Next Article in Journal
Natural Polypropionates in 1999–2020: An Overview of Chemical and Biological Diversity
Next Article in Special Issue
Evaluation of Intestinal Absorption of Dietary Halocynthiaxanthin, a Carotenoid from the Sea Squirt Halocynthia roretzi
Previous Article in Journal
Anti-Hepatocellular Carcinoma (HepG2) Activities of Monoterpene Hydroxy Lactones Isolated from the Marine Microalga Tisochrysis Lutea
Previous Article in Special Issue
Stereochemistry of Astaxanthin Biosynthesis in the Marine Harpacticoid Copepod Tigriopus Californicus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Properties of Carotenoids in Fish Fitness: A Review

1
Marine Biochemistry Laboratory, International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
2
Aquaculture and Fisheries Group, Wageningen University and Research, 6708 WD Wageningen, The Netherlands
*
Author to whom correspondence should be addressed.
Mar. Drugs 2020, 18(11), 568; https://doi.org/10.3390/md18110568
Submission received: 29 September 2020 / Revised: 12 November 2020 / Accepted: 16 November 2020 / Published: 19 November 2020
(This article belongs to the Special Issue Marine Carotenoids)

Abstract

:
Carotenoids, one of the most common types of natural pigments, can influence the colors of living organisms. More than 750 kinds of carotenoids have been identified. Generally, carotenoids occur in organisms at low levels. However, the total amount of carotenoids in nature has been estimated to be more than 100 million tons. There are two major types of carotenoids: carotene (solely hydrocarbons that contain no oxygen) and xanthophyll (contains oxygen). Carotenoids are lipid-soluble pigments with conjugated double bonds that exhibit robust antioxidant activity. Many carotenoids, particularly astaxanthin (ASX), are known to improve the antioxidative state and immune system, resulting in providing disease resistance, growth performance, survival, and improved egg quality in farmed fish without exhibiting any cytotoxicity or side effects. ASX cooperatively and synergistically interacts with other antioxidants such as α-tocopherol, ascorbic acid, and glutathione located in the lipophilic hydrophobic compartments of fish tissue. Moreover, ASX can modulate gene expression accompanying alterations in signal transduction by regulating reactive oxygen species (ROS) production. Hence, carotenoids could be used as chemotherapeutic supplements for farmed fish. Carotenoids are regarded as ecologically friendly functional feed additives in the aquaculture industry.

1. Introduction

It is well known that fish have a high risk of exposure to various environmental stressors and infectious diseases. Accordingly, it is thought that effective supplements, such as probiotics, vaccines, vitamins, peptides, lipids, nucleotides, and disease treatments, such as antibiotics, are needed for successful aquaculture [1,2,3,4,5,6,7,8,9,10,11,12,13,14,15]. Infectious diseases are one of the primary contributors to economic loss in the aquaculture industry [10,16]. Antibiotics have been used to prevent disease and improve feed efficiency in fish [17,18,19]. However, the use of antibiotics in aquaculture entails numerous problems, such as the generation of antibiotic-resistant bacteria, accumulation of residual antibiotics in fish tissues, and side effects on fish health [20]. The prevention of disease is always a priority over medicinal treatment. Therefore, functional feed ingredients from natural sources might be considered as safe agents for diseases prevention in farmed animals and humans.
Carotenoids are lipid-soluble pigments that have conjugated double bonds and are the most common class of pigments that occur in nature. More than 750 kinds of carotenoids have been identified. Carotenoids show robust antioxidative activity and have been widely used in the pharmaceutical, cosmetic, food, and feed industries owing to their biological characteristics without exhibiting any cytotoxicity or side effects [21,22,23,24,25,26,27,28,29,30,31]. Through epidemiological studies and clinical trials, dietary carotenoids in mammals have been found to have a correlation with several positive biological effects. Furthermore, carotenoids have several physiological effects, such as those on the immune system, reproduction, lipid metabolism, photoprotection in skin, diseases such as adiposity, obesity, diabetes, cardiovascular disease, hypertension, atherosclerosis, and cancer, and inflammation [7,11,23,26,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62]. Recently, the global market for carotenoids has been estimated to exceed $1.5 billion USD [63,64].
This brief review summarizes information concerning the biological properties of carotenoids, particularly astaxanthin (ASX), as a functional feed ingredient and supplement in fish. Carotenoids have attracted considerable interest and are expected to be economically valuable in aquaculture. This is because the demand for and production of farmed fish is increasing due to the worldwide decline in ocean fisheries stocks [65]. Improvements in global fish farming could relieve pressure on ocean fisheries and support the global food supply.

2. Sources of Carotenoids

The most common carotenoids are 40-carbon isoprenoids, which contain 11 conjugated double bonds. Carotenoids impart color to living organisms such as plants, birds, fish, crustaceans, and bacteria. Generally, carotenoids occur at low levels in organisms; however, the total amount of carotenoids in nature has been estimated to be more than 100 million tons. [7,21,23,39].
Carotenoids are classified into carotenes (e.g., α-carotene, β-carotene, and lycopene), which are composed of carbon and hydrogen, and xanthophylls (e.g., zeaxanthin, lutein, canthaxanthin “CAX”, and ASX), which also contain oxygen [21,22,25,66,67,68]. Figure 1 shows the structure of specific carotenoids found in fish and shellfish. Most animals cannot produce carotenoids de novo. On the contrary, photosynthetic organisms, such as plants, can produce carotenoids de novo. Salmonids cannot synthesize ketocarotenoids, such as ASX (3,3′-dihydroxy-β, β-carotene-4,4′-dione). The amount of carotenoids deposited in fish muscle depends on the level of carotenoids in the feed [7,21,23]. The ASX level in wild salmonid’s muscle reportedly ranges from 3 to 38 mg/kg [27]. Accordingly, salmonid’s flesh seems to be a good dietary source of ASX.
Many carotenoids, in particular ASX, have been applied to coloration of farmed fish [7,11,27,44,69,70,71,72,73,74]. ASX is a common carotenoid used to enhance the pigmentation of farmed salmon. Enhanced salmon filet pigmentation is a vital quality parameter, in that it can influence the consumer’s decision for purchasing. The sources of carotenoids used in aquaculture are synthetic ASX, such as CAROPHYLL Pink (DSM Nutritional Products Ltd., Kaiseraugst, Switzerland), green microalgae Haematococcus pluvialis, red yeast Xanthophyllomyces dendrorhous (formerly designated Phaffia rhodozyma), bacteria Paracoccus carotinifaciens, krill oil and meal, crayfish oil, etc. The sources of carotenoids mentioned above are summarized in Table 1 [7,11,26,27,58,62,70,73,75,76,77,78,79,80]. The level of ASX in red yeast, krill, and crayfish is low. In contrast, H. pluvialis accumulates high levels of ASX, which can accumulate up to 7% of the dry weight of the alga during its growth in response to stressful cultural conditions under intense sunlight [62,64,78,80,81].
The microscopic picture of Haematococcus sp. that accumulated plenty of ASX in their cells is shown in Figure 2. ASX is known to be usually esterified with fatty acid in microalgae [58]. ASX in Haematococcus contains about 70% of monoesters type, 25% of diesters type, and 5% of non-ester-free type [75]. The major manufacturers which produce ASX from Haematococcus are in the United States (Hawaii), Japan, Israel, and Canada [81].

3. Activities of Carotenoids in Farmed Fish

Besides having clear effects on coloration, carotenoids, such as ASX, are considered semi-essential nutrients that promote optimal survival and growth at relatively low dietary-inclusion levels. Several studies have shown that growth or survival can be significantly improved when fish are fed a diet with carotenoids compared to when they are fed a diet without carotenoids [7,11,27,30,44,46,70]. Generally, ASX has been recommended to use in the level of 50–100 mg/kg diet to enhance muscle pigmentation in fish [82]. Furthermore, greater attention needs to be given to carotenoid biotransformation and absorption, and the detection of carotenoid metabolites in the body [26,58,83]. Several analytical methods have been utilized to investigate the role of carotenoids in the body. For example, high-performance liquid chromatography (HPLC), mass spectrometry (MS), and nuclear magnetic resonance (NMR) have been applied for the detection of carotenoid metabolites [24,25,31,68,84]. In particular, LC time-of-flight MS/MS (LC-TOFMS/MS) is believed to be a useful analytical method for natural chemicals and biological compounds, such as carotenoids [85,86].
As shown in Table 2, carotenoids are reported to have many excellent functions in fish [1,7,8,11,27,44,45,46,70,74,87,88,89,90,91]. In the following sections, the biological effects of carotenoids, such as ASX, on fish fitness, particularly on the immune system, cellular damage, and oxidative stress, are described.

