diseases include pigmentation disorders, such as hypopigmentation due to the absence of melanocytes and hyperpigmentation caused by a metabolic disorder or skin irritation. In addition, one of the biggest concerns is skin cancers (e.g., squamous, basal, and melanoma) with melanoma being the deadliest form in America because of overexposure to UV radiation [35].
In most cases, humans are exposed to UV radiation due to overexposure to sunlight. UV radiation can produce many adverse effects within the cells, including DNA damage, skin pathologies, such as erythema and inflammation, skin aging, and cancer [36]. There are three main components of UV radiation, namely UVA (315–400 nm), UVB (280–315 nm), and UVC (100–280 nm) [37]. UVA can reach the dermis layer of skin, increasing the level of ROS that indirectly induce DNA mutagenesis, which results in skin aging and wrinkling. UVA can act as a carcinogen by shortening telomere in the DNA strand and it has less ability to stimulate melanin production resulting in redness, sun tanning, and freckles. UVB can penetrate the epidermis layer and damage the DNA in skin cells directly and induce skin cancers. UVC is highly bioactive but humans are not exposed to UVC because it is mostly absorbed by the ozone layer. In addition, UV-induced oxidative stress plays a crucial role in causing aging, inflammation, melanogenesis and even cancer which are shown in Figure 1 [9,12,38,39,40,41][9][12][38][39][40][41].
Figure 1. Effect of UV radiation-induced reactive oxygen species (ROS). Accumulation of ROS leads to skin cancer, inflammation, photoaging, wrinkling, and melanogenic through activation of respective signaling pathways.

2. Marine Algae-Derived Compounds in Cosmeceutical Application

Based on the evidence from previous studies, brown algae contribute the most in cosmeceuticals. Some bioactive compounds from brown algae exhibit multiple cosmeceutical activities, including phlorotannin, which possesses several activities, such as anti-melanogenic, antioxidant, anti-inflammation, and anti-aging [12,42,43,44][12][42][43][44]. Likewise, fucoidan, a sulphated polysaccharide isolated from brown algae, contributes to anti-inflammation, anti-melanogenic and anticancer [45,46,47][45][46][47]. Fucoxanthin, a carotenoid isolated from brown, red, green and microalgae exhibit anti-melanogenic, anti-aging and antioxidant activities [48,49,50][48][49][50]. Mycosporine-like amino acids (MAAs), which are commonly found in red and green seaweeds, and microalgae also contribute to antioxidant, anti-inflammation, and anti-aging [51,52,53][51][52][53]. Other examples of bioactive compounds derived from algae, their applications and mode of actions in cosmeceuticals are presented in Table 1. The chemical structures of some prominent bioactive compounds are shown in Figure 2.
/media/item_content/202201/61e913a94bf1fmarinedrugs-18-00323-g004b.png
Figure 2. Chemical structures of bioactive compounds derived from algae. (1) Eckol, (2) Fucosterol, (3) Diphlorethohydroxycarmalol, (4) Mycosporine-glycine, (5) Eleganonal, (6) Phenol, (7) Ascophyllan, (8) Laurinterol, (9) Fucoidan, (10) Eicosapentaenoic acid, (11) Lutein, (12) Sargachromanol E, (13) Fucoxanthin, (14) Astaxanthin, (15) Zeaxanthin, and (16) Lycopene.
Table 1. Bioactive compounds derived from algae and their applications in cosmeceuticals.

Algae Species

Bioactive Compound/Extract

Beneficial Activity

Mechanism of Action

Experimental Model

Reference

Brown algae

Ascophyllum nodosum

Ascophyllan

Anticancer

Inhibit MMP expression

B16 melanoma cells

[54]

Bifurcaria bifurcata

Eleganonal

Antioxidant

DPPH inhibition

In vitro

[55]

Chnoospora implexa

Ethanol extract

Antimicrobial

Bacterial growth inhibition

Staphylococcus aureus, Staphylococcus pyogenes

[56]

Chnoospora minima

Fucoidan

Anti-inflammation

Inhibition of LPS-induced NO production, iNOS, COX-2, and PGE2 levels

RAW macrophages

[47]

Cladosiphon okamuranus

Fucoxanthin

Antioxidant

DPPH inhibition

In vitro

[49]

Colpomenia sinuosa

Ethanol extract

Antimicrobial

Bacterial growth inhibition

S. aureus, S. pyogenes

[56]

Cystoseira barbata

Fat-soluble vitamin and carotenoids

Antioxidant

High fat-soluble vitamin and carotenoid content

In vitro

[57]

Cystoseira foeniculacea

Polyphenol

Antioxidant

DPPH inhibition (EC50 = 5.27 mg/mL)

In vitro

[58]

Cystoseira hakodatensis

Phenol and fucoxanthin

Antioxidant

High total phenolic and fucoxanthin content

In vitro

[59]

Cystoseira osmundacea

Ethanol extract

Antimicrobial

Bacterial growth inhibition

S. pyogenes

[56]

Dictyopteris delicatula

Ethanol extract

Antimicrobial

Bacterial growth inhibition

S. aureus, S. pyogenes

[56]

Dictyota dichotoma

Algae extract

Antimicrobial

Inhibit the synthesis of the peptidoglycan layer of bacterial cell walls

Penicillium purpurescens, Candida albicans, Aspergillus flavus

[60]

Ecklonia cava

Dieckol

Anti-inflammation

Suppression of iNOS and COX-2

Murine BV2 microglia

[61]

Phlorotannin

Anti-melanogenic

Inhibit melanin production

B16F10 melanoma cells

[44]

Phlorotannin

Antioxidant

ROS scavenging potential

Chinese hamster lung fibroblast (V79-4)

[62]

Ecklonia kurome

Phlorotannin

Anti-inflammation

Inhibit hyaluronidase

Assay of HAase (In vitro)

[42]

Ecklonia Stolonifera

Phlorotannin

Anti-aging

Inhibit MMP expression

Human dermal fibroblast cell

[43]

Phlorofucofuroeckol A and B

Anti-inflammation

Inhibition of NO production by downregulating iNOS and prostaglandin E2

LPS stimulated RAW 264.7 cells

[63]

Eisenia arborea

Phlorotannin

Anti-inflammation

Inhibit release of histamine

Rat basophile leukemia cells (RBL-2HE)

[64]

Eisenia bicyclis

Phlorotannin

Anti-inflammation

Inhibit hyaluronidase

Assay of HAase (In vitro)

[42]

Fucus evanescens

Fucoidan

Anticancer

Inhibit cell proliferation

Human malignant melanoma cells

[45]

Fucus vesiculosus

Extract

Anti-aging

Stimulate collagen production

N/A

[8]

Fucoidan

Anti-melanogenic

Inhibit tyrosinase and melanin

B16 murine melanoma cells

[46]

Fucoidan

Anticancer

Decrease melanoma growth

Mice

[65]

Fucoxanthin

Antioxidant

Prevent oxidation formation

In vitro, RAW 264.7 macrophage, Mouse (ex vivo)

[66]

Halopteris scoparia

Ethanol extract

Anti-inflammation

COX-2 inhibition

COX inhibitory screening assay kit

[67]

Himanthalia elongota

Fatty acid and Phenol

Antimicrobial

Bacterial growth inhibition

Escherichia coli, Staphylococcus aureus

[68]

Hizikia fusiformis

Fucosterol

Anti-aging

Inhibit MMP expression

Human dermal fibroblast

[18]

Ethyl acetate extract

Anti-melanogenic

Inhibit tyrosinase and melanin

B16F10 mouse melanoma cells

[69]

Fucoxanthin

Antioxidant

DPPH inhibition

In vitro

[70]

Hydroclathrus clathratus

Ethanol extract

Antimicrobial

Bacterial growth inhibition

S. aureus, S. pyogenes

[56]

Ishige foliacea

Phlorotannin

Anti-melanogenic

Downregulation of tyrosinase and melanin synthesis

B16F10 cells

Zebrafish embryo

[71,72]

[71][72]

Ishige okamurae

Diphlorethohydroxycarmalol

Anti-inflammation

Down-regulation of iNOS and COX-2 expression and NF-κβ activation

Human umbilical vein endothelial cells

[73]

Laminaria japonica

Fucoxanthin

Anti-melanogenic

Suppress tyrosinase activity

UVB- irradiated guinea pig

[48]

Laminaria ochroleuca

Polyphenol

Antioxidant

High total phenolic content and antioxidant capacity

In vitro

[74]

Macrocystis pyrifera

Phlorotannin

Antioxidant

ROS scavenging potential

In vitro

[8]

Hyaluronic acid

Anti-aging

Enhance the production of syndecan-4

N/A

[75]

Padina concrescens

Ethanol extract

Antimicrobial

Bacterial growth inhibition

S. aureus, S. pyogenes

[56]

Padina pavonica

Polyphenol

Antimicrobial

Bacterial growth inhibition

Candida albicans and Mucor ramaniannus

[17]

Acetone extract

Antioxidant

Free radical scavenging activity (IC50 = 691.56 µg L−1)

In vitro

[60]

Padina tetrastromatic

Diterpenes

Antioxidant

DPPH (IC50 = 1.73) & ABTS (IC50 = 2.01) inhibitions

In vitro

[76]

Sulfated polysaccharide

Anti-inflammation

COX-2 and iNOS inhibitions

Paw edema in rats

[77]

Petalonia binghamiae

Ethanol extract

Anti-melanogenic

Inhibit tyrosinase and melanin

B16F10 murine melanoma cells

[78]

Aqueous extract

Antioxidant

Anti-inflammation

DPPH inhibition

COX-2 inhibition

In vitro

In vitro

[67]

Rosenvingea intrincata

Ethanol extract

Antimicrobial

Bacterial growth inhibition

S. aureus, S. pyogenes

[56]

Saccharina latissima

Phenol

Antioxidant

High total phenolic content, DPPH scavenging activity and FRAP

In vitro

[79]

Sargassum fulvellum

Fucoxanthin

Antioxidant

DPPH inhibition

In vitro

[70]

Sargassum furcatum

Methanol extract

Antioxidant

DPPH (EC50 = 0.461) & ABTS (EC50 = 0.266) inhibitions

In vitro

[80]

Sargassum hemiphyllum

Sulfated polysaccharide

Anti-inflammation

Inhibit LPS-induced inflammatory response

RAW 264.7 macrophage cells

[81]

Sargassum henslowianum

Sulfated polysaccharide

Anticancer

Activation of caspase-3

B16 melanoma cells

[82]

Sargassum horridum

Ethanol extract

Antimicrobial

Bacterial growth inhibition

S. aureus, S. pyogenes

[56]

Sargassum horneri

Sargachromanol.E

Anti-aging

Inhibit MMP expression

UVA irradiated dermal fibroblast

[83]

Alginic acid

Anti-inflammation

Inhibit inflammatory response

HaCaT cells

[84]

Sargassum muticum

Tetraprenyltoluquinol chromane meroterpenoid

Anti-aging

ROS scavenging potential

Human dermal fibroblast

[85]

Sargassum polycystum

Ethanol extract

Anti-melanogenic

Inhibit tyrosinase and melanin production

B16F10 melanoma cells

[39]

Sargassum serratifolium

Sargachromenol

Anti-melanogenic

Downregulation of microphthalmia-associated transcription factor

B16F10 melanoma cells

[39]

Sargassum siliquastrum

Fucoxanthin

Antioxidant

Reduced UVB-induced ROS production

Human fibroblast

[86]

Sargassum thunbergi

Thunbergols

Antioxidant

DPPH inhibition

In vitro

[87]

Sargassum vulgare

Methanol extract

Antioxidant

β-carotene bleaching activity

In vitro

[88]

Stoechospermum marginatum

Spatane diterpenoids

Anticancer

Cell growth inhibition

Murine B16F10 melanoma cells

[89]

Turbinaria conoides

Laminarin, alginate, fucoidan

Antioxidant

ROS scavenging potential

N/A

[33]

Turbinaria ornata

Fucoxanthin

Antioxidant

High FRAP value (>10 µM/µg of extract)

In vitro

[90]

Undaria pinnatifida

Fucoxanthin

Anti-aging

MMP expression reduction, VEGF

Mouse

[50]

Ethyl acetate extract

Anti-melanogenic

Down regulate melanin and inhibit tyrosinase

Mouse B16 melanoma cells

[91]

Polyunsaturated fatty acid

Anti-inflammation

N/A

Mouse ear edema and erythema

[92]

Fucoxanthin

Antioxidant

DPPH inhibition

In vitro

[70]

Red algae

Alsidium corallinum

Methanol extract

Antimicrobial

Bacterial growth inhibition

Escherichia coli, Klebsiella pneumoniae, Staphylococcus aureus

[93]

Bangia

Algae extract

Antioxidant

Induce peroxidase and superoxide dismutase to reduce oxidative stress

In vitro

[94]

Bryothamnion triquetrum

Methanol extract

Antioxidant

DPPH (EC50 = 0.357) & ABTS (EC50 = 0.370) inhibitions

In vitro

[80]

Ceramium rubrum

Methanol extract

Antimicrobial

Bacterial growth inhibition

Escherichia coli, Enterococcus faecalis, Staphylococcus aureus

[93]

Chondrocanthus acicularis

Methanol extract

Antimicrobial

Bacterial growth inhibition

E. coli, K. pneumoniae, E. faecalis, S. aureus

[93]

Chondrus canaliculatus

Polysaccharide

Antioxidant

DPPH inhibition

In vitro

[95]

Chondrus crispus

Aqueous extract

Antimicrobial

Bacterial growth inhibition

Salmonella Enteritidis

[96]

Corallina pilulifera

Methanol extract

Anti-aging

Antioxidant

Reduce the expression of gelatinase

Inhibit free radical oxidation

Human dermal fibroblast

Human fibrosarcoma (HT-1080)

[97]

Corallina vancouverensis

Ethanol extract

Antimicrobial

Bacterial growth inhibition

S. aureus, S. pyogenes

[56]

Ganonema farinosum

Ethanol extract

Antimicrobial

Bacterial growth inhibition

S. aureus, S. pyogenes

[56]

Gelidium crinaale

Fat-soluble vitamin and carotenoids

Antioxidant

High fat-soluble vitamin and carotenoid content

In vitro

[57]

Gelidium robustum

Ethanol extract

Antimicrobial

Bacterial growth inhibition

S. aureus, S. pyogenes

[56]

Gracilaria gracilis

Phenol

Antioxidant

ROS scavenging potential

In vitro

[98]

Gracilariopsis lemaneiformis

Sulfated polysaccharide

Antioxidant

DPPH, Superoxide radical assay, hydroxyl radical assay (EC50 = 2.45 mg/mL)

In vitro

[99]

Gracilaria salicornia

2H- chromenyl

Antioxidant

Anti-inflammation

DPPH and ABTS inhibitions

COX-1 inhibition

In vitro

[100]

Jania rubens

Glycosaminoglycan

Anti-aging

Collagen synthesis

Unknown

[75]

