Next Article in Journal
Total Phenolic Levels, In Vitro Antioxidant Properties, and Fatty Acid Profile of Two Microalgae, Tetraselmis marina Strain IMA043 and Naviculoid Diatom Strain IMA053, Isolated from the North Adriatic Sea
Previous Article in Journal
Evaluation of the Preservation and Digestion of Seal Meat Processed with Heating and Antioxidant Seal Meat Hydrolysates
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Astaxanthin from Crustaceans and Their Byproducts: A Bioactive Metabolite Candidate for Therapeutic Application

by
Vida Šimat
1,*,
Nikheel Bhojraj Rathod
2,
Martina Čagalj
1,
Imen Hamed
3 and
Ivana Generalić Mekinić
4
1
University Department of Marine Studies, University of Split, R. Boškovića 37, HR-21000 Split, Croatia
2
Department of Post Harvest Management of Meat, Poultry and Fish, PG Institute of Post Harvest Management (Balasaheb Sawant Konkan Krishi Vidyapeeth, Dapoli), Killa-Roha, Dist. Raigad 402 116, Maharashtra State, India
3
Department of Biotechnology and Food Science, NTNU—Norwegian University of Science and Technology, 7491 Trondheim, Norway
4
Department of Food Technology and Biotechnology, Faculty of Chemistry and Technology, University of Split, R. Boškovića 35, HR-21000 Split, Croatia
*
Author to whom correspondence should be addressed.
Mar. Drugs 2022, 20(3), 206; https://doi.org/10.3390/md20030206
Submission received: 14 February 2022 / Revised: 8 March 2022 / Accepted: 10 March 2022 / Published: 12 March 2022

Abstract

:
In recent years, the food, pharma, and cosmetic industries have shown considerable interest in bioactive molecules of marine origin that show high potential for application as nutraceuticals and therapeutic agents. Astaxanthin, a lipid-soluble and orange-reddish-colored carotenoid pigment, is one of the most investigated pigments. Natural astaxanthin is mainly produced from microalgae, and it shows much stronger antioxidant properties than its synthetic counterpart. This paper aims to summarize and discuss the important aspects and recent findings associated with the possible use of crustacean byproducts as a source of astaxanthin. In the last five years of research on the crustaceans and their byproducts as a source of natural astaxanthin, there are many new findings regarding the astaxanthin content in different species and new green extraction protocols for its extraction. However, there is a lack of information on the amounts of astaxanthin currently obtained from the byproducts as well as on the cost-effectiveness of the astaxanthin production from the byproducts. Improvement in these areas would most certainly contribute to the reduction of waste and reuse in the crustacean processing industry. Successful exploitation of byproducts for recovery of this valuable compound would have both environmental and social benefits. Finally, astaxanthin’s strong biological activity and prominent health benefits have been discussed in the paper.

Graphical Abstract

1. Introduction

The aquaculture and seafood industry productions are rising in line with the increased demand for seafood products. These sectors generate large amounts of byproducts that can be transformed into other products or used for the recovery of parts/byproducts from which high-value compounds could be extracted. The high-value compounds from the seafood industry byproducts include polyunsaturated fatty acids (PUFAs), bioactive peptides, chitin, collagen, pigments, marine enzymes, etc. The byproducts of the crustacean processing industry are considered especially underutilized. Approximately 15 million tons of crustaceans are captured (38%) and aquacultured (62%, mainly in Asian countries) around the world annually [1]. These products are considered a healthy diet choice as they are a good source of various nutrients. However, the yield in crustacean meat production is quite low, from 20 to 25%; thus, up to 80% of the raw materials are wasted [2]. This presents not only a serious ecological problem but also a financial burden for the producers. Organisms used for processing, such as prawns, shrimps, crabs, and lobsters, generate byproducts comprised of heads, exoskeletons, and appendages [3]. These byproducts are composed mainly of organic matter and contain valuable compounds, such as proteins, chitin, chitosan, lipids, carotenoids (pigments), and minerals [4]. The recovery of the valuable compounds from the crustacean byproducts could contribute to the new products/ingredient development and the sustainability of the whole sector as well as have environmental benefits. Carotenoid astaxanthin is a compound of great interest to researchers and to the food, feed, pharmaceutical, and nutraceutical industries. Due to its potent antioxidant properties declared to be stronger than vitamin E, vitamin C, and β-carotene, many health and therapeutic benefits [5,6,7,8,9,10,11] and possible application as a natural coloring in food and feed [12], astaxanthin has become a highly demanded metabolite. Besides, it is used as a feed ingredient in farming systems for obtaining desirable pigmentation of fish and seafood and improving their immunity [13]. Furthermore, its bioactivity has been associated with beneficial health effects for humans, namely its anticardiovascular, anti-inflammatory, and antiaging potentials as well as other favorable cosmetic benefits due to the improvement of skin moisture and elasticity [14]. In 2020, the global astaxanthin market size was estimated at USD 1371.24 million, and it is expected to grow [15]. The main source used for natural astaxanthin industrial production is freshwater microalgae Haematococcus pluvialis, and the cost of its production is estimated at 880 €·kg−1 [16]. The main producer is North America (67%), and the global nutraceutical industry is the biggest consumer of natural astaxanthin [17]. On the other hand, the market value of astaxanthin is determined by the purity of the product and ranges from 2200–6620 €·kg−1 to a high 13.240 €·kg−1 for astaxanthin from H. pluvialis [17]. Nguyen [18] compared the economic, environmental, and social impacts of astaxanthin production by chemical synthesis, fermentation, or isolation from algal material and reported huge differences among the costs of these processes: $2000, $2500, and >$7000 per kg of astaxanthin, respectively. Although the production by chemical synthesis is less expensive (about $1000 per kilo), and it is indicated as the most cost-effective way to obtain this astaxanthin [19], this process does not give a pure compound but a combination of different isoforms that have 20 times lower antioxidant capacity than their natural counterpart. Besides, to date, it has not been approved for human consumption [11,18]. The chemically synthesized astaxanthin is expected to retain its dominance on the market, especially in the feed industry; however, consumers’ focus on naturally extracted astaxanthin products is expected to rise [17]. There is limited information on the production price of natural astaxanthin from crustacean byproducts. It is dependent on the availability of the raw material, extraction methodology, and astaxanthin yield and purity [17]. At the industrial level, astaxanthin is extracted from krill and crustacean byproducts in China [19]. Su et al. [20] calculated that China alone generates 500,000 metric tons of shrimp byproducts and wastes 97 metric tons of natural astaxanthin ($650 million) every year. Evidently, the use of crustacean byproducts for carotenoid extraction has room for improvement and presents a great opportunity for the whole sector.
This paper reviews recent studies on shrimp/crab byproducts as valuable sources of natural astaxanthin (studies are from 2015 to present, found by the keywords search in the electronic databases Scopus and ScienceDirect). The objective is to provide an overview of the scientific knowledge on the chemical and biochemical properties of astaxanthin as well as an overview of its content in crustacean byproducts. To find the limitation of the successful exploitation of astaxanthin, we reviewed papers dealing with extraction protocols in search of the environmentally friendly process or green chemistry techniques for the recovery of this metabolite. The article characterizes the bioactive properties and health benefits of astaxanthin. Special attention has been devoted to data on astaxanthin sourced from crustaceans to identify potential improvements needed for its successful industrial exploitation.

2. The Structure and Division of the Carotenoids from Marine Origin

Carotenoids are a class of lipid-soluble pigments widely distributed in different photosynthetic organisms and some nonphotosynthetic bacteria and fungi, responsible for their red, orange, and yellow colors. These color features are the result of their chemical structure, primarily a long polyenic carbon chain while derivatization of their base structure, among other properties, results in different color hues and tones. They are primarily biosynthesized by plants, algae, yeasts, fungi, archaea, and eubacteria, but they can also be found in animals and humans, who absorb and deposit them through the diet and different metabolic reactions [21,22,23,24,25].
According to the latest available data, approximately 1204 naturally occurring carotenoids have been detected in 722 source organisms [26].

2.1. Chemical Structure and Classes

More than 95% of all known carotenoids are formed by isoprene units (C5 blocks), and based on the number of carbons in their structure, they are classified as C30 (6 isoprenoid units), C40 (8 isoprenoid units), C45 (9 isoprenoid units), and C50 (10 isoprenoid units). The class of C40 is the most abundant in nature as it is synthesized by eukaryotic organisms, bacteria, and archaea. On the other hand, carotenoids with the C30 and C50 structures are synthesized by bacteria and archaea only while those with the C45 structure are synthesized only by some bacteria. The presence of some apocarotenoids, whose normal C40 backbones have been shortened from one and/or both ends, has also been reported. Some of the well-known apocarotenoids are bixin (C25) and crocetin (C20). In some cases, vitamin A is also considered an apocarotenoid since symmetric cleavage of β-carotene gives two equivalent retinal molecules [22,27].
According to the carotenoid-structural features, they are classified into two major classes:
(a)
carotenes—linear hydrocarbons that can be cyclized at the ends (e.g., β-carotene and lycopene), and
(b)
xanthophylls—derivatives of carotenes with one or more oxygen-containing functional groups (oxygenated carotenoids) (e.g., zeaxanthin, lutein, capsanthin, violaxanthin, neoxanthin) [11,22,28,29].
The functional groups of xanthophylls, such as epoxy groups in neoxanthin, violaxanthin, and fucoxanthin, hydroxyl groups in zeaxanthin and lutein, the keto group in astaxanthin, and the methoxy functional group present in spirilloxanthin, are generated by the enzymatic reactions. These functional groups affect the compound solubility (higher polarity) as well as their other chemical and biological functions [22,28,30] (Figure 1).
In nature, carotenoids are found in free forms or esterified with fatty acids (compounds with hydroxyl groups, especially xanthophylls), sugars (crocetin), or proteins (carotenoproteins), which result in increasing their lipophilicity [22,24].
In their polyene chain, some of them contain allenic or acetylenic functional groups, like fucoxanthin and peridinin (allenic carotenoids) or crocoxanthin (acetylene carotenoid) [22]. Most of the naturally present carotenoids are in a more stable trans form despite the presence of multiple double bonds in their structure [22,23].
Their structural diversity is responsible for numerous biochemical and physiological functions associated with this class of compounds.

2.2. Carotenoids from Marine Organisms

Carotenoids are essential pigments, structural and functional components in numerous aquatic organisms (primary carotenoids), or generated after exposure to various conditions (secondary carotenoids) [31,32]. The primary sources of carotenoids are photo-autotrophic organisms; they can be found in their primary consumers (herbivores, e.g., sea urchins, bivalves, crustaceans, and fish) or in secondary consumers (e.g., cephalopods, crustaceans, fish) [30] where they are absorbed directly or modified through the metabolic reactions.
The most important carotenoids from marine organisms are astaxanthin (found in green algae, bacteria, yeast, sea snails, sea urchin, crabs, shrimps, lobsters, shellfishes, starfish, jellyfish, etc.); fucoxanthin (in seaweeds, diatoms, corals, sea urchin, starfish, etc.); β-carotene (in microalgae, seaweeds, shellfish, cyanobacteria, sea urchin, starfish, etc.); lutein (in microalgae, seaweeds, corals, shellfishes, etc.); siphonaxanthin (in green algae); mytiloxanthin (in tunicates, mussels, oysters, etc.); zeaxanthin (in microalgae, seaweeds, corals, shellfish, etc.); saproxanthin and myxol (in bacteria from the family Flavobacteriaceae); halocynthiaxanthin (in sea squirt and sea pineapple); violaxanthin, neoxanthin, and antheraxanthin (in various seaweeds); isorenieratene, renieratene, and renierapurpurin (in sponges), etc. [27,31,32,33].

3. Carotenoids from Crustaceans

The composition of crustacean shells varies with species and seasons; however, it is mainly composed of chitin (15–40%), protein (20–40%), calcium carbonate (20–50%), and lipids (0–14%) with a high content of omega-3 fatty acids, pigments, and other minor components [34,35,36]. These compounds have shown great bioactivity and versatility in applications. Proteins can be used as fertilizer and animal feed and calcium carbonate as a fertilizing material, filler, or white pigment [37] while chitin utilization ranges from the production of biomaterials, food, pharmaceuticals, and cosmetics to water treatments [38,39,40].
In crustaceans, body coloration depends on the specific pigments present in the principal layer of their exoskeleton and the subepidermal chromatophores. Among them, the carotenoids are the most significant, and the most prevalent carotenoid in commercially important crustaceans is astaxanthin [19,20].