3.1. Effects of Important Carotenoids Such as ASX on the Fish Immune System

Carotenoids may help prevent lipid peroxidation, reduce cellular oxidative stress, and reduce inflammatory response in tissues [11,26,28,30,44,45,46,92,93,94]. In short, carotenoids can help prevent chronic stress otherwise caused by a too-strong inflammatory response as part of an overreactive immune system. How? The localized physiological response known as inflammation, in which an affected area may become reddened, swollen, hot, or even painful, is typically part of the body’s normal reaction to injury or infection. When controlled from overreactions, inflammation is and remains an essential part of the innate immune response and helps recruit histamines, white blood cells, prostaglandins, phagocytes, and other cellular mediators to an injured site. Thus, particularly at the early phase of immune responses, inflammation could be considered advantageous, whereas chronic inflammation at a later phase of immune responses could be considered stressful and in need of being prevented.
Carotenoids can convert endogenously produced reactive oxygen species (ROS), such as hydrogen peroxide (H2O2), hydroxyl radicals (OH), and superoxide anions (O2-), into more stable products that cause less damage to cell membranes. The exact dose of carotenoids is relevant and could be important, because the free radicals listed above form an essential part of the innate immune defense system and preferably their effect should only be moderated in situations of overreactions, preventing chronic inflammation. Higher-than-optimal doses of carotenoids could counteract the potentially beneficial effects of these same free radicals released during early inflammatory responses to attack bacterial membranes. After all, professional phagocytes such as neutrophilic granulocytes and macrophages are part of the innate immune system and function primarily with the help of the above-mentioned free radicals to target and destroy pathogens. Indeed, together with nitric oxide (NO), which is induced by cytokine-inducible NO-synthase (iNOS or NOS2), both ROS and reactive nitrogen species (RNS) are essential mediators of the microbicidal activity of these professional phagocytes [94,95]. Overall, the release of RNS and ROS by professional phagocytes, generally referred to as the oxidative burst, is considered an important defense mechanism designed to destroy pathogens and should, therefore, be considered a beneficial reaction that should not be annulled or interfered with by high levels of antioxidants (i.e., carotenoids). Overshoot of this reaction, in contrast, could lead to situations of chronic inflammation that could be counteracted by carotenoids: nitrogen and oxygen radicals should only be considered beneficial up to a point.
Upon overreactions of the immune system, professional phagocytes may end up over-producing nitrogen and oxygen radicals by continuous and chronic activation, contributing to cell dysfunction and cell death beyond that of bacteria. Indeed, in the presence of Fe2+ ions and through the Fenton reaction, H2O2 can be broken down into ·OH, which are the most reactive ROS and can also damage host cell structures (proteins, lipids, and DNA). Further, mitochondrial NO can react with O2- to yield peroxynitrite (ONOO−), a highly potent oxidant that is even more reactive than its precursors. This oxidant can cause nitration (mainly in tyrosine residues) of many proteins, but can also oxidize amino acid residues essential for the functioning of several proteins, including transcription factors and enzymes [94,96,97,98]. Not only in mammals but also in fish, among the professional phagocytes, macrophages appear better equipped to fight potentially damaging and, therefore, negative aspects of oxidation than neutrophils [99]. No matter what, even when produced and (mostly) contained by professional phagocytes, very high concentrations of RNS and/or ROS can be harmful and need to be countered.
As introduced above, carotenoids are classified into carotenes and xanthophylls, the latter including ASX, a major colorant of farmed fish and common carotenoid used to enhance the pigmentation of farmed salmon. Most studies on the effects of carotenoids on the immune system of fish, therefore, have been performed with ASX. In studies targeting farmed fish species, clues on innate immune regulation by ASX frequently come from studying messengers important for the immune system, such as cytokines, and mostly by measuring gene expression levels by real-time quantitative PCR, simply because practical alternatives to measure protein rather than gene expression, such as enzyme-linked immunosorbent assay (ELISA), regretfully have remained scarce [100]. Pro-inflammatory cytokines are the most studied, and most frequently, those are interleukin-1 beta (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNFα), also in fish [101], but the outcomes are difficult to interpret. For example, in a recent study examining effects of dietary CAX (ASX is a metabolite of CAX) on the growth performance and antioxidant status of yellow perch (Perca flavescens), the authors reported an upregulation of IL-1β gene expression in groups fed CAX, compared to that in control fish not receiving CAX [102]. In apparent contrast, exposure of snakehead (Channa argus) hepatocytes to bacterial lipopolysaccharides showed that these cells could be protected from a subsequent inflammatory response by pretreatment with ASX, preventing the upregulation of IL-1β, IL-6, and TNFα [103]. Indeed, a complicating factor in studies on the effects of external factors such as carotenoids on immune responses, is that pro-inflammatory cytokines typically can be produced by a broad range of cells that are not always exclusively immune cells. Maybe, rather than studying up- or down-regulation of a single cytokine gene, complex studies into immune-modulating effects of factors such as carotenoids are better off taking whole-transcriptome approaches [104], more broadly addressing the research question. After all, a reliable analysis of complex physiological changes requires the use of equally complex arrays of gene expression or other bio-assay results before concluding on fish health or disease resistance. Of course, this does not rule out IL-1β as an informative marker, but it does point at the complexity of studies addressing the physiological effects of carotenoids, such as ASX, on the immune system.
Although maybe difficult to explain in immunological detail without further studies on the fish immune system, it seems likely that professional phagocytes such as macrophages, along with their own cell-related antioxidant and cytoprotective pathways that help reduce the oxidative burden, can make use of diet-derived carotenoid antioxidants such as ASX, as they do for ascorbic acid and α-tocopherol [96]. Certainly, since the body concentrations of ROS scavenger molecules, such as ASX and β-carotene, are influenced by dietary intake, it is conceivable that higher concentrations of carotenoids in the diet could help professional phagocytes overcome the side effects caused by excessive oxidation, restrict chronic inflammation and therefore, be beneficial. These conclusions appear supported by studies in several fish species that report on a variety of enhanced immune reactivities after dietary supplementation of ASX and β-carotene [105,106,107,108,109,110]. Thus, like mammals [11,111], carotenoids may be regarded as biogenics with enough potential to modulate ROS/RNS production and immune function in fish.

3.2. Effects of Carotenoids, Particularly ASX, on Cellular Damage and Oxidative Stress in Fish

Several kinds of carotenoids have provitamin A activity, stimulate growth, and improve fish reproduction [13,27,70,112]. Salmonid eggs usually contain high ASX levels, which can enhance the quality of eggs during the initial feeding period of growth. Additionally, ASX can protect salmonid egg membranes from oxidative injury caused by UV radiation [27,54,70]. This photoprotection activity of ASX might be one of the most specific functions of carotenoids in organisms, including fish.
Recently, attention has been given to the anti-stress activity of carotenoids, besides their provitamin A activity. This is because farmed fish are often exposed to environmental stressors in ordinary cultural conditions [20,113,114,115,116,117,118,119,120,121]. Environmental stress in fish is thought to be derived from several factors (stressors), such as diseases, chemicals, acute changes in temperature, heavy metals, and aquacultural conditions.
Several cases regarding environmental stress in fish have been reported. The bacterium Edwardsiella tarda is a virulent intracellular pathogen found in commercial fish species. Edwardsiellosis, caused by E. tarda, is one of the most severe diseases in the Japanese flounder Paralichthys olivaceus. Heat shock protein 70 (HSP70) expression was increased in fish infected with E. tarda, compared to that in control fish [16]. The expression levels of Cu, Zn–superoxide dismutase (Cu, Zn–SOD) in fish increased after infection. In contrast, the expression level of Mn–SOD in fish gradually increased after infection and remained high between 24 and 48 h post-infection [16].
In aquaculture, chemicals such as antiseptics, antibiotics, and parasiticides are often used to prevent or treat diseases [19]. Oxytetracycline (OTC) is an antibiotic that belongs to the tetracycline family and has been used against several bacterial infections in farmed fish. However, high doses of OTC have been known to cause side effects in fish [17,18,19]. The glutathione (GSH) levels in the plasma, liver, muscle, and stomach of OTC-fed fish were higher than those in control fish [20]. Studies have demonstrated that organisms have both enzymatic and non-enzymatic antioxidant defense systems against ROS [98,121,122,123,124,125,126,127,128]. GSH is the major non-protein cellular thiol with reducing and nucleophilic properties [94,98,121,125,129,130]. GSH is also known to be the substrate for glutathione peroxidase (GPX), an antioxidative enzyme that can eliminate the lipid peroxide (LPO) generated within cells [94,121,127,128,130]. Accordingly, OTC-induced stress might increase the metabolic turnover of GSH due to its consumption by scavenging oxidants generated by chemical stress.
The physiological states of fish depend on their environmental temperature. Accordingly, the environmental temperature can induce numerous changes in the fish body. Increased temperature heightens oxygen consumption, resulting in increased ROS production [118,124]. The effect of thermal stressors on redox-related biomarkers in fish has been reported [118]. For example, the plasma LPO levels in fish exposed to heat shock were shown to increase. LPO in stressed fish plasma might be derived from various damaged tissues. The plasma GSH levels in fish initially decreased but returned to basal levels at 17.5 h post-heat shock [118]. Considered one of the primary antioxidative enzymes, the plasma activity of SOD in stressed fish was significantly increased compared with that in the control group after heat shock. Moreover, the expression levels of hepatic GSH and HSP70 gradually increased after heat shock treatment. HSP70 assists in the folding of nascent polypeptide chains, acts as a molecular chaperon or helper molecule, mediates the repair and degradation of denatured proteins, and is involved in the breakdown and replacement of irreparable proteins. HSPs have various species-specific functions and are usually induced during stress. In response to stressors, such as chemicals, heat shock, bacterial pathogens, pollutants, and physiological states, the induction of various HSP families has already been reported in cell lines and various fish tissues [113,115,118,120,131,132,133,134,135,136].
The fish oil used in aquafeed contains plenty of highly unsaturated fatty acids (HUFA), such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), which are readily oxidized. The levels of serum LPO, transaminase activity, and lipids (triglycerides, total cholesterol, and phospholipids) in fish fed oxidized oil were observed to be higher than those in fish provided non-oxidized oil [46]. Oxidized oil usually contains LPO and secondary products. LPO is further decomposed into a variety of ROS that attack cell components and induce stress [94,96,98,122,125,137].
Hence, the above-mentioned results concerning the changing patterns of stress-related biomarkers, such as GSH, SOD, LPO, and HSP70, suggest that the stressors should affect the redox state and induce oxidative stress in fish. These results also suggest that GSH, SOD, and HSP70 might play essential roles in mediating the response and the defense upon oxidative stress in fish. The production of ROS, which are strong oxidants and induce oxidative stress in cells, is already known to increase under certain stressful conditions. The oxidative stress often leads to an increased risk of diseases [35,38,42,46,94,98,118,121,122,123,125,137,138,139,140,141,142]. Accordingly, insufficient ingestion of antioxidants, such as carotenoids and α-tocopherol, might increase an organism’s susceptibility to oxidative stress-related diseases.
The biological effects of carotenoids, such as ASX and lycopene, on oxidative stress in fish have been reported [7,11,27,30,44,45,46,74,107,142,143]. The liver and plasma from fish fed ASX were observed to have a significantly higher level of α-tocopherol and carotenoids than those from control fish fed a non-ASX diet. On the contrary, ASX significantly decreased LPO levels in several tissues. Plasma LPO is a useful indicator of tissue damage due to oxidative stress in the body [144]. ASX, including red yeast, was also found to decrease the level of LPO in fish serum [30,44,46]. Most LPO is found in lipoproteins that contain highly susceptible lipids [54,144]. ASX and α-tocopherol exist in the lipoproteins of fish serum along with several kinds of circulating antioxidants [30,145]. Carotenoids and α-tocopherol are thought to be in biological membranes, which contain a large amount of HUFA. Hence, ASX, α-tocopherol, and other antioxidants might synergistically protect lipoproteins from oxidation, thereby reducing oxidative stress in the body. Additionally, the administration of carotenoids, such as ASX and lycopene, have been observed to enhance the production of antioxidative enzymes, such as SOD and GPX, and the cellular endogenous antioxidants, such as GSH, in fish, mammals, and invertebrate [27,37,43,74,107,142,143,146,147]. The supplementation of carotenoids has also been found to upregulate the expression of HSP in the tissue of several animal species and cultured cells [27,107,146,148,149]. Taken together, these results suggest that dietary carotenoids can improve the antioxidative defense including antioxidative enzymes, cellular endogenous antioxidants, and HSP, and strengthen the ability of resistance against oxidative stress in the body.
ASX is known to be one of the most effective antioxidative compounds owing to its specific structure. Indeed, the antioxidative activity of ASX is 100-fold and 10-fold higher than that of α-tocopherol and β-carotene, respectively [62,150]. ASX has conjugated carbon-carbon double bonds and hydroxyl (–OH) and keto (=O) groups in its structure and shows both lipophilic (hydrophobic, non-polar) and hydrophilic (polar) properties. The hydroxyl groups of ASX are anchored across membranes, with polar functional groups oriented outside the membrane. The backbones of ASX might serve as a molecular wire increasing the mechanical strength of the membrane [28,38,49,151]. Accordingly, membrane-bound antioxidants, such as ASX and α-tocopherol, protect membrane lipids and proteins from oxidation. Carotenoids can improve the cellular antioxidant capacity by regenerating α-tocopherol and ascorbic acid from their corresponding radical forms [94,152]. Carotenoids show cooperative or synergistic interactions with other antioxidants such as α-tocopherol, ascorbic acid, and GSH, located in lipophilic and hydrophobic compartments of tissue [37]. Essentially, among trout fibroblastic cells cultured under ROS-induced oxidative stress, membrane damage was observed in cells without ASX, whereas cells containing ASX had suppressed cellular membrane damage and increased viability [30]. These results suggest that ASX can effectively protect cells against oxidative damage induced by ROS and maintain both membrane dynamics and cellular function.

