227
Views
9
CrossRef citations to date
0
Altmetric
Review

The Role of the Reactive Oxygen Species Scavenger Agent, Astaxanthin, in the Protection of Cisplatin-Treated Patients Against Hearing Loss

, &
Pages 4291-4303 | Published online: 18 Dec 2019

Abstract

Emerging evidence of significant hearing loss occurring shortly after cisplatin administration in cancer patients has stimulated research into the causes and treatment of this side effect. Although the aetiology of cisplatin-induced hearing loss (CIHL) remains unknown, an increasing body of research suggests that it is associated with excessive generation of intracellular reactive oxygen species (ROS) in the cochlea. Astaxanthin, a xanthophyll carotenoid, has powerful anti-oxidant, anti-inflammatory, and anti-apoptotic properties based on its unique cell membrane function, diverse biological activities, and ability to permeate the blood-brain barrier. In this review, we summarize the role of ROS in CIHL and the effect of astaxanthin on inhibiting ROS production. We focus on investigating the mechanism of action of astaxanthin in suppressing excessive production of ROS.

Introduction

Cisplatin, an effective antineoplastic agent commonly used in clinical practice, has many serious adverse effects including nephrotoxic, neurotoxic, and ototoxic effects. These life-long disabling adverse effects are strongly associated with the dosage, frequency, and duration of cisplatin treatment. Cisplatin-induced hearing loss (CIHL), which is permanent and mostly bilateral, can negatively affect academic development and social integration, especially in children.Citation1 To the best of our knowledge, cisplatin ototoxicity has not been studied in detail, and the mechanisms responsible for the degeneration of cochlear structures are not completely understood.

Emerging evidence indicates that excessive production of reactive oxygen species (ROS) contributes to cisplatin ototoxicity. Mechanistically, cisplatin ototoxicity is associated with the absence of glutathione (GSH) and the inhibition of glutathione peroxidase (GSH.Px) and glutathione reductase activities because cisplatin can covalently bind to the sulfhydryl groups of anti-oxidant enzymes, causing enzyme inactivation.Citation2 Increased lipid peroxidation in the cochlea inhibits essential cellular enzymes and membrane transporters, thereby disturbing ion channel function. Increased ROS production eventually results in apoptosis and necroptosis, supporting the hypothesis that ROS play a crucial role in cisplatin ototoxicity and suggesting that inhibiting ROS production could be beneficial for protecting the cochlea and reversing hearing loss.

Astaxanthin is a red carotenoid agent with potent anti-oxidant properties that can scavenge singlet oxygen and free radicals. These properties confer astaxanthin with anti-inflammatory and immunomodulatory activities, protective effects against neuronal damage, anti-aging and anti-cancer activities, and the ability to inhibit cell membrane peroxidation. The anti-oxidant activity of astaxanthin is 10-fold greater than that of zeaxanthin, lutein, canthaxanthin, and β-carotene, and 100-fold greater than that of α-tocopherol.Citation3 Growing evidence suggests that astaxanthin inhibits the development of oxidative stress-associated diseases and mitochondrial dysfunction.Citation4 Moreover, powerful permeation of the blood-brain barrier (BBB) allows astaxanthin to act as a potent neuroprotective agent in mammals.

The use of cisplatin is limited by its ototoxicity and nephrotoxicity. Methods to increase diuresis, such as hydration, have the potential to reduce its nephrotoxicity. However, there are currently no effective FDA-approved treatments for ototoxicity. We reviewed the evidence supporting the ability of astaxanthin to inhibit ROS generation and prevent mitochondrial dysfunction and neurodegeneration. Based on this assessment, we hypothesized that astaxanthin may be effective for the prevention and treatment of CIHL. In this review, we focus on the following topics: (1) The mechanisms underlying cisplatin ototoxicity; (2) astaxanthin-based therapies for diseases related to excessive ROS production; (3) astaxanthin biochemistry and bioactivity; and (4) downstream pathways of astaxanthin contributing to the inhibition of ROS generation.

Mechanisms of Cisplatin Ototoxicity

An increasing body of research suggests that cisplatin ototoxicity is related to cellular hypersensitivity, although the precise cellular and molecular mechanisms remain unclear. Our understanding of the role of cisplatin in ototoxicity is limited; however, research suggests that cisplatin uptake plays a crucial role. A recent study detected residual platinum in the cochleae of mice and cancer patients receiving cisplatin chemotherapy months-to-years after the treatment.Citation5

Cisplatin Transportation

Cisplatin is a square planar complex of a bivalent platinum cation with two cis chloride ligands and two cis ammonia ligands.Citation6 The complex was originally assumed to enter cells by passive diffusion because its uptake is concentration-dependent and non-saturable.Citation7 However, subsequent studies showed that copper transporter 1 (CTR1),Citation8,Citation9 organic cation transporter 2 (OCT2),Citation10 mechanotransduction (MET)Citation11 and copper-extruding P-type ATPases (ATP7A and ATP7B)Citation12 coordinate the cellular uptake of cisplatin. Although there may be other channels involved in cisplatin transportation, they have yet to be identified.Citation13Citation16

CTR1, a high-affinity copper transporter, is highly expressed in outer hair cells, inner hair cells, stria vascularis, and spiral ganglion neurons,Citation8 and contributes to drug entry and cell apoptosis.Citation17 CTR1 is a major entry route for cisplatin in hair cells, and it can enhance the cytotoxicity and cellular uptake of cisplatin in cells and in mouse.Citation8 Coactivity of both CTR1 and OCT2 may lead to secondary damage in the stria vascularis and spiral ganglion.Citation8 Knockout of CTR1 in yeast was reported to increase cisplatin resistance and decrease the intracellular concentration of cisplatin.Citation18 Although increased expression of CTR1 may affect the intracellular concentration and distribution of cisplatin, it does not affect the ability of cisplatin to target DNA.Citation19

OCTs belong to the solute carrier (SLC) 22A family,Citation20 and three isoforms (OCT1–3) have been identified, which are mainly expressed in the kidneys and liver.Citation21Citation23 OCT2 plays a key role in cisplatin transportation and is also expressed in the organ of Corti and stria vascularis.Citation10,Citation24 In OCT1/2 double-knockout (KO) mice, cisplatin shows no ototoxicity and only mild nephrotoxicity.Citation10 OCT2 variants were also reported to impede CIHL in children and adult, which highlights its key role in cisplatin transportation in the cochlea.Citation25

The MET channel is a nonselective cation channel that allows calcium and other ions to cross the membrane.Citation18 In zebrafish, CIHL is associated with functional MET channels, and inhibition of MET channels by quinine or EGTA has protective effects against CIHL.Citation11 Furthermore, zebrafish mutants lacking MET channels are resistant to CIHL.Citation11 These studies suggest that MET channels contribute to the entry of cisplatin into hair cells.

A small increase in the expression of the copper transporter ATP7A resulted in resistance to clinically available platinum drugs in human 2008 ovarian carcinoma cells that may be attributed to ATP7A binding and sequestration of platinum compounds.Citation26 The HSC-4-R, OSC-19-R, and HOC313-R cell lines have acquired resistance to cisplatin that is related to ATP7B overexpression.Citation27 Both ATP7A and ATP7B are expressed in the organ of Corti, stria vascularis and spiral ganglion neurons.Citation28 Inhibition of Na+-K+-ATPase reduces cisplatin accumulation, suggesting the involvement of other transporters in cisplatin resistance.Citation29

It has been suggested that dysfunction of the LRRC8A and LRRC8D subunits of heteromeric volume-regulated anion channels (VRAC) can lead to cisplatin resistance in KCP-4 human epidermoid cancer cell line and human lung adenocarcinoma cells.Citation30,Citation31 In 2015, Planells‐Cases reported that the absence of VRAC is related to cisplatin resistance in haploid KBM7 cells based on genome-wide loss-of-function screening.Citation32 However, the role of VRAC in cisplatin resistance is still unclear. Overexpression of multidrug resistance protein 2 (MRP2) increases the efflux of cisplatin in hepatocellular carcinoma and oesophageal squamous cell carcinoma.Citation33,Citation34

After entry into the cell, cisplatin undergoes aquation to form [Pt (NH3)2Cl (OH2)] + and [Pt (NH3)2(OH2)2]2+ (). The complex interacts with various reactive groups, blocking DNA replication and transcription and resulting in the inhibition of DNA repair and cell cycle progression.Citation7 Platinum-DNA adducts found in the hair cells of the cochlea and the marginal cells of the stria vascularis contribute to the inhibition of cell proliferation.Citation7 DNA damage caused by the formation of DNA adducts induces apoptosis initiated by activation of p53, which activates apoptotic proteins such as Bax or inhibits anti-apoptotic members of the Bcl-2 familyCitation35 (). Cisplatin-induced DNA damage in the cochlea may interfere with signal transduction pathways, including the v-akt murine thymoma viral oncogene homologue (AKT), v-abl Abelson murine leukaemia viral oncogene homologue 1 (c-ABL), p53, and mitogen-activated protein kinase/c-Jun N-terminal kinase/extracellular signal–regulated kinase (MAPK/JNK/ERK) pathways.Citation7,Citation36

Figure 1 Schematic of the proposed mechanism of cisplatin transportation and the generation of ROS in CIHL. Cisplatin is transported into cochlear cells by membrane transporters, including copper transporter 1 (CTR1), organic cation transporter 2 (OCT2), and mechanoelectrical transduction (MET), and is excluded by copper-extruding P-type ATPases (ATP7A and ATP7B), multidrug resistance protein 2 (MRP2), and volume-regulated anion channels (VRAC). Cisplatin induces ROS production in the inner ear via NADPH oxidase (NOX), xanthine oxidase (XO), cytochrome P450 (CYP450), induced nitric oxide synthase (iNOS), and disturbances in the mitochondrial electron transport chain.

Abbreviations: IL, interleukin; STAT1, signal transducer and activator of transcription 1.
Figure 1 Schematic of the proposed mechanism of cisplatin transportation and the generation of ROS in CIHL. Cisplatin is transported into cochlear cells by membrane transporters, including copper transporter 1 (CTR1), organic cation transporter 2 (OCT2), and mechanoelectrical transduction (MET), and is excluded by copper-extruding P-type ATPases (ATP7A and ATP7B), multidrug resistance protein 2 (MRP2), and volume-regulated anion channels (VRAC). Cisplatin induces ROS production in the inner ear via NADPH oxidase (NOX), xanthine oxidase (XO), cytochrome P450 (CYP450), induced nitric oxide synthase (iNOS), and disturbances in the mitochondrial electron transport chain.

ROS in CIHL

Normally, the maintenance of cochlea function requires high metabolic activity in areas such as the stria vascularis, spiral ligament, and spiral prominence, leading to leakage of electrons from the mitochondrial respiratory chain, which react with oxygen (O2) to produce superoxide (O2•). Environmental stimuli increase oxidative stress in the cochlea in association with increased metabolic activity. The metabolic requirements of the cochlea lead to vulnerability to hypoxic events and ischemia-reperfusion injury.Citation37 Cisplatin promotes the generation of ROS by stimulating enzyme systems or deactivating anti-oxidant systems.Citation38,Citation39 This is supported by decreased cochlear GSH and anti-oxidant enzyme activity in rats treated with cisplatin.Citation40 This decrease could result from covalent binding of cisplatin to anti-oxidant enzymes (e.g., superoxide dismutase and catalase), loss of metal cofactors, depletion of anti-oxidant enzymes by increased ROS, and depletion of cochlear anti-oxidant enzyme cofactors such as GSH and NADPH, which protect against the toxicity of ROS and allow the regeneration of GSH.Citation41

NADPH oxidases (NOXs) are membrane-bound, multi-subunit enzyme complexes that face the extracellular space and transfer electrons across the plasma membrane from NADPH to molecular oxygen, generating superoxide in the cell.Citation42 Inactivate NOX is present in an unassembled form in which p22phox and gp91phox are present in the membrane, whereas p47phox, p67phox, and p40phox exist in the cytosol. When NOX is activated by phosphorylated p47phox, the cytosolic subunits translocate to the membrane and convert the oxidase into an assembled and active form that can transfer electrons from the substrate to O2, forming O2•−.Citation43 Some members of the NOX family may be responsible for the bulk of ROS generation in cochlea cells.Citation13,Citation44,Citation45 NOX3-dependent ROS generation may be a target of cisplatin, as indicated by evidence showing that knockdown of NOX3 by trans-tympanic delivery of siRNA attenuates cisplatin ototoxicity in rats.Citation44,Citation46 NOX3-derived ROS can activate signal transducer and activator of transcription 1 and 6 (STAT1 and STAT6), increasing the production of the pro-inflammatory cytokines tumour necrosis factor alpha (TNFα), interleukin (IL)-1b, and IL-6.Citation47,Citation48 These pro-inflammatory mediators exacerbate CIHL by increasing the activity of NOX isoforms including NOX1 and NOX4.Citation45 In addition, NOX3 expression can be suppressed by silencing the transient receptor potential vanilloid 1 (TRPV1) channel, which acts as a key factor contributing to ROS generation in cochlear hair cells.Citation13 siRNA-mediated silencing of STAT1 abolishes cisplatin-induced p53 activation, consistent with a study showing that STAT1 regulates cisplatin-induced hair cell death.Citation48,Citation49 These findings suggest that the TRPV1 and NOX3 signalling pathways may be associated with STAT1, resulting in inflammation and cisplatin-induced hair cell death leading to hearing loss. Increased NOX2 expression in the cochlea increases ROS production in outer hair cells of the basal turn, which is an important factor associated with the vulnerability of outer hair cells.Citation50

The xanthine/xanthine oxidase (XO) system is another active ROS-generating system found in the cochlea that contributes to both superoxide and hydrogen peroxide generation. XO is a xanthine oxidoreductase isoform that catalyses the reduction of O2 into O2•− and H2O2. Its activity may be enhanced by ROS derived from mitochondria and NOX.Citation51 Inhibition of XO in a rat model administered ebselen, a GSH.Px mimetic, decreases cisplatin ototoxicity and nephrotoxicity.Citation52 Depletion of intracellular ATP transforms adenine nucleotides into hypoxanthine and xanthine, which are substrates of XO.Citation53 This is proposed as a principal mechanism underlying oxidative injury.

Disturbance of the mitochondrial electron transport chain system by cisplatin increases ROS generation concomitant with a decrease in the mitochondrial membrane potential (MMP), an indicator of mitochondrial malfunction.Citation54,Citation55 Disruption of Complex I function resulting in elevated ROS production underlies the destruction of sensory hair cells in the cochlea caused by cisplatin.Citation56 In addition, ROS-mediated mitochondrial dysfunction can suppress the amplification of intracellular ROS generation, resulting in hair cell apoptosis and necrosis.Citation57

Evidence supports an association between the toxic effects of cisplatin and cytochrome P450 (CYP450) enzymes.Citation58 However, there is little research on the relation between CYP450 and CHIL. CYP450 is an important source for the generation of ROS and catalytic iron in cisplatin-induced cytotoxicity of the LLC-PK1 cells.Citation59 Liu found that CYP450 subfamily members (CYP2e1) play a pivotal role in cisplatin-induced nephrotoxicity and apoptosis related to the generation of ROS.Citation60 CYP2e1 null mice showed significantly functional and histologic protection against renal injury and decrease of apoptosis by cisplatin.Citation60 Increasing CYP2E1 enhances the toxicity induced by cisplatin in liver injury models both in vitro and in vivo, and the mechanism might associate with accumulating production of ROS and oxidative stress.Citation58 E47 HepG2 cells, overexpress human CYP2E1, were sensitive to cisplatin because of an earlier activation of ERK.Citation61 These results suggest that the generation of ROS by cisplatin in the cochlea is associated with CYP450.

Induced nitric oxide synthase (iNOS) and direct NO generation can be observed in the inner ear (including the spiral ligament, modiolus, spiral limbus, supporting cells, nerve fibres, stria vascularis, hair cells, and spiral ganglion neurons) in response to cisplatin administration.Citation62Citation64 ROS/reactive nitrogen species (RNS), as intracellular second messengers, play a pivotal role in ototoxicity. ROS/RNS can activate downstream signalling pathways, regulate gene expression, and interfere directly with lipids, DNA, RNA, metal cofactors and proteins. Nitrosative stress can react with and suppress ROS generation, overpowering anti-oxidant defence capacities within the inner ear, triggering the pro-apoptotic pathway, and leading to outer hair cell death.Citation65,Citation66

Although our understanding of the mechanism by which ROS exert their effects in the cochlea is still incomplete, it is well established that oxidative stress can cause DNA damage. It is also evident that the DNA damage caused by ROS may be repaired via several pathways, including base excision repair (BER),Citation67Citation69 mismatch repair (MMR),Citation70 and nucleotide excision repair (NER).Citation71 The molecule 8-oxoguanine (8-oxo-G) is well known to cause DNA damage by creating a gap in the DNA, which can be repaired by the enzyme 8-oxoguanine glycosylase (8-OGG1) in mitochondria and nucleus.Citation72 Chronic exposure to a low dose of H2O2 induces a DNA protective response in C2C12 cells by activating transcription factors that enhance the expression of DNA repair enzymes.Citation73,Citation74 An increase in poly-ADP-ribose transferase 1 (PARP1), an enzyme that recruits DNA repair proteins, has been detected in a mouse model of CHIL, and its inhibition is associated with decreased cisplatin-induced cell death.Citation75 HEI-OC1 cells exposed to cisplatin can promote and initiate DNA repair, but cannot prevent or reverse DNA damage,Citation76 possibly because of the absence of some DNA repair pathways in mitochondria and BER can only cope with minor damage or process single nucleotides in the mitochondria.Citation72

Thus, the cisplatin complex leads to reciprocal activation of ROS generation, NOX, XO, mitochondria, CYP450, MMP, and iNOS as well as pro-inflammatory signalling, suggesting that cisplatin initiates a series of vicious cycles (). In turn, ROS enhance lipid peroxidation and DNA damage, finally leading to auditory cell death.

Astaxanthin-Based Therapies for Diseases Related to ROS

Oxidative stress leads to disease conditions that result from increased production of ROS or depletion of the anti-oxidant enzyme system. Mitochondrial disturbance is often involved in the onset of oxidative stress-associated diseases, because the mitochondria are responsible for energy generation and are important sources of ROS. Astaxanthin, which has strong anti-oxidant activity and the ability to maintain metabolic efficiency, is a potent anti-oxidant with the potential to target several health conditions.Citation77

Because nervous system tissues show intense metabolic aerobic activity, rich irrigation with blood vessels, and are abundant in unsaturated fats and iron, they are particularly susceptible to oxidative damage.Citation78 Substantial evidence supports the hypothesis that oxidative stress and impaired mitochondrial efficiency can be causative or at least ancillary factors in the pathogenesis of major neurodegenerative diseases, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s, and amyotrophic lateral sclerosis.Citation79Citation82 Anti-oxidants can improve mitochondrial redox potential, which is a key target of ROS and free radical production.Citation83 A number of studies have shown that diets high in anti-oxidants can reduce these associated risks.Citation84 Synthesized docosahexaenoic acid-acrylated astaxanthin diesters can relieve oxidative stress and inflammasome activation in patients with AD.Citation85 Astaxanthin inhibits the generation of intracellular ROS and protects against 1-methyl-4-phenylpyridinium (MPP+)-induced cytotoxicity in a cellular model of PD, protecting SH-SY5Y cells and substantia nigra neurons from apoptosis in a PD model mouse.Citation80,Citation86 H2O2-stimulated mouse neural progenitor cells pre-treated with astaxanthin show suppression of apoptosis, resulting in cell proliferation.Citation87 Recent rat models show that astaxanthin can ameliorate aluminium-induced impaired memory performance.Citation88 According to studies of rats fed with natural astaxanthin, astaxanthin can cross the BBB in mammals, and its anti-oxidant potential may extend beyond the BBB, allowing it to act as a potent neuroprotective agent.Citation89 In a murine model of ischemic stroke, pre-treatment with astaxanthin decreased ROS production, lipid peroxidation, and cerebral infarction and promoted locomotor function recovery.Citation90 Astaxanthin was therefore suggested as a promising candidate neuroprotective agent in mammals.Citation91

Elevated oxidative stress associated with ROS/RNS and chronic inflammation can aggravate cardiovascular diseases. Emerging in vitro- and in vivo-based evidence indicates that astaxanthin can reduce ROS, RNS, lipid peroxidation, and inflammatory signalling, and activate anti-oxidant enzymes in the heart.Citation92Citation96 Investigations of the effect of astaxanthin on the myocardium of ischemic mice demonstrated that astaxanthin can significantly prevent ischemic myocardial injury by alleviating mitochondrial impairment in mitochondria with increased ROS expression, mitochondrial depolarization, and swelling.Citation97 Astaxanthin promoted recovery of mitochondrial integrity and blocked mitochondria-mediated apoptosis in a homocysteine-induced cardiotoxicity model resulting from overexpression of ROS, loss of MMP, and fragmentation of mitochondria.Citation98 In addition, astaxanthin balanced the expression of Bcl-2 family proteins, thereby suppressing mediators of mitochondrial apoptosis such as PARP1 and caspases.Citation98 Studies also suggest that dietary astaxanthin has beneficial effects during chemotherapy in BALB/c mice, not only because it can counteract induced oxidative stress, but also because it has cardioprotective effects.Citation94 Epidemiological and clinical data indicate that dietary anti-oxidants might protect against cardiovascular disease, reducing the risk of atherosclerosis by decreasing plasma LDL-cholesterol oxidation.Citation99

Astaxanthin may be more effective than vitamin E for preventing and treating non-alcoholic steatohepatitis in mice and humans.Citation100 It protects the liver against acetaminophen hepatotoxicity by alleviating hepatocyte necrosis, blocking ROS generation, inhibiting oxidative stress, and reducing apoptosis.Citation101 Astaxanthin accumulates in the liver, especially in the microsomal and mitochondrial fractions of liver tissues.Citation102 These studies indicate the potential of astaxanthin for the treatment of oxidative stress-related liver and heart diseases.

Astaxanthin also has shown biological activity in dermatology clinical trials, promoting skin health and achieving effective skin cancer chemoprevention.Citation103 Randomized double-blinded, controlled studies reported that astaxanthin can ameliorate skin wrinkles, elasticity, texture, and viscoelasticity affected by aging and associated with oxidative metabolism and subsequent ROS production.Citation104Citation107 A recent study using a rodent model of deep burns revealed that astaxanthin protects against early progression of a burn wound by reducing ROS-induced inflammation and apoptosis.Citation108 Moreover, it was reported that astaxanthin powerfully accelerates wound recovery in mice.Citation109

In addition to its strong antioxidative effect, there is increasing evidence to show that astaxanthin can inhibit the growth of several types of cancer.Citation110 Studies have shown that ROS may be involved in cancer initiation, progression, and proliferation by acting as messengers of the oxidative stress cascade, which can be inhibited by anti-oxidants such as astaxanthin.Citation110,Citation111 In the hamster buccal pouch carcinogenesis model, it was shown that astaxanthin prevents the growth of oral cancer by suppressing cell proliferation and inducing apoptosis.Citation112 In 1,2-dimethyl hydrazine-induced rat colon carcinoma and rat hepatocellular carcinoma CBRH-7919 cells, pre-treatment with astaxanthin exerted anti-cancer effects by inducing cellular apoptosis.Citation113,Citation114 Astaxanthin also enhanced apoptosis of leukaemia K562 cells and interfered with cell cycle progression.Citation115 In ovarian cancer SKOV3 cells, astaxanthin combined with human serum albumin could be a candidate treatment via arresting the cell cycle in the G1 phase and inducing apoptosis.Citation116

These positive effects have been attributed to the ability of astaxanthin to scavenge free radicals, quench singlet oxygen, and inhibit lipid peroxidation. The effect of astaxanthin on protecting cellular membranes against oxidation is related to the protection of the inner part and external surface.Citation117Citation119 Anti-oxidants are indispensable for maintaining cellular health by protecting cellular components against oxidative stress and suppressing ROS-induced inflammation. The effects of anti-oxidants are evident in inflammation-related clinical conditions, such as Crohn’s disease, asthma, and exercise-induced muscle damage.Citation120,Citation121 In 2000, Bennedsen reported that dietary astaxanthin helped reduce the symptoms of ulcerative disease and gastric inflammation.Citation122 Because of these effects, astaxanthin should be considered for the treatment of various ROS-related diseases.

Astaxanthin Biochemistry and Bioactivity

Astaxanthin is the main carotenoid pigment produced in marine animals, and it is present in many types of seafood, such as salmon, trout, shrimp, and lobster. Astaxanthin has three natural stereoisomers [(3S, 3′S), (3R, 3′R), and (3R, 3′S)], which vary in the configuration of the two hydroxyl groups on the molecule (). The presence of the hydroxyl and keto groups on each ionone ring confer unique features, including conversion into an ester, and providing greater anti-oxidant activity and more polar properties than other carotenoids.Citation3,Citation123 Astaxanthin can preserve the integrity of cell membranes by penetrating bilayers and protecting the redox state and functional integrity of mitochondria.Citation124

Figure 2 Chemical structure of astaxanthin (3,3ʹ-dihydroxy-β, β- carotene-4,4R′-dione).

Figure 2 Chemical structure of astaxanthin (3,3ʹ-dihydroxy-β, β- carotene-4,4R′-dione).

Astaxanthin is present in natural sources as an ester of fatty acids or as a conjugate of proteins in foods. Astaxanthin is absorbed into enterocytes through passive diffusion, and is incorporated into chylomicrons to be transferred to the liver. This natural product is then incorporated into low-density lipoprotein and high-density lipoprotein and transported via the circulation. Moreover, the bioavailability of astaxanthin is enhanced when it is taken with dietary lipids.Citation123 Astaxanthin does not turn into vitamin A in the human body; therefore, it is nontoxic if given orally. Even at low concentrations astaxanthin is effective because of its polar features, which optimize the rate and extent of absorption.Citation125 The only study on humans confirmed the bioavailability of astaxanthin administered at a single high dose of 100 mg, and demonstrated that is it transported to the plasma by lipoproteins.Citation126 Astaxanthin can be used as a dietary supplement for human, animal, and fishery consumption. The FDA has validated the use of astaxanthin for food colouring (or colour additive) and different applications in animal and fish food.

Downstream Pathways of Astaxanthin Against the Generation of ROS

Although the anti-oxidant properties of astaxanthin play a crucial role in its biological activity, the concentrations achieved in the blood that protect cells against oxidative injury are well below those required to scavenge free radicals directly. To explain this discrepancy, it has been suggested that astaxanthin increases resistance to oxidative stress by activating signalling pathways associated with cell survival ().

Figure 3 Proposed mechanism by which astaxanthin inhibits ROS and apoptosis. Astaxanthin inhibits apoptosis and scavenges ROS, and modulates various intracellular pathways, predominantly MAPK, PI3K/Akt and Nrf2/ARE. It can also activate the specificity protein 1 (Sp1)–NMDA receptor subunit 1 (NR1) signalling pathway, which leads to cell death.

Abbreviations: AST, astaxanthin; Akt, v-akt murine thymoma viral oncogene homologue; ARE, anti-oxidant response element; CREB, CAMP-responsive element binding protein; ERK, extracellular signal–regulated kinases; JNK, c-Jun N-terminal kinase; Keap1, cytosolic inhibitor of Nrf2; Nrf2, nuclear factor-erythroid 2-related factor 2; NF-κβ, nuclear factor-kappa B subunit; NQO1, NADH Quinone Dehydrogenase1; PTEN, phosphatase and tensin homolog.
Figure 3 Proposed mechanism by which astaxanthin inhibits ROS and apoptosis. Astaxanthin inhibits apoptosis and scavenges ROS, and modulates various intracellular pathways, predominantly MAPK, PI3K/Akt and Nrf2/ARE. It can also activate the specificity protein 1 (Sp1)–NMDA receptor subunit 1 (NR1) signalling pathway, which leads to cell death.

MAPK Pathway

Astaxanthin may help to prevent neurotoxicity by promoting the activation of the AKT/cyclic AMP-responsive element binding protein and ERKs, and blocking the activation of p38 MAPKs, which play pro-apoptotic roles, whereas ERKs have anti-apoptotic roles.Citation127 This is consistent with the observation that astaxanthin decreases ROS production and inflammatory cytokines to inhibit apoptosis and autophagy, which may be related to inactivation of components of the MAPK family, such as p38 MAPK, JNK and ERK, in a model of hepatic ischemia reperfusion injury.Citation128 In addition, inhibition of the TNF-α-mediated JNK signal pathway and phosphorylation of ERK and p38 MAPK are involved in the process of hepatocyte necrosis induced by acetaminophen.Citation101 In a mouse model of smoke-induced impairment of cognitive function, astaxanthin protects the brain against neuroinflammation, synaptic plasticity impairment, and oxidative stress in the cortex and hippocampus by inhibiting p38 MAPK.Citation129 In human umbilical vein endothelial cells (HUVECs), however, Western blot analysis revealed that astaxanthin significantly upregulated p-ERK without affecting p38 MAPK. Although expression of p-Akt was also increased, it was not significant.Citation130 These results suggest that astaxanthin protects cells against oxidative stress-induced apoptosis.

Nrf2/ARE Pathway

Hydroxypropyl-β-cyclodextrin-astaxanthin (CD-A) can lead to the dissociation of nuclear factor-erythroid 2-related factor 2 (Nrf2) from Keap1, which allows Nrf2 to translocate to the nucleus and bind to anti-oxidant response elements (AREs), thereby inducing an endogenous anti-oxidant response caused by heme oxygenase-1 (HO-1) and NAD(P)H Quinone Dehydrogenase 1 (NQO1).Citation131 HO-1 is responsible for many oxidative and cytoprotective functions, and NQO1 has anti-inflammatory effects and can prevent the reduction of quinone.Citation132,Citation133 Astaxanthin pre-treatment significantly increased the expression of Nrf2, HO-1, and NQO1 mRNA, exerting a protective effect against brain injuries. This was demonstrated in a cerebral ischemia rat model, rat hepatocytes, the human retinal pigment epithelial cell line ARPE-19, and early brain injury in a prechiasmatic cistern model of subarachnoid haemorrhage.Citation131,Citation134,Citation135 In HUVECs, astaxanthin activates the Nrf-2/ARE signalling pathway by developing small amounts of ROS, whereas knockdown of Nrf-2 by siRNA inhibits HO-1 mRNA expression.Citation130 However, the direct molecular targets responsible for induction of the Nrf2/HO-1/NQO1 pathway remain undefined, as astaxanthin has an indirect anti-oxidant protective effect against ROS.

PI3K/AKT Pathway

Previous studies indicate that cell survival is affected by intracellular ROS generation through the modulation of the phosphatase and tensin homolog (PTEN)/phosphoinositide 3-kinase (PI3K)/AKT pathway.Citation136 Astaxanthin protects against isoflurane-induced neuroapoptosis in a rat model, as indicated by decreased brain damage, inhibition of caspase-3 activity, and upregulation of the PI3K/AKT pathway.Citation137 Zuluaga recently reported that the generation of ROS induced by stressors (AAPH and t-BuOOH, which are free radical donors that generate a burst of ROS) upregulates PTEN gene expression, which causes cellular apoptosis by deactivating AKT.Citation138 Conversely, astaxanthin treatment significantly suppressed PTEN expression and reduced both eNOS and Bax gene expression in endothelial cells under oxidative stress.Citation138 Astaxanthin can activate the PI3K/Akt pathway, protecting against H2O2-induced oxidative stress through the Nrf2/ARE pathway in ARPE-19 cells.Citation139

Astaxanthin activates the specificity protein 1 (Sp1) and NMDA receptor subunit 1 (NR1) signalling pathway, inhibiting the upregulation and nuclear transfer of Sp1 resulting from MPP+-induced production of intracellular ROS and cytotoxicity in PC12 cells.Citation140

Conclusion

Astaxanthin possesses ROS scavenging and anti-oxidant activities, and thus inhibits oxidative stress-induced mitochondrial dysfunction and ROS production in cells caused by various stimuli. We propose that astaxanthin treatment might be a viable approach for the effective mitigation and prevention of CIHL associated with ROS. Astaxanthin may be an excellent candidate for treating CHIL, as it is a safe nutrient with no toxicity when consumed with food, and it has the ability to pass through the BBB because of its lipid solubility. Future studies should investigate the protective properties and underlying mechanisms of astaxanthin, which may contribute to the use of astaxanthin as an otoprotective agent.

The otoprotective effects of astaxanthin have been examined in a zebrafish model, although research into its otoprotective effects is limited.Citation141 Future studies should focus on the pharmaceutical potential and effects of astaxanthin for the treatment of hearing loss, particularly because astaxanthin can be easily absorbed, and when metabolized, it may have greater biological activity than its free form.Citation3 Future studies should include a therapeutic time window, reliability of drug administration routes, and the optimal dosages of astaxanthin. The design of clinical trials to assess the potential of astaxanthin for the treatment of ototoxicity is warranted based on evidence showing its safety. In addition, the efficacy of astaxanthin for the treatment of neurological diseases should promote clinical trials of this compound for the treatment of other diseases.

Author Contributions

All authors contributed to data analysis, drafting or revising the article, gave final approval for publication, and agree to be accountable for all aspects of the work.

Disclosure

The authors report no conflicts of interest in this work.

References

  • Knight KRG, Kraemer DF, Neuwelt EA. Ototoxicity in children receiving platinum chemotherapy: underestimating a commonly occurring toxicity that may influence academic and social development. J Clin Oncol. 2005;23(34):8588–8596. doi:10.1200/Jco.2004.00.535516314621
  • Sheth S, Mukherjea D, Rybak LP, Ramkumar V. Mechanisms of cisplatin-induced ototoxicity and otoprotection. Front Cell Neurosci. 2017;11:338. doi:10.3389/fncel.2017.0033829163050
  • Ambati RR, Phang SM, Ravi S, Aswathanarayana RG. Astaxanthin: sources, extraction, stability, biological activities and its commercial applications–a review. Mar Drugs. 2014;12(1):128–152. doi:10.3390/md1201012824402174
  • Kuroki T, Ikeda S, Okada T, et al. Astaxanthin ameliorates heat stress-induced impairment of blastocyst development in vitro:–astaxanthin colocalization with and action on mitochondria. J Assist Reprod Genet. 2013;30(5):623–631. doi:10.1007/s10815-013-9987-z23536152
  • Breglio AM, Rusheen AE, Shide ED, et al. Cisplatin is retained in the cochlea indefinitely following chemotherapy. Nat Commun. 2017;8(1):1654. doi:10.1038/s41467-017-01837-129162831
  • Dasari S, Tchounwou PB. Cisplatin in cancer therapy: molecular mechanisms of action. Eur J Pharmacol. 2014;740:364–378. doi:10.1016/j.ejphar.2014.07.02525058905
  • Wang D, Lippard SJ. Cellular processing of platinum anticancer drugs. Nat Rev Drug Discov. 2005;4(4):307–320. doi:10.1038/nrd169115789122
  • More SS, Akil O, Ianculescu AG, Geier EG, Lustig LR, Giacomini KM. Role of the copper transporter, CTR1, in platinum-induced ototoxicity. J Neurosci. 2010;30(28):9500–9509. doi:10.1523/JNEUROSCI.1544-10.201020631178
  • Shen DW, Pouliot LM, Hall MD, Gottesman MM. Cisplatin resistance: a cellular self-defense mechanism resulting from multiple epigenetic and genetic changes. Pharmacol Rev. 2012;64(3):706–721. doi:10.1124/pr.111.00563722659329
  • Ciarimboli G, Deuster D, Knief A, et al. Organic cation transporter 2 mediates cisplatin-induced oto- and nephrotoxicity and is a target for protective interventions. Am J Pathol. 2010;176(3):1169–1180. doi:10.2353/ajpath.2010.09061020110413
  • Thomas AJ, Hailey DW, Stawicki TM, et al. Functional Mechanotransduction Is Required for Cisplatin-Induced Hair Cell Death in the Zebrafish Lateral Line. J Neurosci. 2013;33(10):4405–4414. doi:10.1523/Jneurosci.3940-12.201323467357
  • La Fontaine S, Mercer JF. Trafficking of the copper-ATPases, ATP7A and ATP7B: role in copper homeostasis. Arch Biochem Biophys. 2007;463(2):149–167. doi:10.1016/j.abb.2007.04.02117531189
  • Mukherjea D, Jajoo S, Whitworth C, et al. Short interfering RNA against transient receptor potential vanilloid 1 attenuates cisplatin-induced hearing loss in the rat. J Neurosci. 2008;28(49):13056–13065. doi:10.1523/JNEUROSCI.1307-08.200819052196
  • Takumida M, Ishibashi T, Hamamoto T, Hirakawa K, Anniko M. Age-dependent changes in the expression of klotho protein, TRPV5 and TRPV6 in mouse inner ear. Acta Oto-Laryngol. 2009;129(12):1340–1350. doi:10.3109/00016480902725254
  • Takumida M, Anniko M. Expression of canonical transient receptor potential channel (TRPC) 1-7 in the mouse inner ear. Acta Oto-Laryngol. 2009;129(12):1351–1358. doi:10.3109/00016480902798350
  • Takumida M, Anniko M. Expression of transient receptor potential channel mucolipin (TRPML) and polycystine (TRPP) in the mouse inner ear. Acta Oto-Laryngol. 2010;130(2):196–203. doi:10.3109/00016480903013593
  • Ishida S, Lee J, Thiele DJ, Herskowitz I. Uptake of the anticancer drug cisplatin mediated by the copper transporter Ctr1 in yeast and mammals. P Natl Acad Sci USA. 2002;99(22):14298–14302. doi:10.1073/pnas.162491399
  • Waissbluth S, Daniel SJ. Cisplatin-induced ototoxicity: transporters playing a role in cisplatin toxicity. Hear Res. 2013;299:37–45. doi:10.1016/j.heares.2013.02.00223467171
  • Holzer AK, Samimi G, Katano K, et al. The copper influx transporter human copper transport protein 1 regulates the uptake of cisplatin in human ovarian carcinoma cells. Mol Pharmacol. 2004;66(4):817–823. doi:10.1124/mol.104.00119815229296
  • Budiman T, Bamberg E, Koepsell H, Nagel G. Mechanism of electrogenic cation transport by the cloned organic cation transporter 2 from rat. J Biol Chem. 2000;275(38):29413–29420. doi:10.1074/jbc.M00464520010889205
  • Pelis RM, Wright SH. SLC22, SLC44, and SLC47 transporters-organic anion and cation transporters: molecular and cellular properties. Curr Top Membr. 2014;73:233–261. doi:10.1016/B978-0-12-800223-0.00006-224745985
  • Schaeffeler E, Hellerbrand C, Nies AT, et al. DNA methylation is associated with downregulation of the organic cation transporter OCT1 (SLC22A1) in human hepatocellular carcinoma. Genome Med. 2011;3:82. doi:10.1186/gm29822196450
  • Bleasby K, Castle JC, Roberts CJ, et al. Expression profiles of 50 xenobiotic transporter genes in humans and pre-clinical species: a resource for investigations into drug disposition. Xenobiotica. 2006;36(10–11):963–988. doi:10.1080/0049825060086175117118916
  • Hellberg V, Gahm C, Liu W, Ehrsson H, Rask-Andersen H, Laurell G. Immunohistochemical localization of OCT2 in the cochlea of various species. Laryngoscope. 2015;125(9):E320–E325. doi:10.1002/lary.2530425892279
  • Lanvers-Kaminsky C, Sprowl JA, Malath I, et al. Human OCT2 variant c.808G > T confers protection effect against cisplatin-induced ototoxicity. Pharmacogenomics. 2015;16(4):323–332. doi:10.2217/Pgs.14.18225823781
  • Samimi G, Safaei R, Katano K, et al. Increased expression of the copper efflux transporter ATP7A mediates resistance to cisplatin, carboplatin, and oxaliplatin in ovarian cancer cells. Clin Cancer Res. 2004;10(14):4661–4669. doi:10.1158/1078-0432.CCR-04-013715269138
  • Yoshizawa K, Nozaki S, Kitahara H, Ohara T et al. Copper efflux transporter (ATP7B) contributes to the acquisition of cisplatin-resistance in human oral squamous cell lines. Oncol Rep. 2007;18(4):987–991. PMID:1778636417786364
  • Ding D, He J, Allman BL, et al. Cisplatin ototoxicity in rat cochlear organotypic cultures. Hear Res. 2011;282(1–2):196–203. doi:10.1016/j.heares.2011.08.00221854840
  • Tadini-Buoninsegni F, Sordi G, Smeazzetto S, Natile G, Arnesano F. Effect of cisplatin on the transport activity of P-II-type ATPases. Metallomics. 2017;9(7):960–968. doi:10.1039/c7mt00100b28636017
  • Min XJ, Li H, Hou SC, et al. Dysfunction of volume-sensitive chloride channels contributes to cisplatin resistance in human lung adenocarcinoma cells. Exp Biol Med (Maywood). 2011;236(4):483–491. doi:10.1258/ebm.2011.01029721454376
  • Lee EL, Shimizu T, Ise T, Numata T, Kohno K, Okada Y. Impaired activity of volume-sensitive Cl- channel is involved in cisplatin resistance of cancer cells. J Cell Physiol. 2007;211(2):513–521. doi:10.1002/jcp.2096117186499
  • Planells‐Cases R, Lutter D, Guyader C, et al. Subunit composition of VRAC channels determines substrate specificity and cellular resistance to Pt-based anti-cancer drugs. EMBO J. 2015;34(24):2993–3008. doi:10.15252/embj.20159240926530471
  • Yamasaki M, Makino T, Masuzawa T, et al. Role of multidrug resistance protein 2 (MRP2) in chemoresistance and clinical outcome in oesophageal squamous cell carcinoma. Br J Cancer. 2011;104(4):707–713. doi:10.1038/sj.bjc.660607121206495
  • Korita PV, Wakai T, Shirai Y, et al. Multidrug resistance-associated protein 2 determines the efficacy of cisplatin in patients with hepatocellular carcinoma. Oncol Rep. 2010;23(4):965–972. doi:10.3892/or_0000072120204280
  • Vaseva AV, Moll UM. The mitochondrial p53 pathway. Biochim Biophys Acta. 2009;1787(5):414–420. doi:10.1016/j.bbabio.2008.10.00519007744
  • Rybak LP, Ramkumar V. Ototoxicity. Kidney Int. 2007;72(8):931–935. doi:10.1038/sj.ki.500243417653135
  • Seidman MD, Quirk WS, Nuttall AL, Schweitzer VG. The protective effects of allopurinol and superoxide dismutase-polyethylene glycol on ischemic and reperfusion-induced cochlear damage. Otolaryngol Head Neck Surg. 1991;105(3):457–463. doi:10.1177/0194599891105003181945435
  • Kopke RD, Liu W, Gabaizadeh R, et al. Use of organotypic cultures of Corti’s organ to study the protective effects of antioxidant molecules on cisplatin-induced damage of auditory hair cells. Am J Otol. 1997;18(5):559–571. PMID:9303151 9303151
  • Clerici WJ, Yang LH. Direct effects of intraperilymphatic reactive oxygen species generation on cochlear function. Hearing Res. 1996;101(1–2):14–22. doi:10.1016/S0378-5955(96)00126-8
  • Rybak LP, Husain K, Morris C, Whitworth C, Somani S. Effect of protective agents against cisplatin ototoxicity. Am J Otol. 2000;21(4):513–520. PMID:1091269710912697
  • DeWoskin R, Riviere J. Cisplatin-induced loss of kidney copper and nephrotoxicity is ameliorated by single dose diethyldithiocarbamate, but not mesna. Toxicol Appl Pharmacol. 1992;112(2):182–189. doi:10.1016/0041-008x(92)90186-v1311464
  • Gao HM, Zhou H, Hong JS. NADPH oxidases: novel therapeutic targets for neurodegenerative diseases. Trends Pharmacol Sci. 2012;33(6):295–303. doi:10.1016/j.tips.2012.03.00822503440
  • Turrens JF, Boveris A. Generation of superoxide anion by the NADH dehydrogenase of bovine heart mitochondria. Biochem J. 1980;191(2):421–427. doi:10.1042/bj19104216263247
  • Mukherjea D, Jajoo S, Kaur T, Sheehan KE, Ramkumar V, Rybak LP. Transtympanic administration of short interfering (si)RNA for the NOX3 isoform of NADPH oxidase protects against cisplatin-induced hearing loss in the rat. Antioxid Redox Signal. 2010;13(5):589–598. doi:10.1089/ars.2010.311020214492
  • Kim HJ, Lee JH, Kim SJ, et al. Roles of NADPH oxidases in cisplatin-induced reactive oxygen species generation and ototoxicity. J Neurosci. 2010;30(11):3933–3946. doi:10.1523/JNEUROSCI.6054-09.201020237264
  • Banfi B, Malgrange B, Knisz J, Steger K, Dubois-Dauphin M, Krause KH. NOX3, a superoxide-generating NADPH oxidase of the inner ear. J Biol Chem. 2004;279(44):46065–46072. doi:10.1074/jbc.M40304620015326186
  • Kim HJ, Oh GS, Lee JH, et al. Cisplatin ototoxicity involves cytokines and STAT6 signaling network. Cell Res. 2011;21(6):944–956. doi:10.1038/cr.2011.2721321603
  • Kaur T, Mukherjea D, Sheehan K, Jajoo S, Rybak LP, Ramkumar V. Short interfering RNA against STAT1 attenuates cisplatin-induced ototoxicity in the rat by suppressing inflammation. Cell Death Dis. 2011;2:e180. doi:10.1038/cddis.2011.6321776018
  • Schmitt NC, Rubel EW, Nathanson NM. Cisplatin-induced hair cell death requires STAT1 and is attenuated by epigallocatechin gallate. J Neurosci. 2009;29(12):3843–3851. doi:10.1523/Jneurosci.5842-08.200919321781
  • Qi MH, Qiu Y, Zhou XY, et al. Regional up-regulation of NOX2 contributes to the differential vulnerability of outer hair cells to neomycin. Biochem Bioph Res Co. 2018;500(2):110–116. doi:10.1016/j.bbrc.2018.03.141
  • McNally JS, Saxena A, Cai H, Dikalov S, Harrison DG. Regulation of xanthine oxidoreductase protein expression by hydrogen peroxide and calcium. Arterioscl Throm Vas. 2005;25(8):1623–1628. doi:10.1161/01.ATV.0000170827.16296.6e
  • Lynch ED, Gu R, Pierce C, Kil J. Reduction of acute cisplatin ototoxicity and nephrotoxicity in rats by oral administration of allopurinol and ebselen. Hear Res. 2005;201(1–2):81–89. doi:10.1016/j.heares.2004.08.00215721563
  • Kinugasa Y, Ogino K, Furuse Y, et al. Allopurinol improves cardiac dysfunction after ischemia-reperfusion via reduction of oxidative stress in isolated perfused rat hearts. Circ J. 2003;67(9):781–787. doi:10.1253/circj.67.78112939555
  • Jones QR, Warford J, Rupasinghe HP, Robertson GS. Target-based selection of flavonoids for neurodegenerative disorders. Trends Pharmacol Sci. 2012;33(11):602–610. doi:10.1016/j.tips.2012.08.00222980637
  • Lee JS, Kang SU, Hwang HS, Pyun JH, Choung YH, Kim CH. Epicatechin protects the auditory organ by attenuating cisplatin-induced ototoxicity through inhibition of ERK. Toxicol Lett. 2010;199(3):308–316. doi:10.1016/j.toxlet.2010.09.01320883750
  • Tabuchi K, Nishimura B, Nakamagoe M, Hayashi K, Nakayama M, Hara A. Ototoxicity: mechanisms of cochlear impairment and its prevention. Curr Med Chem. 2011;18(31):4866–4871. doi:10.2174/09298671179753525421919841
  • Cai JY, Yang J, Jones DP. Mitochondrial control of apoptosis: the role of cytochrome c. Bba-Bioenergetics. 1998;1366(1–2):139–149. doi:10.1016/S0005-2728(98)00109-19714780
  • Lu YK, Cederbaum AI. Cisplatin-induced hepatotoxicity is enhanced by elevated expression of cytochrome P450 2E1. Toxicol Sci. 2006;89(2):515–523. doi:10.1093/toxsci/kfj03116251482
  • Liu H, Baliga M, Baliga R. Effect of cytochrome p450 2E1 inhibitors on cisplatin-induced cytotoxicity to renal proximal tubular epithelial cells. Anticancer Res. 2002;22(2a):863–868. PMID:1201466312014663
  • Liu H, Baliga R. Cytochrome P450 2E1 null mice provide novel protection against cisplatin-induced nephrotoxicity and apoptosis. Kidney Int. 2003;63(5):1687–1696. doi:10.1046/j.1523-1755.2003.00908.x12675844
  • Lu Y, Cederbaum A. The mode of cisplatin-induced cell death in CYP2E1-overexpressing HepG2 cells: modulation by ERK, ROS, glutathione, and thioredoxin. Free Radical Bio Med. 2007;43(7):1061–1075. doi:10.1016/j.freeradbiomed.2007.06.02117761302
  • Watanabe K, Hess A, Bloch W, Michel O. Expression of inducible nitric oxide synthase (iNOS/NOS II) in the vestibule of guinea pigs after the application of cisplatin. Anti-Cancer Drug. 2000;11(1):29–32. doi:10.1097/00001813-200001000-00005
  • Watanabe K, Inai S, Jinnouchi K, et al. Nuclear-factor kappa B (NF-kappa B)-inducible nitric oxide synthase (iNOS/NOS II) pathway damages the stria vascularis in cisplatin-treated mice. Anticancer Res. 2002;22(6C):4081–4085. PMID:1255303612553036
  • Ruan RS. Possible roles of nitric oxide in the physiology and pathophysiology of the mammalian cochlea. Ann N Y Acad Sci. 2002;962:260–274. doi:10.1111/j.1749-6632.2002.tb04073.x12076980
  • Rybak LP. Mechanisms of cisplatin ototoxicity and progress in otoprotection. Curr Opin Otolaryngol Head Neck Surg. 2007;15(5):364–369. doi:10.1097/MOO.0b013e3282eee45217823555
  • Rybak LP, Whitworth CA, Mukherjea D, Ramkumar V. Mechanisms of cisplatin-induced ototoxicity and prevention. Hear Res. 2007;226(1–2):157–167. doi:10.1016/j.heares.2006.09.01517113254
  • Bauer NC, Corbett AH, Doetsch PW. The current state of eukaryotic DNA base damage and repair. Nucleic Acids Res. 2015;43(21):10083–10101. doi:10.1093/nar/gkv113626519467
  • Friedberg EC. A history of the DNA repair and mutagenesis field The discovery of base excision repair. DNA Repair (Amst). 2016;37:A35–A39. doi:10.1016/j.dnarep.2015.12.00326861186
  • Miller JH, Goodman MF. Tomas Lindahl: 2015 Nobel Laureate. DNA Repair (Amst). 2016;37:A29–A34. doi:10.1016/j.dnarep.2015.12.00626861184
  • Radman M. Mismatch repair earns Nobel Prize in Chemistry 2015 to Paul Modrich for a biochemical tour de force. DNA Repair (Amst). 2016;37:A22–A28. doi:10.1016/j.dnarep.2015.12.00426861183
  • Van Houten B. A tale of two cities: a tribute to Aziz Sancar’s Nobel Prize in Chemistry for his molecular characterization of NER. DNA Repair (Amst). 2016;37:A3–A13. doi:10.1016/j.dnarep.2015.12.00226861185
  • Cadet J, Davies KJA. Oxidative DNA damage & repair: an introduction. Free Radic Biol Med. 2017;107:2–12. doi:10.1016/j.freeradbiomed.2017.03.03028363603
  • Santa-Gonzalez GA, Gomez-Molina A, Arcos-Burgos M, Meyer JN, Camargo M. Distinctive adaptive response to repeated exposure to hydrogen peroxide associated with upregulation of DNA repair genes and cell cycle arrest. Redox Biol. 2016;9:124–133. doi:10.1016/j.redox.2016.07.00427479053
  • Van Houten B, Santa-Gonzalez GA, Camargo M. DNA repair after oxidative stress: current challenges. Curr Opin Toxicol. 2018;7:9–16. doi:10.1016/j.cotox.2017.10.00929159324
  • Kim HJ, Oh GS, Shen A, et al. Augmentation of NAD(+) by NQO1 attenuates cisplatin-mediated hearing impairment. Cell Death Dis. 2014;5:e1292.24922076
  • Gentilin E, Simoni E, Candito M, Cazzador D, Astolfi L. Cisplatin-induced ototoxicity: updates on molecular targets. Trends Mol Med. 2019;S1471-4914(19):30210–33012. doi:10.1016/j.molmed.2019.08.002
  • Lorenz RT, Cysewski GR. Commercial potential for Haematococcus microalgae as a natural source of astaxanthin. Trends Biotechnol. 2000;18(4):160–167. doi:10.1016/S0167-7799(00)01433-510740262
  • Facchinetti F, Dawson VL, Dawson TM. Free radicals as mediators of neuronal injury. Cell Mol Neurobiol. 1998;18(6):667–682. doi:10.1023/A:10202219191549876873
  • Kim SH, Kim H. Inhibitory effect of astaxanthin on oxidative stress-induced mitochondrial dysfunction-a mini-review. Nutrients. 2018;10(9). doi:10.3390/nu10091137
  • Bae JW, Kim MJ, Jang CG, Lee SY. Protective effects of heme oxygenase-1 against MPP+-induced cytotoxicity in PC-12 cells. Neurol Sci. 2010;31(3):307–313. doi:10.1007/s10072-010-0216-620127499
  • Borlongan CV, Kanning K, Poulos SG, Freeman TB, Cahill DW, Sanberg PR. Free radical damage and oxidative stress in Huntington’s disease. J Fla Med Assoc. 1996;83(5):335–341. PMID: 86669728666972
  • Ferrante RJ, Browne SE, Shinobu LA, et al. Evidence of increased oxidative damage in both sporadic and familial amyotrophic lateral sclerosis. J Neurochem. 1997;69(5):2064–2074.9349552
  • Rebrin I, Zicker S, Wedekind KJ, Paetau-Robinson I, Packer L, Sohal RS. Effect of anti-oxidant-enriched diets on glutathione redox status in tissue homogenates and mitochondria of the senescence-accelerated mouse. Free Radical Bio Med. 2005;39(4):549–557. doi:10.1016/j.freeradbiomed.2005.04.00816043026
  • Chang CH, Chen CY, Chiou JY, Peng RY, Peng CH. Astaxanthine secured apoptotic death of PC12 cells induced by beta-amyloid peptide 25-35: its molecular action targets. J Med Food. 2010;13(3):548–556. doi:10.1089/jmf.2009.129120521980
  • Che HX, Li Q, Zhang TT, et al. Effects of astaxanthin and docosahexaenoic-acid-acylated astaxanthin on Alzheimer’s disease in APP/PS1 double-transgenic mice. J Agr Food Chem. 2018;66(19):4948–4957. doi:10.1021/acs.jafc.8b0098829695154
  • Lee DH, Kim CS, Lee YJ. Astaxanthin protects against MPTP/MPP plus -induced mitochondrial dysfunction and ROS production in vivo and in vitro. Food Chem Toxicol. 2011;49(1):271–280. doi:10.1016/j.fct.2010.10.02921056612
  • Kim JH, Choi W, Lee JH, et al. Astaxanthin inhibits H2O2-mediated apoptotic cell death in mouse neural progenitor cells via modulation of P38 and MEK signaling pathways. J Microbiol Biotechn. 2009;19(11):1355–1363. doi:10.4014/jmb.0906.06003
  • Al-Amin MM, Reza HM, Saadi HM, et al. Astaxanthin ameliorates aluminum chloride-induced spatial memory impairment and neuronal oxidative stress in mice. Eur J Pharmacol. 2016;777:60–69. doi:10.1016/j.ejphar.2016.02.06226927754
  • Tso Mark OM, Lam Tim-Tak. Method of retarding and ameliorating central nervous system and eye damage. Google Patents 1996;167.
  • Shen H, Kuo CC, Chou J, et al. Astaxanthin reduces ischemic brain injury in adult rats. FASEB J. 2009;23(6):1958–1968. doi:10.1096/fj.08-12328119218497
  • Wu W, Wang X, Xiang Q, et al. Astaxanthin alleviates brain aging in rats by attenuating oxidative stress and increasing BDNF levels. Food Funct. 2014;5(1):158–166. doi:10.1039/c3fo60400d24326685
  • Gross GJ, Hazen SL, Lockwood SF. Seven day oral supplementation with Cardax (disodium disuccinate astaxanthin) provides significant cardioprotection and reduces oxidative stress in rats. Mol Cell Biochem. 2006;283(1–2):23–30. doi:10.1007/s11010-006-2217-616444582
  • Pashkow FJ, Watumull DG, Campbell CL. Astaxanthin: a novel potential treatment for oxidative stress and inflammation in cardiovascular disease. Am J Cardiol. 2008;101(10A):58D–68D. doi:10.1016/j.amjcard.2008.02.01018157966
  • 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(7):2721–2725. PMID:2068300420683004
  • Abdelzaher LA, Imaizumi T, Suzuki T, Tomita K, Takashina M, Hattori Y. Astaxanthin alleviates oxidative stress insults-related derangements in human vascular endothelial cells exposed to glucose fluctuations. Life Sci. 2016;150:24–31. doi:10.1016/j.lfs.2016.02.08726924495
  • Alam MN, Hossain MM, Rahman MM, et al. Astaxanthin prevented oxidative stress in heart and kidneys of isoproterenol-administered aged rats. J Diet Suppl. 2018;15(1):42–54. doi:10.1080/19390211.2017.132107828489954
  • Pongkan W, Takatori O, Ni YH, et al. beta-Cryptoxanthin exerts greater cardioprotective effects on cardiac ischemia-reperfusion injury than astaxanthin by attenuating mitochondrial dysfunction in mice. Mol Nutr Food Res. 2017;61(10). doi:10.1002/mnfr.201601077
  • Fan CD, Sun JY, Fu XT, et al. Astaxanthin attenuates homocysteine-induced cardiotoxicity in vitro and in vivo by inhibiting mitochondrial dysfunction and oxidative damage. Front Physiol. 2017;8. doi:10.3389/fphys.2017.01041
  • Gammone MA, Riccioni G, D’Orazio N. Carotenoids: potential allies of cardiovascular health? Food Nutr Res. 2015;59:26762. doi:10.3402/fnr.v59.2676225660385
  • Ni Y, Nagashimada M, Zhuge F, et al. Astaxanthin prevents and reverses diet-induced insulin resistance and steatohepatitis in mice: a comparison with vitamin E. Sci Rep. 2015;5:17192. doi:10.1038/srep1719226603489
  • Zhang J, Zhang S, Bi J, Gu J, Deng Y, Liu C. Astaxanthin pretreatment attenuates acetaminophen-induced liver injury in mice. Int Immunopharmacol. 2017;45:26–33. doi:10.1016/j.intimp.2017.01.02828152447
  • Takahashi K, Watanabe M, Takimoto T, Akiba Y. Uptake and distribution of astaxanthin in several tissues and plasma lipoproteins in male broiler chickens fed a yeast (Phaffia rhodozyma) with a high concentration of astaxanthin. Brit Poultry Sci. 2004;45(1):133–138. doi:10.1080/0007166041000166895015115211
  • 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 Agr Food Chem. 2013;61(16):3842–3851. doi:10.1021/jf304609j23473626
  • 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. doi:10.1186/1743-7075-7-18
  • Tominaga K, Hongo N, Fujishita M, Takahashi Y, Adachi Y. Protective effects of astaxanthin on skin deterioration. J Clin Biochem Nutr. 2017;61(1):33–39. doi:10.3164/jcbn.17-3528751807
  • Yoon HS, Cho HH, Cho S, Lee SR, Shin MH, Chung JH. Supplementating with dietary astaxanthin combined with collagen hydrolysate improves facial elasticity and decreases matrix metalloproteinase-1 and −12 expression: a comparative study with placebo. J Med Food. 2014;17(7):810–816. doi:10.1089/jmf.2013.306024955642
  • Tominaga K, Hongo N, Karato M, Yamashita E. Cosmetic benefits of astaxanthin on humans subjects. Acta Biochim Pol. 2012;59(1):43–47. doi:10.18388/abp.2012_216822428137
  • Fang Q, Guo S, Zhou H, Han R, Wu P, Han 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. doi:10.1038/srep4144028128352
  • Meephansan J, Rungjang A, Yingmema W, Deenonpoe R, Ponnikorn S. Effect of astaxanthin on cutaneous wound healing. Clin Cosmet Investig Dermatol. 2017;10:259–265. doi:10.2147/CCID.S142795
  • Zhang L, Wang H. Multiple mechanisms of anti-cancer effects exerted by astaxanthin. Mar Drugs. 2015;13(7):4310–4330. doi:10.3390/md1307431026184238
  • Cairns RA, Harris IS, Mak TW. Regulation of cancer cell metabolism. Nat Rev Cancer. 2011;11(2):85–95. doi:10.1038/nrc298121258394
  • Kavitha K, Kowshik J, Kishore TK, Baba AB, Nagini S. Astaxanthin inhibits NF-kappaB and Wnt/beta-catenin signaling pathways via inactivation of Erk/MAPK and PI3K/Akt to induce intrinsic apoptosis in a hamster model of oral cancer. Biochim Biophys Acta. 2013;1830(10):4433–4444. doi:10.1016/j.bbagen.2013.05.03223726989
  • Nagendraprabhu P, Sudhandiran G. Astaxanthin inhibits tumor invasion by decreasing extracellular matrix production and induces apoptosis in experimental rat colon carcinogenesis by modulating the expressions of ERK-2, NFkB and COX-2. Invest New Drugs. 2011;29(2):207–224. doi:10.1007/s10637-009-9342-519876598
  • Song XD, Zhang JJ, Wang MR, Liu WB, Gu XB, Lv CJ. Astaxanthin induces mitochondria-mediated apoptosis in rat hepatocellular carcinoma CBRH-7919 cells. Biol Pharm Bull. 2011;34(6):839–844. doi:10.1248/bpb.34.83921628881
  • Zhang X, Zhao WE, Hu L, Zhao L, Huang J. Carotenoids inhibit proliferation and regulate expression of peroxisome proliferators-activated receptor gamma (PPARgamma) in K562 cancer cells. Arch Biochem Biophys. 2011;512(1):96–106. doi:10.1016/j.abb.2011.05.00421620794
  • Su XZ, Chen R, Wang CB, Ouyang XL, Jiang Y, Zhu MY. Astaxanthin combine with human serum albumin to abrogate cell proliferation, migration, and drug-resistant in human ovarian carcinoma SKOV3 cells. Anticancer Agents Med Chem. 2019;19(6):792–801. doi:10.2174/187152061966619022512300330799797
  • Goto S, Kogure K, Abe K, et al. Efficient radical trapping at the surface and inside the phospholipid membrane is responsible for highly potent antiperoxidative activity of the carotenoid astaxanthin. Bba-Biomembranes. 2001;1512(2):251–258. doi:10.1016/S0005-2736(01)00326-111406102
  • Santa-Maria AR, Walter FR, Valkai S, et al. Lidocaine turns the surface charge of biological membranes more positive and changes the permeability of blood-brain barrier culture models. Bba-Biomembranes. 2019;1861(9):1579–1591. doi:10.1016/j.bbamem.2019.07.00831301276
  • Matsushita Y, Suzuki R, Nara E, Yokoyama A, Miyashita K. Anti-oxidant activity of polar carotenoids including astaxanthin-β-glucoside from marine bacterium on PC liposomes. Fish Sci. 2000;66(5):980–985. doi:10.1046/j.1444-2906.2000.00155.x
  • Greene LS. Asthma and oxidant stress - nutritional, environmental, and genetic risk-factors. J Am Coll Nutr. 1995;14(4):317–324. doi:10.1080/07315724.1995.107185168568107
  • Dekkers JC, van Doornen LJP, Kemper HCG. The role of anti-oxidant vitamins and enzymes in the prevention of exercise-induced muscle damage. Sports Med. 1996;21(3):213–238. doi:10.2165/00007256-199621030-000058776010
  • Bennedsen M, Wang X, Willen R, Wadstrom T, Andersen LP. Treatment of H-pylori infected mice with anti-oxidant astaxanthin reduces gastric inflammation, bacterial load and modulates cytokine release by splenocytes. Immunol Lett. 1999;70(3):185–189. doi:10.1016/S0165-2478(99)00145-510656672
  • Higuera-Ciapara I, Felix-Valenzuela L, Goycoolea FM. Astaxanthin: a review of its chemistry and applications. Crit Rev Food Sci. 2006;46(2):185–196. doi:10.1080/10408690590957188
  • Wolf AM, Asoh S, Hiranuma H, et al. Astaxanthin protects mitochondrial redox state and functional integrity against oxidative stress. J Nutr Biochem. 2010;21(5):381–389. doi:10.1016/j.jnutbio.2009.01.01119423317
  • Kidd P. Astaxanthin, cell membrane nutrient with diverse clinical benefits and anti-aging potential. Altern Med Rev. 2011;16(4):355–364.22214255
  • Osterlie M, Bjerkeng B, Liaaen-Jensen S. Plasma appearance and distribution of astaxanthin E/Z and R/S isomers in plasma lipoproteins of men after single dose administration of astaxanthin. J Nutr Biochem. 2000;11(10):482–490. doi:10.1016/S0955-2863(00)00104-2 PMID:2221425511120445
  • Yan TT, Zhao Y, Zhang X, Lin XT. Astaxanthin inhibits acetaldehyde-induced cytotoxicity in sh-sy5y cells by modulating Akt/CREB and p38MAPK/ERK signaling pathways. Mar Drugs. 2016;14(3):56. doi:10.3390/md14030056
  • Li JJ, Wang F, Xia YJ, et al. Astaxanthin pretreatment attenuates hepatic ischemia reperfusion-induced apoptosis and autophagy via the ROS/MAPK pathway in mice. Mar Drugs. 2015;13(6):3368–3387. doi:10.3390/md1306336826023842
  • Yang X, Guo AL, Pang YP, et al. Astaxanthin attenuates environmental tobacco smoke-induced cognitive deficits: a critical role of p38 MAPK. Mar Drugs. 2019;17(1):24. doi:10.3390/md17010024
  • Niu TT, Xuan RR, Jiang LG, et al. Astaxanthin induces the Nrf2/HO-1 anti-oxidant pathway in human umbilical vein endothelial cells by generating trace amounts of ROS. J Agr Food Chem. 2018;66(6):1551–1559. doi:10.1021/acs.jafc.7b0549329381356
  • Wu Q, Zhang XS, Wang HD, et al. Astaxanthin activates nuclear factor erythroid-related factor 2 and the anti-oxidant responsive element (Nrf2-ARE) pathway in the brain after subarachnoid hemorrhage in rats and attenuates early brain injury. Mar Drugs. 2014;12(12):6125–6141. doi:10.3390/md1212612525528957
  • Wu ML, Ho YC, Yet SF. A central role of heme oxygenase-1 in cardiovascular protection. Antioxid Redox Sign. 2011;15(7):1835–1846. doi:10.1089/ars.2010.3726
  • Dinkova-Kostova AT, Talalay P. NAD(P)H:quinoneacceptor oxidoreductase 1 (NQO1), a multifunctional anti-oxidant enzyme and exceptionally versatile cytoprotector. Arch Biochem Biophys. 2010;501(1):116–123. doi:10.1016/j.abb.2010.03.01920361926
  • Pan L, Zhou Y, Li XF, Wan QJ, Yu LH. Preventive treatment of astaxanthin provides neuroprotection through suppression of reactive oxygen species and activation of anti-oxidant defense pathway after stroke in rats. Brain Res Bull. 2017;130:211–220. doi:10.1016/j.brainresbull.2017.01.02428161193
  • Tripathi DN, Jena GB. Astaxanthin intervention ameliorates cyclophosphamide-induced oxidative stress, DNA damage and early hepatocarcinogenesis in rat: role of Nrf2, p53, p38 and phase-II enzymes. Mutat Res-Gen Tox En. 2010;696(1):69–80. doi:10.1016/j.mrgentox.2009.12.014
  • Nakanishi A, Wada Y, Kitagishi Y, Matsuda S. Link between PI3K/AKT/PTEN pathway and NOX protein in diseases. Aging Dis. 2014;5(3):203–211. doi:10.14336/Ad.2014.050020324900943
  • Wang CM, Cai XL, Wen QP. Astaxanthin reduces isoflurane-induced neuroapoptosis via the PI3K/Akt pathway. Mol Med Rep. 2016;13(5):4073–4078. doi:10.3892/mmr.2016.503527035665
  • Zuluaga M, Barzegari A, Letourneur D, Gueguen V, Pavon-Djavid G. Oxidative stress regulation on endothelial cells by hydrophilic astaxanthin complex: chemical, biological, and molecular anti-oxidant activity evaluation. Oxid Med Cell Longev. 2017;2017:1–15. doi:10.1155/2017/8073798
  • Li ZR, Dong X, Liu HL, et al. Astaxanthin protects ARPE-19 cells from oxidative stress via upregulation of Nrf2-regulated Phase II enzymes through activation of PI3K/Akt. Mol Vis. 2013;19:1656–1666.23901249
  • Ye QY, Zhang XD, Huang BX, Zhu YG, Chen XC. Astaxanthin suppresses MPP+-induced oxidative damage in PC12 cells through a Sp1/NR1 signaling pathway. Mar Drugs. 2013;11(4):1019–1034. doi:10.3390/md1104101923538867
  • Takemoto Y, Hirose Y, Sugahara K, Hashimoto M, Hara H, Yamashita H. Protective effect of an astaxanthin nanoemulsion against neomycin-induced hair-cell damage in zebrafish. Auris Nasus Larynx. 2018;45(1):20–25. doi:10.1016/j.anl.2017.02.00128274503