Review Article | Volume: 9, Issue: 1, Jan-Feb, 2021

Astaxanthin: An algae-based natural compound with a potential role in human health-promoting effect: An updated comprehensive review

Jinu Medhi Mohan Chandra Kalita   

Open Access   

Published:  Jan 17, 2021

DOI: 10.7324/JABB.2021.9115
Abstract

Astaxanthin (ASX) is a xanthophyll carotenoid, naturally synthesized by Algae, Yeast, and bacteria. They are not just a powerhouse of antioxidant properties but also possess anti-inflammatory and other bioactive properties. Haematococcus pluvialis is the potential ASX source and shows powerful regulation on many health diseases. Over the decades, numerous studies have emphasized its importance in many human health diseases such as neurodegenerative disorders, cardiovascular diseases, Alzheimer, Parkinson, skin allergies, COVID-19, and many more. The present review discusses available evidence on the functional activities of ASX in various diseases and summarizes the potential factors involved in ASX response and highlights the potential implications.


Keyword:     Astaxanthin Haematococcus pluvialis Antioxidant Anti-inflammatory Human health COVID-19.


Citation:

Medhi J, Kalita MC. Astaxanthin: An algae-based natural compound with a potential role in human health-promoting effect: An updated comprehensive review. J App Biol Biotech. 2021;9(1):114-123. DOI:10.7324/JABB.2021.9115

Copyright: Author(s). This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike license.

HTML Full Text

1. INTRODUCTION

Astaxanthin (ASX) is a carotenoid, occurring naturally in some marine organisms such as microalgae, crustaceans, crayfish, shrimp, krill, trout, and salmon. The potential source of ASX was found in the Haematococcus pluvialis (Chlorophyceae, Volvocales) green microalgae [1]. It is 3,30-dihydroxy-b, b-carotene-4,40-dione and is ubiquitous in nature, belonging to a bright red secondary carotenoid from the same family as b-carotene, lutein, and lycopene. Microalgae/phytoplankton is mainly biosynthesized, gathering in crustaceans and zooplankton, and later in fish, from which it is introduced to greater levels in the food chain. Naturally, several stereoisomers ([3R, 3′S], [3R, 3′R], and [3S, 3′S]) found in ASX and that differ in the structure of the groups of two hydroxyl molecule’s, whereas the 3S, 3’S stereoisomer, most commonly occurs in the salmon and H. pluvialis [1,2] [Figure 1].

ASX has a similar metabolic and physiological function as other carotenoids bear, such as Vitamin C, a-tocopherol, canthaxanthin, zeaxanthin, lutein, and b-carotene; however, ASX is considered as more bioactive compared to others. This is mainly due to the occurrence of hydroxyl and a keto group on each end of the molecule. However, unlike others, it does not convert into Vitamin A. The biosynthetic pathway of ASX was demonstrated in Figure 2.

In marine animals, ASX possesses many important biological functions, including defense against macromolecular oxidation, stress tolerance, immune response, reproductive ability, ultraviolet (UV) light effects, communication, and pigmentation [3]. Due to its specific molecular structure, compared to the other carotenoids, the ASX possess higher antioxidant property [4] such as having both hydroxyl and a keto group, play an important role in neutralizing reactive oxygen species (ROS). ASX protects cell membranes’ integrity by inserting themselves into their bilayers, mitochondria functional integrity, and redox state preservation [5]. Due to its “super-anti-oxidant” property, ASX provides various health benefits and nutraceutical applications to humans, including antidiabetic, kidney protection, atherosclerotic cardiovascular disease (CVD), prevention of liver damage, immunomodulator, neurodegenerative diseases, and many others [Figure 3].

Figure 1: An image of structures of astaxanthin stereoisomers



[Click here to view]

Figure 2: Astaxanthin synthesis pathway



[Click here to view]

Figure 3: Health benefits of astaxanthin



[Click here to view]

1.1. Sources of ASX

ASX naturally originates from algae, crayfish, shrimp, krill, trout, salmon, and yeast [6]. ASX isolated from various microbial origins is demonstrated in Table 1. Besides, commercially, a large amount of ASX is derived from Haematococcus, Phaffia yeast, and through chemical synthesis. Recently, the study by Dave [7] recovered ASX content from Atlantic Shrimp. The potential natural ASX source is H. pluvialis (accumulate up to 5% DW) of ASX. In addition, from a genetically modified Xanthophyllomyces dendrorhous yeast, Chlorella zofinginesis, and Chlorococcum, a very small volume of ASX was isolated [6]. However, natural ASX seems to be more powerful than its synthetic component due to its safety and functional property.

Table 1: Natural microbial sources of astaxanthin

SourcesAstaxanthin (%) on the dry weight basisReferences
Malacostraca
Pandalus clarkia0.015[2]
Pandalus borealis0.12[7,55]
Labyrinthulomycetes
Thraustochytrium sp. CHN-3 (FERM P-18556)0.2[56]
Tremellomycetes
Xanthophyllomyces dendrorhous (VKPM Y2476)0.5[57]
Xanthophyllomyces dendrorhous (JH)0.5[58]
Alphaproteobacteria
Paracoccus carotinifaciens (NITE SD 00017)2.2[55]
Agrobacterium aurantiacum0.01[59]
Florideophyceae
Catenella repens0.02[60]
Ulvophyceae
Ulva lactuca0.01[59]
Enteromorpha intestinalis0.02[60]
Chlorophyceae
Neochloris wimmeri0.6[61]
Chlorella zofingiensis0.001[62]
Chlorococcum0.2[63,64]
H. pluvialis (K-0084)2.7[65]
H. pluvialis (AQSE002)3.4[66]
H. pluvialis (Local isolation)3.6[67]
H. pluvialis (K-0084)3.8[68]
H. pluvialis3.8[69,70]

H. pluvialis: Haematococcus pluvialis

1.2. Effects of H. pluvialis-derived ASX

The secondary metabolite ASX is produced by H. pluvialis in several stress conditions, including enhanced salinity, a higher carbon-to-nitrogen (C:N) ratio and high light. Up to 5% of the ASX can be accumulated by H. pluvialis. Therefore, it is regarded as the potential natural source for the production of carotenoid pigment with high-value [8]. Due to the potential health benefits of naturally derived ASX from H. pluvialis, it is utilized as a dietary supplement from 3.8 to 7.6 mg dosages/day. For more than 15 years, the H. pluvialis-based ASX was frequently used as a nutraceutical supplement without any harmful events [9]. Table 2 represents the H. pluvialis-based ASX on different physiological systems in humans.

Table 2: Haematococcus pluvialis derived astaxanthin derived from role in human health-promoting effect.

Physiological systemSubjectEffect followedMain outcomeReferences
Male fertilityTwenty four healthy male.Idiopathic infertilityReduced[39]
Thirty sub-fertile malePregnancy rate; sperm velocity; oxidation markersIncreased; decreased; Increased[71]
Twenty sub-fertile malePregnancy rate, semen qualityIncreased[72]
Central nervous systemMale and female (Elderly and middle aged)Cog Health battery scoresIncreased[73]
Ten healthy maleTime of response and accuracy of many assignmentsIncreased[74]
Muscle enduranceSixteen non-trained maleAccumulation of lactic acid after a runDecreased[75]
Nineteen non-trained maleActivities of the respiratory and sympathetic nervous systemsIncreased[76]
Twenty non-trained maleExplosiveness/strength/test; Endurance/strength/testImproved; Nil effect[77]
Twenty resistance-trained maleSkeletal muscle damage markersNil effect[78]
Cardiovascular systemMaleLevels of blood plasmaDecreased[38]
Twenty maleTime of blood flowIncreased[79]
Gastric ulcerForty-four functional dyspepsia patientsGastrointestinal discomfort; Inflammatory markersNil effect; Nil effect[80,81]
Immune responseFourteen healthy femaleImmune response; inflammation and oxidative stress markersIncreased; Decreased;[82]
SkinForty-six healthy femaleMoisture and skin elasticityIncreased[83,84]
Healthy male or femaleDermis, corneocyte layer, epidermis, and skin wrinkleIncreased[85]
Eye functionTiredness, blurred vision dryness and eye soreness, andDecreased[86]
Eighty-seven maleAmplitude of eye accommodationIncreased[87]
Forty-nine healthy maleFar visual acuity uncorrectedIncreased[88]
Forty patients with asthenopiaPotency for eye accommodationIncreased[89]
Ten healthy maleFunction of eyeIncreased[90]
Eighteen healthy maleDeep visionIncreased[75]
Anti-oxidationMale (bilateral cataract)The development of hydroperoxide in aqueous humor; antioxidative effects by modifications in the activity of superoxide scavengingSuppressed; Increased[91]

2. VARIOUS BIOLOGICAL PROPERTIES OF ASX

2.1. Antioxidant Property

In a polyene chain, the carotenoids possess long covalently-linked double bonds which are accountable for antioxidant properties. This property scavenges radicals for inhibition of chain reaction and quenching oxygen in a singlet. Compared to other antioxidants, ASX displays greater biological activity since it can bind from inside to outside within the cell membrane. ASX’s polyene chain appears to trap radicals inside the membrane of a cell, whereas the terminal ring of ASX can eliminate radicals both at the interior and in the cell membrane surface. Recently, ASX has drawn much attention from researchers because of its greater antioxidant properties as compared to other naturally occurring carotenoids.

Extrinsic skin damage is caused by various factors, including environmental pollution, poor nutrition, and psychological and physical stress. However, out of all the factors, exposure to UV radiation contributes to 80% and is an important factor for the damage of the skin. UV radiation exposure causes the generation of ROS through oxidative metabolism, leading to skin ageing. It has been established that oxidative stress initiated by ROS depletes the activity of catalases (CAT) in the skin and increases the oxidation of proteins. Further, it initiates the photochemical reactions of ROS by the formation of superoxide anion (O2), singlet oxygen (O2), hydrogen peroxide (H2O2), and hydroxyl radical (OH). UV radiation penetrates the skin and is absorbed by the cells causing the dysfunction of DNA, resulting in the development of photoproducts that inactivates the DNA function, thereby leading to premature skin photoaging. There are many naturally occurring sources available that act as antioxidants which include carotenoids and polyphenols [10]. Recently, Dai et al. [11] investigated the differences of antioxidants between J-aggregates and ASX H-, J-ADC, and H-aggregate-embedded DNA/Chitosan nanoparticles were exposed to H2O2-pretreated Caco-2 cells. H-aggregates had a stronger cytoprotective effect compared with ASX monomers and J-aggregates by facilitating the scavenging of intracellular ROS.

ASX has been recommended to provide beneficial antioxidant protection for people with over-oxidant stress, for example, smokers and obese people [12]. Seventy-eight individuals (39 non-smokers and 39 heavy smokers) were randomly allocated to obtain ASX for 3 weeks at either 5, 20, or 40 mg/day and oxidative stress markers were evaluated. Among smokers, relative to baseline, the oxidative markers isoprostane (ISP) and malondialdehyde (MDA) reduced at all doses. Total antioxidant capacity (TAC) and superoxide dismutase (SOD) ability-assessed antioxidant performance, enhanced, indicating that ASX can help compensate for some of the oxidative damage related to smoking [13]. Various researches have indicated several health benefits from the use of natural ASX in both in vivo and in vitro models based on its chemistry, antioxidant effects, and mechanism of action [14-16].

ASX particularly inhibits the formation of ROS and regulates the oxidative stress-responsive enzymes expression, including heme oxygenase-1 (HO-1). HO-1 is a regulatory mechanism and an oxidative stress marker for the adaptation of cells to oxidative damage [17]. HO-1 is regulated by many factors of transcription, such as nuclear factor erythroid 2-related factor (Nrf2), which enters into the detoxifying mechanism of the enzymes promoter region [4]. Furthermore, ASX has shown to upregulate in irradiated cells of glutathione peroxidase 1, CAT, antioxidative enzymes SOD2, and Nrf2-targeted proteins HO-1. Therefore, ASX has an extensive antioxidant property not only through direct radical scavenging but also through the Nrf2 pathway modulation by activating the cellular antioxidant defense mechanism [18]. Overall, studies conducted by various researchers have shown a significant role of ASX in multiple health benefits based on its mechanism of actions.

2.2. Anti-cancerous Property

Multiple studies have revealed the important role of ASX as anti-oxidative properties without any side effects and toxicity. However, much evidence has suggested its anti-cancerous effects in several cancers types, including leukemia [19], colon carcinogenesis, oral cancer [20], bladder carcinogenesis, and hepatocellular carcinoma [20]. ASX has mediated its effects on different pathways, including cell junction, inflammation, and apoptosis.

The rapid proliferation of cancerous cells results in the formation of tumors. Cancerous cell proliferation facilitates its adhesion to the target tissues, migration, and invasion. These measures differentiate the tumor cell for a metastatic phenotype. Many studies conducted till now shows the effect of ASX on cell proliferation. ASX has an antiproliferative effect on Lewis (mouse lung) lung carcinoma cells, CBRH- 7919 (human hepatoma), and SH-88 (rat breast) [21]. CBRH-7919, however, was a cell line that was most susceptible to ASX with a 39 mM IC50 value.

As compared to other carotenoids, including capsanthin, bixin and, b- carotene on K562 leukemia cells, the growth inhibitory effects of ASX have shown the most effective inhibitory role [19]. ASX has a negative effect on the multiplication of the cancerous cells in oral cancer by controlling the cyclin D1 expression and proliferating cell nuclear antigen [20]. But it decreases the viability of cells in human colon cancer cells HCT-116 [19].

In multicellular organisms, the programmed cell death mechanism that takes place is apoptosis and involves several cellular events such as ultimately cell death, chromosomal DNA fragmentation, cell blebbing, and nuclear fragmentation [22]. However, in tumor cells, if apoptosis occurs, the cancer cell volume tends to decrease. In this case, ASX has shown great importance in the process of apoptosis and thus attracted the interest of many researchers. As the tumor cells are exposed to ASX, it induced modifications in transmembrane potential, respiratory chain, and mitochondrial morphology, and thus regulated the apoptotic mitochondrial proteins such as Bcl- 2 associated X protein (Bax) and B-cell lymphoma 2 (Bcl-2) [19].