3.3. Molecular Mechanisms of Carotenoid’s Actions in the Body

The antioxidative activity shown by carotenoids might not only act by directly scavenging ROS, but also by modulating expression of stress and antioxidative-related proteins. The molecular mechanisms of action shown by carotenoids in the body have been classified into the following four categories: (1) antioxidative and pro-oxidative activities, (2) suppression of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling translation, (3) activation of the nuclear factor erythroid 2-related factor 2 (Nrf2), and (4) interaction with other transcription factors [26,142,153,154]. Transcription factors such as NF-κB and Nrf2 are known to associate with immune reaction, inflammation, and oxidative stress responses. The NF-κB pathway is activated by inflammatory compounds, such as tumor necrosis factor-α (TNF-α) and cytokines, and oxidative stress. On the contrary, the Nrf2 pathway is known to be an important pathway in the defense against ROS-induced oxidative stress in cells [153]. The conjugated double bond in carotenoids can act as a strong antioxidant by donating electrons and reacting with free radicals to convert them to more stable forms. Carotenoids are thought to react with free radicals through the following three reaction mechanisms: (1) electron transfer (oxidation and reduction), (2) hydrogen abstraction (allylic hydrogen atom abstraction), and (3) radical addition (adduct formation) [37,43,49,54,80,94,155,156,157]. In addition, intercellular signaling is often affected by ROS and oxidative stress, resulting in changes in the expression of several genes [122,124,125,129,139,158,159,160].
Accumulated ROS potentially activate signaling cascades involving transcription factors such as NF-κB and activator protein-1 (AP-1), and induce the expression of SODs to neutralize the harmful effects of ROS [74,118,124,158,161,162,163,164,165]. ASX can inhibit ROS-induced expressions of NF-κB, which is involved in the transcription of inducible genes, such as heme oxygenase 1 (HO-1) and iNOS, that regulate inflammatory responses and oxidative stress [58,153]. ROS-induced oxidative stress has been reported to activate several kinases, such as protein kinase A (PKA), mitogen-activated protein kinase (MAPK), c-Jun N-terminal kinase, extracellular signal-regulated kinase, and transcription factor AP-1 [166]. The accumulation of Nrf2 protein can induce the expression of HO-1, and the phosphorylation of EPK and p38 kinase [58]. Oxidative stress is also known to attenuate post-PKA pathways, resulting in inhibited expression of the mature form of steroidogenic acute regulatory protein (STAR). ASX can prevent the downregulation of the expression of STAR caused by ROS [43,58,167]. Thus, under oxidative stress, carotenoids might modulate gene expression accompanying alterations in signal transduction through the regulating ROS formation in the tissue.

4. Conclusions

The demand for and production of farmed fish has increased due to the worldwide decline of ocean fisheries stocks. It is known that almost half of the world’s fishery production is currently based on aquaculture [65]. The total global aquaculture production is estimated to have reached about 82 million tons of aquatic animals ($250 billion USD) in 2018. The aquatic animals farming was dominated by finfish (54 million tons) in 2018 [65]. Aquaculture could be a major source of aquatic dietary proteins by 2050 as it can sustainably enhance food supply. Thus, appropriate monitoring and evaluation methods for impact and risk of aquaculture on the environment need to be considered [65,168,169,170,171]. Additionally, Atlantic salmon (Salmo salar), rainbow trout (Oncorhynchus mykiss), and coho salmon (Oncorhynchus kisutch) are known to be valued salmonid species used in aquaculture worldwide [65,85,114,172,173,174]. Particularly in Japan, coho salmon farming is one of the basic industries in northeastern Pacific coastal areas where the great earthquake and tsunami occurred in 2011 [173,175,176]. Accordingly, there is a desire to improve productivity of coho salmon farming and quality of coho salmon for reconstruction of coastal fisheries in disaster-stricken areas.
Many substances have been used for farmed fish as feed supplements. Carotenoids, being one of them, should be safe for fish and fish consumers. The use of carotenoids in aquaculture should employ an ecologically friendly method. Thus, carotenoids seem to be one of the ideal functional ingredient types in aquaculture. However, some carotenoids have been suggested to act as pro-oxidants under certain conditions [26,28,43,155,156,157,177,178]. It is important to find and maintain an appropriate balance between antioxidants and oxidants in the body [26,34,156,179,180,181,182,183]. In addition, carotenoids have been known to be susceptible to oxidation, so that peroxides perhaps could occur in carotenoid samples during storage. These peroxides might influence the actions of carotenoids as a pro-oxidant [28,156]. Further studies are required to reveal the relationships between antioxidation, oxidative stress, immune stimulation, and gene expression modulated by carotenoids, and their contribution to fish fitness. These studies would provide useful information to establish sustainability in aquaculture.

Author Contributions

The authors wrote and reviewed the manuscript. Both authors have read and agreed to the published version of the manuscript.

Funding

Some studies performed by T.N. were financially supported in part by a Grant-in-Aid for Scientific Research (KAKENHI), a fund from the Core-to-Core Program (A. Advanced Research Networks) entitled “Establishment of international agricultural immunology research-core for a quantum improvement in food safety” from the Japan Society for the Promotion of Science (JSPS), and a fund from the Project of the NARO Bio-oriented Technology Research Advancement Institution (R&D Matching Funds in the Field for Knowledge Integration and Innovation).