3.1. Astaxanthin from Crustaceans

3.1.1. Structure and Biochemistry

Astaxanthin (C40H52O4, 3,3-dihydroxy-β,β-carotene-4,4-dione), an orange-reddish-colored pigment, is widely distributed among the seafood byproducts, so it is often called marine carotenoid [41,42]. Structurally, it is a ketocarotenoid from the group of xanthophylls [7,43], with hydroxyl (OH) and keto (CO) groups at each terminal. This unique chemical structure is responsible for its characteristics and functions (Figure 1) [7,44]. Astaxanthin has two chiral carbons at the 3 and 3′ positions, which allow the formation of various isomers and stereoisomers [20,45,46]. The three main isomers of astaxanthin are two enantiomers (3S, 3′S), (3R, 3′R) and one mesomer (3R, 3′S), depending on the spatial orientation of hydroxyl groups in the chiral carbons [47]. The stereoisomers (3S, 3′S) and (3R, 3′R) are the most abundant in nature [48]. Astaxanthin also occurs in two geometrical isomers, trans- and cis- (E and Z), depending on the configuration of the double bonds in the polyene chain [49].
Astaxanthin is derived from the β-carotene or zeaxanthin by β-carotene hydroxylase and β-carotene ketolase, respectively [50]. In crustaceans, it can be found unesterified (free) or esterified (mono- and diesterified) with various fatty acids [51]. Its structure has a significant impact on its bioavailability, as suggested by Yang et al. [46], who investigated the stability and bioavailability of different astaxanthin derivates (free, mono-, and diesters) by in vitro and in vivo digestion models and concluded that esters with long-chain and saturated fatty acids are the most stable and that esters containing short-chain fatty acids (esters with high unsaturation of fatty acids and monomers in comparison to the dimers) have higher bioavailability. Several studies have also reported their interactions forming lipoproteins and carotenoproteins [41,52]. Besides, a recent study by Yang et al. [43] suggested that encapsulation of astaxanthin (in the starch-based emulsion) imparted its stability and increased accessibility. The bioaccessibility of astaxanthin was found to increase from 50 to 100% by encapsulation in gelatin gel [53], imparting its stability for inclusion in food supplements or matrices. After being ingested, astaxanthin is diffused through the intestine and forms micelles, which are partially absorbed and further stored in the liver. From there, astaxanthin is transported to the tissues via a circulatory mechanism [10].

3.1.2. Sources

Crustaceans can synthesize astaxanthin through the metabolic reactions from other carotenoids, such as β-carotene, lutein, and zeaxanthin, that are ingested (Figure 2), or they can just store it directly by consuming other animals that already performed the bioconversion [31]. Crustaceans accumulate astaxanthin in their tissues, cuticles, hemolymphs, and eggs, where it exists in both forms, free and/or esterified as mono- or diesters with palmitic, oleic, stearic, or linoleic acid. The free astaxanthin circulates in the hemolymph and may be incorporated in the cell membranes where it prevents lipid peroxidation and participates in maintaining membrane structure (Figure 3). On the other hand, esters are mainly stored in the tissue. The structure of astaxanthin can fit the hydrophobic polyene carbon chain inside the bilayer cell membrane while its polar terminal rings are located near its surface [19].
Amongst crustaceans, astaxanthin is widely distributed in shrimps, crawfish, crabs, lobsters, and Antarctic krill byproducts [55,56] (Table 1). In the exoskeleton of crustaceans, astaxanthin exists as carotenoproteins, such as crustacyanin, providing various colors from red, purple, blue/blue-black, to yellow [30,57]. In the complex form (i.e., associated with proteins and lipids e.g., carotenoprotein or carotenolipoprotein), it is blue to green, which, upon denaturation/separation/cleavage and the release of astaxanthin from the complex, imparts a reddish-orange color [56].
Although most of the commercialized astaxanthin originates from the green microalgae H. pluvialis [59], crustaceans are considered its good source, especially if it is recovered from the seafood processing industry byproducts [41,60,61,62]. These byproducts, such as the head, shell, and tail of shrimps, are rich in astaxanthin and are among the most explored sources of natural astaxanthin [4]. Besides, wastewater from shrimp cooking/processing should also be considered as its possible source [56]. A recent literature overview (data reported in the period from 2016 to 2021) of astaxanthin content from different crustaceans and crustacean byproducts is given in Table 1.
The accumulation of astaxanthin in crustaceans has been suggested to play an important role as an immune-stimulating and antioxidant agent. The crustaceans possess endogenous enzymes and nonenzymatic, exogenous compounds that can scavenge free radicals. The generation of reactive oxygen species (ROS) could be induced by high irradiation (mainly ultraviolet), oxygenic photoautotrophy, and the presence of xenobiotics. The major antioxidant enzymes able to detoxify ROS are superoxide dismutase, catalase, and peroxidase. Furthermore, crustaceans possess peroxinectins, proteins involved in the immune defenses, including peroxidase activity. Nonenzymatic, exogenous compounds with antioxidant properties are obtained through dietary intake. Nonenzymatic antioxidants can be divided into hydrophilic compounds, present in aqueous cellular compartments such as vitamin C, glutathione, and lipophilic compounds, such as carotenoids, which can be included in cell membranes and associated with lipoproteins. Hence, carotenoids can regulate immunopathology and play an important role in the modulation and the evolution of immune defenses [57,79].

3.1.3. Extraction

Traditionally, astaxanthin has been extracted from crustacean byproducts by solvent extraction, oil extraction, or microbial fermentation. Due to the harsh processing conditions that characterize these conventional extraction techniques, the yield, quality, and stability of astaxanthin are meager. Considering these limitations, novel, nonthermal techniques, such as supercritical fluid extraction, high-pressure extraction, ultrasonication, and pulsed electric field, have been evaluated (Table 1) [65,77,80,81].
The high-pressure extraction of astaxanthin from shrimp byproducts was reported by Li et al. [74]. Solvation properties of used solvents (ethanol, acetone, dichloromethane) and pressure application (0 to 600 MPa) exhibited a high impact on astaxanthin extraction from shrimp processing byproducts. The application of high-pressure damages cellular membranes and disorders the fiber structure, leading to the higher diffusion of solvent and enhanced astaxanthin extraction. However, pressure increases over 300 MPa showed a negative impact on astaxanthin recovery.
Ultrasound application (23.6% amplitude, 26.3 °C for 13.9 min) improved the extraction of astaxanthin from shrimp shells [80]. Ultrasound-induced cavitation caused fragmentation of the shell matrix, increasing the solubilization of bioactive compounds and their extraction by solvents. Solvent polarity and extraction time had a significant impact on the extracted yield of astaxanthin.
Supercritical fluid extraction employing different solvents is found to be an effective technique for astaxanthin extraction from crustacean byproducts. Optimized conditions (56.88 °C, 215.68 bar, and flow rate of 1.89 mL/min) yielded both free (12.20 µg/g) and conjugated (58.50 µg/g) astaxanthin [77]. This technique is dependent upon the solubility of solute, which is influenced by the temperature and pressure. Besides, solvent selection has a key role in astaxanthin extraction [82,83]. Higher concentrations of ethanol (5, 10, and 15%) resulted in a significant increase of astaxanthin yield (from 26.0 to 34.8 µg/g) [84]. Besides, the application of high pressures (>400 bar) in supercritical fluid extraction hampers its extraction.
Microbial fermentation followed by supercritical extraction from shrimp waste liquid fraction was recently optimized [65]. Results suggested that fermentation of raw material by lactic acid bacteria improved extraction of astaxanthin in comparison to the common supercritical extraction. The fermentation increased the extraction of lipophilic compounds in the liquor and enzymolysis of shrimp shells while neutral proteases resulted in a 3.7-fold higher astaxanthin concentration (134.20 µg/g) [85].
The combined effects of ultrasound- and pulsed electric field-assisted treatment on the extraction of astaxanthin from shrimp byproducts were reported by Gulzar and Benjakul [52]. Disintegration by damaging the cephalothorax was found to increase with an elevation of the electric field strength. Furthermore, ultrasound caused electroporation, improving the mass transfer; thus, the recovery of astaxanthin was recorded as well. The pulsed electric field was found to be a more suitable technique than ultrasonic extraction, in terms of damage caused to the cells.

3.2. Biological Activity of Astaxanthin

Highly reactive oxidative molecules, like free radicals or ROS, through the reactions with various cell-regulating molecules, cause damages that induce different disorders in the human body. These reactions can be effectively inhibited by the application of endogenous and exogenous antioxidants. Among carotenoids, astaxanthin has been recognized as one of the most effective antioxidants due to its unique chemical structure and both lipophilic and hydrophilic properties [7,10,48]. The proposed mechanisms of astaxanthin antioxidant action are electron donation, bonding with free radicals to form less active forms and nonreactive products, transportation of radicals along its own carbon chain outside the cell, inhibition of ROS formation, and metal chelation by two adjacent oxygen atoms on the cyclohexene ring. All of these antioxidant activities have been reported by numerous in vitro and in vivo studies [6,8,23,45,51,86,87,88,89].
In the intense search for novel antibacterial agents that would contribute to the global health issue of bacterial antibiotic resistance that is affecting treatments of the various infections, astaxanthin has been shown as a promising candidate. Generally, the nature and mode of action of antimicrobials is unclear and/or partially explained, but it is known that one of the common antimicrobial mechanisms is based on the ability of antibacterial agents to generate diverse forms of ROS while interacting with cell targets [90,91]. The generated ROS species (e.g., peroxide radical or superoxide anion) are not as reactive as those generated through the Fenton reaction (e.g., hydroxide radical) showing the ability to cause cellular dysfunction and/or cell death [92]. This concept of antibacterial activity through ROS generation has been implicated for carotenoids, among others, and another possible antimicrobial action of astaxanthin is the inhibition of biofilm formation (antibiofilm activity), both in bacteria and fungi [93]. The antimicrobial activity of astaxanthin bacteria has been also widely reported [61,92,93,94,95,96,97,98]. Furthermore, beneficial pharmacological effects of astaxanthin, such as antioxidant, antimicrobial, anticancer, anti-inflammatory, neurodegenerative, gastrointestinal, cardiovascular, antidiabetic, ocular, and skin-protective effects, have been widely reported [6,48]. Generally, scientists are reporting that astaxanthin’s physical and chemical interactions with cell components and/or cell membranes are key features for its high effectiveness in these disorders [48,92], and some results of the novel studies (from 2015 to present) regarding these activities are reported in Table 2.

3.3. Health Benefits, Therapeutic Application, and Safety of Astaxanthin Application in Humans

Considering the health benefits imparted by astaxanthin due to its biological activity as discussed earlier, it is widely distributed among the food web [56]. The wide arrays of bioactivities possessed by astaxanthin from shrimp byproducts have been found to exhibit health benefits both in vitro and in vivo [126,127]. Astaxanthin reduced the release of interleukins, cycloxygenase-2 and nitric oxide generation, and DNA damage induced by radioactivity as well as improved inflammatory-related pathways [110,128,129,130,131,132,133,134]. It has been found to impart a healthy state by reducing oxidative stress, neutralizing singlet oxygen, scavenging free radicals, inhibiting lipid peroxidation, and improving immunity and muscle health [5,7,89,135,136,137]. Several studies have also indicated the ability of astaxanthin from shrimp to reduce obesity, hypertension, hyperlipidemia, and heart-related ailments [138,139,140,141]. These properties have been attributed to its anti-inflammatory properties and reduction of oxidative stress in glucose and lipid metabolism [58,142,143]. The management of blood pressure by astaxanthin was explained by its ability to reduce oxidative stress and vasoconstriction [7,141,144,145,146]. Astaxanthin from natural sources increased endurance, improved resistance to stress, and prevented inflammations and ulcers [145,147].
Astaxanthin gained popularity due to its numerous health benefits and has been proven safe for oral administration [148,149]. Considering its low bioavailability/absorption rate in its free form, it is administered in the ester- or nanoform [100,145,150]. Further, the dosage related to meal timing (before and after meal) and the form (monoester or diester) affects its concentration and availability [151,152]. Astaxanthin imparts protection against a cytokine storm controlling adversaries and pathogenesis caused by COVID-19 [153]. The ability of astaxanthin to enhance immunity by increasing the production of immunoglobulin A responsible for antibody production has also been reported [154]. In addition, an improvement in human skin health measured by skin elasticity (gross, net, and biological) due to supplementation of dietary astaxanthin with collagen hydrolysate was observed after 12 weeks of application [155].
Cardiovascular disorders are responsible for a large number of deaths worldwide [5,156], and since they are often connected with oxidative stress and ROS, astaxanthin can be used in their prevention due to its strong antioxidative activity [126,157,158,159]. Furthermore, the oral administration of astaxanthin reduced cholesterol in mice with immediate spread across the body [160]. It has also been found that astaxanthin reduces stroke incidence and myocardial infarct size [5]. Lowered/delayed oxidation of low-density lipoprotein, reduced oxidative stress, lipid metabolism, anti-inflammatory properties, and improved blood rheology are evidence of its beneficial properties against cardiovascular diseases [10,126,159,161,162,163,164,165,166]. Astaxanthin administered orally at 50 mg/kg body weight improved kidney function in mouse models [167]. Recently, neuroprotection has gained immense importance due to the increase in neurodegenerative diseases. The incidence is linked with high stress causing damage, misfolding, and aggregation leading to the death of neurons [168]. A recent review by Fakhri et al. [169] reported the neuroprotective effects imparted by astaxanthin. The neuroprotective effects of astaxanthin are related to its ability of multitarget treatment and its capability to influence signaling pathways (PI3K/Akt pathways), neuroinflammation, lipid peroxidation, and microcirculation; inhibit amyloid-β peptide, aggregation, and mitochondrial functions; and decrease cell death helpful for treating Parkinson’s and Alzheimer’s diseases as well as neuropathic pain and suppression [169,170,171,172]. The improved cognitive ability of elderly patients evaluated by improved reaction time and low rate of error in the memory-based game was detected for patients that were taking astaxanthin supplements [173]. Furthermore, improved motor skills were also observed after 12 weeks [174].
Astaxanthin from natural sources has been suggested as safe for application with no adverse (mutagenic, carcinogenic, biochemical, and hematological) effects for humans [7,142,147,158]. However, there is a lack of literature on marine astaxanthin therapeutic applications and safety concerns since it is known that supplementation of different amounts and forms (natural and synthetic) exhibits different levels of bioactivities [159,175,176,177]. Considering the safety issues, the allowed levels of astaxanthin in food supplements were 8 mg/day, and acceptable daily intake for adults ranged from 0.034 to 0.2 mg astaxanthin/kg body weight [178]. On the contrary, the highest dosage evaluated (i.e., 700–1000 mg/kg body weight) reported subchronic toxicity in rats [176,179,180]. Furthermore, the application of astaxanthin in the astaxanthin-dimethyldisuccinate form at 100 mg astaxanthin/kg completed diet (138 mg astaxanthin/kg) was recommended for effective coloring in fish and crustaceans without dermal or ocular risk. Considering this, the maximum exposure of the consumer would be 25 mg astaxanthin/kg and 4 mg astaxanthin/kg [181].
There are different applications of natural and synthetic astaxanthin. The main limitation in the industrial application of natural astaxanthin is its price; however, the nutraceutical and pharmaceutic industries use more expensive forms of astaxanthin due to the increase concerns of the consumers for their health and environment [182]. Besides a high price, limited sources of natural astaxanthin stand in the way of widening its application. So far, astaxanthin has been applied as a coloring agent in aquaculture feed [13], food industry, and various cosmetics [19]. It has been widely used as a food supplement and functional ingredient but also as an enhancing agent for food quality and nutritive value [183]. Despite the widely documented, positive effects of astaxanthin, its use in practice is still very limited.

