Open Access

Antioxidant and anti‑inflammatory mechanisms of action of astaxanthin in cardiovascular diseases (Review)

  • Authors:
    • Carolina Parga Martins Pereira
    • Ana Carolina Remondi Souza
    • Andrea Rodrigues Vasconcelos
    • Pietra Sacramento Prado
    • José João Name
  • View Affiliations

  • Published online on: November 4, 2020     https://doi.org/10.3892/ijmm.2020.4783
  • Pages: 37-48
  • Copyright: © Pereira et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cardiovascular diseases are the most common cause of mortality worldwide. Oxidative stress and inflammation are pathophysiological processes involved in the development of cardiovascular diseases; thus, anti‑inflammatory and antioxidant agents that modulate redox balance have become research targets so as to evaluate their molecular mechanisms of action and therapeutic properties. Astaxanthin, a carotenoid of the xanthophyll group, has potent antioxidant properties due to its molecular structure and its arrangement in the plasma membrane, factors that favor the neutralization of reactive oxygen and nitrogen species. This carotenoid also has prominent anti‑inflammatory activity, possibly interrelated with its antioxidant effect, and is also involved in the modulation of lipid and glucose metabolism. Considering the potential beneficial effects of astaxanthin on cardiovascular health evidenced by preclinical and clinical studies, the aim of the present review was to describe the molecular and cellular mechanisms associated with the antioxidant and anti‑inflammatory properties of this carotenoid in cardiovascular diseases, particularly atherosclerosis. The beneficial properties and safety profile of astaxanthin indicate that this compound may be used for preventing progression or as an adjuvant in the treatment of cardiovascular diseases.

1. Introduction

According to data obtained from the World Health Organization, cardiovascular diseases accounted for 31% of all causes of death in 2016, and they are considered as the leading cause of mortality worldwide (1). One of the risk factors for cardiovascular diseases is metabolic syndrome, a multifactorial entity that includes variables such as obesity, dyslipidemia, hypertension and glucose metabolism dysfunctions (2). Evidence connects these variables to increased oxidative stress, with mitochondrial dysfunction, activation of enzymes that produce reactive oxygen and nitrogen species (RONS), and impairment of the activity of antioxidant systems acting as the main triggering mechanisms (2-4).

As oxidative stress and inflammation are interrelated and contribute to the initial events of cardiovascular diseases, antioxidants that modulate redox balance, such as astaxanthin, may be considered as important regulators of inflammatory responses (5) and have become the focus of research to evaluate whether and how they prevent these diseases. Astaxanthin is closely associated with other well-known carotenoids, such as beta-carotene, lutein and zeaxanthin, sharing various of the physiological and metabolic functions attributed to these compounds (6). The two oxygenated groups, hydroxyl (OH) and carbonyl (C=O), in each of its ionone rings explains some of the unique characteristics of astaxanthin, such as its more potent anti-oxidant activity and polar configuration when compared to other carotenoids (6-8). In addition to its antioxidant action, astaxanthin has anti-inflammatory properties and the ability to modulate lipid and glucose metabolism (6,7,9), which are beneficial in the cardiovascular system, preventing disorders such as atherosclerosis, arterial hypertension and dyslipidemia (10-20). Given the extensive evidence supporting its health-promoting properties and safety, astaxanthin was approved as a nutraceutical by the United States Food and Drug Administration in 1999 (21).

Considering the safety of using astaxanthin as a nutraceutical and the scientific evidence of its beneficial effects on cardiovascular physiology, the aim of the present review was to describe the molecular and cellular mechanisms underlying the antioxidant and anti-inflammatory role of astaxanthin in the prevention of cardiovascular diseases.

2. Mechanisms of action of astaxanthin

Antioxidant effect

Cell membrane systems are particularly vulnerable to RONS attacks due to their content of polyun-saturated fatty acids (PUFAs) and their metabolic activities, which endogenously generate other oxidizing metabolites (22).

Astaxanthin protects cell membranes against RONS and oxidative damage. Due to its chemical structure, its polar groups overlap the polar regions of the cell membrane, while the central non-polar region of the molecule fits into the inner non-polar region of the membrane. Thus, this carotenoid may take on a transmembrane alignment in biological membranes, helping to maintain the membrane structure and decrease membrane fluidity, and acting as an antioxidant (17,23,24).

Astaxanthin scavenges RONS and other reactive species (sulfur and carbon) directly, both by donating electrons and by bonding with the free radical to form a non-reactive product (25). In addition, the presence of a series of conjugated bonds in the central non-polar region of astaxanthin enables the molecule to remove free radicals (high-energy electrons) from the cell interior by transporting them along its own carbon chain, resembling a 'lightning rod' for these electrons, so that these are neutralized by other antioxidants located outside the cell membrane, such as vitamin C (25).

The increased susceptibility of membrane lipids and low-density lipoprotein (LDL) to oxidation may trigger the formation of thrombi and the development of atherosclerosis (5). One of the reactive species that induces lipid peroxidation and LDL oxidation is peroxynitrite (ONOO) (26,27), which is neutralized by astaxanthin to form 15-nitroastaxanthin, a compound that also has important antioxidant action (28).

The LDL oxidation time in the presence of astaxanthin has been analyzed in vitro and ex vivo. In the in vitro assays, astaxanthin prolonged LDL oxidation in a dose-dependent manner, in addition to being more effective compared with lutein and α-tocopherol. In turn, the blood samples of individuals who were supplemented daily with 1.8, 3.6, 14.4, or 21.6 mg astaxanthin for 14 days evidenced a significant delay in LDL oxidation when compared to samples collected before supplementation, the greatest effect being obtained with the dose of 14.4 mg (oxidation time increased by 5.0, 26.2, 42.3 and 30.7% with 1.8, 3.6, 14.4 and 21.6 mg astaxanthin, respectively) (Table I) (10). Thus, it was demonstrated that the intake of astaxanthin delayed LDL oxidation, one of the key factors involved in the process of atherosclerosis.

Table I

Clinical studies that have demonstrated the potential beneficial effects of oral astaxanthin supplementation on cardiovascular physiology.

Table I

Clinical studies that have demonstrated the potential beneficial effects of oral astaxanthin supplementation on cardiovascular physiology.

Study typeSubjects (age)Intervention (no. of subjects per group)Mechanism of action evaluatedMain findings(Refs.)
Open-label24 healthy volunteers (mean, 28.2±7.8 years)Control (n=6)a; AST 1.8 mg/day (n=5); AST 3.6 mg/day (n=5); AST 14.4 mg/day (n=3);AST 21.6 mg/day (n=5); 14 daysAntioxidant effectDelayed LDL oxidation time(10)
Randomized, double-blind, placebo-controlled27 overweight and obese adults (BMI>25 kg/m2) (20-55 years)Placebo (n=13); AST 20 mg/day (n=14); 12 weeksAntioxidant effectReduced LDL, ApoB and ApoA1/ApoB ratio relative to baseline; increased TAC and SOD compared to baseline; reduced lipid peroxidation biomarkers (MDA and ISP) compared to baseline.(11)
Randomized, double-blind, placebo-controlled30 healthy individuals (50-69 years)Placebo (n=10); AST 6 mg/day(n=10); AST 12 mg/day (n=10); 12 weeksAntioxidant effectReduced phospholipid hydroperoxide levels in erythrocytes(12)
Randomized, double-blind, placebo-controlled39 healthy men (19-33 years)Placebo (n=19); AST 8 mg/day (n=20); 3 monthsAntioxidant effectReduced plasma lipid peroxidation, particularly 12-hydroxy and 15-hydroxy fatty acids(13)
Open-label, uncontrolled study20 healthy postmenopausal women with high oxidative stress levels (mean, 55.7±4.8 years)AST 12 mg/day (n=20); 8 weeksAntioxidant effectLowered blood pressure; increased antioxidant capacity; reduced vascular resistance in lower limbs and serum adiponectin(14)
Randomized39 smokers (≥20 cigarettes per day) and 39 non-smokers (21-43 years)Control (n=39)b; AST 5 mg/day (n=13); AST 20 mg/day (n=13); AST 40 mg/day (n=13); 3 weeksAntioxidant effectReduced MDA and ISP; increased SOD and TAC(15)
Randomized, double-blind23 overweight (25<BMI≤29.9 kg/m2) and obese (BMI>30 kg/m2) subjects (mean, 25.1±3.7 years)Control (n=10)c; AST 5 mg/day (n=12); AST 20 mg/day (n=11); 3 weeksAntioxidant effectReduced MDA and ISP; increased SOD and TAC(16)
Randomized, double-blind, placebo-controlled61 healthy subjects with triglyceride levels 120-200 mg/dl (25-60 years)Placebo (n=15); AST 6 mg/day (n=15); AST 12 mg/day (n=15); AST 18 mg/day (n=16); 12 weeksLipid metabolismReduced triglyceride levels; increased HDL; increased adiponectin(20)
Randomized, double-blind, placebo-controlled42 healthy young women (20-22 years)Placebo (n=14); AST 2 mg/day (n=14); AST 8 mg/day (n=14); 8 weeks Anti-inflammatoryand antioxidant effectIncreased total number of T and B lymphocytes; increased cytotoxic activity of natural killer cells; reduced biomarkers of oxidative damage 8-hydroxy-2′-deoxyguanosine and C-reactive protein(17)
Single-blind20 healthy adult men (37-67 years)Placebo (n=10); AST 6 mg/day (n=10); 10 daysBlood rheologyReduced blood transit time(18)
Randomized, double-blind, placebo-controlled43 participants with type 2 diabetes (46-62 years)Placebo (n=21); AST 8 mg/day (n=22); 8 weeksLipid and glucose metabolismIncreased adiponectin; reduced visceral fat mass, triglycerides, VLDL, fructosamine and systolic blood pressure(19)

a Individuals without supplementation.

b Non-smoking individuals without supplementation.

c Individuals with normal body weight (20<BMI≤24.9 kg/m2) and without supplementation; AST, astaxanthin; BMI, body mass index; LDL, low-density lipoprotein; HDL, high-density lipoprotein; VLDL, very low-density lipoprotein; MDA, malondialdehyde; SOD, superoxide dismutase; ISP, isoprostane; TAC, total antioxidant capacity.