Acknowledgments

The authors are grateful to Y. Ochiai, H. Shirakawa, and H. Kitazawa at Graduate School of Agricultural Science, Tohoku University, Japan, for their valuable suggestions. T.N. is also grateful to E.M. Donaldson, Scientist Emeritus, and R.H. Devlin at West Vancouver Laboratory, Fisheries and Oceans Canada; G.K. Iwama, President, at Quest University, Canada (former Executive Vice President of the Okinawa Institute of Science and Technology Graduate University, OIST, Japan); L.O.B. Afonso at Faculty of Science, Engineering and Built Environment, Deakin University, Australia; R. Yada, Dean, at Faculty of Land and Food Systems, and J. Richards at Department of Zoology, University of British Columbia, Canada, for giving an opportunity to visit West Vancouver Laboratory, University of British Columbia, and NRC Institute for Marine Biosciences, Canada, to study the biochemical and biophysical features of salmonid fish. The authors would like to thank G. Nishitani at Graduate School of Agricultural Science, Tohoku University, Japan, for providing a picture of microalgae Haematococcus sp.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Meyers, S.P. Developments in world aquaculture, feed formulations, and role of carotenoids. Pure Appl. Chem. 1994, 66, 1069–1076. [Google Scholar] [CrossRef] [Green Version]
  2. Fletcher, T.C. Dietary effects on stress and health. In Fish Stress and Health in Aquaculture; Iwama, G.K., Pickering, A.D., Sumpter, J.P., Schreck, C.B., Eds.; Cambridge University Press: Cambridge, UK, 1997; pp. 223–246. [Google Scholar]
  3. Gatesoupe, F.J. The use of probiotics in aquaculture. Aquaculture 1999, 180, 147–164. [Google Scholar] [CrossRef]
  4. Olafsen, J.A. Interactions between fish larvae and bacteria in marine aquaculture. Aquaculture 2001, 200, 223–247. [Google Scholar] [CrossRef]
  5. Irianto, A.; Austin, B. Probiotics in aquaculture. J. Fish Dis. 2002, 25, 633–642. [Google Scholar] [CrossRef]
  6. Vine, N.G.; Leukes, W.D.; Kaiser, H. Probiotics in marine larviculture. FEMS Microbiol. Rev. 2006, 30, 404–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Nakano, T. Microorganisms. In Micronutrients and Health of Cultured Fish; Nakagawa, H., Sato, M., Eds.; Koseisha Koseikaku: Tokyo, Japan, 2003; pp. 95–106. [Google Scholar]
  8. Pohlenz, C.; Gatlin III, D.M. Interrelationships between fish nutrition and health. Aquaculture 2014, 431, 111–117. [Google Scholar] [CrossRef]
  9. Burr, G.; Gatlin III, D.M.; Ricke, S. Microbial ecology of the gastrointestinal tract of fish and the potential application of prebiotics and probiotics in finfish aquaculture. J. World Aquacult. Soc. 2005, 36, 425–436. [Google Scholar] [CrossRef]
  10. Maita, M. Fish health assessment. In Dietary Supplements for the Health and Quality of Cultured Fish; Nakagawa, H., Sato, M., Gatlin, D.M., III, Eds.; CAB International: Oxford, UK, 2007; pp. 86–108. [Google Scholar]
  11. Nakano, T. Microorganisms. In Dietary Supplements for the Health and Quality of Cultured Fish; Nakagawa, H., Sato, M., Gatlin, D.M., III, Eds.; CAB International: Oxfordshire, UK, 2007; pp. 86–108. [Google Scholar]
  12. Takeuchi, T. Amino acids, peptides. In Dietary Supplements for the Health and Quality of Cultured Fish; Nakagawa, H., Sato, M., Gatlin, D.M., III, Eds.; CAB International: Oxford, UK, 2007; pp. 47–63. [Google Scholar]
  13. Koshio, S. Vitamins. In Dietary Supplements for the Health and Quality of Cultured Fish; Nakagawa, H., Sato, M., Gatlin, D.M., III, Eds.; CAB International: Oxford, UK, 2007; pp. 35–46. [Google Scholar]
  14. Ishikawa, M. Lipids. In Dietary Supplements for the Health and Quality of Cultured Fish; Nakagawa, H., Sato, M., Gatlin, D.M., III, Eds.; CAB International: Oxford, UK, 2007; pp. 64–73. [Google Scholar]
  15. Gatlin III, D.M.; Barrows, F.T.; Brown, P.; Dabrowski, K.; Gaylord, T.G.; Hardy, R.W.; Herman, E.; Hu, G.; Krogdahl, A.; Nelson, R.; et al. Expanding the utilization of sustainable plant products in aquafeeds: A review. Aquacult. Res. 2007, 38, 551–579. [Google Scholar] [CrossRef]
  16. Nakano, T.; Osatomi, K.; Miura, N.; Aikawa-Fukuda, Y.; Kanai, K.; Yoshida, A.; Shirakawa, H.; Yamauchi, A.; Yamaguchi, T.; Ochiai, Y. Effect of bacterial infection on the expression of stress proteins and antioxidative enzymes in Japanese flounder. In Evolution of Marine Coastal Ecosystems under the Pressure of Global Changes; Ceccaldi, H.J., Henocque, Y., Komatsu, T., Prouzet, P., Sautour, B., Yoshida, J., Eds.; Springer Nature: Cham, Switzerland, 2020; pp. 111–127. [Google Scholar]
  17. Yonar, M.E. The effect of lycopene on oxytetracycline-induced oxidative stress and immunosuppression in rainbow trout (Oncorhynchus mykiss, W.). Fish Shellfish Immunol. 2012, 32, 994–1001. [Google Scholar] [CrossRef]
  18. Elia, A.C.; Ciccotelli, V.; Pacini, N.; Dorr, A.J.M.; Gili, M.; Natali, M.; Gasco, L.; Prearo, M.; Abete, M.C. Transferability of oxytetracycline (OTC) from feed to carp muscle and evaluation of the antibiotic effects on antioxidant systems in liver and kidney. Fish Physiol. Biochem. 2014, 40, 1055–1068. [Google Scholar] [CrossRef]
  19. Burridge, L.; Weis, J.S.; Cabello, F.; Pizarro, J.; Bostick, K. Chemical use in aquaculture: A review of current practices and possible environmental effects. Aquaculture 2010, 306, 7–23. [Google Scholar] [CrossRef]
  20. Nakano, T.; Hayashi, S.; Nagamine, N. Effect of excessive doses of oxytetracycline on stress-related biomarker expression in coho salmon. Environ. Sci. Pollut. Res. 2018, 25, 7121–7128. [Google Scholar] [CrossRef] [PubMed]
  21. Takaichi, S. Carotenoids. In Carotenoids-Biological Functions and Diversity; Takaichi, S., Ed.; Shokabo: Tokyo, Japan, 2006; pp. 1–20. [Google Scholar]
  22. Khachik, F. Distribution and metabolism of dietary carotenoids in humans as a criterion for development of nutritional supplements. Pure Appl. Chem. 2006, 78, 1551–1557. [Google Scholar] [CrossRef]
  23. Maoka, T. Diversity and biological functions of natural carotenoids. FFI J. 2015, 220, 118–124. [Google Scholar]
  24. Maoka, T. Recent progress in structural studies of carotenoids in animals and plants. Arch. Biochem. Biophys. 2009, 483, 191–195. [Google Scholar] [CrossRef]
  25. Maoka, T. Experimental techniques for structural elucidation and analysis of natural carotenoids. Oreoscience 2012, 12, 485–501. [Google Scholar] [CrossRef] [Green Version]
  26. Rodriguez-Concepcion, M.; Avalos, J.; Bonet, M.L.; Boronat, A.; Gomez-Gomez, L.; Hornero-Mendez, D.; Limon, M.C.; Melendez-Martinez, A.J.; Olmedilla-Alonso, B.; Palou, A.; et al. A global perspective on carotenoids: Metabolism, biotechnology, and benefits for nutrition and health. Prog. Lipid Res. 2018, 70, 62–93. [Google Scholar] [CrossRef] [Green Version]
  27. 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]
  28. Britton, G. Carotenoid research: History and new perspectives for chemistry in biological systems. Biochim. Biophys. Acta Mol. Cell. Biol. Lipids 2020, 1865, 158699. [Google Scholar] [CrossRef]
  29. Milani, A.; Basirnejad, M.; Shahbazi, S.; Bolhassani, A. Carotenoids: Biochemistry, pharmacology and treatment. Br. J. Pharmacol. 2017, 174, 1290–1324. [Google Scholar] [CrossRef] [Green Version]
  30. Nakano, T.; Wazawa, M.; Yamaguchi, T.; Sato, M.; Iwama, G.K. Positive biological actions of astaxanthin in rainbow trout. Mar. Biotechnol. 2004, 6, S100–S105. [Google Scholar]
  31. Maoka, T. Carotenoids as natural functional pigments. J. Nat. Med. 2020, 74, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Choi, H.D.; Youn, Y.K.; Shin, W.G. Positive effects of astaxanthin of lipid profiles and oxidative stress in overweight subjects. Plant Foods Hum. Nutr. 2011, 66, 363–369. [Google Scholar] [CrossRef] [PubMed]
  33. Bonet, M.L.; Canas, J.A.; Ribot, J.; Palou, A. Carotenoids and their conversion products in the control of adipocyte function, adiposity and obesity. Arch. Biochem. Biophys. 2015, 572, 112–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Aslani, B.A.; Ghobadi, S. Studies on oxidants and antioxidants with a brief glance at their relevance to the immune system. Life Sci. 2016, 146, 163–173. [Google Scholar] [CrossRef]
  35. Aoi, W.; Naito, Y.; Takanami, Y.; Ishii, T.; Kawai, Y.; Akagiri, S.; Kato, Y.; Osawa, T.; Yoshikawa, T. Astaxanthin improves muscle lipid metabolism in exercise via inhibitory effect of oxidative CPT I modification. Biochem. Biophys. Res. Commun. 2008, 366, 892–897. [Google Scholar] [CrossRef]
  36. Amaro, H.M.; Barros, R.; Guedes, A.C.; Sousa-Pinto, I.; Malcata, F.X. Microalgal compounds modulate carcinogenesis in the gastrointestinal tract. Trends Biotechnol. 2013, 31, 92–98. [Google Scholar] [CrossRef]