4. Conclusions

In recent years, studies showed a strong nutraceutical and therapeutic potential of natural astaxanthin. Its powerful antioxidant and other bioactive properties (anti-inflammatory, cytotoxic, antiproliferative, and anticancer activity) as well as favorable safety profile make astaxanthin a promising compound with the ability to prevent or even treat different health conditions. Among them, there is reported evidence of astaxanthin’s ability to delay cardiovascular diseases, adverse neuroprotective effects, and harmful inflammations in the organism while improving lipid and glucose metabolism. Further, astaxanthin has benefits in animal health and aquaculture production. The source of natural astaxanthin has been limited to microalgae (fungi and krill in smaller proportions). Many studies evidence the availability/yield of the astaxanthin from different parts of crustacean byproducts. The main challenge is the unknown cost-effectiveness of the production process as well as the lack of information on the amounts currently obtained from byproducts. There is also a lack of comprehensive studies on the economic aspects of astaxanthin production from crustacean byproduct at the industrial level. Besides, for the successful scale-up of the extraction methodologies and exploitation at the industrial level, there are additional challenges that need to be addressed. Information on the availability of the raw material from processing plants is needed as well as the seasonal effect on astaxanthin content. In addition, it is necessary to distinguish an environmentally friendly process or green extraction method among the promising techniques, such as microwave-assisted, supercritical extraction using CO2 and co-solvents or enzymatic extraction, and optimize/standardize it for testing in the industrial environment. During the extraction processes, the quality of astaxanthin extract should be considered as well as the feasibility of the process. Besides, additional work is needed to determine the structural and biological activity-related mechanisms of natural astaxanthin. Further, adequate doses and encapsulation of crustacean byproduct astaxanthin for supplement and food applications should be established considering its bioavailability as well as research to explain the biological mechanisms associated with its therapeutic potential.

Author Contributions

Conceptualization, V.Š.; writing—original draft preparation, V.Š., N.B.R., M.Č., I.H., and I.G.M.; writing—review and editing, V.Š. and I.G.M.; supervision, V.Š. All authors have read and agreed to the published version of the manuscript.

Funding

This research is supported by the PRIMA program under project BioProMedFood (Project ID 1467). The PRIMA program is supported by the European Union.

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.

