Next Article in Journal
In Vitro Drug Delivery through the Blood–Brain Barrier Using Cold Atmospheric Plasma
Previous Article in Journal
Structural, Thermal and Mechanical Assessment of Green Compounds with Natural Rubber
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Production of Bioactive Peptides from Microalgae and Their Biological Properties Related to Cardiovascular Disease

by
Ranitha Fernando
,
Xiaohong Sun
and
H. P. Vasantha Rupasinghe
*
Department of Plant, Food, and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, NS B2N 5E3, Canada
*
Author to whom correspondence should be addressed.
Macromol 2024, 4(3), 582-596; https://doi.org/10.3390/macromol4030035
Submission received: 7 June 2024 / Revised: 22 July 2024 / Accepted: 1 August 2024 / Published: 12 August 2024

Abstract

:
Microalgae are a substantial group of unicellular prokaryotic and eukaryotic marine organisms. Due to their high protein content of 50–70%, microalgae have the potential to become a sustainable alternative protein source, as well as aiding in the development of bioactive peptide-based nutraceuticals. A series of major steps are involved in the production of peptides from microalgae, which include the disruption of the microalgal cell wall, the hydrolysis of proteins, and the extraction or isolation of peptides derived from hydrolysis. Physical methods of cell wall disruptions are favored due to the ability to obtain high-quality protein fractions for peptide production. Bioactive peptides are protein fragments of two to twenty amino acid residues that have a beneficial impact on the physiological functions or conditions of human health. Strong scientific evidence exists for the in vitro antioxidant, antihypertensive, and anti-atherosclerotic properties of microalgal peptides. This review is aimed at summarizing the methods of producing microalgal peptides, and their role and mechanisms in improving cardiovascular health. The review reveals that the validation of the physiological benefits of the microalgal peptides in relation to cardiovascular disease, using human clinical trials, is required.

1. Introduction

Food-derived bioactive ingredients have been gaining the interest of both researchers and consumers owing to the increasing awareness of the impact of diet on health. Hence, dietary supplements and nutraceuticals have grown exponentially in the past decade [1,2]. Nevertheless, functional ingredients (bioactives) derived from primary food sources are resource-intensive and expensive. Hence, in order to achieve sustainable development, there is a need for innovative food ingredients that are affordable, sustainable, and have functional properties [3,4,5].
Microalgae are an unexploited natural resource for developing sustainable and innovative functional ingredients [6,7,8,9,10,11]. Several studies have shown that microalgae are a potentially significant source of nutrients and natural biologically active compounds (bioactives) [11]. Microalgae biomass is a good source of many bioactives, such as carotenoids, phycobilins, omega-3 fatty acids, polysaccharides, vitamins, sterols, and bioactive peptides [12,13]. Microalgae have the potential to be a sustainable, alternative protein source for functional ingredients, particularly bioactive peptides, with a protein content of 50–70% [14]. The use of microalgae as a source for extracting functional ingredients such as bioactive peptides is economically and environmentally sustainable, as well as requiring relatively low land area (<2.5 m2 per kg of protein) [15].
The production of bioactive peptides requires processing to break down the microalgae cells and then hydrolyze the released proteins [16]. Generally, the processing of microalgae for the extraction of proteins requires a series of steps, which include the breakdown of the cell wall, the hydrolysis of the protein, and the isolation of the peptides. An emerging area of research involves exploring the bioactivities of microalgae-derived peptides and hydrolysates, such as the antioxidant, antihypertensive, immunomodulatory, antidiabetic, antibacterial/antimicrobial, and anticancer activities [17]. This review discusses the emerging technologies reported for protein extraction from microalgae, the hydrolysis of proteins, and their biological properties related to cardiovascular disease.

2. Microalgae as a Protein Source

Algae are a diverse group of photosynthetic organisms that live in oceans and freshwater bodies. The majority of the members of this group are capable of photosynthesis and producing biological compounds using carbon dioxide and other minerals. Algae are divided into two main categories, namely macroalgae and microalgae, depending on morphology, cellular division characteristics and their pigments [7]. Microalgae are single-cell microscopic organisms that are also now recognized as a sustainable source of dietary proteins. The most abundant microalgal divisions are Chlorophyta (green algae), Chrysophyta (golden algae), Bacillariophyta (diatoms), and Cyanophyta (blue-green algae) [18,19], have been cited more than 200,000 species of microalgae, making them an incredibly diverse group.
Microalgae can be easily cultured under laboratory conditions, but it is challenging to achieve high productivity on a large scale. Microalgae cultivation systems can be broadly classified into two categories: open ponds and photobioreactors. Open pond cultivation is widely used in the industry due to its relatively low construction, maintenance, and operational costs. In contrast, a photobioreactor is an enclosed system used to culture microalgae, preventing the direct exchange of materials between the culture and the environment [20]. There is a difference in the annual productivity of microalgae between these two methods. For example, the annual productivity of Chlorella vulgaris is estimated to be 5–7 kg/m2 in open ponds and approximately 15 kg/m2 in photobioreactors [21]. Harvesting microalgae accounts for 20–30% of the total cost of microalgae production due to high energy and capital demands. The commonly used harvesting methods include filtration, centrifugation, flocculation, and flotation [20,22,23]. Economic feasibility is crucial for the viability of microalgal production. The commercialization of microalgae is hindered by insufficient research funding, small-scale operations, and policy implementation challenges [24].
Microalgae-enriched food products are considered novel foods. They face challenges in consumer acceptance due to their fishy taste and smell, as well as consumers’ food neophobia and individual cultural backgrounds [21]. For example, a recent study assessed Spanish consumers’ knowledge and attitude towards microalgae as food. The results suggested that Spanish consumers perceive microalgae as sustainable, environmentally friendly, nutritious, healthy, and safe. The primary reason for not consuming microalgae as food is the lack of available information on microalgae-based food products [25]. Among the macronutrients of microalgae are 40–70% proteins, 12–30% carbohydrates, and 4–20% lipids. Microalgae are also abundant in micronutrients such as 8–14% carotene and considerable amounts of vitamins B1, B2, B3, B6, B12, E, K, and D [26,27]. The protein content of the microalgae differs based on species, growth phase, and light quality, which can be optimized through nutrient and environmental management [7]. Table 1 shows the general composition of different microalgae that have been studied and reported [26].
Among these different species of microalgae, Arthrospira sp. contains the highest amount of proteins [28]. Among the investigated microalgae species, Athrospira sp., Chlorella sp., Scenedesmus obliquus, and Spirulina sp. have been selected for commercial production [26]. Chlorella species are considered in the literature as the most commonly exploited species due to their high protein content (51–58% DW) and the presence of essential amino acids [19].
The quality of the protein depends on the composition of the amino acids, primarily essential amino acids (EAAs) and digestibility [29]. In general, animal sources of protein are considered complete proteins since they are abundant in EAAs, which are not synthesized by humans [19]. Therefore, microalgae are considered a viable dietary protein with EAAs while meeting the FAO requirements and are equally as good as other major protein sources, such as soybean and egg [19]. However, it is also cited that microalgae proteins may have some deficiencies in terms of lower biological values, net protein utilization, low digestibility, and a poorer protein efficiency ratio than the protein standards considered in FAO/WHO references [26]. Also, microalgae can synthesize all 20 common amino acids, and they are capable of delivering EAAs as an unconventional source for human nutrition [30]. For example, Spirulina platensis is considered a food supplement due to its high protein content [31]. Also, cyanobacteria S. platensis has a protein content of 43–63% (dw) with the abundant amino acid profile of leucine, valine, isoleucine, phenylalanine, tyrosine, methionine, cysteine, and tyrosine [26].
Before being considered safe for human consumption, any novel food item is subjected to a series of toxicological tests to prove its harmlessness. Since microalgae are also grouped with unconventional protein sources, they also must go through this process. When considering the available information on algal protein toxicology, none of the algae tested so far have reported a negative effect [7,8,26,32,33,34,35]. However, it was identified that long-term consumption studies are lacking in this area of study.

3. Production of Bioactive Peptides from Microalgae

Microalgae-based extractions for intracellular constituents such as lipids, proteins, carbohydrates, and pigments for biofuel, food, and nutraceutical applications have attracted the attention of many researchers. Producing bioactive peptides from microalgae involves a series of steps that include cell wall disruption to release the intracellular proteins, the hydrolysis of proteins, and extraction or isolation, resulting in peptides (Figure 1).

3.1. Protein Isolate Hydrolysis

The generation of peptides using isolated proteins can be carried out using three methods [33]. These include enzymatic, fermentative, and chemical methods. The most widely used and effective method for generating bioactive peptides is enzymatic hydrolysis [36]. The enzymatic method is desired due to the high specificity for target peptide sequences and does not involve the use of toxic organic solvents. The enzymatic hydrolysis process can be scaled up for industrial processes [36,37]. However, controlling hydrolysis is essential, as otherwise, extensive hydrolysis can lead to the loss of biological activity in the produced bioactive peptides. One disadvantage of enzymatic hydrolysis is the high cost of the enzymes required [36]. The enzymes that are employed are gastrointestinal proteases such as pepsin, pancreatin, and trypsin, proteolytic enzymes from microbial production such as alcalase, neutrase, flavourzyme and plant-sourced proteases such as papain, ficin, bromelain. However, it is more effective to use combinations or cocktails of enzymes to generate bioactive peptides [37,38]. When developing an efficient process for bioactive peptide production, many factors are to be considered, including the nature of the protein, the enzyme–substrate ratio, the pre-treatment of protein before hydrolysis, the degree of hydrolysis, and protease specificity [37,39]. Recently, in silico enzymatic hydrolysis has been used to predict the production of peptide sequences from microalgae using Peptide Cutter. The potential bioactivities of these in silico-generated peptides can be predicted using PeptideRanker, and their novelty can be assessed against the reported peptide sequences in the BIOPEP-UWM database (http://www.uwm.edu.pl/biochemia/index.php/pl/biopep; accessed on 10 September 2023) [40,41,42].
Fermentation is a metabolic process wherein sugars are converted to acids, gases, and alcohol through the action of fermenting microorganisms [6]. The most commonly used microorganisms are bacteria, yeasts, and fungi [6]. This technique is not widely applied when considering peptide production from microalgae. However, fermentation has been used for bioethanol production using microalgae hydrolysate [43]. Fermentation is a common approach to obtaining peptides from dairy products. Here, lactic acid bacteria (LAB) that possess cell wall-bound proteases and intracellular peptidases exert proteolytic activities on milk protein [38,44]. Htet and colleagues have used LAB fermentation to produce 2-pyrone 4, 6-dicarboxylic acid, using microalgae hydrolysate as a fermentation medium [45].
Chemical methods also involve using strong acids and bases to hydrolyze the protein sequence. This is a cheap and secure method. However, it has significant hindrances, such as a loss of functionality in the resulting hydrolysates, a lack of specificity, inferior nutrient quality, and undesirable products [46]. Acid hydrolysis has some disadvantages, such as the oxidation of cysteine and methionine, the deamination of asparagine and glutamine to aspartate and glutamate, and the breakdown of serine and threonine [37]. For these reasons, the hydrolysis of proteins using chemicals is not favored. In the literature, it is shown that the enzymatic method is the most commonly used and sought-after protein hydrolysis method, because of its specificity in cleavage positions in peptide bonds. It increases the reproducibility and predictability of generating peptides. The chemical method is less favored because of its inherent contamination problem and its inability to control hydrolysis, which leads to less reproducibility. The fermentative method lacks research on microalgae. However, since the fermentation method also uses exogenous and endogenous bacterial enzymes for hydrolysis, it also has specificity and, therefore, reproducibility. However, more research in this area is needed to draw a conclusion.

3.2. Isolation of Peptides

Cell disruption helps the release of cellular contents such as lipids, polysaccharides, nucleic acids, and phytochemicals. Hydrolysis helps the breakdown of protein molecules into peptides [47]. After the hydrolysis, the next step is to ensure the separation of peptides from other interfering compounds. Separation or fractionation techniques (centrifugation, precipitation, electrophoresis, ultrafiltration, and chromatographic techniques) help in obtaining pure protein/peptide fractions [48]. Some studies have used this step in between the cell wall disruption and protein hydrolysis step, as it helps to reduce the inferences from the other molecules in the solution [49,50]. However, it can be assumed that performing fractionation after cell wall disruption is better, since it provides an opportunity to obtain protein fractions with fewer interferences from the solutions.
Centrifugation is a technique that uses the gravitational field to separate proteins from other cell constituents, such as membranes, mitochondria, and nuclei, based on their different molecular weight and other physical characteristics [51]. The efficiency of centrifugation can be improved by a density gradient fractionation using sucrose. Precipitation involves the usage of organic solvents, organic polymers, and salts, or pH and temperature adjustments [52]. Electrophoresis is another technique where proteins can be separated based on size, shape, charge, or charge/mass ratio [48]. In electrophoresis, protein molecules are separated based on their charge due to the supplied electric field [53]. Electrophoresis in a free solution or macroporous gel (1–2% agarose) can be used to separate proteins according to net electric charge. Polyacrylamide gels can be used to separate proteins based on the molecular weight of proteins [52]. Ultrafiltration is a membrane separation technique that can be used to isolate macromolecules of 103–106 Da using a semi-permeable membrane. Chromatography is also a fractionation technique that can be used to separate proteins of varying properties. Size exclusion chromatography separates proteins based on size and shape; ion exchange chromatography is based on isoelectric points; reverse-phase chromatography is based on hydrophobicity, and affinity chromatography uses ligands for binding proteins. Even though these methods are extensively used for food-related studies, it is unusual to find these being used in microalgae-related protein extractions. These methods are used in combination with most of those from literature, as a result of the difficulty due to the interactions with the rest of the materials in the solution.

4. Peptides Derived from Microalgae

Bioactive peptides can be defined as protein fragments with health benefits beyond their nutritional values and have a positive impact on the body’s function or condition [40]. The size of the peptide may vary from 2 to 20 amino acid residues, and they possess multifunctional properties [54]. These peptides are not active when they are present within the sequence of the parent protein, but upon release through hydrolysis by proteolytic digestive enzymes or by proteolytic microorganisms, they provide several bioactivities within the human body [55]. Physiologically active peptides are naturally produced during the gastrointestinal digestion of proteins by lactic acid bacteria [55]. Enzymatic hydrolysis is the commonly used method of bioactive peptide production [16]. Gastrointestinal enzymes such as trypsin and pepsin are commonly used. When the hydrolysate contains adequate quantities of the active peptides, the product can be used as a therapeutic substance to manage a specific disease, or else hydrolysates can be used to isolate specific peptides and used in an available drug delivery system (e.g., encapsulation) to deliver to the body. Mass spectrophotometry is a commonly used technique for peptide identification. Table 2 illustrates the reported peptides identified from microalgae and their bioactivities.
The oral administration of protein hydrolysates or bioactive peptides may affect the major body systems, such as the cardiovascular, digestive, immune and nervous systems, depending on the amino acid sequence of the peptides [67]. Bioactive peptides are reported to exhibit antimicrobial, antioxidative, anti-inflammatory, antithrombotic, antihypertensive, and immunomodulatory activities in the human body [35,68,69,70,71]. Most of the body functions are modulated via the interaction of specific amino acid sequences of bioactive peptides, which can be used in a wide range of therapeutic applications in the future [67,69].