  37. Krinsky, N.I.; Johnson, E.J. Carotenoid actions and their relation to health and disease. Mol. Aspects Med. 2005, 26, 459–516. [Google Scholar] [CrossRef]
  38. Kishimoto, Y.; Yoshida, H.; Kondo, K. Potential anti-atherosclerotic properties of astaxanthin. Mar. Drugs 2016, 14, 35. [Google Scholar] [CrossRef]
  39. World Health Organization. IARC, Carotenoids. In IARC Handbooks of Cancer Prevention; International Agency for Research on Cancer, IARC/WHO: Lyon, France, 1998; pp. 15–45. [Google Scholar]
  40. Hussein, G.; Nakamura, M.; Zhao, Q.; Iguchi, H.; Goto, H.; Sankawa, U.; Watanabe, H. Anti-hypertensive and neuroprotecitve effects of astaxanthin in experimental animals. Biol. Pharm. Bull. 2005, 28, 47–52. [Google Scholar] [CrossRef] [Green Version]
  41. Hughes, D.A. Dietary carotenoids and human immune function. Nutrition 2001, 17, 823–827. [Google Scholar] [CrossRef]
  42. Guerin, M.; Huntley, M.E.; Olaizola, M. Haematococcus astaxanthin: Applications for human health and nutrition. Trends Biotechnol. 2003, 21, 210–216. [Google Scholar] [CrossRef]
  43. Gammone, M.A.; Riccioni, G.; D’Orazio, N. Marine carotenoids against oxidative stress: Effects on human health. Mar. Drugs 2015, 13, 6226–6246. [Google Scholar] [CrossRef] [PubMed]
  44. Nakano, T.; Tosa, M.; Takeuchi, M. Improvement of biochemical features in fish health by red yeast and synthetic astaxanthin. J. Agric. Food Chem. 1995, 43, 1570–1573. [Google Scholar] [CrossRef]
  45. Nakano, T.; Miura, Y.; Wazawa, M.; Sato, M.; Takeuchi, M. Red yeast Phaffia rhodozyma reduces susceptibility of liver homogenate to lipid peroxidation in rainbow trout. Fish. Sci. 1999, 65, 961–962. [Google Scholar] [CrossRef] [Green Version]
  46. Nakano, T.; Kanmuri, T.; Sato, M.; Takeuchi, M. Effect of astaxanthin rich red yeast (Phaffia rhodozyma) on oxidative stress in rainbow trout. Biochim. Biophys. Acta 1999, 1426, 119–125. [Google Scholar] [CrossRef]
  47. Miyashita, K.; Hosokawa, M. Focus on a brown seaweed carotenoid, fucoxanthin. FFI J. 2015, 220, 101–109. [Google Scholar]
  48. Rao, A.V.; Rao, L.G. Carotenoids and human health. Pharmacol. Res. 2007, 55, 207–216. [Google Scholar] [CrossRef]
  49. Pashkow, F.J.; Watumull, D.G.; Campbell, C.L. Astaxanthin: A novel potential treatment for oxidative stress and inflammation in cardiovascular disease. Am. J. Cardiol. 2008, 101, S58–S68. [Google Scholar] [CrossRef]
  50. Nishino, H.; Murakoshi, M.; Tokuda, H.; Satomi, Y. Cancer prevention by carotenoids. Arch. Biochem. Biophys. 2009, 483, 165–168. [Google Scholar] [CrossRef]
  51. Zhang, L.; Wang, H. Multiple mechanisms of anti-cancer effects exerted by astaxanthin. Mar. Drugs 2015, 13, 4310–4330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Yoshida, H.; Yanai, H.; Ito, K.; Koikeda, T.; Tsukahara, H.; Tada, N. Administration of natural astaxanthin increase serum HDL-cholesterol and adiponectin in subjects with mild hyperlipidemia. Atherosclerosis 2010, 209, 520–523. [Google Scholar] [CrossRef] [PubMed]
  53. Wu, H.; Niu, H.; Shao, A.; Wu, C.; Dixon, B.J.; 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]
  54. Tomita, Y. Functions and biological activities in animal. In Carotenoids-Biological Functions and Diversity; Takaichi, S., Ed.; Shokabo: Tokyo, Japan, 2006; pp. 67–107. [Google Scholar]
  55. Rowles, J.L., 3rd; Erdman, J.W., Jr. Carotenoids and their role in cancer prevention. Biochim. Biophys. Acta Mol. Cell. Biol. Lipids 2020, 1865, 158613. [Google Scholar] [CrossRef]
  56. Eggersdorfer, M.; Wyss, A. Carotenoids in human nutrition and health. Arch. Biochem. Biophys. 2018, 652, 18–26. [Google Scholar] [CrossRef]
  57. Bae, M.; Kim, M.B.; Park, Y.K.; Lee, J.Y. Health benefits of fucoxanthin in the prevention of chronic diseases. Biochim. Biophys. Acta Mol. Cell. Biol. Lipids 2020, 1865, 158618. [Google Scholar] [CrossRef]
  58. Le Goff, M.; Le Ferrec, E.; Mayer, C.; Mimouni, V.; Lagadic-Gossmann, D.; Schoefs, B.; Ulmann, L. Microalgal carotenoids and phytosterols regulate biochemical mechanisms involved in human health and disease prevention. Biochimie 2019, 167, 106–118. [Google Scholar] [CrossRef]
  59. Zerres, S.; Stahl, W. Carotenoids in human skin. Biochim. Biophys. Acta 2020, 1865, 158588. [Google Scholar] [CrossRef]
  60. Zheng, J.; Manabe, Y.; Sugawara, T. Siphonaxanthin, a carotenoid from green algae Codium cylindricum, protects Ob/Ob mice fed on a high-fat diet against lipotoxicity by ameliorating somatic stresses and restoring anti-oxidative capacity. Nutr. Res. 2020, 77, 29–42. [Google Scholar] [CrossRef]
  61. Komatsu, T.; Sasaki, S.; Manabe, Y.; Hirata, T.; Sugawara, T. Preventive effect of dietary astaxanthin on UVA-induced skin photoaging in hairless mice. PLoS ONE 2017, 12, e0171178. [Google Scholar] [CrossRef] [Green Version]
  62. Mularczyk, M.; Michalak, I.; Marycz, K. Astaxanthin and other nutrients from Haematococcus pluvialis-multifunctional applications. Mar. Drugs 2020, 18, 459. [Google Scholar] [CrossRef] [PubMed]
  63. Nakao, T.; Sakata, S. Carotenoids powder as a food ingredient. FFI J. 2019, 224, 428–431. [Google Scholar]
  64. Sathasivam, R.; Ki, J.S. A review of the biological activities of microalgal carotenoids and their potential use in healthcare and cosmetic industries. Mar. Drugs 2018, 16, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. FAO. The State of World Fishery and Aquaculture 2020 (SOFIA); Food and Agriculture Organization of the United Nations: Rome, Italy, 2020. [Google Scholar] [CrossRef]
  66. Olson, J.A. Vitamin A and carotenoids as antioxidants in a physiological context. J. Nutr. Sci. Vitaminol. 1993, 39, S57–S65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Mannazzu, I.; Landolfo, S.; Lopes de Silva, T.; Buzzini, P. Red yeasts and carotenoid production: Outlining a future for non-conventional yeasts of biotechnological interest. World J. Microbiol. Biotechnol. 2015, 31, 1665–1673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Arathi, B.P.; Sowmya, R.R.; Vijay, K.; Baskaran, V.; Lakshminarayana, R. Metabolomics of carotenids: The challenges and prospects—A review. Trends Food Sci. Technol. 2015, 45, 105–117. [Google Scholar] [CrossRef] [Green Version]
  69. Storebakken, T.; No, H.K. Pigmentation of rainbow trout. Aquaculture 1992, 100, 209–229. [Google Scholar] [CrossRef]
  70. Meyers, S.P. The biological/nutritional role of astaxanthin in salmonids and other aquatic species. In Proceedings of the 1st International Symposium of Natural Colorants for Foods, Nutraceuticals, Beverages and Confectionery, Amherst, MA, USA, 7–10 November 1993; pp. 7–10. [Google Scholar]
  71. Hertrampf, J.W.; Piedad-Pascual, F. Feed carotenoids. In Handbook on Ingredients for Aquaculture Feeds; Kluwer Academic Publishers: Dordrecht, The Netherlands, 2000; pp. 142–156. [Google Scholar]
  72. Baker, R.; Gunther, C. The role of carotenoids in consumer choice and the likely benefits from their inclusion into products for human consumption. Trends Food Sci. Technol. 2004, 15, 484–488. [Google Scholar] [CrossRef]
  73. Faehnrich, B.; Lukas, B.; Humer, E.; Zebeli, Q. Phytogenic pigments in animal nutrition: Potentials and risks. J. Sci. Food Agric. 2016, 96, 1420–1430. [Google Scholar] [CrossRef]
  74. Sahin, K.; Yazlak, H.; Orhan, C.; Tuzcu, M.; Akdemir, F.; Sahin, N. The effect of lycopene on antioxidant status in rainbow trout (Oncorhynchus mykiss) reared under high stocking density. Aquaculture 2014, 418, 132–138. [Google Scholar] [CrossRef]
  75. Lorenz, R.T.; Cysewski, T. Commercial potential for Haematococcus microalgae as a natural source of astaxanthin. Trends Biotechnol. 2000, 18, 160–166. [Google Scholar] [CrossRef]
  76. Lee, C.R.; Pham, M.A.; Lee, S.M. Effect of dietary paprika and lipid levels on growth and skin pigmentation of pale chub (Zacco platypus). Asian Aust. J. Anim. Sci. 2010, 23, 724–732. [Google Scholar] [CrossRef]
  77. Hertrampf, J.W.; Piedad-Pascual, F. Feed yeast. In Handbook on Ingredients for Aquaculture Feeds; Kluwer Academic Publishers: Dordrecht, The Netherlands, 2000; pp. 157–175. [Google Scholar]
  78. Guedes, A.C.; Amaro, H.M.; Malcata, F.X. Microalgae as sources of carotenoids. Mar. Drugs 2011, 9, 625–644. [Google Scholar] [CrossRef] [PubMed]
  79. Buyukcapar, H.M.; Yanar, M.; Yanar, Y. Pigmentation of rainbow trout (Oncorhynchus mykiss) with carotenoids from Marigold flower (Tagetes erecta) and red paper (Capsicum annum). Turk. J. Vet. Anim. Sci. 2007, 31, 7–12. [Google Scholar]
  80. Ambati, R.R.; Phang, S.M.; 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]
  81. Rodriguez-Amaya, D.B. Natural food colorants: Current knowledge and research trends. FFI J. 2020, 225, 111–130. [Google Scholar]