References

  1. FAO. The State of World Fisheries and Aquaculture 2020; Sustainability in Action: Rome, Italy, 2020. [Google Scholar] [CrossRef]
  2. Özogul, F.; Hamed, I.; Özogul, Y.; Regenstein, J.M. Crustacean By-Products. In Encyclopedia of Food Chemistry; Elsevier Inc.: Amsterdam, The Netherlands, 2019; pp. 33–38. [Google Scholar] [CrossRef]
  3. Šimat, V. Valorization of Seafood Processing By-Products. In Valorization of Agri-Food Wastes and By-Products: Recent Trends, Innovations and Sustainability Challenges; Bhat, R., Ed.; Elsevier Inc.: Amsterdam, The Netherlands, 2021; pp. 515–536. [Google Scholar]
  4. Nirmal, N.P.; Santivarangkna, C.; Rajput, M.S.; Benjakul, S. Trends in Shrimp Processing Waste Utilization: An Industrial Prospective. Trends Food Sci. Technol. 2020, 103, 20–35. [Google Scholar] [CrossRef]
  5. Visioli, F.; Artaria, C. Astaxanthin in Cardiovascular Health and Disease: Mechanisms of Action, Therapeutic Merits, and Knowledge Gaps. Food Funct. 2017, 8, 39–63. [Google Scholar] [CrossRef] [PubMed]
  6. Davinelli, S.; Nielsen, M.E.; Scapagnini, G. Astaxanthin in Skin Health, Repair, and Disease: A Comprehensive Review. Nutrients 2018, 10, 522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Fakhri, S.; Abbaszadeh, F.; Dargahi, L.; Jorjani, M. Astaxanthin: A Mechanistic Review on Its Biological Activities and Health Benefits. Pharmacol. Res. 2018, 136, 1–20. [Google Scholar] [CrossRef] [PubMed]
  8. Pereira, C.P.M.; Souza, A.C.R.; Vasconcelos, A.R.; Prado, P.S.; Name, J.J. Antioxidant and Anti-Inflammatory Mechanisms of Action of Astaxanthin in Cardiovascular Diseases (Review). Int. J. Mol. Med. 2021, 47, 37–48. [Google Scholar] [CrossRef] [PubMed]
  9. Raza, S.H.A.; Naqvi, S.R.Z.; Abdelnour, S.A.; Schreurs, N.; Mohammedsaleh, Z.M.; Khan, I.; Shater, A.F.; Abd El-Hack, M.E.; Khafaga, A.F.; Quan, G.; et al. Beneficial Effects and Health Benefits of Astaxanthin Molecules on Animal Production: A Review. Res. Vet. Sci. 2021, 138, 69–78. [Google Scholar] [CrossRef] [PubMed]
  10. Donoso, A.; González-Durán, J.; Muñoz, A.A.; González, P.A.; Agurto-Muñoz, C. Therapeutic Uses of Natural Astaxanthin: An Evidence-Based Review Focused on Human Clinical Trials. Pharmacol. Res. 2021, 166, 105479. [Google Scholar] [CrossRef] [PubMed]
  11. Kumar, S.; Kumar, R.; Kumari, A.; Panwar, A.; Diksha. Astaxanthin: A Super Antioxidant from Microalgae and Its Therapeutic Potential. J. Basic Microbiol. 2021, 1–19. [Google Scholar] [CrossRef] [PubMed]
  12. Mohd Hatta, F.A.; Othman, R. Carotenoids as Potential Biocolorants: A Case Study of Astaxanthin Recovered from Shrimp Waste. In Carotenoids: Properties, Processing and Applications; Elsevier: Amsterdam, The Netherlands, 2020; pp. 289–325. [Google Scholar] [CrossRef]
  13. Lim, K.C.; Yusoff, F.M.; Shariff, M.; Kamarudin, M.S. Astaxanthin as Feed Supplement in Aquatic Animals. Rev. Aquac. 2018, 10, 738–773. [Google Scholar] [CrossRef]
  14. Mezquita, P.C.; Huerta, B.E.B.; Ramírez, J.C.P.; Hinojosa, C.P.O. Milks Pigmentation with Astaxanthin and Determination of Colour Stability during Short Period Cold Storage. J. Food Sci. Technol. 2015, 52, 1634–1641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Grand View Research, Inc. Astaxanthin Market Size, Industry Report, 2021–2028. Available online: https://www.grandviewresearch.com/industry-analysis/global-astaxanthin-market (accessed on 13 February 2022).
  16. Villaró, S.; Ciardi, M.; Morillas-España, A.; Sánchez-Zurano, A.; Acién-Fernández, G.; Lafarga, T. Microalgae Derived Astaxanthin: Research and Consumer Trends and Industrial Use as Food. Foods 2021, 10, 2303. [Google Scholar] [CrossRef] [PubMed]
  17. Athanasios, B.; Zoumboulakis, P. Valuable Products from Algae Using New Magnetic Cultivation and Extraction Techniques; VALUEMAG project (Contract No. H2020-745695). Report on market penetration. 2020. [Google Scholar]
  18. Nguyen, K.D. Astaxanthin: A Comparative Case of Synthetic vs. Natural Production. Chem. Biomol. Eng. Publ. Other Work 2013, 1–9. [Google Scholar]
  19. Stachowiak, B.; Szulc, P. Astaxanthin for the Food Industry. Molecules 2021, 26, 2666. [Google Scholar] [CrossRef] [PubMed]
  20. Su, F.; Huang, B.; Liu, J. The Carotenoids of Shrimps (Decapoda: Caridea and Dendrobranchiata) Cultured in China. J. Crustac. Biol. 2018, 38, 523–530. [Google Scholar] [CrossRef] [Green Version]
  21. Mezzomo, N.; Ferreira, S.R.S. Carotenoids Functionality, Sources, and Processing by Supercritical Technology: A Review. J. Chem. 2016, 2016, 3164312. [Google Scholar] [CrossRef] [Green Version]
  22. Fernandes, A.S.; do Nascimento, T.C.; Jacob-Lopes, E.; De Rosso, V.V.; Zepka, L.Q. Introductory Chapter: Carotenoids—A Brief Overview on Its Structure, Biosynthesis, Synthesis, and Applications. Prog. Carotenoid Res. 2018, 1, 1–16. [Google Scholar] [CrossRef] [Green Version]
  23. Nagarajan, J.; Ramanan, R.N.; Raghunandan, M.E.; Galanakis, C.M.; Krishnamurthy, N.P. Carotenoids; Elsevier Inc.: Amsterdam, The Netherlands, 2017. [Google Scholar] [CrossRef]
  24. Schieber, A.; Weber, F. Carotenoids. In Handbook on Natural Pigments in Food and Beverages: Industrial Applications for Improving Food Color; Woodhead Publishing: Cambridge, UK, 2016; pp. 101–123. [Google Scholar] [CrossRef]
  25. Rivera-Madrid, R.; Carballo-Uicab, V.M.; Cárdenas-Conejo, Y.; Aguilar-Espinosa, M.; Siva, R. Overview of Carotenoids and Beneficial Effects on Human Health. In Carotenoids: Properties, Processing and Applications; Elsevier: Amsterdam, The Netherlands, 2020; pp. 1–40. [Google Scholar] [CrossRef]
  26. Yabuzaki, J. Carotenoids Database. Available online: http://carotenoiddb.jp/ (accessed on 13 February 2022).
  27. Viera, I.; Pérez-Gálvez, A.; Roca, M. Bioaccessibility of Marine Carotenoids. Mar. Drugs 2018, 16, 397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Ngamwonglumlert, L.; Devahastin, S.; Food, A. Carotenoids. In Encyclopedia of Food Chemistry, 1st ed.; Academic Press: Cambridge, MA, USA, 2018; pp. 40–52. [Google Scholar] [CrossRef]
  29. Kiokias, S.; Proestos, C.; Varzakas, T. A Review of the Structure, Biosynthesis, Absorption of Carotenoids-Analysis and Properties of Their Common Natural Extracts. Curr. Res. Nutr. Food Sci. J. 2016, 4, 25–37. [Google Scholar] [CrossRef]
  30. de Carvalho, C.C.C.R.; Caramujo, M.J. Carotenoids in Aquatic Ecosystems and Aquaculture: A Colorful Business with Implications for Human Health. Front. Mar. Sci. 2017, 4, 93. [Google Scholar] [CrossRef] [Green Version]
  31. Maoka, T. Carotenoids in Marine Animals. Mar. Drugs 2011, 9, 278–293. [Google Scholar] [CrossRef] [PubMed]
  32. Galasso, C.; Corinaldesi, C.; Sansone, C. Carotenoids from Marine Organisms: Biological Functions and Industrial Applications. Antioxidants 2017, 6, 96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Miyashita, K.; Hosokawa, M. Health Impact of Marine Carotenoids. J. Food Bioact. 2018, 1, 31–40. [Google Scholar] [CrossRef] [Green Version]
  34. de Queiroz Antonino, R.; Lia Fook, B.; de Oliveira Lima, V.; de Farias Rached, R.; Lima, E.; da Silva Lima, R.; Peniche Covas, C.; Lia Fook, M. Preparation and Characterization of Chitosan Obtained from Shells of Shrimp (Litopenaeus Vannamei Boone). Mar. Drugs 2017, 15, 141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Tan, Y.N.; Lee, P.P.; Chen, W.N. Microbial Extraction of Chitin from Seafood Waste Using Sugars Derived from Fruit Waste-Stream. AMB Express 2020, 10, 17. [Google Scholar] [CrossRef] [PubMed]
  36. Hülsey, M.J. Shell Biorefinery: A Comprehensive Introduction. Green Energy Environ. 2018, 3, 318–327. [Google Scholar] [CrossRef]
  37. Nekvapil, F.; Ganea, I.V.; Ciorîță, A.; Hirian, R.; Ogresta, L.; Glamuzina, B.; Roba, C.; Pinzaru, S.C. Wasted Biomaterials from Crustaceans as a Compliant Natural Product Regarding Microbiological, Antibacterial Properties and Heavy Metal Content for Reuse in Blue Bioeconomy: A Preliminary Study. Materials 2021, 14, 4558. [Google Scholar] [CrossRef] [PubMed]
  38. Arbia, W.; Arbia, L.; Adour, L.; Amrane, A. Chitin Extraction from Crustacean Shells Using Biological Methods—A Review. Food Technol. Biotechnol. 2013, 51, 12–25. [Google Scholar]
  39. Casadidio, C.; Peregrina, D.V.; Gigliobianco, M.R.; Deng, S.; Censi, R.; Di Martino, P. Chitin and Chitosans: Characteristics, Eco-Friendly Processes, and Applications in Cosmetic Science. Mar. Drugs 2019, 17, 369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Morin-Crini, N.; Lichtfouse, E.; Torri, G.; Crini, G. Applications of Chitosan in Food, Pharmaceuticals, Medicine, Cosmetics, Agriculture, Textiles, Pulp and Paper, Biotechnology, and Environmental Chemistry. Environ. Chem. Lett. 2019, 17, 1667–1692. [Google Scholar] [CrossRef] [Green Version]
  41. Ahmadkelayeh, S.; Hawboldt, K. Extraction of Lipids and Astaxanthin from Crustacean By-Products: A Review on Supercritical CO2 Extraction. Trends Food Sci. Technol. 2020, 103, 94–108. [Google Scholar] [CrossRef]
  42. Hamdi, M.; Nasri, R.; Dridi, N.; Li, S.; Nasri, M. Development of Novel High-Selective Extraction Approach of Carotenoproteins from Blue Crab (Portunus Segnis) Shells, Contribution to the Qualitative Analysis of Bioactive Compounds by HR-ESI-MS. Food Chem. 2020, 302, 125334. [Google Scholar] [CrossRef] [PubMed]
  43. Yang, J.; Hua, S.; Huang, Z.; Gu, Z.; Cheng, L.; Hong, Y. Comparison of Bioaccessibility of Astaxanthin Encapsulated in Starch-Based Double Emulsion with Different Structures. Carbohydr. Polym. 2021, 272, 118475. [Google Scholar] [CrossRef] [PubMed]
  44. Seabra, L.M.J.; Pedrosa, L.F.C. Astaxanthin: Structural and Functional Aspects. Rev. Nutr. 2010, 23, 1041–1050. [Google Scholar] [CrossRef] [Green Version]
  45. Brotosudarmo, T.H.P.; Limantara, L.; Setiyono, E.; Heriyanto. Structures of Astaxanthin and Their Consequences for Therapeutic Application. Int. J. Food Sci. 2020, 2020, 2156582. [Google Scholar] [CrossRef] [PubMed]
  46. Yang, L.; Qiao, X.; Gu, J.; Li, X.; Cao, Y.; Xu, J.; Xue, C. Influence of Molecular Structure of Astaxanthin Esters on Their Stability and Bioavailability. Food Chem. 2021, 343, 128497. [Google Scholar] [CrossRef] [PubMed]
  47. Yue, Y.; Kim, B.; Lee, J.-Y. Astaxanthin Structure, Metabolism, and Health Benefits. J. Hum. Nutr. Food Sci. 2013, 1, 1003. [Google Scholar]
  48. Ambati, R.R.; Moi, P.S.; Ravi, S.; Aswathanarayana, R.G. Astaxanthin: Sources, Extraction, Stability, Biological Activities and Its Commercial Applications—A Review. Mar. Drugs 2014, 12, 128–152. [Google Scholar] [CrossRef] [PubMed]
  49. Qiu, D.; Wu, Y.-C.; Zhu, W.-L.; Yin, H.; Yi, L.-T. Identification of Geometrical Isomers and Comparison of Different Isomeric Samples of Astaxanthin. J. Food Sci. 2012, 77, C934–C940. [Google Scholar] [CrossRef] [PubMed]
  50. Saini, R.K.; Nile, S.H.; Park, S.W. Carotenoids from Fruits and Vegetables: Chemistry, Analysis, Occurrence, Bioavailability and Biological Activities. Food Res. Int. 2015, 76, 735–750. [Google Scholar] [CrossRef] [PubMed]
  51. Higuera-Ciapara, I.; Félix-Valenzuela, L.; Goycoolea, F.M. Astaxanthin: A Review of Its Chemistry and Applications. Crit. Rev. Food Sci. Nutr. 2006, 46, 185–196. [Google Scholar] [CrossRef] [PubMed]
  52. Gulzar, S.; Benjakul, S. Impact of Pulsed Electric Field Pretreatment on Yield and Quality of Lipid Extracted from Cephalothorax of Pacific White Shrimp (Litopenaeus Vannamei) by Ultrasound-assisted Process. Int. J. Food Sci. Technol. 2020, 55, 619–630. [Google Scholar] [CrossRef] [Green Version]
  53. Gómez-Guillén, M.C.; Montero, P.; López-Caballero, M.E.; Baccan, G.C.; Gómez-Estaca, J. Bioactive and Technological Functionality of a Lipid Extract from Shrimp (L. Vannamei) Cephalothorax. LWT 2018, 89, 704–711. [Google Scholar] [CrossRef] [Green Version]
  54. Rhodes, A.C.E. Dietary Effects on Carotenoid Composition in the Marine Harpacticoid Copepod Nitokra Lacustris. J. Plankton Res. 2007, 29, i73–i83. [Google Scholar] [CrossRef]