5. Cardiovascular Health Promotion from Microalgae Peptides

5.1. Antioxidant Biopeptides

As mentioned earlier, there are several peptides identified as possessing antioxidant capacity [16,72,73]. Antioxidant peptides contain 5–16 amino acid residues [72]. The advantages of the peptides generated from foods are that they are safe, have a low cost, are easy to absorb and have a relatively high stability [54,68]. Oxidative damage due to elevated levels of reactive oxygen species (ROS) has been implicated in many chronic disorders, such as certain cancers, diabetes mellitus, hypertension, and neurodegenerative and inflammatory diseases, as well as aging [74]. Therefore, the antioxidant activities of bioactive peptides have been shown to contribute to oxidative homeostasis.
The oxidation of proteins can cause modifications to the amino acid R-groups and the polymerization and/or fragmentation of proteins [75]. For example, the oxidation of bovine serum albumin with hydroxyl radicals in the absence of oxygen has resulted in extensive protein cross-linking while in the presence of oxygen, and some alteration of protein size has been observed [76]. The abstraction of alpha-hydrogen atoms to backbone carbons causes the breakage of peptide or amide bonds [76,77]. The oxidative damage to a given amino acid residue by free radical attack is determined in large by its R-group. However, solvent accessibility and the chemical properties of adjacent residues can also influence oxidation [78]. Although all 20 common amino acids are potentially oxidizable, the most vulnerable amino acids to oxidation are those containing either nucleophilic sulfur-containing side chains, i.e., cysteine and methionine, or aromatic side chains, i.e., tryptophan, tyrosine, and phenylalanine, from which hydrogen is easily abstracted. In addition, the imidazole-containing side chain of histidine is also oxidatively labile [77]. The cleavage of proteins, resulting in the peptides, improves the expression of amino acid R-groups, so the antioxidative properties of the peptides are comparatively higher than the proteins. Overall, the mode of the antioxidant function of proteins and peptides is due to their complex reactions of inactivating ROSs, scavenging free radicals, reducing hydroperoxides, chelating prooxidative transition metals, and enzymatically eliminating specific oxidants [77].
Marine microalgae species reported having high diversity and the ability to thrive in extreme environments, such as low light intensities and high oxygen concentrations, which make them more self-protective towards oxidative damage [79]. However, Chlorella sp., Navicula sp., and Spirulina sp. are among the widely studied microalgae species for antioxidative biopeptides (Figure 2). These species have been evaluated for potent antioxidant capacity in vitro and cell-based assays. Antioxidant peptides have been produced by the hydrolysis of Chlorella protothecoides using several extracellular bacterial proteases. The resulting hydrolysates showed high antioxidant activity: 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical scavenging and a significant reduction of hydrogen peroxide and hydroxyl free radical. However, peptide sequences have not been identified in this study [80]. Ko and colleagues have reported the extraction of a hexapeptide (Leu-Asn-Gly-Asp-Val-Trp) from Chlorella ellipsiodea using peptic hydrolysis, which shows peroxyl radical and DPPH radical scavenging activities in vitro, and intracellular radical scavenging activity in monkey kidney cells [64]. Protein hydrolysates produced from C. ellipsiodea and Tetraselmis suecica using different microbial proteases were found to have moderate free radical scavenging capacity [65]. The neutrase and kojizyme hydrolysates of Tetrasel missuecica exhibited the ability to scavenge hydrogen peroxide in African green monkey kidney cells in vitro [65]. When papain, pepsin, α-chymotrypsin, pronase-E, and neutrase were used to hydrolyze Navicula incerta, antioxidant hydrolysates were yielded. For example, two peptides (Pro-Gly-Trp-Asn-Gln-Trp-Phe-Leu and Val-Glu-Val-Leu-Pro-Pro-Ala-Glu-Leu) generated using a papain reaction demonstrated antioxidant effects in HepG2/CYP2E1 cells [56,81]. Moreover, a Chlorella pyrenoidosa-derived peptide mixture showed antioxidant properties in skin fibroblasts. When UV irradiation-exposed skin cells were treated with the Chlorella-derived peptides (5 and 10 mg/L), 100% cytoprotection over a period of 72 h was observed [66]. However, it is not clear whether microalgal peptides were taken into cells to exhibit their antioxidant activity or only acted extracellularly and resulted in cytoprotection. Enzymatic hydrolysis is the most commonly used method of producing antioxidant peptides from microalgae. However, more evidence is required to validate the physiological antioxidant capacity of the microalgae-derived peptides, using in vivo studies of their applications in combating oxidative stress-associated disorders. The additional investigations will help to justify these unicellular organisms as an alternative source of dietary antioxidants to be used in the prevention of an array of human disorders.

5.2. Antihypertensive Biopeptides

Abnormally high blood pressure is one of the major risk factors for CVD [67]. Angiotensin I-converting enzyme (ACE) is one of the main enzymes that are responsible for the blood pressure regulating pathway called the renin–angiotensin system (RAS) and electrolyte homeostasis [82,83]. The RAS and kinin–arginine–nitric oxide system pathways are important for controlling blood pressure [84]. The enzyme renin, which is released by the circulation of the kidney, cleaves the inactive peptide angiotensinogen secreted from the liver, resulting in peptide angiotensin-I. Subsequently, ACE acts on angiotensin-I to produce angiotensin-II and inactivates the vasodilator bradykinin, leading to a rise in blood pressure. Therefore, blood pressure can be controlled by inhibiting either renin or ACE. Therefore, renin and ACE inhibitory factors have become primary targets in the treatment of hypertension. Synthetic ACE inhibitors such as Captopril have been used as antihypertensive drugs. Interestingly, some diseases, genetic or environmental factors, can stimulate the RAS and upregulate the levels of ACE in the human body, and high levels of angiotensin II result in blood vessel constriction, leading to hypertension [85,86]. Bioactive peptides have been shown to inhibit ACE in the renin–angiotensin–aldosterone system (RAAS) (Figure 2). Microalgal peptides play a significant role in the regulation of blood volume, blood pressure, and fluid balance regulation [87]. The most effective antihypertensive peptides seem to contain two to twenty amino acid residues and hydrophobic amino acid residues, such as proline [39,88,89]. The acting mechanisms of microalgae-derived peptides are related to ACE and rennin inhibitions.
In addition, the kinin–nitric oxide (NO) system, a complex network involving peptide kinins, plays a significant role in blood pressure regulation. Kinins, such as bradykinin and kallidin, are generated through the kallikrein–kinin system. Bradykinin activates Ca2+/calmodulin-dependent endothelial nitric oxide synthase (eNOS), which synthesizes nitric oxide (NO) and citrulline from arginine, resulting in vasodilation and a drop in blood pressure. It is shown that eNOS knockout and NO deficiency could result in clinical hypertension [90,91]. The regulation of the kinin–NO system by bioactive peptides is not well explored.
As in the antioxidant peptides, most antihypertensive peptides are also derived from a few microalgal sources, such as Chlorella vulgaris, Chlorella ellipsiodea, Spirulina platensis, and Nannochloropsis oculata. Many extracted peptides are illustrated in Table 2. These antihypertensive peptides have been used in vitro and in some animal studies to evaluate their antihypertensive properties. The peptides produced by the hydrolysis of Chlorella vulgaris using pepsin were found to possess in vitro and in vivo antihypertensive properties [57,59]. The by-products of Chlorella vulgaris processing have been used to produce an 11-amino acid residue peptide (Val-Glu-Cys-Tyr-Gly-Pro-Asn-Arg-Pro-Gln-Phe), which inhibited ACE activity in a dose-dependent manner [49]. The peptide has inhibited ACE activity non-competitively, which could be due to the non-specific binding of the peptide to the ACE protein. Pepsin hydrolysates of Chlorella vulgaris and Spirulina platensis yielded several short-chain peptides that were shown to be effective within 6 h after administration in reducing the high blood pressure of spontaneously hypertensive rats (SHRs) [59]. In addition, microalgal protein hydrolysates derived from Bellerochea malleus exhibited antihypertensive activity, resulting in a decrease of 17 mmHg after five days of oral administration to male SHRs [92]. Compared to the antioxidant activity of microalgae peptides, antihypertensive properties are widely studied. However, similar to antioxidant activity, further studies are needed to establish the physiological importance of the microalgal peptides, especially for the identified antihypertensive peptides in vitro, as in vitro activity does not necessarily translate into physiological antihypertensive effects. Recently, a bioactive hydrolysate was generated using Viscozyme® and Alcalase from the red microalga Porphyridium species, and this hydrolysate inhibited in vitro COX-1 activity and exhibited antihypertensive activity in SHRs [93].

5.3. Anti-Atherosclerotic Biopeptides

Atherogenesis is the process leading to the formation of atherosclerotic plaques in the walls of arteries, which can result in CVDs, such as coronary artery disease, stroke, myocardial infarction and peripheral artery disease [94,95]. Atherosclerosis is a progressive inflammatory disorder of the arterial wall that is a common cause of cardiovascular morbidity and mortality [96]. Increased cardiovascular risk is associated with hypercholesterolemia, hypertension, diabetes mellitus, and cigarette smoking [97,98].
Bioactive peptides have been reported to have antithrombotic and antiatherosclerotic effects [33,99]. Numerous peptides generated from in vivo and in vitro hydrolysis and the enzymatic digestion of milk, soybean, and other primary food sources have shown cardiovascular benefits [100,101,102]. However, only a few studies have researched peptides extracted from microalgae and indicated positive associations with the above-mentioned conditions of atherogenesis and atherosclerosis. Shih and colleagues were able to isolate an undecapeptide (Val-Glu-Cys-Tyr-Gly-Pro-Asn-Arg-Pro-Gln-Phe) from Chlorella pyrenoidosa, which can significantly suppress the levels of E-selectin, intercellular cell adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), monocyte chemoattractant protein (MCP-1), and endothelin-1 (ET-1) gene expression (Figure 2) [103]. The suppression of these expressions has been shown to be a successful method of blocking the development of atherosclerosis [104]. Vo and Kim were able to isolate two peptides P1 (Leu-Asp-Ala-Val-Asn-Arg) and P2 (Met-Met-Leu-Asp) from the gastric enzymatic hydrolysate of Spirulina maxima [105]. P1 and P2 inhibited the production of adhesion molecules, such as P-selectin and E-selectin, through the down-regulation of the expression of early growth response factor 1 (Egr-1) via the histamine receptor and PKCδ-dependent MAPKs activation pathway. It was also found that P1 and P2 have anti-inflammatory activity, by reducing ROS production and inhibiting histamine release and IL-8 expression. These two pieces of research have depicted that micro algae-derived peptide plays a role in adhesion molecule reduction and anti-inflammation. A recent study isolated a novel nonapeptide, Glu-Met-Phe-Gly-Thr-Ser-Ser-Glu-Thr, from the microalgae Isochrysis zhanjiangensis, which demonstrated anti-apoptotic and anti-inflammatory effects in oxidized low-density lipoprotein (ox-LDL)-induced human umbilical vein endothelial cells (HUVECs), suggesting its potential to alleviate atherosclerosis. However, this study only employed the ox-LDL-mediated HUVEC model, lacking an animal model [106]. Reported studies related to the anti-atherosclerotic property of microalgal peptides are very scarce. Therefore, additional research in this field is required to identify the effects that microalgal peptides have on atherogenesis.

6. Concluding Remarks and Future Directions

Algae are divided mainly into two groups, microalgae, and macroalgae. Microalgae are unicellular, microscopic organisms. Microalgae are an incredibly diverse group with around 200,000 species. Microalgae consist of 40–70% proteins, 4–20% carbohydrates, and 8–14% lipids and other nutrients. The literature reveals that the composition of microalgae greatly varies depending on their species, growth phase, light, available nutrients, and environmental stress. For example, Anthrospira species are one of the sources with the highest protein content reported. Chlorella is found to be the genera that are mostly investigated. Interestingly, when looking at the protein content of microalgae, they consist of all essential amino acids; hence, the quality of the proteins meets the FAO/WHO reference values. Because of these factors, microalgae continued to be an untapped, underutilized, and sustainable protein source.
Peptide production comprises four main steps that involve the release of the intracellular protein from cells and the hydrolysis of the protein into smaller peptides. The complexity of the cell walls of microalgae requires vigorous treatments to break down the cell walls. Three types of approaches can be used to disrupt the cell walls, chemical, mechanical, and biological/biochemical. These different techniques have different advantages and disadvantages associated with their specificities. The main disadvantage associated with the chemical treatment is contamination or inability to efficiently remove the chemicals being used for the cell wall breakdown. Heat generation and high associated energy costs are the major drawbacks of the physical or mechanical treatments. Biological or biochemical methods suffer from the costs of commercially available enzymes and low yield compared to the time taken. Regarding the treatments to break down cell walls, the hydrolysis of the protein can be carried out using three methods—enzymatic, fermentative, and chemical methods. The most commonly used method to obtain bioactive peptides is enzymatic hydrolysis, due to its high specificity for the target peptide sequence. Commercially available enzymes have been widely used, such as pepsin, pancreatin, trypsin, alcalase, neutrase, flavourzyme, papain, ficin, and bromelain. Fermentation with microorganisms such as bacteria, yeasts, and fungi is a less costly method compared to enzymatic hydrolysis. The exogenous enzymes of the microorganisms used are utilized for the cleavage of the peptide bonds. Acid and base hydrolysis is the chemical method for protein hydrolysis, with the significant drawback of protein denaturation due to pH changes. The literature has cited many separation or isolation techniques for peptides, including centrifugation, precipitation, electrophoresis, ultrafiltration, and chromatographic techniques. However, many reported investigations have employed chromatographic techniques. Even though other methods are used in food-related research, their usage in the microalgae field is very uncommon.
Many multifunctional properties have been reported in bioactive peptides of sizes ranging from two to twenty amino acids. Among them, antimicrobial, antioxidative, antithrombotic, antihypertensive, and immunomodulatory activities are reported. These peptides have been used as hydrolysates and as isolated peptides to assess their bioactivities. There are several peptides identified as possessing antioxidant capacity, which has been widely studied. The antioxidant activity of peptides derived from microalgae has been investigated to mitigate oxidative stress created by ROSs. Chlorella sp., Navicula sp., and Spirulina sp. are among the widely studied microalgae species for antioxidative biopeptides. It is crucial to identify the amino acid sequences that possess the antioxidant capacity and then evaluate them to produce bulk hydrolysate. With more data regarding the specific sequences derived from microalgae, it is possible to conclude their use in combating oxidative stress-associated diseases. It is also identified that bioactive peptides inhibit ACE in the renin–angiotensin–aldosterone system, indicating their antihypertensive properties. It is also noted that only a few microalgae species have been studied for the extraction of antihypertensive peptides. Among them, Chlorella vulgaris, Chlorella ellipsiodea, Spirulina platensis, and Nannochloropsis oculate are the most prominent. Anti-inflammatory and adhesion molecule reduction properties have been reported in peptides extracted from Chlorella pyrenoidosa and Spirulina maxima.
Through this review, it is identified that long-term human consumption studies on microalgal-based protein and peptide products are lacking; even though they are considered safe due to their natural origin, future research in this area can facilitate the commercialization of microalgal peptide products as food additives or nutraceuticals. Currently, the use of algicidal methods for microalgae cell wall disruption is in the infant stage. Therefore, further research in this area towards safe and sustainable algicidal methods for cell wall disruption may shed light on efficient ways of developing bioactive peptides. For the hydrolysis of protein isolates, fermentation techniques have the potential to develop an economical method compared to the enzymatic method. Therefore, the continuation of research on fermentation applications will help develop new methods to break down microalgal cell walls to extract proteins for health applications. Microalgal peptides need to be further assessed for their bioavailability, antioxidant activity and cytoprotection in targeted tissues and organs. To date, only a few studies have identified the amino acid sequences of microalgal peptides with antioxidant properties. Microalgae peptides have been extensively investigated for their antihypertensive properties. However, further studies are needed to validate the antihypertensive peptides using placebo-controlled randomized human studies. The biostability, bioaccessibility, and bioavailability of microalgae-derived bioactive peptides need to be investigated in detail to establish their full potential in cardiovascular health. In future, the application of bioinformatics, in silico and artificial intelligence tools can be used collectively to predict the production of bioactive peptides using the knowledge derived from microalgae-derived peptides. To expand the market for microalgae peptide-based products, it is important to understand consumer perception toward functional food and nutraceutical products derived from microalgae in future.

Author Contributions

Conceptualization, R.F., X.S. and H.P.V.R.; Methodology, R.F.; Formal Analysis, R.F.; Investigation, R.F.; Resources, H.P.V.R.; Writing—Original Draft Preparation, R.F.; Writing—Review and Editing, X.S. and H.P.V.R.; Visualization, R.F. and H.P.V.R.; Supervision, H.P.V.R.; Project Administration, H.P.V.R.; Funding Acquisition, H.P.V.R. All authors have read and agreed to the published version of the manuscript.