Laurencia caspica

Phenol

Ethanol extract

Antioxidant

Antimicrobial

DPPH inhibition

Bacterial growth inhibition

In vitro

Klebsiella pneumonia

, Pseudomonas aeroginosa

[101]

Laurencia luzonensis

Sesquiterpenes

Antimicrobial

Bacterial growth inhibition

Bacillus megaterium

[12]

Laurenicia obtusa

Polysaccharide

Antioxidant

DPPH (IC50 = 24 µg/mL), FRAP (IC50 = 92 µg/mL),

Hydroxyl radical scavenging activity (IC

50 = 113 µg/mL)

In vitro

[102]

Laurenicia pacifica

Laurinterol

Antimicrobial

Bacterial growth inhibition

Staphylococcus aureus

[9]

Laurencia rigida

Sesquiterpenes

Antimicrobial

Bacterial growth inhibition

Bacillus megaterium

[12]

Meristotheca dakarensis

Glycosaminoglycan

Anti-aging

Collagen synthesis

Unknown

[75]

Osmundaria obtusilo

Methanol extract

Antioxidant

DPPH (EC50 = 0.041 mg/mL), ABTS (EC50 = 0.031 mg/mL), Metal chelating (EC50 = 0.1 mg/mL), folin ciocalteu (EC50 = 0.128 mg/mL)

In vitro

[80]

Palisada flagellifera

Methanol extract

Antioxidant

β-carotene bleaching activity

In vitro

[88]

Palmaria palmata

MAA

Anti-aging

Collagenase inhibition

Clostridium histolyticum

[53]

Polysiphonia howei

Fucoxanthin

Antioxidant

High FRAP value

(>5 µM/µg of extract)

In vitro

[90]

Porphyra haitanensis

Sulfated Polysaccharide

Antioxidant

ROS scavenging potential

Mice

[103]

Porphyra umbilicalis

MAA

Anti-aging

Control expression of MMP

Human dermal fibroblast

[16]

Porphyra sp.

MAA

Anti-aging

Collagenases inhibition

Clostridium histolyticum

[53]

Porphyra yezoensis

MAA

Polyphenol

Phycoerythrin

Antioxidant

Anticancer

Anti-inflammation

ROS scavenging potential and MMP expression

Induce apoptosis

Suppression of mast cells

Human skin fibroblast

HaCaT cells

Rat

[51]

Pterocladia capillacea

Sulfated polysaccharide

Antimicrobial

N/A

Staphylococcus aureus

Escherichia coli

[104]

Pyropia columbia

Phenol

Antioxidant

DPPH, β-carotene bleaching and ABTS inhibitions

Piaractus mesopotamicus

[105]

Pyropia yezoensis

Polysaccharide

Anti-aging

Promote collagen synthesis

Human dermal fibroblast

[106]

Rhodomela confervoides

Polyphenol

Antimicrobial

Bacterial growth inhibition

Candida albicans

Mucor ramaniannus

[17]

Bromophenol

Antioxidant

DPPH inhibition

In vitro

[107]

Schizymenia dubyi

Phenol

Anti-melanogenic

Inhibit tyrosinase activity

In vitro

[39]

Green algae

Bryopsis plumose

Polysaccharide

Antioxidant

ROS scavenging potential

In vitro

[108]

Chaetomorpha antennia

Fucoxanthin

Antioxidant

DPPH inhibition (63.77%)

In vitro

[109]

Chlamydomonas hedleyi

MAA

Antioxidant

Anti-aging

Anti-inflammation

ROS scavenging potential

Increase UV-suppressed genes (procollagen C proteinase enhancer and elastin) expression

Reduce COX-2 and involucrin expression

In vitro

HaCaT cells

HaCaT cells

[52]

Cladophora sp.

Ethanol extract

Antimicrobial

Bacterial growth inhibition

S. aureus, S. pyogenes

[56]

Codium amplivesiculatum

Ethanol extract

Antimicrobial

Bacterial growth inhibition

S. aureus, S. pyogenes

[56]

Codium cuneatum

Ethanol extract

Antimicrobial

Bacterial growth inhibition

S. aureus, S. pyogenes

[56]

Codium fragile

Sterol

Anti-inflammation

Reduces the expression of COX-2, iNOS, and TNF-α

Mice

[110]

Codium simulans

Ethanol extract

Antimicrobial

Bacterial growth inhibition

S. aureus, S. pyogenes

[56]

Entromorpha intestinalis

Chloroform and methanol extract

Antioxidant

SOD activity is reduced

Labidochromis caeruleus

[111]

Enteromorpha linza

Polysaccharide

Antioxidant

ROS scavenging potential

In vitro

[108]

Gayralia oxysperma

Fucoxanthin

Antioxidant

High FRAP value

(>6 µM/µg of extract)

In vitro

[90]

Ulva dactilifera

Ethanol extract

Antimicrobial

Bacterial growth inhibition

S. aureus, Streptococcus pyogenes

[56]

Ulva fasciata

Fucoxanthin

Antioxidant

DPPH inhibition (83.95%)

In vitro

[109]

Ulva lactuca

Phycocolloids

Anti-inflammation

N/A

N/A

[75]

Ulva pertusa

Polysaccharide

Antioxidant

ROS scavenging potential

In vitro

[108]

Ulva prolifera

Phenol and flavonoid

Antioxidant

DPPH inhibition, high phenolic and flavonoid contents

In vitro

[112]

Ulva rigida

Phenol

Antioxidant

DPPH inhibition

In vitro

[113]

Ulva sp.

Sulfated polysaccharide

Anti-aging

Increase hyaluronan production

Human dermal fibroblast

[114]

Microalgae/Cyanobacteria

Anabaena vaginicola

Lycopene

Antioxidant

Anti-aging

N/A

In vitro

[115]

Arthrospira platensis

Methanol extracts of exopolysaccharides

Antioxidant

N/A

In vitro

[115]

Chlorella fusca

Sporopollenin

Anti-aging

Protect cells from UV radiation

N/A

[116]

Chlorella minutissima

MAA

Anti-aging

Protect cells from UV radiation

N/A

[116]

Chlorella sorokiniana

MAA

Anti-aging

Protect cells from UV radiation

N/A

[116]

Lutein

Anti-aging

Reduce UV induced damage

N/A

[33]

Chlorella vulgaris

Hot water extract

Anti-aging

Reduced activity of SOD

Human diploid fibroblast

[117]

Anti-inflammation

Downregulated mRNA expression levels of IL-4 and IFN-γ

NC/Nga mice

[118]

Dunaliella salina

β-carotene

Antioxidant

Protect against oxidative stress

Rat

[119]

β-cryptoxanthin

Anti-inflammation

Reduced the production of IL-1β, IL-6, TNF-α, the protein expression of iNOS and COX-2

LPS-stimulated RAW 264.7 cells

[120]

Haematococcus pluvialis

Astaxanthin (carotenoid)

Anti-aging

Inhibit MMP expression

Mice and human dermal fibroblasts

[121]

Anticancer

ROS scavenging potential

Mice

[122]

Nannochloropsis granulata

Carotenoid

Antioxidant

DPPH inhibition

In vitro

[123]

Nannochloropsis oculata

Zeaxanthin

Anti-melanogenic

Inhibit tyrosinase

In vitro

[124]

Nitzschia sp.

Fucoxanthin

Antioxidant

Reduced oxidative stress

Human Glioma Cells

[125]

Nostoc sp.

MAA

Antioxidant

ROS scavenging potential

In vitro

[126]

Odontella aurita

EPA

Antioxidant

Reduce oxidative stress

Rat

[127]

Planktochlorella nurekis

Fatty acid

Antimicrobial

Bacterial growth inhibition

Campylobacter jejuni, E. coli, Salmonella enterica var.