  55. Montoya, J.M.; Velazco Mata, S.; Acosta, J.L.; Herrera Cabrera, B.E.; López Valdez, L.G.; Reyes, C.; Barrales Cureño, J.H. Obtaining of Astaxanthin from Crab Exosqueletons and Shrimp Head Shells. Biointerface Res. Appl. Chem. 2021, 11, 13516–13523. [Google Scholar] [CrossRef]
  56. Routray, W.; Dave, D.; Cheema, S.K.; Ramakrishnan, V.V.; Pohling, J. Biorefinery Approach and Environment-Friendly Extraction for Sustainable Production of Astaxanthin from Marine Wastes. Crit. Rev. Biotechnol. 2019, 39, 469–488. [Google Scholar] [CrossRef] [PubMed]
  57. Babin, A.; Motreuil, S.; Teixeira, M.; Bauer, A.; Rigaud, T.; Moreau, J.; Moret, Y. Origin of the Natural Variation in the Storage of Dietary Carotenoids in Freshwater Amphipod Crustaceans. PLoS ONE 2020, 15, e0231247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Kishimoto, Y.; Yoshida, H.; Kondo, K. Potential Anti-Atherosclerotic Properties of Astaxanthin. Mar. Drugs 2016, 14, 35. [Google Scholar] [CrossRef] [PubMed]
  59. Shah, M.M.R.; Liang, Y.; Cheng, J.J.; Daroch, M. Astaxanthin-Producing Green Microalga Haematococcus Pluvialis: From Single Cell to High Value Commercial Products. Front. Plant Sci. 2016, 7, 531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Nunes, A.N.; Roda, A.; Gouveia, L.F.; Fernández, N.; Bronze, M.R.; Matias, A.A. Astaxanthin Extraction from Marine Crustacean Waste Streams: An Integrate Approach between Microwaves and Supercritical Fluids. ACS Sustain. Chem. Eng. 2021, 9, 3050–3059. [Google Scholar] [CrossRef]
  61. Dalei, J.; Sahoo, D. Extraction and Characterisation of Astaxanthin from the Crustacean Shell Waste from Shrimp Processing Industries. Int. J. Pharm. Sci. Res. 2015, 6, 2532–2537. [Google Scholar] [CrossRef]
  62. Hu, J.; Lu, W.; Lv, M.; Wang, Y.; Ding, R.; Wang, L. Extraction and Purification of Astaxanthin from Shrimp Shells and the Effects of Different Treatments on Its Content. Rev. Bras. Farmacogn. 2019, 29, 24–29. [Google Scholar] [CrossRef]
  63. Liu, Z.; Liu, Q.; Zhang, D.; Wei, S.; Sun, Q.; Xia, Q.; Shi, W.; Ji, H.; Liu, S. Comparison of the Proximate Composition and Nutritional Profile of Byproducts and Edible Parts of Five Species of Shrimp. Foods 2021, 10, 2603. [Google Scholar] [CrossRef] [PubMed]
  64. Messina, C.M.; Manuguerra, S.; Arena, R.; Renda, G.; Ficano, G.; Randazzo, M.; Fricano, S.; Sadok, S.; Santulli, A. In Vitro Bioactivity of Astaxanthin and Peptides from Hydrolisates of Shrimp (Parapenaeus Longirostris) By-Products: From the Extraction Process to Biological Effect Evaluation, as Pilot Actions for the Strategy “From Waste to Profit”. Mar. Drugs 2021, 19, 216. [Google Scholar] [CrossRef] [PubMed]
  65. Cabanillas-Bojórquez, L.A.; Gutiérrez-Grijalva, E.P.; González-Aguilar, G.A.; López-Martinez, L.X.; Castillo-López, R.I.; Bastidas-Bastidas, P.D.J.; Heredia, J.B. Valorization of Fermented Shrimp Waste with Supercritical CO2 Conditions: Extraction of Astaxanthin and Effect of Simulated Gastrointestinal Digestion on Its Antioxidant Capacity. Molecules 2021, 26, 4465. [Google Scholar] [CrossRef] [PubMed]
  66. de Silva, M.P.K.S.K.; Senaarachchi, W.A.R.K. Efficiency of Biotransformation of Shellfish Waste to Carotenoprotein by Autolysis and Crab-Shrimp Endo-Enzymes. J. Aquat. Food Prod. Technol. 2021, 30, 526–534. [Google Scholar] [CrossRef]
  67. Dave, D.; Liu, Y.; Pohling, J.; Trenholm, S.; Murphy, W. Astaxanthin Recovery from Atlantic Shrimp (Pandalus Borealis) Processing Materials. Bioresour. Technol. Rep. 2020, 11, 100535. [Google Scholar] [CrossRef]
  68. Cheong, J.Y.; Muskhazli, M.; Nor Azwady, A.A.; Ahmad, S.A.; Adli, A.A. Three Dimensional Optimisation for the Enhancement of Astaxanthin Recovery from Shrimp Shell Wastes by Aeromonas Hydrophila. Biocatal. Agric. Biotechnol. 2020, 27, 101649. [Google Scholar] [CrossRef]
  69. Nunes, A.N.; Rodaa, A.; Matias, A.A. Recovery of Astaxanthin Pigments from Marine Crustacean Waste Streams Using Supercritical Fluid Technology. In Book of Abstracts The European Summer School in High Pressure Technology; Verlag der Technischen Universität: Graz, Austria, 2019; pp. 24–28. [Google Scholar]
  70. da Silva, A.K.N.; Rodrigues, B.D.; da Silva, L.H.M.; da Rodrigues, A.M.C. Drying and Extraction of Astaxanthin from Pink Shrimp Waste (Farfantepenaeus Subtilis): The Applicability of Spouted Beds. Food Sci. Technol. 2018, 38, 454–461. [Google Scholar] [CrossRef] [Green Version]
  71. Chintong, S.; Phatvej, W.; Rerk-Am, U.; Waiprib, Y.; Klaypradit, W. In Vitro Antioxidant, Antityrosinase, and Cytotoxic Activities of Astaxanthin from Shrimp Waste. Antioxidants 2019, 8, 128. [Google Scholar] [CrossRef] [Green Version]
  72. Irna, C.; Jaswir, I.; Othman, R.; Jimat, D.N. Comparison between High-Pressure Processing and Chemical Extraction: Astaxanthin Yield from Six Species of Shrimp Carapace. J. Diet. Suppl. 2018, 15, 805–813. [Google Scholar] [CrossRef]
  73. Darachai, P.; Limpawattana, M.; Hawangjoo, M.; Klaypradit, W. Effects of Shrimp Waste Types and Their Cooking on Properties of Extracted Astaxanthin and Its Characteristics in Liposomes. J. Food Nutr. Res. 2019, 7, 530–536. [Google Scholar] [CrossRef]
  74. Li, J.; Sun, W.; Ramaswamy, H.S.; Yu, Y.; Zhu, S.; Wang, J.; Li, H. High Pressure Extraction of Astaxanthin from Shrimp Waste (Penaeus Vannamei Boone): Effect on Yield and Antioxidant Activity. J. Food Process Eng. 2017, 40, e12353. [Google Scholar] [CrossRef]
  75. Gómez-Estaca, J.; Calvo, M.M.; Álvarez-Acero, I.; Montero, P.; Gómez Guillén, M.C. Characterization and Storage Stability of Astaxanthin Esters, Fatty Acid Profile and a -Tocopherol of Lipid Extract from Shrimp (L. Vannamei) Waste with Potential Applications as Food Ingredient. Food Chem. 2017, 216, 37–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Antunes-Valcareggi, S.A.; Ferreira, S.R.S.; Hense, H. Enzymatic Hydrolysis of Blue Crab (Callinectes Sapidus) Waste Processing to Obtain Chitin, Protein, and Astaxanthin-Enriched Extract. Int. J. Environ. Agric. Res. 2017, 3, 81–92. [Google Scholar]
  77. Shazana, A.R.; Masturah, M.; Badlishah, S.B.; Rashidi, O.; Russly, A.R. Optimisation of Supercritical Fluid Extraction of Astaxanthin From Penaeus Monodon Waste Using Ethanol-Modified Carbon Dioxide. J. Eng. Sci. Technol. 2016, 11, 722–736. [Google Scholar]
  78. Soultani, G.; Strati, I.F. Assessment of Functional Lipid Constituents of Red (Aristaeomorpha Foliacea) and Pink (Parapenaeus Longirostris) Shrimps. J. Aquac. Res. Dev. 2016, 7, 452. [Google Scholar] [CrossRef] [Green Version]
  79. Caramujo, M.-J.; de Carvalho, C.C.C.R.; Silva, S.J.; Carman, K.R. Dietary Carotenoids Regulate Astaxanthin Content of Copepods and Modulate Their Susceptibility to UV Light and Copper Toxicity. Mar. Drugs 2012, 10, 998–1018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Sharayei, P.; Azarpazhooh, E.; Zomorodi, S.; Einafshar, S.; Ramaswamy, H.S. Optimization of Ultrasonic-Assisted Extraction of Astaxanthin from Green Tiger (Penaeus semisulcatus) Shrimp Shell. Ultrason. Sonochem. 2021, 76, 105666. [Google Scholar] [CrossRef]
  81. Gulzar, S.; Benjakul, S. Ultrasound Waves Increase the Yield and Carotenoid Content of Lipid Extracted from Cephalothorax of Pacific White Shrimp (Litopenaeus vannamei). Eur. J. Lipid Sci. Technol. 2018, 120, 1700495. [Google Scholar] [CrossRef]
  82. Sánchez-Camargo, A.P.; Almeida Meireles, M.Â.; Lopes, B.L.F.; Cabral, F.A. Proximate Composition and Extraction of Carotenoids and Lipids from Brazilian Redspotted Shrimp Waste (Farfantepenaeus paulensis). J. Food Eng. 2011, 102, 87–93. [Google Scholar] [CrossRef]
  83. Sánchez-Camargo, A.P.; Martinez-Correa, H.A.; Paviani, L.C.; Cabral, F.A. Supercritical CO2 Extraction of Lipids and Astaxanthin from Brazilian Redspotted Shrimp Waste (Farfantepenaeus paulensis). J. Supercrit. Fluids 2011, 56, 164–173. [Google Scholar] [CrossRef] [Green Version]
  84. Sánchez-Camargo, A.P.; Meireles, M.Â.A.; Ferreira, A.L.K.; Saito, E.; Cabral, F.A. Extraction of ω-3 Fatty Acids and Astaxanthin from Brazilian Redspotted Shrimp Waste Using Supercritical CO2+ethanol Mixtures. J. Supercrit. Fluids 2012, 61, 71–77. [Google Scholar] [CrossRef]
  85. Wang, W.; Liu, M.; Fawzy, S.; Xue, Y.; Wu, M.; Huang, X.; Yi, G.; Lin, Q. Effects of Dietary Phaffia Rhodozyma Astaxanthin on Growth Performance, Carotenoid Analysis, Biochemical and Immune-Physiological Parameters, Intestinal Microbiota, and Disease Resistance in Penaeus Monodon. Front. Microbiol. 2021, 12, 762689. [Google Scholar] [CrossRef] [PubMed]
  86. Kidd, P. Astaxanthin, Cell Membrane Nutrient with Diverse Clinical Benefits and Anti-Aging Potential. Altern. Med. Rev. 2011, 16, 355–364. [Google Scholar] [PubMed]
  87. Naguib, Y.M.A. Antioxidant Activities of Astaxanthin and Related Carotenoids. J. Agric. Food Chem. 2000, 48, 1150–1154. [Google Scholar] [CrossRef] [PubMed]
  88. Martin, H.D.; Ruck, C.; Schmidt, M.; Sell, S.; Beutner, S.; Mayer, B.; Walsh, R. Chemistry of Carotenoid Oxidation and Free Radical Reactions. Pure Appl. Chem. 1999, 71, 2253–2262. [Google Scholar] [CrossRef]
  89. Nishida, Y.; Yamashita, E.; Miki, W. Quenching Activities of Common Hydrophilic and Lipophilic Antioxidants against Singlet Oxygen Using Chemiluminescence Detection System. Carotenoid Sci. 2007, 11, 16–20. [Google Scholar]
  90. Memar, M.Y.; Ghotaslou, R.; Samiei, M.; Adibkia, K. Antimicrobial Use of Reactive Oxygen Therapy: Current Insights. Infect. Drug Resist. 2018, 11, 567–576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Li, H.; Zhou, X.; Huang, Y.; Liao, B.; Cheng, L.; Ren, B. Reactive Oxygen Species in Pathogen Clearance: The Killing Mechanisms, the Adaption Response, and the Side Effects. Front. Microbiol. 2021, 11, 622534. [Google Scholar] [CrossRef] [PubMed]
  92. Aribisala, J.O.; Nkosi, S.; Idowu, K.; Nurain, I.O.; Makolomakwa, G.M.; Shode, F.O.; Sabiu, S. Astaxanthin-Mediated Bacterial Lethality: Evidence from Oxidative Stress Contribution and Molecular Dynamics Simulation. Oxid. Med. Cell. Longev. 2021, 2021, 7159652. [Google Scholar] [CrossRef] [PubMed]
  93. Karpiński, T.M.; Ożarowski, M.; Alam, R.; Łochyńska, M.; Stasiewicz, M. What Do We Know about Antimicrobial Activity of Astaxanthin and Fucoxanthin? Mar. Drugs 2022, 20, 36. [Google Scholar] [CrossRef] [PubMed]
  94. Suganya, V.; Asheeba, S. Antioxidant and Antimicrobial Activity of Astaxanthin Isolated from Three Varieties of Crabs. Int. J. Recent Sci. Res. 2015, 6, 6753–6758. [Google Scholar]
  95. Ushakumari, U.N.; Ramanujan, R. Isolation of Astaxanthin from Marine Yeast and Study of Its Pharmacological Activity. Int. Curr. Pharm. J. 2013, 2, 67–69. [Google Scholar] [CrossRef] [Green Version]
  96. Mageswari, A.; Subramanian, P.; Srinivasan, R.; Karthikeyan, S.; Gothandam, K.M. Astaxanthin from Psychrotrophic Sphingomonas Faeni Exhibits Antagonism against Food-Spoilage Bacteria at Low Temperatures. Microbiol. Res. 2015, 179, 38–44. [Google Scholar] [CrossRef] [PubMed]
  97. Irna, C.; Jaswir, I.; Othman, R.; Jimat, D.N. Antioxidant and Antimicrobial Activities of Astaxanthin from Penaeus Monodon in Comparison between Chemical Extraction and High Pressure Processing (HPP). Int. Food Res. J. 2017, 24, 508–513. [Google Scholar]
  98. Rather, A.H.; Singh, S.; Choudhary, S. Antibacterial Activity of Haematococcus Pluvialis Crude Astaxanthin Extract. J. Drug Deliv. Ther. 2021, 11, 28–30. [Google Scholar] [CrossRef]
  99. Guan, L.; Liu, J.; Yu, H.; Tian, H.; Wu, G.; Liu, B.; Dong, P.; Li, J.; Liang, X. Water-Dispersible Astaxanthin-Rich Nanopowder: Preparation, Oral Safety and Antioxidant Activity in Vivo. Food Funct. 2019, 10, 1386–1397. [Google Scholar] [CrossRef] [PubMed]
  100. Hu, Q.; Hu, S.; Fleming, E.; Lee, J.Y.; Luo, Y. Chitosan-Caseinate-Dextran Ternary Complex Nanoparticles for Potential Oral Delivery of Astaxanthin with Significantly Improved Bioactivity. Int. J. Biol. Macromol. 2020, 151, 747–756. [Google Scholar] [CrossRef] [PubMed]