LDL oxidation is also associated with the development of endothelial dysfunction in patients with diabetes mellitus, which increases the risk of cardiovascular complications (29). Endothelial dysfunction consists of impaired vessel relaxation dependent on endothelial factors, such as the production of nitric oxide (NO) by endothelial NO synthase (eNOS) (30). One of the pathways responsible for this type of dysfunction is the binding of oxidized LDL to its endothelial receptor, lectin-like ox-LDL receptor 1 (LOX-1), thus favoring oxidative stress, which leads to increased lipid peroxidation and eNOS inactivation (31). Supplementation with 10 mg/kg astaxanthin for 42 days in a diabetic rat model increased artery relaxation by significantly lowering oxidized the levels of LDL, LOX-1 receptor and lipid peroxidation in the aorta, in addition to increasing eNOS, demonstrating that astaxanthin may have therapeutic potential for the treatment of endothelial dysfunction in diabetic patients (32).

Erythrocytes have a large amount of PUFAs and high concentrations of oxygen and ferrous ions (Fe2+), which makes these cells more susceptible to oxidative changes in the lipid bilayer, leading to compromised cell stability, oxygen transport and blood rheological properties (33). The oxidation of erythrocytes is associated with both the formation of atheromas and the occurrence of intraplate hemorrhage during the development of atherosclerosis (34). As regards lipid peroxidation in erythrocytes, daily supplementation with 6 or 12 mg of astaxanthin for 12 weeks in healthy individuals demonstrated that this carotenoid is incorporated and distributed into these blood cells. When incorporated into erythrocytes, astaxanthin exerted antioxidant effects on the cell membrane by significantly reducing the levels of phospholipid hydroperoxides, which are the primary products of phospholipid oxidation (14.9 pmol/ml for the placebo group and 8.0 and 9.7 pmol/ml for the 6 and 12 mg astaxanthin groups, respectively). In that study, the two doses of astaxanthin used had similar effects when compared to placebo, suggesting that the intake of 6 mg of this carotenoid is sufficient to inhibit oxidative stress in erythrocytes (Table I) (12). A significant reduction in oxidative damage by lipid peroxidation was also observed, as the plasma levels of 12- and 15-hydroxy fatty acids (P=0.048 and P=0.047, respectively) decreased after 3 months of supplementation with 8 mg astaxanthin in healthy men (Table I) (13).

Blood rheology is important for cardiovascular homeostasis. Astaxanthin was shown to reduce blood transit time in a hypertensive rat model (35) as well as in humans (18). In the latter study, individuals receiving supplementation with 6 mg astaxanthin for 10 days had a significantly faster blood transit time compared with that before supplementation and with that in the placebo group (47.6±4.2 vs. 54.2±6.7 sec for the treated and placebo groups, respectively; P<0.05; Table I) (18). One hypothesis for the improvement of blood rheology by astaxanthin is its antioxidant effect on the intra- and extracellular environment and the consequent increase in the flexibility of the erythrocyte membrane conferred by the structural arrangement of astaxanthin in the membrane.

Other factors that affect the blood flow velocity, such as plasma viscosity and vasodilation, affect peripheral vascular resistance and may contribute to hypertension and its main cardiac complication, myocardial hypertrophy (36). Studies of spontaneously hypertensive rats (SHR) reported that astaxanthin supplementation significantly reduced systolic pressure and induced significant histological changes in the aorta associated with decreased vascular stiffness and blood pressure (37-39). This response was caused by increased endothelial cell-dependent vasodilation due to the greater bioavailability of NO, as well as the remodeling of the arteries. The increase in NO was caused by the reduced production of superoxide anion radicals released by NADPH oxidase, which is one of the antioxidant effects of astaxanthin (37,38). Chen et al (2020) reported that this carotenoid also participated in the remodeling of the smooth muscle cells of the vessels, reducing their proliferation and the damage caused by oxidative stress. Astaxanthin lowered RONS levels by increasing the activity of antioxidant enzymes and regulating mitochondrial dynamics, mitophagy and mitochondrial biogenesis, which are important for the maintenance of mitochondrial and cellular metabolism (39).

The high bioavailability of NO due to lower oxidative stress promoted by astaxanthin was also associated with its antithrombogenic effects. In a stroke-prone SHR model, astax-anthin significantly downregulated the oxidative stress marker 8-hydroxy-2′-deoxyguanosine (8-OHdG) in the urine, lowered systolic blood pressure and suppressed thrombogenesis in the cerebral veins. The observed antithrombogenic effect may have been due to vasodilation and inhibition of platelet aggregation caused by an increased bioavailability of NO (40). In a murine model of thrombosis treated with astaxanthin in the form of the CDX-085 prodrug, an increase of ~20% in the blood flow of the carotid artery was observed before the occurrence of endothelial dysfunction, as was a delay in the formation of occlusive thrombi. These results were due to the increase in NO and the decrease in ONOO. The authors suggested that the increase in blood flow was due to vasodilation caused by increased release of NO by endothelial cells and reduced platelet activation triggered by the antioxidant effect (41).

Some of the vascular benefits promoted by the antioxidant effect of astaxanthin were reported in an open study of 20 postmenopausal women who had a high rate of oxidative stress (14). After 8 weeks of supplementation with 12 mg astaxanthin, there was a significant reduction of 4.64 and 6.93% in the systolic and diastolic pressure values, respectively, possibly resulting from the reduction in the vascular tone due to the action of the carotenoid in the endothelium. Reduced vascular resistance in the lower limbs (3.7% increase in the ankle-brachial pressure index), a 4.58% increase in antioxidant capacity, and improvement of some physical and mental symptoms, such as tired eye sensation and difficulty sleeping, were also observed (14).

In addition to the protective role of astaxanthin in the lipid oxidation process, this carotenoid affects the activity of antioxidant enzymes involved in lipid metabolism, such as thioredoxin reductase (TrxR) and paraoxonase-1. TrxR is an antioxidant enzyme involved in the reduction of thioredoxin, lipid hydroperoxides and hydrogen peroxide (42). A previous study demonstrated that thioredoxin in its oxidized form was associated with the degree of severity of chronic heart failure and with the resulting oxidative stress (43). Paraoxonase-1 binds to serum high-density lipoprotein (HDL) and is responsible for protecting both LDL and HDL from oxidation, as well as for breaking down oxidized lipids (44). The effect of astaxanthin on these two enzymes was evaluated in rabbits fed a cholesterol-rich diet (45). The authors found that astaxanthin (100 and 500 mg/100 g of feed) reduced the amount of oxidized protein, possibly due to changes in the activities of TrxR and paraoxonase-1, but the in vitro evaluation only demonstrated the direct action of this molecule on TrxR (45).

Choi et al (11,16) also conducted two randomized and double-blind clinical studies on overweight or obese individuals to demonstrate the antioxidant effects of astaxanthin (Table I). In one study, volunteers who received 5 and 20 mg astaxanthin for 3 weeks exhibited lower oxidative stress biomarkers associated with lipid peroxidation compared with prior to treatment, with a 34.6 and 35.2% reduction in malondialdehyde (MDA) levels, and 64.9 and 64.7% reduction in isoprostane (ISP) levels, respectively. An increase in the activity of the antioxidant defense system was also observed, with a 193 and 194% increase in superoxide dismutase (SOD) and a 121 and 125% increase in total antioxidant capacity at the doses of 5 and 20 mg, respectively, compared with the data prior to treatment (16). No significant differences were observed between the results obtained with the two doses, indicating that the clinical effects of this carotenoid are not dose-dependent. Later, the same authors analyzed lipid profile, oxidative stress and antioxidant system parameters (11). After 12 weeks of supplementation with 20 mg astaxanthin, the same results in oxidative stress and the antioxidant system as in the previous study were observed. Regarding the lipid profile, there was a significant reduction of 10.4% in the LDL concentration, 7.59% in ApoB and 8.22% in the ApoA1/ApoB ratio (considered an index of the risk of heart attack) compared to the placebo group values (11). Therefore, these studies demonstrated that astaxanthin reduces oxidative stress and modulates the lipid profile in overweight and obese individuals, mitigating the risk of developing cardiovascular diseases.

Astaxanthin also has potent detoxifying and antioxidant effects in smokers (15). The free radicals induced by smoking have been strongly associated with increased oxidative stress, contributing to the increased susceptibility of smokers to the pathogenesis of cardiovascular diseases. This group of individuals requires a higher daily intake of antioxidants compared with non-smokers to reduce the consequences of prolonged exposure to toxins present in cigarettes. After 3 weeks, supplementation with different doses of astaxanthin (5, 20 and 40 mg) in active smokers prevented oxidative damage by suppressing lipid peroxidation and stimulating the activity of the antioxidant system (Table I) (15). This effect was confirmed by the significant reduction in serum MDA and ISP levels and the increased SOD activity and total antioxidant capacity in the three astaxanthin groups compared with the indices prior to treatment (15). The authors also observed that the serum concentration of astaxanthin in the groups treated with 20 and 40 mg was similar, showing that there was saturation of its absorption and that smaller doses, such as 5 mg, may have the necessary antioxidant effect for these individuals. However, placebo-controlled studies with larger groups and longer interventions may help determine the optimal dosage for smokers.

Several preclinical studies have demonstrated that astaxanthin also exerts an indirect antioxidant effect by activating transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2), and increasing the expression of its antioxidant target genes, such as phase II biotransformation enzymes (46-52). A study with a model of coronary microembolization in rats revealed that supplementation with astaxanthin drastically attenuated the induction of cardiac dysfunction, myocardial infarction and cardiomyocyte apoptosis, which was associated with the suppression of oxidative stress via activation of Nrf2/heme oxygenase-1 signaling (47).

Thus, astaxanthin may accumulate in the blood plasma and, through its antioxidant action, it helps reduce the levels of RONS responsible for LDL oxidation and lipid peroxidation; it increases the bioavailability of NO, enabling its vasodilator and antithrombogenic effects; it increases the activity of antioxidant enzymes; and it ensures the stability of blood rheological properties, thus avoiding the loss of erythrocyte flexibility and the increase in plasma viscosity, factors that affect the blood flow velocity. These actions of astaxanthin against early events of atherosclerotic plaque formation and arterial dysfunction may delay the progression of cardiovascular diseases (Fig. 1).

Anti-inflammatory effect

Inflammation plays an important role in the development of cardiovascular diseases and other comorbidities, such as hypertension, hypercholesterolemia, type 2 diabetes, chronic kidney disease and obesity (53). Astaxanthin exerts a marked anti-inflammatory effect, which may be interrelated with its antioxidant effect and contributes to physiological changes that benefit cardiovascular function (Fig. 1) (53-58).

Atherosclerosis is a degenerative and chronic disease that affects large- and medium-caliber arteries. Atherogenesis, the initial phase of the atherosclerotic process, is character-ized by the accumulation of LDL in the subendothelial layer of the vascular wall, which is responsible for inflammation mediated by the innate and adaptive immune responses (59). The anti-inflammatory effects promoted by astaxanthin are evidenced in its role in atherosclerosis prevention, as will be detailed below.

The epitopes generated from enzymatic or non-enzymatic oxidation of LDL are the main damage-associated molecular patterns recognized by macrophages and are responsible for the onset of the inflammatory cascade, with the release of cytokines and chemokines that recruit more resident vascular macrophages and monocytes from the blood. Macrophages bind to oxidized LDL via scavenger receptors, such as SR-A, SR-B2 (CD36) and LOX-1 (59). The expression of these receptors is controlled by nuclear factor-κB (NF-κB), the main mediator of the inflammatory response, which is activated by pattern recognition receptors and pro-inflammatory cytokines (60). In inflammatory states, macrophages produce excessive amounts of pro-inflammatory mediators, such as cytokines, chemokines, NO, cyclooxygenase-2 (COX-2) and matrix metalloproteinases (MMPs). MMPs are responsible for the degradation of most extracellular matrix proteins and mediate the tissue remodeling associated with atherosclerosis (5).

In vitro and in vivo studies have evaluated the effect of astaxanthin on the formation of atherosclerotic plaques (54-58,61-82). Supplementation with 10 µM astaxanthin significantly reduced the expression of the SR-A and CD36 scavenger receptors in the THP-1 macrophage line and reduced the total activity of MMPs, as reflected by reduced protein expression of MMP-9 and MMP-2 and of the mRNA levels of five MMPs (62). Astaxanthin at this concentration also reduced the gene expression of pro-inflammatory markers, such as interleukin (IL)-1β, IL-6, tumor necrosis factor-α (TNF-α), inducible nitric oxide synthase (iNOS) and COX-2 (62). These results corroborate those of other studies indicating that astaxanthin reduces the expression of pro-inflammatory mediators in macrophages (62-65) and other cell types, such as microglia, endothelial vascular cells and human neutrophils (66-69).

The significant decreases in the levels of MMPs and proinflammatory cytokines may result from the suppression of the NF-κB transcription factor by astaxanthin (54-58,62,64,68,70,71). NF-κB is frequently activated at inflammation sites associated with various pathologies, particularly cardiovascular diseases, in the etiology of which the increased expression of its pro-inflammatory target genes plays a key role (72). The inflammatory pathway of NF-κB is, at least in part, regulated by oxidative stress (72). Astaxanthin inhibits the activity of IκB kinase, a complex responsible for the control of NF-κB activation. This maintains NF-κB inactive in the cell cytoplasm, and its pro-inflammatory target genes, such as TNF-α, IL-1β and iNOS, are downregulated (64).

Cholesterol uptake is balanced by the transfer of this molecule from macrophages to free apolipoproteins A1 or to HDL, the latter being responsible for the reverse cholesterol transport process. When cholesterol uptake exceeds cholesterol efflux in macrophages, lipid droplets accumulate in the cytoplasm, forming foam cells, the main markers of atherosclerosis (Fig. 2) (59). The progression of cholesterol accumulation may lead to its precipitation in the form of crystals, which activate the inflammasome, leading to cell death by apoptosis or necrosis (83). The atherosclerotic plaque is separated from the bloodstream by a fibrous layer, which, upon rupture, initiates intraluminal thrombosis, the initial event of stroke and other coronary syndromes (59).

Reverse cholesterol transport consists in HDL removing excess cholesterol from peripheral tissues and transporting it to the liver, where it is degraded by bile juice and excreted in the feces, thus preventing the accumulation of cholesterol in macrophages (79). Both the liver and intestine synthesize apolipoprotein A-I (apoA-I) and apoA-II in the plasma, which incorporate free cholesterol and phospholipids through the ATP-binding cassette A1 (ABCA1) hepatic transporter, originating from nascent HDL. In peripheral tissues, nascent HDL molecules recruit free cholesterol from foam cells via the macrophage ABCA1 transporter (80). Of note, this reverse cholesterol transport was also observed in the lymphatic system, which is largely responsible for the removal of cholesterol from different tissues (79). Finally, mature HDL can transport cholesterol directly to the liver via the SR-B1 scavenger receptor or can transfer cholesteryl esters to very low-density lipoprotein (VLDL) through the cholesteryl ester transfer protein (81). These lipoproteins are absorbed in the liver by their specific receptors, which is likely the predominant pathway in humans. Once in the liver, cholesterol is secreted into the bile via the ABCG5 and ABCG8 transporters. Some of these molecules can be reabsorbed by the intestine and reach the bloodstream again, while the rest is excreted in the feces (80).

In atherosclerosis, apolipoproteins are oxidized by the enzyme myeloperoxidase, which is expressed in macrophages during the inflammatory process, compromising cholesterol efflux via ABCA1 (81). In individuals with heart disease, elevated levels of apoA-I modified by myeloperoxidase were identified, and their HDL molecules were dysfunctional in performing reverse cholesterol transport. In addition, an association was observed between increased cholesterol efflux via ABCA1 and reduced risk of cardiovascular diseases (OR=0.30; 95% CI: 0.14-0.66; P<0.003) (82). Thus, the oxidation of apolipoproteins by macrophage myeloperoxidase is a determining factor in HDL dysfunction in cholesterol transport and, therefore, in the risk of cardiovascular diseases.

The effects of astaxanthin on reverse cholesterol transport have been demonstrated in vivo. In wild-type and ApoE−/− mice, astaxanthin increased cholesterol efflux from peripheral tissues to the liver and its subsequent excretion in feces (61). In addition, in the ApoE−/− model, this carotenoid promoted a significant decline in plasma total cholesterol, triglycerides and non-HDL cholesterol, and reduced the atherosclerotic plaque area of the aortic sinus and the cholesterol concentration in the aorta compared to controls. Thus, astaxanthin may exert antiatherosclerotic effects by increasing the activity of the reverse cholesterol transport pathway, but the molecular mechanisms underlying this action remain elusive (61).

In addition to its function in reverse cholesterol transport, astaxanthin is involved in certain lipid metabolism steps, a finding corroborated by a randomized, placebo-controlled clinical study of 61 adult individuals with moderate hyperlipidemia (Table I) (20). In that study, daily supplementation with 6, 12 or 18 mg astaxanthin for 12 weeks led to an improvement in the lipid profile of the patients. Triglycerides were reduced by 25.2 and 23.8% (P<0.05) by doses of 12 and 18 mg, respectively, whereas HDL was increased by 10.6% (P<0.05) and 15.4% (P<0.01) by doses of 6 and 12 mg, respectively. Doses of 12 and 18 mg also significantly increased serum adiponectin (P<0.01 and P<0.05, respectively), a protein secreted by adipocytes with important functions in the cardiovascular and endocrine systems associated with its anti-inflammatory, atheroprotective and insulin-sensitizing actions (20).

Inflammation is also involved in the pathophysiology of metabolic syndrome, a multifactorial disorder associated with glucose and lipid metabolism disorders. This disease has risk factors that are also strongly associated with the development of cardiovascular complications, including type 2 diabetes, dyslipidemia, hypertension and abdominal fat deposition (84). In this context, astaxanthin has been found to be promising in the improvement of glucose and lipid metabolism in a randomized, placebo-controlled clinical study with 43 diabetic patients aged 46-62 years (Table I) (19). In agreement with a previous clinical study (20), supplementation with astaxanthin (8 mg/day for 8 weeks) significantly increased serum adiponectin (47±14 vs. 45±13 and 36±15 µg/ml compared with placebo and base-line, respectively; P<0.05) and improved the lipid profile of the patients, as shown by the reductions in the levels of triglycerides (128±52 vs. 150±85 and 156±90 mg/dl compared with placebo and baseline, respectively; P<0.05) and VLDL (27±16 vs. 31±16 mg/dl compared with placebo; P<0.05). Furthermore, astaxanthin marginally reduced fasting glucose levels (8.3±2.7 vs. 9.4±3.2 mmol/l compared with placebo; P=0.057) and significantly increased serum fructosamine levels (5.8±3.8 vs. 7.32±4.31 and 7.36±4.2 µmol/l compared with placebo and baseline, respectively; P<0.05), an important marker in the control of diabetes that reflects the mean concentration of blood glucose. Patients receiving astaxanthin supplementation also exhibited lower visceral fat deposition (11.2±3.4 vs. 11.85±3.8% compared with placebo; P<0.05) and systolic blood pressure (132±18 vs. 133±19 and 143±27 mmHg compared with placebo and baseline, respectively; P<0.05) (19).

Disorders characterized by ischemia/reperfusion, including myocardial infarction, stroke and peripheral vascular disease, are among the most frequent causes of morbidity and mortality worldwide (85). Ischemia/reperfusion is a complex inflammatory process associated with high levels of oxidative stress in the affected tissue (86). In rodents with hepatic lesions induced by ischemia/reperfusion, astaxanthin not only reduced oxidative stress and histopathological damage (87,88) but also exerted a significant anti-inflammatory effect, attenuating the release of inflammatory cytokines through the mitogen-activated protein kinase (MAPK) pathway (88,89). Furthermore, astaxanthin exerted an anti-inflammatory and antioxidant effect in the context of myocardial injury due to ischemia/reperfusion in rabbits by significantly reducing the activation of the complement system associated with the reduced deposition of C-reactive protein and the membrane attack complex in the injured area of the myocardium (90).

In mice with non-alcoholic steatohepatitis (NASH) induced by a high-lipid diet, supplementation with astaxanthin (0.02% in the diet, ~20 mg/kg body weight) significantly improved several liver parameters: it reduced liver inflammation, decreased the proportion of pro-inflammatory M1-type macrophages, reduced stellate cell activation, and attenuated liver fibrosis, the accumulation and peroxidation of hepatic lipids and insulin resistance (91). Additionally, astaxanthin was more effective in preventing and treating NASH and improving liver inflammation and fibrosis compared with vitamin E (standard NASH treatment). The same study also corroborated the potential of astaxanthin to improve NASH in 12 individuals receiving this carotenoid as a supplement (12 mg/day; control: placebo) for 24 weeks (91).

The effects of astaxanthin on the relief of liver injury was shown to be correlated to its positive effects on the intestinal microbiota and consequent reduction of inflammation (92). In fact, a growing body of evidence indicates that gut microbiota plays a key role in the pathogenesis of inflammatory disorders and cardiovascular diseases, and alterations in its composition (dysbiosis) have been associated with heart failure, hypertension, atherosclerosis and metabolic syndrome (93-95). Several recent in vivo studies revealed that astaxanthin supplementation improved gut microbiota composition, which may contribute to its local and systemic anti-inflammatory and antioxidant effects (92,96-99). The beneficial effect of astaxanthin on gut microbiota has been shown to be correlated with the mitigation of cardiovascular disease-related pathologies and risk factors, such as obesity (100), insulin resistance (99) and alcoholic fatty liver disease (92).

The anti-inflammatory and antioxidant effects of astaxanthin were also confirmed by a randomized clinical trial in 42 healthy young women receiving placebo or astaxanthin at 2 or 8 mg/day (Table I) (17). Compared with placebo, after 8 weeks of treatment, 2 mg astaxanthin significantly lowered the levels of the plasma inflammatory marker C-reactive protein (unspecified values; P<0.05). Astaxanthin improved the immune responses of the participants, as evidenced by the increased cytotoxic activity of natural killer cells (8 mg dose, 67.9±3.0 vs. 57.8±2.7% lysis; P<0.05), levels of T and B lymphocytes (2 mg dose, 75.7±1.6 vs. 70.6±1.5 and 13.1±0.5 vs. 10.7±0.5%, respectively; P<0.05), and the production of interferon (IFN)-γ and IL-6 (dose of 8 mg, 9.55 vs. 4.68 and 25.2 vs. 13.6 pg/ml, respectively; P<0.05). Finally, starting at 4 weeks, both doses drastically reduced the plasma levels of 8-OHdG (unspecified values; P<0.01), a biomarker of oxidative DNA damage (17).

Although macrophages are the main type of immune cell found in atherosclerotic plaques, T lymphocytes also contribute to the development of the disease (101). In fact, the inflammatory response mediated by T lymphocytes plays a crucial role in the etiology of cardiovascular diseases, contributing to atherosclerosis, heart failure and myocardial infarction (102-108). For example, T helper cells can be activated by LDL particles in the arterial wall and trigger inflammation through an autoimmune response, contributing to the development of atherosclerotic plaques (106,108,109). Similarly, self-reactive T helper cells may target cardiomyocytes, contributing to the development of heart failure (103).

Astaxanthin was shown to be effective not only in preventing oxidative stress in T lymphocytes (110-115), but also in modulating their activity (115-123). In the aforementioned clinical study on healthy young women (17), astaxanthin supplementation stimulated mitogen-induced lymphoproliferation and increased the subpopulation of T lymphocytes, without changing the populations of T killer or T helper cells, as well as increased the response to tuberculin, an indicator of T lymphocyte function. In a mouse model of NASH, astaxanthin reduced T helper and T killer cell recruitment to the liver, contributing to the improvement of inflammation and insulin resistance (91). Supplementation for 45 days with fish oil containing astaxanthin (1 mg/kg of body weight) significantly reduced the proliferative capacity of T lymphocytes in response to mitogens and RONS production when compared with fish oil alone (115). In in vitro studies with peripheral blood mononuclear cells from patients with asthma and allergic rhinitis, it was demonstrated that astaxanthin significantly suppressed the activation of T lymphocytes induced by phytohemagglutinin (116,122). Another in vitro and ex vivo study with cultured lymphocytes demonstrated that astaxanthin stimulated their immune response and increased the production of IL-2 and IFN-γ, without inducing cytotoxicity (117). The administration of astaxanthin in mice prevented renal fibrosis by mechanisms involving stimulation of T killer cell recruitment and increased production of IFN-γ (118). In cats, astaxanthin increased the immune response mediated by total T lymphocytes and T helper cells (119).

Therefore, astaxanthin was shown to exert a clear modulatory effect on T lymphocytes, overall improving their immune response or downregulating their potentially pathological immune activation. However, the role of T lymphocyte modulation by astaxanthin in the risk and progression of cardiovascular diseases remain to be fully elucidated.

In summary, inflammation plays a key role in the pathophysiology of cardiovascular diseases and their risk factors, while astaxanthin exerts beneficial anti-inflammatory effects. The mechanism of action of this carotenoid involves inhibition of the NF-κB and MAPK signaling pathways, which suppresses the inflammatory process and stimulates reverse cholesterol transport, thereby attenuating the formation of foam cells (Fig. 1).

3. Clinical evidence of the prevention of cardiovascular diseases by astaxanthin

The potential beneficial effects of oral astaxanthin supplementation on cardiovascular physiology were evidenced in 11 clinical studies, summarized in Table I. Of these 11 studies, 6 were randomized placebo-controlled studies (11-13,17,19,20), 1 was single-blinded (18), 2 were open-label (10,14), and 2 were randomized but lacked a placebo group (15,16). A total of 6 studies evaluated the metabolic and oxidative changes promoted by astaxanthin in healthy individuals, while 5 investigated individuals who had one element of the metabolic syndrome, namely obesity, dyslipidemia or type 2 diabetes. In addition, the dose of astaxanthin ranged between 1.8 and 40 mg, and the duration of supplementation varied between 10 days and 12 weeks, reflecting a wide variety of these two parameters. Despite encompassing a small population with a total of 417 individuals, the results of those studies indicated that the beneficial effect of astaxanthin on cardiovascular health was mainly due to its antioxidant and anti-inflammatory properties, its ability to modulate lipid and glucose metabolism, and its role in the maintenance of blood rheological properties.

4. Conclusion

Based on preclinical and clinical evidence, the antioxidant and anti-inflammatory effects of astaxanthin appear to delay the progression of cardiovascular diseases. As an antioxidant, astaxanthin reduces oxidative stress, increases the bioavailability of NO and the activity of antioxidant enzymes, and maintains the rheological properties of the blood. Its anti-inflammatory properties involve modulating the NF-κB and MAPK signaling pathways, reducing the release of pro-inflammatory cytokines and increasing reverse cholesterol transport by HDL, thereby attenuating the accumulation of cholesterol in foam cells and the formation of atherosclerotic plaques. These properties of astaxanthin, together with its favorable safety profile, make this compound a promising option for the prevention and/or adjuvant treatment of cardiovascular diseases.

Funding

No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

CPMP and JJN contributed to the conception of the study and critically reviewed the article. CPMP contributed to the design of the manuscript and figure preparation and editing. CPMP, ACRS, ARV, PSP and JJN contributed to the data acquisition and analysis and drafted the manuscript. All authors have approved all aspects of the present study and agree to be fully accountable for ensuring the integrity and accuracy of the work. All the authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

All the authors declare that they have no competing interests.

Acknowledgments

The authors would like to thank Paula Mitie Hirata for the technical assistance with figure editing.

References

1 

World Health Organization Cardiovascular diseases (CVDs): Journal. 2017, https://www.who.int/en/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds)urisimplehttps://www.who.int/en/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds). Accessed June 10, 2020.

2 

Spahis S, Borys JM and Levy E: Metabolic syndrome as a multi-faceted risk factor for oxidative stress. Antioxid Redox Signal. 26:445–461. 2017. View Article : Google Scholar

3 

Vona R, Gambardella L, Cittadini C, Straface E and Pietraforte D: Biomarkers of oxidative stress in metabolic syndrome and associated diseases. Oxid Med Cell Longev. 2019:82672342019. View Article : Google Scholar : PubMed/NCBI

4 

Puddu P, Puddu GM, Galletti L, Cravero E and Muscari A: Mitochondrial dysfunction as an initiating event in atherogenesis: A plausible hypothesis. Cardiology. 103:137–141. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Kishimoto Y, Yoshida H and Kondo K: Potential anti-atherosclerotic properties of astaxanthin. Mar Drugs. 14:352016. View Article : Google Scholar :

6 

Guerin M, Huntley ME and Olaizola M: Haematococcus astaxanthin: Applications for human health and nutrition. Trends Biotechnol. 21:210–216. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Hussein G, Sankawa U, Goto H, Matsumoto K and Watanabe H: Astaxanthin, a carotenoid with potential in human health and nutrition. J Nat Prod. 69:443–449. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Zhang L and Wang H: Multiple mechanisms of anti-cancer effects exerted by astaxanthin. Mar Drugs. 13:4310–4330. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Maoka T and Etoh H: Some biological functions of carotenoids in Japanese food. Functional Foods of the East. Shi J, Ho CT and Shahidi F: CRC Press; Boca Raton, FL: pp. 85–97. 2010, View Article : Google Scholar

10 

Iwamoto T, Hosoda K, Hirano R, Kurata H, Matsumoto A, Miki W, Kamiyama M, Itakura H, Yamamoto S and Kondo K: Inhibition of low-density lipoprotein oxidation by astaxanthin. J Atheroscler Thromb. 7:216–222. 2000. View Article : Google Scholar

11 

Choi HD, Youn YK and Shin WG: Positive effects of astaxanthin on lipid profiles and oxidative stress in overweight subjects. Plant Foods Hum Nutr. 66:363–369. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Nakagawa K, Kiko T and Miyazawa T, Carpentero Burdeos G, Kimura F, Satoh A and Miyazawa T: Antioxidant effect of astaxanthin on phospholipid peroxidation in human erythrocytes. Br J Nutr. 105:1563–1571. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Karppi J, Rissanen TH, Nyyssönen K, Kaikkonen J, Olsson AG, Voutilainen S and Salonen JT: Effects of astaxanthin supplementation on lipid peroxidation. Int J Vitam Nutr Res. 77:3–11. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Iwabayashi M, Fujioka N, Nomoto K, Miyazaki R, Takahashi H, Hibino S, Takahashi Y, Nishikawa K, Nishida M and Yonei Y: Efficacy and safety of eight-week treatment with astaxanthin in individuals screened for increased oxidative stress burden. Anti Aging Med. 6:15–21. 2009. View Article : Google Scholar

15 

Kim JH, Chang MJ, Choi HD, Youn YK, Kim JT, Oh JM and Shin WG: Protective effects of Haematococcus astaxanthin on oxidative stress in healthy smokers. J Med Food. 14:1469–1475. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Choi HD, Kim JH, Chang MJ, Kyu-Youn Y and Shin WG: Effects of astaxanthin on oxidative stress in overweight and obese adults. Phytother Res. 25:1813–1818. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Park JS, Chyun JH, Kim YK, Line LL and Chew BP: Astaxanthin decreased oxidative stress and inflammation and enhanced immune response in humans. Nutr Metab (Lond). 7:182010. View Article : Google Scholar

18 

Miyawaki H, Takahashi J, Tsukahara H and Takehara I: Effects of astaxanthin on human blood rheology. J Clin Biochem Nutr. 43:69–74. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Mashhadi NS, Zakerkish M, Mohammadiasl J, Zarei M, Mohammadshahi M and Haghighizadeh MH: Astaxanthin improves glucose metabolism and reduces blood pressure in patients with type 2 diabetes mellitus. Asia Pac J Clin Nutr. 27:341–346. 2018.PubMed/NCBI

20 

Yoshida H, Yanai H, Ito K, Tomono Y, Koikeda T, Tsukahara H and Tada N: Administration of natural astaxanthin increases serum HDL-cholesterol and adiponectin in subjects with mild hyperlipidemia. Atherosclerosis. 209:520–523. 2010. View Article : Google Scholar

21 

Lorenz RT and Cysewski GR: Commercial potential for Haematococcus microalgae as a natural source of astaxanthin. Trends Biotechnol. 18:160–167. 2000. View Article : Google Scholar : PubMed/NCBI

22 

Hulbert AJ, Pamplona R, Buffenstein R and Buttemer WA: Life and death: Metabolic rate, membrane composition, and life span of animals. Physiol Rev. 87:1175–1213. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Barros MP, Pinto E, Colepicolo P and Pedersén M: Astaxanthin and peridinin inhibit oxidative damage in Fe(2+)-loaded liposomes: Scavenging oxyradicals or changing membrane permeability? Biochem Biophys Res Commun. 288:225–232. 2001. View Article : Google Scholar : PubMed/NCBI

24 

McNulty HP, Byun J, Lockwood SF, Jacob RF and Mason RP: Differential effects of carotenoids on lipid peroxidation due to membrane interactions: X-ray diffraction analysis. Biochim Biophys Acta. 1768:167–174. 2007. View Article : Google Scholar

25 

Kidd P: Astaxanthin, cell membrane nutrient with diverse clinical benefits and anti-aging potential. Altern Med Rev. 16:355–364. 2011.

26 

Liaudet L, Rosenblatt-Velin N and Pacher P: Role of peroxynitrite in the cardiovascular dysfunction of septic shock. Curr Vasc Pharmacol. 11:196–207. 2013.PubMed/NCBI

27 

Halliwell B: Free radicals and antioxidants: A personal view. Nutr Rev. 52:253–265. 1994. View Article : Google Scholar : PubMed/NCBI

28 

Maoka T, Tokuda H, Suzuki N, Kato H and Etoh H: Anti-oxidative, anti-tumor-promoting, and anti-carcinogensis activities of nitroastaxanthin and nitrolutein, the reaction prod-ucts of astaxanthin and lutein with peroxynitrite. Mar Drugs. 10:1391–1399. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Donahoe SM, Stewart GC, McCabe CH, Mohanavelu S, Murphy SA, Cannon CP and Antman EM: Diabetes and mortality following acute coronary syndromes. JAMA. 298:765–775. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Cai H and Harrison DG: Endothelial dysfunction in cardiovas-cular diseases: The role of oxidant stress. Circ Res. 87:840–844. 2000. View Article : Google Scholar : PubMed/NCBI

31 

Xu X, Gao X, Potter BJ, Cao JM and Zhang C: Anti-LOX-1 rescues endothelial function in coronary arterioles in atheroscle-rotic ApoE knockout mice. Arterioscler Thromb Vasc Biol. 27:871–877. 2007. View Article : Google Scholar : PubMed/NCBI

32 

Zhao ZW, Cai W, Lin YL, Lin QF, Jiang Q, Lin Z and Chen LL: Ameliorative effect of astaxanthin on endothelial dysfunction in streptozotocin-induced diabetes in male rats. Arzneimittelforschung. 61:239–246. 2011. View Article : Google Scholar : PubMed/NCBI

33 

da Silva Garrote-Filho M, Bernardino-Neto M and Penha-Silva N: Influence of erythrocyte membrane stability in atherosclerosis. Curr Atheroscler Rep. 19:172017. View Article : Google Scholar : PubMed/NCBI

34 

Pasterkamp G and Virmani R: The erythrocyte: A new player in atheromatous core formation. Heart. 88:115–116. 2002. View Article : Google Scholar : PubMed/NCBI

35 

Hussein G, Goto H, Oda S, Iguchi T, Sankawa U, Matsumoto K and Watanabe H: Antihypertensive potential and mechanism of action of astaxanthin: II. Vascular reactivity and hemorheology in spontaneously hypertensive rats. Biol Pharm Bull. 28:967–971. 2005. View Article : Google Scholar : PubMed/NCBI

36 

Becker RC: The role of blood viscosity in the development and progression of coronary artery disease. Cleve Clin J Med. 60:353–358. 1993. View Article : Google Scholar : PubMed/NCBI

37 

Hussein G, Goto H, Oda S, Sankawa U, Matsumoto K and Watanabe H: Antihypertensive potential and mechanism of action of astaxanthin: III. Antioxidant and histopathological effects in spontaneously hypertensive rats. Biol Pharm Bull. 29:684–688. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Monroy-Ruiz J, Sevilla MÁ, Carrón R and Montero MJ: Astaxanthin-enriched-diet reduces blood pressure and improves cardiovascular parameters in spontaneously hypertensive rats. Pharmacol Res. 63:44–50. 2011. View Article : Google Scholar

39 

Chen Y, Li S, Guo Y, Yu H, Bao Y, Xin X, Yang H, Ni X, Wu N and Jia D: Astaxanthin attenuates hypertensive vascular remodeling by protecting vascular smooth muscle cells from oxidative stress-induced mitochondrial dysfunction. Oxid Med Cell Longev. 2020:46291892020. View Article : Google Scholar : PubMed/NCBI

40 

Sasaki Y, Kobara N, Higashino S, Giddings JC and Yamamoto J: Astaxanthin inhibits thrombosis in cerebral vessels of stroke-prone spontaneously hypertensive rats. Nutr Res. 31:784–789. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Khan SK, Malinski T, Mason RP, Kubant R, Jacob RF, Fujioka K, Denstaedt SJ, King TJ, Jackson HL, Hieber AD, et al: Novel astaxanthin prodrug (CDX-085) attenuates thrombosis in a mouse model. Thromb Res. 126:299–305. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Nordberg J and Arnér ES: Reactive oxygen species, antioxidants, and the mammalian thioredoxin system. Free Radic Biol Med. 31:1287–1312. 2001. View Article : Google Scholar : PubMed/NCBI

43 

Jekell A, Hossain A, Alehagen U, Dahlström U and Rosén A: Elevated circulating levels of thioredoxin and stress in chronic heart failure. Eur J Heart Fail. 6:883–890. 2004. View Article : Google Scholar : PubMed/NCBI

44 

Aviram M: Introduction to the serial review on paraoxonases, oxidative stress, and cardiovascular diseases. Free Radic Biol Med. 37:1301–1303. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Augusti PR, Quatrin A, Somacal S, Conterato GM, Sobieski R, Ruviaro AR, Maurer LH, Duarte MM, Roehrs M and Emanuelli T: Astaxanthin prevents changes in the activities of thioredoxin reductase and paraoxonase in hypercholesterolemic rabbits. J Clin Biochem Nutr. 51:42–49. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Cui G, Li L, Xu W, Wang M, Jiao D, Yao B, Xu K, Chen Y, Yang S, Long M, et al: Astaxanthin protects ochratoxin a-induced oxidative stress and apoptosis in the heart via the Nrf2 pathway. Oxid Med Cell Longev. 2020:76391092020. View Article : Google Scholar : PubMed/NCBI

47 

Xue Y, Sun C, Hao Q and Cheng J: Astaxanthin ameliorates cardiomyocyte apoptosis after coronary microembolization by inhibiting oxidative stress via Nrf2/HO-1 pathway in rats. Naunyn Schmiedebergs Arch Pharmacol. 392:341–348. 2019. View Article : Google Scholar

48 

Wu Q, Zhang XS, Wang HD, Zhang X, Yu Q, Li W, Zhou ML and Wang XL: Astaxanthin activates nuclear factor erythroid-related factor 2 and the antioxidant responsive element (Nrf2-ARE) pathway in the brain after subarachnoid hemorrhage in rats and attenuates early brain injury. Mar Drugs. 12:6125–6141. 2014. View Article : Google Scholar : PubMed/NCBI

49 

Kavitha K, Thiyagarajan P, Rathna Nandhini J, Mishra R and Nagini S: Chemopreventive effects of diverse dietary phyto-chemicals against DMBA-induced hamster buccal pouch carcinogenesis via the induction of Nrf2-mediated cytoprotective antioxidant, detoxification, and DNA repair enzymes. Biochimie. 95:1629–1639. 2013. View Article : Google Scholar : PubMed/NCBI

50 

Tripathi DN and Jena GB: Astaxanthin intervention ameliorates cyclophosphamide-induced oxidative stress, DNA damage and early hepatocarcinogenesis in rat: Role of Nrf2, p53, p38 and phase-II enzymes. Mutat Res. 696:69–80. 2010. View Article : Google Scholar

51 

Saw CL, Yang AY, Guo Y and Kong AN: Astaxanthin and omega-3 fatty acids individually and in combination protect against oxidative stress via the Nrf2-ARE pathway. Food Chem Toxicol. 62:869–875. 2013. View Article : Google Scholar : PubMed/NCBI

52 

Wen X, Huang A, Hu J, Zhong Z, Liu Y, Li Z, Pan X and Liu Z: Neuroprotective effect of astaxanthin against glutamate-induced cytotoxicity in HT22 cells: Involvement of the Akt/GSK-3β pathway. Neuroscience. 303:558–568. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Visioli F and Artaria C: Astaxanthin in cardiovascular health and disease: Mechanisms of action, therapeutic merits, and knowledge gaps. Food Funct. 8:39–63. 2017. View Article : Google Scholar

54 

Li J, Dai W, Xia Y, Chen K, Li S, Liu T, Zhang R, Wang J, Lu W, Zhou Y, et al: Astaxanthin inhibits proliferation and induces apoptosis of human hepatocellular carcinoma cells via inhibition of NF-κB P65 and Wnt/B-catenin in vitro. Mar Drugs. 13:6064–6081. 2015. View Article : Google Scholar : PubMed/NCBI

55 

Kochi T, Shimizu M, Sumi T, Kubota M, Shirakami Y, Tanaka T and Moriwaki H: Inhibitory effects of astaxanthin on azoxymethane-induced colonic preneoplastic lesions in C57/BL/KsJ-db/db mice. BMC Gastroenterol. 14:2122014. View Article : Google Scholar : PubMed/NCBI

56 

Kavitha K, Kowshik J, Kishore TK, Baba AB and Nagini S: Astaxanthin inhibits NF-κB and Wnt/β-catenin signaling path-ways via inactivation of Erk/MAPK and PI3K/Akt to induce intrinsic apoptosis in a hamster model of oral cancer. Biochim Biophys Acta. 1830:4433–4444. 2013. View Article : Google Scholar : PubMed/NCBI

57 

Yasui Y, Hosokawa M, Mikami N, Miyashita K and Tanaka T: Dietary astaxanthin inhibits colitis and colitis-associated colon carcinogenesis in mice via modulation of the inflammatory cyto-kines. Chem Biol Interact. 193:79–87. 2011. View Article : Google Scholar : PubMed/NCBI

58 

Nagendraprabhu P and Sudhandiran G: Astaxanthin inhibits tumor invasion by decreasing extracellular matrix production and induces apoptosis in experimental rat colon carcinogenesis by modulating the expressions of ERK-2, NFkB and COX-2. Invest New Drugs. 29:207–224. 2011. View Article : Google Scholar

59 

Moroni F, Ammirati E, Norata GD, Magnoni M and Camici PG: The role of monocytes and macrophages in human atherosclerosis, plaque neoangiogenesis, and atherothrombosis. Mediators Inflamm. 2019:74343762019. View Article : Google Scholar : PubMed/NCBI

60 

Hashizume M and Mihara M: Blockade of IL-6 and TNF-α inhibited oxLDL-induced production of MCP-1 via scavenger receptor induction. Eur J Pharmacol. 689:249–254. 2012. View Article : Google Scholar : PubMed/NCBI

61 

Zou TB, Zhu SS, Luo F, Li WQ, Sun XR and Wu HF: Effects of astaxanthin on reverse cholesterol transport and atherosclerosis in mice. Biomed Res Int. 2017:46259322017. View Article : Google Scholar : PubMed/NCBI

62 

Kishimoto Y, Tani M, Uto-Kondo H, Iizuka M, Saita E, Sone H, Kurata H and Kondo K: Astaxanthin suppresses scavenger receptor expression and matrix metalloproteinase activity in macrophages. Eur J Nutr. 49:119–126. 2010. View Article : Google Scholar

63 

Santos SD, Cahú TB, Firmino GO, de Castro CC, Carvalho LB Jr, Bezerra RS and Filho JL: Shrimp waste extract and astaxanthin: Rat alveolar macrophage, oxidative stress and inflammation. J Food Sci. 77:H141–H146. 2012. View Article : Google Scholar : PubMed/NCBI

64 

Lee SJ, Bai SK, Lee KS, Namkoong S, Na HJ, Ha KS, Han JA, Yim SV, Chang K, Kwon YG, et al: Astaxanthin inhibits nitric oxide production and inflammatory gene expression by suppressing I(kappa)B kinase-dependent NF-kappaB activation. Mol Cells. 16:97–105. 2003.PubMed/NCBI

65 

Franceschelli S, Pesce M, Ferrone A, De Lutiis MA, Patruno A, Grilli A, Felaco M and Speranza L: Astaxanthin treatment confers protection against oxidative stress in U937 cells stimulated with lipopolysaccharide reducing O2-production. PLoS One. 9:e883592014. View Article : Google Scholar

66 

Macedo RC, Bolin AP, Marin DP and Otton R: Astaxanthin addition improves human neutrophils function: In vitro study. Eur J Nutr. 49:447–457. 2010. View Article : Google Scholar : PubMed/NCBI

67 

Choi SK, Park YS, Choi DK and Chang HI: Effects of astaxanthin on the production of NO and the expression of COX-2 and iNOS in LPS-stimulated BV2 microglial cells. J Microbiol Biotechnol. 18:1990–1996. 2008.

68 

Kim YJ, Kim YA and Yokozawa T: Protection against oxidative stress, inflammation, and apoptosis of high-glucose-exposed proximal tubular epithelial cells by astaxanthin. J Agric Food Chem. 57:8793–8797. 2009. View Article : Google Scholar : PubMed/NCBI

69 

Abdelzaher LA, Imaizumi T, Suzuki T, Tomita K, Takashina M and Hattori Y: Astaxanthin alleviates oxidative stress insults-related derangements in human vascular endothelial cells exposed to glucose fluctuations. Life Sci. 150:24–31. 2016. View Article : Google Scholar : PubMed/NCBI

70 

Speranza L, Pesce M, Patruno A, Franceschelli S, de Lutiis MA, Grilli A and Felaco M: Astaxanthin treatment reduced oxidative induced pro-inflammatory cytokines secretion in U937: SHP-1 as a novel biological target. Mar Drugs. 10:890–899. 2012. View Article : Google Scholar : PubMed/NCBI

71 

Jones WK, Brown M, Wilhide M, He S and Ren X: NF-kappaB in cardiovascular disease: Diverse and specific effects of a 'general' transcription factor? Cardiovasc Toxicol. 5:183–202. 2005. View Article : Google Scholar

72 

Pashkow FJ, Watumull DG and Campbell CL: Astaxanthin: A novel potential treatment for oxidative stress and inflammation in cardiovascular disease. Am J Cardiol. 101:58D–68D. 2008. View Article : Google Scholar : PubMed/NCBI

73 

Ghosh S, May MJ and Kopp EB: NF-kappa B and Rel proteins: Evolutionarily conserved mediators of immune responses. Annu Rev Immunol. 16:225–260. 1998. View Article : Google Scholar : PubMed/NCBI

74 

Woronicz JD, Gao X, Cao Z, Rothe M and Goeddel DV: IkappaB kinase-beta: NF-kappaB activation and complex formation with IkappaB kinase-alpha and NIK. Science. 278:866–869. 1997. View Article : Google Scholar : PubMed/NCBI

75 

Mercurio F, Zhu H, Murray BW, Shevchenko A, Bennett BL, Li J, Young DB, Barbosa M, Mann M, Manning A and Rao A: IKK-1 and IKK-2: Cytokine-activated IkappaB kinases essential for NF-kappaB activation. Science. 278:860–866. 1997. View Article : Google Scholar : PubMed/NCBI

76 

Zandi E, Rothwarf DM, Delhase M, Hayakawa M and Karin M: The IkappaB kinase complex (IKK) contains two kinase subunits, IKKalpha and IKKbeta, necessary for IkappaB phosphorylation and NF-kappaB activation. Cell. 91:243–252. 1997. View Article : Google Scholar : PubMed/NCBI

77 

DiDonato JA, Hayakawa M, Rothwarf DM, Zandi E and Karin M: A cytokine-responsive IkappaB kinase that activates the transcription factor NF-kappaB. Nature. 388:548–554. 1997. View Article : Google Scholar : PubMed/NCBI

78 

Ghosh S and Karin M: Missing pieces in the NF-kappaB puzzle. Cell. 109(Suppl): S81–S96. 2002. View Article : Google Scholar : PubMed/NCBI

79 

Wang HH, Garruti G, Liu M, Portincasa P and Wang DQ: Cholesterol and lipoprotein metabolism and atherosclerosis: Recent advances in reverse cholesterol transport. Ann Hepatol. 16(Suppl 1): S27–S42. 2017. View Article : Google Scholar : PubMed/NCBI

80 

Khera AV and Rader DJ: Future therapeutic directions in reverse cholesterol transport. Curr Atheroscler Rep. 12:73–81. 2010. View Article : Google Scholar : PubMed/NCBI

81 

Tall AR and Yvan-Charvet L: Cholesterol, inflammation and innate immunity. Nat Rev Immunol. 15:104–116. 2015. View Article : Google Scholar : PubMed/NCBI

82 

Shao B, Tang C, Sinha A, Mayer PS, Davenport GD, Brot N, Oda MN, Zhao XQ and Heinecke JW: Humans with atherosclerosis have impaired ABCA1 cholesterol efflux and enhanced high-density lipoprotein oxidation by myeloperoxidase. Circ Res. 114:1733–1742. 2014. View Article : Google Scholar : PubMed/NCBI

83 

Clarke MC and Bennett MR: Cause or consequence: What does macrophage apoptosis do in atherosclerosis? Arterioscler Thromb Vasc Biol. 29:153–155. 2009. View Article : Google Scholar

84 

Monteiro R and Azevedo I: Chronic inflammation in obesity and the metabolic syndrome. Mediators Inflamm. 2010:2896452010. View Article : Google Scholar : PubMed/NCBI

85 

Kalogeris T, Baines CP, Krenz M and Korthuis RJ: Cell biology of ischemia/reperfusion injury. Int Rev Cell Mol Biol. 298:229–317. 2012. View Article : Google Scholar : PubMed/NCBI

86 

Sanderson TH, Reynolds CA, Kumar R, Przyklenk K and Hüttemann M: Molecular mechanisms of ischemia-reperfusion injury in brain: Pivotal role of the mitochondrial membrane potential in reactive oxygen species generation. Mol Neurobiol. 47:9–23. 2013. View Article : Google Scholar :

87 

Curek GD, Cort A, Yucel G, Demir N, Ozturk S, Elpek GO, Savas B and Aslan M: Effect of astaxanthin on hepatocellular injury following ischemia/reperfusion. Toxicology. 267:147–153. 2010. View Article : Google Scholar

88 

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. 13:3368–3387. 2015. View Article : Google Scholar : PubMed/NCBI

89 

Cai X, Chen Y, Xie X, Yao D, Ding C and Chen M: Astaxanthin prevents against lipopolysaccharide-induced acute lung injury and sepsis via inhibiting activation of MAPK/NF-κB. Am J Transl Res. 11:1884–1894. 2019.

90 

Lauver DA, Lockwood SF and Lucchesi BR: Disodium disuccinate astaxanthin (Cardax) attenuates complement activation and reduces myocardial injury following ischemia/reperfusion. J Pharmacol Exp Ther. 314:686–692. 2005. View Article : Google Scholar : PubMed/NCBI

91 

Ni Y, Nagashimada M, Zhuge F, Zhan L, Nagata N, Tsutsui A, Nakanuma Y, Kaneko S and Ota T: Astaxanthin prevents and reverses diet-induced insulin resistance and steatohepatitis in mice: A comparison with vitamin E. Sci Rep. 5:171922015. View Article : Google Scholar : PubMed/NCBI

92 

Liu H, Liu M, Fu X, Zhang Z, Zhu L, Zheng X and Liu J: Astaxanthin prevents alcoholic fatty liver disease by modulating mouse gut microbiota. Nutrients. 10:12982018. View Article : Google Scholar :

93 

Lyu Y, Wu L, Wang F, Shen X and Lin D: Carotenoid supple-mentation and retinoic acid in immunoglobulin A regulation of the gut microbiota dysbiosis. Exp Biol Med (Maywood). 243:613–620. 2018. View Article : Google Scholar

94 

Tang WH, Kitai T and Hazen SL: Gut microbiota in cardiovascular health and disease. Circ Res. 120:1183–1196. 2017. View Article : Google Scholar : PubMed/NCBI

95 

Saad MJ, Santos A and Prada PO: Linking gut microbiota and inflammation to obesity and insulin resistance. Physiology (Bethesda). 31:283–293. 2016.

96 

Wu L, Lyu Y, Srinivasagan R, Wu J, Ojo B, Tang M, El-Rassi GD, Metzinger K, Smith BJ, Lucas EA, et al: Astaxanthin-shifted gut microbiota is associated with inflammation and metabolic homeostasis in mice. J Nutr. 150:2687–2698. 2020. View Article : Google Scholar : PubMed/NCBI

97 

Ou W, Liao Z, Yu G, Xu H, Liang M, Mai K and Zhang Y: The effects of dietary astaxanthin on intestinal health of juvenile tiger puffer takifugu rubripes in terms of antioxidative status, inflammatory response and microbiota. Aquaculture Nutrition. 25:466–476. 2018.

98 

Zhang L, Cao W, Gao Y, Yang R, Zhang X, Xu J and Tang Q: Astaxanthin (ATX) enhances the intestinal mucosal functions in immunodeficient mice. Food Funct. 11:3371–3381. 2020. View Article : Google Scholar : PubMed/NCBI

99 

Gao Y, Yang L, Chin Y, Liu F, Li RW, Yuan S, Xue C, Xu J and Tang Q: Astaxanthin n-octanoic acid diester ameliorates insulin resistance and modulates gut microbiota in high-fat and high-sucrose dietfed mice. Int J Mol Sci. 21:21492020. View Article : Google Scholar

100 

Wang J, Liu S, Wang H, Xiao S, Li C, Li Y and Liu B: Xanthophyllomyces dendrorhous-derived astaxanthin regulates lipid metabolism and gut microbiota in obese mice induced by a high-fat diet. Mar Drugs. 17:3372019. View Article : Google Scholar :

101 

Linton MRF, Yancey PG, Davies SS, Jerome WG, Linton EF, Song WL, Doran AC and Vickers KC: The role of lipids and lipoproteins in atherosclerosis. Endotext Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, et al: MDText Com, Inc; South Dartmouth, MA: 2000

102 

Strassheim D, Dempsey EC, Gerasimovskaya E, Stenmark K and Karoor V: Role of inflammatory cell subtypes in heart failure. J Immunol Res. 2019:21640172019. View Article : Google Scholar : PubMed/NCBI

103 

Gröschel C, Sasse A, Röhrborn C, Monecke S, Didié M, Elsner L, Kruse V, Bunt G, Lichtman AH, Toischer K, et al: T helper cells with specificity for an antigen in cardiomyocytes promote pressure overload-induced progression from hypertrophy to heart failure. Sci Rep. 7:159982017. View Article : Google Scholar : PubMed/NCBI

104 

Fukunaga T, Soejima H, Irie A, Sugamura K, Oe Y, Tanaka T, Nagayoshi Y, Kaikita K, Sugiyama S, Yoshimura M, et al: Relation between CD4+ T-cell activation and severity of chronic heart failure secondary to ischemic or idiopathic dilated cardio-myopathy. Am J Cardiol. 100:483–488. 2007. View Article : Google Scholar : PubMed/NCBI

105 

Kallikourdis M, Martini E, Carullo P, Sardi C, Roselli G, Greco CM, Vignali D, Riva F, Ormbostad Berre AM, Stølen TO, et al: T cell costimulation blockade blunts pressure overload-induced heart failure. Nat Commun. 8:146802017. View Article : Google Scholar : PubMed/NCBI

106 

Gisterå A and Hansson GK: The immunology of atherosclerosis. Nat Rev Nephrol. 13:368–380. 2017. View Article : Google Scholar

107 

Swirski FK and Nahrendorf M: Leukocyte behavior in athero-sclerosis, myocardial infarction, and heart failure. Science. 339:161–166. 2013. View Article : Google Scholar

108 

Stemme S, Faber B, Holm J, Wiklund O, Witztum JL and Hansson GK: T lymphocytes from human atherosclerotic plaques recognize oxidized low density lipoprotein. Proc Natl Acad Sci USA. 92:3893–3897. 1995. View Article : Google Scholar : PubMed/NCBI

109 

Frostegård J, Ulfgren AK, Nyberg P, Hedin U, Swedenborg J, Andersson U and Hansson GK: Cytokine expression in advanced human atherosclerotic plaques: Dominance of pro-inflammatory (Th1) and macrophage-stimulating cytokines. Atherosclerosis. 145:33–43. 1999. View Article : Google Scholar : PubMed/NCBI

110 

Tripathi DN and Jena GB: Intervention of astaxanthin against cyclophosphamide-induced oxidative stress and DNA damage: A study in mice. Chem Biol Interact. 180:398–406. 2009. View Article : Google Scholar : PubMed/NCBI

111 

Bolin AP, Guerra BA, Nascimento SJ and Otton R: Changes in lymphocyte oxidant/antioxidant parameters after carbonyl and antioxidant exposure. Int Immunopharmacol. 14:690–697. 2012. View Article : Google Scholar : PubMed/NCBI

112 

Bolin AP, Macedo RC, Marin DP, Barros MP and Otton R: Astaxanthin prevents in vitro auto-oxidative injury in human lymphocytes. Cell Biol Toxicol. 26:457–467. 2010. View Article : Google Scholar : PubMed/NCBI

113 

Campoio TR, Oliveira FA and Otton R: Oxidative stress in human lymphocytes treated with fatty acid mixture: Role of carotenoid astaxanthin. Toxicol In Vitro. 25:1448–1456. 2011. View Article : Google Scholar : PubMed/NCBI

114 

Pilinska MA, capital Ka CD, Rushkovsky SR and Dybska OB: Genoprotective properties of astaxanthin revealed by ionizing radiation exposure in vitro on human peripheral blood lymphocytes. Probl Radiac Med Radiobiol. 21:141–148. 2016.In English, Ukrainian. View Article : Google Scholar : PubMed/NCBI

115 

Otton R, Marin DP, Bolin AP, de Cássia Santos Macedo R, Campoio TR, Fineto C Jr, Guerra BA, Leite JR, Barros MP and Mattei R: Combined fish oil and astaxanthin supplementation modulates rat lymphocyte function. Eur J Nutr. 51:707–718. 2012. View Article : Google Scholar

116 

Mahmoud FF, Haines DD, Abul HT, Abal AT, Onadeko BO and Wise JA: In vitro effects of astaxanthin combined with ginkgolide B on T lymphocyte activation in peripheral blood mononuclear cells from asthmatic subjects. J Pharmacol Sci. 94:129–136. 2004. View Article : Google Scholar : PubMed/NCBI

117 

Lin KH, Lin KC, Lu WJ, Thomas PA, Jayakumar T and Sheu JR: Astaxanthin, a carotenoid, stimulates immune responses by enhancing IFN-γ and IL-2 secretion in primary cultured lymphocytes in vitro and ex vivo. Int J Mol Sci. 17:442015. View Article : Google Scholar

118 

Diao W, Chen W, Cao W, Yuan H, Ji H, Wang T, Chen W, Zhu X, Zhou H, Guo H and Zhao X: Astaxanthin protects against renal fibrosis through inhibiting myofibroblast activation and promoting CD8+ T cell recruitment. Biochim Biophys Acta Gen Subj. 1863:1360–1370. 2019. View Article : Google Scholar

119 

Park JS, Mathison BD, Hayek MG, Massimino S, Reinhart GA and Chew BP: Astaxanthin stimulates cell-mediated and humoral immune responses in cats. Vet Immunol Immunopathol. 144:455–461. 2011. View Article : Google Scholar : PubMed/NCBI

120 

Chew BP, Wong MW, Park JS and Wong TS: Dietary beta-carotene and astaxanthin but not canthaxanthin stimulate splenocyte function in mice. Anticancer Res. 19:5223–5227. 1999.

121 

Jyonouchi H, Hill RJ, Tomita Y and Good RA: Studies of immunomodulating actions of carotenoids. I. Effects of beta-carotene and astaxanthin on murine lymphocyte functions and cell surface marker expression in in vitro culture system. Nutr Cancer. 16:93–105. 1991. View Article : Google Scholar : PubMed/NCBI

122 

Mahmoud FF, Haines D, Al-Awadhi R, Arifhodzic N, Abal A, Azeamouzi C, Al-Sharah S and Tosaki A: In vitro suppression of lymphocyte activation in patients with seasonal allergic rhinitis and pollen-related asthma by cetirizine or azelastine in combination with ginkgolide B or astaxanthin. Acta Physiol Hung. 99:173–184. 2012. View Article : Google Scholar : PubMed/NCBI

123 

Jyonouchi H, Zhang L and Tomita Y: Studies of immunomodulating actions of carotenoids. II. Astaxanthin enhances in vitro antibody production to T-dependent antigens without facilitating polyclonal B-cell activation. Nutr Cancer. 19:269–280. 1993. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2021
Volume 47 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Pereira CP, Souza AC, Vasconcelos AR, Prado PS and Name JJ: Antioxidant and anti‑inflammatory mechanisms of action of astaxanthin in cardiovascular diseases (Review). Int J Mol Med 47: 37-48, 2021
APA
Pereira, C.P., Souza, A.C., Vasconcelos, A.R., Prado, P.S., & Name, J.J. (2021). Antioxidant and anti‑inflammatory mechanisms of action of astaxanthin in cardiovascular diseases (Review). International Journal of Molecular Medicine, 47, 37-48. https://doi.org/10.3892/ijmm.2020.4783
MLA
Pereira, C. P., Souza, A. C., Vasconcelos, A. R., Prado, P. S., Name, J. J."Antioxidant and anti‑inflammatory mechanisms of action of astaxanthin in cardiovascular diseases (Review)". International Journal of Molecular Medicine 47.1 (2021): 37-48.
Chicago
Pereira, C. P., Souza, A. C., Vasconcelos, A. R., Prado, P. S., Name, J. J."Antioxidant and anti‑inflammatory mechanisms of action of astaxanthin in cardiovascular diseases (Review)". International Journal of Molecular Medicine 47, no. 1 (2021): 37-48. https://doi.org/10.3892/ijmm.2020.4783