[128]

Porphyridium sp.

Sulfated polysaccharide

Anti-inflammation

Antioxidant

Inhibit proinflammatory modulator

Inhibited oxidative damage

Unknown

3T3 cells

[103]

Rhodella reticulata

Sulfated polysaccharide

Antioxidant

ROS scavenging potential

In vitro

[103]

Skeletonema marinoi

Polyunsaturated aldehyde and fatty acid

Anticancer

Inhibit cell proliferation

Human melanoma cells (A2058)

[129]

Spirulina platensis

β-carotene and phycocyanin

Antioxidant

Anti-inflammation

Inhibit lipid peroxidation

Inhibit TNF-α and IL-6 expressions

Mouse

Human dermal fibroblast cells (CCD-986sk)

[130]

Ethanol extract

Antimicrobial

Bacterial growth inhibition

E. coli, Pseudomonas aeruginosa, Bacillus subtilis, and Aspergillus niger

[131]

Synechocystis spp.

Fatty acids and phenols

Antimicrobial

Bacterial growth inhibition

E. coli and S. aureus

[68]

References

  1. Kligman, D. Cosmeceuticals. Dermatol. Clin. 2000, 18, 609–615.
  2. Dureja, H.; Kaushik, D.; Gupta, M.; Kumar, V.; Lather, V. Cosmeceuticals: An emerging concept. Indian J. Pharm. 2005, 37, 155–159.
  3. Smit, N.; Vicanova, J.; Pavel, S. The Hunt for Natural Skin Whitening Agents. Int. J. Mol. Sci. 2009, 10, 5326–5349.
  4. Bos, J.D.; Meinardi, M.M. The 500 Dalton rule for the skin penetration of chemical compounds and drugs. Exp. Dermatol. 2000, 9, 165–169.
  5. Kerdudo, A.; Burger, P.; Merck, F.; Dingas, A.; Rolland, Y.; Michel, T.; Fernandez, X. Development of a natural ingredient–Natural preservative: A case study. Comptes Rendus Chimie 2016, 19, 1077–1089.
  6. Barrett, J. Chemical Exposures: The Ugly Side of Beauty Products. Environ Health Perspect. 2005, 113, A24.
  7. Global Market Value for Natural Cosmetics in 2018–2027|Statista. Available online: https://www.statista.com/statistics/673641/global-market-value-for-natural-cosmetics/ (accessed on 18 November 2019).
  8. Ariede, M.B.; Candido, T.M.; Jacome, A.L.M.; Velasco, M.V.R.; de Carvalho, J.C.M.; Baby, A.R. Cosmetic attributes of algae—A review. Algal Res. 2017, 25, 483–487.
  9. Wang, H.M.D.; Chen, C.C.; Huynh, P.; Chang, J.S. Exploring the potential of using algae in cosmetics. Bioresour. Technol. 2015, 184, 355–362.
  10. Fu, W.; Nelson, D.R.; Yi, Z.; Xu, M.; Khraiwesh, B.; Jijakli, K.; Chaiboonchoe, A.; Alzahmi, A.; Al-Khairy, D.; Brynjolfsson, S.; et al. Bioactive compounds from microalgae: Current development and prospects. Stud. Nat. Prod. Chem. 2017, 54, 199–225.
  11. Freile-Pelegrín, Y.; Robledo, D. Bioactive phenolic compounds from algae. In Bioactive Compounds from Marine Foods: Plant and Animal Sources, 1st ed.; Hernández-Ledesma, B., Herrero, M., Eds.; John Wiley & Sons, Ltd.: Chichester, UK, 2014; pp. 113–129.
  12. Thomas, N.V.; Kim, S.K. Beneficial effects of marine algal compounds in cosmeceuticals. Mar. Drugs 2013, 11, 146–164.
  13. Fernando, I.S.; Nah, J.W.; Jeon, Y.J. Potential anti-inflammatory natural products from marine algae. Environ. Toxicol. Pharm. 2016, 48, 22–30.
  14. 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.
  15. Talero, E.; García-Mauriño, S.; Ávila-Román, J.; Rodríguez-Luna, A.; Alcaide, A.; Motilva, V. Bioactive compounds isolated from microalgae in chronic inflammation and cancer. Mar. Drugs 2015, 13, 6152–6209.
  16. Wang, H.M.D.; Li, X.C.; Lee, D.J.; Chang, J.S. Potential biomedical applications of marine algae. Bioresour. Technol. 2017, 244, 1407–1415.
  17. Saidani, K.; Bedjou, F.; Benabdesselam, F.; Touati, N. Antifungal activity of methanolic extracts of four Algerian marine algae species. Afr. J. Biotechnol. 2012, 11, 9496–9500.
  18. 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.
  19. Martins, A.; Vieira, H.; Gaspar, H.; Santos, S. Marketed marine natural products in the pharmaceutical and cosmeceutical industries: Tips for success. Mar. Drugs 2014, 12, 1066–1101.
  20. Nayaka, S.; Toppo, K.; Verma, S. Adaptation in Algae to Environmental Stress and Ecological Conditions. In Plant Adaptation Strategies in Changing Environment; Springer: Singapore, 2017; pp. 103–115.
  21. Katz, A.; Waridel, P.; Shevchenko, A.; Pick, U. Salt-induced changes in the plasma membrane proteome of the halotolerant alga Dunaliella salina as revealed by blue native gel electrophoresis and nano-LC-MS/MS analysis. Mol. Cell. Proteom. 2007, 6, 1459–1472.
  22. Sydney, E.B.; Sydney, A.C.N.; de Carvalho, J.C.; Soccol, C.R. Potential carbon fixation of industrially important microalgae. In Biofuels from Algae; Elsevier: Amsterdam, The Netherlands, 2019; pp. 67–88.
  23. Usher, P.K.; Ross, A.B.; Camargo-Valero, M.A.; Tomlin, A.S.; Gale, W.F. An overview of the potential environmental impacts of large-scale microalgae cultivation. Biofuels 2014, 5, 331–349.
  24. Álvarez-Gómez, F.; Korbee, N.; Casas-Arrojo, V.; Abdala-Díaz, R.T.; Figueroa, F.L. UV photoprotection, cytotoxicity and immunology capacity of red algae extracts. Molecules 2019, 24, 341.
  25. Khanra, S.; Mondal, M.; Halder, G.; Tiwari, O.N.; Gayen, K.; Bhowmick, T.K. Downstream processing of microalgae for pigments, protein and carbohydrate in industrial application: A review. Food Bioprod. Process. 2018, 110, 60–84.
  26. Pagels, F.; Guedes, A.C.; Amaro, H.M.; Kijjoa, A.; Vasconcelos, V. Phycobiliproteins from cyanobacteria: Chemistry and biotechnological applications. Biotechnol. Adv. 2019, 37, 422–443.
  27. Ambati, R.R.; Gogisetty, D.; Aswathanarayana, R.G.; Ravi, S.; Bikkina, P.N.; Bo, L.; Yuepeng, S. Industrial potential of carotenoid pigments from microalgae: Current trends and future prospects. Crit. Rev. Food Sci. Nutr. 2019, 59, 1880–1902.
  28. Galasso, C.; Corinaldesi, C.; Sansone, C. Carotenoids from marine organisms: Biological functions and industrial applications. Antioxidant 2017, 6, 96.
  29. Torres, P.; Santos, J.P.; Chow, F.; dos Santos, D.Y. A comprehensive review of traditional uses, bioactivity potential, and chemical diversity of the genus Gracilaria (Gracilariales, Rhodophyta). Algal Res. 2019, 37, 288–306.
  30. Jahan, A.; Ahmad, I.Z.; Fatima, N.; Ansari, V.A.; Akhtar, J. Algal bioactive compounds in the cosmeceutical industry: A review. Phycologia 2017, 56, 410–422.
  31. Siezen, R.J. Microbial sunscreens. Microb. Biotechnol. 2011, 4, 1–7.
  32. Muñoz, R.; Gonzalez-Fernandez, C. (Eds.) Microalgae-Based Biofuels and Bioproducts: From Feedstock Cultivation to End-Products; Woodhead Publishing: Duxford, UK, 2017.
  33. Berthon, J.Y.; Nachat-Kappes, R.; Bey, M.; Cadoret, J.P.; Renimel, I.; Filaire, E. Marine algae as attractive source to skin care. Free Radic. Res. 2017, 51, 555–567.
  34. Tabassum, N.; Hamdani, M. Plants used to treat skin diseases. Pharm. Rev. 2014, 8, 52–60.
  35. Skin Cancer|Skin Cancer Facts|Common Skin Cancer Types. Available online: https://www.cancer.org/cancer/skin-cancer.html (accessed on 27 August 2018).
  36. Tan, L.T.; Mahendra, C.K.; Yow, Y.Y.; Chan, K.G.; Khan, T.M.; Lee, L.H.; Goh, B.H. Streptomyces sp. MUM273b: A mangrove-derived potential source for antioxidant and UVB radiation protectants. Microbiol. Open 2019, 8, e859.
  37. D’Orazio, J.; Jarrett, S.; Amaro-Ortiz, A.; Scott, T. UV radiation and the skin. Int. J. Mol. Sci. 2013, 14, 12222–12248.
  38. Amaro-Ortiz, A.; Yan, B.; D’Orazio, J.A. Ultraviolet radiation, aging and the skin: Prevention of damage by topical cAMP manipulation. Molecules 2014, 19, 6202–6219.
  39. 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.
  40. Hwang, H.; Chen, T.; Nines, R.G.; Shin, H.C.; Stoner, G.D. Photochemoprevention of UVB-induced skin carcinogenesis in SKH-1 mice by brown algae polyphenols. Int. J. Cancer 2006, 119, 2742–2749.
  41. Mahendra, C.K.; Tan, L.T.H.; Yap, W.H.; Chan, C.K.; Pusparajah, P.; Goh, B.H. An optimized cosmetic screening assay for ultraviolet B (UVB) protective property of natural products. Prog. Drug Discov. Biomed. Sci. 2019, 2.
  42. Shibata, T.; Fujimoto, K.; Nagayama, K.; Yamaguchi, K.; Nakamura, T. Inhibitory activity of brown algal phlorotannins against hyaluronidase. Int. J. Food Sci. Technol. 2002, 37, 703–709.
  43. Joe, M.J.; Kim, S.N.; Choi, H.Y.; Shin, W.S.; Park, G.M.; Kang, D.W.; 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.
  44. Yoon, N.Y.; Eom, T.K.; Kim, M.M.; Kim, S.K. Inhibitory effect of phlorotannins isolated from Ecklonia cava on mushroom tyrosinase activity and melanin formation in mouse B16F10 melanoma cells. J. Agric. Food Chem. 2009, 57, 4124–4129.
  45. Anastyuk, S.; Shervchenko, N.; Ermakova, S.; Vishchuk, O.; Nazarenko, E.; Dmitrenok, P.; Zvyagintseva, T. Anticancer activity in vitro of a fucoidan from the brown algae Fucus evanescens and its low-molecular fragments, structurally characterized by tandem mass-spectrometry. Carbohydr. Polym. 2012, 87, 186–194.
  46. Wang, Z.-J.; Xu, W.; Liang, J.-W.; Wang, C.-S.; Kang, Y. Effect of fucoidan on B16 murine melanoma cell melanin formation and apoptosis. Afr. J. Tradit. Complement. Altern. Med. 2017, 14, 149–155.
  47. Fernando, I.S.; Sanjeewa, K.A.; Samarakoon, K.W.; Lee, W.W.; Kim, H.S.; Kang, N.; Ranasinghe, P.; Lee, H.S.; Jeon, Y.J. A fucoidan fraction purified from Chnoospora minima; a potential inhibitor of LPS-induced inflammatory responses. Int. J. Boil. Macromol. 2017, 104, 1185–1193.
  48. Shimoda, H.; Tanaka, J.; Shan, S.J.; Maoka, T. Anti-pigmentary activity of fucoxanthin and its influence on skin mRNA expression of melanogenic molecules. J. Pharm. Pharm. 2010, 62, 1137–1145.
  49. Mise, T.; Ueda, M.; Yasumoto, T. Production of fucoxanthin-rich powder from Cladosiphon okamuranus. Adv. J. Food Sci. Technol. 2011, 3, 73–76.
  50. Urikura, I.; Sugawara, T.; Hirata, T. Protective effect of fucoxanthin against UVB-induced skin photoaging in hairless mice. Biosci. Biotechnol. Biochem. 2011, 75, 757–760.
  51. Sakai, S.; Komura, Y.; Nishimura, Y.; Sugawara, T.; Hirata, T. Inhibition of mast cell degranulation by phycoerythrin and its pigment moiety phycoerythrobilin, prepared from Porphyra yezoensis. Food Sci. Technol. Res. 2011, 17, 171–177.
  52. Suh, S.S.; Hwang, J.; Park, M.; Seo, H.; Kim, H.S.; Lee, J.; Moh, S.; Lee, T.K. Anti-inflammation activities of mycosporine-like amino acids (MAAs) in response to UV radiation suggest potential anti-skin aging activity. Mar. Drugs 2014, 12, 5174–5187.
  53. Hartmann, A.; Gostner, J.; Fuchs, J.E.; Chaita, E.; Aligiannis, N.; Skaltsounis, L.; Ganzera, M. Inhibition of collagenase by mycosporine-like amino acids from marine sources. Planta Medica 2015, 81, 813–820.
  54. Abu, R.; Jiang, Z.; Ueno, M.; Isaka, S.; Nakazono, S.; Okimura, T.; Cho, K.; Yamaguchi, K.; Kim, D.; Oda, T. Anti-metastatic effects of the sulfated polysaccharide ascophyllan isolated from Ascophyllum nodosum on B16 melanoma. Biochem. Biophys. Res. Commun. 2015, 458, 727–732.
  55. Silva, J.; Alves, C.; Freitas, R.; Martins, A.; Pinteus, S.; Ribeiro, J.; Gaspar, H.; Alfonso, A.; Pedrosa, R. Antioxidant and Neuroprotective Potential of the Brown Seaweed Bifurcaria bifurcata in an in vitro Parkinson’s Disease Model. Mar. Drugs 2019, 17, 85.
  56. Muñoz-Ochoa, M.; Murillo-Álvarez, J.I.; Zermeño-Cervantes, L.A.; Martínez-Díaz, S.; Rodríguez-Riosmena, R. Screening of extracts of algae from Baja California Sur, Mexico as reversers of the antibiotic resistance of some pathogenic bacteria. Eur. Rev. Med. Pharmacol. Sci. 2010, 14, 739–747.
  57. Panayotova, V.; Merzdhanova, A.; Dobreva, D.A.; Zlatanov, M.; Makedonski, L. Lipids of black sea algae: Unveiling their potential for pharmaceutical and cosmetic applications. J. IMAB–Ann. Proc. Sci. Pap. 2017, 23, 1747–1751.
  58. Messina, C.M.; Renda, G.; Laudicella, V.A.; Trepos, R.; Fauchon, M.; Hellio, C.; Santulli, A. From ecology to biotechnology, study of the defense strategies of algae and halophytes (from Trapani Saltworks, NW Sicily) with a focus on antioxidants and antimicrobial properties. Int. J. Mol. Sci. 2019, 20, 881.
  59. Airanthi, M.W.A.; Hosokawa, M.; Miyashita, K. Comparative antioxidant activity of edible Japanese brown seaweeds. J. Food Sci. 2011, 76, C104–C111.
  60. Kosanić, M.; Ranković, B.; Stanojković, T. Brown macroalgae from the Adriatic Sea as a promising source of bioactive nutrients. J. Food Meas. Charact. 2019, 13, 330–338.
  61. Jung, W.K.; Heo, S.J.; Jeon, Y.J.; Lee, C.M.; Park, Y.M.; Byun, H.G.; Choi, Y.H.; Park, S.G.; Choi, I.W. Inhibitory effects and molecular mechanism of dieckol isolated from marine brown alga on COX-2 and iNOS in microglial cells. J. Agric. Food Chem. 2009, 57, 4439–4446.
  62. Kang, K.A.; Lee, K.H.; Chae, S.; Koh, Y.S.; Yoo, B.S.; Kim, J.H.; Ham, Y.M.; Baik, J.S.; Lee, N.H.; Hyun, J.W. Triphlorethol-A from Ecklonia cava protects V79-4 lung fibroblast against hydrogen peroxide induced cell damage. Free Radic. Res. 2005, 39, 883–892.
  63. Lee, M.S.; Kwon, M.S.; Choi, J.W.; Shin, T.; No, H.K.; Choi, J.S.; Byun, D.S.; Kim, J.I.; Kim, H.R. Anti-inflammatory activities of an ethanol extract of Ecklonia stolonifera in lipopolysaccharide-stimulated RAW 264.7 murine macrophage cells. J. Agric. Food Chem. 2012, 60, 9120–9129.
  64. Sugiura, Y.; Takeuchi, Y.; Kakinuma, M.; Amano, H. Inhibitory effects of seaweeds on histamine release from rat basophile leukemia cells (RBL-2H3). Fish. Sci. 2006, 72, 1286–1291.
  65. Teas, J.; Irhimeh, M.R. Melanoma and brown seaweed: An integrative hypothesis. J. Appl. Phycol. 2017, 29, 941–948.
  66. Zaragozá, M.C.; López, D.P.; Sáiz, M.; Poquet, M.; Pérez, J.; Puig-Parellada, P.; Marmol, F.; Simonetti, P.; Gardana, C.; Lerat, Y.; et al. Toxicity and antioxidant activity in vitro and in vivo of two Fucus vesiculosus extracts. J. Agric. Food Chem. 2008, 56, 7773–7780.
  67. Campos, A.M.; Matos, J.; Afonso, C.; Gomes, R.; Bandarra, N.M.; Cardoso, C. Azorean macroalgae (Petalonia binghamiae, Halopteris scoparia and Osmundea pinnatifida) bioprospection: A study of fatty acid profiles and bioactivity. Int. J. Food Sci. Technol. 2018, 54, 880–890.
  68. Plaza, M.; Santoyo, S.; Jaime, L.; Reina, G.G.B.; Herrero, M.; Señoráns, F.J.; Ibáñez, E. Screening for bioactive compounds from algae. J. Pharm. Biomed. Anal. 2010, 51, 450–455.
  69. Choi, E.O.; Kim, H.S.; Han, M.H.; Choi, Y.H.; Park, C.; Kim, B.W.; Hwang, H.J. Effects of Hizikia fusiforme fractions on melanin synthesis in B16F10 melanoma cells. J. Life Sci. 2013, 23, 1495–1500.
  70. Yan, X.; Chuda, Y.; Suzuki, M.; Nagata, T. Fucoxanthin as the major antioxidant in Hijikia fusiformis, a common edible seaweed. Biosci. Biotechnol. Biochem. 1999, 63, 605–607.
  71. Kim, K.N.; Yang, H.M.; Kang, S.M.; Kim, D.; Ahn, G.; Jeon, Y.J. Octaphlorethol A isolated from Ishige foliacea inhibits α-MSH-stimulated induced melanogenesis via ERK pathway in B16F10 melanoma cells. Food Chem. Toxicol. 2013, 59, 521–526.
  72. Kim, K.N.; Yang, H.M.; Kang, S.M.; Ahn, G.N.; Roh, S.W.; Lee, W.; Kim, D.K.; Jeon, Y.J. Whitening effect of octaphlorethol A isolated from Ishige foliacea in an in vivo zebrafish model. J. Microbiol. Biotechnol. 2015, 25, 448–451.
  73. Heo, S.J.; Hwang, J.Y.; Choi, J.I.; Lee, S.H.; Park, P.J.; Kang, D.H.; Oh, C.; Kim, D.W.; Han, J.S.; Jeon, Y.J.; et al. Protective effect of diphlorethohydroxycarmalol isolated from Ishige okamurae against high glucose-induced-oxidative stress in human umbilical vein endothelial cells. Food Chem. Toxicol. 2010, 48, 1448–1454.
  74. Del Olmo, A.; Picon, A.; Nuñez, M. High pressure processing for the extension of Laminaria ochroleuca (kombu) shelf-life: A comparative study with seaweed salting and freezing. Innov. Food Sci. Emerg. Technol. 2019, 52, 420–428.
  75. SpecialChem—The Universal Selection Source: Cosmetics Ingredients. Available online: https://cosmetics.specialchem.com/ (accessed on 5 May 2020).
  76. Antony, T.; Chakraborty, K. Xenicanes attenuate pro-inflammatory 5-lipoxygenase: Prospective natural anti-inflammatory leads from intertidal brown seaweed Padina tetrastromatica. Med. Chem. Res. 2019, 28, 591–607.
  77. Mohsin, S.; Kurup, G.M. Mechanism underlying the anti-inflammatory effect of sulphated polysaccharide from Padina tetrastromatica against carrageenan induced paw edema in rats. Biomed. Prev. Nutr. 2011, 1, 294–301.
  78. Yoon, H.S.; Koh, W.B.; Oh, Y.S.; Kim, I.J. The Anti-Melanogenic Effects of Petalonia binghamiae extarcts in α-melanocyte stimulating hormone-induced B16/F10 murine melanoma cells. J. Korean Soc. Appl. Biol. Chem. 2009, 52, 564–567.
  79. Sappati, P.K.; Nayak, B.; VanWalsum, G.P.; Mulrey, O.T. Combined effects of seasonal variation and drying methods on the physicochemical properties and antioxidant activity of sugar kelp (Saccharina latissima). J. Appl. Phycol. 2019, 31, 1311–1332.
  80. Vasconcelos, J.B.; de Vasconcelos, E.R.; Urrea-Victoria, V.; Bezerra, P.S.; Reis, T.N.; Cocentino, A.L.; Navarro, D.M.; Chow, F.; Areces, A.J.; Fujii, M.T. Antioxidant activity of three seaweeds from tropical reefs of Brazil: Potential sources for bioprospecting. J. Appl. Phycol. 2019, 31, 835–846.
  81. Hwang, P.A.; Chien, S.Y.; Chan, Y.L.; Lu, M.K.; Wu, C.H.; Kong, Z.L.; Wu, C.J. Inhibition of lipopolysaccharide (LPS)-induced inflammatory responses by Sargassum hemiphyllum sulfated polysaccharide extract in RAW 264.7 macrophage cells. J. Agric. Food Chem. 2011, 59, 2062–2068.
  82. Ale, M.T.; Maruyama, H.; Tamauchi, H.; Mikkelsen, J.D.; Meyer, A.S. Fucose-containing sulfated polysaccharides from brown seaweeds inhibit proliferation of melanoma cells and induce apoptosis by activation of caspase-3 in vitro. Mar. Drugs 2011, 9, 2605–2621.
  83. Kim, J.A.; Ahn, B.N.; Kong, C.S.; Kim, S.K. The chromene sargachromanol E inhibits ultraviolet A-induced ageing of skin in human dermal fibroblasts. Br. J. Dermatol. 2013, 168, 968–976.
  84. Fernando, I.S.; Jayawardena, T.U.; Sanjeewa, K.A.; Wang, L.; Jeon, Y.J.; Lee, W.W. Anti-inflammatory potential of alginic acid from Sargassum horneri against urban aerosol-induced inflammatory responses in keratinocytes and macrophages. Ecotoxicol. Environ. Saf. 2018, 160, 24–31.
  85. Balboa, E.M.; Li, Y.X.; Ahn, B.N.; Eom, S.H.; Domínguez, H.; Jiménez, C.; Rodríguez, J. Photodamage attenuation effect by a tetraprenyltoluquinol chromane meroterpenoid isolated from Sargassum muticum. J. Photochem. Photobiol. B Biol. 2015, 148, 51–58.
  86. Heo, S.-J.; Jeon, Y.-J. Protective effect of fucoxanthin isolated from Sargassum siliquastrum on UV-B induced cell damage. J. Photochem. Photobiol. B 2009, 95, 101–107.
  87. Seo, Y.; Park, K.E.; Kim, Y.A.; Lee, H.J.; Yoo, J.S.; Ahn, J.W.; Lee, B.J. Isolation of tetraprenyltoluquinols from the brown alga Sargassum thunbergii. Chem. Pharm. Bull. 2006, 54, 1730–1733.
  88. Santos, J.P.; Torres, P.B.; dos Santos, D.Y.; Motta, L.B.; Chow, F. Seasonal effects on antioxidant and anti-HIV activities of Brazilian seaweeds. J. Appl. Phycol. 2018, 31, 1333–1341.
  89. Velatooru, L.R.; Baggu, C.B.; Janapala, V.R. Spatane diterpinoid from the brown algae, Stoechospermum marginatum induces apoptosis via ROS induced mitochondrial mediated caspase dependent pathway in murine B16F10 melanoma cells. Mol. Carcinog. 2016, 55, 2222–2235.
  90. Kelman, D.; Posner, E.K.; McDermid, K.J.; Tabandera, N.K.; Wright, P.R.; Wright, A.D. Antioxidant activity of Hawaiian marine algae. Mar. Drugs 2012, 10, 403–416.
  91. Kim, M.; Kim, D.; Yoon, H.; Lee, W.; Lee, N.; Hyun, C. Melanogenesis inhibitory activity of Korean Undaria pinnatifida in mouse B16 melanoma cells. Interdiscip. Toxicol. 2014, 7, 89–92.
  92. Khan, M.N.A.; Yoon, S.J.; Choi, J.S.; Park, N.G.; Lee, H.H.; Cho, J.Y.; Hong, Y.K. Anti-edema effects of brown seaweed (Undaria pinnatifida) extract on phorbol 12-myristate 13-acetate-induced mouse ear inflammation. Am. J. Chin. Med. 2009, 37, 373–381.
  93. Rhimou, B.; Hassane, R.; José, M.; Nathalie, B. The antibacterial potential of the seaweeds (Rhodophyceae) of the Strait of Gibraltar and the Mediterranean Coast of Morocco. Afr. J. Biotechnol. 2010, 9, 6365–6372.
  94. Wang, W.J.; Li, X.L.; Zhu, J.Y.; Liang, Z.R.; Liu, F.L.; Sun, X.T.; Wang, F.J.; Shen, Z.G. Antioxidant response to salinity stress in freshwater and marine Bangia (Bangiales, Rhodophyta). Aquat. Bot. 2019, 154, 35–41.
  95. Jaballi, I.; Sallem, I.; Feki, A.; Cherif, B.; Kallel, C.; Boudawara, O.; Jamoussi, K.; Mellouli, L.; Nasri, M.; Amara, I.B. Polysaccharide from a Tunisian red seaweed Chondrus canaliculatus: Structural characteristics, antioxidant activity and in vivo hemato-nephroprotective properties on maneb induced toxicity. Int. J. Biol. Macromol. 2019, 123, 1267–1277.
  96. Kulshreshtha, G.; Borza, T.; Rathgeber, B.; Stratton, G.S.; Thomas, N.A.; Critchley, A.; Hafting, J.; Prithiviraj, B. Red seaweeds Sarcodiotheca gaudichaudii and Chondrus crispus down regulate virulence factors of Salmonella enteritidis and induce immune responses in Caenorhabditis elegans. Front. Microbiol. 2016, 7, 421.
  97. Ryu, B.; Qian, Z.J.; Kim, M.M.; Nam, K.W.; Kim, S.K. Anti-photoaging activity and inhibition of matrix metalloproteinase (MMP) by marine red alga, Corallina pilulifera methanol extract. Radiat. Phys. Chem. 2009, 78, 98–105.
  98. Francavilla, M.; Franchi, M.; Monteleone, M.; Caroppo, C. The red seaweed Gracilaria gracilis as a multi products source. Mar. Drugs 2013, 11, 3754–3776.
  99. Wang, X.; Zhang, Z.; Wu, Y.; Sun, X.; Xu, N. Synthesized sulfated and acetylated derivatives of polysaccharide extracted from Gracilariopsis lemaneiformis and their potential antioxidant and immunological activity. Int. J. Boil. Macromol. 2019, 124, 568–572.
  100. Antony, T.; Chakraborty, K. First report of antioxidative 2H-chromenyl derivatives from the intertidal red seaweed Gracilaria salicornia as potential anti-inflammatory agents. Nat. Prod. Res. 2019.
  101. Moshfegh, A.; Salehzadeh, A.; Shandiz, S.A.S.; Shafaghi, M.; Naeemi, A.S.; Salehi, S. Phytochemical analysis, antioxidant, anticancer and antibacterial properties of the Caspian Sea red macroalgae, Laurencia caspica. Iran. J. Sci. Technol. Trans. A Sci. 2019, 43, 49–56.
  102. Lajili, S.; Ammar, H.H.; Mzoughi, Z.; Amor, H.B.H.; Muller, C.D.; Majdoub, H.; Bouraoui, A. Characterization of sulfated polysaccharide from Laurencia obtusa and its apoptotic, gastroprotective and antioxidant activities. Int. J. Biol. Macromol. 2019, 126, 326–336.
  103. De Jesus Raposo, M.; de Morais, A.; de Morais, R. Marine polysaccharides from algae with potential biomedical applications. Mar. Drugs 2015, 13, 2967–3028.
  104. Pimentel, F.B.; Alves, R.C.; Rodrigues, F.; PP Oliveira, M.B. Macroalgae-derived ingredients for cosmetic industry—An Update. Cosmetics 2017, 5, 2.
  105. Cian, R.E.; Bacchetta, C.; Rossi, A.; Cazenave, J.; Drago, S.R. Red seaweed Pyropia columbina as antioxidant supplement in feed for cultured juvenile Pacú (Piaractus mesopotamicus). J. Appl. Phycol. 2019, 31, 1455–1465.
  106. Kim, C.R.; Kim, Y.M.; Lee, M.K.; Kim, I.H.; Choi, Y.H.; Nam, T.J. Pyropia yezoensis peptide promotes collagen synthesis by activating the TGF-β/Smad signaling pathway in the human dermal fibroblast cell line Hs27. Int. J. Mol. Med. 2017, 39, 31–38.
  107. Li, K.; Li, X.M.; Gloer, J.B.; Wang, B.G. New nitrogen-containing bromophenols from the marine red alga Rhodomela confervoides and their radical scavenging activity. Food chem. 2012, 135, 868–872.
  108. Zhang, Z.; Wang, F.; Wang, X.; Liu, X.; Hou, Y.; Zhang, Q. Extraction of the polysaccharides from five algae and their potential antioxidant activity in vitro. Carbohydr. Polym. 2010, 82, 118–121.
  109. Premalatha, M.; Dhasarathan, P.; Theriappan, P. Phytochemical characterization and antimicrobial efficiency of seaweed samples, Ulva fasciata and Chaetomorpha antennina. Int. J. Pharm. Biol. Sci. 2011, 2, 288–293.
  110. Lee, C.; Park, G.H.; Ahn, E.M.; Kim, B.A.; Park, C.I.; Jang, J.H. Protective effect of Codium fragile against UVB-induced pro-inflammatory and oxidative damages in HaCaT cells and BALB/c mice. Fitoterapia 2013, 86, 54–63.
  111. Pezeshk, F.; Babaei, S.; Abedian Kenari, A.; Hedayati, M.; Naseri, M. The effect of supplementing diets with extracts derived from three different species of macroalgae on growth, thermal stress resistance, antioxidant enzyme activities and skin colour of electric yellow cichlid (Labidochromis caeruleus). Aquac. Nutr. 2019, 25, 436–443.
  112. Farasat, M.; Khavari-Nejad, R.A.; Nabavi, S.M.B.; Namjooyan, F. Antioxidant properties of two edible green seaweeds from northern coasts of the Persian Gulf. Jundishapur. J. Nat. Pharm. Prod. 2013, 8, 47.
  113. Fernandes, H.; Salgado, J.M.; Martins, N.; Peres, H.; Oliva-Teles, A.; Belo, I. Sequential bioprocessing of Ulva rigida to produce lignocellulolytic enzymes and to improve its nutritional value as aquaculture feed. Bioresour. Technol. 2019, 281, 277–285.
  114. Adrien, A.; Bonnet, A.; Dufour, D.; Baudouin, 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.
  115. Mourelle, M.L.; Gómez, C.P.; Legido, J.L. The potential use of marine microalgae and cyanobacteria in cosmetics and thalassotherapy. Cosmetics 2017, 4, 46.
  116. José de Andrade, C.; Maria de Andrade, L. An overview on the application of genus Chlorella in biotechnological processes. J. Adv. Res. Biotechnol. 2017, 2, 1–9.
  117. Makpol, S.; Yeoh, T.W.; Ruslam, F.A.C.; Arifin, K.T.; Yusof, Y.A.M. Comparative effect of Piper betle, Chlorella vulgaris and tocotrienol-rich fraction on antioxidant enzymes activity in cellular ageing of human diploid fibroblasts. BMC Complement. Altern. Med. 2013, 13, 210.
  118. Kang, H.; Lee, C.H.; Kim, J.R.; Kwon, J.Y.; Seo, S.G.; Han, J.G.; Kim, B.; Kim, J.; Lee, K.W. Chlorella vulgaris attenuates dermatophagoides farinae-induced atopic dermatitis-like symptoms in NC/Nga mice. Int. J. Mol. Sci. 2015, 16, 21021–21034.
  119. Murthy, K.; Vanitha, A.; Rajesha, J.; Swamy, M.; Sowmya, P.; Ravishankar, G. In vivo antioxidant activity of carotenoids from Dunaliella salina—A green microalga. Life Sci. 2005, 76, 1381–1390.
  120. Yang, D.J.; Lin, J.T.; Chen, Y.C.; Liu, S.C.; Lu, F.J.; Chang, T.J.; Wang, M.; Lin, H.W.; Chang, Y.Y. Suppressive effect of carotenoid extract of Dunaliella salina alga on production of LPS-stimulated pro-inflammatory mediators in RAW264. 7 cells via NF-κB and JNK inactivation. J. Funct. Foods 2013, 5, 607–615.
  121. Shin, J.; Kim, J.E.; Pak, K.J.; Kang, J.I.; Kim, T.S.; Lee, S.Y.; Yeo, I.H.; Park, J.H.Y.; Kim, J.H.; Kang, N.J.; et al. A Combination of soybean and Haematococcus extract alleviates ultraviolet B-induced photoaging. Int. J. Mol. Sci. 2017, 18, 682.
  122. Rao, A.R.; Sindhuja, H.N.; Dharmesh, S.M.; Sankar, K.U.; Sarada, R.; Ravishankar, G.A. Effective inhibition of skin cancer, tyrosinase, and antioxidative properties by astaxanthin and astaxanthin esters from the green alga Haematococcus pluvialis. J. Agric. Food Chem. 2013, 61, 3842–3851.
  123. Banskota, A.H.; Sperker, S.; Stefanova, R.; McGinn, P.J.; O’Leary, S.J. Antioxidant properties and lipid composition of selected microalgae. J. Appl. Phycol. 2019, 31, 309–318.
  124. Shen, C.T.; Chen, P.Y.; Wu, J.J.; Lee, T.M.; Hsu, S.L.; Chang, C.M.J.; Young, C.C.; Shieh, C.J. Purification of algal anti-tyrosinase zeaxanthin from Nannochloropsis oculate using supercritical anti-solvent precipitation. J. Supercrit. Fluids 2011, 55, 955–962.
  125. Wu, H.L.; Fu, X.Y.; Cao, W.Q.; Xiang, W.Z.; Hou, Y.J.; Ma, J.K.; Wang, Y.; Fan, C.D. Induction of apoptosis in human glioma cells by fucoxanthin via triggering of ROS-mediated oxidative damage and regulation of MAPKs and PI3K-AKT pathways. J. Agric. Food Chem. 2019, 67, 2212.
  126. Rastogi, R.P.; Sonani, R.R.; Madamwar, D.; Incharoensakdi, A. Characterization and antioxidant functions of mycosporine-like amino acids in the cyanobacterium Nostoc sp. R76DM. Algal Res. 2016, 16, 110–118.
  127. Haimeur, A.; Ulmann, L.; Mimouni, V.; Guéno, F.; Pineau-Vincent, F.; Meskini, N.; Tremblin, G. The role of Odontella aurita, a marine diatom rich in EPA, as a dietary supplement in dyslipidemia, platelet function and oxidative stress in high-fat fed rats. Lipids Health Dis. 2012, 11, 147.
  128. Shannon, E.; Abu-Ghannam, N. Antibacterial derivatives of marine algae: An overview of pharmacological mechanisms and applications. Mar. Drugs 2016, 14, 81.
  129. Lauritano, C.; Andersen, J.H.; Hansen, E.; Albrigtsen, M.; Escalera, L.; Esposito, F.; Helland, K.; Hanssen, K.Ø.; Romano, G.; Ianora, A. Bioactivity screening of microalgae for antioxidant, anti-inflammatory, anticancer, anti-diabetes, and antibacterial activities. Front. Mar. Sci. 2016, 3, 68.
  130. Wu, Q.; Liu, L.; Miron, A.; Klímová, B.; Wan, D.; Kuča, K. The antioxidant, immunomodulatory, and anti-inflammatory activities of Spirulina: An overview. Arch. Toxicol. 2016, 90, 1817–1840.
  131. El-Sheekh, M.M.; Daboor, S.M.; Swelim, M.A.; Mohamed, S. Production and characterization of antimicrobial active substance from Spirulina platensis. Iran. J. Microbiol. 2014, 6, 112–119.
More