  101. Feng, Z.Z.; Li, M.Y.; Wang, Y.T.; Zhu, M.J. Astaxanthin from Phaffia Rhodozyma: Microencapsulation with Carboxymethyl Cellulose Sodium and Microcrystalline Cellulose and Effects of Microencapsulated Astaxanthin on Yogurt Properties. LWT 2018, 96, 152–160. [Google Scholar] [CrossRef]
  102. Tirado, D.F.; Palazzo, I.; Scognamiglio, M.; Calvo, L.; Della Porta, G.; Reverchon, E. Astaxanthin Encapsulation in Ethyl Cellulose Carriers by Continuous Supercritical Emulsions Extraction: A Study on Particle Size, Encapsulation Efficiency, Release Profile and Antioxidant Activity. J. Supercrit. Fluids 2019, 150, 128–136. [Google Scholar] [CrossRef]
  103. Montanari, E.; Di Meo, C.; Coviello, T.; Gueguen, V.; Pavon-Djavid, G.; Matricardi, P. Intracellular Delivery of Natural Antioxidants via Hyaluronan Nanohydrogels. Pharmaceutics 2019, 11, 532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Weintraub, S.; Shpigel, T.; Harris, L.G.; Schuster, R.; Lewis, E.C.; Lewitus, D.Y. Astaxanthin-Based Polymers as New Antimicrobial Compounds. Polym. Chem. 2017, 8, 4182–4189. [Google Scholar] [CrossRef]
  105. Karuppusamy, S.; Kim, H.; Saravana, P.S.; Chun, B.S.; Kang, H.W. Astaxanthin-Alpha Tocopherol Nanoemulsion Formulation by Emulsification Methods: Investigation on Anticancer, Wound Healing, and Antibacterial Effects. Colloids Surf. B Biointerfaces 2018, 172, 170–179. [Google Scholar] [CrossRef]
  106. Sukmawati; Fawwaz, M.; Pratama, M.; Hasrawati, A. Potential of Astaxanthin from Asian Tiger Shrimp (Penaeus Monodon) Shell Extract as an Antibacterial and Anti-Inflammatory. J. Glob. Pharma Technol. 2019, 11, 2017–2022. [Google Scholar]
  107. Kohandel, Z.; Farkhondeh, T.; Aschner, M.; Pourbagher-Shahri, A.M.; Samarghandian, S. Anti-Inflammatory Action of Astaxanthin and Its Use in the Treatment of Various Diseases. Biomed. Pharmacother. 2022, 145, 112179. [Google Scholar] [CrossRef] [PubMed]
  108. Peng, Y.J.; Lu, J.W.; Liu, F.C.; Lee, C.H.; Lee, H.S.; Ho, Y.J.; Hsieh, T.H.; Wu, C.C.; Wang, C.C. Astaxanthin Attenuates Joint Inflammation Induced by Monosodium Urate Crystals. FASEB J. 2020, 34, 11215–11226. [Google Scholar] [CrossRef]
  109. Hwang, Y.H.; Hong, S.G.; Mun, S.K.; Kim, S.J.; Lee, S.J.; Kim, J.J.; Kang, K.Y.; Yee, S.T. The Protective Effects of Astaxanthin on the OVA-Induced Asthma Mice Model. Molecules 2017, 22, 2019. [Google Scholar] [CrossRef] [Green Version]
  110. Bharathiraja, S.; Manivasagan, P.; Bui, N.Q.; Oh, Y.O.; Lim, I.G.; Park, S.; Oh, J. Cytotoxic Induction and Photoacoustic Imaging of Breast Cancer Cells Using Astaxanthin-Reduced Gold Nanoparticles. Nanomaterials 2016, 6, 78. [Google Scholar] [CrossRef] [Green Version]
  111. Zhang, X.; Yin, W.; Qi, Y.; Li, X.; Zhang, W.; He, G. Microencapsulation of Astaxanthin in Alginate Using Modified Emulsion Technology: Preparation, Characterization, and Cytostatic Activity. Can. J. Chem. Eng. 2017, 95, 412–419. [Google Scholar] [CrossRef]
  112. Haung, H.Y.; Wang, Y.C.; Cheng, Y.C.; Kang, W.; Hu, S.H.; Liu, D.; Xiao, C.; Wang, H.M.D.; Ali, D. A Novel Oral Astaxanthin Nanoemulsion from Haematococcus Pluvialis Induces Apoptosis in Lung Metastatic Melanoma. Oxid. Med. Cell. Longev. 2020, 2020, 2647670. [Google Scholar] [CrossRef]
  113. Ni, X.; Yu, H.; Wang, S.; Zhang, C.; Shen, S. Astaxanthin Inhibits PC-3 Xenograft Prostate Tumor Growth in Nude Mice. Mar. Drugs 2017, 15, 66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Cui, L.; Xu, F.; Wang, M.; Li, L.; Qiao, T.; Cui, H.; Li, Z.; Sun, C. Dietary Natural Astaxanthin at an Early Stage Inhibits N-Nitrosomethylbenzylamine-Induced Esophageal Cancer Oxidative Stress and Inflammation via Downregulation of NFκB and COX2 in F344 Rats. Onco. Targets. Ther. 2019, 12, 5087–5096. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Ohno, T.; Shimizu, M.; Shirakami, Y.; Miyazaki, T.; Ideta, T.; Kochi, T.; Kubota, M.; Sakai, H.; Tanaka, T.; Moriwaki, H. Preventive Effects of Astaxanthin on Diethylnitrosamine-Induced Liver Tumorigenesis in C57/BL/KsJ-Db/Db Obese Mice. Hepatol. Res. 2016, 46, E201–E209. [Google Scholar] [CrossRef] [PubMed]
  116. Yuan, L.; Liang, P.; Qu, Y.; An, T.; Wang, J.; Deng, X.; Bai, L.; Shen, P.; Bai, D. Protective Effect of Astaxanthin against SnS2 Nanoflowers Induced Testes Toxicity by Suppressing RIPK1-RIPK3-MLKL Signaling in Mice. Food Chem. Toxicol. 2020, 145, 111736. [Google Scholar] [CrossRef] [PubMed]
  117. Wu, Y.C.; Huang, H.H.; Wu, Y.J.; Manousakas, I.; Yang, C.C.; Kuo, S.M. Therapeutic and Protective Effects of Liposomal Encapsulation of Astaxanthin in Mice with Alcoholic Liver Fibrosis. Int. J. Mol. Sci. 2019, 20, 4057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Chiu, C.H.; Chang, C.C.; Lin, S.T.; Chyau, C.C.; Peng, R.Y. Improved Hepatoprotective Effect of Liposome-Encapsulated Astaxanthin in Lipopolysaccharide-Induced Acute Hepatotoxicity. Int. J. Mol. Sci. 2016, 17, 1128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Zhang, J.; Zhang, S.; Bi, J.; Gu, J.; Deng, Y.; Liu, C. Astaxanthin Pretreatment Attenuates Acetaminophen-Induced Liver Injury in Mice. Int. Immunopharmacol. 2017, 45, 26–33. [Google Scholar] [CrossRef] [PubMed]
  120. Özbeyli, D.; Gürler, E.B.; Buzcu, H.; Çilingir-Kaya, Ö.T.; Çam, M.E.; Yüksel, M. Astaxanthin Alleviates Oxidative Damage in Acute Pancreatitis via Direct Antioxidant Mechanisms. Turkish J. Gastroenterol. 2020, 31, 706–712. [Google Scholar] [CrossRef]
  121. Zhuge, F.; Ni, Y.; Wan, C.; Liu, F.; Fu, Z. Anti-Diabetic Effects of Astaxanthin on an Stz-Induced Diabetic Model in Rats. Endocr. J. 2021, 68, 451–459. [Google Scholar] [CrossRef] [PubMed]
  122. Akduman, H.; Tayman, C.; Çakir, U.; Çakir, E.; Dilli, D.; Türkmenoğlu, T.T.; Gönel, A. Astaxanthin Prevents Lung Injury Due to Hyperoxia and Inflammation. Comb. Chem. High Throughput Screen. 2021, 24, 1243–1250. [Google Scholar] [CrossRef]
  123. Li, H.; Li, J.; Hou, C.; Li, J.; Peng, H.; Wang, Q. The Effect of Astaxanthin on Inflammation in Hyperosmolarity of Experimental Dry Eye Model in Vitro and in vivo. Exp. Eye Res. 2020, 197, 108113. [Google Scholar] [CrossRef] [PubMed]
  124. Tominaga, K.; Hongo, N.; Fujishita, M.; Takahashi, Y.; Adachi, Y. Protective Effect of Astaxanthin on Skin Deterioration. J. Clin. Biochem. Nutr. 2017, 61, 33–39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Lee, Y.S.; Jeon, S.H.; Ham, H.J.; Lee, H.P.; Song, M.J.; Hong, J.T. Improved Anti-Inflammatory Effects of Liposomal Astaxanthin on a Phthalic Anhydride-Induced Atopic Dermatitis Model. Front. Immunol. 2020, 11, 1–9. [Google Scholar] [CrossRef] [PubMed]
  126. Yaqoob, Z.; Arshad, M.S.; Imran, M.; Munir, H.; Qaisrani, T.B.; Khalid, W.; Asghar, Z.; Suleria, H.A.R. Mechanistic Role of Astaxanthin Derived from Shrimp against Certain Metabolic Disorders. Food Sci. Nutr. 2022, 10, 12–20. [Google Scholar] [CrossRef] [PubMed]
  127. Faraone, I.; Sinisgalli, C.; Ostuni, A.; Armentano, M.F.; Carmosino, M.; Milella, L.; Russo, D.; Labanca, F.; Khan, H. Astaxanthin Anticancer Effects Are Mediated through Multiple Molecular Mechanisms: A Systematic Review. Pharmacol. Res. 2020, 155, 104689. [Google Scholar] [CrossRef] [PubMed]
  128. Grimmig, B.; Kim, S.-H.; Nash, K.; Bickford, P.C.; Douglas Shytle, R. Neuroprotective Mechanisms of Astaxanthin: A Potential Therapeutic Role in Preserving Cognitive Function in Age and Neurodegeneration. GeroScience 2017, 39, 19–32. [Google Scholar] [CrossRef] [PubMed]
  129. Kim, S.; Lim, J.; Kim, H. Astaxanthin Inhibits Mitochondrial Dysfunction and Interleukin-8 Expression in Helicobacter Pylori-Infected Gastric Epithelial Cells. Nutrients 2018, 10, 1320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Zhang, M.; Cui, Z.; Cui, H.; Wang, Y.; Zhong, C. Astaxanthin Protects Astrocytes against Trauma-Induced Apoptosis through Inhibition of NKCC1 Expression via the NF-ΚB Signaling Pathway. BMC Neurosci. 2017, 18, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Liu, X.; Song, M.; Gao, Z.; Cai, X.; Dixon, W.; Chen, X.; Cao, Y.; Xiao, H. Stereoisomers of Astaxanthin Inhibit Human Colon Cancer Cell Growth by Inducing G2/M Cell Cycle Arrest and Apoptosis. J. Agric. Food Chem. 2016, 64, 7750–7759. [Google Scholar] [CrossRef] [PubMed]
  132. Wu, H.; Niu, H.; Shao, A.; Wu, C.; Dixon, B.; Zhang, J.; Yang, S.; Wang, Y. Astaxanthin as a Potential Neuroprotective Agent for Neurological Diseases. Mar. Drugs 2015, 13, 5750–5766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Choi, S.-K.; Park, Y.-S.; Choi, D.-K.; Chang, H.-I. Effects of Astaxanthin on the Production of NO and the Expression of COX-2 and INOS in LPS-Stimulated BV2 Microglial Cells. J. Microbiol. Biotechnol. 2008, 18, 1990–1996. [Google Scholar] [PubMed]
  134. Santocono, M.; Zurria, M.; Berrettini, M.; Fedeli, D.; Falcioni, G. Influence of Astaxanthin, Zeaxanthin and Lutein on DNA Damage and Repair in UVA-Irradiated Cells. J. Photochem. Photobiol. B Biol. 2006, 85, 205–215. [Google Scholar] [CrossRef] [PubMed]
  135. Wong, S.K.; Ima-Nirwana, S.; Chin, K. Effects of Astaxanthin on the Protection of Muscle Health (Review). Exp. Ther. Med. 2020, 20, 2941–2952. [Google Scholar] [CrossRef] [PubMed]
  136. Kamath, B.S.; Srikanta, B.M.; Dharmesh, S.M.; Sarada, R.; Ravishankar, G.A. Ulcer Preventive and Antioxidative Properties of Astaxanthin from Haematococcus Pluvialis. Eur. J. Pharmacol. 2008, 590, 387–395. [Google Scholar] [CrossRef] [PubMed]
  137. Iwasaki, T.; Tawara, A. Effects of Astaxanthin on Eyestrain Induced by Accommodative Dysfunction. J. Eye 2006, 23, 829. [Google Scholar]
  138. Manivasagan, P.; Bharathiraja, S.; Santha Moorthy, M.; Mondal, S.; Seo, H.; Dae Lee, K.; Oh, J. Marine Natural Pigments as Potential Sources for Therapeutic Applications. Crit. Rev. Biotechnol. 2018, 38, 745–761. [Google Scholar] [CrossRef] [PubMed]
  139. Kim, J.H.; Chang, M.J.; Choi, H.D.; Youn, Y.-K.; Kim, J.T.; Oh, J.M.; Shin, W.G. Protective Effects of Haematococcus Astaxanthin on Oxidative Stress in Healthy Smokers. J. Med. Food 2011, 14, 1469–1475. [Google Scholar] [CrossRef] [PubMed]
  140. Bhuvaneswari, S.; Arunkumar, E.; Viswanathan, P.; Anuradha, C.V. Astaxanthin Restricts Weight Gain, Promotes Insulin Sensitivity and Curtails Fatty Liver Disease in Mice Fed a Obesity-Promoting Diet. Process Biochem. 2010, 45, 1406–1414. [Google Scholar] [CrossRef]
  141. Hussein, G.; Goto, H.; Oda, S.; Iguchi, T.; Sankawa, U.; Matsumoto, K.; Watanabe, H. Antihypertensive Potential and Mechanism of Action of Astaxanthin: II. Vascular Reactivity and Hemorheology in Spontaneously Hypertensive Rats. Biol. Pharm. Bull. 2005, 28, 967–971. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Satoh, A.; Tsuji, S.; Okada, Y.; Murakami, N.; Urami, M.; Nakagawa, K.; Ishikura, M.; Katagiri, M.; Koga, Y.; Shirasawa, T. Preliminary Clinical Evaluation of Toxicity and Efficacy of A New Astaxanthin-Rich Haematococcus Pluvialis Extract. J. Clin. Biochem. Nutr. 2009, 44, 280–284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Palozza, P.; Barone, E.; Mancuso, C.; Picci, N. The Protective Role of Carotenoids against 7-Keto-Cholesterol Formation in Solution. Mol. Cell. Biochem. 2008, 309, 61–68. [Google Scholar] [CrossRef] [PubMed]
  144. Zhang, J.; Wang, Q.; Zhao, S.; Ji, X.; Qiu, J.; Wang, J.; Zhou, Y.; Cai, Q.; Zhang, J.; Gao, H. Astaxanthin Attenuated Pressure Overload-Induced Cardiac Dysfunction and Myocardial Fibrosis: Partially by Activating SIRT1. Biochim. Biophys. Acta Gen. Subj. 2017, 1861, 1715–1728. [Google Scholar] [CrossRef] [PubMed]
  145. Satoh, T. Astaxanthin. In Nutraceuticals; Elsevier: Amsterdam, The Netherlands, 2016; pp. 531–539. [Google Scholar] [CrossRef]
  146. Preuss, H.G.; Echard, B.; Bagchi, D.; Perricone, N.V.; Yamashita, E. Astaxanthin Lowers Blood Pressure and Lessens the Activity of the Renin-Angiotensin System in Zucker Fatty Rats. J. Funct. Foods 2009, 1, 13–22. [Google Scholar] [CrossRef]
