Next Article in Journal
Nutritional Intake in Hospitalized Patients Receiving Texture-Modified Diets: Preliminary Results from an Observational Study
Previous Article in Journal
Cooking Skills in Health Professionals: A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Interactions of Astaxanthin and Omega-3 Fat in Health and Disease

Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, 1955 East West Road, Honolulu, HI 96822, USA
Dietetics 2025, 4(3), 39; https://doi.org/10.3390/dietetics4030039
Submission received: 8 July 2025 / Revised: 6 August 2025 / Accepted: 29 August 2025 / Published: 8 September 2025

Abstract

Elevated oxidative stress and chronic inflammation are major pathological factors underlying numerous diseases, including cardiovascular and neurodegenerative diseases. Astaxanthin, an antioxidant carotenoid, and very long-chain omega-3 polyunsaturated fatty acids, such as eicosapentaenoic acid and docosahexaenoic acid, are commonly found in seafood and exhibit beneficial effects on a myriad of diseases. The two powerful nutrients often work together, and this interaction is particularly beneficial for various aspects of health, primarily due to their complementary antioxidant and anti-inflammatory properties. The current understanding of the molecular and cellular mechanisms by which powerful duos exhibit protective effects, and their potential interactive effects on cardiometabolic and neurodegenerative diseases, is discussed.

1. Introduction

Diet and dietary factors are key modifiable factors that influence health and disease risks. Bioactive food components, including carotenoids, polyphenols, phytosterols, fiber, and very long-chain omega-3 polyunsaturated fatty acids (omega-3 PUFAs), are known to provide beneficial effects [1]. Astaxanthin (Ast), an oxygenated carotenoid, and omega-3 PUFAs are frequently found in several seafoods, and the intake of both is known to be protective against various diseases [2,3,4]. This literature review examines current understanding of how Ast and omega-3 PUFAs act together to reduce oxidative stress and inflammatory responses through complementary mechanisms, providing beneficial effects on health and disease, particularly in cardiometabolic and neurodegenerative conditions. The distinctive roles of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), the two most common omega-3 PUFAs contained in seafood, are also discussed.

2. Methods

This narrative review examined potential interactive effects of Ast and omega-3 PUFAs on health and diseases. The relevant literature was searched on PubMed, ResearchGate, and Web of Science using search terms that included Ast, omega-3 fat, oxidative stress, and inflammation. Health benefits of Ast and omega-3 PUFAs on cardiometabolic diseases were examined using terms such as hyperlipidemia, cardiovascular diseases, fatty liver, hepatic steatosis, insulin resistance, and type 2 diabetes. Their benefits on neurodegenerative conditions were searched using terms that included Alzheimer’s disease, Parkinson’s disease, and dementia. Effects of different omega-3 PUFAs were searched using terms that included EPA, docosapentaenoic acid (DPA), and DHA.

3. Food Sources of Ast and Omega-3 PUFAs

3.1. Dietary Sources of Ast

Ast is a xanthophyll carotenoid that is responsible for the characteristic reddish-orange color in certain marine organisms such as Antarctic krill, shrimp, crab, lobster, trout, and salmon. These organisms obtain their Ast by consuming specific types of microalgae, yeast, or smaller fish. Ast is also commonly used in aquaculture for pigmentation as well as for the health of aquatic animals [5]. Ast has a long polyene chain with two terminal beta-ionone rings with hydroxyl groups and keto groups at both ends. The hydroxyl groups can be esterified with various fatty acids (FAs), and Ast is present as free, mono-esters or di-esters. Additionally, various geometric and meso isomers of Ast are also present in foods and supplements, with different forms exhibiting distinctive stability, bioavailability, and functions [6].
Antarctic krill and shrimp contain 30–40 mg/kg Ast, primarily in the form of Ast-esters. The common esterified FAs are myristic acid (C14:0), palmitic acid (PA, C16:0), oleic acid (C18:1), EPA (C20:5), and DHA (C22:6) [7]. Salmon and trout contain mostly free Ast in their muscle tissues in the range of 6–38 mg/kg flesh. Wild-caught salmon contains higher amounts of Ast, with Sockeye salmon containing 26–38 mg/kg of flesh and Atlantic farmed salmon containing 6–8 mg/kg of flesh [4]. In contrast to wild-caught salmon mostly containing free forms of Ast, farm-raised salmon contains Ast-ester forms in addition to the free forms, most likely due to their diets [8]. Additionally, different isoforms of Ast are present between the wild-caught and farm-raised salmon [8]. Various dietary supplement products, containing 2–12 mg per capsule, are available. They are mostly derived from microalgae [Haematococcus pluvialis (H. pluvialis)], yeast (Phaffia rhodozyma), or chemical synthesis. H. pluvialis mostly contains Ast-esters, while free Ast is found in yeast and synthetic form [9]. It has been shown that Ast-esters, especially diester forms, are more stable than free Ast [6]. The Food and Drug Administration has placed Ast, H. pluvialis extract containing Ast-esters, on the GRAS (Generally Recognized As Safe) list.

3.2. Dietary Sources of Omega-3 PUFAs

FAs are not only energy substrates but also provide important functional roles, such as structural components and precursors for signaling molecules. FA synthesis is not a major pathway in adult humans consuming typical Western diets; FAs are mainly derived from dietary lipids, which are mostly present as triacylglycerols (TAGs) and a small fraction as phospholipids (PLs) [10]. FAs are classified based on the carbon chain length as well as the presence, number, and stereochemistry of double bonds. Humans need to consume two essential FAs: omega-6 linoleic acid (C18:2) and omega-3 alpha-linolenic acid (ALA, C18:3). These can be further elongated and desaturated into omega-6 arachidonic acid (ARA, C20:4) and omega-3 EPA (C20:5), DPA (C22:5), and DHA (C22:6), respectively. Genetic differences in elongases and desaturases influence the functionality and metabolism of PUFAs [11]. ALA conversion into EPA, DPA, and DHA in the human body is not efficient. Consequently, FA composition in cellular TAGs and PLs mostly reflects dietary FA intake, especially for the omega-3 PUFAs [10]. Depending on the types of FAs consumed, they have differential impacts on health such that a high intake of omega-3 PUFAs is beneficial, while saturated FAs cause harmful effects [12].
In general, cold-water, oily fish such as mackerel, herring, salmon, and sardines are the richest sources of omega-3 PUFAs. These fish, however, do not produce omega-3 PUFAs. Rather, they accumulate omega-3 PUFAs by consuming microalgae, phytoplankton, smaller fish, and crustaceans. Other sources of omega-3 PUFAs include seaweeds and crustaceans. Omega-3 PUFA content in fish varies mainly due to their diets; for example, wild-caught salmon contains a greater amount of EPA and DHA, while farm-raised salmon contains a higher amount of omega-6 PUFAs [4]. Cooked Atlantic salmon provides 1.57–1.83 g of EPA plus DHA per three ounces, while canned sardines contain approximately 1.19 g per three ounces. For more detailed information, please refer to the U.S. Department of Agriculture, FoodData Central Website.
Current intake of seafood, a primary source of omega-3 PUFAs, falls below recommended levels in most countries [13,14], and dietary supplements are available. While fish oil has been the main source of omega-3 PUFAs, the use of alternatives, such as krill oil and algae oil, has been on the rise [15,16]. Algae oil, notably, provides an excellent omega-3 PUFA source for vegetarians and vegans. These oils typically contain 20% to 40% of omega-3 PUFAs out of their total FAs [17,18]. Beyond supplements, a high-purity prescription form of an ethyl ester of EPA, icosapent ethyl, is available due to its well-documented health benefits [19].

3.3. Stability and Bioavailability of Ast and Omega-3 PUFAs

The conjugated double bonds in omega-3 PUFAs are susceptible to oxidation by light, temperature, oxygen, and other oxidants during processing, storage, and cooking, producing oxidized lipids that can be harmful [20]. While the polyene chain in Ast may also be oxidized, leading to its degradation, Ast is known to reduce the oxidation of omega-3 PUFAs in foods [21]. Although cooking generally increases their bioavailability, these two nutrients are sensitive to heat, oxygen, and light and can be degraded during high-temperature cooking for prolonged periods [8]. It has been reported that cooking 85 g of salmon to an internal temperature of 71 °C did not impact Ast levels in both wild-caught and farm-raised salmon [4]. However, Ast levels were lower in processed (canned/pouch) compared to unprocessed (fresh/frozen) forms of salmon. While baking and steaming are mostly recommended, frying fish and shellfish, especially with margarine, is strongly discouraged [22]. Among different forms of Ast, Ast-esters are known to be more stable than free forms [23]. Additionally, the Z-isomers, which are present in wild salmon, have been shown to be less stable than the all-E-isomer. However, the Z-isomers are known to have greater bioavailability than the all-E-isomer [24]. FAs, including omega-3 PUFAs, have higher bioavailability. It has been shown that omega-3 PUFAs in the PL form, as found in krill oil, may offer superior bioavailability compared to the TAG forms in fish oil [25].