Funding

Financial support from the Natural Sciences and Engineering Research Council (NSERC grant number RGPIN-2023-03324) of Canada.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Vicentini, A.; Liberatore, L.; Mastrocola, D. Functional Foods: Trends and Development. Ital. J. Food Sci. 2016, 28, 338–352. [Google Scholar]
  2. Yadav, S.; Malik, K.; Moore, J.M.; Kamboj, B.R.; Malik, S.; Malik, V.K.; Arya, S.; Singh, K.; Mahanta, S.; Bishnoi, D.K. Valorisation of Agri-Food Waste for Bioactive Compounds: Recent Trends and Future Sustainable Challenges. Molecules 2024, 29, 2055. [Google Scholar] [CrossRef] [PubMed]
  3. Granato, D.; Barba, F.J.; Kovačević, D.B.; Lorenzo, J.M.; Cruz, A.G.; Putnik, P. Functional Foods: Product Development, Technological Trends, Efficacy Testing, and Safety. Annu. Rev. Food Sci. Technol. 2020, 11, 93–118. [Google Scholar] [CrossRef]
  4. Neenu, R.; Tiwari, S.; Jethani, H.; Chauhan, V.S. The Commercial Microalgae-Based Foods. In Handbook of Food and Feed from Microalgae: Production, Application, Regulation, and Sustainability; Elsevier: Amsterdam, The Netherlands, 2023; pp. 489–507. ISBN 978-0-323-99196-4. [Google Scholar]
  5. Bhatnagar, P.; Gururani, P.; Joshi, S.; Singh, Y.P.; Vlaskin, M.S.; Kumar, V. Enhancing the Bio-Prospects of Microalgal-Derived Bioactive Compounds in Food Industry: A Review. In Biomass Conversion Biorefinery; Springer: Berlin/Heidelberg, Germany, 2023. [Google Scholar] [CrossRef]
  6. Khan, M.I.; Shin, J.H.; Kim, J.D. The Promising Future of Microalgae: Current Status, Challenges, and Optimization of a Sustainable and Renewable Industry for Biofuels, Feed, and Other Products. Microb. Cell Factories 2018, 17, 36. [Google Scholar] [CrossRef]
  7. Villarruel-Lopez, A.; Ascencio, F.; Nuno, K. Microalgae, a Potential Natural Functional Food Source—A Review. Pol. J. Food Nutr. Sci. 2017, 67, 251–263. [Google Scholar] [CrossRef]
  8. Abd El-Hack, M.E.; Abdelnour, S.; Alagawany, M.; Abdo, M.; Sakr, M.A.; Khafaga, A.F.; Mahgoub, S.A.; Elnesr, S.S.; Gebriel, M.G. Microalgae in Modern Cancer Therapy: Current Knowledge. Biomed. Pharmacother. 2019, 111, 42–50. [Google Scholar] [CrossRef] [PubMed]
  9. Mosibo, O.K.; Ferrentino, G.; Udenigwe, C.C. Microalgae Proteins as Sustainable Ingredients in Novel Foods: Recent Developments and Challenges. Foods 2024, 13, 733. [Google Scholar] [CrossRef]
  10. Zhou, L.; Li, K.; Duan, X.; Hill, D.; Barrow, C.; Dunshea, F.; Martin, G.; Suleria, H. Bioactive Compounds in Microalgae and Their Potential Health Benefits. Food Biosci. 2022, 49, 101932. [Google Scholar] [CrossRef]
  11. Papadaki, S.; Tricha, N.; Panagiotopoulou, M.; Krokida, M. Innovative Bioactive Products with Medicinal Value from Microalgae and Their Overall Process Optimization through the Implementation of Life Cycle Analysis—An Overview. Mar. Drugs 2024, 22, 152. [Google Scholar] [CrossRef]
  12. Saha, S.; Murray, P. Exploitation of Microalgae Species for Nutraceutical Purposes: Cultivation Aspects. Fermentation 2018, 4, 46. [Google Scholar] [CrossRef]
  13. Çelekli, A.; Özbal, B.; Bozkurt, H. Challenges in Functional Food Products with the Incorporation of Some Microalgae. Foods 2024, 13, 725. [Google Scholar] [CrossRef]
  14. Plaza, M.; Herrero, M.; Cifuentes, A.; Ibáñez, E. Innovative Natural Functional Ingredients from Microalgae. J. Agric. Food Chem. 2009, 57, 7159–7170. [Google Scholar] [CrossRef]
  15. Caporgno, M.P.; Mathys, A. Trends in Microalgae Incorporation into Innovative Food Products with Potential Health Benefits. Front. Nutr. 2018, 5, 58. [Google Scholar] [CrossRef] [PubMed]
  16. Siddhnath; Surasani, V.K.R.; Singh, A.; Singh, S.M.; Hauzoukim; Murthy, L.N.; Baraiya, K.G. Bioactive Compounds from Micro-Algae and Its Application in Foods: A Review. Discov. Food 2024, 4, 27. [Google Scholar] [CrossRef]
  17. O’Connor, J.; Garcia-Vaquero, M.; Meaney, S.; Tiwari, B.K. Bioactive Peptides from Algae: Traditional and Novel Generation Strategies, Structure-Function Relationships, and Bioinformatics as Predictive Tools for Bioactivity. Mar. Drugs 2022, 20, 317. [Google Scholar] [CrossRef] [PubMed]
  18. Norton, T.A.; Melkonian, M.; Andersen, R.A. Algal Biodiversity. Phycologia 1996, 35, 308–326. [Google Scholar] [CrossRef]
  19. Bleakley, S.; Hayes, M. Algal Proteins: Extraction, Application, and Challenges Concerning Production. Foods 2017, 6, 33. [Google Scholar] [CrossRef]
  20. Tan, J.S.; Lee, S.Y.; Chew, K.W.; Lam, M.K.; Lim, J.W.; Ho, S.-H.; Show, P.L. A Review on Microalgae Cultivation and Harvesting, and Their Biomass Extraction Processing Using Ionic Liquids. Bioengineered 2020, 11, 116–129. [Google Scholar] [CrossRef] [PubMed]
  21. Yang, S.; Wang, Y.; Wang, J.; Cheng, K.; Liu, J.; He, Y.; Zhang, Y.; Mou, H.; Sun, H. Microalgal Protein for Sustainable and Nutritious Foods: A Joint Analysis of Environmental Impacts, Health Benefits and Consumer’s Acceptance. Trends Food Sci. Technol. 2024, 143, 104278. [Google Scholar] [CrossRef]
  22. Uduman, N.; Qi, Y.; Danquah, M.K.; Forde, G.M.; Hoadley, A. Dewatering of Microalgal Cultures: A Major Bottleneck to Algae-Based Fuels. J. Renew. Sustain. Energy 2010, 2, 012701. [Google Scholar] [CrossRef]
  23. Barros, A.I.; Gonçalves, A.L.; Simões, M.; Pires, J.C.M. Harvesting Techniques Applied to Microalgae: A Review. Renew. Sustain. Energy Rev. 2015, 41, 1489–1500. [Google Scholar] [CrossRef]
  24. Loke Show, P. Global Market and Economic Analysis of Microalgae Technology: Status and Perspectives. Bioresour. Technol. 2022, 357, 127329. [Google Scholar] [CrossRef] [PubMed]
  25. Lafarga, T.; Rodríguez-Bermúdez, R.; Morillas-España, A.; Villaró, S.; García-Vaquero, M.; Morán, L.; Sánchez-Zurano, A.; González-López, C.V.; Acién-Fernández, F.G. Consumer Knowledge and Attitudes towards Microalgae as Food: The Case of Spain. Algal Res. 2021, 54, 102174. [Google Scholar] [CrossRef]
  26. Becker, E.W. Micro-Algae as a Source of Protein. Biotechnol. Adv. 2007, 25, 207–210. [Google Scholar] [CrossRef]
  27. Guo, X.; Wang, Q.; Wu, Y.; Liu, X.; Gong, Z. Comprehensive Insights into Microalgae Proteins: Nutritional Profiles and Innovative Applications as Sustainable Alternative Proteins in Health and Food Sciences. Food Hydrocoll. 2024, 154, 110112. [Google Scholar] [CrossRef]
  28. Capelli, B.; Cysewski, G. Potential Health Benefits of Spirulina Microalgae: A Review of the Existing Literature. Nutrafoods 2010, 9, 19–26. [Google Scholar] [CrossRef]
  29. Boisen, S.; Eggum, B.O. Critical Evaluation of in Vitro Methods for Estimating Digestibility in Simple-Stomach Animals. Nutr. Res. Rev. 1991, 4, 141–162. [Google Scholar] [CrossRef]
  30. Spolaore, P.; Joannis-Cassan, C.; Duran, E.; Isambert, A. Commercial Applications of Microalgae. J. Biosci. Bioeng. 2006, 101, 87–96. [Google Scholar] [CrossRef]