  82. Pereira da Costa, D.; Campos Miranda-Filho, K. The use of carotenoid pigments as food additives for aquatic organisms and their functional roles. Rev. Aquaculture 2019, 12, 1567–1578. [Google Scholar] [CrossRef]
  83. Widjaja-Adhi, M.A.K.; Golczak, M. The molecular aspects of absorption and metabolism of carotenoids and retinoids in vertebrates. Biochim. Biophys. Acta Mol. Cell. Biol. Lipids 2020, 1865, 158571. [Google Scholar] [CrossRef]
  84. Takaichi, S. Separation and analytical methods. In Carotenoids-Biological Functions and Diversity; Takaichi, S., Ed.; Shokabo: Tokyo, Japan, 2006; pp. 157–199. [Google Scholar]
  85. Nakano, T.; Shirakawa, H.; Yeo, G.; Devlin, R.H.; Soga, T. Metabolome profiling of growth hormone transgenic coho salmon by capillary electrophoresis time-of-flight mass spectrometry. In Oceanography Challenges to Future Earth; Komatsu, T., Ceccaldi, H.J., Yoshida, J., Prouzet, P., Henocque, Y., Eds.; Springer Nature: Cham, Switzerland, 2019; pp. 223–234. [Google Scholar]
  86. Hirayama, A.; Kami, K.; Sugimoto, M.; Sugawara, M.; Toki, N.; Onozuka, H.; Kinoshita, T.; Saito, N.; Ochiai, A.; Tomita, M.; et al. Quantitative metabolome profiling of colon and stomach cancer microenvironment by capillary electrophoresis time-of-flight mass spectrometry. Cancer Res. 2009, 69, 4918–4925. [Google Scholar] [CrossRef] [Green Version]
  87. Miki, W. Carotenoids. In Micronutrients and Health of Cultured Fish; Nakagawa, H., Sato, M., Eds.; Koseisha Koseikaku: Tokyo, Japan, 2003; pp. 42–53. [Google Scholar]
  88. Karnaukhov, V.N. Carotenoids: Recent progress, problems and prospects. Comp. Biochem. Physiol. 1990, 95B, 1–20. [Google Scholar] [CrossRef]
  89. Ahmadi, M.R.; Bazyar, A.A.; Safi, S.; Ytrestoyl, T.; Bjerkeng, B. Effects of dietary astaxanthin supplementation on reproductive characteristics of rainbow trout (Oncorhynchus mykiss). J. Appl. Ichthyol. 2006, 22, 388–394. [Google Scholar] [CrossRef]
  90. Maita, M.; Kiron, V. Nutrition and health in fish. In Nutrition and Feeding in Fish and Crustaceans; Watanabe, T., Ed.; Koseisha Koseikaku: Tokyo, Japan, 2009; pp. 251–269. [Google Scholar]
  91. Rossi, L.T.; Sharpen, A.R.; Zimmermann, J.A.; Olivero, C.R.; Zbrun, M.V.; Frizzo, L.S.; Signorini, M.L.; Bacchetta, C.; Cian, R.E.; Cazenave, J.; et al. Intestinal microbiota modulation in juvenile Pacú (Piaractus mesopotamicus) by supplementation with Pyropia columbina and β-carotene. Aquacult. Int. 2020, 28, 1001–1016. [Google Scholar] [CrossRef]
  92. Von Lintig, J.; Moon, J.; Lee, J.; Ramkumar, S. Carotenoid metabolism at the intestinal barrier. Biochim. Biophys. Acta Mol. Cell. Biol. Lipids 2020, 1865. [Google Scholar] [CrossRef] [PubMed]
  93. Oroian, M.; Escriche, I. Antioxidants: Characterization, natural sources, extraction and analysis. Food Res. Int. 2015, 74, 10–36. [Google Scholar] [CrossRef] [PubMed]
  94. Pisoschi, A.M.; Pop, A. The role of antioxidants in the chemistry of oxidative stress: A review. Eur. J. Med. Chem. 2015, 97, 55–74. [Google Scholar] [CrossRef] [PubMed]
  95. Secombes, C.J. The nonspecific immune system: Cellular defenses. In The Fish Immune System; Iwama, G.K., Nakanishi, T., Eds.; Academic Press: San Diego, CA, USA, 1996; pp. 63–103. [Google Scholar]
  96. Virág, L.; Jaén, R.I.; Regdon, Z.; Boscá, L.; Prieto, P. Self-defense of macrophages against oxidative injury: Fighting for their own survival. Redox Biol. 2019, 26, 101261. [Google Scholar] [CrossRef]
  97. Taniguchi, N.; Endo, T. Cross talk of SOD, NO, and glutathione metabolism. In Biochemistry in Oxidative Stress and Redox; Taniguchi, N., Yodoi, J., Eds.; Kyoritsu Shuppan: Tokyo, Japan, 2000; pp. 1–11. [Google Scholar]
  98. Sies, H.; Berndt, C.; Jones, D.P. Oxidative Stress. Annu. Rev. Biochem. 2017, 86, 715–748. [Google Scholar] [CrossRef]
  99. Forlenza, M.; Scharsack, J.P.; Kachamakova, N.M.; Taverne-Thiele, A.J.; Rombout, J.H.W.M.; Wiegertjes, G.F. Differential contribution of neutrophilic granulocytes and macrophages to nitrosative stress in a host-parasite animal model. Moll. Immunol. 2008, 45, 3178–3189. [Google Scholar] [CrossRef] [PubMed]
  100. Dixon, B.; Barreda, D.R.; Sunyer, J.O. Perspective on the development and validation of Ab reagents to fish immune proteins for the correct assessment of immune function. Front. Immunol. 2018, 9, 2957. [Google Scholar] [CrossRef]
  101. Zou, J.; Secombes, C.J. The function of fish cytokines. Biology 2016, 5, 23. [Google Scholar] [CrossRef]
  102. Abd El-Gawad, E.A.; Wang, H.-P.; Yao, H. Diet supplemented with synthetic carotenoids: Effects on growth performance and biochemical and immunological parameters of yellow perch (Perca flavescens). Front. Physiol. 2019, 10, 1056. [Google Scholar] [CrossRef] [PubMed]
  103. Li, M.Y.; Sun, L.; Niu, X.T.; Chen, X.M.; Tian, J.X.; Kong, Y.D.; Wang, G.Q. Astaxanthin protects lipopolysaccharide-induced inflammatory response in Channa argus through inhibiting NF-kappa B and MAPKs signaling pathways. Fish Shellfish Immunol. 2019, 86, 280–286. [Google Scholar] [CrossRef] [PubMed]
  104. Petit, J.; David, L.; Dirks, R.; Wiegertjes, G.F. Genomic and transcriptomic approaches to study immunology in cyprinids: What is next? Dev. Comp. Immunol. 2017, 75, 48–62. [Google Scholar] [CrossRef] [PubMed]
  105. Liu, F.; Qu, Y.K.; Wang, A.M.; Yu, Y.B.; Yang, W.P.; Lv, F.; Nie, Q. Effects of carotenoids on the growth performance, biochemical parameters, immune responses and disease resistance of yellow catfish (Pelteobagrus fulvidraco) under high-temperature stress. Aquaculture 2019, 503, 293–303. [Google Scholar] [CrossRef]
  106. Lim, K.C.; Yusoff, F.M.; Shariff, M.; Kamarudin, M.S.; Nagao, N. Dietary supplementation of astaxanthin enhances hemato-biochemistry and innate immunity of Asian seabass Lates calcarifer (Bloch, 1790). Aquaculture 2019, 512, 734339. [Google Scholar] [CrossRef]
  107. Cheng, C.H.; Guo, Z.X.; Ye, C.X.; Wang, A.L. Effect of dietary astaxanthin on the growth performance, non-specific immunity, and antioxidant capacity of pufferfish (Takifugu obscurus) under high temperature stress. Fish Physiol. Biochem. 2018, 44, 209–218. [Google Scholar] [CrossRef]
  108. Anbazahan, S.M.; Mari, L.S.; Yogeshwari, G.; Jagruthi, C.; Thirumurugan, R.; Arockiaraj, J.; Velanganni, A.A.; Krishnamoorthy, P.; Balasundaram, C.; Harikrishnan, R. Immune response and disease resistance of carotenoids supplementation diet in Cyprinus carpio against Aeromonas hydrophila. Fish Shellfish Immunol. 2014, 40, 9–13. [Google Scholar] [CrossRef]
  109. Abdollahi, Y.; Ahmadifard, N.; Agh, N.; Rahmanifarah, K.; Hejazi, M.A. β-Carotene-enriched Artemia as a natural carotenoid improved skin pigmentation and enhanced the mucus immune responses of platyfish Xiphophorus maculatus. Aquacult. Int. 2019, 27, 1847–1858. [Google Scholar] [CrossRef]
  110. Alishahi, M.; Karamifar, M.; Mesbah, M. Effects of astaxanthin and Dunaliella salina on skin carotenoids, growth performance and immune response of Astronotus ocellatus. Aquacult. Int. 2015, 23, 1239–1248. [Google Scholar] [CrossRef]
  111. Villena, J.; Alvarez, S.; Kitazawa, H. Introduction. In Probiotics: Immunobiotics and immunogenics; Kitazawa, H., Villena, J., Alvarez, S., Eds.; CRC Press: Boca Raton, FL, USA, 2014; pp. 1–11. [Google Scholar]
  112. Katsuyama, M. Carotenoids as vitamin A precursors. In Carotenoids in Marine Organisms; Miki, W., Ed.; Koseisha Koseikaku: Tokyo, Japan, 1993; pp. 23–34. [Google Scholar]
  113. Iwama, G.K.; Afonso, L.O.B.; Vijayan, M.M. Stress in fishes. In The Physiology of Fishes, 3rd ed.; Evans, D.H., Claiborne, J.B., Eds.; CRC Press: Boca Raton, FL, USA, 2006; pp. 319–342. [Google Scholar]
  114. Nakano, T.; Afonso, L.O.; Beckman, B.R.; Iwama, G.K.; Devlin, R.H. Acute physiological stress down-regulates mRNA expressions of growth-related genes in coho salmon. PLoS ONE 2013, 8, e71421. [Google Scholar] [CrossRef]
  115. Basu, N.; Nakano, T.; Grau, E.G.; Iwama, G.K. The effects of cortisol on heat shock protein 70 levels in two fish species. Gen. Comp. Endocrinol. 2001, 124, 97–105. [Google Scholar] [CrossRef] [PubMed]
  116. Barton, B.A.; Iwama, G.K. Physiological change in fish from stress in aquaculture with emphasis on the response and effects of corticosteroids. Annu. Rev. Fish Dis. 1991, 1, 3–26. [Google Scholar] [CrossRef]
  117. Wu, H.; Aoki, A.; Arimoto, T.; Nakano, T.; Ohnuki, H.; Murata, M.; Ren, H.; Endo, H. Fish stress become visible: A new attempt to use biosensor for real-time monitoring fish stress. Biosens. Bioelectron. 2015, 67, 503–510. [Google Scholar] [CrossRef] [PubMed]