4. Digestion, Absorption, and Transport of Ast and Omega-3 PUFAs

Ast is a lipid-soluble compound, and its digestion, absorption, and transport are closely related to lipids and lipoprotein metabolism. Dietary lipids enhance both digestion and absorption of Ast [26]. TAGs are digested by lipases and colipases into FAs and MAGs, and PLs by phospholipases and esterases into FAs and lysophospholipids (lyso-PLs) in the stomach and small intestine (Figure 1). Ast-esters are digested into FAs and Ast, potentially by esterases, which are then incorporated into the micelles. Bile salts assist the digestion and absorption of both nutrients by acting as emulsifiers and aiding micelle formation. Micelles bring lipid-soluble nutrients close to the brush border membrane across the unstirred water layer, enabling the subsequent absorption of the digestion products—FAs, MAGs, lyso-PLs, and Ast—into the enterocytes mostly via passive diffusion [27]. Several transporter proteins, such as cluster-determinant 36 and scavenger receptor class B type 1, have been suggested to mediate the absorption of FAs and Ast via the facilitative diffusion process [28,29]. FAs and MAGs are esterified into TAGs, and lyso-PLs are re-esterified with FAs to form PLs. TAGs, PLs, and other lipid-soluble nutrients, including fat-soluble vitamins and carotenoids, are then packaged into chylomicrons within the enterocytes. Ast is incorporated into chylomicrons, most likely in the PL monolayer enveloping the lipoprotein particles. Chylomicrons exit into the lymphatic system and are slowly released into the systemic blood through the thoracic duct, initially bypassing the liver.
Lipoprotein lipase (LPL) hydrolyzes TAGs in TAG-rich lipoproteins into FAs, MAGs, and glycerol, which are taken up by cells. LPL is mainly expressed in adipose tissues and skeletal muscle. However, adipose LPL activity is increased in the post-prandial condition; hence, the majority of dietary lipids are stored in the adipose tissues [30]. After LPL-mediated hydrolysis of TAGs, chylomicrons become chylomicron remnants, which are taken up by the liver. The liver packages lipid-soluble nutrients, including vitamin E and carotenoids, into very low-density lipoprotein (VLDL), an endogenous lipid transport mechanism. VLDL becomes low-density lipoprotein (LDL) after LPL-mediated TAG hydrolysis. It is estimated that 60% of FAs in hepatic TAG are derived from circulating FAs, with lesser contributions from de novo lipogenesis (25%) and diet (15%) in humans [31]. Adipose tissue is the major storage site of energy, where FAs and MAGs, derived from LPL-mediated hydrolysis of TAG in chylomicron and VLDL, are re-esterified into TAGs and stored in lipid droplets of adipocytes. When systemic energy demands increase during fasting or exercise, adipocytes hydrolyze TAG through lipolysis and release FAs and glycerol to the systemic blood. In adults consuming a typical Western diet, the majority of FAs in adipose TAG are derived from dietary sources and therefore, FAs in the blood reflect mostly FA compositions in the diet [10].
Ast is incorporated into the PL monolayer that surrounds the hydrophobic core of the lipoprotein particles, and studies have reported that 36–64% of blood Ast is found in TAG-rich lipoproteins (chylomicrons/VLDLs), 28–29% in LDL, and 21–24% in HDL [27,32]. Cells acquire Ast by taking up LDL particles through LDL receptor-mediated endocytosis or from HDL particles, possibly via passive diffusion or a receptor-mediated facilitated diffusion process. Ast is known to inhibit oxidation of LDL and other lipoprotein particles [33], a mechanism through which Ast provides beneficial impacts on atherosclerotic cardiovascular diseases.

5. Mechanisms Involved in the Beneficial Actions of Ast and Omega-3 PUFAs

5.1. Antioxidant Action of Ast

The unique structure of Ast, a series of conjugated double-bond chains with polar ionone rings at both ends, allows it to act as a strong antioxidant, effectively scavenging free radicals and reactive oxygen and nitrogen species (RONS). Studies have shown that Ast is a more potent antioxidant than other carotenoids (alpha-carotene, beta-carotene, and lutein) and vitamin E [34,35,36]. Ast is relatively more polar than other carotenoids and spans PL monolayers in lipoproteins and bilayers in cellular membranes, where Ast effectively detoxifies free radicals and other RONS in both the hydrophobic core (FA tails) and surface [35] (Figure 2). Additionally, Ast activates the natural antioxidant defense systems by inducing the expression and activity of antioxidative enzymes through nuclear factor erythroid 2-related factor 2 (NRF2), a key transcription factor that mediates cellular defense systems against oxidative stress and induces the expression levels of antioxidant enzymes, including glutathione peroxidase (GPX), superoxide dismutase (SOD), and catalase (CAT) [37,38].
Ast may also exert beneficial impacts on cellular functions by maintaining membrane integrity through the prevention of peroxidation of PUFAs in membrane PLs. While beta-carotene and lycopene (nonpolar carotenoids) disordered the membrane bilayer and increased lipid peroxidation in model membranes enriched with PUFAs, Ast preserved membrane structure and decreased membrane lipid peroxidation [39]. This is crucial in the prevention of numerous diseases, including atherosclerosis and neurodegenerative diseases, where the accumulation of oxidized lipids plays a crucial role. Additionally, studies have shown that Ast can counteract ferroptosis, a novel form of iron-mediated lipid peroxidation in PUFAs in membrane PLs and cell death, thereby decreasing cardiotoxicity and atherosclerosis, as well as neurotoxicity [40,41].

5.2. Omega-3 PUFA-Mediated Suppression of Oxidative Stress

Although conjugated double bonds in omega-3 PUFAs may react with RONS, acting as a sink for oxidative damage, they are also susceptible to lipid peroxidation. Upon incorporation into membrane PLs, omega-3 PUFAs increase membrane fluidity and lipid raft formation and therefore affect the localization, function, and signaling of membrane-bound proteins, including insulin receptors and those involved in oxidative stress responses [12] (Figure 2). Further, omega-3 PUFAs can reduce oxidative stress through suppression of inflammation, activation of NRF2, and induction of antioxidant enzymes [42].

5.3. Anti-Inflammatory Actions of Ast

Ast is known to inhibit the inflammatory responses through multiple mechanisms, including suppression of the canonical proinflammatory signaling pathways by directly scavenging RONS, inducing NRF2 activity and the expression of antioxidant genes, and decreasing mitochondrial dysfunction. Ast inhibited phosphorylation and degradation of inhibitor of kappa B (IκB) by activating IκB kinase, which prevented nuclear translocation of nuclear factor kappa B (NF-κB), the canonical transcription factor that increases the expression of proinflammatory genes [43]. Additionally, Ast suppressed mitogen-activated protein kinases (MAPKs) [44]. Through these actions, Ast decreased the expression of proinflammatory cytokines [tumor necrosis factor-alpha (TNFα), interleukin-1 beta (IL-1β), interleukin-6 (IL-6)] and other genes [cyclooxygenase 2 (COX-2) and inducible nitric oxide synthase (iNOS)] in various cell types such as macrophages [43,44]. There is an antagonistic crosstalk between the two transcription factors such that NRF2 activation suppresses NF-κB activity and vice versa, especially in the context of oxidative stress and inflammation [45]. Studies have shown that Ast suppressed the expression levels of proinflammatory cytokines through NRF2-dependent and -independent mechanisms [46,47].
Ast is also present in organelle membranes, including mitochondria, where ROS are generated during oxidative metabolism, and high levels of ROS cause mitochondrial dysfunction. In macrophages, Ast blocked endotoxin-induced mitochondrial dysfunction and proinflammatory responses [48]. Furthermore, Ast not only decreased proinflammatory M1 macrophages but also induced macrophage polarization toward more anti-inflammatory phenotypes in peritoneal macrophages [49] and in the liver of mouse models with metabolic dysfunction-associated steatohepatitis [47], improving insulin resistance and hepatic steatosis, respectively.

5.4. Anti-Inflammatory Actions of Omega-3 PUFAs

Different types of FAs have distinctive roles in health and disease, and the health benefits of omega-3 PUFAs are at least in part mediated by their anti-inflammatory actions. Multiple mechanisms are involved in omega-3 PUFA-mediated suppression of inflammation. EPA, DPA, and DHA are used for the synthesis of lipid mediators, including specialized proresolving mediators (SPMs; resolvins, maresins, and protectins) that play crucial roles during the resolution of inflammatory responses [12]. Lipid mediators, including eicosanoids and SPMs, are generated from omega-3 and omega-6 PUFAs through cyclooxygenases, lipoxygenases, and cytochrome P450 oxidases. Omega-3 PUFAs not only compete with omega-6 arachidonic acid (ARA) for the same enzymatic systems, thereby reducing the production of proinflammatory eicosanoids from ARA, but also increase the production of less inflammatory or even anti-inflammatory eicosanoids from EPA [12]. Anti-inflammatory lipid mediators reduce inflammation by decreasing further immune cell infiltration, enhancing the phagocytic activity of macrophages, or inducing a switch to more resolutory M2-macrophages. They also reduce inflammatory responses by suppressing the expression of proinflammatory cytokines (TNFα, IL-1β, and IL-6) and chemokines.
FAs also affect cellular signaling pathways by directly acting as ligands for the membrane receptors. It is well known that saturated FAs, especially PA, activate the canonical proinflammatory signaling pathways via Toll-like receptors (TLRs). Upon activation by PA, TLR4 activates nuclear translocation of NF-κB through stimulation of IκB kinase and degradation of IκBα and IκBβ, which results in the liberation of NF-κB [12]. TLR4 also acts through p38, ERK1/2, and JNK MAPKs that mediate the proinflammatory responses by phosphorylating transcription factors such as activator protein 1. Omega-3 PUFAs have been shown to suppress proinflammatory responses by inhibiting TLR4 or antagonizing PA-mediated stimulation of TLR4 [50,51].
Omega-3 PUFA-mediated activation of free fatty acid receptors (FFAR4), a member of G-protein coupled receptors, is also known to be involved in their beneficial anti-inflammatory actions. FFAR4 is abundantly expressed in immune cells and adipocytes. In macrophages, omega-3 PUFAs act through the FFAR4 to inhibit the canonical proinflammatory signaling pathways, including NF-κB and JNK MAPKs [52]. Omega-3 PUFA-mediated stimulation of FFAR4 has been shown to reduce vascular inflammation and thrombus formation [53].

5.5. Interactive Effects of Ast and Omega-3 PUFAs in Reducing Oxidative Stress and Inflammation

Both AST and omega-3 PUFAs individually contribute to reducing oxidative stress and inflammatory responses. Further, Ast not only prevents oxidation of omega-3 PUFAs in foods, increasing their storage stability, but also suppresses peroxidation of omega-3 PUFAs in lipoproteins and PLs in cellular membranes [21]. When combined, they can provide a more robust defense against free radicals, protecting cells and tissues from damage, and potentially can have additive or synergistic antioxidant effects via the NRF2 pathway, as shown in a hepatic cell line [54]. The antagonistic crosstalk between NF-κB and NRF2 suggests that the two nutrients potentially work together to reduce inflammation.

5.6. Other Mechanisms Mediating the Beneficial Actions of Ast and Omega-3 PUFAs

In addition to providing health benefits by reducing oxidative stress and inflammation, both Ast and omega-3 PUFAs are known to confer beneficial effects through modulation of gut microbiota and gut-derived metabolites. Ast-mediated increases in the abundance of beneficial Akkermansia muciniphila and short-chain FAs are thought to be involved in the beneficial effects of Ast [55,56,57]. Similarly, omega-3 PUFAs also exert protective actions at least in part by improving gut microbiome: increased richness, diversity, and abundance of short-chain FA-producing genera, Bifidobaterium, Roseburia, and Lactobacillu while decreasing LPS-producing Bacteroidetes [58,59].
AST has been shown to enhance transcriptional activity of peroxisome proliferator-activated receptor-gamma [60], a nuclear receptor that plays a fundamental role in adipocyte development and their metabolic and endocrine properties, insulin sensitivity, and the regulation of inflammatory responses [61]. Therefore, Ast may enhance the thermogenic capacity and secretory profiles in adipose tissues, providing protection against cardiometabolic diseases. Omega-3 PUFAs are also known to enhance the browning of white fat through the FFAR4 as well as the production of bioactive lipid species, which in turn exert beneficial impacts on obesity and its associated metabolic diseases [62].
Both Ast and omega-3 PUFAs are known to suppress endoplasmic reticulum stress, leading to a reduction in the proinflammatory responses, oxidative stress, and apoptosis in several models [63,64]. Other mechanisms involved in the beneficial effects of Ast and omega-3 PUFAs include their regulation of autophagy through activation of AMP-activated protein kinase and insulin signaling activities [65,66].

6. Health Benefits of Ast and Omega-3 PUFAs

6.1. Beneficial Impacts of Ast

Oxidative stress and chronic inflammation are the major pathological factors in various diseases such as cardiometabolic diseases, neurodegenerative diseases, and ageing-associated macular diseases. Numerous studies, mostly in preclinical animal models, have shown that Ast provides protection against cardiometabolic diseases by alleviating hyperlipidemia, hepatic steatosis, and insulin resistance through reducing oxidative stress and inflammation [67,68,69]. Ast also reduced hepatic injuries through downregulation of MAPK-mediated apoptosis and autophagy [44,70]. Another study, using a diet-induced model of fatty liver disease, found that Ast reduced liver inflammation, improved insulin sensitivity, and was more effective than vitamin E [49]. Ast also exerted a protective effect on cardiac dysfunction and necrosis caused by sepsis by inhibiting inflammation [71].
Chronic neuroinflammatory response involving neuronal and immune cells occurs in the central nervous system (CNS) during the development of neurodegenerative diseases. Proinflammatory mediators (TNFα, IL-6, and NO) released by immune cells, including the tissue-resident macrophage microglia, increase oxidative stress, causing cytotoxicity in the CNS [72]. Ast has a unique ability to cross the blood–brain barrier and exerts neuroprotective effects directly within the CNS through its potent antioxidative and anti-inflammatory actions [72]. Ast also enhances mitochondrial function, while suppressing neuronal cell death. Therefore, Ast has garnered significant interest for its potential therapeutic role in neurodegenerative diseases. For the efficacy of Ast in neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, and ageing-associated decline in cognitive function and dementia, readers are referred to recently published reviews [73,74].

6.2. Beneficial Impacts of Omega-3 PUFAs: Distinctive Roles of EPA and DHA

Omega-3 PUFAs also exert beneficial impacts on various diseases. Their health benefits for cardiometabolic and neurodegenerative diseases are well known, and for further information, readers are referred to previous reviews on these topics [12,62,75]. While generally positive associations between the intake of omega-PUFAs and reduced disease risks have been reported, whether increasing their intake provides protective effects on health and the underlying mechanisms that mediate their beneficial impacts remains uncertain. Moreover, although EPA (20:5) can be converted into DHA (C22:6), the two omega-3 PUFAs are known to exhibit distinct tissue distribution patterns, metabolism, functional properties, and contribution to disease risks [76]. Current understanding of the differential roles of EPA and DHA, the two omega-3 PUFAs enriched in seafood, is discussed below.
DHA is the most abundant PUFA in the nervous system, especially the brain and retina, constituting up to 60% of the total omega-3 PUFAs [76]. DHA contributes to the structural formation and functional capabilities during the development of the CNS. Studies using model membranes have shown that EPA and DHA associate with distinct regions of the membrane, and DHA is more incorporated into lipid rafts than EPA [77]. In the brain, DHA enhances and stabilizes lipid rafts containing high levels of cholesterol and sphingomyelin. Moreover, DHA and its derivatives, including D-class neuroprotectins, provide neuroprotective actions against neurodegenerative diseases and injuries [78]. Preclinical studies in neuronal cell culture models have shown that differential impacts of EPA, DHA, and the ratio of EPA to DHA on amyloid accumulation, cellular stress signaling, and apoptosis [79,80,81]. However, a recently published systematic review and meta-analysis reported that although omega-3 PUFA supplementation reduced the progression of cognitive decline among patients with Alzheimer’s disease, there were no differences among EPA, DHA, and EPA/DHA mixed formulations [82], and more studies are needed to assess potential distinct roles of DHA compared to EPA in neurodegenerative diseases.
In the retina, a high concentration of DHA is essential for maintaining the fluidity, integrity, and correct morphology of photoreceptor cell membranes [83]. The rapid motion of DHA is thought to be necessary for rapid conformational changes in rhodopsin during light perception. Accordingly, a recent systematic review on randomized controlled clinical trials supports a crucial role of omega-3 PUFA supplementation, especially DHA, in visual health during early life [84]. The results from animal studies showed that a PL form of DHA (lysophosphatidylcholine-DHA), but not omega-3 PUFA supplements in TAG forms, increased retinal DHA, indicating its higher bioavailability [85]. However, whether it leads to improvement in visual functions in humans requires further investigation.
Despite a similar reduction in TAG levels, EPA alone, compared to the combined use of EPA and DHA, showed a further significant risk reduction in cardiovascular diseases, an implication of distinctive roles between EPA and DHA [86,87]. While DHA, compared to EPA, creates a more fluid membrane, EPA has been shown to suppress membrane cholesterol crystalline domain formation more effectively through a potent antioxidant mechanism in hyperglycemia [88]. Further, EPA, compared to DHA, more effectively suppressed oxidation of HDL, small-dense LDL, and model membranes [89]. Different lipid mediators generated from EPA, compared to DPA and DHA, may also contribute to the better actions of EPA than mixed formulations of omega-3 PUFAs.

6.3. Interactions of Ast and Omega-3 PUFAs in Reducing Disease Risks

The complementary actions of Ast and omega-3 PUFAs to counteract oxidative stress and inflammatory responses suggest that combined intake of both nutrients may be better than each (Figure 3). Accordingly, several studies have shown that combinations of Ast and DHA exhibit better protective effects than each in animal models of neurodegenerative diseases [90,91]. Moreover, Ast and DHA exhibited potential synergistic beneficial effects on dysregulated plasma metabolic profiles induced by undernutrition during the gestation and lactation periods [92]. Krill oil contains both Ast and omega-3 PUFAs. However, krill oil contains omega-3 PUFAs in PL forms compared to the TAG forms in fish oil, and it also contains variable amounts of EPA and DHA per serving [93]. Whether supplementation of krill oil is better than fish oil in reducing disease risks remains undetermined.

7. Conclusions

Ast and omega-3 PUFAs are commonly present in selected seafood, and they are known to exert beneficial impacts on health. The interactions of Ast and omega-3 PUFAs are manifold. Dietary lipids, such as FAs esterified in the forms of TAG and PLs, enhance the digestion and absorption of Ast. The transport of Ast is also closely related to lipoprotein metabolism. Therefore, dietary lipids enhance the bioavailability of Ast. Furthermore, the polyene chain in omega-3 PUFAs can be oxidized and degraded, and Ast, as a strong antioxidant, can prevent this oxidation both in foods and the body.
Both Ast and omega-3 PUFAs inhibit oxidative stress and inflammation, the major pathological factors underlying various diseases, including cardiometabolic and neurodegenerative diseases, through complementary mechanisms. Ast is relatively polar compared to other carotenoids and can span the PL monolayers of lipoprotein particles and bilayers of cellular membranes, preventing lipid peroxidation by scavenging free radicals and RONS. Ast also suppresses oxidative stress by stimulating the transcriptional activity of NRF2, thereby increasing expression levels of antioxidant enzymes. Omega-3 PUFAs suppress inflammation through multiple mechanisms. Upon incorporation into membrane PLs, omega-3 PUFAs increase membrane fluidity and lipid raft formation, affecting cellular signaling activities. Additionally, they serve as precursors for the production of anti-inflammatory lipid mediators and reduce the production of proinflammatory eicosanoids by competing with omega-6 ARA for the same enzymes. Omega-3 PUFAs also suppress inflammation by antagonizing the proinflammatory TLR4 signaling pathways and by acting as ligands for FFAR4. Through these various mechanisms, omega-3 PUFAs suppress the canonical proinflammatory NF-κB and MAPK signaling pathways. An antagonistic crosstalk exists between NRF2 and NF-κB, the two primary transcription factors that mediate antioxidant responses and proinflammatory responses, respectively. Therefore, Ast activation of NRF2 can suppress inflammation while omega-3 PUFA inhibition of NF-κB can enhance antioxidant capacity, clearly indicating complementary actions of the two bioactive food components.
The interactive actions of Ast and omega-3 PUFAs create a powerful duo, working in concert to exert profound antioxidative and anti-inflammatory effects throughout the body. While numerous studies have shown beneficial impacts of each in various disease models, most results are derived from preclinical models, and studies that have examined the interrelationships of their beneficial effects, especially in humans, are limited. Various isomers of Ast and omega-3 PUFAs, EPA and DHA in the form of TAG or PLs, are present in food, and they are known to exhibit distinctive bioavailability or functional roles. Moreover, food sources also contain many other components, and future studies to understand the complexity of the food matrix on health and disease are warranted. The potential additive or synergistic effects of Ast and omega-3 PUFAs extend to various other areas, including eye health, skin health, and ageing-associated health issues, where oxidative stress and chronic inflammation play crucial roles in their pathogenesis.

Funding

This work was supported by NIH P20GM139753 (Project Leader 2), Hawaii Community Foundation (MedRes_2024_00006265), and USDA Hatch Project (HAW02063-H).

Conflicts of Interest

The author declares no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ALAAlpha-linolenic acid
AP-1Activator protein 1
ARAArachidonic acid
AstAstaxanthin
CATCatalase
CMChylomicron
CMDsCardiometabolic diseases
CNSCentral nervous system
DAGDiacylglycerol
DHADocosahexaenoic acid
DPADocosapentaenoic acid
EPAEicosapentaenoic acid
FAsFatty acids
FFARsFree fatty acid receptors
GPXGlutathione peroxidase
HDLHigh-density lipoprotein
IL-1βInterleukin-1 beta
IL-6Interleukin-6
LDLLow-density lipoprotein
Lyso-PLsLyso-phospholipids
MAGMonoacylglycerol
MAPKsMitogen-activated protein kinases
NF-κBNuclear factor-kappa B
NRF2Nuclear factor erythroid 2-related factor 2
Omega-3 PUFAsVery long-chain omega-3 polyunsaturated fatty acids
PAPalmitic acid
PLsPhospholipids
PUFAsPolyunsaturated fatty acids
RONSReactive oxygen and nitrogen species
SODSuperoxide dismutase
SPMsSpecialized proresolving mediators
TAGTriacylglycerol
TLRsToll-like receptors
TNFαTumor necrosis factor-alpha

References

  1. Abuajah, C.I.; Ogbonna, A.C.; Osuji, C.M. Functional components and medicinal properties of food: A review. J. Food Sci. Technol. 2015, 52, 2522–2529. [Google Scholar] [CrossRef]
  2. Polotow, T.G.; Poppe, S.C.; Vardaris, C.V.; Ganini, D.; Guariroba, M.; Mattei, R.; Hatanaka, E.; Martins, M.F.; Bondan, E.F.; Barros, M.P. Redox Status and Neuro Inflammation Indexes in Cerebellum and Motor Cortex of Wistar Rats Supplemented with Natural Sources of Omega-3 Fatty Acids and Astaxanthin: Fish Oil, Krill Oil, and Algal Biomass. Mar. Drugs 2015, 13, 6117–6137. [Google Scholar] [CrossRef]
  3. Liu, Y.; Robinson, A.M.; Su, X.Q.; Nurgali, K. Krill Oil and Its Bioactive Components as a Potential Therapy for Inflammatory Bowel Disease: Insights from In Vivo and In Vitro Studies. Biomolecules 2024, 14, 447. [Google Scholar] [CrossRef]
  4. Lu, P.; Reisdorph, N.; Hendricks, A.E.; Reisdorph, R.; Quinn, K.; Armstrong, M.; Michel, C.; Doenges, K.A.; Frank, D.N.; Campbell, W.W.; et al. Characterization of astaxanthin isomers in different types of salmon filets and human plasma after salmon consumption. Food Chem. 2025, 482, 144024. [Google Scholar] [CrossRef]
  5. Elbahnaswy, S.; Elshopakey, G.E. Recent progress in practical applications of a potential carotenoid astaxanthin in aquaculture industry: A review. Fish Physiol. Biochem. 2024, 50, 97–126. [Google Scholar] [CrossRef] [PubMed]
  6. Liu, X.; Xie, J.; Zhou, L.; Zhang, J.; Chen, Z.; Xiao, J.; Cao, Y.; Xiao, H. Recent advances in health benefits and bioavailability of dietary astaxanthin and its isomers. Food Chem. 2023, 404, 134605. [Google Scholar] [CrossRef] [PubMed]
  7. Grynbaum, M.D.; Hentschel, P.; Putzbach, K.; Rehbein, J.; Krucker, M.; Nicholson, G.; Albert, K. Unambiguous detection of astaxanthin and astaxanthin fatty acid esters in krill (Euphausia superba Dana). J. Sep. Sci. 2005, 28, 1685–1693. [Google Scholar] [CrossRef]
  8. Chitchumroonchokchai, C.; Failla, M.L. Bioaccessibility and intestinal cell uptake of astaxanthin from salmon and commercial supplements. Food Res. Int. 2017, 99, 936–943. [Google Scholar] [CrossRef]
  9. Molino, A.; Rimauro, J.; Casella, P.; Cerbone, A.; Larocca, V.; Chianese, S.; Karatza, D.; Mehariya, S.; Ferraro, A.; Hristoforou, E.; et al. Extraction of astaxanthin from microalga Haematococcus pluvialis in red phase by using generally recognized as safe solvents and accelerated extraction. J. Biotechnol. 2018, 283, 51–61. [Google Scholar] [CrossRef] [PubMed]
  10. Strawford, A.; Antelo, F.; Christiansen, M.; Hellerstein, M.K. Adipose tissue triglyceride turnover, de novo lipogenesis, and cell proliferation in humans measured with 2H2O. Am. J. Physiol. Endocrinol. Metab. 2004, 286, E577–E588. [Google Scholar] [CrossRef]
  11. Cormier, H.; Rudkowska, I.; Lemieux, S.; Couture, P.; Julien, P.; Vohl, M.C. Effects of FADS and ELOVL polymorphisms on indexes of desaturase and elongase activities: Results from a pre-post fish oil supplementation. Genes Nutr. 2014, 9, 437. [Google Scholar] [CrossRef]
  12. Toncan, F.; Raj, R.R.; Lee, M.J. Dynamics of Fatty Acid Composition in Lipids and Their Distinct Roles in Cardiometabolic Health. Biomolecules 2025, 15, 696. [Google Scholar] [CrossRef]
  13. Thompson, M.; Hein, N.; Hanson, C.; Smith, L.M.; Anderson-Berry, A.; Richter, C.K.; Stessy, B.K.; Kusi, A.A.; Kris-Etherton, P.; Skulas-Ray, A.C.; et al. Omega-3 Fatty Acid Intake by Age, Gender, and Pregnancy Status in the United States: National Health and Nutrition Examination Survey 2003–2014. Nutrients 2019, 11, 177. [Google Scholar] [CrossRef]
  14. Hu, X.F.; Chan, H.M. Seafood Consumption and Its Contribution to Nutrients Intake among Canadians in 2004 and 2015. Nutrients 2020, 13, 77. [Google Scholar] [CrossRef] [PubMed]
  15. Kwantes, J.M.; Grundmann, O. A brief review of krill oil history, research, and the commercial market. J. Diet. Suppl. 2015, 12, 23–35. [Google Scholar] [CrossRef]
  16. Ramos-Romero, S.; Torrella, J.R.; Pages, T.; Viscor, G.; Torres, J.L. Edible Microalgae and Their Bioactive Compounds in the Prevention and Treatment of Metabolic Alterations. Nutrients 2021, 13, 563. [Google Scholar] [CrossRef] [PubMed]
  17. Kutzner, L.; Ostermann, A.I.; Konrad, T.; Riegel, D.; Hellhake, S.; Schuchardt, J.P.; Schebb, N.H. Lipid Class Specific Quantitative Analysis of n-3 Polyunsaturated Fatty Acids in Food Supplements. J. Agric. Food Chem. 2017, 65, 139–147. [Google Scholar] [CrossRef] [PubMed]
  18. Koch, E.; Kampschulte, N.; Schebb, N.H. Comprehensive Analysis of Fatty Acid and Oxylipin Patterns in n3-PUFA Supplements. J. Agric. Food Chem. 2022, 70, 3979–3988. [Google Scholar] [CrossRef]
  19. Nelson, S.D.; Munger, M.A. Icosapent ethyl for treatment of elevated triglyceride levels. Ann. Pharmacother. 2013, 47, 1517–1523. [Google Scholar] [CrossRef]
  20. Albert, B.B.; Cameron-Smith, D.; Hofman, P.L.; Cutfield, W.S. Oxidation of marine omega-3 supplements and human health. BioMed Res. Int. 2013, 2013, 464921. [Google Scholar] [CrossRef]
  21. Wang, H.; He, W.; Mahukpego, D.D.; Zhang, H.; Dwi, N.R.; Tang, C.; Guo, X.; Yu, Y.; Zhao, Q.; Qin, Y.; et al. Astaxanthin improved the storage stability of docosahexaenoic acid-enriched eggs by inhibiting oxidation of non-esterified poly-unsaturated fatty acids. Food Chem. 2022, 381, 132256. [Google Scholar] [CrossRef] [PubMed]
  22. Tan, K.; Lim, L.; Peng, Y.; Cheong, K.L. Effects of food processing on the lipid nutritional quality of commercially important fish and shellfish. Food Chem. 2023, 20, 101034. [Google Scholar] [CrossRef]
  23. 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]
  24. Honda, M.; Murakami, K.; Osawa, Y.; Kawashima, Y.; Hirasawa, K.; Kuroda, I. Z-Isomers of Astaxanthin Exhibit Greater Bioavailability and Tissue Accumulation Efficiency than the All-E-Isomer. J. Agric. Food Chem. 2021, 69, 3489–3495. [Google Scholar] [CrossRef]
  25. Ahn, S.H.; Lim, S.J.; Ryu, Y.M.; Park, H.R.; Suh, H.J.; Han, S.H. Absorption rate of krill oil and fish oil in blood and brain of rats. Lipids Health Dis. 2018, 17, 162. [Google Scholar] [CrossRef]
  26. Mercke, O.J.; Lignell, A.; Pettersson, A.; Hoglund, 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]
  27. Coral-Hinostroza, G.N.; Ytrestoyl, T.; Ruyter, B.; Bjerkeng, B. Plasma appearance of unesterified astaxanthin geometrical E/Z and optical R/S isomers in men given single doses of a mixture of optical 3 and 3′R/S isomers of astaxanthin fatty acyl diesters. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2004, 139, 99–110. [Google Scholar] [CrossRef]
  28. Yang, C.; Hassan, Y.I.; Liu, R.; Zhang, H.; Chen, Y.; Zhang, L.; Tsao, R. Anti-Inflammatory Effects of Different Astaxanthin Isomers and the Roles of Lipid Transporters in the Cellular Transport of Astaxanthin Isomers in Caco-2 Cell Monolayers. J. Agric. Food Chem. 2019, 67, 6222–6231. [Google Scholar] [CrossRef]
  29. Liu, X.; Zhang, J.; Chen, Z.; Xiao, J.; Zhou, A.; Fu, Y.; Cao, Y. Cluster-determinant 36 (CD36) mediates intestinal absorption of dietary astaxanthin and affects its secretion. Food Res. Int. 2023, 173, 113328. [Google Scholar] [CrossRef] [PubMed]
  30. Wang, H.; Eckel, R.H. Lipoprotein lipase: From gene to obesity. Am. J. Physiol. Endocrinol. Metab. 2009, 297, E271–E288. [Google Scholar] [CrossRef] [PubMed]
  31. Cohen, J.C.; Horton, J.D.; Hobbs, H.H. Human fatty liver disease: Old questions and new insights. Science 2011, 332, 1519–1523. [Google Scholar] [CrossRef] [PubMed]
  32. Osterlie, M.; Bjerkeng, B.; Liaaen-Jensen, S. Plasma appearance and distribution of astaxanthin E/Z and R/S isomers in plasma lipoproteins of men after single dose administration of astaxanthin. J. Nutr. Biochem. 2000, 11, 482–490. [Google Scholar] [CrossRef]
  33. 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]
  34. Naguib, Y.M. Antioxidant activities of astaxanthin and related carotenoids. J. Agric. Food Chem. 2000, 48, 1150–1154. [Google Scholar] [CrossRef]
  35. Goto, S.; Kogure, K.; Abe, K.; Kimata, Y.; Kitahama, K.; Yamashita, E.; Terada, H. Efficient radical trapping at the surface and inside the phospholipid membrane is responsible for highly potent antiperoxidative activity of the carotenoid astaxanthin. Biochim. Biophys. Acta 2001, 1512, 251–258. [Google Scholar] [CrossRef]
  36. Hosaka, S.; Obuki, M.; Nakajima, J.; Suzuki, M. Comparative study of antioxidants as quenchers or scavengers of reactive oxygen species based on quenching of MCLA-dependent chemiluminescence. Luminescence 2005, 20, 419–427. [Google Scholar] [CrossRef] [PubMed]
  37. Ranga, R.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]
  38. Kong, D.; Liu, Y.; Li, L.; Wang, H.; Li, K.; Zheng, G. Astaxanthin ameliorates oxidative stress in lens epithelial cells by regulating GPX4 and ferroptosis. Chem. Biol. Interact. 2023, 383, 110684. [Google Scholar] [CrossRef]
  39. McNulty, H.; Jacob, R.F.; Mason, R.P. Biologic activity of carotenoids related to distinct membrane physicochemical interactions. Am. J. Cardiol. 2008, 101, 20D–29D. [Google Scholar] [CrossRef]
  40. Jin, M.; Chen, X.; Zheng, L.; Peng, Y.; Lin, M.; Liang, K.; Liu, X.; Xu, Z.; Yang, Y.; Wei, B.; et al. Astaxanthin-loaded polylactic acid-glycolic acid nanoparticles alleviates atherosclerosis by suppressing macrophage ferroptosis via the NRF2/SLC7A11/GPX4 pathway. Arch. Biochem. Biophys. 2025, 765, 110316. [Google Scholar] [CrossRef] [PubMed]
  41. Yin, B.; Ren, J.; Liu, X.; Zhang, Y.; Zuo, J.; Wen, R.; Pei, H.; Lu, M.; Zhu, S.; Zhang, Z.; et al. Astaxanthin mitigates doxorubicin-induced cardiotoxicity via inhibiting ferroptosis and autophagy: A study based on bioinformatic analysis and in vivo/vitro experiments. Front. Pharmacol. 2025, 16, 1524448. [Google Scholar] [CrossRef]
  42. Davinelli, S.; Medoro, A.; Intrieri, M.; Saso, L.; Scapagnini, G.; Kang, J.X. Targeting NRF2-KEAP1 axis by Omega-3 fatty acids and their derivatives: Emerging opportunities against aging and diseases. Free Radic. Biol. Med. 2022, 193, 736–750. [Google Scholar] [CrossRef] [PubMed]
  43. Lee, S.J.; Bai, S.K.; Lee, K.S.; Namkoong, S.; Na, H.J.; Ha, K.S.; Han, J.A.; Yim, S.V.; Chang, K.; Kwon, Y.G.; et al. Astaxanthin inhibits nitric oxide production and inflammatory gene expression by suppressing I(kappa)B kinase-dependent NF-kappaB activation. Mol. Cells 2003, 16, 97–105. [Google Scholar] [CrossRef] [PubMed]
  44. Li, J.; Xia, Y.; Liu, T.; Wang, J.; Dai, W.; Wang, F.; Zheng, Y.; Chen, K.; Li, S.; Abudumijiti, H.; et al. Protective effects of astaxanthin on ConA-induced autoimmune hepatitis by the JNK/p-JNK pathway-mediated inhibition of autophagy and apoptosis. PLoS ONE 2015, 10, e0120440. [Google Scholar] [CrossRef]
  45. Gao, W.; Guo, L.; Yang, Y.; Wang, Y.; Xia, S.; Gong, H.; Zhang, B.K.; Yan, M. Dissecting the Crosstalk Between Nrf2 and NF-kappaB Response Pathways in Drug-Induced Toxicity. Front. Cell Dev. Biol. 2021, 9, 809952. [Google Scholar] [CrossRef] [PubMed]
  46. Kobayashi, E.H.; Suzuki, T.; Funayama, R.; Nagashima, T.; Hayashi, M.; Sekine, H.; Tanaka, N.; Moriguchi, T.; Motohashi, H.; Nakayama, K.; et al. Nrf2 suppresses macrophage inflammatory response by blocking proinflammatory cytokine transcription. Nat. Commun. 2016, 7, 11624. [Google Scholar] [CrossRef]
  47. Farruggia, C.; Kim, M.B.; Bae, M.; Lee, Y.; Pham, T.X.; Yang, Y.; Han, M.J.; Park, Y.K.; Lee, J.Y. Astaxanthin exerts anti-inflammatory and antioxidant effects in macrophages in NRF2-dependent and independent manners. J. Nutr. Biochem. 2018, 62, 202–209. [Google Scholar] [CrossRef]
  48. Sun, L.; Kim, S.; Mori, R.; Miyaji, N.; Nikawa, T.; Hirasaka, K. Astaxanthin Exerts Immunomodulatory Effect by Regulating SDH-HIF-1alpha Axis and Reprogramming Mitochondrial Metabolism in LPS-Stimulated RAW264.7 Cells. Mar. Drugs 2022, 20, 660. [Google Scholar] [CrossRef]
  49. Ni, Y.; Nagashimada, M.; Zhuge, F.; Zhan, L.; Nagata, N.; Tsutsui, A.; Nakanuma, Y.; Kaneko, S.; Ota, T. Astaxanthin prevents and reverses diet-induced insulin resistance and steatohepatitis in mice: A comparison with vitamin E. Sci. Rep. 2015, 5, 17192. [Google Scholar] [CrossRef]
  50. Lee, J.Y.; Ye, J.; Gao, Z.; Youn, H.S.; Lee, W.H.; Zhao, L.; Sizemore, N.; Hwang, D.H. Reciprocal modulation of Toll-like receptor-4 signaling pathways involving MyD88 and phosphatidylinositol 3-kinase/AKT by saturated and polyunsaturated fatty acids. J. Biol. Chem. 2003, 278, 37041–37051. [Google Scholar] [CrossRef] [PubMed]
  51. Nakakuki, M.; Kawano, H.; Notsu, T.; Imada, K. Eicosapentaenoic acid suppresses palmitate-induced cytokine production by modulating long-chain acyl-CoA synthetase 1 expression in human THP-1 macrophages. Atherosclerosis 2013, 227, 289–296. [Google Scholar] [CrossRef] [PubMed]
  52. Oh, D.Y.; Talukdar, S.; Bae, E.J.; Imamura, T.; Morinaga, H.; Fan, W.; Li, P.; Lu, W.J.; Watkins, S.M.; Olefsky, J.M. GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects. Cell 2010, 142, 687–698. [Google Scholar] [CrossRef]
  53. Li, X.; Ballantyne, L.L.; Che, X.; Mewburn, J.D.; Kang, J.X.; Barkley, R.M.; Murphy, R.C.; Yu, Y.; Funk, C.D. Endogenously generated omega-3 fatty acids attenuate vascular inflammation and neointimal hyperplasia by interaction with free fatty acid receptor 4 in mice. J. Am. Heart Assoc. 2015, 4, e001856. [Google Scholar] [CrossRef]
  54. Saw, C.L.; Yang, A.Y.; Guo, Y.; Kong, A.N. Astaxanthin and omega-3 fatty acids individually and in combination protect against oxidative stress via the Nrf2-ARE pathway. Food Chem. Toxicol. 2013, 62, 869–875. [Google Scholar] [CrossRef]
  55. Liu, H.; Liu, M.; Fu, X.; Zhang, Z.; Zhu, L.; Zheng, X.; Liu, J. Astaxanthin Prevents Alcoholic Fatty Liver Disease by Modulating Mouse Gut Microbiota. Nutrients 2018, 10, 1298. [Google Scholar] [CrossRef]
  56. Wang, J.; Liu, S.; Wang, H.; Xiao, S.; Li, C.; Li, Y.; Liu, B. Xanthophyllomyces dendrorhous-Derived Astaxanthin Regulates Lipid Metabolism and Gut Microbiota in Obese Mice Induced by A High-Fat Diet. Mar. Drugs 2019, 17, 337. [Google Scholar] [CrossRef]
  57. Wu, L.; Lyu, Y.; Srinivasagan, R.; Wu, J.; Ojo, B.; Tang, M.; El-Rassi, G.D.; Metzinger, K.; Smith, B.J.; Lucas, E.A.; et al. Astaxanthin-Shifted Gut Microbiota Is Associated with Inflammation and Metabolic Homeostasis in Mice. J. Nutr. 2020, 150, 2687–2698. [Google Scholar] [CrossRef]
  58. Cui, C.; Li, Y.; Gao, H.; Zhang, H.; Han, J.; Zhang, D.; Li, Y.; Zhou, J.; Lu, C.; Su, X. Modulation of the gut microbiota by the mixture of fish oil and krill oil in high-fat diet-induced obesity mice. PLoS ONE 2017, 12, e0186216. [Google Scholar] [CrossRef] [PubMed]
  59. Machate, D.J.; Figueiredo, P.S.; Marcelino, G.; Guimaraes, R.C.A.; Hiane, P.A.; Bogo, D.; Pinheiro, V.A.Z.; Oliveira, L.C.S.; Pott, A. Fatty Acid Diets: Regulation of Gut Microbiota Composition and Obesity and Its Related Metabolic Dysbiosis. Int. J. Mol. Sci. 2020, 21, 4093. [Google Scholar] [CrossRef] [PubMed]
  60. Inoue, M.; Tanabe, H.; Matsumoto, A.; Takagi, M.; Umegaki, K.; Amagaya, S.; Takahashi, J. Astaxanthin functions differently as a selective peroxisome proliferator-activated receptor gamma modulator in adipocytes and macrophages. Biochem. Pharmacol. 2012, 84, 692–700. [Google Scholar] [CrossRef]
  61. Lee, M.J.; Jash, S.; Jones, J.E.C.; Puri, V.; Fried, S.K. Rosiglitazone remodels the lipid droplet and britens human visceral and subcutaneous adipocytes ex vivo. J. Lipid Res. 2019, 60, 856–868. [Google Scholar] [CrossRef]
  62. Raj, R.R.; Lofquist, S.; Lee, M.J. Remodeling of Adipose Tissues by Fatty Acids: Mechanistic Update on Browning and Thermogenesis by n-3 Polyunsaturated Fatty Acids. Pharm. Res. 2023, 40, 467–480. [Google Scholar] [CrossRef] [PubMed]
  63. Shi, H.; Zhao, Y. Astaxanthin inhibits apoptosis in a cell model of tauopathy by attenuating endoplasmic reticulum stress and unfolded protein response. Eur. J. Pharmacol. 2024, 983, 176962. [Google Scholar] [CrossRef]
  64. Begum, G.; Kintner, D.; Liu, Y.; Cramer, S.W.; Sun, D. DHA inhibits ER Ca2+ release and ER stress in astrocytes following in vitro ischemia. J. Neurochem. 2012, 120, 622–630. [Google Scholar] [CrossRef]
  65. Kim, S.H.; Kim, H. Astaxanthin Modulation of Signaling Pathways That Regulate Autophagy. Mar. Drugs 2019, 17, 546. [Google Scholar] [CrossRef]
  66. Gwon, D.H.; Hwang, T.W.; Ro, J.Y.; Kang, Y.J.; Jeong, J.Y.; Kim, D.K.; Lim, K.; Kim, D.W.; Choi, D.E.; Kim, J.J. High Endogenous Accumulation of omega-3 Polyunsaturated Fatty Acids Protect against Ischemia-Reperfusion Renal Injury through AMPK-Mediated Autophagy in Fat-1 Mice. Int. J. Mol. Sci. 2017, 18, 2081. [Google Scholar] [CrossRef]
  67. 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]
  68. Kim, B.; Farruggia, C.; Ku, C.S.; Pham, T.X.; Yang, Y.; Bae, M.; Wegner, C.J.; Farrell, N.J.; Harness, E.; Park, Y.K.; et al. Astaxanthin inhibits inflammation and fibrosis in the liver and adipose tissue of mouse models of diet-induced obesity and nonalcoholic steatohepatitis. J. Nutr. Biochem. 2017, 43, 27–35. [Google Scholar] [CrossRef]
  69. Radice, R.P.; Limongi, A.R.; Viviano, E.; Padula, M.C.; Martelli, G.; Bermano, G. Effects of astaxanthin in animal models of obesity-associated diseases: A systematic review and meta-analysis. Free Radic. Biol. Med. 2021, 171, 156–168. [Google Scholar] [CrossRef] [PubMed]
  70. Li, J.; Wang, F.; Xia, Y.; Dai, W.; Chen, K.; Li, S.; Liu, T.; Zheng, Y.; Wang, J.; Lu, W.; et al. Astaxanthin Pretreatment Attenuates Hepatic Ischemia Reperfusion-Induced Apoptosis and Autophagy via the ROS/MAPK Pathway in Mice. Mar. Drugs 2015, 13, 3368–3387. [Google Scholar] [CrossRef] [PubMed]
  71. Xie, W.J.; Hou, G.; Wang, L.; Wang, S.S.; Xiong, X.X. Astaxanthin suppresses lipopolysaccharide‑induced myocardial injury by regulating MAPK and PI3K/AKT/mTOR/GSK3beta signaling. Mol. Med. Rep. 2020, 22, 3338–3346. [Google Scholar]
  72. Medoro, A.; Davinelli, S.; Milella, L.; Willcox, B.J.; Allsopp, R.C.; Scapagnini, G.; Willcox, D.C. Dietary Astaxanthin: A Promising Antioxidant and Anti-Inflammatory Agent for Brain Aging and Adult Neurogenesis. Mar. Drugs 2023, 21, 643. [Google Scholar] [CrossRef]
  73. Adiguzel, E.; Ulger, T.G. A marine-derived antioxidant astaxanthin as a potential neuroprotective and neurotherapeutic agent: A review of its efficacy on neurodegenerative conditions. Eur. J. Pharmacol. 2024, 977, 176706. [Google Scholar] [CrossRef] [PubMed]
  74. Utomo, N.P.; Pinzon, R.T.; Latumahina, P.K.; Damayanti, K.R.S. Astaxanthin and improvement of dementia: A systematic review of current clinical trials. Cereb. Circ. Cogn. Behav. 2024, 7, 100226. [Google Scholar] [CrossRef] [PubMed]
  75. Bertoni, C.; Pini, C.; Mazzocchi, A.; Agostoni, C.; Brambilla, P. The Role of Alpha-Linolenic Acid and Other Polyunsaturated Fatty Acids in Mental Health: A Narrative Review. Int. J. Mol. Sci. 2024, 25, 12479. [Google Scholar] [CrossRef]
  76. Dyall, S.C. Long-chain omega-3 fatty acids and the brain: A review of the independent and shared effects of EPA, DPA and DHA. Front. Aging Neurosci. 2015, 7, 52. [Google Scholar] [CrossRef]
  77. Williams, J.A.; Batten, S.E.; Harris, M.; Rockett, B.D.; Shaikh, S.R.; Stillwell, W.; Wassall, S.R. Docosahexaenoic and eicosapentaenoic acids segregate differently between raft and nonraft domains. Biophys. J. 2012, 103, 228–237. [Google Scholar] [CrossRef]
  78. Echeverria, F.; Valenzuela, R.; Catalina Hernandez-Rodas, M.; Valenzuela, A. Docosahexaenoic acid (DHA), a fundamental fatty acid for the brain: New dietary sources. Prostaglandins Leukot. Essent. Fat. Acids 2017, 124, 1–10. [Google Scholar] [CrossRef]
  79. Zhang, Y.P.; Brown, R.E.; Zhang, P.C.; Zhao, Y.T.; Ju, X.H.; Song, C. DHA, EPA and their combination at various ratios differently modulated Abeta(25–35)-induced neurotoxicity in SH-SY5Y cells. Prostaglandins Leukot. Essent. Fat. Acids 2018, 136, 85–94. [Google Scholar] [CrossRef] [PubMed]
  80. Ceccarini, M.R.; Ceccarelli, V.; Codini, M.; Fettucciari, K.; Calvitti, M.; Cataldi, S.; Albi, E.; Vecchini, A.; Beccari, T. The Polyunsaturated Fatty Acid EPA, but Not DHA, Enhances Neurotrophic Factor Expression through Epigenetic Mechanisms and Protects against Parkinsonian Neuronal Cell Death. Int. J. Mol. Sci. 2022, 23, 16176. [Google Scholar] [CrossRef]
  81. Wang, Y.W.; Li, Q.; Li, X.Y.; Zhao, Y.C.; Wang, C.C.; Xue, C.H.; Wang, Y.M.; Zhang, T.T. A Comparative Study about the Neuroprotective Effects of DHA-Enriched Phosphatidylserine and EPA-Enriched Phosphatidylserine against Oxidative Damage in Primary Hippocampal Neurons. Mar. Drugs 2023, 21, 410. [Google Scholar] [CrossRef]
  82. Calderon, M.E.; Zachariah, S.S.; Salazar Ore, J.V.; Borges-Sosa, O.A.; Srinivas, S.; Mareddy, N.S.R.; Manzoor, T.; Di, V.M.; Al, S.Y.; Taneja, R.; et al. The effects of omega-3, DHA, EPA, Souvenaid(R) in Alzheimer’s disease: A systematic review and meta-analysis. Neuropsychopharmacol. Rep. 2024, 44, 545–556. [Google Scholar]
  83. Swinkels, D.; Baes, M. The essential role of docosahexaenoic acid and its derivatives for retinal integrity. Pharmacol. Ther. 2023, 247, 108440. [Google Scholar] [CrossRef] [PubMed]
  84. Tabilo, C.; Squella, V.; Illesca, P.; Munoz, Y.; Farias, C.; Valenzuela, R. Impact of n-3 polyunsaturate fatty acids supplementation on visual health throughout the life cycle: A systematic review. Prostaglandins Leukot. Essent. Fat. Acids 2025, 206, 102686. [Google Scholar] [CrossRef] [PubMed]
  85. Sugasini, D.; Yalagala, P.C.R.; Subbaiah, P.V. Efficient Enrichment of Retinal DHA with Dietary Lysophosphatidylcholine-DHA: Potential Application for Retinopathies. Nutrients 2020, 12, 3114. [Google Scholar] [CrossRef]
  86. Sherratt, S.C.R.; Libby, P.; Budoff, M.J.; Bhatt, D.L.; Mason, R.P. Role of Omega-3 Fatty Acids in Cardiovascular Disease: The Debate Continues. Curr. Atheroscler. Rep. 2023, 25, 1–17. [Google Scholar] [CrossRef]
  87. Dinu, M.; Sofi, F.; Lotti, S.; Colombini, B.; Mattioli, A.V.; Catapano, A.L.; Casula, M.; Baragetti, A.; Wong, N.D.; Steg, P.G.; et al. Effects of omega-3 fatty acids on coronary revascularization and cardiovascular events: A meta-analysis. Eur. J. Prev. Cardiol. 2024, 31, 1863–1875. [Google Scholar] [CrossRef]
  88. Mason, R.P.; Jacob, R.F. Eicosapentaenoic acid inhibits glucose-induced membrane cholesterol crystalline domain formation through a potent antioxidant mechanism. Biochim. Biophys. Acta 2015, 1848, 502–509. [Google Scholar] [CrossRef]
  89. Sherratt, S.C.R.; Mason, R.P. Eicosapentaenoic acid inhibits oxidation of high density lipoprotein particles in a manner distinct from docosahexaenoic acid. Biochem. Biophys. Res. Commun. 2018, 496, 335–338. [Google Scholar] [CrossRef]
  90. Wang, C.C.; Shi, H.H.; Xu, J.; Yanagita, T.; Xue, C.H.; Zhang, T.T.; Wang, Y.M. Docosahexaenoic acid-acylated astaxanthin ester exhibits superior performance over non-esterified astaxanthin in preventing behavioral deficits coupled with apoptosis in MPTP-induced mice with Parkinson’s disease. Food Funct. 2020, 11, 8038–8050. [Google Scholar] [CrossRef]
  91. Hegde, P.S.; Agni, M.B.; Rai, P.; Sadananda, M.; Mirajkar, A.M.; Kumar, B.M.; Ranade, A.V.; Gowda, K.M.D. Unraveling the synergistic effects of Astaxanthin and DHA on perinatal undernutrition-induced oxidative stress and cognitive deficit. Learn. Behav. 2025, 1–18. [Google Scholar] [CrossRef] [PubMed]
  92. Hegde, P.S.; Agni, M.B.; Rai, P.; Upadhyay, S.S.; Aravind, A.; Keshava Prasad, T.S.; Gowda, K.M.D. Supplementation of diet with Astaxanthin and DHA prevents gestational and lactational undernourishment-induced metabolic derangements in dams: A metabolomic approach. J. Dev. Orig. Health Dis. 2024, 15, e30. [Google Scholar] [CrossRef] [PubMed]
  93. Ulven, S.M.; Holven, K.B. Comparison of bioavailability of krill oil versus fish oil and health effect. Vasc. Health Risk Manag. 2015, 11, 511–524. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Digestion, absorption, and transport of astaxanthin (Ast) and omega-3 PUFAs in the human body. (A). Digestion and absorption of Ast and omega-3 PUFAs. Ast-esters are digested into Ast and fatty acids (FAs) by esterases and lipases. Dietary lipids are digested into FAs, monoacylglycerols (MAGs) and lyso-phospholipids. The digestion products are absorbed into the enterocytes from micelles. (B). Transprot of Ast and omega-3 PUFAs. Lipid-soluble nutrients are delivered through lipoproteins, which are composed of a hydrophobic core surrounded by a surface layer of phospholipids (PLs), free cholesterol, and apolipoproteins. FAs are esterified into triacylglycerols (TAGs), which are then present in the core, while Ast is known to be incorporated into the surrounding PL monolayer. Chylomicrons (CMs) deliver dietary lipids and lipid-soluble nutrients to non-hepatic tissues, and are then taken up by the liver as CM remnants following lipoprotein lipase (LPL)-mediated hydrolysis of TAGs. The liver packages lipids and lipid-soluble nutrients into very low-density lipoprotein (VLDL). After LPL hydrolyzes TAGs, VLDL becomes low-density lipoprotein (LDL). Ast is taken up into cells through LDL receptor-mediated endocytosis of LDL particles or passive diffusion from high-density lipoprotein (HDL). Apo-A1: apoprotein-A1; Lyso-PLs: lysophospholipids.
Figure 1. Digestion, absorption, and transport of astaxanthin (Ast) and omega-3 PUFAs in the human body. (A). Digestion and absorption of Ast and omega-3 PUFAs. Ast-esters are digested into Ast and fatty acids (FAs) by esterases and lipases. Dietary lipids are digested into FAs, monoacylglycerols (MAGs) and lyso-phospholipids. The digestion products are absorbed into the enterocytes from micelles. (B). Transprot of Ast and omega-3 PUFAs. Lipid-soluble nutrients are delivered through lipoproteins, which are composed of a hydrophobic core surrounded by a surface layer of phospholipids (PLs), free cholesterol, and apolipoproteins. FAs are esterified into triacylglycerols (TAGs), which are then present in the core, while Ast is known to be incorporated into the surrounding PL monolayer. Chylomicrons (CMs) deliver dietary lipids and lipid-soluble nutrients to non-hepatic tissues, and are then taken up by the liver as CM remnants following lipoprotein lipase (LPL)-mediated hydrolysis of TAGs. The liver packages lipids and lipid-soluble nutrients into very low-density lipoprotein (VLDL). After LPL hydrolyzes TAGs, VLDL becomes low-density lipoprotein (LDL). Ast is taken up into cells through LDL receptor-mediated endocytosis of LDL particles or passive diffusion from high-density lipoprotein (HDL). Apo-A1: apoprotein-A1; Lyso-PLs: lysophospholipids.
Dietetics 04 00039 g001
Figure 2. Astaxanthin (Ast) and omega-3 PUFAs reduce oxidative stress and inflammation through complementary mechanisms. Ast reduces oxidative stress by acting as an antioxidant or by increasing nuclear factor erythroid 2-related factor 2 (NRF2) activity and inducing the expression of antioxidant enzymes. Very long-chain omega-3 polyunsaturated fatty acids (omega-3 PUFAs) suppress the canonical inflammatory signaling pathways, nuclear factor kappa B (NF-κB), and mitogen-activated protein kinases (MAPKs). Additionally, by antagonizing the proinflammatory Toll-like receptor 4 (TLR4) or directly acting as ligands for the free fatty acid receptor 4 (FFAR4), omega-3 PUFAs inhibit inflammatory responses. Inflammation is closely linked to oxidative stress, which may be at least in part due to an antagonistic crosstalk between NRF2 and NF-κB, explaining Ast-mediated inhibition of inflammation and omega-3 PUFA-mediated suppression of oxidative stress. IκB: inhibitor of kappa B; SPMs: specialized proresolving mediators; RONS: reactive oxygen and nitrogen species; AP-1: activator protein 1; GPX: glutathione peroxidase; SOD: superoxide dismutase; CAT: catalase; TNFα: tumor necrosis factor-alpha; IL-6: interleukin-6; IL-1β: interleukin-1 beta; COX2: cyclooxygenase 2; iNOS: inducible nitric oxide synthase.
Figure 2. Astaxanthin (Ast) and omega-3 PUFAs reduce oxidative stress and inflammation through complementary mechanisms. Ast reduces oxidative stress by acting as an antioxidant or by increasing nuclear factor erythroid 2-related factor 2 (NRF2) activity and inducing the expression of antioxidant enzymes. Very long-chain omega-3 polyunsaturated fatty acids (omega-3 PUFAs) suppress the canonical inflammatory signaling pathways, nuclear factor kappa B (NF-κB), and mitogen-activated protein kinases (MAPKs). Additionally, by antagonizing the proinflammatory Toll-like receptor 4 (TLR4) or directly acting as ligands for the free fatty acid receptor 4 (FFAR4), omega-3 PUFAs inhibit inflammatory responses. Inflammation is closely linked to oxidative stress, which may be at least in part due to an antagonistic crosstalk between NRF2 and NF-κB, explaining Ast-mediated inhibition of inflammation and omega-3 PUFA-mediated suppression of oxidative stress. IκB: inhibitor of kappa B; SPMs: specialized proresolving mediators; RONS: reactive oxygen and nitrogen species; AP-1: activator protein 1; GPX: glutathione peroxidase; SOD: superoxide dismutase; CAT: catalase; TNFα: tumor necrosis factor-alpha; IL-6: interleukin-6; IL-1β: interleukin-1 beta; COX2: cyclooxygenase 2; iNOS: inducible nitric oxide synthase.
Dietetics 04 00039 g002
Figure 3. Interactive actions of Ast and omega-3 PUFAs in health and disease. Ast and omega-3 PUFAs are known to reduce disease risks by suppressing oxidative stress and inflammation through complementary mechanisms. Therefore, combined intake of both bioactive nutrients may exert additive or synergistic effects on health. Omega-3 PUFAs: very long-chain omega-3 polyunsaturated fatty acids; EPA: eicosapentaenoic acid; DPA: docosapentaenoic acid; DHA: docosahexaenoic acid. Structures of Ast isomers and omega-3 PUFAs are derived from PubChem (https://pubchem.ncbi.nlm.nih.gov/), accessed on 10 July 2025.
Figure 3. Interactive actions of Ast and omega-3 PUFAs in health and disease. Ast and omega-3 PUFAs are known to reduce disease risks by suppressing oxidative stress and inflammation through complementary mechanisms. Therefore, combined intake of both bioactive nutrients may exert additive or synergistic effects on health. Omega-3 PUFAs: very long-chain omega-3 polyunsaturated fatty acids; EPA: eicosapentaenoic acid; DPA: docosapentaenoic acid; DHA: docosahexaenoic acid. Structures of Ast isomers and omega-3 PUFAs are derived from PubChem (https://pubchem.ncbi.nlm.nih.gov/), accessed on 10 July 2025.
Dietetics 04 00039 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lee, M.-J. Interactions of Astaxanthin and Omega-3 Fat in Health and Disease. Dietetics 2025, 4, 39. https://doi.org/10.3390/dietetics4030039

AMA Style

Lee M-J. Interactions of Astaxanthin and Omega-3 Fat in Health and Disease. Dietetics. 2025; 4(3):39. https://doi.org/10.3390/dietetics4030039

Chicago/Turabian Style

Lee, Mi-Jeong. 2025. "Interactions of Astaxanthin and Omega-3 Fat in Health and Disease" Dietetics 4, no. 3: 39. https://doi.org/10.3390/dietetics4030039

APA Style

Lee, M.-J. (2025). Interactions of Astaxanthin and Omega-3 Fat in Health and Disease. Dietetics, 4(3), 39. https://doi.org/10.3390/dietetics4030039

Article Metrics

Back to TopTop