  31. Templeton, D.W.; Laurens, L.M.L. Nitrogen-to-Protein Conversion Factors Revisited for Applications of Microalgal Biomass Conversion to Food, Feed and Fuel. Algal Res. 2015, 11, 359–367. [Google Scholar] [CrossRef]
  32. Fan, X.; Bai, L.; Zhu, L.; Yang, L.; Zhang, X. Marine Algae-Derived Bioactive Peptides for Human Nutrition and Health. J. Agric. Food Chem. 2014, 62, 9211–9222. [Google Scholar] [CrossRef]
  33. Ejike, C.E.C.C.; Ezeorba, T.P.C.; Ajah, O.; Udenigwe, C.C. Big Things, Small Packages: An Update on Microalgae as Sustainable Sources of Nutraceutical Peptides for Promoting Cardiovascular Health. Glob. Chall. 2023, 7, 2200162. [Google Scholar] [CrossRef] [PubMed]
  34. Ejike, C.; Collins, S.; Balasuriya, N.; Swanson, A.; Mason, B.; Udenigwe, C. Prospects of Microalgae Proteins in Producing Peptide-Based Functional Foods for Promoting Cardiovascular Health. Trends Food Sci. Technol. 2017, 59, 30–36. [Google Scholar] [CrossRef]
  35. Kim, S.K.; Ngo, D.H.; Vo, T.S. Marine Fish-Derived Bioactive Peptides as Potential Antihypertensive Agents, 1st ed.; Elsevier Inc.: Amsterdam, The Netherlands, 2012; Volume 65, ISBN 9780124160033. [Google Scholar]
  36. Saadi, S.; Saari, N.; Anwar, F.; Abdul Hamid, A.; Ghazali, H.M. Recent Advances in Food Biopeptides: Production, Biological Functionalities and Therapeutic Applications. Biotechnol. Adv. 2015, 33, 80–116. [Google Scholar] [CrossRef] [PubMed]
  37. Nasri, M. Protein Hydrolysates and Biopeptides: Production, Biological Activities, and Applications in Foods and Health Benefits. A Review, 1st ed.; Elsevier Inc.: Amsterdam, The Netherlands, 2017; Volume 81. [Google Scholar]
  38. Pihlanto, A. Bioactive Peptides: Functionality and Production. Agro Food Ind. Hi-Tech 2006, 17, 24–26. [Google Scholar] [CrossRef]
  39. Udenigwe, C.C.; Aluko, R.E. Food Protein-Derived Bioactive Peptides: Production, Processing, and Potential Health Benefits. J. Food Sci. 2012, 77, R11–R24. [Google Scholar] [CrossRef] [PubMed]
  40. Kose, A.; Oncel, S.S. Design of Melanogenesis Regulatory Peptides Derived from Phycocyanin of the Microalgae Spirulina platensis. Peptides 2022, 152, 170783. [Google Scholar] [CrossRef] [PubMed]
  41. Hayes, M.; Mora, L.; Lucakova, S. Identification of Bioactive Peptides from Nannochloropsis Oculata Using a Combination of Enzymatic Treatment, in Silico Analysis and Chemical Synthesis. Biomolecules 2022, 12, 1806. [Google Scholar] [CrossRef] [PubMed]
  42. Tejano, L.A.; Peralta, J.P.; Yap, E.E.S.; Panjaitan, F.C.A.; Chang, Y.-W. Prediction of Bioactive Peptides from Chlorella Sorokiniana Proteins Using Proteomic Techniques in Combination with Bioinformatics Analyses. Int. J. Mol. Sci. 2019, 20, 1786. [Google Scholar] [CrossRef]
  43. Al Abdallah, Q.; Nixon, B.T.; Fortwendel, J.R. The Enzymatic Conversion of Major Algal and Cyanobacterial Carbohydrates to Bioethanol. Front. Energy Res. 2016, 4, 36. [Google Scholar] [CrossRef]
  44. Beermann, C.; Hartung, J. Physiological Properties of Milk Ingredients Released by Fermentation. Food Funct. 2013, 4, 185–199. [Google Scholar] [CrossRef]
  45. Htet, A.N.; Noguchi, M.; Ninomiya, K.; Tsuge, Y.; Kuroda, K.; Kajita, S.; Masai, E.; Katayama, Y.; Shikinaka, K.; Otsuka, Y.; et al. Application of Microalgae Hydrolysate as a Fermentation Medium for Microbial Production of 2-Pyrone 4,6-Dicarboxylic Acid. J. Biosci. Bioeng. 2018, 125, 717–722. [Google Scholar] [CrossRef] [PubMed]
  46. Ovissipour, M.; Safari, R.; Motamedzadegan, A.; Shabanpour, B. Chemical and Biochemical Hydrolysis of Persian Sturgeon (Acipenser persicus) Visceral Protein. Food Bioprocess Technol. 2012, 5, 460–465. [Google Scholar] [CrossRef]
  47. Nirmal, N.; Khanashyam, A.C.; Shah, K.; Awasti, N.; Sajith Babu, K.; Ucak, İ.; Afreen, M.; Hassoun, A.; Tuanthong, A. Plant Protein-Derived Peptides: Frontiers in Sustainable Food System and Applications. Front. Sustain. Food Syst. 2024, 8, 1292297. [Google Scholar] [CrossRef]
  48. Martínez-Maqueda, D.; Hernández-Ledesma, B.; Amigo, L.; Miralles, B.; Gómez-Ruiz, J.Á. Extraction/Fractionation Techniques for Proteins and Peptides and Protein Digestion. In Proteomics in Foods: Principles and Applications; Toldrá, F., Nollet, L.M.L., Eds.; Springer: Boston, MA, USA, 2013; pp. 21–50. ISBN 978-1-4614-5626-1. [Google Scholar]
  49. Sheih, I.C.; Fang, T.J.; Wu, T.K. Isolation and Characterisation of a Novel Angiotensin I-Converting Enzyme (ACE) Inhibitory Peptide from the Algae Protein Waste. Food Chem. 2009, 115, 279–284. [Google Scholar] [CrossRef]
  50. Zambrowicz, A.; Timmer, M.; Polanowski, A.; Lubec, G.; Trziszka, T. Manufacturing of Peptides Exhibiting Biological Activity. Amino Acids 2013, 44, 315–320. [Google Scholar] [CrossRef]
  51. Scopes, R.K. Protein Purification: Principles and Practice; Springer Science & Business Media: Berlin, Germany, 2013. [Google Scholar]
  52. Ersson, B.; Rydén, L.; Janson, J.-C. Introduction to Protein Purification. In Protein Purification: Principles, High Resolution Methods, and Applications; Wiley: Hoboken, NJ, USA, 2011; pp. 1–22. [Google Scholar]
  53. Liu, S.; Li, Z.; Yu, B.; Wang, S.; Shen, Y.; Cong, H. Recent advances on protein separation and purification methods. Adv. Colloid Interface Sci. 2020, 284, 102254. [Google Scholar] [CrossRef]
  54. Martinez-Villaluenga, C.; Peñas, E.; Frias, J. Bioactive Peptides in Fermented Foods: Production and Evidence for Health Effects. Fermented Foods Health Dis. Prev. 2017, 23–47. [Google Scholar] [CrossRef]
  55. Mora, L.; Aristoy, M.-C.; Toldrá, F. Bioactive Peptides. Encycl. Food Chem. 2019, 381–389. [Google Scholar] [CrossRef]
  56. Kang, K.H.; Ryu, B.M.; Kim, S.K.; Qian, Z.J. Characterization of Growth and Protein Contents from Microalgae Navicula Incerta with the Investigation of Antioxidant Activity of Enzymatic Hydrolysates. Food Sci. Biotechnol. 2011, 20, 183–191. [Google Scholar] [CrossRef]
  57. Sheih, I.C.; Wu, T.K.; Fang, T.J. Antioxidant Properties of a New Antioxidative Peptide from Algae Protein Waste Hydrolysate in Different Oxidation Systems. Bioresour. Technol. 2009, 100, 3419–3425. [Google Scholar] [CrossRef]
  58. Sheih, I.C.; Fang, T.J.; Wu, T.K.; Lin, P.H. Anticancer and Antioxidant Activities of the Peptide Fraction from Algae Protein Waste. J. Agric. Food Chem. 2010, 58, 1202–1207. [Google Scholar] [CrossRef] [PubMed]
  59. Suetsuna, K.; Chen, J.-R. Identification of Antihypertensive Peptides from Peptic Digest of Two Microalgae, Chlorella Vulgaris and Spirulina Platensis. Mar. Biotechnol. 2001, 3, 305–309. [Google Scholar] [CrossRef]
  60. 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]
  61. 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]
  62. Suetsuna, K.; Maekawa, K.; Chen, J.R. Antihypertensive Effects of Undaria pinnatifida (Wakame) Peptide on Blood Pressure in Spontaneously Hypertensive Rats. J. Nutr. Biochem. 2004, 15, 267–272. [Google Scholar] [CrossRef] [PubMed]
  63. Saito, M.; Nagoya, K.; HAGino, H.; HAWAi, M. Antihypertensive Effect of Oligopeptides Derived from Nori on Rats. Jpn. J. Med. Pharm. Sci 2000, 43, 529–538. [Google Scholar]
  64. Ko, S.C.; Kim, D.; Jeon, Y.J. Protective Effect of a Novel Antioxidative Peptide Purified from a Marine Chlorella ellipsoidea Protein against Free Radical-Induced Oxidative Stress. Food Chem. Toxicol. 2012, 50, 2294–2302. [Google Scholar] [CrossRef]
  65. Lee, S.H.; Chang, D.U.; Lee, B.J.; Jeon, Y.J. Antioxidant Activity of Solubilized Tetraselmis Suecica and Chlorella Ellipsoidea by Enzymatic Digests. J. Food Sci. Nutr. 2009, 14, 21–28. [Google Scholar] [CrossRef]
  66. Shih, M.F.; Cherng, J.Y. Protective Effects of Chlorella-Derived Peptide Against UVC-Induced Cytotoxicity through Inhibition of Caspase-3 Activity and Reduction of the Expression of Phosphorylated FADD and Cleaved PARP-1 in Skin Fibroblasts. Molecules 2012, 17, 9116–9128. [Google Scholar] [CrossRef]
  67. Yurika, N.; Montuori, E.; Lauritano, C. Marine Microalgal Products with Activities against Age-Related Cardiovascular Diseases. Mar. Drugs 2024, 22, 229. [Google Scholar] [CrossRef]
  68. Bhat, Z.F.; Kumar, S.; Bhat, H.F. Bioactive Peptides of Animal Origin: A Review. J. Food Sci. Technol. 2015, 52, 5377–5392. [Google Scholar] [CrossRef] [PubMed]
  69. Hong, F.; Ming, L.; Yi, S.; Zhanxia, L.; Yongquan, W.; Chi, L. The Antihypertensive Effect of Peptides: A Novel Alternative to Drugs? Peptides 2008, 29, 1062–1071. [Google Scholar] [CrossRef] [PubMed]
  70. Li, G.H.; Le, G.W.; Shi, Y.H.; Shrestha, S. Angiotensin I-Converting Enzyme Inhibitory Peptides Derived from Food Proteins and Their Physiological and Pharmacological Effects. Nutr. Res. 2004, 24, 469–486. [Google Scholar] [CrossRef]
  71. Elbandy, M. Anti-Inflammatory Effects of Marine Bioactive Compounds and Their Potential as Functional Food Ingredients in the Prevention and Treatment of Neuroinflammatory Disorders. Molecules 2023, 28, 2. [Google Scholar] [CrossRef] [PubMed]
  72. Sarmadi, B.H.; Ismail, A. Antioxidative Peptides from Food Proteins: A Review. Peptides 2010, 31, 1949–1956. [Google Scholar] [CrossRef] [PubMed]
  73. Senadheera, T.R.L.; Dave, D.; Shahidi, F. Antioxidant Potential and Physicochemical Properties of Protein Hydrolysates from Body Parts of North Atlantic Sea Cucumber (Cucumaria frondosa). Food Prod. Process. Nutr. 2021, 3, 3. [Google Scholar] [CrossRef]
  74. Valko, M.; Leibfritz, D.; Moncol, J.; Cronin, M.T.D.; Mazur, M.; Telser, J. Free Radicals and Antioxidants in Normal Physiological Functions and Human Disease. Int. J. Biochem. Cell Biol. 2007, 39, 44–84. [Google Scholar] [CrossRef]
  75. King, T.A.; Kandemir, J.M.; Walsh, S.J.; Spring, D.R. Photocatalytic Methods for Amino Acid Modification. Chem. Soc. Rev. 2021, 50, 39–57. [Google Scholar] [CrossRef] [PubMed]
  76. Dean, R.T.; Fu, S.; Stocker, R.; Davies, M.J. Biochemistry and Pathology of Radical-Mediated Protein Oxidation. Biochem. J. 1997, 324, 1–18. [Google Scholar] [CrossRef]
  77. Elias, R.J.; Kellerby, S.S.; Decker, E.A. Antioxidant Activity of Proteins and Peptides. Crit. Rev. Food Sci. Nutr. 2008, 48, 430–441. [Google Scholar] [CrossRef]
  78. Davies, M.J. Radical-Mediated Protein Oxidation. In The Pathology of Protein Oxidation; Oxford Academic: Oxford, UK, 1997; pp. 207–218. [Google Scholar]
  79. Guedes, A.C.; Amaro, H.M.; Malcata, F.X. Microalgae as Sources of High Added-Value Compounds-a Brief Review of Recent Work. Biotechnol. Prog. 2011, 27, 597–613. [Google Scholar] [CrossRef]
  80. Olena, Z.; Yang, Y.; Yin, T.T.; Yan, X.T.; Rao, H.L.; Xun, X.; Dong, X.; Wu, C.L.; He, H.L. Simultaneous Preparation of Antioxidant Peptides and Lipids from Microalgae by Pretreatment with Bacterial Proteases. Bioresour. Technol. 2022, 348, 126759. [Google Scholar] [CrossRef]
  81. Kang, K.-H.; Qian, Z.-J.; Ryu, B.; Karadeniz, F.; Kim, D.; Kim, S.-K. Antioxidant Peptides from Protein Hydrolysate of Microalgae Navicula Incerta and Their Protective Effects in Hepg2/CYP2E1 Cells Induced by Ethanol. Phytother. Res. 2012, 26, 1555–1563. [Google Scholar] [CrossRef] [PubMed]
  82. Barrett, K.E.; Barman, S.M.; Boitano, S.; Brooks, H. Ganong’s Review of Medical Physiology. In LANGE Basic Science; McGraw-Hill: New York, NY, USA, 2012. [Google Scholar]
  83. Cutrell, S.; Alhomoud, I.S.; Mehta, A.; Talasaz, A.H.; Van Tassell, B.; Dixon, D.L. ACE-Inhibitors in Hypertension: A Historical Perspective and Current Insights. Curr. Hypertens. Rep. 2023, 25, 243–250. [Google Scholar] [CrossRef]
  84. Chen, Z.-Y.; Peng, C.; Jiao, R.; Wong, Y.M.; Yang, N.; Huang, Y. Anti-Hypertensive Nutraceuticals and Functional Foods. J. Agric. Food Chem. 2009, 57, 4485–4499. [Google Scholar] [CrossRef] [PubMed]
  85. Unger, T. The Role of the Renin-Angiotensin System in the Development of Cardiovascular Disease. Am. J. Cardiol. 2002, 89, 3–9. [Google Scholar] [CrossRef]
  86. Alper, A.B.; Calhoun, D.A. Contemporary Management of Refractory Hypertension. Curr. Hypertens. Rep. 1999, 1, 402–407. [Google Scholar] [CrossRef]
  87. Fitzgerald, C.; Gallagher, E.; Tasdemir, D.; Hayes, M. Heart Health Peptides from Macroalgae and Their Potential Use in Functional Foods. J. Agric. Food Chem. 2011, 59, 6829–6836. [Google Scholar] [CrossRef]
  88. García, M.C.; Puchalska, P.; Esteve, C.; Marina, M.L. Vegetable Foods: A Cheap Source of Proteins and Peptides with Antihypertensive, Antioxidant, and Other Less Occurrence Bioactivities. Talanta 2013, 106, 328–349. [Google Scholar] [CrossRef] [PubMed]
  89. Hernández-Ledesma, B.; Del Mar Contreras, M.; Recio, I. Antihypertensive Peptides: Production, Bioavailability and Incorporation into Foods. In Advances in Colloid and Interface Science; Elsevier: Amsterdam, The Netherlands, 2011; Volume 165, pp. 23–35. [Google Scholar]
  90. Huang, P.L.; Huang, Z.; Mashimo, H.; Bloch, K.D.; Moskowitz, M.A.; Bevan, J.A.; Fishman, M.C. Hypertension in Mice Lacking the Gene for Endothelial Nitric Oxide Synthase. Nature 1995, 377, 239–242. [Google Scholar] [CrossRef]
  91. Thomas, G.D.; Zhang, W.; Victor, R.G. Nitric Oxide Deficiency as a Cause of Clinical Hypertension: Promising New Drug Targets for Refractory Hypertension. J. Am. Med. Assoc. 2001, 285, 2055–2057. [Google Scholar] [CrossRef] [PubMed]
  92. Barkia, I.; Al-Haj, L.; Abdul Hamid, A.; Zakaria, M.; Saari, N.; Zadjali, F. Indigenous Marine Diatoms as Novel Sources of Bioactive Peptides with Antihypertensive and Antioxidant Properties. Int. J. Food Sci. Technol. 2019, 54, 1514–1522. [Google Scholar] [CrossRef]
  93. Hayes, M.; Aluko, R.E.; Aurino, E.; Mora, L. Generation of Bioactive Peptides from Porphyridium Sp. and Assessment of Their Potential for Use in the Prevention of Hypertension, Inflammation and Pain. Mar. Drugs 2023, 21, 422. [Google Scholar] [CrossRef] [PubMed]
  94. Zilversmit, D.B. Atherogenesis: A Postprandial Phenomenon. Circulation 1979, 60, 473–485. [Google Scholar] [CrossRef] [PubMed]
  95. Munro, J.M.; Cotran, R.S. Biology of Disease. The Pathogenesis of Atherosclerosis: Atherogenesis and Inflammation. Lab. Investig. 1988, 58, 249–261. [Google Scholar] [PubMed]
  96. Channon, K.M. The Endothelium and the Pathogenesis of Atherosclerosis. Medicine 2006, 34, 173–177. [Google Scholar] [CrossRef]
  97. Ribeiro, F.; Alves, A.J.; Duarte, J.A.; Oliveira, J. Is Exercise Training an Effective Therapy Targeting Endothelial Dysfunction and Vascular Wall Inflammation? Int. J. Cardiol. 2010, 141, 214–221. [Google Scholar] [CrossRef] [PubMed]
  98. Yusuf, S.; Joseph, P.; Rangarajan, S.; Islam, S.; Mente, A.; Hystad, P.; Brauer, M.; Kutty, V.R.; Gupta, R.; Wielgosz, A.; et al. Modifiable Risk Factors, Cardiovascular Disease, and Mortality in 155 722 Individuals from 21 High-Income, Middle-Income, and Low-Income Countries (PURE): A Prospective Cohort Study. Lancet 2020, 395, 795–808. [Google Scholar] [CrossRef] [PubMed]
  99. Senadheera, T.R.L.; Hossain, A.; Shahidi, F. Marine Bioactives and Their Application in the Food Industry: A Review. Appl. Sci. 2023, 13, 12088. [Google Scholar] [CrossRef]
  100. Marcone, S.; Belton, O.; Fitzgerald, D.J. Milk-Derived Bioactive Peptides and Their Health Promoting Effects: A Potential Role in Atherosclerosis. Br. J. Clin. Pharmacol. 2017, 83, 152–162. [Google Scholar] [CrossRef]
  101. Singh, B.P.; Vij, S.; Hati, S. Functional Significance of Bioactive Peptides Derived from Soybean. Peptides 2014, 54, 171–179. [Google Scholar] [CrossRef] [PubMed]
  102. Sirtori, C.R.; Galli, C.; Anderson, J.W.; Arnoldi, A. Nutritional and Nutraceutical Approaches to Dyslipidemia and Atherosclerosis Prevention: Focus on Dietary Proteins. Atherosclerosis 2009, 203, 8–17. [Google Scholar] [CrossRef] [PubMed]
  103. Shih, M.; Chen, L.; Cherng, J. Chlorella 11-Peptide Inhibits the Production of Macrophage-Induced Adhesion Molecules and Reduces Endothelin-1 Expression and Endothelial Permeability. Mar. Drugs 2013, 11, 3861–3874. [Google Scholar] [CrossRef] [PubMed]
  104. Winkles, J.A.; Alberts, G.F.; Brogi, E.; Libby, P. Endothelin-1 and Endothelin Receptor mRNA Expression in Normal and Atherosclerotic Human Arteries. Biochem. Biophys. Res. Commun. 1993, 191, 1081–1088. [Google Scholar] [CrossRef] [PubMed]
  105. Vo, T.S.; Kim, S.K. Down-Regulation of Histamine-Induced Endothelial Cell Activation as Potential Anti-Atherosclerotic Activity of Peptides from Spirulina Maxima. Eur. J. Pharm. Sci. 2013, 50, 198–207. [Google Scholar] [CrossRef]
  106. Pei, Y.; Lui, Y.; Cai, S.; Zhou, C.; Hong, P.; Qian, Z.-J. A Novel Peptide Isolated from Microalgae Isochrysis Zhanjiangensis Exhibits Anti-Apoptosis and Anti-Inflammation in Ox-LDL Induced HUVEC to Improve Atherosclerosis. Plant Foods Hum. Nutr. 2022, 77, 181–189. [Google Scholar] [CrossRef]
Figure 1. The standard process for the production of bioactive peptides from microalgae.
Figure 1. The standard process for the production of bioactive peptides from microalgae.
Macromol 04 00035 g001
Figure 2. Potential biological properties related to cardiovascular disease of microalgae-derived bioactive peptides. ROSs, reactive oxygen species; ACE, angiotensin-converting enzyme; RAAS, renin–angiotensin–aldosterone system, ICAM-1, intercellular cell adhesion molecule-1; VCAM-1, vascular cell adhesion molecule-1; MCP-1, monocyte chemoattractant protein-1; ET-1, endothelin-1.
Figure 2. Potential biological properties related to cardiovascular disease of microalgae-derived bioactive peptides. ROSs, reactive oxygen species; ACE, angiotensin-converting enzyme; RAAS, renin–angiotensin–aldosterone system, ICAM-1, intercellular cell adhesion molecule-1; VCAM-1, vascular cell adhesion molecule-1; MCP-1, monocyte chemoattractant protein-1; ET-1, endothelin-1.
Macromol 04 00035 g002
Table 1. Macronutrient composition of different microalgae (% of dry matter).
Table 1. Macronutrient composition of different microalgae (% of dry matter).
AlgaProteinCarbohydratesLipids
Anabaena cylindrica43–5625–304–7
Aphanizomenon flos-aquae62233
Arthrospira maxima60–7113–166–7
Chlamydomonas rheinhardii481721
Chlorella pyrenoidosa57262
Chlorella vulgaris51–5812–1714–22
Dunaliella salina57326
Euglena gracilis39–6114–1814–20
Porphyridium cruentum28–3940–579–14
Scenedesmus obliquus50–5610–1712–14
Spirogyra sp.6–2033–6411–21
Spirulina platensis46–638–144–9
Synechococcus sp.631511
Source: [26].
Table 2. Examples of bioactive peptides derived from microalgae.
Table 2. Examples of bioactive peptides derived from microalgae.
Amino Acid Sequence of Bioactive Peptide Source of PeptideReported BioactivityHydrolytic MethodReference
Glu-, Asp-, Lys-, Arg-Navicula incertaAntioxidativeEnzymatic–Pepsin[56]
Pro-Gly-Trp-Asn-Gln-Trp-Phe-Leu Val-Glu-Val-Leu-Pro-Pro-Ala-Glu-Leu,
Val-Glu-Val-Leu-Pro-Pro-Ala-Glu-Leu
Navicula incertaHepatic fibrosis inhibitory effect (Cytotoxicity in HepG2/CYP2E1 cells)Enzymatic–Papain[56]
Val-Glu-Cys-Iyr-Gly-Pro-Asn-Arg-Pro-Glu-PheChlorella vulgarisAntioxidativeEnzymatic–Pepsin[57]
Val-Glu-Cys-Tyr-Gly-Pro-Asn-Arg-Pro-Glu-PheChlorella vulgarisACE inhibitory, Superoxide radical quenchingEnzymatic–Pepsin[49]
Val-Glu-Cys-Iyr-Gly-Pro-Asn-Arg-Pro-Glu-PheChlorella vulgarisAnti-proliferationEnzymatic–Pepsin[58]
Ile-Val-Val-GluChlorella vulgarisACE-I inhibitoryEnzymatic–Pepsin[59]
Ile-Ala-GluSpirulina platensisACE-I inhibitoryEnzymatic–Pepsin[59]
Ala-Ile-Tyr-LysUndaria pinnatifidaAntihypertensiveEnzymatic–Pepsin[60]
Val-TyrUndaria pinnatifidaAntihypertensiveEnzymatic–Proteases[61]
Tyr-His, Lys-Trp, Lys-Tyr, Lys-Phe, Phe-Tyr, Val-Trp, Val-Phe, Ile-Tyr, Ile-Trp, Val-TyrUndaria pinnatifidaAntihypertensiveHot water extract [62]
Ala-Lys-Tyr-Ser-TyrPorphyra yezoensisAntihypertensivePepsin[63]
Leu-Asn-Gly-Asp-Val-TrpChlorella ellipsiodeaAntioxidant activitiesPepsin[64]
Pro-Gly-Trp-Asn-Gln-Trp-Phe-Leu and Val-Glu-Val-Leu-Pro-Pro-Ala-Glu-LeuNavicula incertaAntioxidant activitiesPapain, pepsin, α-chymotrypsin, pronase-E, and neutrase[65]
Peptide mixtureChlorella pyrenoidosaAntioxidant activities-[66]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Fernando, R.; Sun, X.; Rupasinghe, H.P.V. Production of Bioactive Peptides from Microalgae and Their Biological Properties Related to Cardiovascular Disease. Macromol 2024, 4, 582-596. https://doi.org/10.3390/macromol4030035

AMA Style

Fernando R, Sun X, Rupasinghe HPV. Production of Bioactive Peptides from Microalgae and Their Biological Properties Related to Cardiovascular Disease. Macromol. 2024; 4(3):582-596. https://doi.org/10.3390/macromol4030035

Chicago/Turabian Style

Fernando, Ranitha, Xiaohong Sun, and H. P. Vasantha Rupasinghe. 2024. "Production of Bioactive Peptides from Microalgae and Their Biological Properties Related to Cardiovascular Disease" Macromol 4, no. 3: 582-596. https://doi.org/10.3390/macromol4030035

Article Metrics

Back to TopTop