  147. Capelli, B.; Talbott, S.; Ding, L. Astaxanthin Sources: Suitability for Human Health and Nutrition. Funct. Foods Heal. Dis. 2019, 9, 430. [Google Scholar] [CrossRef] [Green Version]
  148. Li, J.; Guo, C.; Wu, J. Astaxanthin in Liver Health and Disease: A Potential Therapeutic Agent. Drug Des. Devel. Ther. 2020, 14, 2275–2285. [Google Scholar] [CrossRef] [PubMed]
  149. Mercke Odeberg, J.; Lignell, Å.; Pettersson, A.; Höglund, P. Oral Bioavailability of the Antioxidant Astaxanthin in Humans Is Enhanced by Incorporation of Lipid Based Formulations. Eur. J. Pharm. Sci. 2003, 19, 299–304. [Google Scholar] [CrossRef]
  150. Zhou, Q.; Xu, J.; Yang, L.; Gu, C.; Xue, C. Thermal Stability and Oral Absorbability of Astaxanthin Esters from Haematococcus Pluvialis in Balb/c Mice. J. Sci. Food Agric. 2019, 99, 3662–3671. [Google Scholar] [CrossRef] [PubMed]
  151. Ranga Rao, A.; Raghunath Reddy, R.L.; Baskaran, V.; Sarada, R.; Ravishankar, G.A. Characterization of Microalgal Carotenoids by Mass Spectrometry and Their Bioavailability and Antioxidant Properties Elucidated in Rat Model. J. Agric. Food Chem. 2010, 58, 8553–8559. [Google Scholar] [CrossRef] [PubMed]
  152. Fukami, H.; Namikawa, K.; Sugiura-Tomimori, N.; Sumida, M.; Katano, K.; Nakao, M. Chemical Synthesis of Astaxanthin N-Octanoic Acid Monoester and Diester and Evaluation of Their Oral Absorbability. J. Oleo Sci. 2006, 55, 653–656. [Google Scholar] [CrossRef] [Green Version]
  153. Talukdar, J.; Dasgupta, S.; Nagle, V.; Bhadra, B. COVID-19: Potential of Microalgae Derived Natural Astaxanthin As Adjunctive Supplement in Alleviating Cytokine Storm. SSRN Electron. J. 2020. [Google Scholar] [CrossRef]
  154. Niu, T.; Zhou, J.; Wang, F.; Xuan, R.; Chen, J.; Wu, W.; Chen, H. Safety Assessment of Astaxanthin from Haematococcus Pluvialis: Acute Toxicity, Genotoxicity, Distribution and Repeat-Dose Toxicity Studies in Gestation Mice. Regul. Toxicol. Pharmacol. 2020, 115, 104695. [Google Scholar] [CrossRef]
  155. Yoon, H.-S.; Cho, H.H.; Cho, S.; Lee, S.-R.; Shin, M.-H.; Chung, J.H. Supplementing with Dietary Astaxanthin Combined with Collagen Hydrolysate Improves Facial Elasticity and Decreases Matrix Metalloproteinase-1 and -12 Expression: A Comparative Study with Placebo. J. Med. Food 2014, 17, 810–816. [Google Scholar] [CrossRef]
  156. Giordano, F.J. Oxygen, Oxidative Stress, Hypoxia, and Heart Failure. J. Clin. Investig. 2005, 115, 500–508. [Google Scholar] [CrossRef] [PubMed]
  157. Zhang, Z.-W.; Xu, X.-C.; Liu, T.; Yuan, S. Mitochondrion-Permeable Antioxidants to Treat ROS-Burst-Mediated Acute Diseases. Oxid. Med. Cell. Longev. 2016, 2016, 6859523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Yamashita, E. Let Astaxanthin Be Thy Medicine. PharmaNutrition 2015, 3, 115–122. [Google Scholar] [CrossRef]
  159. Choi, H.D.; Youn, Y.K.; Shin, W.G. Positive Effects of Astaxanthin on Lipid Profiles and Oxidative Stress in Overweight Subjects. Plant Foods Hum. Nutr. 2011, 66, 363–369. [Google Scholar] [CrossRef] [PubMed]
  160. Ryu, S.K.; King, T.J.; Fujioka, K.; Pattison, J.; Pashkow, F.J.; Tsimikas, S. Effect of an Oral Astaxanthin Prodrug (CDX-085) on Lipoprotein Levels and Progression of Atherosclerosis in LDLR−/− and ApoE−/− Mice. Atherosclerosis 2012, 222, 99–105. [Google Scholar] [CrossRef]
  161. Kato, T.; Kasai, T.; Sato, A.; Ishiwata, S.; Yatsu, S.; Matsumoto, H.; Shitara, J.; Murata, A.; Shimizu, M.; Suda, S.; et al. Effects of 3-Month Astaxanthin Supplementation on Cardiac Function in Heart Failure Patients with Left Ventricular Systolic Dysfunction-A Pilot Study. Nutrients 2020, 12, 1896. [Google Scholar] [CrossRef]
  162. Maria, A.G.; Graziano, R.; Nicolantonio, D. Carotenoids: Potential Allies of Cardiovascular Health? Food Nutr. Res. 2015, 59, 26762. [Google Scholar] [CrossRef] [Green Version]
  163. Park, J.S.; Chyun, J.H.; Kim, Y.K.; Line, L.L.; Chew, B.P. Astaxanthin Decreased Oxidative Stress and Inflammation and Enhanced Immune Response in Humans. Nutr. Metab. 2010, 7, 18. [Google Scholar] [CrossRef] [Green Version]
  164. Yoshida, H.; Yanai, H.; Ito, K.; Tomono, Y.; Koikeda, T.; Tsukahara, H.; Tada, N. Administration of Natural Astaxanthin Increases Serum HDL-Cholesterol and Adiponectin in Subjects with Mild Hyperlipidemia. Atherosclerosis 2010, 209, 520–523. [Google Scholar] [CrossRef] [PubMed]
  165. Miyawaki, H.; Takahashi, J.; Tsukahara, H.; Takehara, I. Effects of Astaxanthin on Human Blood Rheology. J. Clin. Biochem. Nutr. 2008, 43, 69–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Iwamoto, T.; Hosoda, K.; Hirano, R.; Kurata, H.; Matsumoto, A.; Miki, W.; Kamiyama, M.; Itakura, H.; Yamamoto, S.; Kondo, K. Inhibition of Low-Density Lipoprotein Oxidation by Astaxanthin. J. Atheroscler. Thromb. 2000, 7, 216–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Liu, G.; Shi, Y.; Peng, X.; Liu, H.; Peng, Y.; He, L. Astaxanthin Attenuates Adriamycin-Induced Focal Segmental Glomerulosclerosis. Pharmacology 2015, 95, 193–200. [Google Scholar] [CrossRef] [PubMed]
  168. Mohan, V.; Edamakanti, C.R.; Sharma, V. Role of Neuroinflammation in Neurodegenerative Disorders. In The Molecular Immunology of Neurological Diseases; Elsevier: Amsterdam, The Netherlands, 2021; pp. 41–49. [Google Scholar] [CrossRef]
  169. Fakhri, S.; Aneva Yosifova, I.; Farzaei, M.H.; Sobarzo-Sánchez, E. The Neuroprotective Effects of Astaxanthin: Therapeutic Targets and Clinical Perspective. Molecules 2019, 24, 2640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Zarneshan, S.N.; Fakhri, S.; Farzaei, M.H.; Khan, H.; Saso, L. Astaxanthin Targets PI3K/Akt Signaling Pathway toward Potential Therapeutic Applications. Food Chem. Toxicol. 2020, 145, 111714. [Google Scholar] [CrossRef] [PubMed]
  171. Rahman, S.O.; Panda, B.P.; Parvez, S.; Kaundal, M.; Hussain, S.; Akhtar, M.; Najmi, A.K. Neuroprotective Role of Astaxanthin in Hippocampal Insulin Resistance Induced by Aβ Peptides in Animal Model of Alzheimer’s Disease. Biomed. Pharmacother. 2019, 110, 47–58. [Google Scholar] [CrossRef] [PubMed]
  172. Xu, L.; Zhu, J.; Yin, W.; Ding, X. Astaxanthin Improves Cognitive Deficits from Oxidative Stress, Nitric Oxide Synthase and Inflammation through Upregulation of PI3K/Akt in Diabetes Rat. Int. J. Clin. Exp. Pathol. 2015, 8, 6083–6094. [Google Scholar] [PubMed]
  173. Katagiri, M.; Satoh, A.; Tsuji, S.; Shirasawa, T. Effects of Astaxanthin-Rich Haematococcus Pluvialis Extract on Cognitive Function: A Randomised, Double-Blind, Placebo-Controlled Study. J. Clin. Biochem. Nutr. 2012, 51, 102–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Ito, N.; Seki, S.; Ueda, F. The Protective Role of Astaxanthin for UV-Induced Skin Deterioration in Healthy People—A Randomized, Double-Blind, Placebo-Controlled Trial. Nutrients 2018, 10, 817. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Imai, A.; Oda, Y.; Ito, N.; Seki, S.; Nakagawa, K.; Miyazawa, T.; Ueda, F. Effects of Dietary Supplementation of Astaxanthin and Sesamin on Daily Fatigue: A Randomized, Double-Blind, Placebo-Controlled, Two-Way Crossover Study. Nutrients 2018, 10, 281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Katsumata, T.; Ishibashi, T.; Kyle, D. A Sub-Chronic Toxicity Evaluation of a Natural Astaxanthin-Rich Carotenoid Extract of Paracoccus Carotinifaciens in Rats. Toxicol. Rep. 2014, 1, 582–588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Iwabayashi, M.; Fujioka, N.; Nomoto, K.; Miyazaki, R.; Takahashi, H.; Hibino, S.; Takahashi, Y.; Nishikawa, K.; Nishida, M.; Yonei, Y. Efficacy and Safety of Eight-Week Treatment with Astaxanthin in Individuals Screened for Increased Oxidative Stress Burden. ANTI-AGING Med. 2009, 6, 15–21. [Google Scholar] [CrossRef] [Green Version]
  178. Turck, D.; Castenmiller, J.; de Henauw, S.; Hirsch-Ernst, K.I.; Kearney, J.; Maciuk, A.; Mangelsdorf, I.; McArdle, H.J.; Naska, A.; Pelaez, C.; et al. Safety of Astaxanthin for Its Use as a Novel Food in Food Supplements. EFSA J. 2020, 18, e05993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Lin, Y.-J.; Lin, J.-Y.; Wang, D.-S.; Chen, C.-H.; Chiou, M.-H. Safety Assessment of Astaxanthin Derived from Engineered Escherichia Coli K-12 Using a 13-Week Repeated Dose Oral Toxicity Study and a Prenatal Developmental Toxicity Study in Rats. Regul. Toxicol. Pharmacol. 2017, 87, 95–105. [Google Scholar] [CrossRef] [PubMed]
  180. Buesen, R.; Schulte, S.; Strauss, V.; Treumann, S.; Becker, M.; Gröters, S.; Carvalho, S.; van Ravenzwaay, B. Safety Assessment of [3S, 3′S]-Astaxanthin—Subchronic Toxicity Study in Rats. Food Chem. Toxicol. 2015, 81, 129–136. [Google Scholar] [CrossRef] [PubMed]
  181. Bampidis, V.; Azimonti, G.; de Bastos, M.L.; Christensen, H.; Dusemund, B.; Kouba, M.; Kos Durjava, M.; López-Alonso, M.; López Puente, S.; Marcon, F.; et al. Assessment of the Application for Renewal of Authorisation of Selenomethionine Produced by Saccharomyces Cerevisiae NCYC R397 for All Animal Species. EFSA J. 2019, 17, e05539. [Google Scholar] [CrossRef] [PubMed]
  182. Jannel, S.; Caro, Y.; Bermudes, M.; Petit, T. Novel Insights into the Biotechnological Production of Haematococcus Pluvialis-Derived Astaxanthin: Advances and Key Challenges to Allow Its Industrial Use as Novel Food Ingredient. J. Mar. Sci. Eng. 2020, 8, 789. [Google Scholar] [CrossRef]
  183. Lima, S.G.M.; Freire, M.C.L.C.; da Oliveira, V.S.; Solisio, C.; Converti, A.; de Lima, Á.A.N. Astaxanthin Delivery Systems for Skin Application: A Review. Mar. Drugs 2021, 19, 511. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structures and sources of carotenoids.
Figure 1. Chemical structures and sources of carotenoids.
Marinedrugs 20 00206 g001
Figure 2. The pathways for β-carotene conversion to astaxanthin as proposed by Rhodes [54]. Orange arrows indicate the pathway proposed for most crustaceans. A light blue arrow with a dotted line is a hypothesized pathway for lutein conversion to astaxanthin; yellow arrows indicate an alternative conversion pathway proposed for crustaceans that may not rely on echinenone and canthaxanthin as intermediates.
Figure 2. The pathways for β-carotene conversion to astaxanthin as proposed by Rhodes [54]. Orange arrows indicate the pathway proposed for most crustaceans. A light blue arrow with a dotted line is a hypothesized pathway for lutein conversion to astaxanthin; yellow arrows indicate an alternative conversion pathway proposed for crustaceans that may not rely on echinenone and canthaxanthin as intermediates.
Marinedrugs 20 00206 g002
Figure 3. Orientation of astaxanthin in the cell membrane (adapted from Kishimoto et al. [58]).
Figure 3. Orientation of astaxanthin in the cell membrane (adapted from Kishimoto et al. [58]).
Marinedrugs 20 00206 g003
Table 1. Crustaceans and their byproducts as a source of astaxanthin (review of studies from 2015–2022).
Table 1. Crustaceans and their byproducts as a source of astaxanthin (review of studies from 2015–2022).
SpeciesByproductExtraction ProcedureAstaxanthin ContentSalient FindingReference
Shrimps and prawns (Litopenaeus vannamei (L.v), Macrobrachium rosenbergii (M.r), Penaeus monodon (P.m), Fenneropenaeus chinensis (F.c), and Penaeus japonicus (P.j))Head, shell, and tailSolvent extraction using dichloromethane: methanol (1:3, v/v)19.2 µg/g (L.v.), 15.7 µg/g (M.r.), 2.9 µg/g (P.m.), 7.1 µg/g (F.c.), and 5.8 µg/g (P.j.)The byproduct yield was 44.06–62.53%.[63]
Brown crab (Cancer pagurus)Crab shellsMicrowave (MW) pretreatment in ethanol (at 140 °C and 300 W, during 90 s) followed by supercritical fluid extraction (SFE) (500 bar, 40 °C, and 13 wt % ethanol content, 30 min)1023 µg/g dry extractIn comparison to conventional extraction, the SFE conditions after MW pretreatment gave the best results.[60]