ASX is of great importance in oral cancer [20]. By up-regulating pro-apoptotic p-Bcl-2-associated death promoter and BAX, down-regulating the anti-apoptotic Bcl-2 expression, it induces mitochondrial apoptosis mediated by caspase, followed by the movement of cytochrome c and Smac/Diablo into the cytosol and polymerase (ADP-ribose) polymerase cleavage. However, in hepatocellular carcinoma, ASX has regaled the apoptosis pathway by increasing the non-metastasis 23-1 and BAX level while decreasing the B-cell lymphoma-extra-large, c-Myc, and Bcl-2 expression [21]. Thus, this reveals an efficient role of ATX in inducing mitochondria-mediated apoptosis in cells of cancer.

2.3. Cardiovascular Health Diseases

ASX also offers great support for CVD in that its antioxidative and anti-inflammatory properties can significantly shorten blood transmit times. Oxidative stress has appeared as a widespread mechanism for atherosclerosis in particular [23]. It helps in reducing inflammation and improves lipid and glucose metabolism. In a 12-week dose-response randomized clinical trial (RCT), the use of ASX derived from natural algae doses of 6, 12, and 18 mg/day demonstrated a substantial reduction in levels of serum triglycerides (TG), while an increase in serum adiponectin and serum high-density lipoprotein (HDL)-cholesterol levels among adults with mild hyperlipidemia [15]. The effects of a 3-month supplementation period of 8 mg/day of ASX on ASX absorption and lipid peroxidation were examined in a randomized study of healthy men aged 19–33 years. The findings showed that compared to the placebo group, ASX supplementation led to an increase in levels of plasma ASX (P < 0.001). This study indicated that bowel encapsulated ASX absorption was well-tolerated and effective and that ASX supplementation in healthy men reduced in vivo oxidation of selected fatty acids [15].

In another research, 5 and 20 mg/day of ASX ingestion for 3 weeks showed increased SOD and TAC blood levels while decreased MDA and ISP levels among obese and overweight adults. In addition, regular consumption of ASX led to a significant decrease in the levels of LDL-cholesterol and apolipoprotein B [12]. A pilot clinical study conducted by Kato et al. [24] on 16 patients with a risk of heart failure who were taking 12 mg of ASX supplementation daily for 3 months [24]. Researchers have assessed 6-min walk distance, blood pressure, BMI, left ventricular ejection fraction, heart rate, and numerous oxidative stress markers, including urinary 8-hydroxy-2-deoxyguanosine, biological antioxidant potential, and the diacron-reactive oxygen metabolites (d-ROM). Throughout the study, levels of plasma ASX increased significantly. As a result, it decreased the oxidative stress levels demonstrated by a decrease in dROM levels. This research showed improvements in cardiac contractility and exercise tolerance in left ventricular systolic dysfunction patients.

As discussed earlier, ASX does not act as a pro-oxidant, and therefore it is an effective antioxidant. Similarly, in another study, ASX concentration has significantly decreased the serum TG and increased serum HDL-cholesterol levels along with an increase in adiponectin levels in adults with milk hyperlipidemia [25]. However, the mechanism behind the relationship between adiponectin and ASX is unclear. Some researchers have suggested that the ASX’s anti-inflammatory properties can be due to the presence of an increased level of adiponectin as adiponectin itself have its own anti-inflammatory effects. Further studies are required, however, to define specific dosages and to evaluate overall efficacy as a potential preventive measure against CVD. Various bioactivities of ASX reported by several studies were demonstrated in Table 3.

Table 3: Bioactivities of astaxanthin demonstrated by various studies

Biological activitiesReferences
Anticancer[92]
Antiproliferative activity[93]
Bone disease[94]
Immunoregulatory activity[95]
Antimicrobial activity[92,96,97]
Cytotoxic activity[98]
Antityrosinase activity[98]
Neuroprotection[99-101]
Immune response[82,102]
Cardiovascular prevention[15,103-105]
Anti-diabetes[50,106,107]
Anti-hepatoprotective[108]
Anti-gastric activity[109,110]
Anti-inflammatory[105,111-114]
Anti-skin cancer[115-117]
Protection from UV rays[117]
Antioxidant activity[11,13,18,69,95,97,98,117-123]

2.4. Diabetes

A recent human epidemiological study indicates an inverse correlation between carotenoid content in plasma and diabetes mellitus (DM) incidence [26]. In an RCT, 54 Type 2 diabetes (T2D) patients received a placebo and ASX 12 or 6 mg daily for 8 weeks. Ingestion of daily ASX supplementation significantly reduced tumor necrosis factor-a (TNF-a) and plasma IL-6 levels and further decrease in HbA1c (%) and blood glucose levels. Specifically, a higher dosage supplementation significantly reduced the levels of LDL, total cholesterol, plasma TG, and clotting factors such as factor VII and plasminogen activator inhibitor [27]. Data from ten RCTs of the recent meta-analysis found that fasting blood glucose levels were significantly decreased by ASX supplementation [28]. Forty-four participants with T2D received 8 mg of ASX for 8 weeks or placebo in an RCT. Adiponectin, visceral fat, systolic blood pressure, serum TG, and very-low-density lipoprotein were significantly decreased in the group receiving ASX than placebo [29]. Studies conducted in the past revealed a positive association between carotenoids intake and decreased T2D risk [30]. Recently, a study by Chen et al. [31] reported, kidney-targeted evaluation showed that ASX liposomes modified by the glucose-PEG600-DSPE ligand could be transported directly to the glomerular mesangial cell membrane through overexpressed GLUT1 and achieved excellent delivery of kidney-targeted drugs. However, more human research is required to know the role of ASX in diabetes. The immune system, for instance, induces inflammation through releasing cytokines carotenoids and chemokines such as ASX modulate the function of the immune system through reducing cytokines and chemokines through their mechanisms of antioxidant. In addition, evidence indicates that ASX has protective effects against oxidative damage in other conditions, such as neurodegenerative diseases, cardiovascular diseases, diabetes, cancers and obesity [32].

2.5. Liver Function

The liver is vital to lipids and glucose metabolism [33]. There is multifactorial growth of liver stenosis and fatty liver, but oxidative stress is strongly involved in the pathogenesis [34,35]. Studies have confirmed that lipid oxidation products interact with the liver, which further increases the levels of ROS, insulin resistance, and accumulation of lipids and thereby causes liver damage [34]. To inhibit the lipid peroxidation level by ASX, female long-Evans rats were treated with carbon tetrachloride (CCl4) orally (1 mg/kg) and subsequently treated with ASX (10 mg/kg) every day for 2 weeks. The findings of this experiment showed that CCl4 administered in rats increased plasma alanine aminotransferase, aspartate aminotransferase, and alkaline phosphatase activities which were normalized by ASX treatment compared to control rats [36,37]. An RCT performed among obese Korean adults demonstrated that treatment of ASX significantly improved antioxidant markers, including the capacity of total antioxidant (by 121% and 125%) and SOD (by 193% and 194%) and further reduced the ASX blood oxidative stress markers including ISPs (by 64.9% and 64.7%) and MDA (by 34.6% and 35.2%) [12]. Another RCT among Finnish men (placebo [n = 20] vs. ASX, 8 mg/day, 3 months [n = 20]) demonstrated a significant reduction in plasma lipid peroxidation markers, such as 12-hydroxy fatty acids (by 36%) and 15-hydroxy fatty acids (by 60%) [38]. Such human studies have demonstrated beneficial effects of treatment dependent on ASX on oxidative stress, but the effects in the liver are hypothesis.

2.6. Male Infertility

To achieve potential oocytes fertilization, human sperms have to undergo a number of structural changes which is known as capacitation. ROS production demonstrates a major role in triggering a broad acrosome reaction (AR) to occur in the cells. There are many intrinsic and extrinsic factors which trigger the formation of ROS in semen such as activated leukocytes causing inflammation, immature spermatozoa, sertoli cells, exposure to radiation, and inactive lifestyle. The outer sperm membrane undergoes depletion of cholesterol during the capacitation phase [39], which increases both acrosome exocytosis and membrane fusibility followed by downstream signaling processes such as ROS generation, protein serine, and tyrosine phosphorylation (Tyr-P). Higher levels of ROS have induced oxidative stress on sperms, which is being correlated to reduced motility and damage of sperm DNA.

Further, ASX has been shown to enhance parameters of human sperm such as AR-pattern and Tyr-P head without influencing the cell redox process [40]. It demonstrates the important role of a controlled ROS generation in the proper functioning of the sperm, thus providing ROS with the main role in the process of maturation, other than the detrimental factor previously evaluated by the sperm [10].

2.7. Skin Health

Because of its diverse roles in skin biology, collaborative evidence exists that ASX has substantial nutraceutical applications in the dermatology field. In damage and ageing to human skin, oxidative stress plays a significant role. The two factors that cause skin ageing and damage are the generation of ROS and exposure to solar UV radiation.

Oxidant skin ageing events include elastin in the dermal skin layer, the formation of matrix metalloproteinases (MMPs) which degrade collagen, reduced antioxidant production, inflammatory response, and DNA damage [4]. As discussed earlier, the expression of stress-responsive oxidative enzymes such as HO-1 was modulated by ASX and prevented the development of ROS. The latest study also reported in rats that ASX induces reepithelialization during the early burn wounds [41].

This process involves the decreased form of nicotinamide adenine dinucleotide phosphate oxidase (Nox) and free radical regulation of development through the influence of xanthine oxidase; both respond to the ROS generation [41]. It is well understood that by releasing pro-inflammatory mediators, keratinocytes play a significant role in response to photodamage after UV exposure. By reducing the inflammatory cytokine expression, development of UV-induced reactive nitrogen species, and apoptosis in keratinocytes, ASX therapy has been suggested to avoid the traumatic effects of UV. A potential reduction in cyclooxygenase-2 and inducible nitric oxide (iNOS) levels caused by ASX and reduced keratinocyte release of prostaglandin E2 after UV irradiation [15].

ASX’s inhibitory impact on iNOS production has substantial consequences for anti-inflammatory development medications for skin diseases such as atopic dermatitis (AD) and psoriasis. A chronic inflammatory skin condition is called AD, and it is related to causative factors, including abnormalities in immunology that lead to skin lesions. The latest study showed that in an AD animal model, the gene expression of many pro-inflammatory biomarkers such as TNF-a, interleukin (IL)-6, and IL-1b had been inhibited by ASX [42].

A variety of researchers investigated ASX’s nuclear factor-kappa B (NF-kB) inhibition. Specifically, ASX has been documented to have a powerful capacity to degrade of IpBa through its inhibitory effect on the activity of NkB kinase (IKK) and inhibit the nuclear translocation of the NF-pB p65 subunit [43]. Recent research on 44 healthy individuals has demonstrated that a collagen hydrolysate (2 mg/day) and ASX (2 mg/day) combination for 3 months develops barrier integrity and elasticity in the skin of a human. These developments were specifically correlated with molecular changes, including lower levels in the collagen-degrading enzyme MMP-1 expression, elastin-degrading enzyme MMP-12, and induction of procollagen type I [4]. To further confirm the previous studies findings, a high-level evidence-based systematic review by Loke et al. [44] reported ASX substantially protects the skin without any adverse reactions from UV-induced skin damage.

2.8. Brain Health

ASX has also gained a lot of attention from researchers across the globe for its significant impact on neurological pathologies treatment or prevention, including amyotrophic lateral sclerosis, dementia, Huntington’s disease, Parkinson disease, and Alzheimer disease. Although various diseases have different causative agents, the characteristics are mostly same such as the redox metals release combining with oxygen and levels of ROS in cells of neuronal increasing caused by mitochondrial stress and the redox metals release combining with oxygen.

This can cause enhance in protein aggregation that activates cells of microglia. Neuroinflammation contributes to the release of activated cytokines and chemokine’s, creating an oxidative stress environment within the neurons. Many researchers have revealed the role of ASX in delaying the ROS process and alleviating the pathophysiology of various neurodegenerative diseases. ASX induces nerve cell degeneration by increasing growth-associated protein 43, microtubule-associated protein 2, brain-derived neutrophil factor, and gene expression of a glial fibrillary acidic protein [45]. These proteins are responsible for brain recovery.

ASX has also shown its significant role in cognitive impairment. Chemobrain is a cognitive disorder which mostly occurs in cancer patients leading to inability to concentrate, memory impairment, and slow processing speed [46]. El- Agamy et al. have shown the significant effect of ASX as a protective shield to doxorubicin which induces the impairment of cognitive functions [46]. A systematic review of RCTs demonstrated that in middle-aged adults, ASX significantly improved verbal episodic memory performance [47].

ASX has shown neuroprotection and memory-enhancing effects by removing the oxidative stress and mitigating the enhanced activity of acetylcholinesterase.

2.9. Immune System

As discussed earlier, ASX role is an antioxidant, which helps to explain its effect in improving immunity and decreasing inflammation. In a recent study, the authors suggested the potent role of ASX to promote antibody production and oxidative and immune function balance. The study was conducted on a group of male soccer players who received either 4 mg of placebo or ASX for 90 days. Results revealed increased levels of immunoglobulin’s A in salivary glands following ASX supplementation, accompanied by improved pro-oxidant-antioxidant balance [48]. Yet another study examined the effect of ASX compared to b-carotene on the production of antibodies by cells of the immune system (peripheral blood mononuclear cells). ASX enhanced antibody production in response to stimuli, whereas b-carotene did not show a potential impact on the production of human Ig [49].

2.10. Treatment of COVID-19

The algal derived - ASX has been potentially used for the treatment of certain viral infections. Chan et al. [50] reported that diet containing 0.05 percent ASX provided plasma-retained glutathione content for diabetic rats, significantly reduced plasminogen activator inhibitor-1, significantly decreased plasma CRP levels, and reduced MCP-1, TNF-a, IL-6, and ROS. These findings showed that ASX might potentially reduce oxidative, inflammatory, and coagulatory stress, which may have a similar useful impact in patients with COVID-19 who have comorbidities such as diabetes. Kubo et al. [51] reported that ASX prevents oxidative damage through Nrf2 pathway activation in mice model. Recently, Talukdar et al. [52] reported that ASX-derived naturally has considerable potential as a co-adjunctive supplement, desiring required clinical help against COVID-19 for its efficacy and benefit.

2.11. Safety Assessment

ASX has been consumed for nearly 20 years as a dietary supplement [53]. Novel Foods and Food Allergens (NDA) were requested to provide a suggestion on ASX safety with a limit of 8 mg/day when used as a food supplement, taking into account the total combined consumption of ASX from all sources of food. In 2014, in the form of an application submitted pursuant to regulation (EC) No 258/1997, the NDA Panel evaluated the protection of the new ASX-rich ingredient derived from H. pluvialis. In that view, the NDA Panel considered that 0.034 mg/kg body weight (bw) was the appropriate daily intake (ADI) for ASX set by the EFSA FEEDAP Panel in 2014. In 2019, the FEEDAP Panel adopted an opinion on the safety and efficacy of ATX-DMDS (dimethyldisuccinate (EFSA FEEDAP Panel, 2019), a colouring feed additive, for salmonids, crustaceans and other fish. In that assessment, a new ADI of 0.2 mg ASX/kg bw was extracted by the FEEDAP Panel that replaced 2014 ADI definition of 0.034 mg/kg bw [54].

2.12. Future Directions

A variety of compounds are produced by the microalgae, which is useful for human health. ASX is a reddish pigment group of chemicals known as carotenoids. At present, only a fraction of carotenoids is used commercially. There are many opportunities for future study, including isolation of ASX from the novel organisms with a high level of quantity and study into rare carotenoids. In addition, ASX is frequently isolated from the microalgae only. However, in the future, isolation of ASX from several macroalgae species can be attempted. The present market demand for natural ASX is not fulfilled. Therefore, we need to find out several optimization processes for the maximum production of ASX.


3. CONCLUSION

In the prevention and treatment of chronic health diseases such as CVD, cancers, skin diseases, neurological disorders, and COVID-19, ASX has shown an effective role. Several mechanisms of action have been discussed for human health-related diseases. Although there are still some voids in reproducing the efficient roles of ASX in human health as compared to animal studies, and therefore further development in these fields can have a profound effect on the commercial deployment of H. pluvialis ASX products.


4. AUTHOR CONTRIBUTIONS

All authors made substantial contributions to conception and design, acquisition of data, or analysis and interpretation of data; took part in drafting the article or revising it critically for important intellectual content; agreed to submit to the current journal; gave final approval of the version to be published; and agree to be accountable for all aspects of the work.


5. FUNDING

There is no funding to report.


6. CONFLICTS OF INTEREST

The authors report no conflicts of interest in this work.


7. ETHICAL APPROVALS

Not applicable.

REFERENCES

1. Shah MM, Liang Y, Cheng JJ, Daroch M. Astaxanthin-producing green microalga Haematococcus pluvialis:From single cell to high value commercial products. Front Plant Sci 2016;7:531. [CrossRef]

2. Meyers SP, Bligh D. Characterization of astaxanthin pigments from heat-processed crawfish waste. J Agric Food Chem 1981;29:505-8. [CrossRef]

3. Lim KC, Yusoff FM, Shariff M, Kamarudin SM. Astaxanthin as feed supplement in aquatic animals. Rev Aquac 2018;10:738-73. [CrossRef]

4. Davinelli S, Bertoglio JC, Polimeni A, Scapagnini G. Cytoprotective polyphenols against chronological skin aging and cutaneous photodamage. Curr Pharm Design 2018;24:99-105. [CrossRef]

5. Sztretye M, Dienes B, Gönczi M, Czirják T, Csernoch L, Dux L, et al. Astaxanthin:A potential mitochondrial-targeted antioxidant treatment in diseases and with aging. Oxid Med Cell Longev 2019;2019:1-14. [CrossRef]

6. Ambati R, Phang SM, Ravi S, Aswathanarayana RG. Astaxanthin:Sources, extraction, stability, biological activities and its commercial applications-a review. Mar Drugs 2014;12:128-52. [CrossRef]

7. Dave D, Liu Y, Pohling J, Trenholm S, Murphy W. Astaxanthin recovery from Atlantic shrimp (Pandalus borealis) processing materials. Bioresour Technol Rep 2020;11:100535. [CrossRef]

8. Wayama M, Ota S, Matsuura H, Nango N, Hirata A, Kawano S. Three-dimensional ultrastructural study of oil and astaxanthin accumulation during encystment in the green alga Haematococcus pluvialis. PLoS One 2013;8:e53618. [CrossRef]

9. Yang Y, Kim B, Lee JY. Astaxanthin structure, metabolism, and health benefits. J Hum Nutr Food Sci 2013;1:1003.

10. Davinelli S, Bertoglio JC, Zarrelli A, Pina R, Scapagnini G. A randomized clinical trial evaluating the efficacy of an anthocyanin-maqui berry extract (Delphinol®) on oxidative stress biomarkers. J Am Coll Nutr 2015;34:28-33. [CrossRef]

11. Dai M, Li C, Yang Z, Sui Z, Li J, Dong P, et al. The astaxanthin aggregation pattern greatly influences its antioxidant activity:A comparative study in caco-2 cells. Antioxidants 2020;9:126. [CrossRef]

12. Choi HD, Kim JH, Chang MJ, Kyu-Youn Y, Shin WG. Effects of astaxanthin on oxidative stress in overweight and obese adults. Phytother Res 2011;25:1813-8. [CrossRef]

13. Kim JH, Chang MJ, Choi HD, Youn YK, Kim JT, Oh JM, et al. Protective effects of Haematococcus astaxanthin on oxidative stress in healthy smokers. J Med Food 2011;14:1469-75. [CrossRef]

14. Hussein G, Sankawa U, Goto H, Matsumoto K, Watanabe H. Astaxanthin, a carotenoid with potential in human health and nutrition. J Nat Prod 2006;69:443-9. [CrossRef]

15. Yoshida H, Yanai H, Ito K, Tomono Y, Koikeda T, Tsukahara H, et al. Administration of natural astaxanthin increases serum HDL-cholesterol and adiponectin in subjects with mild hyperlipidemia. Atherosclerosis 2010;209:520-3. [CrossRef]

16. Guerin M, Huntley ME, Olaizola M. Haematococcus astaxanthin:Applications for human health and nutrition. TRENDS Biotechnol 2003;21:210-6. [CrossRef]

17. Camera E, Mastrofrancesco A, Fabbri C, Daubrawa F, Picardo M, Sies H, et al. Astaxanthin, canthaxanthin and b-carotene differently affect UVA-induced oxidative damage and expression of oxidative stress-responsive enzymes. Exp Dermatol 2009;18:222-31. [CrossRef]

18. Xue XL, Han XD, Li Y, Chu XF, Miao WM, Zhang JL, et al. Astaxanthin attenuates total body irradiation-induced hematopoietic system injury in mice via inhibition of oxidative stress and apoptosis. Stem Cell Res Ther 2017;8:7. [CrossRef]

19. Zhang L, Wang H. Multiple mechanisms of anti-cancer effects exerted by astaxanthin. Mar Drugs 2015;13:4310-30. [CrossRef]

20. Kowshik J, Nivetha R, Ranjani S, Venkatesan P, Selvamuthukumar S, Veeravarmal V, et al. Astaxanthin inhibits hallmarks of cancer by targeting the PI3K/NF-kB/STAT3 signalling axis in oral squamous cell carcinoma models. IUBMB Life 2019;71:1595-610. [CrossRef]

21. Shao Y, Ni Y, Yang J, Lin X, Li J, Zhang L. Astaxanthin inhibits proliferation and induces apoptosis and cell cycle arrest of mice H22 hepatoma cells. Med Sci Monit 2016;22:2152. [CrossRef]

22. Hormozi M, Ghoreishi S, Baharvand P. Astaxanthin induces apoptosis and increases activity of antioxidant enzymes in LS-180 cells. Artif Cells Nanomed Biotechnol 2019;47:891-5. [CrossRef]

23. Hewlings SJ. Human health benefits of astaxanthin derived from Haematococcus pluvialis:A review. Ec Nutr 2019;10:902-16.

24. Kato T, Kasai T, Sato A, Ishiwata S, Yatsu S, Matsumoto H, et al. Effects of 3-month astaxanthin supplementation on cardiac function in heart failure patients with left ventricular systolic dysfunction-a pilot study. Nutrients 2020;12:1896. [CrossRef]

25. Szentesi P, Csernoch L, Dux L, Keller-Pintér A. Changes in redox signaling in the skeletal muscle with aging. Oxid Med Cell Longev 2019;2019:1-12. [CrossRef]

26. Akbaraly TN, Fontbonne A, Favier A, Berr C. Plasma carotenoids and onset of dysglycemia in an elderly population:Results of the epidemiology of vascular ageing study. Diabetes Care 2008;31:1355-9. [CrossRef]

27. Chan K, Chen S, Chen P. Astaxanthin attenuated thrombotic risk factors in Type 2 diabetic patients. J Funct Foods 2019;53:22-7. [CrossRef]

28. Ursoniu S, Sahebkar A, Serban MC, Banach M. Lipid profile and glucose changes after supplementation with astaxanthin:A systematic review and meta-analysis of randomized controlled trials. Arch Med Sci 2015;11:253. [CrossRef]

29. Mashhadi NS, Zakerkish M, Mohammadiasl J, Zarei M, Mohammadshahi M, Haghighizadeh MH. Astaxanthin improves glucose metabolism and reduces blood pressure in patients with Type 2 diabetes mellitus. Asia Pac J Clin Nutr 2018;27:341.

30. Coyne T, Ibiebele TI, Baade PD, McClintock CS, Shaw JE. Metabolic syndrome and serum carotenoids:Findings of a cross-sectional study in Queensland, Australia. Br J Nutr 2009;102:1668-77. [CrossRef]

31. Chen Z, Li W, Shi L, Jiang L, Li M, Zhang C, et al. Kidney-targeted astaxanthin natural antioxidant nanosystem for diabetic nephropathy therapy. Eur J Pharm Biopharm 2020;156:143-54. [CrossRef]

32. Roohbakhsh A, Karimi G, Iranshahi M. Carotenoids in the treatment of diabetes mellitus and its complications:A mechanistic review. Biomed Pharmacother 2017;91:31-42. [CrossRef]

33. Bechmann LP, Gastaldelli A, Vetter D, Patman GL, Pascoe L, Hannivoort RA, et al. Glucokinase links krüppel-like factor 6 to the regulation of hepatic insulin sensitivity in nonalcoholic fatty liver disease. Hepatology 2012;55:1083-93. [CrossRef]

34. Takaki A, Kawai D, Yamamoto K. Multiple hits, including oxidative stress, as pathogenesis and treatment target in non-alcoholic steatohepatitis (NASH). Int J Mol Sci 2013;14:20704-28. [CrossRef]

35. Reddy JK, Rao MS. Lipid metabolism and liver inflammation. II. Fatty liver disease and fatty acid oxidation. Am J Physiol Gastrointest Liver Physiol 2006;290:G852-8. [CrossRef]

36. Islam MA, Al Mamun MA, Faruk M, Ul Islam MT, Rahman MM, Alam MN, et al. Astaxanthin ameliorates hepatic damage and oxidative stress in carbon tetrachloride-administered rats. Pharmacogn Res 2017;9:84. [CrossRef]

37. Kang JO, Kim SJ, Kim H. Effect of astaxanthin on the hepatotoxicity, lipid peroxidation and antioxidative enzymes in the liver of CCl4-treated rats. Methods Find Exp Clin Pharmacol 2001;23:79. [CrossRef]

38. Karppi J, Rissanen TH, Nyyssönen K, Kaikkonen J, Olsson AG, Voutilainen S, et al. Effects of astaxanthin supplementation on lipid peroxidation. Int J Vitam Nutr Res 2007;77:3-11. [CrossRef]

39. Andrisani A, DonàG, Tibaldi E, Brunati AM, Sabbadin C, Armanini D, et al. Astaxanthin improves human sperm capacitation by inducing lyn displacement and activation. Mar Drugs 2015;13:5533-51. [CrossRef]

40. Gadallah MR. Role of Anti-oxidants in Male Fertility. Crimson;2018.

41. Fang Q, Guo S, Zhou H, Han R, Wu P, Hana C. Astaxanthin protects against early burn-wound progression in rats by attenuating oxidative stress-induced inflammation and mitochondria-related apoptosis. Sci Rep 2017;7:41440. [CrossRef]

42. Park JH, Yeo IJ, Han JH, Suh JW, Lee HP, Hong JT. Anti-inflammatory effect of astaxanthin in phthalic anhydride-induced atopic dermatitis animal model. Exp Dermatol 2018;27:378-85. [CrossRef]

43. Singh KN, Patil S, Barkate H. Protective effects of astaxanthin on skin:Recent scientific evidence, possible mechanisms, and potential indications. J Cosmet Dermatol 2020;19:22-7. [CrossRef]

44. Xiang Q, De Deyn ML, Loke W, Foo NX, Chan HW, Yeo WS. Effects of astaxanthin supplementation on skin health:A systematic review of clinical studies. J Diet Suppl 2020:1-14. [CrossRef]

45. Galasso C, Orefice I, Pellone P, Cirino P, Miele R, Ianora A, et al. On the neuroprotective role of astaxanthin:New perspectives?Mar Drugs 2018;16:247. [CrossRef]

46. El-Agamy SE, Abdel-Aziz AK, Wahdan S, Esmat A, Azab SS. Astaxanthin ameliorates doxorubicin-induced cognitive impairment (chemobrain) in experimental rat model:Impact on oxidative, inflammatory, and apoptotic machineries. Mol Neurobiol 2018;55:5727-40. [CrossRef]

47. Nouchi R, Suiko T, Kimura E, Takenaka H, Murakoshi M, Uchiyama A, et al. Effects of lutein and astaxanthin intake on the improvement of cognitive functions among healthy adults:A systematic review of randomized controlled trials. Nutrients 2020;12:617. [CrossRef]

48. Baralic I, Andjelkovic M, Djordjevic B, Dikic N, Radivojevic N, Suzin-Zivkovic V, et al. Effect of astaxanthin supplementation on salivary IgA, oxidative stress, and inflammation in young soccer players. Evid Based Complement Alternat Med 2015;2015:1-9. [CrossRef]

49. Lin KH, Lin KC, Lu WJ, Thomas PA, Jayakumar T, Sheu JR. Astaxanthin, a carotenoid, stimulates immune responses by enhancing IFN-g and IL-2 secretion in primary cultured lymphocytes in vitro and ex vivo. Int J Mol Sci 2015;17:44. [CrossRef]

50. Chan K, Pen PJ, Yin M. Anticoagulatory and antiinflammatory effects of astaxanthin in diabetic rats. J Food Sci 2012;77:H76-80. [CrossRef]

51. Kubo H, Asai K, Kojima K, Sugitani A, Kyomoto Y, Okamoto A, et al. Astaxanthin suppresses cigarette smoke-induced emphysema through Nrf2 activation in mice. Mar Drugs 2019;17:673. [CrossRef]

52. Talukdar J, Bhadra B, Dasgupta S, Nagle V. Potential of Natural Astaxanthin in Alleviating the Risk of Cytokine Storm and Improve Health in COVID-19:A Scoping Review. Irvine, CA:Research Square;2020. [CrossRef]

53. Capelli B, Talbott S, Ding L. Astaxanthin sources:Suitability for human health and nutrition. Funct Foods Health Dis 2019;9:430. [CrossRef]

54. Turck D, Castenmiller J, de Henauw S, Hirsch-Ernst KI, Kearney J, Maciuk A, et al. Safety of astaxanthin for its use as a novel food in food supplements. EFSA J 2020;18:5993. [CrossRef]

55. Bories G, Brantom P, de Barbera JB, Chesson A, Cocconcelli PS, Debski B, et al. Safety and efficacy of panaferd-AX (red carotenoid rich bacterium Paracoccus carotinifaciens) as feed additive for salmon and trout. EFSA J 2007;546:1-30.

56. Yamaoka Y. Microorganism and production of carotinoid compounds thereby. Patents 2008, Number:US 7,374,908.

57. de la Fuente JL, Rodrrguez-Saiz M, Schleissner C, Díez B, Peiro E, Barredo JL. High-titer production of astaxanthin by the semi-industrial fermentation of xanthophyllomyces dendrorhous. J Biotechnol 2010;148:144-6. [CrossRef]

58. Kim JH, Kang SW, Kim SW, Chang HI. High-level production of astaxanthin by xanthophyllomyces dendrorhous mutant JH1 using statistical experimental designs. Biosci Biotechnol Biochem 2005;69:1743-8. [CrossRef]

59. Yokoyama A, Adachi K, Shizuri Y. New carotenoid glucosides, astaxanthin glucoside and adonixanthin glucoside, isolated from the astaxanthin-producing marine bacterium, Agrobacterium aurantiacum. J Nat Prod 1995;58:1929-33. [CrossRef]

60. Banerjee K, Ghosh R, Homechaudhuri S, Mitra A. Biochemical composition of marine macroalgae from Gangetic Delta at the apex of Bay of Bengal. Afr J Basic Appl Sci 2009;1:96-104.

61. Orosa M, Torres E, Fidalgo P, Abalde J. Production and analysis of secondary carotenoids in green algae. J Appl Phycol 2000;12:553-6. [CrossRef]

62. Wang Y, Peng J. Growth-associated biosynthesis of astaxanthin in heterotrophic Chlorella zofingiensis (Chlorophyta). World J Microbiol Biotechnol 2008;24:1915-22. [CrossRef]

63. Zhang DH, Lee YK. Enhanced accumulation of secondary carotenoids in a mutant of the green alga, Chlorococcum sp. J Appl Phycol 1997;9:459-63. [CrossRef]

64. Zhang DH, Ng YK, Phang SM. Composition and accumulation of secondary carotenoids in Chlorococcum sp. J Appl Phycol 1997;9:147. [CrossRef][CrossRef] [CrossRef]

65. Wang J, Han D, Sommerfeld MR, Lu C, Hu Q. Effect of initial biomass density on growth and astaxanthin production of Haematococcus pluvialis in an outdoor photobioreactor. J Appl Phycol 2013;25:253-60. [CrossRef]

66. Olaizola M. Commercial production of astaxanthin from Haematococcus pluvialis using 25,000-liter outdoor photobioreactors. J Appl Phycol 2000;12:499-506. [CrossRef]

67. Torzillo G, Goksan T, Faraloni C, Kopecky J, Masojídek J. Interplay between photochemical activities and pigment composition in an outdoor culture of Haematococcus pluvialis during the shift from the green to red stage. J Appl Phycol 2003;15:127-36. [CrossRef]

68. Aflalo C, Meshulam Y, Zarka A, Boussiba S. On the relative efficiency of two-vs. One-stage production of astaxanthin by the green alga Haematococcus pluvialis. Biotechnol Bioeng 2007;98:300-5. [CrossRef]

69. Rao AR, Reddy RL, Baskaran V, Sarada R, Ravishankar GA. Characterization of microalgal carotenoids by mass spectrometry and their bioavailability and antioxidant properties elucidated in rat model. J Agric Food Chem 2010;58:8553-9. [CrossRef]

70. Ranga R, Sarada AR, Baskaran V, Ravishankar GA. Identification of carotenoids from green alga Haematococcus pluvialis by HPLC and LC-MS (APCI) and their antioxidant properties. J Microbiol Biotechnol 2009;19:1333-41.

71. Comhaire FH, El Garem Y, Mahmoud A, Eertmans F, Schoonjans F. Combined conventional/antioxidant “astaxanthin“treatment for male infertility:A double blind, randomized trial. Asian J Androl 2005;7:257-62. [CrossRef]

72. Elgarem Y, Lignell A, Comhaire FH. Supplementation with astaxanthin (astacarox) improves semen quality in infertile men. In:Proceedings of the 13th International Carotenoid Symposium. United States:IEEE;2002.

73. Katagiri M, Satoh A, Tsuji S, Shirasawa T. Effects of astaxanthin-rich Haematococcus pluvialis extract on cognitive function:A randomised, double-blind, placebo-controlled study. J Clin Biochem Nutr 2012;51:102-7. [CrossRef]

74. Satoh A, Tsuji S, Okada Y, Murakami N, Urami M, Nakagawa K, et al. Preliminary clinical evaluation of toxicity and efficacy of a new astaxanthin-rich Haematococcus pluvialis extract. J Clin Biochem Nutr 2009;44:280-4. [CrossRef]

75. Sawaki K, Yoshigi H, Aoki K, Natsue K, Akito A, Kesatoki K, et al. Sports performance benefits from taking natural astaxanthin characterized by visual acuity and muscle fatigue improvements in humans. J Clin Ther Med 2002;18:73-88.

76. Nagata A, Tajima T, Takahashi J. Effect of astaxanthin 5 mg on anti fatigue and task performance of human. Carotenoid Sci 2006;10:102-6.

77. Lignell A. Medicament for Improvement of Duration of Muscle Function or Treatment of Muscle Disorders or Diseases;2001. Available from:https://europepmc.org/article/pat/wo9911251.

78. Bloomer RJ, Fry A, Schilling B, Chiu L, Hori N, Weiss L. Astaxanthin supplementation does not attenuate muscle injury following eccentric exercise in resistance-trained men. Int J Sport Nutr Exerc Metab 2005;15:401-12. [CrossRef]

79. Miyawaki H, Takahashi J, Tsukahara H, Takehara I. Effects of astaxanthin on human blood rheology. J Clin Biochem Nutr 2008;43:69-74. [CrossRef]

80. Andersen LP, Holck S, Kupcinskas L, Kiudelis G, Jonaitis L, Janciauskas D, et al. Gastric inflammatory markers and interleukins in patients with functional dyspepsia treated with astaxanthin. FEMS Immunol Med Microbiol 2007;50:244-8. [CrossRef]

81. Kupcinskas L, Lafolie P, Lignell Å, Kiudelis G, Jonaitis L, Adamonis K, et al. Efficacy of the natural antioxidant astaxanthin in the treatment of functional dyspepsia in patients with or without Helicobacter pylori infection:A prospective, randomized, double blind, and placebo-controlled study. Phytomedicine 2008;15:391-9. [CrossRef]

82. Park JS, Chyun JH, Kim YK, Line LL, Chew BP. Astaxanthin decreased oxidative stress and inflammation and enhanced immune response in humans. Nutr Metab 2010;7:18. [CrossRef]

83. Seki T, Sueki H, Kono H, Kaoru S, Eiji Y. Effects of astaxanthin from Haematococcus pluvialis on human skin-patch test;skin repeated application test;effect on wrinkle reduction. Fragrance J 2001;12:98-103.

84. Yamashita E. Cosmetic benefi of dietary supplements containing astaxanthin and tocotrienol on human skin. Food Style 2002;21:112-7.

85. Tominaga K, Hongo N, Karato M, Yamashita E. Cosmetic benefits of astaxanthin on humans subjects. Acta Biochim Pol 2012;59:43-7. [CrossRef]

86. Nagaki Y, Hayasaka S, Yamada T, Hayasaka Y, Sanada M, Uonomi T. Effects of astaxanthin on accommodation, critical flicker fusion, and pattern visual evoked potential in visual display terminal workers. J Tradit Med 2002;19:170-3.

87. Nagaki Y, Mihara M, Tsukahara H, Yoshimoto T, Masuda K. The supplementation effect of astaxanthin on accommodation and asthenopia. J Clin Ther Med 2006;22:41-54.

88. Nakamura A, Isobe R, Otaka Y, Abematsu Y, Nakata D, Honma C, et al. Changes in visual function following peroral astaxanthin. Jpn J Clin Ophthalmol 2004;58:1051-4.

89. Shiratori K, Ogami K, Nitta T. The effects of astaxanthin on accommodation and asthenopia efficacy identification study in healthy volunteers. Clin Med 2005;21:637-50.

90. Iwasaki T, Tawara A. Effects of astaxanthin on eyestrain induced by accommodative dysfunction. J Eye 2006;23:829.

91. Hashimoto H, Arai K, Hayashi S, Okamoto H, Takahashi J, Chikuda M, et al. Effects of astaxanthin on antioxidation in human aqueous humor. J Clin Biochem Nutr 2013;53:13-6. [CrossRef]

92. Marrez DA, Naguib MM, Sultan Y, Higazy AM. Antimicrobial and anticancer activities of Scenedesmus obliquus metabolites. Heliyon 2019;5:e01404. [CrossRef]

93. Sila A, Ayed-Ajmi Y, Sayari N, Nasri M, Martinez-Alvarez O, Bougatef A. Antioxidant and anti-proliferative activities of astaxanthin extracted from the shell waste of deep-water pink shrimp (Parapenaeus longirostris). Nat Prod J 2013;3:82-9. [CrossRef]

94. Valenti MT, Perduca M, Romanelli MG, Mottes M, Carbonare LD. A potential role for astaxanthin in the treatment of bone diseases (review). Mol Med Rep 2020;22:1695-701. [CrossRef]

95. Sun W, Xing L, Lin H, Leng K, Zhai Y, Liu X. Assessment and comparison of in vitro immunoregulatory activity of three astaxanthin stereoisomers. J Ocean Univ China 2016;15:283-7. [CrossRef]

96. Weintraub S, Shpigel T, Harris LG, Schuster R, Lewis EC, Lewitus DY. Astaxanthin-based polymers as new antimicrobial compounds. Polymer Chem 2017;8:4182-9. [CrossRef]

97. Irna C, Jaswir I, Othman R, Jimat DN. Comparison between high-pressure processing and chemical extraction:Astaxanthin yield from six species of shrimp carapace. J Diet Suppl 2018;15:805-13. [CrossRef]

98. Chintong S, Phatvej W, Rerk-Am U, Waiprib Y, Klaypradit M. In vitro antioxidant, antityrosinase, and cytotoxic activities of astaxanthin from shrimp waste. Antioxidants 2019;8:128. [CrossRef]

99. Chang CH, Chen CY, Chiou JY, Peng RY, Peng CH. Astaxanthine secured apoptotic death of PC12 cells induced by b-amyloid peptide 25-35:Its molecular action targets. J Med Food 2010;13:548-56. [CrossRef]

100. Lu YP, Liu SY, Sun H, Wu XM, Li JJ, Zhu L. Neuroprotective effect of astaxanthin on H2O2-induced neurotoxicity in vitro and on focal cerebral ischemia in vivo. Brain Res 2010;1360:40-8. [CrossRef]

101. Fakhri S, Aneva IY, Farzaei MH, Sobarzo-Sánchez E. The neuroprotective effects of astaxanthin:Therapeutic targets and clinical perspective. Molecules 2019;24:2640. [CrossRef]

102. Park JS, Mathison BD, Hayek MG, Massimino S, Reinhart GA, Chew BP. Astaxanthin stimulates cell-mediated and humoral immune responses in cats. Vet Immunol Immunopathol 2011;144:455-61. [CrossRef]

103. Nakao R, Nelson OL, Park JS, Mathison BD, Thompson PA, Chew BP. Effect of astaxanthin supplementation on inflammation and cardiac function in BALB/c mice. Anticancer Res 2010;30:2721-5.

104. Iizuka M, Ayaori M, Uto-Kondo H, Yakushiji E, Takiguchi S, Nakaya K, et al. Astaxanthin enhances ATP-binding cassette transporter A1/G1 expressions and cholesterol efflux from macrophages. J Nutr Sci Vitaminol 2012;58:96-104. [CrossRef]

105. Chew W, Mathison BD, Kimble LL, Mixter PF, Chew BP. Astaxanthin decreases inflammatory biomarkers associated with cardiovascular disease in human umbilical vein endothelial cells. Am J Adv Food Sci Technol 2013;1:1-17. [CrossRef]

106. Gross GJ, Lockwood SF. Acute and chronic administration of disodium disuccinate astaxanthin (CardaxTM) produces marked cardioprotection in dog hearts. Mol Cell Biochem 2005;272:221-7. [CrossRef]

107. Dong LY, Jin J, Lu G, Kang XL. Astaxanthin attenuates the apoptosis of retinal ganglion cells in db/db mice by inhibition of oxidative stress. Mar Drugs 2013;11:960-74. [CrossRef]

108. Turkez H, Geyikoglu F, Yousef MI. Beneficial effect of astaxanthin on 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced liver injury in rats. Toxicol Ind Health 2013;29:591-9. [CrossRef][CrossRef]

109. Kamath BS, Srikanta BM, Dharmesh SM, Sarada R, Ravishankar GA. Ulcer preventive and antioxidative properties of astaxanthin from Haematococcus pluvialis. Eur J Pharmacol 2008;590:387-95. [CrossRef]

110. Bennedsen M, Wang X, Willen R, Wadström T, Andersen LP. Treatment of H. pylori infected mice with antioxidant astaxanthin reduces gastric inflammation, bacterial load and modulates cytokine release by splenocytes. Immunol Lett 2000;70:185-9. [CrossRef]

111. Bhuvaneswari S, Yogalakshmi B, Sreeja S, Anuradha CV. Astaxanthin reduces hepatic endoplasmic reticulum stress and nuclear factor-kB-mediated inflammation in high fructose and high fat diet-fed mice. Cell Stress Chaperones 2014;19:183-91. [CrossRef]

112. Park JS, Mathison BD, Hayek MG, Zhang J, Reinhart GA, Chew BP. Astaxanthin modulates age-associated mitochondrial dysfunction in healthy dogs. J Anim Sci 2013;91:268-75. [CrossRef]

113. Carreira JT, Mingoti GZ, Rodrigues LH, Silva C, Perri SH, Koivisto MB. Impact of proximal cytoplasmic droplets on quality traits and in-vitro embryo production efficiency of cryopreserved bull spermatozoa. Acta Vet Scand 2012;54:1. [CrossRef]

114. Tomofuji T, Ekuni D, Azuma T, Irie K, Endo Y, Kasuyama K, et al. Involvement of toll-like receptor 2 and 4 in association between dyslipidemia and osteoclast differentiation in apolipoprotein E deficient rat periodontium. Lipids Health Dis 2013;12:1. [CrossRef]

115. Maoka T, Tokuda H, Suzuki N, Kato H, Etoh H. Anti-oxidative, anti-tumor-promoting, and anti-carcinogensis activities of nitroastaxanthin and nitrolutein, the reaction products of astaxanthin and lutein with peroxynitrite. Mar Drugs 2012;10:1391-9. [CrossRef]

116. Huangfu J, Liu J, Sun Z, Wang M, Jiang Y, Chen ZY, et al. Antiaging effects of astaxanthin-rich alga Haematococcus pluvialis on fruit flies under oxidative stress. J Agric Food Chem 2013;61:7800-4. [CrossRef]

117. Rao AR, Sindhuja HN, Dharmesh SM, Sankar KU, Sarada R, Ravishankar GA. Effective inhibition of skin cancer, tyrosinase, and antioxidative properties by astaxanthin and astaxanthin esters from the green alga Haematococcus pluvialis. J Agric Food Chem 2013;61:3842-51. [CrossRef]

118. Nakagawa K, Kiko T, Miyazawa T, Burdeos GC, Kimura F, Satoh A, et al. Antioxidant effect of astaxanthin on phospholipid peroxidation in human erythrocytes. Br J Nutr 2011;105:1563-71. [CrossRef]

119. Yang Y, Seo JM, Nguyen A, Pham TX, Park HJ, Park Y, et al. Astaxanthin-rich extract from the green alga Haematococcus pluvialis lowers plasma lipid concentrations and enhances antioxidant defense in apolipoprotein E knockout mice. J Nutr 2011;141:1611-7. [CrossRef]

120. Ishiki M, Nishida Y, Ishibashi H, Wada T, Fujisaka S, Takikawa A, et al. Impact of divergent effects of astaxanthin on insulin signaling in L6 cells. Endocrinology 2013;154:2600-12. [CrossRef]

121. Dose J, Matsugo S, Yokokawa H, Koshida Y, Okazaki S, Seidel U, et al. Free radical scavenging and cellular antioxidant properties of astaxanthin. Int J Mol Sci 2016;17:103. [CrossRef]

122. Rao AR, Baskaran V, Sarada R, Baskaran V. In vivo bioavailability and antioxidant activity of carotenoids from microalgal biomass A repeated dose study. Food Res Int 2013;54:711-7. [CrossRef]

123. Choi HD, Kang HE, Yang SH, Lee MG, Shin WG. Pharmacokinetics and first-pass metabolism of astaxanthin in rats. Br J Nutr 2011;105:220-7. [CrossRef]

Reference

1. Shah MM, Liang Y, Cheng JJ, Daroch M. Astaxanthin-producing green microalga Haematococcus pluvialis: From single cell to high value commercial products. Front Plant Sci 2016;7:531. https://doi.org/10.3389/fpls.2016.00531

2. Meyers SP, Bligh D. Characterization of astaxanthin pigments from heat-processed crawfish waste. J Agric Food Chem 1981;29:505-8. https://doi.org/10.1021/jf00105a017

3. Lim KC, Yusoff FM, Shariff M, Kamarudin SM. Astaxanthin as feed supplement in aquatic animals. Rev Aquac 2018;10:738-73. https://doi.org/10.1111/raq.12200

4. Davinelli S, Bertoglio JC, Polimeni A, Scapagnini G. Cytoprotective polyphenols against chronological skin aging and cutaneous photodamage. Curr Pharm Design 2018;24:99-105. https://doi.org/10.2174/1381612823666171109102426

5. Sztretye M, Dienes B, Gönczi M, Czirják T, Csernoch L, Dux L, et al. Astaxanthin: A potential mitochondrial-targeted antioxidant treatment in diseases and with aging. Oxid Med Cell Longev 2019;2019:1-14. https://doi.org/10.1155/2019/3849692

6. Ambati R, Phang SM, Ravi S, Aswathanarayana RG. Astaxanthin: Sources, extraction, stability, biological activities and its commercial applications-a review. Mar Drugs 2014;12:128-52. https://doi.org/10.3390/md12010128

7. Dave D, Liu Y, Pohling J, Trenholm S, Murphy W. Astaxanthin recovery from Atlantic shrimp (Pandalus borealis) processing materials. Bioresour Technol Rep 2020;11:100535. https://doi.org/10.1016/j.biteb.2020.100535

8. Wayama M, Ota S, Matsuura H, Nango N, Hirata A, Kawano S. Three-dimensional ultrastructural study of oil and astaxanthin accumulation during encystment in the green alga Haematococcus pluvialis. PLoS One 2013;8:e53618. https://doi.org/10.1371/journal.pone.0053618

9. Yang Y, Kim B, Lee JY. Astaxanthin structure, metabolism, and health benefits. J Hum Nutr Food Sci 2013;1:1003.

10. Davinelli S, Bertoglio JC, Zarrelli A, Pina R, Scapagnini G. A randomized clinical trial evaluating the efficacy of an anthocyanin-maqui berry extract (Delphinol®) on oxidative stress biomarkers. J Am Coll Nutr 2015;34:28-33. https://doi.org/10.1080/07315724.2015.1080108

11. Dai M, Li C, Yang Z, Sui Z, Li J, Dong P, et al. The astaxanthin aggregation pattern greatly influences its antioxidant activity: A comparative study in caco-2 cells. Antioxidants 2020;9:126. https://doi.org/10.3390/antiox9020126

12. Choi HD, Kim JH, Chang MJ, Kyu-Youn Y, Shin WG. Effects of astaxanthin on oxidative stress in overweight and obese adults. Phytother Res 2011;25:1813-8. https://doi.org/10.1002/ptr.3494

13. Kim JH, Chang MJ, Choi HD, Youn YK, Kim JT, Oh JM, et al. Protective effects of Haematococcus astaxanthin on oxidative stress in healthy smokers. J Med Food 2011;14:1469-75. https://doi.org/10.1089/jmf.2011.1626

14. Hussein G, Sankawa U, Goto H, Matsumoto K, Watanabe H. Astaxanthin, a carotenoid with potential in human health and nutrition. J Nat Prod 2006;69:443-9. https://doi.org/10.1021/np050354+

15. Yoshida H, Yanai H, Ito K, Tomono Y, Koikeda T, Tsukahara H, et al. Administration of natural astaxanthin increases serum HDL-cholesterol and adiponectin in subjects with mild hyperlipidemia. Atherosclerosis 2010;209:520-3. https://doi.org/10.1016/j.atherosclerosis.2009.10.012

16. Guerin M, Huntley ME, Olaizola M. Haematococcus astaxanthin: Applications for human health and nutrition. TRENDS Biotechnol 2003;21:210-6. https://doi.org/10.1016/S0167-7799(03)00078-7

17. Camera E, Mastrofrancesco A, Fabbri C, Daubrawa F, Picardo M, Sies H, et al. Astaxanthin, canthaxanthin and β-carotene differently affect UVA-induced oxidative damage and expression of oxidative stress-responsive enzymes. Exp Dermatol 2009;18:222-31. https://doi.org/10.1111/j.1600-0625.2008.00790.x

18. Xue XL, Han XD, Li Y, Chu XF, Miao WM, Zhang JL, et al. Astaxanthin attenuates total body irradiation-induced hematopoietic system injury in mice via inhibition of oxidative stress and apoptosis. Stem Cell Res Ther 2017;8:7. https://doi.org/10.1186/s13287-016-0464-3

19. Zhang L, Wang H. Multiple mechanisms of anti-cancer effects exerted by astaxanthin. Mar Drugs 2015;13:4310-30. https://doi.org/10.3390/md13074310

20. Kowshik J, Nivetha R, Ranjani S, Venkatesan P, Selvamuthukumar S, Veeravarmal V, et al. Astaxanthin inhibits hallmarks of cancer by targeting the PI3K/NF-κΒ/STAT3 signalling axis in oral squamous cell carcinoma models. IUBMB Life 2019;71:1595-610. https://doi.org/10.1002/iub.2104

21. Shao Y, Ni Y, Yang J, Lin X, Li J, Zhang L. Astaxanthin inhibits proliferation and induces apoptosis and cell cycle arrest of mice H22 hepatoma cells. Med Sci Monit 2016;22:2152. https://doi.org/10.12659/MSM.899419

22. Hormozi M, Ghoreishi S, Baharvand P. Astaxanthin induces apoptosis and increases activity of antioxidant enzymes in LS-180 cells. Artif Cells Nanomed Biotechnol 2019;47:891-5. https://doi.org/10.1080/21691401.2019.1580286

23. Hewlings SJ. Human health benefits of astaxanthin derived from Haematococcus pluvialis: A review. Ec Nutr 2019;10:902-16.

24. Kato T, Kasai T, Sato A, Ishiwata S, Yatsu S, Matsumoto H, et al. Effects of 3-month astaxanthin supplementation on cardiac function in heart failure patients with left ventricular systolic dysfunction-a pilot study. Nutrients 2020;12:1896. https://doi.org/10.3390/nu12061896

25. Szentesi P, Csernoch L, Dux L, Keller-Pintér A. Changes in redox signaling in the skeletal muscle with aging. Oxid Med Cell Longev 2019;2019:1-12. https://doi.org/10.1155/2019/4617801

26. Akbaraly TN, Fontbonne A, Favier A, Berr C. Plasma carotenoids and onset of dysglycemia in an elderly population: Results of the epidemiology of vascular ageing study. Diabetes Care 2008;31:1355-9. https://doi.org/10.2337/dc07-2113

27. Chan K, Chen S, Chen P. Astaxanthin attenuated thrombotic risk

factors in Type 2 diabetic patients. J Funct Foods 2019;53:22-7. https://doi.org/10.1016/j.jff.2018.12.012

28. Ursoniu S, Sahebkar A, Serban MC, Banach M. Lipid profile and glucose changes after supplementation with astaxanthin: A systematic review and meta-analysis of randomized controlled trials. Arch Med Sci 2015;11:253. https://doi.org/10.5114/aoms.2015.50960

29. Mashhadi NS, Zakerkish M, Mohammadiasl J, Zarei M, Mohammadshahi M, Haghighizadeh MH. Astaxanthin improves glucose metabolism and reduces blood pressure in patients with Type 2 diabetes mellitus. Asia Pac J Clin Nutr 2018;27:341.

30. Coyne T, Ibiebele TI, Baade PD, McClintock CS, Shaw JE. Metabolic syndrome and serum carotenoids: Findings of a cross-sectional study in Queensland, Australia. Br J Nutr 2009;102:1668-77. https://doi.org/10.1017/S000711450999081X

31. Chen Z, Li W, Shi L, Jiang L, Li M, Zhang C, et al. Kidney-targeted astaxanthin natural antioxidant nanosystem for diabetic nephropathy therapy. Eur J Pharm Biopharm 2020;156:143-54. https://doi.org/10.1016/j.ejpb.2020.09.005

32. Roohbakhsh A, Karimi G, Iranshahi M. Carotenoids in the treatment of diabetes mellitus and its complications: A mechanistic review. Biomed Pharmacother 2017;91:31-42. https://doi.org/10.1016/j.biopha.2017.04.057

33. Bechmann LP, Gastaldelli A, Vetter D, Patman GL, Pascoe L, Hannivoort RA, et al. Glucokinase links krüppel-like factor 6 to the regulation of hepatic insulin sensitivity in nonalcoholic fatty liver disease. Hepatology 2012;55:1083-93. https://doi.org/10.1002/hep.24793

34. Takaki A, Kawai D, Yamamoto K. Multiple hits, including oxidative stress, as pathogenesis and treatment target in non-alcoholic steatohepatitis (NASH). Int J Mol Sci 2013;14:20704-28. https://doi.org/10.3390/ijms141020704

35. Reddy JK, Rao MS. Lipid metabolism and liver inflammation. II. Fatty liver disease and fatty acid oxidation. Am J Physiol Gastrointest Liver Physiol 2006;290:G852-8. https://doi.org/10.1152/ajpgi.00521.2005

36. Islam MA, Al Mamun MA, Faruk M, Ul Islam MT, Rahman MM, Alam MN, et al. Astaxanthin ameliorates hepatic damage and oxidative stress in carbon tetrachloride-administered rats. Pharmacogn Res 2017;9:84. https://doi.org/10.4103/pr.pr_26_17

37. Kang JO, Kim SJ, Kim H. Effect of astaxanthin on the hepatotoxicity, lipid peroxidation and antioxidative enzymes in the liver of CCl4- treated rats. Methods Find Exp Clin Pharmacol 2001;23:79. https://doi.org/10.1358/mf.2001.23.2.627931

38. Karppi J, Rissanen TH, Nyyssönen K, Kaikkonen J, Olsson AG, Voutilainen S, et al. Effects of astaxanthin supplementation on lipid peroxidation. Int J Vitam Nutr Res 2007;77:3-11. https://doi.org/10.1024/0300-9831.77.1.3

39. Andrisani A, Donà G, Tibaldi E, Brunati AM, Sabbadin C, Armanini D, et al. Astaxanthin improves human sperm capacitation by inducing lyn displacement and activation. Mar Drugs 2015;13:5533-51. https://doi.org/10.3390/md13095533

40. Gadallah MR. Role of Anti-oxidants in Male Fertility. Crimson; 2018.

41. Fang Q, Guo S, Zhou H, Han R, Wu P, Hana C. Astaxanthin protects against early burn-wound progression in rats by attenuating oxidative stress-induced inflammation and mitochondria-related apoptosis. Sci Rep 2017;7:41440. https://doi.org/10.1038/srep41440

42. Park JH, Yeo IJ, Han JH, Suh JW, Lee HP, Hong JT. Anti-inflammatory effect of astaxanthin in phthalic anhydride-induced atopic dermatitis animal model. Exp Dermatol 2018;27:378-85. https://doi.org/10.1111/exd.13437

43. Singh KN, Patil S, Barkate H. Protective effects of astaxanthin on skin: Recent scientific evidence, possible mechanisms, and potential indications. J Cosmet Dermatol 2020;19:22-7. https://doi.org/10.1111/jocd.13019

44. Xiang Q, De Deyn ML, Loke W, Foo NX, Chan HW, Yeo WS. Effects of astaxanthin supplementation on skin health: A systematic review of clinical studies. J Diet Suppl 2020:1-14. https://doi.org/10.1080/19390211.2020.1739187

45. Galasso C, Orefice I, Pellone P, Cirino P, Miele R, Ianora A, et al. On the neuroprotective role of astaxanthin: New perspectives? Mar Drugs 2018;16:247. https://doi.org/10.3390/md16080247

46. El-Agamy SE, Abdel-Aziz AK, Wahdan S, Esmat A, Azab SS. Astaxanthin ameliorates doxorubicin-induced cognitive impairment (chemobrain) in experimental rat model: Impact on oxidative, inflammatory, and apoptotic machineries. Mol Neurobiol 2018;55:5727-40. https://doi.org/10.1007/s12035-017-0797-7

47. Nouchi R, Suiko T, Kimura E, Takenaka H, Murakoshi M, Uchiyama A, et al. Effects of lutein and astaxanthin intake on the improvement of cognitive functions among healthy adults: A systematic review of randomized controlled trials. Nutrients 2020;12:617. https://doi.org/10.3390/nu12030617

48. Baralic I, Andjelkovic M, Djordjevic B, Dikic N, Radivojevic N, Suzin-Zivkovic V, et al. Effect of astaxanthin supplementation on salivary IgA, oxidative stress, and inflammation in young soccer players. Evid Based Complement Alternat Med 2015;2015:1-9. https://doi.org/10.1155/2015/783761

49. Lin KH, Lin KC, Lu WJ, Thomas PA, Jayakumar T, 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 2015;17:44. https://doi.org/10.3390/ijms17010044

50. Chan K, Pen PJ, Yin M. Anticoagulatory and antiinflammatory effects of astaxanthin in diabetic rats. J Food Sci 2012;77:H76-80. https://doi.org/10.1111/j.1750-3841.2011.02558.x

51. Kubo H, Asai K, Kojima K, Sugitani A, Kyomoto Y, Okamoto A, et al. Astaxanthin suppresses cigarette smoke-induced emphysema through Nrf2 activation in mice. Mar Drugs 2019;17:673. https://doi.org/10.3390/md17120673

52. Talukdar J, Bhadra B, Dasgupta S, Nagle V. Potential of Natural Astaxanthin in Alleviating the Risk of Cytokine Storm and Improve Health in COVID-19: A Scoping Review. Irvine, CA: Research Square; 2020. https://doi.org/10.21203/rs.3.rs-26458/v1

53. Capelli B, Talbott S, Ding L. Astaxanthin sources: Suitability for human health and nutrition. Funct Foods Health Dis 2019;9:430. https://doi.org/10.31989/ffhd.v9i6.584

54. Turck D, Castenmiller J, de Henauw S, Hirsch-Ernst KI, Kearney J, Maciuk A, et al. Safety of astaxanthin for its use as a novel food in food supplements. EFSA J 2020;18:5993. https://doi.org/10.2903/j.efsa.2020.5993

55. Bories G, Brantom P, de Barbera JB, Chesson A, Cocconcelli PS, Debski B, et al. Safety and efficacy of panaferd-AX (red carotenoid rich bacterium Paracoccus carotinifaciens) as feed additive for salmon and trout. EFSA J 2007;546:1-30.

56. Yamaoka Y. Microorganism and production of carotinoid compounds thereby. Patents 2008, Number: US 7,374,908.

57. de la Fuente JL, Rodrrguez-Saiz M, Schleissner C, Díez B, Peiro E, Barredo JL. High-titer production of astaxanthin by the semi-industrial fermentation of xanthophyllomyces dendrorhous. J Biotechnol 2010;148:144-6. https://doi.org/10.1016/j.jbiotec.2010.05.004

58. Kim JH, Kang SW, Kim SW, Chang HI. High-level production of astaxanthin by xanthophyllomyces dendrorhous mutant JH1 using statistical experimental designs. Biosci Biotechnol Biochem 2005;69:1743-8. https://doi.org/10.1271/bbb.69.1743

59. Yokoyama A, Adachi K, Shizuri Y. New carotenoid glucosides, astaxanthin glucoside and adonixanthin glucoside, isolated from the astaxanthin-producing marine bacterium, Agrobacterium aurantiacum. J Nat Prod 1995;58:1929-33. https://doi.org/10.1021/np50126a022

60. Banerjee K, Ghosh R, Homechaudhuri S, Mitra A. Biochemical composition of marine macroalgae from Gangetic Delta at the apex of Bay of Bengal. Afr J Basic Appl Sci 2009;1:96-104.

61. Orosa M, Torres E, Fidalgo P, Abalde J. Production and analysis of secondary carotenoids in green algae. J Appl Phycol 2000;12:553-6. https://doi.org/10.1023/A:1008173807143

62. Wang Y, Peng J. Growth-associated biosynthesis of astaxanthin in heterotrophic Chlorella zofingiensis (Chlorophyta). World J Microbiol Biotechnol 2008;24:1915-22. https://doi.org/10.1007/s11274-008-9692-8

63. Zhang DH, Lee YK. Enhanced accumulation of secondary carotenoids in a mutant of the green alga, Chlorococcum sp. J Appl Phycol 1997;9:459-63. https://doi.org/10.1023/A:1007902103419

64. Zhang DH, Ng YK, Phang SM. Composition and accumulation of secondary carotenoids in Chlorococcum sp. J Appl Phycol 1997;9:147. https://doi.org/10.1023/A:1007926528388

65. Wang J, Han D, Sommerfeld MR, Lu C, Hu Q. Effect of initial biomass density on growth and astaxanthin production of Haematococcus pluvialis in an outdoor photobioreactor. J Appl Phycol 2013;25:253-60. https://doi.org/10.1007/s10811-012-9859-4

66. Olaizola M. Commercial production of astaxanthin from Haematococcus pluvialis using 25,000-liter outdoor photobioreactors.

J Appl Phycol 2000;12:499-506. https://doi.org/10.15358/0935-0381-2000-10-499

67. Torzillo G, Goksan T, Faraloni C, Kopecky J, Masojídek J. Interplay between photochemical activities and pigment composition in an outdoor culture of Haematococcus pluvialis during the shift from the green to red stage. J Appl Phycol 2003;15:127-36. https://doi.org/10.1023/A:1023854904163

68. Aflalo C, Meshulam Y, Zarka A, Boussiba S. On the relative efficiency of two-vs. One-stage production of astaxanthin by the green alga Haematococcus pluvialis. Biotechnol Bioeng 2007;98:300-5. https://doi.org/10.1002/bit.21391

69. Rao AR, Reddy RL, Baskaran V, Sarada R, Ravishankar GA. Characterization of microalgal carotenoids by mass spectrometry and their bioavailability and antioxidant properties elucidated in rat model. J Agric Food Chem 2010;58:8553-9. https://doi.org/10.1021/jf101187k

70. Ranga R, Sarada AR, Baskaran V, Ravishankar GA. Identification of carotenoids from green alga Haematococcus pluvialis by HPLC and LC-MS (APCI) and their antioxidant properties. J Microbiol Biotechnol 2009;19:1333-41.

71. Comhaire FH, El Garem Y, Mahmoud A, Eertmans F, Schoonjans F. Combined conventional/antioxidant "astaxanthin"treatment for male infertility: A double blind, randomized trial. Asian J Androl 2005;7:257-62. https://doi.org/10.1111/j.1745-7262.2005.00047.x

72. Elgarem Y, Lignell A, Comhaire FH. Supplementation with astaxanthin (astacarox) improves semen quality in infertile men. In: Proceedings of the 13th International Carotenoid Symposium. United States: IEEE; 2002.

73. Katagiri M, Satoh A, Tsuji S, Shirasawa T. Effects of astaxanthin-rich Haematococcus pluvialis extract on cognitive function: A randomised, double-blind, placebo-controlled study. J Clin Biochem Nutr 2012;51:102-7. https://doi.org/10.3164/jcbn.D-11-00017

74. Satoh A, Tsuji S, Okada Y, Murakami N, Urami M, Nakagawa K, et al. Preliminary clinical evaluation of toxicity and efficacy of a new astaxanthin-rich Haematococcus pluvialis extract. J Clin Biochem Nutr 2009;44:280-4. https://doi.org/10.3164/jcbn.08-238

75. Sawaki K, Yoshigi H, Aoki K, Natsue K, Akito A, Kesatoki K, et al. Sports performance benefits from taking natural astaxanthin characterized by visual acuity and muscle fatigue improvements in humans. J Clin Ther Med 2002;18:73-88.

76. Nagata A, Tajima T, Takahashi J. Effect of astaxanthin 5 mg on anti fatigue and task performance of human. Carotenoid Sci 2006;10:102-6.

77. Lignell A. Medicament for Improvement of Duration of Muscle Function or Treatment of Muscle Disorders or Diseases; 2001. Available from: https://europepmc.org/article/pat/wo9911251.

78. Bloomer RJ, Fry A, Schilling B, Chiu L, Hori N, Weiss L. Astaxanthin supplementation does not attenuate muscle injury following eccentric exercise in resistance-trained men. Int J Sport Nutr Exerc Metab 2005;15:401-12. https://doi.org/10.1123/ijsnem.15.4.401

79. Miyawaki H, Takahashi J, Tsukahara H, Takehara I. Effects of astaxanthin on human blood rheology. J Clin Biochem Nutr 2008;43:69-74. https://doi.org/10.3164/jcbn.2008048

80. Andersen LP, Holck S, Kupcinskas L, Kiudelis G, Jonaitis L, Janciauskas D, et al. Gastric inflammatory markers and interleukins in patients with functional dyspepsia treated with astaxanthin. FEMS Immunol Med Microbiol 2007;50:244-8. https://doi.org/10.1111/j.1574-695X.2007.00257.x

81. Kupcinskas L, Lafolie P, Lignell Å, Kiudelis G, Jonaitis L, Adamonis K, et al. Efficacy of the natural antioxidant astaxanthin in the treatment of functional dyspepsia in patients with or without Helicobacter pylori infection: A prospective, randomized, double blind, and placebo-controlled study. Phytomedicine 2008;15:391-9. https://doi.org/10.1016/j.phymed.2008.04.004

82. Park JS, Chyun JH, Kim YK, Line LL, Chew BP. Astaxanthin decreased oxidative stress and inflammation and enhanced immune response in humans. Nutr Metab 2010;7:18. https://doi.org/10.1186/1743-7075-7-18

83. Seki T, Sueki H, Kono H, Kaoru S, Eiji Y. Effects of astaxanthin from Haematococcus pluvialis on human skin-patch test; skin repeated application test; effect on wrinkle reduction. Fragrance J 2001;12:98-103.

84. Yamashita E. Cosmetic benefi of dietary supplements containing astaxanthin and tocotrienol on human skin. Food Style 2002;21:112-7.

85. Tominaga K, Hongo N, Karato M, Yamashita E. Cosmetic benefits of astaxanthin on humans subjects. Acta Biochim Pol 2012;59:43-7. https://doi.org/10.18388/abp.2012_2168

86. Nagaki Y, Hayasaka S, Yamada T, Hayasaka Y, Sanada M, Uonomi T. Effects of astaxanthin on accommodation, critical flicker fusion, and pattern visual evoked potential in visual display terminal workers. J Tradit Med 2002;19:170-3.

87. Nagaki Y, Mihara M, Tsukahara H, Yoshimoto T, Masuda K. The supplementation effect of astaxanthin on accommodation and asthenopia. J Clin Ther Med 2006;22:41-54.

88. Nakamura A, Isobe R, Otaka Y, Abematsu Y, Nakata D, Honma C, et al. Changes in visual function following peroral astaxanthin. Jpn J Clin Ophthalmol 2004;58:1051-4.

89. Shiratori K, Ogami K, Nitta T. The effects of astaxanthin on accommodation and asthenopia efficacy identification study in healthy volunteers. Clin Med 2005;21:637-50.

90. Iwasaki T, Tawara A. Effects of astaxanthin on eyestrain induced by accommodative dysfunction. J Eye 2006;23:829.

91. Hashimoto H, Arai K, Hayashi S, Okamoto H, Takahashi J, Chikuda M, et al. Effects of astaxanthin on antioxidation in human aqueous humor. J Clin Biochem Nutr 2013;53:13-6. https://doi.org/10.3164/jcbn.13-6

92. Marrez DA, Naguib MM, Sultan Y, Higazy AM. Antimicrobial and anticancer activities of Scenedesmus obliquus metabolites. Heliyon 2019;5:e01404. https://doi.org/10.1016/j.heliyon.2019.e01404

93. Sila A, Ayed-Ajmi Y, Sayari N, Nasri M, Martinez-Alvarez O, Bougatef A. Antioxidant and anti-proliferative activities of astaxanthin extracted from the shell waste of deep-water pink shrimp (Parapenaeus longirostris). Nat Prod J 2013;3:82-9. https://doi.org/10.2174/2210315511303020002

94. Valenti MT, Perduca M, Romanelli MG, Mottes M, Carbonare LD. A potential role for astaxanthin in the treatment of bone diseases (review). Mol Med Rep 2020;22:1695-701. https://doi.org/10.3892/mmr.2020.11284

95. Sun W, Xing L, Lin H, Leng K, Zhai Y, Liu X. Assessment and comparison of in vitro immunoregulatory activity of three astaxanthin stereoisomers. J Ocean Univ China 2016;15:283-7. https://doi.org/10.1007/s11802-016-2716-3

96. Weintraub S, Shpigel T, Harris LG, Schuster R, Lewis EC, Lewitus DY. Astaxanthin-based polymers as new antimicrobial compounds. Polymer Chem 2017;8:4182-9. https://doi.org/10.1039/C7PY00663B

97. Irna C, Jaswir I, Othman R, Jimat DN. Comparison between high-pressure processing and chemical extraction: Astaxanthin yield from six species of shrimp carapace. J Diet Suppl 2018;15:805-13. https://doi.org/10.1080/19390211.2017.1387885

98. Chintong S, Phatvej W, Rerk-Am U, Waiprib Y, Klaypradit M. In vitro antioxidant, antityrosinase, and cytotoxic activities of astaxanthin from shrimp waste. Antioxidants 2019;8:128. https://doi.org/10.3390/antiox8050128

99. Chang CH, Chen CY, Chiou JY, Peng RY, Peng CH. Astaxanthine secured apoptotic death of PC12 cells induced by β-amyloid peptide 25-35: Its molecular action targets. J Med Food 2010;13:548-56. https://doi.org/10.1089/jmf.2009.1291

100. Lu YP, Liu SY, Sun H, Wu XM, Li JJ, Zhu L. Neuroprotective effect of astaxanthin on H2O2-induced neurotoxicity in vitro and on focal cerebral ischemia in vivo. Brain Res 2010;1360:40-8. https://doi.org/10.1016/j.brainres.2010.09.016

101. Fakhri S, Aneva IY, Farzaei MH, Sobarzo-Sánchez E. The neuroprotective effects of astaxanthin: Therapeutic targets and clinical perspective. Molecules 2019;24:2640. https://doi.org/10.3390/molecules24142640

102. Park JS, Mathison BD, Hayek MG, Massimino S, Reinhart GA, Chew BP. Astaxanthin stimulates cell-mediated and humoral immune responses in cats. Vet Immunol Immunopathol 2011;144:455-61. https://doi.org/10.1016/j.vetimm.2011.08.019

103. Nakao R, Nelson OL, Park JS, Mathison BD, Thompson PA, Chew BP. Effect of astaxanthin supplementation on inflammation and cardiac function in BALB/c mice. Anticancer Res 2010;30:2721-5.

104. Iizuka M, Ayaori M, Uto-Kondo H, Yakushiji E, Takiguchi S, Nakaya K, et al. Astaxanthin enhances ATP-binding cassette transporter A1/G1 expressions and cholesterol efflux from

macrophages. J Nutr Sci Vitaminol 2012;58:96-104. https://doi.org/10.3177/jnsv.58.96

105. Chew W, Mathison BD, Kimble LL, Mixter PF, Chew BP. Astaxanthin decreases inflammatory biomarkers associated with cardiovascular disease in human umbilical vein endothelial cells. Am J Adv Food Sci Technol 2013;1:1-17. https://doi.org/10.7726/ajafst.2013.1001

106. Gross GJ, Lockwood SF. Acute and chronic administration of disodium disuccinate astaxanthin (CardaxTM) produces marked cardioprotection in dog hearts. Mol Cell Biochem 2005;272:221-7. https://doi.org/10.1007/s11010-005-7555-2

107. Dong LY, Jin J, Lu G, Kang XL. Astaxanthin attenuates the apoptosis of retinal ganglion cells in db/db mice by inhibition of oxidative stress. Mar Drugs 2013;11:960-74. https://doi.org/10.3390/md11030960

108. Turkez H, Geyikoglu F, Yousef MI. Beneficial effect of astaxanthin on 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced liver injury in rats. Toxicol Ind Health 2013;29:591-9. https://doi.org/10.1177/0748233711434959

109. Kamath BS, Srikanta BM, Dharmesh SM, Sarada R, Ravishankar GA. Ulcer preventive and antioxidative properties of astaxanthin from Haematococcus pluvialis. Eur J Pharmacol 2008;590:387-95. https://doi.org/10.1016/j.ejphar.2008.06.042

110. Bennedsen M, Wang X, Willen R, Wadström T, Andersen LP. Treatment of H. pylori infected mice with antioxidant astaxanthin reduces gastric inflammation, bacterial load and modulates cytokine release by splenocytes. Immunol Lett 2000;70:185-9. https://doi.org/10.1016/S0165-2478(99)00145-5

111. Bhuvaneswari S, Yogalakshmi B, Sreeja S, Anuradha CV. Astaxanthin reduces hepatic endoplasmic reticulum stress and nuclear factor-κB-mediated inflammation in high fructose and high fat diet-fed mice. Cell Stress Chaperones 2014;19:183-91. https://doi.org/10.1007/s12192-013-0443-x

112. Park JS, Mathison BD, Hayek MG, Zhang J, Reinhart GA, Chew BP. Astaxanthin modulates age-associated mitochondrial dysfunction in healthy dogs. J Anim Sci 2013;91:268-75. https://doi.org/10.2527/jas.2012-5341

113. Carreira JT, Mingoti GZ, Rodrigues LH, Silva C, Perri SH, Koivisto MB. Impact of proximal cytoplasmic droplets on quality traits and in-vitro embryo production efficiency of cryopreserved bull spermatozoa. Acta Vet Scand 2012;54:1. https://doi.org/10.1186/1751-0147-54-1

114. Tomofuji T, Ekuni D, Azuma T, Irie K, Endo Y, Kasuyama K, et al. Involvement of toll-like receptor 2 and 4 in association between dyslipidemia and osteoclast differentiation in apolipoprotein E deficient rat periodontium. Lipids Health Dis 2013;12:1. https://doi.org/10.1186/1476-511X-12-1

115. Maoka T, Tokuda H, Suzuki N, Kato H, Etoh H. Anti-oxidative, anti-tumor-promoting, and anti-carcinogensis activities of nitroastaxanthin and nitrolutein, the reaction products of astaxanthin and lutein with peroxynitrite. Mar Drugs 2012;10:1391-9. https://doi.org/10.3390/md10061391

116. Huangfu J, Liu J, Sun Z, Wang M, Jiang Y, Chen ZY, et al. Antiaging effects of astaxanthin-rich alga Haematococcus pluvialis on fruit flies under oxidative stress. J Agric Food Chem 2013;61:7800-4. https://doi.org/10.1021/jf402224w

117. Rao AR, Sindhuja HN, Dharmesh SM, Sankar KU, Sarada R, Ravishankar GA. Effective inhibition of skin cancer, tyrosinase, and antioxidative properties by astaxanthin and astaxanthin esters from the green alga Haematococcus pluvialis. J Agric Food Chem 2013;61:3842-51. https://doi.org/10.1021/jf304609j

118. Nakagawa K, Kiko T, Miyazawa T, Burdeos GC, Kimura F, Satoh A, et al. Antioxidant effect of astaxanthin on phospholipid peroxidation in human erythrocytes. Br J Nutr 2011;105:1563-71. https://doi.org/10.1017/S0007114510005398

119. Yang Y, Seo JM, Nguyen A, Pham TX, Park HJ, Park Y, et al. Astaxanthin-rich extract from the green alga Haematococcus pluvialis lowers plasma lipid concentrations and enhances antioxidant defense in apolipoprotein E knockout mice. J Nutr 2011;141:1611-7. https://doi.org/10.3945/jn.111.142109

120. Ishiki M, Nishida Y, Ishibashi H, Wada T, Fujisaka S, Takikawa A, et al. Impact of divergent effects of astaxanthin on insulin signaling in L6 cells. Endocrinology 2013;154:2600-12. https://doi.org/10.1210/en.2012-2198

121. Dose J, Matsugo S, Yokokawa H, Koshida Y, Okazaki S, Seidel U, et al. Free radical scavenging and cellular antioxidant properties of astaxanthin. Int J Mol Sci 2016;17:103. https://doi.org/10.3390/ijms17010103

122. Rao AR, Baskaran V, Sarada R, Baskaran V. In vivo bioavailability and antioxidant activity of carotenoids from microalgal biomass A repeated dose study. Food Res Int 2013;54:711-7. https://doi.org/10.1016/j.foodres.2013.07.067

123. Choi HD, Kang HE, Yang SH, Lee MG, Shin WG. Pharmacokinetics and first-pass metabolism of astaxanthin in rats. Br J Nutr 2011;105:220-7. https://doi.org/10.1017/S0007114510003454

Article Metrics
101 Views 240 Downloads 341 Total

Year

Month

Related Search

By author names

Similar Articles

Bioactivity assessment of endophytic fungi associated with Centella asiatica and Murraya koengii

Archana Nath, Jyoti Pathak and SR Joshi

Asparagus racemosus extract increases the life span in Drosophila melanogaster

K. V. Kiran Kumar, K. S. Prasanna, J. S. Ashadevi

Comparative three way analysis of biochemical responses in cereal and millet crops under salinity stress

Ritika Bhatt, Prem Prakash Asopa, Santosh Sihag, Rakesh Sharma, Sumita Kachhwaha, S.L. Kothari

Impact of Phyllanthus amarus extract on antioxidant enzymes in Drosophila melanogaster

N. Manasa, J. S. Ashadevi

Alterations in antioxidant defense system in hepatic and renal tissues of rats following aspartame intake

Saeed A. Alwaleedi

Ten different brewing methods of green tea: comparative antioxidant study

Naila Safdar, Amina Sarfaraz, Zehra Kazmi, Azra Yasmin

Medicinal and Functional Values of Thyme (Thymus vulgaris L.) Herb

Eqbal M. A. Dauqan, Aminah Abdullah

Buffalo (Bubalus bubilis) colostrum and milk fat globule membrane fractions are potent antioxidants

N. Brijesha, H. S. Aparna

Effect of extraction solvents on phenolic contents and antioxidant capacities of Artocarpus chaplasha and Carissa carandas fruits from Bangladesh

Gouri Dhar, Sonam Akther, Afrin Sultana, Uchen May, Mohammed Moinul Islam, Mowri Dhali, Dwaipayan Sikdar

In Vitro Antidiabetic and Anti-oxidant Activities of Methanol Extract of Tinospora Sinensis

Anindita Banerjee, Bithin Maji, Sandip Mukherjee, Kausik Chaudhuri, Tapan Seal

Chemical composition, antioxidant and antimicrobial activities of the essential oil of Vetiveria nigritana (Benth.) Stapf roots from Burkina Faso

Zenabou Semde, Jean Koudou, Cheikna Zongo, Gilles Figueredo, Marius K. Somda, Leguet Ganou, Alfred S. Traore

Crosstalk of brassinosteroids with other phytohormones under various abiotic stresses

Farhan Ahmad, Ananya Singh, Aisha Kamal

In vitro propagation by axillary shoot proliferation, assessment of antioxidant activity, and genetic fidelity of micropropagated Paederia foetida L.

Biswaranjan Behera, Priyajeet Sinha, Sushanto Gouda, Sakti K. Rath, Durga P. Barik, Padan K. Jena, Pratap C. Panda, Soumendra K. Naik

Effect of growth hormones in induction of callus, antioxidants, and antibacterial activity in Nerium odorum

Avinash Prakasha, S Umesha

Aroclor 1254 induced oxidative stress and histopathological changes in mice liver

Jalpa Raja, Shweta Pathak, Rahul Kundu

A study of endophytic fungi Neofusicoccum ribis from Gandaria (Bouea macrophylla Griffith) as enzyme inhibitor, antibacterial, and antioxidant

Trisanti Anindyawati, Praptiwi

Determination of phytochemical, antioxidant, antimicrobial, and protein binding qualities of hydroethanolic extract of Celastrus paniculatus

Vijay Kumar¥, Simranjeet Singh¥, Arjun Singh¥, Amit Kumar Dixit¥, Bhavana Shrivastava, Sapna Avinash Kondalkar, Joginder Singh, Ravindra Singh, Gurpreet Kaur Sidhu, Rajesh Partap Singh, Varanasi Subhose, Om Prakash

Effect of Trema guineensis leaves (celtidaceae) on glucose-induced hypertension in Wistar rats

Balakiyém Kadissoli, P A Mouzou, T Pakoussi, K Eklu-Gadegbeku, A K Aklikokou, M Gbeassor

Phytochemical analysis, antimicrobial and antioxidant activities of Aidia borneensis leaf extracts

Zulhamizan Awang-Jamil, Aida Maryam Basri, Norhayati Ahmad, Hussein Taha

Comparative chemical and biological investigations of three Saudi Astragalus species

Mohamed A. Ashour

Phytochemical analysis and antioxidant potential of Ocimum gratissimum Linn (Lamiaceae) commonly consumed in the Republic of Benin

Hinnoutondji Wilfrid Kpètèhoto, Abdou Madjid Olatoundé Amoussa, Roch Christian Johnson, Eustache Enock Meinsan Houéto , Franck Maurille Zinsou Mignanwandé, Hounnankpon Yédomonhan, Frédéric Loko, Honoré Bankolé, Latifou Lagnika

Pleurotus pulmonarius (Fr.) Quel. (Pleurotaceae): In vitro antioxidant evaluation and the isolation of a steroidal isoprenoid

Blessing Onyinye Okonkwo, Ozadheoghene Eriarie Afieroho, Emeka Daniel Ahanonu, Lambert Okwubie, Kio Anthony Abo

Study of the changes in the growth, protein, and bioactive profile of Chlorella emersonii KJ725233 in response to sodium and ammonium nitrate

Sneha Sunil Sawant, Varsha Kelkar-Mane

Determination of phenolic content and antioxidant capacity of Launaea resedifolia from Algerian Sahara

Amina Bouguerra, Mohamed Hadjadj, Mesaouda Dekmouche, Zhour Rahmani, Houssine Dendougui

Investigation of morphological, phytochemical, and enzymatic characteristics of Anethum graveolens L. using selenium in combination with humic acid and fulvic acid

Parviz Samavatipour, Vahid Abdossi, Reza Salehi, Saeed Samavat,Alireza Ladan Moghadam

A study on the salinity stress effects on the biochemical traits of seedlings and its relationship with resistance toward sensitive and tolerant flax genotypes

Yousef Alaei, Seyed Kamal Kazemitabar, Mohammad Zaefi Zadeh, Hamid Najafi Zarini, Gaffar Kiani

Nyctanthes arbor-tristis: Comprehensive review on its pharmacological, antioxidant, and anticancer activities

Smita Parekh, Anjali Soni

Anti-quorum sensing, antibacterial, antioxidant activities, and phytoconstituents analysis of medicinal plants used in Benin: Acacia macrostachya (Rchb. ex DC.)

Mounirou Tchatchedre, Abdou Madjid O. Amoussa, Ménonvè Atindehou, Aminata P. Nacoulma, Ambaliou Sanni, Martin kiendrebeogo, Latifou Lagnika

Studies on the mechanism of desiccation tolerance in the resurrection fern Adiantum raddianum

Tumkur Govindaraju Banupriya, Chandraiah Ramyashree, Devaraja Akash, Neeragunda Shivaraj Yathish, Ramasandra Govindarao Sharthchandra

Antioxidant and antihyperlipidemic effects of aqueous seed extract of Daucus carota L. in triton ×100-induced hyperlipidemic mice

Habibu Tijjani, Abubakar Mohammed, Sani Muktar, Saminu Musa, Yusuf Abubakar, Adegbenro Peter Adegunloye, Ahmed Adebayo Ishola, Enoch Banbilbwa Joel, Carrol Domkat Luka, Adamu Jibril Alhassan

Role of glutathione reductase and catalase enzyme in antioxidant defense mechanism in controlling fluoride-induced oxidative stress

Komal Sharma, Mamta Choudhary, Khushbu Verma

Biological activities and phytochemicals of Hyptis capitata grown in East Kalimantan, Indonesia

Irawan Wijaya Kusuma, Rahmini, Enos Tangke Arung, Arif Yudo Pramono, Erwin, Supomo

Mycelial biomass, antioxidant, and myco-actives of mycelia of abalone mushroom Pleurotus cystidiosus in liquid culture

Kent Garcia, Chester Jhae Garcia, Reynante Bustillos, Rich Milton Dulay

Salt stress, its impacts on plants and the strategies plants are employing against it: A review

Zeenat Mushtaq, Shahla Faizan, Basit Gulzar

Impact of oxidizing, reducing, and stabilizing agents on the inhibitory properties of Cyamopsis tetragonoloba trypsin inhibitor

Preeti Patidar, Mahima Golani, Sumati Hajela, Krishnan Hajela

Identification of highest L-Methioninase enzyme producers among soil microbial isolates, with potential antioxidant and anticancer properties

D. Kavya, Varalakshmi Kilingar Nadumane

Linalool protects hippocampal CA1 neurons and improves functional outcomes following experimental ischemia/reperfusion in rats

Vishal Airao, Prakruti Buch, Tejas Sharma, Devendra Vaishnav, Sachin Parmar

Modification of the time of incubation in colorimetric method for accurate determination of the total antioxidants capacity using 2,2-diphenyl-1-picrylhydrazyl stable free radical

Abhipsa Bal, Samar Gourav Pati, Falguni Panda, Biswaranjan Paital

HR-LC-MS based profiling of phytochemicals from methanol extracts of leaves and bark of Myristica dactyloides Gaertn. from Western Ghats of Karnataka, India

Kuppuru Mallikarjunaiah Marulasiddaswamy, Bettadapura Rameshgowda Nuthan,, Channarayapatna-Ramesh Sunilkumar, Shrisha Naik Bajpe,, Kigga Kaadappa Sampath Kumara, Shailasree Sekhar, Kukkundoor Ramachandra Kini

Preclinical evaluation of anticataract activity of Mentha spicata leaves on isolated goat lens by an in vitro model

Shreya Mohandas, Saahiba Thaleshwari, Myrene Roselyn Dsouza

Green synthesis, characterizations, and in vitro biological evaluation of Cu (II) complexes of quercetin with N ^ N ligands

Tanu Srivastava, Sunil Kumar Mishra, Om Prakash Tiwari

Optimization of extraction conditions of phytochemical compounds in “Xiem” banana peel powder using response surface methodology

Ngo Van Tai, Mai Nhat Linh, Nguyen Minh Thuy

Antioxidative, antiproliferative, and apoptosis effect of Coleus tuberosus flesh and peel ethanol extracts on cervical cancer cell lines

Mutiara Nugraheni, Windarwati Windarwati, Badraningsih Lastariwati

Comparative study of hydroalcoholic extracts of Bryophyllum pinnatum and Macrotyloma uniflorum for their antioxidant, antiurolithiatic, and wound healing potential

Chetna Faujdar, Priyadarshini

Qualitative and quantitative analysis of Precocene II, estimation of enzymatic, nonenzymatic antioxidant, and cytotoxic potentials of methyl jasmonate-elicited shoot culture of Ageratum conyzoides Linn.

Selvaraj Vasantharani, Ramaraj Thirugnanasampandan, Gunasekaran Bhuvaneswari

Suppression of the RAGE gene expression in RAW 264.7 murine leukemia cell line by ethyl acetate extract of Mikania micrantha (L.) Kunth.

Alex Zohmachhuana, Malsawmdawngliana Tlaisun, Vabeiryureilai Mathipi, Lalrinzuali Khawlhring, Joyce Sudandara Priya

A review on fish peptides isolated from fish waste with their potent bioactivities

Ayusman Behera, Rajashree Das, Pranati Patnaik, Jyotirmaya Mohanty, Gargee Mohanty

Quantification of phytochemicals and in vitro antioxidant activities from various parts of Euphorbia neriifolia Linn.

Priya Chaudhary, Pracheta Janmeda

Influence of soaking and germination treatments on the nutritional, anti-nutritional, and bioactive composition of pigeon pea (Cajanus cajan L.)

Qurat Ul Eain Hyder Rizvi, Krishan Kumar, Naseer Ahmed, Ajar Nath Yadav, Divya Chauhan, Priyanka Thakur, Sumaira Jan, Imran Sheikh

Impact of diverse processing treatments on nutritional and anti-nutritional characteristics of soybean (Glycine max L.)

Priyanka Thakur, Krishan Kumar, Naseer Ahmed, Ajar Nath Yadav, Sunil Kumar, Qurat Ul Eain Hyder Rizvi, Divya Chauhan, Sumaira Jan

Cathelicidin-HR from Hoplobatrachus rugulosus: an antioxidant peptide that performs a protective effect against UV/H2O2 -induced DNA damage

Piyachat Wiriyaampaiwong, Chutima Karnmongkol, Arpaporn Punpad, Nattapong Srisamoot, Wutti Rattanavichai, Alongkod Tanomtong, Sakda Daduang,, Sompong Klaynongsruang,, Anupong Tankrathok,

Woodfordia fruticosa (Linn.) Kurz’s fungal endophyte Mucor souzae’s secondary metabolites, kaempferol and quercetin, bestow biological activities

Kavyashree Doreswamy, Priyanka Shenoy, Sneha Bhaskar, Ramachandra K. Kini, Shailasree Sekhar

Characterization of the crude extract of Portulaca oleracea and the determination of the polyphenol oxidase kinetics in the presence of Cu and Zn

Omar Mohammad Atrooz, Shada Zaher Al-Maitah

Effect of diverse fermentation treatments on nutritional composition, bioactive components, and anti-nutritional factors of finger millet (Eleusine coracana L.)

Sumaira Jan, Krishan Kumar, Ajar Nath Yadav, Naseer Ahmed, Priyanka Thakur, Divya Chauhan, Qurat-Ul-Eain Hyder Rizvi, Harcharan Singh Dhaliwal

Optimization of active antioxidative defatted Canarium indicum L. (Canary) protein hydrolysate production

Cintya Nurul Apsari,, Ilma Nugrahani, Sukrasno, Tutus Gusdinar

Elemental, nutritional, and phytochemical profiling and antioxidant activity of Cordia obliqua Willd. (Clammy Cherry): An important underutilized forest tree of East India

Mamta Naik#,, Shashikanta Behera#,,, Sadhni Induar, Swaraj K. Babu, Pradeep K. Naik

Effects of enzymatic hydrolysis on the antioxidant activity of protein hydrolysate derived from the larvae of black soldier fly (Hermetia illucens L.)

Muhammad Yusuf Abduh,, Diah Ayu Prawitasari,, Ula Aulia Fitrian,, Mochamad Firmansyah,

Evaluation of functional characteristics of roselle seed and its use as a partial replacement of wheat flour in soft bread making

Nguyen Minh Thuy, Nguyen Bao Tram, Dinh Gia Cuong, Huynh Khanh Duy, Ly Thanh An, Vo Quoc Tien, Tran Ngoc Giau, Ngo Van Tai

Total phenolic, flavonoid contents, and antioxidant activity of three selected Portulaca grandiflora mutants in MV8 generation as a result of recurrent irradiation technique

Waras Nurcholis,, Syarifah Iis Aisyah, Regina Agritena Mayrischa Saraswati, Yoshua Shandy Yudha

Insights into the impact of spermidine in reducing salinity stress in Gerbera jamesonii

Javeria Uzma, Sai Krishna Talla, Praveen Mamidala

In vitro antioxidant and antibacterial potential of biosynthesized yttrium oxide nanoparticles using floral extract of Illicium verum

Karthikeyan Kandasamy, Premkumar Kumpati

Effect of combined NPK fertilizer on polyphenol contents and antioxidant activity in methanol extract of Curcuma xanthorhiza

Minarni Minarni, Rayandra Asyhar, Amira Amandanisa, Sintya Ainun, Yoshua Shandy Yudha, I Made Artika,, Waras Nurcholis,

Dehydration kinetics of green banana slices, characterization of optimized product based on physicochemical, nutritional, optical, and sensory attributes

Ram Kaduji Gadhave, Ravneet Kaur, Rahul Das, Kamlesh Prasad

Assessment of morpho-agronomic and yield attributes in gamma-irradiated mutants of Kalanamak rice (Oryza sativa L.)

Tanmai Mishra, Anjali Singh, Virendra Kumar Madhukar, Ashutosh Kumar Verma, Shambhavi Mishra, Rajveer Singh Chauhan

Metabolic profile, bioactivities, and variations in chemical constituents of essential oils of twenty mango ginger (Curcuma amada) accessions

Jyotirmayee Lenka, Snehalata Khuntia, Basudeba Kar, Suprava Sahoo

Phenolic compounds and in vitro antioxidant activity of spray-dried and freeze-dried aqueous extracts of sea cucumber (Holothuria tubulosa)

Fadna Aatab, Fatima Bellali, Fatima Zahra Aboudamia, Ahmed Errhif, Mariem Kharroubi

Optimization of pasteurization process of the ready-to-drink beverage from Hong Quan (Flacourtia jangomas) fruit by response surface methodology

Tan Duy Nguyen,, Tuyen Thi Xuan Vo,, Khang Nghia Tran,

Increasing polyphenol antioxidant in Orthosiphon stamineus Benth leaves with fermentation extraction by Saccharomyces cerevisiae ATCC-9763

Muhammad Aria Chandra, Khaswar Syamsu, Laksmi Ambarsari, Nurul Fatimah, Waras Nurcholis,

Phytochemical composition and antiproliferative activity of Opuntia elatior Mill.: In vitro and in silico studies on breast cancer cell line MCF-7

Foram Patel, Khushali Upadhyay, Denni Mammen, Elizabeth Robin, A.V. Ramachandran, Darshee Baxi

Bioactive properties of the extracts of peels, pomace, seeds, and essential oils of Citrus limon and Citrus aurantifolia

Folasade Oluwatobi, Olakunle Afolabi, Pius Okiki, Funmilayo Adeniyi, Oghenerobor Akpor

High resolution-liquid chromatograph mass spectrometer characterization of bioactive compounds in pineapple wastes: Valorization of antioxidant and enzymatic activity

Suman Polaki, Sourav Nayak, K. Sampad Kumar, Rabi Prasad B

Recent important insight into nutraceuticals potential of pigmented rice cultivars: A promising ingredient for future food

Le Thi Kim Loan, Bui The Vinh, Ngo Van Tai

Solid-state fermentation of pigment producing endophytic fungus Fusarium solani from Madiwala lake and its toxicity studies

Bhoomika Prakash Poornamath, Suma Sarojini, Saranya Jayaram, Soma Biswas, Anand Kaloor, Mridul Umesh

Elucidation of antioxidant compounds recovery capacity from “Cam” purple rice bran by different sustainable extraction techniques

Le Thi Kim Loan, Bui The Vinh, Ngo Van Tai

Secondary metabolite profiles, antimicrobial and antioxidant activities of callus, and leaves extract of Piper sarmentosum Roxb.

Junairiah Junairiah, Listijani Suhargo, Tri Nurhariyati, Nabilah Istighfari Zuraidassanaaz

Antimicrobial, anti-inflammatory, and anticancer activities of leaves extracts of Filicium decipiens

Atiyaparveen I Basarikatti, Sanjay Mishra, Vijaykumar Uppar, Basavaraj Padmashali

Emerging microplastic contamination in ecosystem: An urge for environmental sustainability

Akanksha Saini, Jai Gopal Sharma

Arsenic contamination in the food chain: A threat to food security and human health

Siril Singh, Rajni Yadav, Sheenu Sharma, Anand Narain Singh

Biotechnological potential of secondary metabolites: Current status and future challenges

Sofia Sharief Khan, Divjot Kour, Seema Ramniwas, Shaveta Singh, Sanjeev Kumar, Satvinder Kour, Roshi Sharma, Harpreet Kour, Shafaq Rasool, Sarvesh Rustagi, Sangram Singh, Kundan Kumar Chaubey, Ashutosh Kumar Rai, Ajar Nath Yadav