  118. Nakano, T.; Kameda, M.; Shoji, Y.; Hayashi, S.; Yamaguchi, T.; Sato, M. Effect of severe environmental thermal stress on redox state in salmon. Redox Biol. 2014, 2, 772–776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Wu, H.; Fujii, Y.; Nakano, T.; Arimoto, T.; Murata, M.; Matsumoto, H.; Yoshiura, Y.; Ohnuki, H.; Endo, H. Development of a novel enhanced biosensor system for real-time monitoring of fish stress using a self-assembled monolayer. Biosensors 2019, 19, 1585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Nakano, T.; Wu, H.; Arimoto, T.; Endo, H.; Yamaguchi, T.; Ochiai, Y. Some artificially applied stressors have a positive effect on fish fitness. In Proceedings of the JSFS 85th Anniversary-Commemorative International Symposium “Fisheries Science for Future Generations”, Tokyo, Japan, 22–24 September 2017; Available online: http://www.jsfs.jp/office/annual_meeting/meeting-program/85th/proceeding/pdfs/06010.pdf (accessed on 15 August 2020).
  121. Livingstone, D.R. Contaminant-stimulated reactive oxygen species production and oxidative damage in aquatic organisms. Mar. Pollut. Bull. 2001, 42, 656–666. [Google Scholar] [CrossRef]
  122. Valavanidis, A.; Vlahogianni, T.; Dassenakis, M.; Scoullos, M. Molecular biomarkers of oxidative stress in aquatic organisms in relation to toxic environmental pollutants. Ecotoxicol. Environ. Saf. 2006, 64, 178–189. [Google Scholar] [CrossRef]
  123. Martinez-Alvarez, R.M.; Morales, A.E.; Sanz, A. Antioxidant defenses in fish: Biotic and abiotic factors. Rev. Fish Biol. Fish. 2005, 15, 75–88. [Google Scholar] [CrossRef]
  124. Lushchak, V.I. Environmentally induced oxidative stress in aquatic animals. Aquat. Toxicol. 2011, 101, 13–30. [Google Scholar] [CrossRef]
  125. Lesser, M.P. Oxidative stress in marine environments: Biochemistry and physiological ecology. Annu. Rev. Physiol. 2006, 68, 253–278. [Google Scholar] [CrossRef] [Green Version]
  126. Nakano, T.; Sato, M.; Takeuchi, M. Superoxide dismutase activity in the skin of fish. J. Fish Biol. 1993, 43, 492–496. [Google Scholar] [CrossRef]
  127. Nakano, T.; Sato, M.; Takeuchi, M. Partial purification and properties of glutathione peroxidase from carp hepatopancreas. Comp. Biochem. Physiol. 1992, 102B, 31–35. [Google Scholar] [CrossRef]
  128. Nakano, T.; Sato, M.; Takeuchi, M. Glutathione peroxidase of fish. J. Food Sci. 1992, 57, 1116–1119. [Google Scholar] [CrossRef]
  129. Arrigo, A.P. Gene expression and the thiol redox state. Free Radic. Biol. Med. 1999, 27, 936–944. [Google Scholar] [CrossRef]
  130. Sies, H. Glutathione and its role in cellular functions. Free Radic. Biol. Med. 1999, 27, 916–921. [Google Scholar] [CrossRef]
  131. Iwama, G.K.; Thomas, P.T.; Forsyth, R.B.; Vijayan, M.M. Heat shock protein expression in fish. Rev. Fish Biol. Fish. 1998, 8, 35–56. [Google Scholar] [CrossRef]
  132. Hochachka, P.W.; Somero, G.N. Temperature. In Biochemical Adaptation; Hochachka, P.W., Somero, G.N., Eds.; Oxford University Press: Oxford, UK, 2002; pp. 290–449. [Google Scholar]
  133. Basu, N.; Todgham, A.E.; Ackerman, P.A.; Bibeau, M.R.; Nakano, K.; Shulte, P.M.; Iwama, G.K. Heat shock protein genes and their functional significance in fish. Gene 2002, 295, 173–183. [Google Scholar] [CrossRef]
  134. Roberts, R.J.; Agius, C.; Saliba, C.; Bossier, P.; Sung, Y.Y. Heat shock proteins (chaperones) in fish and shellfish and their potential role in relation to fish health: A review. J. Fish Dis. 2010, 33, 789–801. [Google Scholar] [CrossRef]
  135. Feder, M.E.; Hofmann, G.E. Heat-shock proteins, molecular chaperones, and the stress response. Annu. Rev. Physiol. 1999, 61, 243–282. [Google Scholar] [CrossRef] [Green Version]
  136. Deane, E.E.; Woo, N.Y.S. Advances and perspectives on the regulation and expression of piscine heat shock proteins. Rev. Fish Biol. Fish. 2011, 21, 153–185. [Google Scholar] [CrossRef]
  137. Regoli, F.; Giuliani, M.E. Oxidative pathways of chemical toxicity and oxidative stress biomarkers in marine organisms. Mar. Environ. Res. 2014, 93, 106–117. [Google Scholar] [CrossRef] [PubMed]
  138. Srikanth, K.; Pereira, E.; Duarte, A.C.; Ahmad, I. Glutathione and its dependent enzymes’ modulatory responses to toxic metals and metalloids in fish-a review. Environ. Sci. Pollut. Res. 2013, 20, 2133–2149. [Google Scholar] [CrossRef] [PubMed]
  139. Ho, E.; Galougahi, K.K.; Liu, C.C.; Bhindi, R.; Figtree, G.A. Biological markers of oxidative stress: Applications to cardiovascular research and practice. Redox Biol. 2013, 1, 483–491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Ameur, W.B.; De Lapuente, J.; Megdiche, Y.E.; Barhoumi, B.; Trabelsi, S.; Camps, L.; Serret, J.; Ramos-Lopez, D.; Gonzalez-Linares, J.; Driss, M.R.; et al. Oxidative stress, genotoxicity and histopathology biomarker responses in mullet (Mugil cephalus) and sea bass (Dicentrarchus labrax) liver from Bizerte Lagoon (Tunisia). Mar. Pollut. Bull. 2012, 64, 241–251. [Google Scholar] [CrossRef] [PubMed]
  141. Heise, K.; Puntarulo, S.; Portner, H.O.; Abele, D. Production of reactive oxygen species by isolated mitochondria of the Antarctic bivalve Laternula elliptica (King and Broderip) under heat stress. Comp. Biochem. Physiol. 2003, 134C, 79–90. [Google Scholar] [CrossRef]
  142. Dawood, M.A.O.; Abdel-Tawwab, M.; Abdel-Latif, H.M.R. Lycopene reduces the impacts of aquatic environmental pollutants and physical stressors in fish. Rev. Aquac. 2020, 12, 2511–2526. [Google Scholar] [CrossRef]
  143. Pan, C.H.; Chien, Y.H.; Wang, Y.J. Antioxidant defence to ammonia stress of characins (Hyphessobrycon eques Steindachner) fed diets supplemented with carotenoids. Aquac. Nutr. 2011, 17, 258–266. [Google Scholar] [CrossRef]
  144. Hata, Y. Lipid hydroperoxide and disease. In Lipid Peroxides and Nutrition; Igarashi, O., Kaneda, T., Fukuba, H., Mino, M., Eds.; Koseikan: Tokyo, Japan, 1986; pp. 169–214. [Google Scholar]
  145. Ando, S. Carotenoid transport in fishes. In Carotenoids in Marine Organisms; Miki, W., Ed.; Koseisha Koseikaku: Tokyo, Japan, 1993; pp. 49–58. [Google Scholar]
  146. Tan, K.; Zhang, H.; Lim, L.-S.; Ma, H.; Li, S.; Zheng, H. Roles of carotenoids in invertebrate immunology. Front. Immunol. 2020, 10. [Google Scholar] [CrossRef]
  147. Al-Amin, M.M.; Akhter, S.; Hasan, A.T.; Alam, T.; Nageeb Hasan, S.M.; Saifullah, A.R.; Shohel, M. The antioxidant effect of astaxanthin is higher in young mice than aged: A region specific study on brain. Metab. Brain Dis. 2015, 30, 1237–1246. [Google Scholar] [CrossRef]
  148. Müller, L.; Catalano, A.; Simone, R.; Cittadini, A.; Fröhlich, K.; Böhm, V.; Palozza, P. Antioxidant capacity of tomato seed oil in solution and its redox properties in cultured macrophages. J. Agric. Food Chem. 2013, 61, 346–354. [Google Scholar] [CrossRef]
  149. Fleischmann, C.; Bar-Ilan, N.; Horowitz, M.; Bruchim, Y.; Deuster, P.; Heled, Y. Astaxanthin supplementation impacts the cellular HSP expression profile during passive heating. Cell Stress Chaperones 2020, 25, 549–558. [Google Scholar] [CrossRef] [PubMed]
  150. Miki, W. Biological functions and activities of animal carotenoids. Pure Appl. Chem. 1991, 63, 141–146. [Google Scholar] [CrossRef]
  151. Skibsted, L.H. Carotenoids in antioxidant networks, colorants or radical scavengers. J. Agric. Food Chem. 2012, 60, 2409–2417. [Google Scholar] [CrossRef] [PubMed]
  152. Bast, A.; Haenen, G.R.; van den Berg, R.; van den Berg, H. Antioxidant effects of carotenoids. Int. J. Vitam. Nutr. Res. 1998, 68, 399–403. [Google Scholar] [PubMed]
  153. Kaulmann, A.; Bohn, T. Carotenoids, inflammation, and oxidative stress-implications of cellular signaling pathways and relation to chronic disease prevention. Nutr. Res. 2014, 34, 907–929. [Google Scholar] [CrossRef] [PubMed]
  154. Niu, T.; Xuan, R.; Jiang, L.; Wu, W.; Zhen, Z.; Song, Y.; Hong, L.; Zheng, K.; Zhang, J.; Xu, Q.; et al. Astaxanthin induces the Nrf2/HO-1 antioxidant pathway in human umbilical vein endothelial cells by generating trace amounts of ROS. J. Agric. Food Chem. 2018, 66, 1551–1559. [Google Scholar] [CrossRef]
  155. Young, A.J.; Lowe, G.M. Antioxidant and prooxidant properties of carotenoids. Arch. Biochem. Biophys. 2001, 385, 20–27. [Google Scholar] [CrossRef]
  156. Ribeiro, D.; Freitas, M.; Silva, A.M.S.; Carvalho, F.; Fernandes, E. Antioxidant and pro-oxidant activities of carotenoids and their oxidation products. Food Chem. Toxicol. 2018, 120, 681–699. [Google Scholar] [CrossRef]
  157. Jomova, K.; Valko, M. Health protective effects of carotenoids and their interactions with other biological antioxidants. Eur. J. Med. Chem. 2013, 70, 102–110. [Google Scholar] [CrossRef]
  158. Droge, W. Free radicals in the physiological control of cell function. Physiol. Rev. 2002, 82, 47–95. [Google Scholar] [CrossRef]