Shrimp (Parapenaeus longirostris)Exoskeleton, including cephalothorax and abdominal partsExtraction using fish oil (CVO) and different fatty acid ethyl esters (TFA) and by SFE (350 bar, 40 °C, 30 min of static extraction followed by dynamic
extraction with a CO2 flow 2.5 L/min for 2 h)
CVO: 149.1 ± 0.8 µg/g
TFA: 160.1 ± 8.9 µg/g
The highest astaxanthin yields were obtained for wet byproducts, extracted with ethyl esters fatty acids at a 2.0 ratio.[64]
Shrimp (Litopenaeus vannamei)Fermented shrimp exoskeletonSFE (300 bar, 60 °C, and 6 mL/min)12.62%, 0.52 µg/gExtracts showed antioxidant activity in vitro.[65]
Tiger prawn (Penaeus monodon) and mud crab (Scylla serrata)DiscardsAutolysis at 55 °C for 20 min on a hot plate with continuous stirring35.76 ± 6.74 μg/gThe highest astaxanthin amount was found when the 60:20 shrimp:crab ratio was used.[66]
Blue crab (Portunus segnis)ShellsConventional extraction, enzymatic extraction,
Soxhlet, maceration
5045 μg/g extractThe highest amount of total carotenoid content was found for combined enzyme-assisted extraction and maceration in hexane/isopropanol (50/50; v/v).[42]
Atlantic shrimp (Pandalus borealis)ShellsUAE solvent extraction by acetone, hexane/isopropanol 3:2 (v/v), and methanol for 5 min at 25 °C270.04, 284.48, and 57.34 mg/gHexane/isopropanol extraction resulted in the highest amount of extracted astaxanthin.[67]
Shrimp (species not determined)ShellsDegradation by Aeromonas hydrophila2.14 ± 0.13 μg/mlThe optimized culture media for higher astaxanthin recovery is characterized by the following conditions: pH 7.0, monosodium glutamate 3% (w/v), glucose (1% w/v) and 30 °C.[68]
Brown crab (Cancer pagurus)ResiduesSupercritical fluid extraction (500 bar, 40 °C, 30 min, 50 g/min)5.18 µg/gOptimized conditions yielded a 1.5-fold higher content of astaxanthin.[69]
Pink shrimp (Farfantepenaeus subtilis)Shrimp waste pasteExtraction using palm olein (90 mL/2.5 g) at 50, 60, and 70 °C26.38 µg/g (50 °C), 28.62 µg/g (60 °C), and 29.18 µg/g (70 °C)Extraction at 70 °C yielded 50.42% astaxanthin.[70]
Shrimp (Litopenaeus vannamei)ShellsShrimp shells, dried under vacuum (40 °C and 175 MPa), were extracted by ethanol28.9 µg/gThe obtained isolate exhibited high antioxidative activity, no toxic effect up to 160 µg/mL on human fibroblast cells, and anti-tyrosinase (12.2 µg/mL) properties.[71]
Shrimps (Parapenaeopsis sculptili, Metapenaeus lysianassa, Macrobrachium rosenbergii, Metapenaeopsis hardwickii, Penaeus merguiensis, and Penaeus monodon)CarapaceExtraction using acetone and methanol (7:3 v/v) and high-pressure processing (HPP) (210 MPa, 10 min)46.95 µg/mL (conventional) 68.26 µg/mL (HPP)HPP improved astaxanthin extraction by around 45%. P. monodon yielded the highest astaxanthin with a shorter extraction time.[72]
Shrimp (Procambarus clarkia)ShellsExtracted using ethanol (1:7) for 20 min at 50 °C using ultrasound (40 kHz) and dried under a vacuum43.7 µg/gExtraction using optimized conditions increased purity by 250 times, exhibiting great application abilities.[62]
Shrimp (species not determined)Fresh head, cooked head, fresh shell and cooked shellExtraction by cooking at 90 °C for 15 min3.64 mg/g (fresh head),
2.38 mg/g (cooked head),
14.65 mg/g (fresh shell), 11.76 mg/g (cooked shell)
Fresh shells contained the highest amount of astaxanthin, and cooking slightly impacted its content.[73]
Shrimp (Penaeus vannamei Boone)ShellsHPE using acetone, dichloromethane, and ethanolRange from 42.3–72.9 μg/g depending on applied pressure and timeHPE resulted in higher extraction yield with improved antioxidant activity.[74]
Shrimp (Litopenaeus vannamei)Cephalothorax, cuticles, pleopods, and tailsLipid extraction for
30 min with ethyl acetate (10 g/50 mL)
7 ± 1 mg/gValorization of shrimp byproducts by the production of an extract rich in bioactive compounds, such as astaxanthin, PUFAs, and α-tocopherol.[75]
Blue crab (Callinectes sapidus)Crab byproductsEnzymatic hydrolysis with alcalase and bromelainRange from 12.0–97.7 μg/g residueProduction of chitin and astaxanthin-enriched extract using enzymatic hydrolysis.[76]
Tiger shrimp (Penaeus monodon)Shrimp wasteSupercritical fluid extraction using carbon dioxide with 15% (v/v) ethanol58.50 ± 2.62 µg/g astaxanthin and 12.20 ± 4.16 µg/g free astaxanthinUse of modeling to determine the best extraction conditions, which were 215.68 bar, 56.88 °C, and 1.89 mL/min for 120 min.[77]
Red (Aristaeomorpha foliacea) and pink shrimp (Parapenaeus longirostris)Muscle and cephalothoraxSolvent extraction using Bligh and Dyer methodFor A. foliacea of total carotenoids: 34.73 ± 0.87 (muscle) and 37.55 ± 0.64 (cephalothorax) % (w/w).
For P. longirostris of total carotenoids: 34.32 ± 0.58 (muscle), 49.08 ± 0.82 (cephalothorax) % (w/w).
Analysis showed higher content of PUFAs (mainly omega-3) and high concentrations of carotenoids (astaxanthin followed by lutein).[78]
Table 2. Biological activity of astaxanthin (review of studies from 2015–2022).
Table 2. Biological activity of astaxanthin (review of studies from 2015–2022).
ActivityForm of Astaxanthin and Its ActionReference
AntioxidantBetter activity of isolated astaxanthin from crabs in comparison to the standard compound investigated by scavenging activity against hydrogen peroxide and 2,2-diphenyl-1-picryl hydrazyl (DPPH) radicals, as reducing power and metal-ion-chelating ability.[94]
In vivo antioxidant efficiency on the alcohol-induced oxidative damage in mice of the water-dispersible, astaxanthin-rich nanopowder.[99]
Improved antioxidant properties of astaxanthin biopolymer nanoparticles in comparison to the free compound tested by in vitro scavenging activity against 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS).[100]
Higher antioxidant activity of microencapsulated astaxanthin from Phaffia rhodozyma.[101]
Applied supercritical emulsions extraction technology resulted in encapsulated astaxanthin in ethyl cellulose with good antioxidant activity.[102]
Effectiveness of astaxanthin in form of nanohydrogels in the neutralization of ROS in vitro.[103]
AntimicrobialThe extent of ROS involvement in antibacterial activity against S. aureus, B. cereus, P. aeroginosa, and E. coli[92]
High activity of astaxanthin isolate from crabs against E. coli detected using the agar diffusion method.[94]
Confirmed antagonism of the astaxanthin methanolic isolate from Sphingomonas faeni against common food-borne pathogens.[96]
Good antimicrobial activity of astaxanthin from crustacean shell byproducts against Escherichia coli, Bacillus, Staphylococcus, and Pseudomonas.[61]
Good antimicrobial activity of astaxanthin from Penaeus monodon against four bacteria (E. coli, E. aerogenes, S. aureus, and B. subtilis), especially for extracts obtained by high-pressure processing.[97]
Effectiveness of astaxanthin from H. pluvialis against E. coli, Salmonella typhi, Vibrio cholera, and S. aureus).[98]
Astaxanthin in bioactive polymers showed significant reduction of bacterial growth and biofilm formation, especially against MRSA.[104]
The good activity of astaxanthin-alpha tocopherol nanoemulsions through the disruption of the integrity of the bacterial cell membrane detected by MIC, MBC, and disk diffusion methods.[105]
Astaxanthin from Asian tiger shrimp shell showed good activity in killing and growth inhibition of E. coli, Pseudomonas aeroginosa, Salmonella typhi, S. aureus, and Streptococcus mutans bacteria.[106]
Anti-inflammatoryEffective in various diseases (diabetes mellitus, Alzheimer’s and Parkinson’s diseases, neuropathic pain, kidney-related diseases, hepatitis, dry eye disease, atopic dermatitis, and inflammatory bowel disease)[107]
The activity is demonstrated by recording suppression of proinflammatory cytokines and inflammatory mediator production in rats with monosodium urate crystal-induced arthritis.[108]
Astaxanthin alleviated the status of epilepticus-induced hippocampal injury in rats and improved cognitive dysfunction.[108]
The anti-inflammatory effect of orally administered astaxanthin was confirmed in mice with ovalbumin-induced asthma.[109]
Cytotoxic, antiproliferative, and anticancer activityAstaxanthin-alpha tocopherol nanoemulsions showed cytotoxicity as a measure of cell viability of four cell lines (CT26, HeLa, Panc1, and T24) and showed a significant decrease in viability after 1 and 2 days of exposure.[105]
Dose-dependent toxicity and antiproliferative effect of gold nanoparticles synthesized using astaxanthin against human breast cancer cells (MDA-MB-231).[110]
Microencapsulated astaxanthin showed inhibition of lipid peroxidation and significant cytostatic activity on adipose-derived stem cells.[111]
Oral treatment of astaxanthin nanoemulsion demonstrated a chemotherapy effect in mice with lung metastatic melanoma by triggering apoptosis.[112]
Astaxanthin administered intragastrically in mice with PC-3 xenograft prostate tumor significantly inhibited its growth.[113]
Astaxanthin suppressed the occurrence of N-nitrosomethylbenzylamine-induced esophageal cancer in rats through antioxidant and anti-inflammation capacity increase.[114]
Significant inhibition of the development of liver cell adenoma and hepatocellular carcinoma in diethylnitrosamine-treated mice by ameliorating serum adiponectin level and improving oxidative stress.[115]
Effect on subchronic testis injury induced by SnS2 nanoflowers in mice; treatment attenuates testicular ultrastructure alterations and histopathological injury and alleviated testicular inflammation, oxidative stress, apoptosis, and necroptosis.[116]
Astaxanthin-alpha tocopherol nanoemulsions showed wound healing potential through scratch assay on HeLa, CT26, and T24 cells.[105]
HepatoprotectiveAstaxanthin-rich nanopowder prepared by nanoencapsulation and freeze-drying showed in vivo antioxidant effect on the alcohol-induced oxidative damage in mice, making the hepatic injury less severe.[99]
Astaxanthin-loaded liposomes provided therapeutic and reparative effects on mice with alcoholic liver fibrosis.[117]
Astaxanthin encapsulated within liposomes caused a reduction of lipopolysaccharide-induced acute hepatotoxicity in rats.[118]
Astaxanthin pretreatment reduces the effect of acetaminophen-induced liver injury in mice by reduction of ROS generation, inhibition of oxidative stress, and reduction of apoptosis[119]
Protection from pancreatic damage and reduces oxidative stress in rats with acute pancreatitis.[120]
AntidiabeticSignificant decrease of total cholesterol and blood glucose levels and increase of high-density lipoprotein cholesterol levels in rats.[121]
Oral administration of astaxanthin reduced lung damage in rat pups with bronchopulmonary dysplasia (induced by hyperoxia and lipopolysaccharide).[122]
Eye healthProtective effect against dry eye disease in vitro on human corneal epithelial cells cultures and in vivo in mice.[123]
Skin healthProtective effects on age-related skin deterioration and environmentally induced damage.[124]
Liposomal astaxanthin showed antidermatotic effects in mice with phthalic anhydride-induced atopic dermatitis.[125]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Šimat, V.; Rathod, N.B.; Čagalj, M.; Hamed, I.; Generalić Mekinić, I. Astaxanthin from Crustaceans and Their Byproducts: A Bioactive Metabolite Candidate for Therapeutic Application. Mar. Drugs 2022, 20, 206. https://doi.org/10.3390/md20030206

AMA Style

Šimat V, Rathod NB, Čagalj M, Hamed I, Generalić Mekinić I. Astaxanthin from Crustaceans and Their Byproducts: A Bioactive Metabolite Candidate for Therapeutic Application. Marine Drugs. 2022; 20(3):206. https://doi.org/10.3390/md20030206

Chicago/Turabian Style

Šimat, Vida, Nikheel Bhojraj Rathod, Martina Čagalj, Imen Hamed, and Ivana Generalić Mekinić. 2022. "Astaxanthin from Crustaceans and Their Byproducts: A Bioactive Metabolite Candidate for Therapeutic Application" Marine Drugs 20, no. 3: 206. https://doi.org/10.3390/md20030206

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop