Next Article in Journal
A Review of Climate Change Impact Studies on Harmful Algal Blooms
Previous Article in Journal
Biochemical and Elemental Composition of Pelagic Sargassum Biomass Harvested across the Caribbean
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Seaweeds as a Source of Functional Proteins

by
Punniamoorthy Thiviya
1,
Ashoka Gamage
2,
Nalin Suranjith Gama-Arachchige
3,
Othmane Merah
4,5,* and
Terrence Madhujith
6,*
1
Postgraduate Institute of Agriculture, University of Peradeniya, Peradeniya 20400, Sri Lanka
2
Chemical and Process Engineering, Faculty of Engineering, University of Peradeniya, Peradeniya 20400, Sri Lanka
3
Department of Botany, Faculty of Science, University of Peradeniya, Peradeniya 20400, Sri Lanka
4
Laboratoire de Chimie Agro-Industrielle (LCA), Institut National de la Recherche Agronomique, Université de Toulouse, CEDEX 4, 31030 Toulouse, France
5
Département Génie Biologique, IUT Paul Sabatier, Université Paul Sabatier, 32000 Auch, France
6
Department of Food Science and Technology, Faculty of Agriculture, University of Peradeniya, Peradeniya 20400, Sri Lanka
*
Authors to whom correspondence should be addressed.
Phycology 2022, 2(2), 216-243; https://doi.org/10.3390/phycology2020012
Submission received: 12 April 2022 / Revised: 2 May 2022 / Accepted: 9 May 2022 / Published: 11 May 2022

Abstract

:
Protein is one of the major macronutrients essential in human nutrition. Protein sources especially animal sourced proteins are expensive, thus much work has been carried out to explore alternative protein sources. Seaweeds, or macroalgae, are emerging as one of the alternative protein sources. They are rich in protein with an excellent amino acid profile comparable to the other conventional protein sources. Seaweed protein contains bioactive components, such as free amino acids, peptides, lectins, and phycobiliproteins, including phycoerythrin and phycocyanin, among others. Seaweed proteins have been proved for their antihypertensive, antidiabetic, antioxidant, anti-inflammatory, antitumoral, antiviral, antimicrobial, and many other beneficial functional properties. Therefore, seaweed proteins can be a natural alternative source for functional food development. This paper discusses the compositional and nutritional aspects of seaweed protein, protein extraction techniques, functional properties of various seaweed proteins, as well as their safety for new product development and functional food applications.

1. Introduction

Traditional agriculture is no longer enough to meet the global food demand as the world population keeps rising, and as a result, protein is one of the main nutrients that will be in short supply in the near future. Therefore, alternative, unconventional protein sources and production methods are necessary to fulfil the global protein requirement and to improve the nutritional status of the global population [1,2].
Seaweeds are emerging as one of the alternative protein sources with several benefits over traditional high-protein crops in relation to nutritional value, productivity, protein yield per unit area, and the need for arable land, freshwater, and artificial fertilizer [3]. Algae are aquatic organisms categorized as microalgae and macroalgae based on their size. Multicellular macroalgae are referred to as seaweeds. Macroalgae are grown predominantly in the marine environment and classified into three main taxonomic groups based on color or pigments and their habitat: brown algae (Phaeophyta), red algae (Rhodophyta), and green algae (Chlorophyta) [4,5,6]. Seaweeds include more than 10,000 species, though only 145 species are harvested for human consumption for their flavor, texture, or culinary versatility, including Enteromorpha, Monostroma, Caulerpa, Laminaria, Undaria, Hizikia, Palmaria, and Porphyra [7]. More examples are listed below (Table 1 and Figure 1).
Edible seaweeds contain polysaccharides (starch, laminarin, floridoside, cellulose, hemicellulose, and hydrocolloids such as agar, alginate, and carrageenan), proteins, minerals (Na, Fe, Mg, Ca, I, K, Zn, F, and Se), vitamins (A, B1, B2, B9 (folic acid), B12, C, D, E, and K), antioxidants (vitamin C and E, polyphenols, sulphated polysaccharides, carotenoids, sterols, phlorotannins, catechins and proteins), polyphenols (catechins, flavonols, and phlorotannins) and low amounts of fat, which are predominantly mono and polyunsaturated fatty acids with low caloric value [5,9,12,13,14,15].
Many countries produce seaweeds, and Chile, China, Korea, and Japan are the largest producers. Seaweeds are produced in two ways: (1) wild capture (from natural marine systems) and (2) aquaculture (controlled system). According to FAO statistics, 33.3 million MT of aquatic plants, primarily marine macroalgae, were produced globally in 2018, of which 0.95 million MT (2.9%) were produced by wild capture and 32.4 million MT (about 97.1%) from aquaculture [16]. The data also show that wild capture is declining, whereas aquaculture of seaweeds has steadily increased in the last ten years [16], due to increasing concern for the protection of a high biodiversity ecosystem as they play an essential role in water purification, coastal erosion protection, carbon fixation, and nursery habitats for several species [17,18].
Currently, several protein-rich macroalgae, such as U. lactuca (Chlorophyta), U. pinnatifida, F. serratus (Phaeophyceae), Neopyropia tenera (formerly Porphyra tenera), C. crispus, and P. palmata (Rhodophyta) (17–44% of proteins), are approved by the European Food and Safety Authority for human consumption [19].
Seaweeds are widely used as human food, animal feed, dietary substitutes, gelling agents, stabilizers in food preparations, thickening agents (hydrocolloids such as agar, alginate, and carrageenan), and additives for functional foods. Some seaweeds have potential pharmaceutical and medicinal uses against cancer, allergies, diabetes, oxidative stress, inflammation, thrombosis, obesity, lipidemia, hypertension, iodine deficiency, and other degenerative ailments. Seaweeds have also been exploited as an ingredient in cosmetics (oil, peptides, amino acids, vitamins, amongst others) [20,21,22]. In addition, seaweeds can also be used in fertilizer and biofuel like biodiesel, bioethanol, biomethane, biogas, bio-oils, and hydrogen gas preparation [23,24].
Seaweeds have been used in human diet since ancient times and are traditionally consumed as raw, dried, baked, pickled form, or mixed with other food products (in soups, stews, bread, salads, and snacks) in East Asia, particularly in China and Japan [8,25,26].
Apart from satisfying hunger and providing energy and nutrition, the other functions of food are modulating physiological systems, preventing diseases, reducing health risks, and improving human well-being. In this context, the development of functional foods is becoming a primary focus of new product development [27].
Seaweeds are consumed for their functional benefits beyond their nutritional value and are highly marketed as “functional foods” or “nutraceuticals” [28,29]. Though these terms have no legal status in many countries, much of the literature defines them as “foods that contain bioactive compounds, or phytochemicals, that may benefit health beyond their nutritional value” [28].
Seaweeds are rich in protein (up to 47%) and contain bioactive compounds such as peptides, glycoproteins, lectins, mycosporine-like amino acids, and phycobiliproteins [30]. At present, seaweed proteins or purified protein fractions are rarely used in functional food development. Therefore, this paper discusses the seaweed as a potential source of functional proteins with regards to protein composition and amino acid profile, nutritional aspects of seaweed protein, protein extraction techniques, functional properties of various seaweed proteins, applications, as well as their safety in the interest of new product development or functional food applications.

2. Protein Content of Seaweeds

Seaweeds contain up to 47% of protein on a dry weight basis, which is close to the protein content of traditional protein sources such as meat, egg, soybean, and milk [31]. Generally, the protein content is low in brown algae (4–24% of dry weight) and high in red (8–47% of dry weight) and green algae (9–33% of dry weight) and can be comparable to other protein sources such as soybean (38% of dry weight) [32,33]. Table 2 summarizes the protein content of some edible seaweed in three phyla (Table 2).
The content of protein, peptides, and amino acids varies with the species, season, maturity, and environmental factors [43,51,52,53]. Moreover, differently processed samples, protocols used for protein evaluation, and nitrogen-to-protein conversion factors affect the accuracy of protein quantification, such that it is difficult to compare the results from different studies [54,55]. The crude protein content of seaweeds is widely calculated from total nitrogen content (N × 6.25). Studies showed that the traditional nitrogen to protein conversion factor of 6.25 leads to an overestimation of the protein content of seaweed and the true nitrogen to protein conversion factor for each seaweed species is found in the range of 3–5 [54,55,56,57].

3. Quality of Protein in Seaweeds

The quality of a protein can be determined by amino acid composition, proportion or ratios, digestibility, and bioavailability [29,58]. Since different methods with various standard patterns are used to assess protein quality, it is difficult to compare the published data [58].

3.1. Amino Acid Composition

The amino acid composition is essential to determine the protein quality of seaweeds and to ensure the adequate intake of essential amino acids [59]. Seaweeds contain all the amino acids required for human nutrition, especially glycine, alanine, arginine, proline, glutamic, aspartic acids, and almost all essential amino acids [60]. Table 3 shows the essential amino acid (EAA) composition of selected species of seaweeds.
The most abundant essential amino acids in seaweeds are leucine, valine, threonine, and the aromatic amino acid phenylalanine, of which leucine makes up the highest amount in most seaweeds (Table 3). The most abundant EAA in brown seaweed, H. elongata (sea spaghetti), A. esculenta (Irish wakame), and red seaweed, P. umbilicalis (nori), is leucine [67]. De Bhowmick and Hayes found that leucine is the most predominant amino acid in A. esculenta and F. serratus, and valine, followed by leucine in U. lactuca (formerly U. fasciata) and P. palmata [68].
Aspartic acid and glutamic acid together constitute a large proportion of total amino acids in many seaweeds [69,70], notably in A. nodosum (38.22% of total amino acids) [71], P. palmata (25.42%), C. crispus (38.62%) [52], Ulva spp. (formerly Enteromorpha spp.) (28.11%), Gracilaria spp. (25.82%) [72], Fucus spp. (22–44%), U. rotundata (32%), and U. rigida (26%) [37]. The abundance of these amino acids is responsible for the special flavors and tastes of seaweed, and glutamic acid is the main component in the taste sensation of “umami” [5,64].
Various factors, such as species, preservation method, extraction method, seasonal variation, and the environment in which they grow, influence the amino acid composition of seaweeds [29,53,73,74].

3.2. Amino Acid Ratio

Proportion or ratios of amino acids, such as the ratio of EAA to non-essential amino acids or total amino acids (EAA/NEAA or EAA/total AA), essential amino acid index (EAAI), and amino acid score (AAS), have been used to assess the protein quality of seaweeds in various studies [29].
Based on Table 3, seaweeds consist of 37.7–48.4% of EAA, and the ratio of EAA to NEAA is in the range of 0.65–1.61. Though red seaweed generally contains a higher amount of protein (Table 2), the concentration of EAAs per gram of protein or the ratio of EAA/NEAA is lower in red seaweed (Table 3) [62].
Almost half of the total amino acids (about 40–50%) in seaweeds consists of EAA [42], which is close to the value of soya (39%) and egg protein (47%) [31]. Furthermore, the ratio of EAA/NEAA is used to evaluate the distribution of amino acids in seaweed proteins [29]. The EAA/total AA ratio or EAA percentage for three brown seaweeds, A. nodosum, F. vesiculosus, and Bifurcaria bifurcata, were reported as 38.87, 40.99, and 39.99%, respectively [36]. The under-exploited edible seaweeds in India, such as Acanthophora spicifera, G. edulis (Rhodophyta), Padina gymnospora (Phaeophyceae), U. lactuca (formerly U. fasciata), and Ulva flexuosa (formerly Enteromorpha flexuosa) (Chlorophyta) were reported to contain 41–50% (0.41–0.50) of EAA in total AA with an EAA/NEAA score of 0.72–1.02 [75]. Similar results also have been recorded for Hypnea japonica, Hypnea charoides (Rhodophyta), U. lactuca (formerly U. fasciata) (Chlorophyta) (36.2–40.2%) [76], and P. palmata (Rhodophyta) (40.9–42.1%) [55]. Norziah and Ching also stated that the ratio of EAA to total AA is 0.4 in Gracilaria changgi [77]. The EAA/total AA and EAA/NEAA ratios for brown seaweed Sargassum polycystum were reported as 0.5 and 1.0, respectively [78]. Rosemary et al. reported that red seaweeds, G. corticata and G. edulis, have a good EAA/NEAA ratio (0.62 and 1.19, respectively) and EAA/total AA ratio (0.29 and 0.54, respectively) [46].

3.3. Amino Acid Score

Amino acid score (AAS) or chemical score is calculated using the following equation:
AAS   ( % ) = mg   AA   in   1   g   of   the   seaweed   protein   tested mg   AA   in   1   g   reference   protein × 100
The amino acid composition in the tested sample is compared with a reference protein, the amino acid requirement pattern defined by the FAO/WHO/UNU for children (3–10 years of age) or adults [61], or the respective amino acid content for eggs or leguminous plants (soybean) [42,57,77]. The AAS evaluates the actual abundance of individual EAA in food material and relates it to dietary requirements or a reference protein, and thereby, it is also possible to determine the limiting amino acid. The lowest score (<0.100) obtained for the essential amino acids in a tested protein is the “most limiting amino acid”, which means the concentration of the corresponding amino acid is lower than the reference standard [40,79].
The sulfur amino acids (methionine and cysteine), lysine, and tryptophan are often limiting amino acids in seaweed protein [80]. This composition pattern varies with the species. In comparison, leucine and isoleucine in red algae, and methionine, cysteine, and lysine in brown algae, are often deficient amino acids [40,52].
Limiting amino acids in H. japonica, H. charoides, and U. lactuca (formerly U. fasciata) are methionine (0.24–0.79 AAS) and lysine (0.68–0.80 AAS) [65]. Machado et al. identified methionine (58.4–93.4% AAS) as limited EAA in Porphyra dioica, P. umbilicalis, A. vermiculophyllum (previously G. vermiculophylla) (Rhodophyta), and U. rigida (Chlorophyta) [29]. The most limiting amino acid in U. australis (formerly U. pertusa) and U. intestinalis (formerly E. intestinalis) was lysine, followed by leucine, despite the fact that leucine was present at a higher amount in both species (49.7–52.0 mg/g protein) [64].
When egg protein is used as a reference protein, seaweeds have better sources for isoleucine, threonine, and valine (>1.00 or 100% EAA score), and are deficient in methionine (0.5 or 50%), whatever the phylum considered [42]. In another study, lysine was identified as a limiting amino acid in G. changgi [77].
Seaweeds can be a good source of high-quality protein as it consists of a high concentration of EAA (40–50% of total amino acids) with an excellent EAA profile close to that of egg protein and a higher AAS than other plant-based protein, except soy, which has an AAS of 1.00 [11]. Therefore, seaweeds can be an alternative source of traditional protein in human food and animal feeds.

3.4. Digestibility and Bioavailability

Protein quality does not solely depend on the amino acid profile. Even with an excellent amino acid profile, the protein may have lower nutritional value if the digestibility is low due to poor bioavailability [81]. Therefore, the bioavailability of proteins is an essential factor in determining protein quality, and can be described as the degree to which amino acids or small peptides from a test protein consumed by a living organism are finally transported across the intestinal membrane and into the body [82]. As bioavailability includes digestibility and absorption mechanisms [82], studies examining the bioavailability of protein are required to incorporate in vivo experiments.
Using animal assay, in vivo protein digestibility can be determined by measuring the amount of nitrogen absorbed (the difference between the nitrogen intake and nitrogen recovered from the feces) relative to the nitrogen intake [83]. However, compared to the in vivo method, the digestibility of seaweeds has been widely studied based on rapid and cost-effective in vitro methods [65,84,85], which also give information about the bioavailability of food proteins [82]. In vitro digestibility relies on several proteolytic enzymes such as pepsin, pancreatin, trypsin, chymotrypsin, or a mixture of these enzymes [50,65,86,87]. This enzymatic approach is based on in vitro simulations of human digestion, and most methods use mammalian gastric and/or pancreatic and intestinal enzymes in the assay [83,86,87].
Red seaweed has the highest digestibility among the three phyla [65,85], whose values are comparable with some plant sources, including grains (69–84%), legumes (72–92%), fruits (72–92%), and vegetables (68–80%), and slightly lower than animal protein sources (casein and whey) [1,85]. Table 4 shows the relative in vitro digestibility of selected seaweeds using a multi-enzyme assay (porcine pancreatic trypsin, bovine pancreatic chymotrypsin, and porcine intestinal peptidase) at pH 8.0 and 37 °C.
However, the values reported for in vitro digestibility of seaweeds are highly variable depending on the particular assay used [37,85,86]. Further, the in vitro digestibility of seaweed proteins can differ according to the species and seasonal variations in glycoprotein and antinutritional factors such as phenolic molecules or polysaccharides [37,65]. This lack of consistency makes the resulting data challenging to compare.
In vitro digestibility (using multi-enzyme assay; pepsin, trypsin, and chymotrypsin at neutral pH) of A. esculenta, F. serratus, F. vesiculosus (Phaeophyceae), U. lactuca (formerly U. fasciata) (Chlorophyta), P. palmata, and Asparagopsis taxiformis (Rhodophyta) were quite similar (0.77–0.82) [68]. The digestibility of U. pinnatifida (brown) was recorded as 17% and 66.6% when using pepsin (acidic pH, 37 °C) and pancreatin (pH 7.6, 37 °C), respectively [37,88]. When using the combination of pepsin and pancreatin, the digestibility of P. columbina (formerly P. columbina) was reported as 74.3% [50]. Table 5 shows the digestibility of selected seaweeds using various assays.
Though the seaweeds contain a higher amount of protein, the inhibitory effect of macroalgal compounds like glycoprotein and other antinutritional factors (phenolic compounds or polysaccharides) reduces the digestibility [1,37].
Polysaccharides, notably soluble fibers (xylan and carrageenan) and their interaction with proteins or proteolytic enzymes, reduce protein hydrolysis in seaweed [84,85]. Fleurence also suggested that the glycosylation level of the protein fraction or glycoprotein content may decrease the rate of hydrolysis by enzymes such as trypsin and chymotrypsin and influence the digestibility of Ulva armoricana protein [37]. Further, Marrion et al. reported that the presence of higher soluble fiber reduced the digestibility of P. palmata and G. longissima (formerly G. verrucosa) [84].
Oxidized phenolic compounds may react with amino acids and proteins, thereby inhibiting the activity of proteolytic enzymes. A strong negative correlation between total phenolic content and in vitro digestibility of seaweed protein has been reported in various studies [65,85]. Typically, brown seaweed contains a higher amount of phenolic compounds, including catechins, flavanols, and phlorotannins, which greatly influence protein digestibility [1].
However, these in vitro studies only give an approximation of the true protein digestibility. Therefore, biological evaluation using human and animal feeding studies (in vivo protein digestibility) is required for an accurate prediction of the nutritional value of seaweed protein.

4. Protein Extraction Methods

Seaweed has poor protein digestibility in its raw, unprocessed form because of its complex cell wall, which poses a physical barrier in the absence of digestive enzymes in the gastrointestinal tract, and greatly emphasizes the need for protein extraction techniques to improve their digestibility [52]. Protein can be extracted from dried seaweed powders using conventional or classical methods, including physical, chemical, enzymatic methods, and other novel methods [42,71], then the protein can be collected by centrifugation or filtration. The dissolved protein can be obtained by various recovery and purification techniques, such as ultrafiltration, chromatography techniques, dialysis, and/or precipitation using ammonium sulphate followed by centrifugation [89,90,91]. High-purity proteins are not required to produce regular food and feed but are essential for functional food development. Chromatography is the main purification technique that uses molecular exclusion, ion exchange, affinity, and hydrophobic interactions [92]. The extracted protein can be preserved by preconcentration and drying techniques (freeze-drying and oven drying at 40 °C) [89,90,91]. Figure 2 illustrates the basic steps involved in protein extraction from seaweed.
Physical cell disruption methods include grinding followed by aqueous treatment, osmotic shock, or high shear force, and the extraction is carried out based on the hydrophilic property of most proteins. Chemical methods include acidic (HCl) and alkaline (NaOH) treatment, which significantly improves the solubility and extraction of highly water-insoluble proteins from seaweeds. However, these two methods are relatively time-consuming, and the efficiency of extraction (29–59% of protein content) is limited [1,93]. Proteins in seaweed species are bound to non-protein components such as polysaccharides and polyphenols [79]. Therefore, cell disruption techniques are required to enhance the efficiency, extraction rate, and yield. Enzymes such as polysaccharidases (cellulase, hemicellulose, xylanase, k-carrageenase, β-agarase, β-glucanase, amylases, arabinase) or proteases can be applied before protein extraction in order to degrade polysaccharides, such as carrageenans, alginates, ulvans, xylans, galactans, cellulose, fucoidan and laminarin, and improve the protein yield by up to 67% [1,71,94].
Many non-conventional novel protein extraction techniques have been used for protein extraction: microwave-assisted extraction, supercritical fluid extraction, ultra-high-pressure extraction, pressurized liquid extraction, pulsed electric field, and ultrasound-assisted extraction [1,71,95].
The amount of protein that can be extracted from seaweeds depends on various factors, such as the species, seasonal variation, pre-treatment, the combination of extraction methods, preservation method, extraction method, and its processing parameters, such as extraction temperature, time, and pH [3,71,96]. The most influencing factor in seaweed protein extraction is the complex rigid cell wall, and its composition varies with species. In addition, seaweed proteins are bound to non-protein components such as polysaccharides (alginates, agar, carrageenan) and polyphenols that influence protein extraction efficiency [91,97]. Seaweeds are seasonal, and fresh seaweeds can deteriorate quickly after harvest. Therefore, seaweeds are preserved by freezing or freeze-, sun-, vacuum-, or air-drying at different temperatures to ensure year-round availability [3]. The methods used for protein extraction and the processing parameters also have an effect on the amino acid profile [74,98].
Compared to the physical method (sonication), the pH-shift method (using NaOH and HCl) recovered more protein from P. umbilicalis and Saccharina latissima, and a higher EAA percentage (42.2–42.6%) from P. umbilicalis, S. latissima, and U. lactuca (formerly U. fasciata) [99]. When compared to the high-pressure processing and autoclave method, the classical method (cell lysis induced by osmotic shock and sonication) produced a higher protein yield for C. crispus (35.2%), Fucus vesiculosis (35.1%), and A. esculenta (18.2%) with a good EAA profile [52]. The brown seaweed Macrocystis pyrifera (74.6%) yielded significantly more protein than the red seaweed Chondracanthus chamissoi (36.1%) when using enzyme-assisted extraction with cellulase [91]. A combination of various protein extraction methods has been used to obtain higher protein yields. Sequential extraction using acid treatment followed by alkaline treatment has also increased protein extraction (59.76%) [71]. For P. palmata, up to 90% of the protein was extracted using a combination of enzymes followed by the N-acetyl-L-cysteine-assisted alkaline extraction method [31].

5. Functional Properties of Seaweed Proteins and Their Role in Health

Functional foods can be defined as foods and food components that provide a health-promoting benefit beyond basic nutrition and energy [28]. “Let food be your medicine and medicine be your food” is a popular quote by the father of medicine, Hippocrates. Many studies have confirmed a direct relationship between diet and health, and the regular inclusion of functional ingredients in has an impact on the quality of life [100]. Seaweeds contain several bioactive compounds, including polysaccharides, polyphenols, lipids, polyunsaturated fatty acids (PUFAs), sterols, proteins, dietary fiber, pigments, and vitamins [101,102]. Several studies have revealed that the seaweeds are an excellent source of various proteins (amino acids, peptides, phycobiliproteins, and lectins) with interesting biological properties, such as antihypertensive, antioxidant, antidiabetic, anti-inflammatory, antitumoral, antiviral, and antimicrobial [20,32,103]. Table 6 summarizes the bioactive compounds and their functional properties for selected seaweeds.

5.1. Amino Acids

Amino acids are building blocks of polypeptides and proteins, and the amino acid composition varies with seaweed species. Amino acids serve as essential precursors for the synthesis of low molecular-weight substances (e.g., NO, polyamines, glutathione, creatine, carnitine, carnosine, thyroid hormones, serotonin, melanin, melatonin, and heme) with enormous physiological roles, including regulating nutrient transport and metabolism, cell-to-cell communication, gene expression, protein phosphorylation, antioxidative defense, immune function, reproduction, lactation, fetal and postnatal growth and development, tissue regeneration, neurotransmission, acid-base balance, homeostasis, intestinal microbial growth, and metabolism, among many others [134].
In general, glycine, alanine, arginine, proline, aspartic acid, and glutamic acid make up a larger portion, and cysteine, methionine, and tyrosine are found in lower concentrations in seaweeds [58]. Supplementation with amino acids has a beneficial effect on disease management, e.g., methionine for patients with multiple sclerosis; arginine has a neuroprotective effect after brain ischemia injury and in infertility; histidine improves insulin sensitivity in hyper-insulinemia; glycine alleviates liver and lung injury; tryptophan improves sleep disorders and depression [29,134]. Glutamic acid plays an important role in key physiological functions, including maintaining brain function and mental activity. Aspartic acid helps to initiate important metabolic pathways like the Krebs and urea cycles [58]. However, elevated amino acid levels and their products, such as ammonia, homocysteine, and asymmetric dimethylarginine, are pathogenic factors for neurological disorders, oxidative stress, and cardiovascular disease. Therefore, it is vital to maintain an optimal amino acid balance in the diet and circulation for whole-body homeostasis [134].

5.2. Peptides

Peptides that are 2–20 amino acids in length can be linear, cyclic, depsipeptides, dipeptides (carnosine, almazole D), tripeptides (glutathione), pentapeptides (galaximide), hexapeptides, oligopeptides, and phycobiliproteins [32,79]. These isolated bioactive peptides have hormone-like properties that are inactive within the parental proteins, but become activated upon release during fermentation or hydrolysis [1,128]. Based on their structural properties, amino acid composition, and sequences, they can display a wide range of biological functions, including antihypertensive (ACE inhibitory), antioxidant, antidiabetic (DPP-IV inhibitory, α-amylase inhibitory), appetite suppression, antitumoral, antimicrobial, antiviral, opioid agonistic, immunomodulatory, prebiotic, opioid, mineral binding, tyrosinase inhibitory, anticoagulatory, anti-thrombotic and hypocholesterolemic effects [1,27,32,135,136].
Hypertension is one of the major risk factors for cardiovascular disease (CVD) [27,137]. Renin and ACE are the two key enzymes in the renin-angiotensin system (RAS), which regulates peripheral blood pressure. ACE catalyzes the conversion of angiotensin-I to a potent vasoconstrictor, angiotensin-II, and degrades the vasodilator peptides bradykinin [121,138]. Thus, inhibition of ACE is one of the key therapeutic approaches in the management of hypertension (Figure 3) [27].
To date, a number of ACE inhibitory or antihypertensive macroalgal peptide hydrolysates have been identified [137]. Paiva et al. revealed that ACE inhibitory peptides from U. rigida have potential therapeutic benefits for the prevention and/or treatment of hypertension and its related diseases [121]. ACE inhibitory peptides have also been reported in P. columbina (formerly P. columbina), P. palmata, N. tenera (formerly P. tenera), N. yezoensis (formerly P. yezoensis, S. chordalis, M. japonica (Rhodophyta), S. fusiforme (formerly H. fusiformis), U. pinnatifida (Phaeophyceae), Ulva prolifera (formerly Enteromorpha prolifera), and U. intestinalis (formerly E. intestinalis) (Chlorophyta) [27,107,118,119,122,137,139].
Furthermore, the recent outbreak of SARS-CoV-2 (or 2019-nCoV) responsible for the COVID-19 pandemic, enters host cells through an interaction between the spike viral protein and angiotensin-converting enzyme 2 (ACE 2) [140]. ACE inhibitory peptides with antiviral activity in edible seaweeds (U. pinnatifida, S. fusiforme, Porphyra spp.) could exert a protective effect against COVID-19 by reducing the dominance of the ACE/Ang II/ATR1 axis [141].
A few studies have reported the antidiabetic potential of seaweed protein/peptides that inhibit the α-amylase, α-glucosidase, and DPP-IV [112,113]. One therapeutic approach for type 2 diabetes mellitus (T2DM) management is to lower blood glucose levels by inhibiting the key enzymes involved in intestinal carbohydrate digestion (α-amylase and α-glucosidase). Two α-amylase inhibitory peptides have been identified in proteolytic enzyme hydrolysates of seaweed laver (Porphyra spp.) that can prevent postprandial hyperglycemia [142]. Another, newer, therapeutic approach for T2DM is to inhibit DPP IV as an insulin regulatory strategy. Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are the two incretin hormones that stimulate glucose-induced insulin secretion, inhibit postprandial glucagon release, and delay gastric emptying, which results in lower blood glucose level. DPP-IV inactivates GLP-1 and GIP, resulting in the loss of their insulinotropic potential in vivo. Hence, DPP-IV inhibitors prevent the degradation of GLP-1 and GIP and enhance its insulinotropic effects, and thus, can be used in the management of T2DM [113,143]. Figure 4 illustrates the simplified mechanism of DPP-IV inhibitors and antidiabetic activity.
Seaweeds can be a natural source of DPP-IV inhibitors. Studies have reported that protein hydrolysates of P. palmata have DPP-IV inhibitory activities that are useful for the management of T2DM [111,144] and obesity [111]. Oral administration of P. palmata protein hydrolysate derived from Alcalase and Flavourzyme reduced food intake by streptozotocin-induced diabetic mice and showed antihyperglycemic effects [112].
Reactive oxygen species (ROS) contribute to the development of chronic diseases, including cardiovascular diseases, cancer, diabetes mellitus, cataracts, and neurodegenerative disorders [116]. ROS includes free radical species, such as superoxide anions, hydroxyl radicals, and singlet oxygen, and non-radical species, such as hydrogen peroxide (H2O2), generated during the metabolic process [145]. The antioxidant activity of bioactive peptides is attributed to the hydrophobicity of valine, leucine, isoleucine, glycine, methionine, proline, and alanine and some aromatic amino acids (tyrosine, histidine, tryptophan, and phenylalanine) [116]. They exert a protective effect on the body by binding free radicals and other reactive oxygen compounds. Furthermore, the regulation of oxidative stress is an essential factor in tumor development and anticancer therapies [146]. Protein hydrolysates or peptides and amino acids exhibit multiple antioxidant properties. Two antioxidant peptides, such as carnosine and glutathione, generally present in high concentrations in animal muscle, have been found in seaweed [116]. Antioxidant peptides have been isolated from several species of macroalgae, including Scytosiphon lomentaria [147], Ecklonia cava, Sargassum coreanum (Phaeophyceae) [148], P. palmata [49], and P. columbina (formerly P. columbina) (Rhodophyta) [118]. Antioxidant and anticancer bioactivity have also been reported in Sri Lankan seaweed, and the highest value was reported for Caulerpa racemose [149]. N. yezoensis (formerly P. yezoensis), G. pusillum, and many other seaweed species have been studied for their antioxidant properties [130].
Antimicrobial peptides have been identified in S. longicruris (formerly L. longicruris) against S. aureus, and cyclic depsipeptide from Bryopsis spp. demonstrated activity against M. tuberculosis [104,119]. Protein hydrolysates from P. columbina (formerly P. columbina) also have immunosuppressive, antihypertensive, and antioxidant capacities [50].
Furthermore, inhibition of platelet-activating factor acetyl-hydrolase (PAF-AH) has been reported for peptides derived from P. palmata that could prevent high blood pressure and atherosclerosis [114]. PAF-AH plays an active role in atherosclerotic development and progression [114]. PAF-AH is thought to be involved in the generation of pro-inflammatory mediators, such as lysophosphatidylcholine (LPC) and oxidized non-esterified fatty acids (NEFA) [144,146]. In addition, macroalgae peptides from different species display many other biological activities (Table 6).

5.3. Lectins

Lectins and phycobiliproteins are two groups of functionally active proteins in seaweeds [150]. Lectins are proteins, glycoproteins, or hemagglutinin proteins that reversibly bind specific mono- or oligo-saccharides [126,128]. Lectins have been found in red and green algae, such as Eucheuma spp., Solieria filiformis, Enantiocladia duperreyi, Pterocladiella capillacea, Gracilaria cornea, Gracilaria ornate, Bryothamnion spp., M. amakusaensis (formerly E. amakusaense) (Rhodophyta), Ulva spp. (formerly Enteromorpha spp.), and C. fragile (Chlorophyta) [32,60,128]. Lectins are involved in numerous biological processes, such as host-pathogen interactions, intercellular communication, recognizing and binding carbohydrates, induction of apoptosis, metastasis, and cell differentiation in cancer cells [128,131]. These proteins also have other bioactive properties, including antibiotic, antibacterial, antifungal, anti-inflammatory, mitogenic, cytotoxic, antinociceptive, anticancer, fibroblast, human platelet aggregation inhibition, antiviral, and anti-human immunodeficiency virus (anti-HIV) activities [30,38,60,128,151].
Lectins from red algae Alsidium triquetrum (formerly Bryothamnion triquetrum), P. capillacea, Hypnea cervicornis, S. filiformis, and green seaweed C. cupressoides have demonstrated anti-inflammatory activities in different studies [152]. Lectin is the only seaweed protein reported as an antibacterial in the literature [119,150]. The lectins found in Alsidium seaforthii (formerly Bryothamnion seaforthii) and Hypnea musciformis show bactericidal activity, especially inhibiting the growth of S. aureus and P. aeruginosa [153]. Lectin extracted from red seaweed showed antibacterial activity against six pathogenic Gram-negative species, including Serratia marcescens, Salmonella typhi, Klebsiella pneumoniae, Enterobacter aerogenes, Proteus spp., and P. aeruginosa [60]. The lectin isolated from B. seaforthii has a pro-healing property responsible for accelerating the healing of skin wounds [154]. Because of these antimicrobial properties, lectins are used to treat many pathologies such as cancer and chronic bacterial diseases, chronic otitis, tonsillitis, cystic fibrosis, periodontal diseases, and urinary tract infections [153].
Lectins have the ability to precipitate glycoprotein and agglutinate red blood cells [30,60]. Further, lectins have displayed antiviral effects against human immunodeficiency, hepatitis C, and SARS-CoV viruses, mainly by preventing entry of the virus into host cells, and thereby, their propagation [125]. Griffithsin, a highly potent broad-spectrum antiviral lectin from Griffithsia spp. has an antiviral effect against HIV [155], SARS-CoV, and Middle East respiratory syndrome coronavirus (MERS-CoV) [156]. Lectins have also been widely studied for their antiviral activity against SARS-CoV-2 (or 2019-nCoV)since they can inhibit coronavirus infectivity by specifically binding to the spike glycoprotein. Glycoproteins, especially the spike protein of SARS-CoV-2, are involved in cell adhesion and invasion, morphogenesis, and modulation of immune response processes. This spike protein mediates viral adhesion through human ACE 2. Lectins that bind SARS-CoV-2 spike protein via their ability to recognize glycans can inhibit the adhesion of coronavirus and impair the initial steps of viral pathogenesis [157]. Many studies have highlighted lectins from seaweeds and their potential antiviral therapeutic activity against SARS-CoV and SARS-CoV-2 (COVID 19) [124,158]. Thus, seaweed lectins should be considered when developing new antiviral approaches because of their antiviral properties.

5.4. Phycobilliproteins

Phycobiliproteins are the only water-soluble algal pigments in red seaweeds [32]. Phycobiliproteins are the most abundant proteins in red seaweeds, representing nearly 50% of the total protein content [159]. Phycobiliproteins are grouped into the following four groups: phycoerythrin (purple), phycocyanin (blue), phycoerythrocyanins (purple), and allophycocyanin (bluish-green), whereas phycoerythrin is the main pigment [30,160]. Phycobiliproteins have been reported in many species, including Porphyra spp. [160], Gracilaria canaliculate (formerly Gracilaria crassa) [161], P. palmata [162], and G. tikvahiae [131]. Phycoerythrin has been reported in G. gracilis [163], Grateloupia turuturu [164], G. pusillum, and Rhodymenia pseudopalmata [30]. Furthermore, extraction of phycocyanin has been reported for C. crispus, G. gracilis, and Gelidium amansii with many bioactivities, including anticancer activity, anti-inflammatory effect, antioxidative, and anti-irradiative effects [165].
Phycobiliprotein has become popular for its biological activities, including antioxidant, ACE inhibitory, antitumoral, antidiabetic, immunomodulating, anti-inflammatory, liver-protecting, antiviral, anticancer, antiatherosclerosis, antihyperlipidemic activities, lipase activity inhibitor, serum lipid reducing agent, and obstructing absorption of environmental pollutants into the body [110,128,164,166]. Other than these, it is also beneficial for preventing or treating gastric ulcers and neurodegenerative diseases caused by oxidative stress (Alzheimer’s and Parkinson’s) due to their antioxidant effects [20,32].
Phycocyanin improves the immune system and has several other bioactivities, including in vitro anticancer activity, chemotherapy sensitiveness, photosensitized tumor suppressor activity, anti-inflammatory effects, antioxidative, anti-irradiative, and neuroprotective effects [165].

5.5. Free Amino Acids

The free amino acid fraction in seaweeds mainly consists of taurine, alanine, ornithine, citrulline, hydroxyproline, and aminobutyric acid [131]. Taurine content varies with the species. Red algae contains taurine in high concentrations, however it is rarely found in green and brown algae [40,167]. Seaweeds such as N. yezoensis (formerly P. yezoensis), N. tenera (formerly P. tenera), Gloiopeltis tenax, Gloiopeltis furcate, Gracilaria textorii, A. vermiculophyllum (formerly G. vermiculophylla) (Rhodophyta), U. pinnatifida, S. japonica (formerly L. japonica), and Sargassum confusum (Phaeophyceae) contain a high amount of taurine [10,129,167,168] and can be used in functional foods that contain naturally occurring taurine [40,167,169]. Taurine plays an important role in physiological functions such as bile-acid conjugation, retinal and neurological development, osmoregulation, antioxidant, a modulator of intracellular calcium level, and immune function [169]. In addition, taurine acts as an antioxidant and protects against the toxicity of various heavy metals, including lead and cadmium, by preventing their absorption in the stomach [128]. Taurine also has antihypertensive and hypocholesterolemic activities by reducing the secretion of serum lipids and apolipoprotein (very low-density lipoprotein, VLDL, and intermediate-density lipoproteins, IDL) [38,170].
In addition to taurine, macroalgae contain unusual amino acids, such as laminine, kanoids (kainic and domoic acid), and mycosporine-like amino acids with bioactivity [38,131]. Many macroalgae species, including Digenea simplex, C. armata, P. palmata, among others, contain kanoid amino acids (kainic and domoic acids), and extraction from D. simplex has been commercialized [127,131]. Kanoid amino acids are reported to have insecticidal, neuroexcitatory and anthelmintic properties [131]. In Japan, D. simplex and C. armata extracts contain kanoids and have been used for centuries as anthelmintic agents to treat ascariasis (a disease in humans caused by the parasitic roundworm). They also act as central nervous system stimulants and assist in neurophysiological disorders such as Alzheimer’s disease, Parkinson’s disease, and epilepsy. However, they become neurotoxins when safe levels are exceeded [38]. Laminine, a choline-like basic amino acid, has been isolated from S. angustata (formerly L. angustata) and C. armata, and can depress the contraction of excited smooth muscles and exert a transitory hypotensive effect [131].

6. Applications

Nowadays, consumers are more health-conscious, and as a result, they seek more and more natural sources to treat or prevent health issues. The functional food market has been growing over the years and can be categorized as fortified, enriched, altered, or enhanced commodities (naturally enhancing one of the components) [171,172,173]. In this context, the food industry has emerged to develop and market a diverse group of functional food products using natural ingredients [128].
Over the last few decades, seaweeds have been widely studied for their bioactive compounds. Hydrocolloids like agar, alginates and carrageenan, and alkaloids, carotenoids, polyphenols, terpenes, tocopherols, laminarin, and fucoidan, among many others, have all been used as functional ingredients in bakery, dairy, fish, meat, and vegetable-based products [11,32]. Seaweed contains a considerable amount of protein that can be used to fulfil nutritional requirements or to treat malnutrition [9]. N. tenera (formerly P. tenera), N. yezoensis (formerly P. yezoensis), P. columbina (formerly P. columbina), and P. umbilicalis, Gracilaria spp., Sargassum wightii, Eucheuma spp., and many other species have been used in different varieties of seaweed-based foods and beverages such as wine, instant soup, noodles, jam, jelly, tea, porridge, soft cheese, sausages, among others, to enhance their nutritional value [32,174].
Wheat flour used for making pasta and noodles has relatively low protein content (10% to 15%) and lacks some EAA, such as lysine, threonine, and methionine [175]. In such cases, seaweed or macroalgal supplementation can improve the protein quality of the bakery foods, pasta, and cereals due to their essential amino acid content. Bakery, pasta, and cereal products are widely consumed food products, which can be used as food vehicles for the delivery of bioactive compounds. In Wales, United Kingdom, Porphyra species are traditionally used to make a dish known as laverbread. Wakame (U. pinnatifida)-incorporated pasta was reported to have improved bio-functional properties and enhanced interactions between starch granules and the protein matrix [176]. Seaweeds such as Caulerpa racemosa, U. lactuca (formerly U. fasciata), Chnoospora minima, P. gymnospora, and A. spicifera have an excellent amino acid profile rich in lysine and methionine. Hence, they are utilized in highly nutritive food formulations with cereals and legumes (seaweed-based bread, biscuits, and idly) to balance the amino acid profile and provide a balanced diet to the individual [174]. However, consumer acceptance of seaweed-containing foods in the Western world is limited due to undesirable sensory characteristics. Whole-wheat bread containing C. crispus and A. nodosum was acceptable at lower concentrations (2% and 4%, respectively) with no significant changes in protein content [12].
Few studies mention the application of seaweed protein in the development of functional food. Proteins, peptides, and amino acids derived from seaweeds are used as nutraceuticals but are best utilized during the dietary consumption of seaweeds [177]. Food applications of seaweed-derived proteins, peptides, and amino acids have become popular in the last few decades since most of these components have anti-inflammatory, antioxidant, antitumor, anti-aging, and protective activity [60], although at present, macroalgal proteins or purified protein fractions are rarely used as ingredients in the food industry [10].
S. japonica (formerly L. japonica) has been used as a flavor enhancer in Japanese cooking for many years [40]. The high content of free amino acids such as glutamic acid, aspartic acid, alanine, and glycine has been described as responsible for the unique flavor of seaweeds [29]. Free glutamic acid and aspartic acid (to some extent) are the main components in the taste sensation of ‘umami’ [178], whereas glycine and alanine give a sweet flavor [77]. The enzymatic (bromelain) hydrolysis of protein from Gracilaria fisheri yields a roasted seafood-like flavor that can be used as a flavoring agent in the food industry [179].
Mainly in Japan, several seaweed-derived peptides containing functional foods are currently commercialized and approved as Foods for Specified Health Uses (FOSHU). Foods containing peptides from nori and wakame are approved for antihypertensive claims [146]. Peptides derived from P. palmata protein can be incorporated into bread to enrich its renin-inhibitory capacity without affecting the texture or sensory properties of the bread to a large degree [180].
Currently, considerable interest has been mounting for finding alternative sources of synthetic antioxidants for application in food [148]. Synthetic antioxidants/preservatives and other bioactive additives are highly used in processed food. These can oxidize functional components in the food, resulting in increased oxidative stress that can lead to hypertension and cardiovascular diseases. Natural bioactive compounds extracted from natural commodities like seaweeds can be used to replace these synthetic additives [174]. P. palmata-derived peptides have potential applications as health-promoting ingredients and food preservatives because of their antioxidant activity [116].
Various polysaccharides are present on cell surfaces, and as a result, many cells, including microbes, lymphocytes, tumor cells, and erythrocytes, are selectively agglutinated by lectins. Lectins have been used as specific probes in immunological, cell biology, membrane structure, biomarker, drug delivery/targeting, mitogenic, antitumor, and cancer studies. Lectins specifically agglutinate human blood groups, which has led to their use in assays for blood typing [127,151].
The phycobiliproteins are an interesting group of high-value (approximately $5000/g) macroalgal products [181]. Despite their lower stability in heat and light, phycobiliproteins are used as natural colorants for food (phycocyanin) and cosmetics (phycocyanin and phycoerythrin) [60,182]. Phycobiliproteins are used as colorants in many food products, including fermented milk, ice creams, desserts, milkshakes, jelly gum, and coated soft candies [60,183].
R-phycoerythrin is currently derived from a species of Porphyra [181] and can be used as a food colorant [159]. Phycobiliproteins from G. longissima (formerly G. verrucosa), U. lactuca (formerly U. fasciata), and S. wightii can be used as natural food colorants for jelly without a change in color for more than three months [184]. Phycoerythrin, the most abundant phycobiliprotein in G. turuturu, has many applications, such as natural colorant, fluorescent probe, antioxidant, antitumoral, and antidiabetic compounds [164]. In general, phycobiliproteins are used as labels or markers in immunolabeling experiments, fluorescence microscopy, and diagnostics. R-phycoerythrin is a powerful and highly sensitive fluorescent reagent used as labels or markers for antibodies, receptors, and other biological molecules in a fluorescence-activated cell sorter [60,183,185].
Only a few studies mention the applications of protein derived from seaweeds in functional food products, due in part to the fact that acceptability, toxicity, allergenicity, and microbial studies are required before they can be safely utilized.

7. Safety

Although seaweeds have gained much interest in food industrial applications based on their nutritive values, several factors limit their widespread usage, including the accumulation of toxic heavy metals, allergenicity (phycobiliprotein and phycolectins in red seaweeds), contamination with pathogens, and toxic synthetic compounds such as cyanotoxins (neurotoxin and hepatotoxin), amino acids (kainic acid), and radioisotopes [28]. Therefore, it is essential to determine their toxicological profile for the safety of the consumers.
Heavy metals such as arsenic, cadmium, and mercury, and microorganisms such as Salmonella have been identified as major hazards associated with seaweeds [169,186]. Seaweed absorbs heavy metals from seawater depending on various factors, such as species, location, season, wave exposure, temperature, salinity, light intensity, pH, nitrogen availability, and the age of the plant [187]. Chen et al. identified ten metals and metalloids—Al, Mn, As, Cu, Cr, Ni, Cd, Se, Pb, and Hg—in 295 dried brown and red seaweeds [188]. Generally, heavy metal concentrations in seaweeds are found below the toxic level. However, bioaccumulation of arsenic, lead, and cadmium beyond a hazardous level is the main risk for seaweed harvested from the wild and can result in allergies, hyperpigmentation, and cancer. Arsenic is a known carcinogen, and commonly consumed seaweeds were reported to contain high levels of arsenic, primarily in organic forms [189]. Iodine, a component of thyroid hormones, plays a role in metabolism, and iodine deficiency causes goiter and hypothyroidism. Seaweeds such as L. digitata and S. japonica (formerly L. japonica) are a good source of iodine and can be used to prevent goiter. However, iodine-rich products such as dried seaweed can cause excessive iodine intake and induce hyper- or hypothyroidism [186,189].
Microplastics have become an emerging environmental pollutant because of their persistence, ubiquity, toxic potential, and distribution in various environments, including soil, lakes, rivers, sea surface water, and marine sediments. Microplastics have been reported to attach to marine macroalgae, which can be transferred and accumulated between organisms of different trophic levels in the marine food web, and consequently, affect human health [190,191].
Apart from the bioactive properties of lectins, the hemagglutinating mechanism is a reason for toxicity. The hemagglutination by lectins leads to growth retardation in animals, probably because of their ability to bind with specific receptor sites on the surface of the intestinal epithelial cells, resulting in impairment of nutrient absorption [192].
Though seaweed peptides are predicted as non-toxic, several sequences have been reported as allergenic [193]. Gammaridean amphipods and caprellid amphipods inhabit Porphyra spp. (nori) and can mix with nori sheets during harvesting and are consequently found in dried nori sheets, resembling small stones. Hence, dried Porphyra spp. (nori) products may contain amphipod allergens that can cause severe allergic reactions, particularly in crustacean-allergic people [169,194]. Amino acids such as kainic acid in dulse (P. palmata) and some other red algae (D. simplex) are structurally similar to glutamate (a neurotransmitter in the brain) and become neurotoxins at excessive levels. Food allergy is one of the main concerns for food safety; however, the potential allergenicity of proteins from macroalgae has not yet been fully explored [186,195].

8. Future Perspectives

Currently, exploitation of seaweed protein for human consumption is rare. Many studies have stated that seaweed has many biological properties such as antioxidant, antihypertensive, antioxidant, antidiabetic, antiviral, antimicrobial, among others. Although several publications are available on the quality of seaweed protein and its potential functional properties, only a few clinical studies have reached logical conclusions about actual functional food products [100].
Applications of new technologies need to be focused on identifying useful health-promoting compounds and eliminating chemical/microbiological risks and other current issues [196].
Since conventional protein extraction methods may require the use of non-negligible amounts of solvents, other environmentally friendly reagents or nonthermal techniques (membrane technologies such as ultrafiltration or nanofiltration) and economically viable processes should be investigated to produce, extract, and purify algal protein [1,95].
In addition, about half of all oxygen production on the planet comes from algae, which is another reason for saying “our lives depend on algae” [197]. Therefore, sustainable seaweed production should be carried out. Policies supporting seaweed production in aquaculture as a replacement for wild harvest and repopulation of natural sites are required to avoid an environmental crisis caused by the overexploitation of wild seaweed [18].

9. Conclusions

Seaweeds, also known as macroalgae or marine algae, are rich in protein, containing up to 40% protein with an excellent AA profile and high digestibility that is comparable, or even superior, to animal protein sources. More than 50% of the total amino acids are EAA in most seaweeds. Seaweed protein contains a number of bioactive components, including amino acids, free amino acids (especially taurine, laminine, kainic and domoic acids), peptides, phycobiliproteins (phycoerythrin and phycocyanin), and lectins. These bioactive compounds have many health benefits, including antihypertensive, antioxidant, antidiabetic, antiatherosclerosis, anti-inflammatory, antitumoral, antimicrobial, antiviral, and neuroprotective effects, among others. More than ever, people are concerned about their diet and health, resulting in a huge demand for high value functional foods that have been developed using natural sources instead of synthetic compounds. Developing functional foods using proteins derived from seaweeds has become more popular in the last decade. Seaweeds are a source of natural food additives include colorants, flavoring agents, antioxidants or preservatives, and many other compounds with health claims. Currently, protein derived from seaweed is rarely used as ingredients in the food industry, and acceptability, toxicity, allergenicity, and microbial studies have yet to be conducted for seaweed.

Author Contributions

Writing—original draft, P.T., A.G., O.M. and T.M. Writing—review and editing, P.T., A.G., N.S.G.-A., O.M. and T.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not Applicable.

Conflicts of Interest

The authors declare no conflict of interest. No conflict, informed consent, or human or animal rights are applicable to this study.

Abbreviations

AAAmino acid(s)
ACEAngiotensin-I-converting enzyme
Ang II Angiotensin II
DPP-IV Dipeptidyl peptidase IV
EAAEssential amino acid(s)
FAO Food and Agriculture Organization of the United Nations
GIPGlucose-dependent insulinotropic poly-peptide
GLP-1Glucagon-like peptide-1
MERS-CoV Middle East respiratory syndrome coronavirus
NEAANon-essential amino acid(s)
PAF-AHPlatelet-activating factor acetylhydrolase
SARS-CoV Severe acute respiratory syndrome coronavirus
T2DMType 2 diabetes mellitus
WHOWorld Health Organization

References

  1. Bleakley, S.; Hayes, M. Algal Proteins: Extraction, Application, and Challenges Concerning Production. Foods 2017, 6, 33. [Google Scholar] [CrossRef] [Green Version]
  2. Henchion, M.; Hayes, M.; Mullen, A.M.; Fenelon, M.; Tiwari, B. Future Protein Supply and Demand: Strategies and Factors Influencing a Sustainable Equilibrium. Foods 2017, 6, 53. [Google Scholar] [CrossRef] [Green Version]
  3. Wijers, T.; Hylkema, A.; Visser, T.; Timmermans, K. Effects of Preservation on Protein Extraction in Four Seaweed Species. J. Appl. Phycol. 2020, 32, 3401–3409. [Google Scholar] [CrossRef]
  4. Ferrara, L. Seaweeds: A Food for Our Future. J. Food Chem. Nanotechnol. 2020, 6, 56–64. [Google Scholar] [CrossRef]
  5. MacArtain, P.; Gill, C.I.R.; Brooks, M.; Campbell, R.; Rowland, I.R. Nutritional Value of Edible Seaweeds. Nutr. Rev. 2007, 65, 535–543. [Google Scholar] [CrossRef]
  6. Rajauria, G.; Cornish, L.; Ometto, F.; Msuya, F.E.; Villa, R. Chapter 12—Identification and Selection of Algae for Food, Feed, and Fuel Applications. In Seaweed Sustainability; Tiwari, B.K., Troy, D.J., Eds.; Academic Press: San Diego, CA, USA, 2015; pp. 315–345. ISBN 978-0-12-418697-2. [Google Scholar]
  7. Baweja, P.; Kumar, S.; Sahoo, D.; Levine, I. Chapter 3—Biology of Seaweeds. In Seaweed in Health and Disease Prevention; Fleurence, J., Levine, I., Eds.; Academic Press: San Diego, CA, USA, 2016; pp. 41–106. ISBN 978-0-12-802772-1. [Google Scholar]
  8. Mahadevan, K. Chapter 13—Seaweeds: A Sustainable Food Source. In Seaweed Sustainability; Tiwari, B.K., Troy, D.J., Eds.; Academic Press: San Diego, CA, USA, 2015; pp. 347–364. ISBN 978-0-12-418697-2. [Google Scholar]
  9. Pandey, A.K.; Chauhan, O.P.; Semwal, A.D. Seaweeds. Def. Life Sci. J. 2020, 5, 315–322. [Google Scholar] [CrossRef]
  10. Fleurence, J.; Morançais, M.; Dumay, J. 9—Seaweed Proteins. In Proteins in Food Processing, 2nd ed.; Yada, R.Y., Ed.; Woodhead Publishing Series in Food Science, Technology and Nutrition; Woodhead Publishing: Sawston, UK, 2018; pp. 245–262. ISBN 978-0-08-100722-8. [Google Scholar]
  11. Shannon, E.; Abu-Ghannam, N. Seaweeds as Nutraceuticals for Health and Nutrition. Phycologia 2019, 58, 563–577. [Google Scholar] [CrossRef] [Green Version]
  12. Lamont, T.; McSweeney, M. Consumer Acceptability and Chemical Composition of Whole-Wheat Breads Incorporated with Brown Seaweed (Ascophyllum Nodosum) or Red Seaweed (Chondrus Crispus). J. Sci. Food Agric. 2021, 101, 1507–1514. [Google Scholar] [CrossRef]
  13. Lomartire, S.; Marques, J.C.; Gonçalves, A.M.M. An Overview to the Health Benefits of Seaweeds Consumption. Mar. Drugs 2021, 19, 341. [Google Scholar] [CrossRef]
  14. Sabeena Farvin, K.H.; Jacobsen, C. Phenolic Compounds and Antioxidant Activities of Selected Species of Seaweeds from Danish Coast. Food Chem. 2013, 138, 1670–1681. [Google Scholar] [CrossRef]
  15. Fernández-Segovia, I.; Lerma-García, M.J.; Fuentes, A.; Barat, J.M. Characterization of Spanish Powdered Seaweeds: Composition, Antioxidant Capacity and Technological Properties. Food Res. Int. 2018, 111, 212–219. [Google Scholar] [CrossRef] [PubMed]
  16. FAO. FAO Yearbook. Fishery and Aquaculture Statistics 2018/FAO Annuaire. Statistiques des Pêches et de L’aquaculture 2018/FAO anuario. Estadísticas de Pesca y Acuicultura 2018; FAO Yearbook of Fishery and Aquaculture Statistics; FAO: Rome, Italy, 2020; ISBN 978-92-5-133371-6. [Google Scholar]
  17. Kraan, S. Chapter 3—Seaweed Resources, Collection, and Cultivation with Respect to Sustainability. In Sustainable Seaweed Technologies; Torres, M.D., Kraan, S., Dominguez, H., Eds.; Advances in Green and Sustainable Chemistry; Elsevier: Amsterdam, The Netherlands, 2020; pp. 89–102. ISBN 978-0-12-817943-7. [Google Scholar]
  18. Buschmann, A.H.; Camus, C.; Infante, J.; Neori, A.; Israel, Á.; Hernández-González, M.C.; Pereda, S.V.; Gomez-Pinchetti, J.L.; Golberg, A.; Tadmor-Shalev, N.; et al. Seaweed Production: Overview of the Global State of Exploitation, Farming and Emerging Research Activity. Eur. J. Phycol. 2017, 52, 391–406. [Google Scholar] [CrossRef]
  19. Geada, P.; Moreira, C.; Silva, M.; Nunes, R.; Madureira, L.; Rocha, C.M.R.; Pereira, R.N.; Vicente, A.A.; Teixeira, J.A. Algal Proteins: Production Strategies and Nutritional and Functional Properties. Bioresour. Technol. 2021, 332, 125125. [Google Scholar] [CrossRef] [PubMed]
  20. 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]
  21. Nagarajan, M.; Rajesh Kumar, R.; Meenakshi Sundaram, K.; Sundararaman, M. Marine Biotechnology: Potentials of Marine Microbes and Algae with Reference to Pharmacological and Commercial Values. In Plant Biology and Biotechnology: Volume II: Plant Genomics and Biotechnology; Bahadur, B., Venkat Rajam, M., Sahijram, L., Krishnamurthy, K.V., Eds.; Springer India: New Delhi, India, 2015; pp. 685–723. ISBN 978-81-322-2283-5. [Google Scholar]
  22. 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. [Google Scholar] [CrossRef]
  23. Cassani, L.; Lourenço-Lopes, C.; Barral-Martinez, M.; Chamorro, F.; Garcia-Perez, P.; Simal-Gandara, J.; Prieto, M.A. Thermochemical Characterization of Eight Seaweed Species and Evaluation of Their Potential Use as an Alternative for Biofuel Production and Source of Bioactive Compounds. Int. J. Mol. Sci. 2022, 23, 2355. [Google Scholar] [CrossRef]
  24. Chen, H.; Zhou, D.; Luo, G.; Zhang, S.; Chen, J. Macroalgae for Biofuels Production: Progress and Perspectives. Renew. Sustain. Energy Rev. 2015, 47, 427–437. [Google Scholar] [CrossRef]
  25. Nisizawa, K.; Noda, H.; Kikuchi, R.; Watanabe, T. The Main Seaweed Foods in Japan. Hydrobiologia 1987, 151, 5–29. [Google Scholar] [CrossRef]
  26. Ganesan, A.R.; Tiwari, U.; Rajauria, G. Seaweed Nutraceuticals and Their Therapeutic Role in Disease Prevention. Food Sci. Hum. Wellness 2019, 8, 252–263. [Google Scholar] [CrossRef]
  27. Admassu, H.; Gasmalla, M.A.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]
  28. Wells, M.L.; Potin, P.; Craigie, J.S.; Raven, J.A.; Merchant, S.S.; Helliwell, K.E.; Smith, A.G.; Camire, M.E.; Brawley, S.H. Algae as Nutritional and Functional Food Sources: Revisiting Our Understanding. J. Appl. Phycol. 2017, 29, 949–982. [Google Scholar] [CrossRef] [PubMed]
  29. Machado, M.; Machado, S.; Pimentel, F.B.; Freitas, V.; Alves, R.C.; Oliveira, M.B.P.P. Amino Acid Profile and Protein Quality Assessment of Macroalgae Produced in an Integrated Multi-Trophic Aquaculture System. Foods 2020, 9, 1382. [Google Scholar] [CrossRef] [PubMed]
  30. Pliego-Cortés, H.; Wijesekara, I.; Lang, M.; Bourgougnon, N.; Bedoux, G. Chapter Nine—Current Knowledge and Challenges in Extraction, Characterization and Bioactivity of Seaweed Protein and Seaweed-Derived Proteins. Adv. Bot. Res. 2020, 95, 289–326. [Google Scholar] [CrossRef]
  31. Naseri, A.; Marinho, G.S.; Holdt, S.L.; Bartela, J.M.; Jacobsen, C. Enzyme-Assisted Extraction and Characterization of Protein from Red Seaweed Palmaria palmata. Algal Res. 2020, 47, 101849. [Google Scholar] [CrossRef]
  32. 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]
  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. Pereira, L. A Review of the Nutrient Composition of Selected Edible Seaweeds. In Seaweed: Ecology, Nutrient Composition and Medicinal Uses; Nova Science Publishers, Inc.: Hauppauge, NY, USA, 2011; pp. 15–47. ISBN 978-1-61470-878-0. [Google Scholar]
  35. Marsham, S.; Scott, G.W.; Tobin, M.L. Comparison of Nutritive Chemistry of a Range of Temperate Seaweeds. Food Chem. 2007, 100, 1331–1336. [Google Scholar] [CrossRef]
  36. Lorenzo, J.M.; Agregán, R.; Munekata, P.E.S.; Franco, D.; Carballo, J.; Şahin, S.; Lacomba, R.; Barba, F.J. Proximate Composition and Nutritional Value of Three Macroalgae: Ascophyllum Nodosum, Fucus Vesiculosus and Bifurcaria Bifurcata. Mar. Drugs 2017, 15, 360. [Google Scholar] [CrossRef] [Green Version]
  37. Fleurence, J. Seaweed Proteins: Biochemical, Nutritional Aspects and Potential Uses. Trends Food Sci. Technol. 1999, 10, 25–28. [Google Scholar] [CrossRef]
  38. Harnedy, P.A.; FitzGerald, R.J. Bioactive Proteins, Peptides, and Amino Acids from Macroalgae. J. Phycol. 2011, 47, 218–232. [Google Scholar] [CrossRef]
  39. Dickson, R.; Ryu, J.-H.; Liu, J.J. Optimal Plant Design for Integrated Biorefinery Producing Bioethanol and Protein from Saccharina Japonica: A Superstructure-Based Approach. Energy 2018, 164, 1257–1270. [Google Scholar] [CrossRef]
  40. Dawczynski, C.; Schubert, R.; Jahreis, G. Amino Acids, Fatty Acids, and Dietary Fibre in Edible Seaweed Products. Food Chem. 2007, 103, 891–899. [Google Scholar] [CrossRef]
  41. Nagappan, T.; Vairappan, C.S. Nutritional and Bioactive Properties of Three Edible Species of Green Algae, Genus Caulerpa (Caulerpaceae). J. Appl. Phycol. 2014, 26, 1019–1027. [Google Scholar] [CrossRef]
  42. Dumay, J.; Morançais, M. Chapter 9—Proteins and Pigments. In Seaweed in Health and Disease Prevention; Fleurence, J., Levine, I., Eds.; Academic Press: San Diego, CA, USA, 2016; pp. 275–318. ISBN 978-0-12-802772-1. [Google Scholar]
  43. Afonso, C.; Correia, A.P.; Freitas, M.V.; Baptista, T.; Neves, M.; Mouga, T. Seasonal Changes in the Nutritional Composition of Agarophyton vermiculophyllum (Rhodophyta, Gracilariales) from the Center of Portugal. Foods 2021, 10, 1145. [Google Scholar] [CrossRef]
  44. Kulshreshtha, G.; Burlot, A.-S.; Marty, C.; Critchley, A.; Hafting, J.; Bedoux, G.; Bourgougnon, N.; Prithiviraj, B. Enzyme-Assisted Extraction of Bioactive Material from Chondrus Crispus and Codium Fragile and Its Effect on Herpes Simplex Virus (HSV-1). Mar. Drugs 2015, 13, 558–580. [Google Scholar] [CrossRef] [Green Version]
  45. Tabarsa, M.; Rezaei, M.; Ramezanpour, Z.; Waaland, J.R. Chemical Compositions of the Marine Algae Gracilaria Salicornia (Rhodophyta) and Ulva Lactuca (Chlorophyta) as a Potential Food Source. J. Sci. Food Agric. 2012, 92, 2500–2506. [Google Scholar] [CrossRef]
  46. Rosemary, T.; Arulkumar, A.; Paramasivam, S.; Mondragon-Portocarrero, A.; Miranda, J.M. Biochemical, Micronutrient and Physicochemical Properties of the Dried Red Seaweeds Gracilaria Edulis and Gracilaria Corticata. Molecules 2019, 24, 2225. [Google Scholar] [CrossRef] [Green Version]
  47. Francavilla, M.; Manara, P.; Kamaterou, P.; Monteleone, M.; Zabaniotou, A. Cascade Approach of Red Macroalgae Gracilaria Gracilis Sustainable Valorization by Extraction of Phycobiliproteins and Pyrolysis of Residue. Bioresour. Technol. 2015, 184, 305–313. [Google Scholar] [CrossRef]
  48. Patarra, R.F.; Paiva, L.; Neto, A.I.; Lima, E.; Baptista, J. Nutritional Value of Selected Macroalgae. J. Appl. Phycol. 2011, 23, 205–208. [Google Scholar] [CrossRef] [Green Version]
  49. Wang, T.; Jónsdóttir, R.; Kristinsson, H.G.; Hreggvidsson, G.O.; Jónsson, J.Ó.; Thorkelsson, G.; Ólafsdóttir, G. Enzyme-Enhanced Extraction of Antioxidant Ingredients from Red Algae Palmaria palmata. LWT—Food Sci. Technol. 2010, 43, 1387–1393. [Google Scholar] [CrossRef]
  50. Cian, R.E.; Fajardo, M.A.; Alaiz, M.; Vioque, J.; González, R.J.; Drago, S.R. Chemical Composition, Nutritional and Antioxidant Properties of the Red Edible Seaweed Porphyra Columbina. Int. J. Food Sci. Nutr. 2014, 65, 299–305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Kumar, S.; Sahoo, D.; Levine, I. Assessment of Nutritional Value in a Brown Seaweed Sargassum Wightii and Their Seasonal Variations. Algal Res. 2015, 9, 117–125. [Google Scholar] [CrossRef]
  52. O’ Connor, J.; Meaney, S.; Williams, G.A.; Hayes, M. Extraction of Protein from Four Different Seaweeds Using Three Different Physical Pre-Treatment Strategies. Molecules 2020, 25, 2005. [Google Scholar] [CrossRef] [PubMed]
  53. Zhou, A.Y.; Robertson, J.; Hamid, N.; Ma, Q.; Lu, J. Changes in Total Nitrogen and Amino Acid Composition of New Zealand Undaria pinnatifida with Growth, Location and Plant Parts. Food Chem. 2015, 186, 319–325. [Google Scholar] [CrossRef]
  54. Biancarosa, I.; Espe, M.; Bruckner, C.G.; Heesch, S.; Liland, N.; Waagbø, R.; Torstensen, B.; Lock, E.J. Amino Acid Composition, Protein Content, and Nitrogen-to-Protein Conversion Factors of 21 Seaweed Species from Norwegian Waters. J. Appl. Phycol. 2017, 29, 1001–1009. [Google Scholar] [CrossRef]
  55. Bjarnadóttir, M.; Aðalbjörnsson, B.V.; Nilsson, A.; Slizyte, R.; Roleda, M.Y.; Hreggviðsson, G.Ó.; Friðjónsson, Ó.H.; Jónsdóttir, R. Palmaria palmata as an Alternative Protein Source: Enzymatic Protein Extraction, Amino Acid Composition, and Nitrogen-to-Protein Conversion Factor. J. Appl. Phycol. 2018, 30, 2061–2070. [Google Scholar] [CrossRef]
  56. Lourenço, S.O.; Barbarino, E.; De-Paula, J.C.; Pereira, L.O.D.S.; Marquez, U.M.L. Amino Acid Composition, Protein Content and Calculation of Nitrogen-to-Protein Conversion Factors for 19 Tropical Seaweeds. Phycol. Res. 2002, 50, 233–241. [Google Scholar] [CrossRef]
  57. Shuuluka, D.; Bolton, J.J.; Anderson, R.J. Protein Content, Amino Acid Composition and Nitrogen-to-Protein Conversion Factors of Ulva Rigida and Ulva Capensis from Natural Populations and Ulva Lactuca from an Aquaculture System, in South Africa. J. Appl. Phycol. 2013, 25, 677–685. [Google Scholar] [CrossRef]
  58. Černá, M. Chapter 24—Seaweed Proteins and Amino Acids as Nutraceuticals. Adv. Food Nutr. Res. 2011, 64, 297–312. [Google Scholar] [CrossRef]
  59. Cherry, P.; O’Hara, C.; Magee, P.J.; McSorley, E.M.; Allsopp, P.J. Risks and Benefits of Consuming Edible Seaweeds. Nutr. Rev. 2019, 77, 307–329. [Google Scholar] [CrossRef] [Green Version]
  60. Pangestuti, R.; Kim, S.-K. Chapter 6—Seaweed Proteins, Peptides, and Amino Acids. In Seaweed Sustainability; Tiwari, B.K., Troy, D.J., Eds.; Academic Press: San Diego, CA, USA, 2015; pp. 125–140. ISBN 978-0-12-418697-2. [Google Scholar]
  61. Joint FAO/WHO/UNU Protein and Amino Acid Requirements in Human Nutrition; World Health Organization: Geneva, Switzerland, 2007; ISBN 978-92-4-120935-9.
  62. Cofrades, S.; López-Lopez, I.; Bravo, L.; Ruiz-Capillas, C.; Bastida, S.; Larrea, M.T.; Jiménez-Colmenero, F. Nutritional and Antioxidant Properties of Different Brown and Red Spanish Edible Seaweeds. Food Sci. Technol. Int. 2010, 16, 361–370. [Google Scholar] [CrossRef] [PubMed]
  63. Vieira, E.F.; Soares, C.; Machado, S.; Correia, M.; Ramalhosa, M.J.; Oliva-teles, M.T.; Paula Carvalho, A.; Domingues, V.F.; Antunes, F.; Oliveira, T.A.C.; et al. Seaweeds from the Portuguese Coast as a Source of Proteinaceous Material: Total and Free Amino Acid Composition Profile. Food Chem. 2018, 269, 264–275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Benjama, O.; Masniyom, P. Nutritional Composition and Physicochemical Properties of Two Green Seaweeds (Ulva Pertusa and U. Intestinalis) from the Pattani Bay in Southern Thailand. Songklanakarin J. Sci. Technol. 2011, 33, 575–583. [Google Scholar]
  65. 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]
  66. Chan, P.T.; Matanjun, P. Chemical Composition and Physicochemical Properties of Tropical Red Seaweed, Gracilaria Changii. Food Chem. 2017, 221, 302–310. [Google Scholar] [CrossRef]
  67. Mohammed, H.O.; O’Grady, M.N.; O’Sullivan, M.G.; Hamill, R.M.; Kilcawley, K.N.; Kerry, J.P. An Assessment of Selected Nutritional, Bioactive, Thermal and Technological Properties of Brown and Red Irish Seaweed Species. Foods 2021, 10, 2784. [Google Scholar] [CrossRef]
  68. De Bhowmick, G.; Hayes, M. In Vitro Protein Digestibility of Selected Seaweeds. Foods 2022, 11, 289. [Google Scholar] [CrossRef]
  69. Astorga-España, M.S.; Rodríguez-Galdón, B.; Rodríguez-Rodríguez, E.M.; Díaz-Romero, C. Amino Acid Content in Seaweeds from the Magellan Straits (Chile). J. Food Compos. Anal. 2016, 53, 77–84. [Google Scholar] [CrossRef]
  70. Bikker, P.; Stokvis, L.; Van Krimpen, M.M.; Van Wikselaar, P.G.; Cone, J.W. Evaluation of Seaweeds from Marine Waters in Northwestern Europe for Application in Animal Nutrition. Anim. Feed. Sci. Technol. 2020, 263, 114460. [Google Scholar] [CrossRef]
  71. Kadam, S.U.; Álvarez, C.; Tiwari, B.K.; O’Donnell, C.P. Extraction and Characterization of Protein from Irish Brown Seaweed Ascophyllum Nodosum. Food Res. Int. 2017, 99, 1021–1027. [Google Scholar] [CrossRef]
  72. Kazir, M.; Abuhassira, Y.; Robin, A.; Nahor, O.; Luo, J.; Israel, A.; Golberg, A.; Livney, Y.D. Extraction of Proteins from Two Marine Macroalgae, Ulva sp. and Gracilaria sp., for Food Application, and Evaluating Digestibility, Amino Acid Composition and Antioxidant Properties of the Protein Concentrates. Food Hydrocoll. 2019, 87, 194–203. [Google Scholar] [CrossRef]
  73. Bak, U.G.; Nielsen, C.W.; Marinho, G.S.; Gregersen, Ó.; Jónsdóttir, R.; Holdt, S.L. The Seasonal Variation in Nitrogen, Amino Acid, Protein and Nitrogen-to-Protein Conversion Factors of Commercially Cultivated Faroese Saccharina latissima. Algal Res. 2019, 42, 101576. [Google Scholar] [CrossRef]
  74. Uribe, E.; Vega-Gálvez, A.; García, V.; Pastén, A.; López, J.; Goñi, G. Effect of Different Drying Methods on Phytochemical Content and Amino Acid and Fatty Acid Profiles of the Green Seaweed, Ulva spp. J. Appl. Phycol. 2019, 31, 1967–1979. [Google Scholar] [CrossRef]
  75. Ramu Ganesan, A.; Subramani, K.; Shanmugam, M.; Seedevi, P.; Park, S.; Alfarhan, A.H.; Rajagopal, R.; Balasubramanian, B. A Comparison of Nutritional Value of Underexploited Edible Seaweeds with Recommended Dietary Allowances. J. King Saud Univ.—Sci. 2020, 32, 1206–1211. [Google Scholar] [CrossRef]
  76. Wong, K.H.; Cheung, P.C.K. Nutritional Evaluation of Some Subtropical Red and Green Seaweeds: Part I —Proximate Composition, Amino Acid Profiles and Some Physico-Chemical Properties. Food Chem. 2000, 71, 475–482. [Google Scholar] [CrossRef]
  77. Norziah, M.H.; Ching, C.Y. Nutritional Composition of Edible Seaweed Gracilaria Changgi. Food Chem. 2000, 68, 69–76. [Google Scholar] [CrossRef]
  78. Nazarudin, M.F.; Alias, N.H.; Balakrishnan, S.; Wan Hasnan, W.N.I.; Noor Mazli, N.A.I.; Ahmad, M.I.; Md Yasin, I.-S.; Isha, A.; Aliyu-Paiko, M. Chemical, Nutrient and Physicochemical Properties of Brown Seaweed, Sargassum Polycystum C. Agardh (Phaeophyceae) Collected from Port Dickson, Peninsular Malaysia. Molecules 2021, 26, 5216. [Google Scholar] [CrossRef]
  79. Conde, E.; Balboa, E.M.; Parada, M.; Falqué, E. 4—Algal Proteins, Peptides and Amino Acids. In Functional Ingredients from Algae for Foods and Nutraceuticals; Domínguez, H., Ed.; Woodhead Publishing Series in Food Science, Technology and Nutrition; Woodhead Publishing: Sawston, UK, 2013; pp. 135–180. ISBN 978-0-85709-512-1. [Google Scholar]
  80. Galland-Irmouli, A.-V.; Fleurence, J.; Lamghari, R.; Luçon, M.; Rouxel, C.; Barbaroux, O.; Bronowicki, J.-P.; Villaume, C.; Guéant, J.-L. Nutritional Value of Proteins from Edible Seaweed Palmaria Palmata (Dulse). J. Nutr. Biochem. 1999, 10, 353–359. [Google Scholar] [CrossRef]
  81. Bejosano, F.P.; Corke, H. Protein Quality Evaluation of Amaranthus Wholemeal Flours and Protein Concentrates. J. Sci. Food Agric. 1998, 76, 100–106. [Google Scholar] [CrossRef]
  82. Kies, C. Bioavailability: A Factor in Protein Quality. J. Agric. Food Chem. 1981, 29, 435–440. [Google Scholar] [CrossRef]
  83. Swaisgood, H.E.; Catignani, G.L. Protein Digestibility: In Vitro Methods of Assessment. In Advances in Food and Nutrition Research; Kinsella, J.E., Ed.; Academic Press: Cambridge, MA, USA, 1991; Volume 35, pp. 185–236. [Google Scholar]
  84. Marrion, O.; Fleurence, J.; Schwertz, A.; Guéant, J.-L.; Mamelouk, L.; Ksouri, J.; Villaume, C. Evaluation of Protein in Vitro Digestibility of Palmaria Palmata and Gracilaria Verrucosa. J. Appl. Phycol. 2005, 17, 99–102. [Google Scholar] [CrossRef]
  85. Tibbetts, S.M.; Milley, J.E.; Lall, S.P. Nutritional Quality of Some Wild and Cultivated Seaweeds: Nutrient Composition, Total Phenolic Content and in Vitro Digestibility. J. Appl. Phycol. 2016, 28, 3575–3585. [Google Scholar] [CrossRef]
  86. Mišurcová, L.; Kráčmar, S.; Klejdus, B.; Vacek, J. Nitrogen Content, Dietary Fiber, and Digestibility in Algal Food Products. Czech. J. Food Sci. 2010, 28, 27–35. [Google Scholar] [CrossRef] [Green Version]
  87. Machů, L.; Mišurcová, L.; Samek, D.; Hrabě, J.; Fišera, M. In Vitro Digestibility of Different Commercial Edible Algae Products. J. Aquat. Food Prod. Technol. 2014, 23, 423–435. [Google Scholar] [CrossRef]
  88. Fujiwara-Arasaki, T.; Mino, N.; Kuroda, M. The Protein Value in Human Nutrition of Edible Marine Algae in Japan. In Proceedings of the Eleventh International Seaweed Symposium, Qingdao, China, 19–25 June 1983; Bird, C.J., Ragan, M.A., Eds.; Springer: Dordrecht, The Netherlands, 1984; pp. 513–516. [Google Scholar]
  89. Echave, J.; Fraga-Corral, M.; Garcia-Perez, P.; Popović-Djordjević, J.; Avdović, E.H.; Radulović, M.; Xiao, J.; Prieto, M.A.; Simal-Gandara, J. Seaweed Protein Hydrolysates and Bioactive Peptides: Extraction, Purification, and Applications. Mar. Drugs 2021, 19, 500. [Google Scholar] [CrossRef] [PubMed]
  90. Gajaria, T.K.; Suthar, P.; Baghel, R.S.; Balar, N.B.; Sharnagat, P.; Mantri, V.A.; Reddy, C.R.K. Integration of Protein Extraction with a Stream of Byproducts from Marine Macroalgae: A Model Forms the Basis for Marine Bioeconomy. Bioresour. Technol. 2017, 243, 867–873. [Google Scholar] [CrossRef]
  91. Vásquez, V.; Martínez, R.; Bernal, C. Enzyme-Assisted Extraction of Proteins from the Seaweeds Macrocystis pyrifera and Chondracanthus chamissoi: Characterization of the Extracts and Their Bioactive Potential. J. Appl. Phycol. 2019, 31, 1999–2010. [Google Scholar] [CrossRef]
  92. Amorim, M.L.; Soares, J.; Coimbra, J.S.D.R.; De Oliviera Leite, M.; Albino, L.F.T.; Martins, M.A. Microalgae Proteins: Production, Separation, Isolation, Quantification, and Application in Food and Feed. Crit. Rev. Food Sci. Nutr. 2021, 61, 1976–2002. [Google Scholar] [CrossRef]
  93. Naseri, A.; Jacobsen, C.; Sejberg, J.J.P.; Pedersen, T.E.; Larsen, J.; Hansen, K.M.; Holdt, S.L. Multi-Extraction and Quality of Protein and Carrageenan from Commercial Spinosum (Eucheuma denticulatum). Foods 2020, 9, 1072. [Google Scholar] [CrossRef]
  94. Nadeeshani, H.; Hassouna, A.; Lu, J. Proteins Extracted from Seaweed Undaria pinnatifida and Their Potential Uses as Foods and Nutraceuticals. Crit. Rev. Food Sci. Nutr. 2021, 1–17. [Google Scholar] [CrossRef]
  95. Tremblay, A.; Beaulieu, L. Extraction Technologies for Proteins and Peptides. In Recent Advances in Micro and Macroalgal Processing; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2021; pp. 141–162. ISBN 978-1-119-54265-0. [Google Scholar]
  96. Abdollahi, M.; Axelsson, J.; Carlsson, N.-G.; Nylund, G.M.; Albers, E.; Undeland, I. Effect of Stabilization Method and Freeze/Thaw-Aided Precipitation on Structural and Functional Properties of Proteins Recovered from Brown Seaweed (Saccharina latissima). Food Hydrocoll. 2019, 96, 140–150. [Google Scholar] [CrossRef]
  97. Cermeño, M.; Kleekayai, T.; Amigo-Benavent, M.; Harnedy-Rothwell, P.; FitzGerald, R.J. Current Knowledge on the Extraction, Purification, Identification, and Validation of Bioactive Peptides from Seaweed. Electrophoresis 2020, 41, 1694–1717. [Google Scholar] [CrossRef]
  98. Badmus, U.O.; Taggart, M.A.; Boyd, K.G. The Effect of Different Drying Methods on Certain Nutritionally Important Chemical Constituents in Edible Brown Seaweeds. J. Appl. Phycol. 2019, 31, 3883–3897. [Google Scholar] [CrossRef] [Green Version]
  99. Harrysson, H.; Hayes, M.; Eimer, F.; Carlsson, N.-G.; Toth, G.B.; Undeland, I. Production of Protein Extracts from Swedish Red, Green, and Brown Seaweeds, Porphyra umbilicalis Kützing, Ulva Lactuca Linnaeus, and Saccharina latissima (Linnaeus) J. V. Lamouroux Using Three Different Methods. J. Appl. Phycol. 2018, 30, 3565–3580. [Google Scholar] [CrossRef] [Green Version]
  100. Tanna, B.; Mishra, A. Metabolites Unravel Nutraceutical Potential of Edible Seaweeds: An Emerging Source of Functional Food. Compr. Rev. Food Sci. Food Saf. 2018, 17, 1613–1624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Brown, E.M.; Allsopp, P.J.; Magee, P.J.; Gill, C.I.; Nitecki, S.; Strain, C.R.; McSorley, E.M. Seaweed and Human Health. Nutr. Rev. 2014, 72, 205–216. [Google Scholar] [CrossRef]
  102. Saikia, S.; Mahnot, N.K.; Sahu, R.K.; Kalita, J. Edible Seaweeds as Potential Source of Nutraceuticals. In Marine Niche: Applications in Pharmaceutical Sciences: Translational Research; Nathani, N.M., Mootapally, C., Gadhvi, I.R., Maitreya, B., Joshi, C.G., Eds.; Springer: Singapore, 2020; pp. 183–201. ISBN 9789811550171. [Google Scholar]
  103. Flórez-Fernández, N.; Torres, M.D.; Braz, L.; Grenha, A.; Loret, E.P.; Domínguez, H. Seaweed and Sea Anemones Proteins as a Source of New Pharmaceutical Active Principles. In Marine Niche: Applications in Pharmaceutical Sciences: Translational Research; Nathani, N.M., Mootapally, C., Gadhvi, I.R., Maitreya, B., Joshi, C.G., Eds.; Springer: Singapore, 2020; pp. 203–219. ISBN 9789811550171. [Google Scholar]
  104. Dmitrenok, A.; Iwashita, T.; Nakajima, T.; Sakamoto, B.; Namikoshi, M.; Nagai, H. New Cyclic Depsipeptides from the Green Alga Bryopsis Species; Application of a Carboxypeptidase Hydrolysis Reaction to the Structure Determination. Tetrahedron 2006, 62, 1301–1308. [Google Scholar] [CrossRef]
  105. Deng, Z.; Liu, Y.; Wang, J.; Wu, S.; Geng, L.; Sui, Z.; Zhang, Q. Antihypertensive Effects of Two Novel Angiotensin I-Converting Enzyme (ACE) Inhibitory Peptides from Gracilariopsis lemaneiformis (Rhodophyta) in Spontaneously Hypertensive Rats (SHRs). Mar. Drugs 2018, 16, 299. [Google Scholar] [CrossRef] [Green Version]
  106. Kumagai, Y.; Kitade, Y.; Kobayashi, M.; Watanabe, K.; Kurita, H.; Takeda, H.; Yasui, H.; Kishimura, H. Identification of ACE Inhibitory Peptides from Red Alga Mazzaella japonica. Eur. Food Res. Technol. 2020, 246, 2225–2231. [Google Scholar] [CrossRef]
  107. Kitade, Y.; Miyabe, Y.; Yamamoto, Y.; Takeda, H.; Shimizu, T.; Yasui, H.; Kishimura, H. Structural Characteristics of Phycobiliproteins from Red Alga Mazzaella japonica. J. Food Biochem. 2018, 42, e12436. [Google Scholar] [CrossRef] [Green Version]
  108. Qu, W.; Ma, H.; Pan, Z.; Luo, L.; Wang, Z.; He, R. Preparation and Antihypertensive Activity of Peptides from Porphyra Yezoensis. Food Chem. 2010, 123, 14–20. [Google Scholar] [CrossRef]
  109. Jiao, K.; Gao, J.; Zhou, T.; Yu, J.; Song, H.; Wei, Y.; Gao, X. Isolation and Purification of a Novel Antimicrobial Peptide from Porphyra Yezoensis. J. Food Biochem. 2019, 43, e12864. [Google Scholar] [CrossRef] [PubMed]
  110. Furuta, T.; Miyabe, Y.; Yasui, H.; Kinoshita, Y.; Kishimura, H. Angiotensin I Converting Enzyme Inhibitory Peptides Derived from Phycobiliproteins of Dulse Palmaria palmata. Mar. Drugs 2016, 14, 32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. McLaughlin, C.M.; Harnedy-Rothwell, P.A.; Lafferty, R.A.; Sharkey, S.; Parthsarathy, V.; Allsopp, P.J.; McSorley, E.M.; FitzGerald, R.J.; O’Harte, F.P.M. Macroalgal Protein Hydrolysates from Palmaria palmata Influence the ‘Incretin Effect’ in Vitro via DPP-4 Inhibition and Upregulation of Insulin, GLP-1 and GIP Secretion. Eur. J. Nutr. 2021, 60, 4439–4452. [Google Scholar] [CrossRef]
  112. McLaughlin, C.M.; Sharkey, S.J.; Harnedy-Rothwell, P.; Parthsarathy, V.; Allsopp, P.J.; McSorley, E.M.; FitzGerald, R.J.; O’Harte, F.P.M. Twice Daily Oral Administration of Palmaria palmata Protein Hydrolysate Reduces Food Intake in Streptozotocin Induced Diabetic Mice, Improving Glycaemic Control and Lipid Profiles. J. Funct. Foods 2020, 73, 104101. [Google Scholar] [CrossRef]
  113. Harnedy, P.A.; O’Keeffe, M.B.; FitzGerald, R.J. Purification and Identification of Dipeptidyl Peptidase (DPP) IV Inhibitory Peptides from the Macroalga Palmaria palmata. Food Chem. 2015, 172, 400–406. [Google Scholar] [CrossRef]
  114. Fitzgerald, C.; Gallagher, E.; O’Connor, P.; Prieto, J.; Mora-Soler, L.; Grealy, M.; Hayes, M. Development of a Seaweed Derived Platelet Activating Factor Acetylhydrolase (PAF-AH) Inhibitory Hydrolysate, Synthesis of Inhibitory Peptides and Assessment of Their Toxicity Using the Zebrafish Larvae Assay. Peptides 2013, 50, 119–124. [Google Scholar] [CrossRef]
  115. Fitzgerald, C.; Mora-Soler, L.; Gallagher, E.; O’Connor, P.; Prieto, J.; Soler-Vila, A.; Hayes, M. Isolation and Characterization of Bioactive Pro-Peptides with in Vitro Renin Inhibitory Activities from the Macroalga Palmaria palmata. J. Agric. Food Chem. 2012, 60, 7421–7427. [Google Scholar] [CrossRef]
  116. Harnedy, P.A.; O’Keeffe, M.B.; FitzGerald, R.J. Fractionation and Identification of Antioxidant Peptides from an Enzymatically Hydrolysed Palmaria palmata Protein Isolate. Food Res. Int. 2017, 100, 416–422. [Google Scholar] [CrossRef] [Green Version]
  117. Cermeño, M.; Stack, J.; Tobin, P.R.; O’Keeffe, M.B.; Harnedy, P.A.; Stengel, D.B.; FitzGerald, R.J. Peptide Identification from a Porphyra Dioica Protein Hydrolysate with Antioxidant, Angiotensin Converting Enzyme and Dipeptidyl Peptidase IV Inhibitory Activities. Food Funct. 2019, 10, 3421–3429. [Google Scholar] [CrossRef]
  118. Cian, R.E.; Martínez-Augustin, O.; Drago, S.R. Bioactive Properties of Peptides Obtained by Enzymatic Hydrolysis from Protein Byproducts of Porphyra Columbina. Food Res. Int. 2012, 49, 364–372. [Google Scholar] [CrossRef]
  119. Beaulieu, L.; Bondu, S.; Doiron, K.; Rioux, L.-E.; Turgeon, S.L. Characterization of Antibacterial Activity from Protein Hydrolysates of the Macroalga Saccharina Longicruris and Identification of Peptides Implied in Bioactivity. J. Funct. Foods 2015, 17, 685–697. [Google Scholar] [CrossRef]
  120. Liu, X.; Wang, C.-Y.; Shao, C.-L.; Wei, Y.-X.; Wang, B.-G.; Sun, L.-L.; Zheng, C.-J.; Guan, H.-S. Chemical Constituents from Sargassum Pallidum (Turn.) C. Agardh. Biochem. Syst. Ecol. 2009, 37, 127–129. [Google Scholar] [CrossRef]
  121. Paiva, L.; Lima, E.; Neto, A.I.; Baptista, J. Isolation and Characterization of Angiotensin I-Converting Enzyme (ACE) Inhibitory Peptides from Ulva Rigida, C. Agardh Protein Hydrolysate. J. Funct. Foods 2016, 26, 65–76. [Google Scholar] [CrossRef]
  122. Suetsuna, K.; Nakano, T. Identification of an Antihypertensive Peptide from Peptic Digest of Wakame (Undaria pinnatifida). J. Nutr. Biochem. 2000, 11, 450–454. [Google Scholar] [CrossRef]
  123. Sato, M.; Hosokawa, T.; Yamaguchi, T.; Nakano, T.; Muramoto, K.; Kahara, T.; Funayama, K.; Kobayashi, A.; Nakano, T. Angiotensin I-Converting Enzyme Inhibitory Peptides Derived from Wakame (Undaria pinnatifida) and Their Antihypertensive Effect in Spontaneously Hypertensive Rats. J. Agric. Food Chem. 2002, 50, 6245–6252. [Google Scholar] [CrossRef]
  124. Barre, A.; Van Damme, E.J.M.; Simplicien, M.; Le Poder, S.; Klonjkowski, B.; Benoist, H.; Peyrade, D.; Rougé, P. Man-Specific Lectins from Plants, Fungi, Algae and Cyanobacteria, as Potential Blockers for SARS-CoV, MERS-CoV and SARS-CoV-2 (COVID-19) Coronaviruses: Biomedical Perspectives. Cells 2021, 10, 1619. [Google Scholar] [CrossRef]
  125. Cheung, R.C.F.; Wong, J.H.; Pan, W.; Chan, Y.S.; Yin, C.; Dan, X.; Ng, T.B. Marine Lectins and Their Medicinal Applications. Appl. Microbiol. Biotechnol. 2015, 99, 3755–3773. [Google Scholar] [CrossRef]
  126. Vanderlei, E.S.O.; Patoilo, K.K.N.R.; Lima, N.A.; Lima, A.P.S.; Rodrigues, J.A.G.; Silva, L.M.C.M.; Lima, M.E.P.; Lima, V.; Benevides, N.M.B. Antinociceptive and Anti-Inflammatory Activities of Lectin from the Marine Green Alga Caulerpa cupressoides. Int. Immunopharmacol. 2010, 10, 1113–1118. [Google Scholar] [CrossRef]
  127. Smit, A.J. Medicinal and Pharmaceutical Uses of Seaweed Natural Products: A Review. J. Appl. Phycol. 2004, 16, 245–262. [Google Scholar] [CrossRef]
  128. Mendis, E.; Kim, S.-K. Chapter 1—Present and Future Prospects of Seaweeds in Developing Functional Foods. In Advances in Food and Nutrition Research; Kim, S.-K., Ed.; Marine Medicinal Foods; Academic Press: Cambridge, MA, USA, 2011; Volume 64, pp. 1–15. [Google Scholar]
  129. Wang, F.; Guo, X.-Y.; Zhang, D.-N.; Wu, Y.; Wu, T.; Chen, Z.-G. Ultrasound-Assisted Extraction and Purification of Taurine from the Red Algae Porphyra yezoensis. Ultrason. Sonochem. 2015, 24, 36–42. [Google Scholar] [CrossRef] [PubMed]
  130. Torres, M.D.; Flórez-Fernández, N.; Domínguez, H. Integral Utilization of Red Seaweed for Bioactive Production. Mar. Drugs 2019, 17, 314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Holdt, S.L.; Kraan, S. Bioactive Compounds in Seaweed: Functional Food Applications and Legislation. J. Appl. Phycol. 2011, 23, 543–597. [Google Scholar] [CrossRef]
  132. Mittal, R.; Tavanandi, H.A.; Mantri, V.A.; Raghavarao, K.S.M.S. Ultrasound Assisted Methods for Enhanced Extraction of Phycobiliproteins from Marine Macro-Algae, Gelidium pusillum (Rhodophyta). Ultrason. Sonochem. 2017, 38, 92–103. [Google Scholar] [CrossRef] [PubMed]
  133. Jones, J.H. A Short Guide to Abbreviations and Their Use in Peptide Science. J. Pept. Sci. 1999, 5, 465–471. [Google Scholar] [CrossRef]
  134. Wu, G. Functional Amino Acids in Nutrition and Health. Amino Acids 2013, 45, 407–411. [Google Scholar] [CrossRef] [Green Version]
  135. Samarakoon, K.; Jeon, Y.-J. Bio-Functionalities of Proteins Derived from Marine Algae—A Review. Food Res. Int. 2012, 48, 948–960. [Google Scholar] [CrossRef]
  136. Pimentel, F.B.; Alves, R.C.; Harnedy, P.A.; FitzGerald, R.J.; Oliveira, M.B.P.P. Macroalgal-Derived Protein Hydrolysates and Bioactive Peptides: Enzymatic Release and Potential Health Enhancing Properties. Trends Food Sci. Technol. 2019, 93, 106–124. [Google Scholar] [CrossRef]
  137. Hayes, M. Chapter 14—Seaweeds: A Nutraceutical and Health Food. In Seaweed Sustainability; Tiwari, B.K., Troy, D.J., Eds.; Academic Press: San Diego, CA, USA, 2015; pp. 365–387. ISBN 978-0-12-418697-2. [Google Scholar]
  138. Turner, J.M.; Kodali, R. Should Angiotensin-Converting Enzyme Inhibitors Ever Be Used for the Management of Hypertension? Curr. Cardiol. Rep. 2020, 22, 95. [Google Scholar] [CrossRef]
  139. Sun, S.; Xu, X.; Sun, X.; Zhang, X.; Chen, X.; Xu, N. Preparation and Identification of ACE Inhibitory Peptides from the Marine Macroalga Ulva Intestinalis. Mar. Drugs 2019, 17, 179. [Google Scholar] [CrossRef] [Green Version]
  140. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.-H.; Nitsche, A.; et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020, 181, 271–280.e8. [Google Scholar] [CrossRef] [PubMed]
  141. Tamama, K. Potential Benefits of Dietary Seaweeds as Protection against COVID-19. Nutr. Rev. 2021, 79, 814–823. [Google Scholar] [CrossRef] [PubMed]
  142. Admassu, H.; Gasmalla, M.A.A.; Yang, R.; Zhao, W. Identification of Bioactive Peptides with α-Amylase Inhibitory Potential from Enzymatic Protein Hydrolysates of Red Seaweed (Porphyra spp). J. Agric. Food Chem. 2018, 66, 4872–4882. [Google Scholar] [CrossRef] [PubMed]
  143. Singh, A.-K.; Jatwa, R.; Purohit, A.; Ram, H. Synthetic and Phytocompounds Based Dipeptidyl Peptidase-IV (DPP-IV) Inhibitors for Therapeutics of Diabetes. J. Asian Nat. Prod. Res. 2017, 19, 1036–1045. [Google Scholar] [CrossRef] [PubMed]
  144. Harnedy, P.A.; FitzGerald, R.J. In Vitro Assessment of the Cardioprotective, Anti-Diabetic and Antioxidant Potential of Palmaria palmata Protein Hydrolysates. J. Appl. Phycol. 2013, 25, 1793–1803. [Google Scholar] [CrossRef]
  145. Kim, E.-Y.; Choi, Y.H.; Nam, T.-J. Identification and Antioxidant Activity of Synthetic Peptides from Phycobiliproteins of Pyropia Yezoensis. Int. J. Mol. Med. 2018, 42, 789–798. [Google Scholar] [CrossRef] [Green Version]
  146. 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]
  147. Ahn, C.-B.; Jeon, Y.-J.; Kang, D.-S.; Shin, T.-S.; Jung, B.-M. Free Radical Scavenging Activity of Enzymatic Extracts from a Brown Seaweed Scytosiphon Lomentaria by Electron Spin Resonance Spectrometry. Food Res. Int. 2004, 37, 253–258. [Google Scholar] [CrossRef]
  148. Heo, S.-J.; Park, E.-J.; Lee, K.-W.; Jeon, Y.-J. Antioxidant Activities of Enzymatic Extracts from Brown Seaweeds. Bioresour. Technol. 2005, 96, 1613–1623. [Google Scholar] [CrossRef]
  149. Lakmal, H.C.; Samarakoon, K.W.; Lee, W.; Lee, J.-H.; Abeytunga, D.T.U.; Lee, H.-S.; Jeon, Y.-J. Anticancer and Antioxidant Effects of Selected Sri Lankan Marine Algae. J. Natl. Sci. Found. Sri Lanka 2014, 42, 315–323. [Google Scholar] [CrossRef] [Green Version]
  150. Silva, A.; Silva, S.A.; Carpena, M.; Garcia-Oliveira, P.; Gullón, P.; Barroso, M.F.; Prieto, M.A.; Simal-Gandara, J. Macroalgae as a Source of Valuable Antimicrobial Compounds: Extraction and Applications. Antibiotics 2020, 9, 642. [Google Scholar] [CrossRef] [PubMed]
  151. Singh, R.S.; Walia, A.K. Lectins from Red Algae and Their Biomedical Potential. J. Appl. Phycol. 2018, 30, 1833–1858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Fontenelle, T.P.C.; Lima, G.C.; Mesquita, J.X.; De Souza Lopes, J.L.; De Brito, T.V.; Das Chagas Vieira Júnior, F.; Sales, A.B.; Aragão, K.S.; Souza, M.H.L.P.; Dos Reis Barbosa, A.L.; et al. Lectin Obtained from the Red Seaweed Bryothamnion Triquetrum: Secondary Structure and Anti-Inflammatory Activity in Mice. Int. J. Biol. Macromol. 2018, 112, 1122–1130. [Google Scholar] [CrossRef] [PubMed]
  153. Vasconcelos, M.A.; Arruda, F.V.S.; Carneiro, V.A.; Silva, H.C.; Nascimento, K.S.; Sampaio, A.H.; Cavada, B.; Teixeira, E.H.; Henriques, M.; Pereira, M.O. Effect of Algae and Plant Lectins on Planktonic Growth and Biofilm Formation in Clinically Relevant Bacteria and Yeasts. BioMed Res. Int. 2014, 2014, e365272. [Google Scholar] [CrossRef] [Green Version]
  154. Gonzaga do Nascimento-Neto, L.; Carneiro, R.F.; Da Silva, S.R.; Da Silva, B.R.; Arruda, F.V.S.; Carneiro, V.A.; Do Nascimento, K.S.; Saker-Sampaio, S.; Da Silva, V.A.; Porto, A.L.F.; et al. Characterization of Isoforms of the Lectin Isolated from the Red Algae Bryothamnion Seaforthii and Its Pro-Healing Effect. Mar. Drugs 2012, 10, 1936–1954. [Google Scholar] [CrossRef] [Green Version]
  155. Mori, T.; O’Keefe, B.R.; Sowder, R.C.; Bringans, S.; Gardella, R.; Berg, S.; Cochran, P.; Turpin, J.A.; Buckheit, R.W.; McMahon, J.B.; et al. Isolation and Characterization of Griffithsin, a Novel HIV-Inactivating Protein, from the Red Alga Griffithsia sp. J. Biol. Chem. 2005, 280, 9345–9353. [Google Scholar] [CrossRef] [Green Version]
  156. Lusvarghi, S.; Bewley, C.A. Griffithsin: An Antiviral Lectin with Outstanding Therapeutic Potential. Viruses 2016, 8, 296. [Google Scholar] [CrossRef]
  157. Nascimento da Silva, L.C.; Mendonça, J.S.P.; de Oliveira, W.F.; Batista, K.L.R.; Zagmignan, A.; Viana, I.F.T.; dos Santos Correia, M.T. Exploring Lectin–Glycan Interactions to Combat COVID-19: Lessons Acquired from Other Enveloped Viruses. Glycobiology 2021, 31, 358–371. [Google Scholar] [CrossRef]
  158. O’Keefe, B.R.; Giomarelli, B.; Barnard, D.L.; Shenoy, S.R.; Chan, P.K.S.; McMahon, J.B.; Palmer, K.E.; Barnett, B.W.; Meyerholz, D.K.; Wohlford-Lenane, C.L.; et al. Broad-Spectrum in Vitro Activity and in Vivo Efficacy of the Antiviral Protein Griffithsin against Emerging Viruses of the Family Coronaviridae. J. Virol. 2010, 84, 2511–2521. [Google Scholar] [CrossRef] [Green Version]
  159. Dumay, J.; Morançais, M.; Munier, M.; Le Guillard, C.; Fleurence, J. Chapter Eleven—Phycoerythrins: Valuable Proteinic Pigments in Red Seaweeds. Adv. Bot. Res. 2014, 71, 321–343. [Google Scholar] [CrossRef]
  160. 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]
  161. Sudhakar, M.P.; Jagatheesan, A.; Perumal, K.; Arunkumar, K. Methods of Phycobiliprotein Extraction from Gracilaria Crassa and Its Applications in Food Colourants. Algal Res. 2015, 8, 115–120. [Google Scholar] [CrossRef]
  162. Lee, D.; Nishizawa, M.; Shimizu, Y.; Saeki, H. Anti-Inflammatory Effects of Dulse (Palmaria Palmata) Resulting from the Simultaneous Water-Extraction of Phycobiliproteins and Chlorophyll a. Food Res. Int. 2017, 100, 514–521. [Google Scholar] [CrossRef]
  163. Nguyen, H.P.T.; Morançais, M.; Déléris, P.; Fleurence, J.; Nguyen-Le, C.T.; Vo, K.H.; Dumay, J. Purification of R-Phycoerythrin from a Marine Macroalga Gracilaria Gracilis by Anion-Exchange Chromatography. J. Appl. Phycol. 2020, 32, 553–561. [Google Scholar] [CrossRef]
  164. Le Guillard, C.; Dumay, J.; Donnay-Moreno, C.; Bruzac, S.; Ragon, J.-Y.; Fleurence, J.; Bergé, J.-P. Ultrasound-Assisted Extraction of R-Phycoerythrin from Grateloupia Turuturu with and without Enzyme Addition. Algal Res. 2015, 12, 522–528. [Google Scholar] [CrossRef] [Green Version]
  165. Cotas, J.; Leandro, A.; Pacheco, D.; Gonçalves, A.M.M.; Pereira, L. A Comprehensive Review of the Nutraceutical and Therapeutic Applications of Red Seaweeds (Rhodophyta). Life 2020, 10, 19. [Google Scholar] [CrossRef] [Green Version]
  166. Huang, C.-H.; Chen, W.-C.; Gao, Y.-H.; Chen, G.-W.; Lin, H.-T.V.; Pan, C.-L. Enzyme-Assisted Method for Phycobiliproteins Extraction from Porphyra and Evaluation of Their Bioactivity. Processes 2021, 9, 560. [Google Scholar] [CrossRef]
  167. Kawasaki, A.; Ono, A.; Mizuta, S.; Kamiya, M.; Takenaga, T.; Murakami, S. The Taurine Content of Japanese Seaweed. Adv. Exp. Med. Biol. 2017, 975 Pt 2, 1105–1112. [Google Scholar] [CrossRef]
  168. Hwang, E.-S.; Ki, K.-N.; Chung, H.-Y. Proximate Composition, Amino Acid, Mineral, and Heavy Metal Content of Dried Laver. Prev. Nutr. Food Sci. 2013, 18, 139–144. [Google Scholar] [CrossRef] [Green Version]
  169. Bito, T.; Teng, F.; Watanabe, F. Bioactive Compounds of Edible Purple Laver Porphyra sp. (Nori). J. Agric. Food Chem. 2017, 65, 10685–10692. [Google Scholar] [CrossRef]
  170. Mochizuki, H.; Takido, J.; Oda, H.; Yokogoshi, H. Improving Effect of Dietary Taurine on Marked Hypercholesterolemia Induced by a High-Cholesterol Diet in Streptozotocin-Induced Diabetic Rats. Biosci. Biotechnol. Biochem. 1999, 63, 1984–1987. [Google Scholar] [CrossRef] [PubMed]
  171. Roberfroid, M.B. Global View on Functional Foods: European Perspectives. Br. J. Nutr. 2002, 88, S133–S138. [Google Scholar] [CrossRef] [PubMed]
  172. Siró, I.; Kápolna, E.; Kápolna, B.; Lugasi, A. Functional Food. Product Development, Marketing and Consumer Acceptance—A Review. Appetite 2008, 51, 456–467. [Google Scholar] [CrossRef] [PubMed]
  173. Spence, J.T. Challenges Related to the Composition of Functional Foods. J. Food Compos. Anal. 2006, 19, S4–S6. [Google Scholar] [CrossRef]
  174. Kumar, Y.; Tarafdar, A.; Badgujar, P.C. Seaweed as a Source of Natural Antioxidants: Therapeutic Activity and Food Applications. J. Food Qual. 2021, 2021, 5753391. [Google Scholar] [CrossRef]
  175. Siddiqi, R.A.; Singh, T.P.; Rani, M.; Sogi, D.S.; Bhat, M.A. Diversity in Grain, Flour, Amino Acid Composition, Protein Profiling, and Proportion of Total Flour Proteins of Different Wheat Cultivars of North India. Front. Nutr. 2020, 7, 141. [Google Scholar] [CrossRef]
  176. Prabhasankar, P.; Ganesan, P.; Bhaskar, N.; Hirose, A.; Stephen, N.; Gowda, L.R.; Hosokawa, M.; Miyashita, K. Edible Japanese Seaweed, Wakame (Undaria pinnatifida) as an Ingredient in Pasta: Chemical, Functional and Structural Evaluation. Food Chem. 2009, 115, 501–508. [Google Scholar] [CrossRef]
  177. Qin, Y. 6—Applications of Bioactive Seaweed Substances in Functional Food Products. In Bioactive Seaweeds for Food Applications; Qin, Y., Ed.; Academic Press: Cambridge, MA, USA, 2018; pp. 111–134. ISBN 978-0-12-813312-5. [Google Scholar]
  178. Mouritsen, O.G.; Duelund, L.; Petersen, M.A.; Hartmann, A.L.; Frøst, M.B. Umami Taste, Free Amino Acid Composition, and Volatile Compounds of Brown Seaweeds. J. Appl. Phycol. 2019, 31, 1213–1232. [Google Scholar] [CrossRef]
  179. Laohakunjit, N.; Selamassakul, O.; Kerdchoechuen, O. Seafood-like Flavour Obtained from the Enzymatic Hydrolysis of the Protein by-Products of Seaweed (Gracilaria sp.). Food Chem. 2014, 158, 162–170. [Google Scholar] [CrossRef]
  180. Cardoso, S.M.; Pereira, O.R.; Seca, A.M.L.; Pinto, D.C.G.A.; Silva, A.M.S. Seaweeds as Preventive Agents for Cardiovascular Diseases: From Nutrients to Functional Foods. Mar. Drugs 2015, 13, 6838–6865. [Google Scholar] [CrossRef] [Green Version]
  181. Suganya, T.; Varman, M.; Masjuki, H.H.; Renganathan, S. Macroalgae and Microalgae as a Potential Source for Commercial Applications along with Biofuels Production: A Biorefinery Approach. Renew. Sustain. Energy Rev. 2016, 55, 909–941. [Google Scholar] [CrossRef]
  182. Griffiths, M.; Harrison, S.T.L.; Smit, M.; Maharajh, D. Major Commercial Products from Micro- and Macroalgae. In Algae Biotechnology: Products and Processes; Bux, F., Chisti, Y., Eds.; Green Energy and Technology; Springer International Publishing: Cham, Switzerland, 2016; pp. 269–300. ISBN 978-3-319-12334-9. [Google Scholar]
  183. 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]
  184. Jayasinghe, P.S.; Pahalawattaarachchi, V.; Ranaweera, K.K.D.S. Seaweed Extract as Natural Food Colouring Agents in Jelly Dessert on Chemical Microbiological and Sensory Quality. In Proceedings of the National Aquatic Resources Research and Development Agency (NARA), Colombo, Sri Lanka, 29 March 2016; pp. 121–124. [Google Scholar]
  185. Spolaore, P.; Joannis-Cassan, C.; Duran, E.; Isambert, A. Commercial Applications of Microalgae. J. Biosci. Bioeng. 2006, 101, 87–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Den Banach, J.L.; Hil, E.F.H.; Van der Fels-Klerx, H.J. Food Safety Hazards in the European Seaweed Chain. Compr. Rev. Food Sci. Food Saf. 2020, 19, 332–364. [Google Scholar] [CrossRef]
  187. Arulkumar, A.; Nigariga, P.; Paramasivam, S.; Rajaram, R. Metals Accumulation in Edible Marine Algae Collected from Thondi Coast of Palk Bay, Southeastern India. Chemosphere 2019, 221, 856–862. [Google Scholar] [CrossRef]
  188. Chen, Q.; Pan, X.-D.; Huang, B.-F.; Han, J.-L. Distribution of Metals and Metalloids in Dried Seaweeds and Health Risk to Population in Southeastern China. Sci. Rep. 2018, 8, 3578. [Google Scholar] [CrossRef] [Green Version]
  189. Taylor, V.F.; Li, Z.; Sayarath, V.; Palys, T.J.; Morse, K.R.; Scholz-Bright, R.A.; Karagas, M.R. Distinct Arsenic Metabolites Following Seaweed Consumption in Humans. Sci. Rep. 2017, 7, 3920. [Google Scholar] [CrossRef]
  190. Seng, N.; Lai, S.; Fong, J.; Saleh, M.F.; Cheng, C.; Cheok, Z.Y.; Todd, P.A.; Seng, N.; Lai, S.; Fong, J.; et al. Early Evidence of Microplastics on Seagrass and Macroalgae. Mar. Freshw. Res. 2020, 71, 922–928. [Google Scholar] [CrossRef]
  191. Li, Q.; Feng, Z.; Zhang, T.; Ma, C.; Shi, H. Microplastics in the Commercial Seaweed Nori. J. Hazard. Mater. 2020, 388, 122060. [Google Scholar] [CrossRef]
  192. Naidu, K.A.; Tewari, A.; Joshi, H.V.; Viswanath, S.; Ramesh, H.P.; Rao, S.V. Evaluation of Nutritional Quality and Food Safety of Seaweeds of India. J. Food Saf. 1993, 13, 77–90. [Google Scholar] [CrossRef]
  193. Garcia-Vaquero, M.; Mora, L.; Hayes, M. In Vitro and in Silico Approaches to Generating and Identifying Angiotensin-Converting Enzyme I Inhibitory Peptides from Green Macroalga Ulva Lactuca. Mar. Drugs 2019, 17, 204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Motoyama, K.; Hamada, Y.; Nagashima, Y.; Shiomi, K. Allergenicity and Allergens of Amphipods Found in Nori (Dried Laver). Food Addit. Contam. 2007, 24, 917–922. [Google Scholar] [CrossRef] [PubMed]
  195. Polikovsky, M.; Fernand, F.; Sack, M.; Frey, W.; Müller, G.; Golberg, A. In Silico Food Allergenic Risk Evaluation of Proteins Extracted from Macroalgae Ulva sp. with Pulsed Electric Fields. Food Chem. 2019, 276, 735–744. [Google Scholar] [CrossRef] [PubMed]
  196. Cho, T.J.; Rhee, M.S. Health Functionality and Quality Control of Laver (Porphyra, Pyropia): Current Issues and Future Perspectives as an Edible Seaweed. Mar. Drugs 2020, 18, 14. [Google Scholar] [CrossRef] [Green Version]
  197. Chapman, R.L. Algae: The World’s Most Important “Plants”—An Introduction. Mitig. Adapt. Strateg. Glob. Chang. 2013, 18, 5–12. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Some edible algae species. (A) Ulva lectuca; (B) Codium fragile; (C) Caulerpa sertularioides; (D) Halimeda spp.; (E) P. palmata; (F) Hypnea pannosa (G) Padina spp.; (H) Sargassum spp. Photo courtesy by Piyumika Madhushani.
Figure 1. Some edible algae species. (A) Ulva lectuca; (B) Codium fragile; (C) Caulerpa sertularioides; (D) Halimeda spp.; (E) P. palmata; (F) Hypnea pannosa (G) Padina spp.; (H) Sargassum spp. Photo courtesy by Piyumika Madhushani.
Phycology 02 00012 g001
Figure 2. Simplified diagram for protein extraction from seaweeds.
Figure 2. Simplified diagram for protein extraction from seaweeds.
Phycology 02 00012 g002
Figure 3. Mechanism of ACE inhibition and antihypertension.
Figure 3. Mechanism of ACE inhibition and antihypertension.
Phycology 02 00012 g003
Figure 4. Antidiabetic activity of DPP-IV inhibitors.
Figure 4. Antidiabetic activity of DPP-IV inhibitors.
Phycology 02 00012 g004
Table 1. Edible species of seaweeds.
Table 1. Edible species of seaweeds.
Green AlgaeRed AlgaeBrown Algae
Caulerpa spp.Champia compressaAlaria esculenta
Codium spp.Chondrus crispusAscophyllum nodosum
Enteromorpha spp.Eucheuma denticulatum (formerly Eucheuma spinosum)Durvillaea antarctica
Monostroma spp.Gelidiella acerosaEisenia bicyclis
Ulva spp. (formerly
Enteromorpha spp.)
Gracilaria corticataFucus serratus
Ulva lactuca (formerly Ulva fasciata) (sea lettuce)Gracilaria edulisFucus vesiculosus
Ulva australis (formerly Ulva pertusa)Gracilariopsis longissima (formerly Gracilaria verrucosa)Himanthalia elongata
Mastocarpus stellatusLaminaria digitata
Osmundea pinnatifidaLaminaria hyperborea
Palmaria palmata (dulse)Postelsia palmiformis
Porphyra spp. (nori)Saccharina japonica (formerly Laminaria japonica)
Porphyra laciniataPadina spp.
Porphyra umbilicalisSargassum fusiforme
Pyropia columbina (formerly Porphyra columbina)Sargassum muticum
Solieria robustaSargassum swartzii
Sargassum vulgare
Stoechospermum marginatum
Undaria pinnatifida
Undaria undarioides
Source: Mahadevan, 2015 [8]; Pandey et al., 2020 [9]; Fleurence et al., 2018 [10]; Shannon and Abu-Ghannam [11].
Table 2. The protein content of selected edible seaweeds on a dry weight basis.
Table 2. The protein content of selected edible seaweeds on a dry weight basis.
Seaweed Species or GenusProtein (% of Dry Mass)Reference
Phaeophyceae (Brown Algae)
A. nodosum3–15[10]
A. esculenta9–20[34]
F. serratus3–11[35]
F. vesiculosus12.9[36]
Fucus spp.3–11[37]
H. elongata6–11[38]
L. digitata8–15[10,35]
S. japonica (formerly L. japonica) (kombu)12[39]
U. pinnatifida (wakame)11–24[10,40]
Chlorophyta (Green Algae)
Caulerpa lentillifera19.38[41]
Cladophora rupestris29.8[35]
Ulva intestinalis (formerly Enteromorpha intestinalis)10–18[38]
U. lactuca (formerly U. fasciata) (sea lettuce)8.7–32.7[10,35]
U. australis (formerly U. pertusa)17.5–26.0[10]
Ulva rigida15–25[38]
Ulva rotundata (formerly Ulva pseudorotundata)10.0[42]
Rhodophyta (Red Algae)
Agarophyton vermiculophyllum (previously Gracilaria vermiculophylla)17.0%[43]
C. crispus21–27[10,44]
Gracilaria spp.7–13[45]
G. corticata22.8[46]
G. edulis25.3[46]
Gracilaria salicornia9.58[45]
Gracilaria gracilis31–45[42,47]
O. pinnatifida20.6–27.3[35,48]
P. palmata (dulse)8–35[10,49]
Porphyra spp. (nori or purple laver)33–50[35]
P. columbina (formerly P. columbina)25[50]
N. tenera (formerly P. tenera) (nori)33–47[10]
P. umbilicalis (nori)15–37[38]
Table 3. Comparison of EAA composition (mg/g protein) of selected edible seaweeds with FAO/WHO/UNU, 2007.
Table 3. Comparison of EAA composition (mg/g protein) of selected edible seaweeds with FAO/WHO/UNU, 2007.
Protein
(% dw)
HisIleLeuLysMet + Cys 2MetCysPhe + Tyr 3PheTyrThrTrpValEAA/
NEAA
EAA%
FAO/WHO/UNU, 2007
AA Scoring Pattern 1 [61]
153059452216638 23639
Brown Seaweeds
U. Pinnatifida [53]13.168.250.878.569.6NR30.9NR93.247.146.142.7NR35NRNR
U. pinnatifida [62]12.521.647.3895873.17.365.8110.449.46153.6NR31.1NR48.4
H. elongata [62]520.243.679.360.968.84.364.5113.755.957.854.8NR28.2NR47
A. nodosum [63] a7.649.84858.814.311.2NRNR46.630.715.962.45.854137.7
Green Seaweeds
U. australis (formerly
U. pertusa) [64]
15.48.625.95230.1NRNRNR59.636.722.934.8NR39.10.7242
U. intestinalis (formerly
E. intestinalis) [64]
17.97.425.349.719.6NRNRNR52.135.916.241.7NR40.50.6740
U. rigida [29]10.230.746.18249.419.9NRNR93.9NRNR50.88.860.10.6940.8
U. lactuca (formerly
U. fasciata) [65]
7.113.14072.646.46.16.1093.457.136.362NR70.1NRNR
Red Seaweeds
P. umbilicalis [62]3915.736.576.856.175.98.767.29346.846.257.8NR12.3NR42.4
P. palmata [31]15.218.56581107.834.7286.793.3NRNR47.4NR143.60.8947
P. columbina (formerly
P. columbina) [50] a
24.612.627.173.860.135.716.818.962.53725.559.16.358.50.65NR
Gracilaria changii [66]12.6NR42.353.448.716.1NRNR63.5NRNR49.3NR39.11.61NR
A. vermiculophyllum (formerly
G. vermiculophylla) [29]
13.410.754.984.554.412.9NRNR90.8NRNR58.24.064.10.6740.1
1 FAO/WHO/UNU, 2007 amino acid scoring pattern for adults in mg/g protein. 2 Sulfur amino acids (methionine + cysteine). 3 Aromatic amino acids (phenylalanine + tyrosine). NEAA, non-essential amino acids. Amino acids are represented using the three-letter abbreviation code: His, histidine; Ile, isoleucine; Leu, leucine; Met, methionine; Cys, cysteine; Phe, phenylalanine; Tyr, Tyrosine; Thr, threonine; Trp, tryptophan; Val, valine. NR, Not Reported. a The data was originally expressed as g/100 g protein.
Table 4. Relative digestibility of selected seaweeds.
Table 4. Relative digestibility of selected seaweeds.
SeaweedsDigestibility (%) 1
Red Seaweed
H. charoides [65]88.7
H. japonica [65]88.9
P. palmata [85]85.8
C. crispus [85]84.2
Sarcodiotheca gaudichaudii [85]86.7
Green Seaweed
U. lactuca (formerly U. fasciata) [65] (green seaweed)85.7
Brown Seaweed
A. nodosum [85]78.7
F. vesiculosus [85]78.8
A. esculenta [85]79.2
1 Relative digestibility is expressed as a percentage compared with casein digestibility (100%). The in vitro digestibility was determined by multi-enzyme hydrolysis using porcine pancreatic trypsin, bovine pancreatic chymotrypsin, and porcine intestinal peptidase at pH 8 and 37 °C.
Table 5. Relative digestibility of selected seaweeds using various assays.
Table 5. Relative digestibility of selected seaweeds using various assays.
SeaweedsDigestibility (%) 1Reference
PepsinPancreatinPepsin+
Pancreatin
Pronase
U. australis (formerly U. pertusa) (green)17.066.6-94.8[37,88] a
U. pinnatifida (brown)23.948.1-87.2
N. tenera (formerly P. tenera) (red)56.756.1-78.4
S. japonica (formerly L. japonica) (brown)39.054.0-83.9
P. palmata (red)-56.0--
P. columbina (formerly P. columbina) (red)--74.3-[50] b
P. palmata (red)87.484.987.3-[86] c
N. tenera (formerly P. tenera) (red)73.265.970.2-
E. bicyclis (brown)57.673.257.1-
Sargassum fusiforme (formerly Hizikia fusiformis) (brown)51.865.851.8-
S. japonica (formerly L. japonica) (brown)70.276.172.1-
U. pinnatifida (brown)69.187.568.6-
1 Relative digestibility is expressed as a percentage compared with casein digestibility (100%). The in vitro digestibility was determined by various assays using, a pepsin in acidic pH, pancreatin in pH 7.6, and pronase in pH 8.6 at 37 °C; b not reported; c pepsin at pH below 1.7 and 40 °C followed by pancreatin at pH 7.5 and 40 °C for 24 h.
Table 6. Seaweed protein exhibits potential bioactivities.
Table 6. Seaweed protein exhibits potential bioactivities.
SeaweedBioactive CompoundsPropertiesReferences
Bryopsis spp. (green)Cyclic
depsipeptide
Antimicrobial activity against Mycobacterium tuberculosis[104]
Gracilariopsis lemaneiformis (red)TGAPCR, FQIN [M(O)] CILRAngiotensin-I-converting enzyme (ACE) inhibitory activity[105]
Mazzaella japonica (red)YRD, VSEGLD,
TIMPHPR, GGPAT, SSNDYPI,
SRIYNVKSNG, VDAHY, CPYDWV, YGDPDHY, NLGN, DFGVPGHEP
ACE inhibitory activity[106]
YRD, LDY, LRY, VY, LF, FYACE inhibitory activity[107]
Neopyropia yezoensis (formerly Porphyra yezoensis) (red)Di- and tripeptides
TPDSEAL
ACE inhibition/antihypertensive activity
Antimicrobial activity against Staphylococcus aureus
[108]

[109]
P. palmata (dulse) (red)Peptides derived from phycobiliproteins:
YRD, AGGEY, VYRT, VDHY, IKGHY, LKNPG, LDY, LRY, FEQDWAS
ACE inhibition, antioxidant,[110]
Alcalase, bromelain,
and Promod-derived hydrolysates
dipeptidyl peptidase IV (DPP-IV) inhibitory activities,[111]
Alcalase/Flavourzyme hydrolysatesAntihyperglycemic/antidiabetic potential[112]
Peptides: ILAP,
LLAP, MAGVDHI
DPP-IV inhibitory activities[113]
Papain hydrolysates: NIGKPlatelet-activating factor acetyl-hydrolase (PAF-AH) inhibitory peptides[114]
IRLIIVLMPILMARenin inhibitory activity[115]
SDITRPGGQMAntioxidant[116]
P. dioica (red)Peptides
DYYKR, YLVA
Antioxidant, ACE inhibition, DPP-IV inhibitory activities[117]
P. columbina (formerly P. columbina) (red)PeptidesACE inhibitory, immunosuppressive, antioxidant properties[118]
Saccharina longicruris (formerly Laminaria longicruris) (brown)TITLDVEPSDTIDGVK, ISGLIYEETR, MALSSLPR,
ILVLQSNQIR, ISAILPSR,
IGNGGELPR, LPDAALNR,
EAESSLTGGNGCAK, QVHPDTGISK
Antibacterial activities[119]
Sargassum pallidum (brown)Dipeptides
(aurantiamide, aurantiamideacetate, dia-aurantiamide)
Antibiotic activity in vitro against S. aureus, Staphylococcus epidermidis, and Pseudomonas aeruginosa[120]
Sargassum thunbergia (brown)Iodo-amino acidsPossibly helps in human thyroid metabolism[79]
U. rigida (green)Peptides
ACE inhibition[121]
U. pinnatifida (brown)Di-, tri-, and tetrapeptides
VYIY, AWFY, VW, IW, LW,
YNKLKFYG, YKYY
ACE inhibition/ antihypertensive activity, antioxidant[122,123]
Boodlea coacta (green),
Griffithsia spp. (red)
Lectins
griffithsin
Antiviral effects against human immunodeficiency virus (HIV), Hepatitis C virus and SARS-CoV/ e SARS-CoV-2 by preventing the entry into the host cells[124,125]
Caulerpa cupressoides (green)LectinsAntinociceptive and anti-inflammatory activities[126]
C. fragile (green),
Eucheuma serra (red)
LectinsMitogenic activities, lipogenic activity[127]
Mimica amakusaensis (formerly Eucheuma amakusaense) (red),
Ulva spp. (formerly Enteromorpha spp.) (green)
LectinsInduce apoptosis, metastasis, and cell differentiation in cancer cells, antibiotic, anti-inflammatory, anti-HIV activity, and human platelet aggregation inhibition[128]
N. yezoensis (formerly P. yezoensis) (red)TaurineAntioxidant[129,130]
Saccharina angustata (formerly Laminaria angustata) (brown),
Chondria armata (red)
LaminineHypertensive effect, depress contraction of smooth muscles[131]
C. crispus, Gelidium pusillum, Dasysiphonia japonica (formerly Heterosiphonia japonica),
P. palmata (red)
PhycobiliproteinsAntioxidant, antidiabetic, antitumor, anti-inflammatory, neuro-protective, and hepato-protective properties[132]
Gracilaria tikvahiae,
P. palmata (red)
Phycobilliproteins
(phycocyanins and allphycocyanins)
Anti-inflammatory, liver-protecting, antiviral, antitumor, antiatherosclerosis, lipase activity inhibitor, serum lipid reducing agent, and antioxidant[128]
Abbreviation of amino acids as per Jones, 1999 [133]: A, Ala; R, Arg; N, Asn; D, Asp; C, Cys; Q, Gln; E, Glu; G, Gly; H, His; I, Ile; L, Leu; K, Lys; M, Met; F, Phe; P, Pro; S, Ser; T, Thr; W, Trp; Y, Tyr; V, Val.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Thiviya, P.; Gamage, A.; Gama-Arachchige, N.S.; Merah, O.; Madhujith, T. Seaweeds as a Source of Functional Proteins. Phycology 2022, 2, 216-243. https://doi.org/10.3390/phycology2020012

AMA Style

Thiviya P, Gamage A, Gama-Arachchige NS, Merah O, Madhujith T. Seaweeds as a Source of Functional Proteins. Phycology. 2022; 2(2):216-243. https://doi.org/10.3390/phycology2020012

Chicago/Turabian Style

Thiviya, Punniamoorthy, Ashoka Gamage, Nalin Suranjith Gama-Arachchige, Othmane Merah, and Terrence Madhujith. 2022. "Seaweeds as a Source of Functional Proteins" Phycology 2, no. 2: 216-243. https://doi.org/10.3390/phycology2020012

Article Metrics

Back to TopTop