  159. Allen, R.G.; Tresini, M. Oxidative stress and gene regulation. Free Radic. Biol. Med. 2000, 28, 463–499. [Google Scholar] [CrossRef]
  160. Burri, B.J. Carotenoids and gene expression. Nutrition 2000, 16, 577–578. [Google Scholar] [CrossRef]
  161. Suzuki, Y.J.; Forman, H.J.; Sevanian, A. Oxidants as stimulators of signal transduction. Free Radic. Biol. Med. 1997, 22, 269–285. [Google Scholar] [CrossRef]
  162. Schwarz, K.B. Oxidative stress during viral infection: A review. Free Radic. Biol. Med. 1996, 21, 641–649. [Google Scholar] [CrossRef]
  163. Osatomi, K.; Kanai, K.; Hara, K.; Ishihara, T. Changes in Cu, Zn-SOD activity in Japanese flounder Paralichtys olivaceus with bacterial infection. Nippon Suisan Gakkaishi 2002, 68, 207–213. [Google Scholar] [CrossRef] [Green Version]
  164. Maehara, K.; Hasegawa, T.; Xiao, H.; Takeuchi, A.; Abe, R.; Isobe, K. Cooperative interaction of NF-kB and C/EBP binding sites is necessary for manganese superoxide dismutase gene transcription mediated by lipopolysaccharide and interferon-γ. FEBS Lett. 1999, 449, 115–119. [Google Scholar] [CrossRef] [Green Version]
  165. Brady, T.C.; Chang, L.Y.; Day, B.J.; Crapo, J.D. Extracellular superoxide dismutase is upregulated with inducible nitric oxide synthase after NF-κB activation. Am. J. Physiol. Lung Cell Mol. Physiol. 1997, 273, L1002–L1006. [Google Scholar] [CrossRef]
  166. Qadri, I.; Iwahashi, M.; Capasso, J.M.; Hopken, M.W.; Flores, S.; Schaack, J.; Simon, F.R. Induced oxidative stress and activated expression of manganese superoxide dismutase during hepatitis C virus replication: Role of JNK, p38 MAPK and AP-1. Biochem. J. 2004, 378, 919–928. [Google Scholar] [CrossRef]
  167. Wang, J.Y.; Lee, Y.J.; Chou, M.C.; Chang, R.; Chiu, C.H.; Liang, Y.J.; Wu, L.S. Astaxanthin protects steroidogenesis from hydrogen peroxide-induced oxidative stress in mouse Leydig cells. Mar. Drugs 2015, 13, 1375–1388. [Google Scholar] [CrossRef] [Green Version]
  168. Stentiford, G.D.; Bateman, I.J.; Hinchliffe, S.J.; Bass, D.; Hartnell, R.; Santos, E.M.; Devlin, M.J.; Feist, S.W.; Taylor, N.G.H.; Verner-Jeffreys, D.W.; et al. Sustainable aquaculture through the one health lens. Nature Food 2020, 1, 468–474. [Google Scholar] [CrossRef]
  169. Naylor, R.L.; Hardy, R.W.; Bureau, D.P.; Chiu, A.; Elliott, M.; Farrell, A.P.; Forster, I.; Gatlin, D.M.; Goldburg, R.J.; Hua, K.; et al. Feeding aquaculture in an era of finite resources. Proc. Natl. Acad. Sci. USA 2009, 106, 15103–15110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Naylor, R.L.; Goldburg, R.J.; Primavera, J.H.; Kautsky, N.; Beveridge, M.C.M.; Clay, J.; Folke, C.; Lubchenco, J.; Mooney, H.; Troell, M. Effect of aquaculture on world fish supplies. Nature 2000, 405, 1017–1024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Costello, C.; Cao, L.; Gelcich, S.; Cisneros-Mata, M.A.; Free, C.M.; Froehlich, H.E.; Golden, C.D.; Ishimura, G.; Maier, J.; Macadam-Somer, I.; et al. The future of food from the sea. Nature 2020. [Google Scholar] [CrossRef] [PubMed]
  172. FAO. FAO-Yearbook of Fishery and Aquaculture tatistics 2017; Food and Agriculture Organization of the United Nations: Rome, Italy, 2019; pp. 81–84. Available online: http://www.fao.org/3/ca5495t/CA5495T.pdf (accessed on 15 August 2020).
  173. DBJ. Monthly Topics on Global Fish Farming; Development Bank of Japan: Tokyo, Japan, 2014; pp. 7–32. [Google Scholar]
  174. Nakano, T.; Shoji, Y.; Shirakawa, H.; Suda, Y.; Yamaguchi, T.; Sato, M.; Devlin, R.H. Daily expression patterns of growth-related genes in growth hormone transgenic coho salmon, Oncorhynchus kisutch. La Mer 2011, 49, 111–117. [Google Scholar]
  175. Nakano, T. Rising to the challenge of reconstructing the coastal fisheries environment following the massive tsunami in Japan: The national 10-year “Tohoku Ecosystem-Associated Marine Sciences (TEAMS)”. In Marine Productivity: Perturbations and Resilience of Socio-Ecosystems; Ceccaldi, H.J., Henocque, Y., Koike, Y., Komatsu, T., Stora, G., Tusseau-Vuillemin, M.H., Eds.; Springer International Publishin: Cham, Switzerland, 2015; pp. 63–68. [Google Scholar]
  176. Ushio, H.; Kitazawa, D.; Mizuno, H. An innovation of salmonid aquaculture for reconstruction following the Great East Japan Earthquake. In eReconstruction and Renovation Efforts Following the Great East Japan Earthquake by Development of New Technologies; Takuchi, T., Sato, M., Watabe, S., Eds.; Koseisha Koseikaku: Tokyo, Japan, 2017; pp. 43–52. [Google Scholar]
  177. Edge, R.; Truscott, T.G. Prooxidant and antioxidant reaction mechanisms of carotene and radical interactions with vitamins E and C. Nutrition 1997, 13, 992–994. [Google Scholar] [CrossRef]
  178. Burton, G.W.; Ingold, K.U. β-Carotene: An unusual type of lipid antioxidant. Science 1984, 224, 569–573. [Google Scholar] [CrossRef]
  179. Nakano, T.; Shiba, Y.; Yamaguchi, T.; Sato, M.; Ohshima, H. Natural marine products inhibit formation of 8-chloroguanosine mediated by hypochlorous acid, myeloperoxidase and activated neutrophils. Free Radic. Biol. Med. 2012, 53, S95. [Google Scholar] [CrossRef]
  180. Suzuki, T.; Nakano, T.; Masuda, M.; Ohshima, H. Epigallocatechin gallate markedly enhances formation of 8-oxo-7,8-dihydro-2’-deoxyguanosine with hypochlorous acid. Free Radic. Biol. Med. 2004, 36, 1087–1093. [Google Scholar] [CrossRef]
  181. Ohshima, H.; Yoshie, Y.; Auriol, S.; Gilibert, I. Antioxidant and pro-oxidant actions of flavonoids: Effects on DNA damage induced by nitric oxide, peroxynitrite and nitroxyl anion. Free Radic. Biol. Med. 1998, 25, 1057–1065. [Google Scholar] [CrossRef]
  182. Masuda, M.; Suzuki, T.; Friesen, M.D.; Ravanat, J.L.; Cadet, J.; Pignatelli, B.; Nishino, H.; Ohshima, H. Chlorination of guanosine and other nucleosides by hypochlorous acid and myeloperoxidase of activated human neutrophils: Catalysis by nicotine and trimethylamine. J. Biol. Chem. 2001, 276, 40486–40496. [Google Scholar] [CrossRef] [Green Version]
  183. Nakano, T.; Masuda, M.; Suzuki, T.; Ohshima, H. Inhibition by polyphenolic phytochemicals and sulfurous compounds of the formation of 8-chloroguanosine mediated by hypochlorous acid, human myeloperoxidase, and activated human neutrophils. Biosci. Biotechnol. Biochem. 2012, 76, 2208–2213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Structure of specific carotenoids in fish. (A) β-Carotene, (B) canthaxanthin, (C) astaxanthin. Carotenoids are classified into carotenes (e.g., β-carotene), which are composed of carbon and hydrogen, and xanthophylls (e.g., canthaxanthin and astaxanthin), which also contain oxygen.
Figure 1. Structure of specific carotenoids in fish. (A) β-Carotene, (B) canthaxanthin, (C) astaxanthin. Carotenoids are classified into carotenes (e.g., β-carotene), which are composed of carbon and hydrogen, and xanthophylls (e.g., canthaxanthin and astaxanthin), which also contain oxygen.
Marinedrugs 18 00568 g001
Figure 2. Picture of Haematococcus sp. Haematococcus produces and accumulates high levels of astaxanthin under conditions of intense sunlight. Credit: Dr. G. Nishitani.
Figure 2. Picture of Haematococcus sp. Haematococcus produces and accumulates high levels of astaxanthin under conditions of intense sunlight. Credit: Dr. G. Nishitani.
Marinedrugs 18 00568 g002
Table 1. Sources of carotenoids in farmed fish.
Table 1. Sources of carotenoids in farmed fish.
Source
Synthetic astaxanthin (CAROPHYLL Pink, DSM Nutritional Products Ltd.)
Synthetic canthaxanthin (CAROPHYLL Red, DSM Nutritional Products Ltd.)
Red yeast Xanthophyllomyces dendrnrhous (formerly designated Phaffia rhodozyma)
Green microalgae Haematococcus pluvialis
Bacteria Paracoccus carotinifaciens
Krill Euphausia superba
Alga Spirulina spp.
Crustacean exoskeleton and meal
Red pepper paprika Capsicum annuum L
Marigold
Table 2. General biological functions of carotenoids in fish.
Table 2. General biological functions of carotenoids in fish.
Biological Function
Antioxidant activity
Anti-stress
Anti-inflammatory
Egg quality
Growth performance
Immune system
Lipid metabolism
Liver function
Muscle pigmentation
Photoprotection
Provitamin A activity
Reproduction
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Nakano, T.; Wiegertjes, G. Properties of Carotenoids in Fish Fitness: A Review. Mar. Drugs 2020, 18, 568. https://doi.org/10.3390/md18110568

AMA Style

Nakano T, Wiegertjes G. Properties of Carotenoids in Fish Fitness: A Review. Marine Drugs. 2020; 18(11):568. https://doi.org/10.3390/md18110568

Chicago/Turabian Style

Nakano, Toshiki, and Geert Wiegertjes. 2020. "Properties of Carotenoids in Fish Fitness: A Review" Marine Drugs 18, no. 11: 568. https://doi.org/10.3390/md18110568

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