Open access peer-reviewed chapter

Role of Carotenoids in Parkinson’s Diseases

Written By

Fengjuan Jiao

Submitted: 19 June 2023 Reviewed: 22 June 2023 Published: 18 October 2023

DOI: 10.5772/intechopen.112311

From the Edited Volume

Dietary Carotenoids - Sources, Properties, and Role in Human Health

Edited by Akkinapally Venketeshwer Rao and Leticia Rao

Chapter metrics overview

54 Chapter Downloads

View Full Metrics

Abstract

Parkinson’s disease (PD) is the second most common neurodegenerative disease, which is characterized by a progressive loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) of the middle brain. Excessive reactive oxygen species (ROS) accumulation due to mitochondrial dysfunction or inflammation is the main factor contributing to the degeneration of dopaminergic neurons. In the preclinical and clinical studies, carotenoids and their major components including vitamin and astaxanthin were found to have antioxidant, anti-inflammatory, autophagy-promoting, and mitochondrial dysfunction improving functions. This chapter focuses on the current status of research on carotenoids and their major components in PD, which can provide help for the prevention and treatment of PD.

Keywords

  • carotenoids
  • Parkinson’s disease
  • vitamins
  • astaxanthin
  • antioxidant
  • anti-inflammatory

1. Introduction

Parkinson’s disease (PD) is the second most common neurodegenerative disease characterized primarily by movement disorders, which affects about 1% of the world’s people over the age of 60. It is predicted that more than 14 million people will be affected by PD by 2040 [1]. Clinically, PD is mainly characterized by motor symptoms including resting tremor, bradykinesia, muscle rigidity, and gait abnormalities, and some nonmotor neurological symptoms including olfactory disorders, constipation, depression, and cognitive decline. The major pathological hallmarks of PD are the selective loss of the dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the appearance of Lewis bodies (LBs) in the cytoplasm of neurons. The LBs are eosinophilic inclusions composed of α-synuclein, ubiquitin, synphilin-1, neurofilament proteins, Parkin and other proteins that can lead to degeneration of dopaminergic neurons, with a consequent decrease in dopamine levels and increases of oxidative stress [2, 3].

Currently, drugs are also the main treatment strategies for PD, which can improve some symptoms of PD patients and delay the progression of PD through dopaminergic stimulation or cholinergic and glutamatergic inhibition. However, the improvement of symptoms may disappear and/or motor abnormalities may appear over time, with the disease progression or the treatment discontinuation. In recent years, increased attention is given to the patient’s diet, and the use of food supplements and functional food rich in antioxidants plays a very important role in the treatment and prevention of PD. Recently, carotenoids have received increased attention for their anti-oxidative and anti-apoptotic activities in PD. In this chapter, we discuss the pharmacological profiles of carotenoids and the protective roles of carotenoids in PD.

Advertisement

2. Overview of carotenoids

Carotenoids are one of the most widespread and prevalent lipid-soluble pigments in nature, which are synthesized by various plants, microalgae, bacteria, and fungi. However, animals cannot synthesize carotenoids and they must obtain them through diet to meet their nutritional requirements. Carotenoids are especially abundant in vegetables, yellow-orange fruits, dark green leafy vegetables, and internal fat and egg yolks of terrestrial animals.

To date, more than 750 different chemical structures of carotenoids have been determined [4]. Carotenoids usually contain a hydrocarbon backbone of 40 carbon atoms, an acyclic C40H56 structure consisting of eight isoprene units. The C40H56 structure has a long central light-absorbing conjugated polyene compound constituting the chromophoric system that absorbs light from the visible region. The central structure can be modified by desaturation, cyclization of one or both ends, and addition of oxygen-containing functional groups such as hydroxy, epoxy, and/or oxo groups [5]. All of these modifications contribute to the enormous diversity of carotenoids in nature. According to the modifications of oxygen-containing functional groups, carotenoids are classified as carotenes and xanthophylls, with only the latter containing the functional groups [6]. Polar xanthophylls, such as astaxanthin, β-cryptoxanthin, lutein, and zeaxanthin, are usually located in the lipid bilayer membranes, whereas nonpolar carotenoids, such as α-carotene, β-carotene, and lycopene, are usually located in the inner part of a cell membrane [7, 8].

Advertisement

3. Biological functions of carotenoids

In higher plants, algae, fungi, and bacteria, carotenoids (mainly xanthophylls) are located in specific pigment-protein complexes and act as accessory pigments to harvest light for photosynthesis [9]. Moreover, carotenoids also act as photoprotectors by quenching energy excesses under high light stress and preventing the formation of highly reactive, destructive, and potentially lethal singlet oxygen species. Additionally, carotenoids in plants also play a vital ecological role by contributing to the color of fruit and flower, which is an important signal to attract insects and animals for pollination and seed dispersal [10]. Besides these functions in plant and microorganisms, carotenoids are key compounds for human health. Vitamin A deficiency is still a cause of blindness in children and may be associated with reduced immune function, gastrointestinal disease, and measles [11]. α-, β-, and γ-carotene, as well as β-cryptoxanthin, were found to have provitamin A activity and are precursors for the synthesis of retinoids, retinol (vitamin A), retinal, and retinoic acid, with β-carotene being the main precursor of vitamin A [12, 13]. In addition, zeaxanthin and lutein are the pigment components of the macula that can prevent age-related macular degeneration [14]. Additionally, the Age-related Eye Disease Study (AREDS) showed that a formulation containing β-carotene, zinc, vitamin C, and vitamin E had grateful benefits in the prevention of age-related macular degeneration (AMD) [15]. Etminan et al. found that serum lycopene levels are inversely correlated with the incidence of prostate cancer [16]. Recently, growing evidence has shown that carotenoids can reduce oxidative damage by neutralizing ROS, exerting anti-inflammatory and anti-apoptotic effects in vitro and in vivo in many diseases [17].

3.1 Antioxidant activity

Several forms of reactive oxygen species (ROS), such as superoxide (O2 ~ -), hydrogen peroxide (H2O2), and hydroxyl radicals (HO.), are produced during various metabolic processes in cells or under exogenous stress. Besides, the ROS generation can also trigger mitochondrial dysfunction and protein misfolding in the endoplasmic reticulum (ER), which further augments ROS production [18]. Together, these free radicals can cause the cleavage of DNA, peroxidize lipids, alter enzyme activity, and kill cells [19]. The antioxidant effect of carotenoids has been well demonstrated in a number of in vitro studies, including lipids in homogeneous solution, liposomes, isolated membranes, nuclear, and intact cells. This antioxidant activity is related to the chemical structures of the carotenoids [20]. For example, β-carotene can reduce the nuclear damage induced by xanthine oxidase/hypoxanthine, as well as inhibit the lipid peroxidation induced either by enzymatic sources or by nonenzymatic sources of oxy radicals [21, 22]. In addition, carotenoids can protect cells from certain oxidative stresses by activating endogenous antioxidant enzyme activities and reducing DNA damage. In a mouse model of streptozotocin-induced Alzheimer’s disease (AD), β-carotene at a dose of 2.05 mg/kg was found to attenuate streptozotocin-induced cognitive deficit via increase of glutathione peroxidase (GSH), superoxide dismutase (SOD), and catalase (CAT) activities and inhibition of acetylcholinesterase (AChE) [23]. Crocetin, a carotenoid isolated from the Chinese herbal medicine Crocus sativus L. (saffron), exerts cardioprotective effects by increasing SOD and GSH activities in norepinephrine-induced cardiac hypertrophy in rat models [24]. Crocin, another carotenoid component of saffron, has also been shown to prevent rat pheochromocytoma (PC12) cell death induced by serum/glucose deprivation via enhancement of SOD activity [25]. At low concentrations, β-cryptoxanthin (1 and 4 μM) can protect HeLa and Caco-2 cells from damage induced by H2O2 or by visible light in the presence of a photosensitizer through enhancement of DNA repair [26]. However, a number of in vitro and in vivo studies have found that carotenoids may lose their effectiveness as antioxidants at high concentrations or high partial pressures of oxygen [27]. Lycopene and β-carotene are protective against xanthine/xanthine oxidase-induced DNA damage only at relatively low concentrations (1–3 μM), and these levels are comparable with those in the plasma of individuals consuming a carotenoid-rich diet. However, at higher concentrations (4–10 μM), lycopene and β-carotene contribute to increased DNA damage [28]. However, another in vitro study showed that both lycopene and β-carotene at concentrations ranging from 0.25 to 10 μM significantly inhibit strand breakage induced by 4-hydroxyestradiol (4-OHE2)/copper sulfate (catechol estrogens) by up to approximately 90% in plasmid DNA [29]. The antioxidant activity of aspartate aminotransferase (AST) is associated with the activation of mitogen-activated protein kinase (MAPK), nuclear factor erythroid 2-related factor 2 (NRF2)/antioxidant response element (ARE), and phosphatidylinositol 3-kinase (PI3K/AKT) pathways [30, 31, 32]. The Keap1-Nrf2 [Kelch-like ECH-associated protein 1-nuclear factor-like 2] pathway plays an important role in protecting cells against oxidative and xenobiotic stresses. Furthermore, NRF2 is a transcription factor that regulates the expression of a wide array of cytoprotective antioxidant genes that have been implicated in several diseases [33]. Recently, Nan et al. discovered that AST firmly binds to the active pocket of KEAP1 to form the KEAP1-NRF2-AST complex via computer molecular simulation. This result suggested that AST may competitively inhibit the binding of NRF2 to the Kelch domain of KEAP, which could be the possible reason for the astaxanthin-mediated mechanism of antioxidant action of NRF2 [34]. The antioxidant properties of astaxanthin (ASTX) are related to the two oxygenated groups on each of its two rings [35]. It has been shown that ASTX not only quenches ROS, but also promotes the nuclear translocation of NRF2, thereby increasing the expression of endogenous antioxidants [36, 37].

3.2 Anti-inflammatory activity

Inflammation is a normal and crucial response to tissue injury caused by foreign organisms such as pathogens, dust particles, and viruses, which is initiated by the synthesis and secretion of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-6, IL-12, and interferon γ (IFN-γ) in macrophages [38]. The binding of pro-inflammatory cytokines to the receptors triggers the MAPK signaling pathway, which ultimately results in the activation of nuclear factor-kappa B (NF-κB) and the c-Jun [39]. Subsequently, these transcription factors activate the expression of genes including cytokines (TNF-α and IL-1β), chemokines, adhesion molecules, and inducible effector enzymes such as inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2), thereby generating a feedforward loop of the inflammatory response [40]. Growing evidence has highlighted the importance of inflammation in the development of chronic diseases such as neurodegeneration, cancer, diabetes, cardiovascular disease, and inflammatory bowel disease [41]. In the last decade, some carotenoids have been shown to have anti-inflammatory effects in vivo andin vitro. The ability of lutein to inhibit inflammation has been demonstrated in mice and cellular models of retinopathy and in a clinical trial studying retinal health [42, 43, 44]. Additionally, it has been shown that lutein reduces pro-inflammatory cytokine release by suppressing the activation of the NF-κB pathway and Nrf2 in the presence of many oxidative stressors [45, 46]. Crocin and crocetin were shown to effectively reduce lipopolysaccharide (LPS)-induced nitric oxide (NO) release, production of TNF-α, IL-1β, and intracellular ROS, and NF-κB activation in cultured rat brain microglial cells [47]. Astaxanthin, a member of the xanthophyll family, has an anti-inflammatory effect similar to other carotenoids. Astaxanthin (AST) can improve the sensory and motor function of spinal cord injury (SCI) rats by reducing the expression of N-methyl-D-aspartate receptor subunit 2B (NR2B) and phospho-p38 (p-p38) MAPK as inflammatory signaling mediators and tumor necrosis factor-α (TNF-α) as an inflammatory cytokine [48]. In another rat model, AST could decrease the tissue damage and mechanical pain after SCI through its anti-inflammatory and antioxidant effects [49]. Furthermore, AST also exhibits its anti-inflammatory actions by inhibiting cyclooxygenase 1 (COX-1) and NO in LPS-stimulated BV2 microglial cells [50]. In streptozotocin-induced diabetic animal models, it has also been found that astaxanthin reduced hippocampal and retinal inflammation by reducing activity of NF-κB and/or decreased the expression of pro-inflammatory factors, ameliorating cognitive impairment, retinal oxidative stress, and depression [51, 52, 53]. Crocin and crocetin effectively inhibited LPS-induced nitric oxide (NO) release, TNF-α, IL-1β, and intracellular ROS production, and NF-κB activation in cultured rat brain microglia. These results suggest that crocin and crocetin may provide neuroprotection by reducing the production of various pro-inflammatory factors and ROS from activated microglia [47]. Fucoxanthin (Fx), another member of the marine xanthophylls, exerts anti-inflammatory effects against LPS stimulation. In LPS-activated BV-2 microglia, fucoxanthin dose-dependently inhibited the secretion of pro-inflammatory mediators including IL-6, TNF-α, ROS, prostaglandin (PG) E2, and NO productions, and also blocked Akt/NF-κB and MAPKs/AP-1 pathways [54]. Lycopene, one of the more abundant carotenoids in tomatoes, has been demonstrated to ameliorate Aβ-induced learning and memory deficits, LPS-induced depression-like behaviors, and D-galactose-induced cognitive impairments by downregulating the expression of inflammatory cytokines and mediating Nrf2/NF-κB transcriptional pathway in animal models [55, 56, 57].

3.3 Anti-apoptotic activity

Apoptosis, also known as programmed death, usually occurs during development and aging as an important mechanism for maintaining cell populations in tissues. In addition, apoptosis also occurs as a defense mechanism when the individuals are in immune reactions or when cells are damaged by noxious agents. Excessive apoptosis is associated with many human diseases including neurodegenerative diseases, cancer, ischemic injury, and autoimmune diseases [58, 59]. Many key apoptotic proteins are involved in two major apoptotic pathways, which are the intrinsic mitochondrial pathway and the extrinsic death receptor pathway [59]. An increasing number of researchers have highlighted either anti-apoptotic or pro-apoptotic effects of some carotenoids depending on the pathological condition. It has been found that lycopene exhibits mainly anti-apoptotic effects in neurological diseases. For example, lycopene can inhibit apoptosis via downregulation of pro-apoptotic proteins including Bcl-2-associated X protein (Bax) and upregulation of anti-apoptotic proteins including B-cell lymphoma 2 (Bcl-2) and B-cell lymphoma-extra large (Bcl-xL), thereby protecting against neuronal damage [60, 61, 62]. Additionally, lycopene inhibits mitochondrial damage-induced neuronal apoptosis through blocking the opening of mitochondrial permeability transition pore (mtPTP), thereby reducing mitochondrial depolarization and the release of cytochrome c [63, 64, 65]. However, in most tumor studies, lycopene may exhibit pro-apoptotic effects. It has been shown that lycopene promotes apoptosis in oral cancer via PI3K/AKT/m-TOR (mammalian target of rapamycin) signaling pathway [66]. Furthermore, lycopene and β-carotene inhibit cell proliferation, arrest the cell cycle in different phases, and increase apoptosis in human breast cancer cell lines [67]. Lycopene increases intrinsic apoptosis via increase of active caspase-3 and the Bax-to-Bcl-2 ratio in pancreatic cancer PANC-1 cells [68]. Similarly, the pro-apoptotic activity of fucoxanthin is one of the main factors responsible for its anticancer properties. In various tumor cells and animal models, fucoxanthin can induce G1 cell-cycle arrest and attribute to the molecular regulation facilitating apoptosis [69]. AST has been reported to enhance BAD phosphorylation through activation of the PI3K/AKT survival pathway and further downregulated the expression of cytochrome c and caspases-3/9 [70]. However, it has also been reported that AST can induce the intrinsic apoptotic pathway in a hamster model of oral cancer via inactivation of extracellular signal-regulated kinase (Erk)/MAPK and PI3K/Akt cascades, which ultimately inhibit the NF-κB and Wnt/β-catenin signaling pathways [71]. Recent studies have shown that the anti-apoptotic and pro-apoptotic activities of other carotenoids, such as crocin, lutein, and crocetin, play critical roles in normal cell survival and tumor cell clearance [72, 73, 74, 75, 76, 77, 78].

3.4 Regulation of autophagy

Autophagy is a catabolic process necessary for intracellular clearance of damaged organelles, protein complexes, and the recycling of nutritional building blocks. Dysfunction of autophagy has been shown to be associated with the pathogenesis of various diseases such as aging, cancer, and neurodegenerative diseases [79]. It has been shown that some carotenoids are able to regulate autophagy in cellular and animal models. Astaxanthin has been found to downregulate the level of autophagy in carbon tetrachloride (CCL4)-induced liver fibrosis mice and hepatic ischemia-reperfusion (IR) injury mice [80, 81]. However, a recent study shows that astaxanthin prevents acetaminophen (APAP)-induced liver injury (AILI) via improvement of oxidative stress and enhancement of autophagy [82]. It has been recently demonstrated that lutein attenuated autophagosome formation by regulating mTOR-mediated pathway, which in turn decreases cobalt chloride-induced autophagy in rat Müller cells, while it induced autophagy via upregulation of autophagy-related genes including autophagy-related 4A cysteine peptidase (ATG4A), autophagy-related 5 (ATG5), autophagy-related 7 (ATG7), autophagy-related 12 (ATG12), and beclin-1 (BENC1) in rat intestinal epithelial (IEC-6) cells [83, 84]. In an animal model of myocardial ischemia/reperfusion injury, crocin administration during ischemia significantly promoted autophagy and was accompanied by the activation of AMPK. In contrast, crocin inhibited autophagy during reperfusion and was accompanied by activation of Akt [85]. In addition, in a model of murine traumatic brain injury, fucoxanthin protects neuronal cells from death through the activation of the Nrf2-autophagy pathway [86]. The regulation of autophagy by carotenoids is still controversial, and the precise molecular mechanism of this regulation is still unclear.

Advertisement

4. Roles of carotenoids in Parkinson’s diseases

Inflammation and oxidative stress are two important factors that contribute to neuronal degeneration in PD [87]. Dietary antioxidants of carotenoids have been suggested as neuroprotective agents for PD based on their property of reducing oxidative damage [88]. In a prospective cohort study that included 38,937 women and 45,837 men, the dietary intake of β-carotene was associated with a lower risk of PD in a 14.9-year follow-up [89]. Another clinical study showed that total carotenoids, β-carotene, and lutein-zeaxanthin, are shown to slow down the rate of parkinsonian sign progression in older adults [90]. Epidemiological studies have shown that serum α-carotene, β-carotene, and lycopene levels are significantly decreased in PD patients, and decreased serum carotenoid levels are also associated with motor dysfunction, suggesting that carotenoids may have a neuroprotective effect in PD [89, 91].

4.1 Astaxanthin

Astaxanthin is an important natural carotenoid that is widely distributed in marine organisms, such as microalgae, krill, shrimp, crab, and salmon [92]. Currently, accumulating evidence has revealed that astaxanthin plays a protective role in PD through its antioxidant, anti-apoptotic, and anti-inflammatory activities. In docosahexenoic acid hydroperoxide (DHA-OOH) or 6-hydroxydopamine (6-OHDA)-treated dopaminergic SH-SY5Y cells, astaxanthin treatment significantly attenuates intracellular ROS production and mitochondrial dysfunction. In addition, astaxanthin also inhibits oxidative stress-induced neuronal cell damage mainly via attenuating activation of p38 MAPK and expression of cleaved caspase-9 and caspase-3 [93, 94, 95]. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is a neurotoxin that targets specifically dopaminergic neurons and has been widely used to develop animal models of PD. MPTP-induced Parkinsonism exhibits key pathological features, such as oxidative stress, mitochondrial dysfunction, and loss of dopaminergic neurons, in the substantia nigra [96]. In MPP+ (1-methyl-4-phenylpyridinium)-induced PC12 cells, astaxanthin pretreatment inhibited the increase of ROS and NADPH oxidase 2 (NOX2) and increased the expression of NRF2 and heme oxygenase 1 (HO-1), suggesting that astaxanthin treatment could inhibit MPP + -induced oxidative stress through the activation of HO-1/NOX2 axis [97]. Subsequently, it was found that astaxanthin could inhibit MPP + -induced oxidative damage by inhibition of the Sp1 (activated transcription factor 1)/NR1 (N-methyl-D-aspartate (NMDA) receptor subunit 1) pathway [98]. Astaxanthin attenuates MPTP/MPP + -induced oxidative damage by inhibiting ROS production and preventing mitochondrial Δψm collapse, which in turn protects against MPTP/MPP + -induced neuronal damage [99]. Both in young and aged mice, astaxanthin (30 mg/kg b.w.) can protect against MPTP-induced damage to nigrostriatal dopaminergic neurons. However, astaxanthin was less efficacious in protection against the MPTP-induced loss of tyrosine hydroxylase (TH) in neurons in the aged mice compared to the young mice, which suggested that aging may be a critical factor to consider during the development of novel therapeutics for neurodegenerative disorders like PD [100]. A recent study has reported that AST inhibits endoplasmic reticulum (ER) stress and protects against MPP+/MPTP-induced neuronal apoptosis by targeting the microRNA-7 (miR-7)/α-synuclein axis [101]. Additionally, AST reduces the abnormal mitochondrial membrane depolarization induced by excitatory glutamate injury in primary cortical neurons and prevents neuronal death by regulating ionotropic glutamate subtype receptors such as NMDA, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), kainic acid (KA), cytosolic secondary calcium rise, and mitochondrial calcium buffering [102]. AST is distributed in nature not only as nonesterified AST but also as fatty acid acylated AST esters [103]. It was shown that nonesterified astaxanthin significantly attenuated neuronal damage in mice with PD by enhancing its antioxidant and anti-inflammatory capacities [99, 100, 104]. Administration of AST can inhibit apoptosis of dopaminergic neurons in the brain of MPTP-induced mice via mitochondria-mediated pathway and JNK and P38 MAPK pathways, and in particular, DHA-AST had a better performance compared with nonesterified AST [105].

Inflammation is one of the pathogenic mechanisms of pathogenesis in PD. Peroxynitrite produced by activated microglia was found to be cytotoxic to dopaminergic neurons, while astaxanthin can protect the mitochondrial respiratory chain from peroxynitrite toxicity [106]. Overall, these results suggested that astaxanthin might act as a potent therapeutic to prevent oxidative stress, apoptosis, and inflammation, subsequently preventing the loss of dopaminergic neurons in the SNpc.

4.2 Lycopene

Lycopene, an aliphatic hydrocarbon carotenoid found mainly in the ripened tomatoes, has strong antioxidant property [107]. Because lycopene can cross the blood-brain barrier (BBB), it is considered a potentially useful agent in the management of neurodegenerative diseases [108]. In an in vivo model, the administration of lycopene protected against MPTP-induced depletion of striatal dopamine (DA) and its metabolites in a dose-dependent manner. In addition, lycopene also reversed MPTP-induced oxidative stress, motor defects, and apoptosis in PD mice [109]. Lycopene reduced ROS production by improving the abnormal opening of the mitochondrial permeability transition pore and the impaired mitochondrial membrane potential, which in turn ameliorated MPP + -induced apoptosis in SH-SY5Y cells [110]. Moreover, it was found that lycopene not only exerts its antioxidant activity by reducing the malondialdehyde (MDA) content in the substantia nigra and striatum and increasing the activity of SOD, glutathione peroxidase (GSH-Px), and CAT enzymes, but also reduces the autophagy activity in dopaminergic neurons and has a protective effect on rotenone-induced neuronal death [111, 112].

4.3 β-Carotene

β-Carotene is a carotenoid found in yellow-orange fruits and vegetables. A higher intake of β-carotene-containing foods contributes to a lower risk of PD in men and women [113, 114]. The relationship between serum β -carotene levels and the risk of PD is currently controversial in clinical studies. Some studies have reported that serum β-carotene levels are significantly lower in PD patients, while other studies suggest that there was no significant difference in serum β -carotene levels between PD patients and control groups [91, 115, 116]. Pretreatment of mice with large doses of β -carotene prevented the loss of GSH caused by MPTP and partially protected dopaminergic nigrostriatal neurons from damage in mice killed 30 days after injection with MPTP [117]. However, another group of investigators showed that large oral doses of β -carotene failed to prevent the neurotoxicity of MPTP in marmosets [118]. A recent study showed that β-carotene-loaded nanoparticles improved motor function, memory, and survival in Drosophila melanogaster, and also restored oxidative stress indicators including catalase (CAT), superoxide dismutase (SOD), ROS, and thiobarbituric acid reactive substances (TBARS), dopamine levels, and AChE activity after exposure to rotenone [119].

4.4 Lutein

Lutein, a carotenoid of the xanthophyll family, is found in some vegetables such as spinach, kale, and broccoli, as well as in marigold flowers, and is often used as a source of supplemental micronutrients [120]. In a rotenone-induced PD model of Drosophila melanogaster, administration of luteolin-loaded nanoparticles ameliorated rotenone-induced motor impairment and decreased survival. It also inhibited rotenone-induced reductions in dopamine levels and acetylcholinesterase activity and increases in oxidative stress indicators [121]. Additionally, lutein significantly reversed MPTP-induced neuronal apoptosis by inhibiting the activation of pro-apoptotic markers such as Bax, caspases-3, −8, and − 9 and enhancing the expression of the anti-apoptotic marker Bcl-2, as well as increasing dopamine levels in the striatum of mice. In addition, luteolin ameliorated MPTP-induced oxidative stress, mitochondrial dysfunction, and motor impairment, suggesting a neuroprotective effect of luteolin on dopaminergic neurons [122].

4.5 Fucoxanthin

Fucoxanthin is a natural carotenoid produced mainly by brown macroalgae and microdiatoms, which have many bioactive functions in neuroprotective agents [123]. Both in vivo and in vitro toxicity assays reveal that fucoxanthin lacks any relevant toxic effects, and a number of increasing studies demonstrate the potential benefits of fucoxanthin in neuroprotection [124, 125]. It has been showed that fucoxanthin exhibits concentration-dependent agonistic effects on dopamine D3 and D4 receptors with half maximal effective concentrations (EC50) of 16.87 and 81.87 μM, respectively. Mechanistically, fucoxanthin binds with the Ser196 and Thr115 of D3 receptors and Ser196 and Asp115 of D4 receptors with a low binding energy and H-bond interactions to exert its agonistic effects. These results suggest that fucoxanthin may be a potential D3/D4 agonist for the treatment of PD [126]. In addition, fucoxanthin reversed MPTP-induced activation of microglia and increase in pro-inflammatory cytokines in the SNpc region, and ameliorated α synuclein abnormal accumulation and motor impairment in PD mice [127]. Keap1/Nrf2 is the major pathway regulating antioxidant responses, which can protect against oxidative stress-induced cellular damage [86, 128]. Fucoxanthin blocks the interaction between Keap1 and Nrf2 by binding to the hydrophobic region of Keap1 pocket, which suppressed 6-hydroxydopamine (6-OHDA)-induced accumulation of ROS, the disruption of mitochondrial membrane potential, and apoptosis. Additionally, in 6-OHDA-induced PC12 cells, fucoxanthin also upregulated the expression of Keap1 downstream antioxidant enzymes in a dose-dependent manner, including nicotinamide heme oxygenase-1 (HO-1), glutamate-cysteine ligase modification subunit (GCLM), and glutamate-cysteine ligase catalytic subunit (GCLC) [129]. Furthermore, fucoxanthin can prevent neurotoxicity and suppress the high concentration of levodopa (L-DOPA)-induced cell apoptosis in the 6-OHDA-induced PC12 cells by improving the reduction in mitochondrial membrane potential, suppressing ROS expression, and inhibiting activation of ERK/JNK-c-Jun pathway and expression of cleaved caspase-3 [130].

4.6 Crocin and crocetin

Crocin and crocetin are two main carotenoids commonly found in Crocus sativus L. (saffron), which have been shown to have antioxidant and neuroprotective properties in various animal models [131, 132]. Similarly, crocin was also found to significantly alleviate rotenone-induced behavioral alterations, reduce oxidative stress, and enhance the levels of antioxidant enzymes and mitochondrial enzymes in the striatal region in a rotenone-induced PD mouse model. Furthermore, crocin differentially restores cholinergic function, dopamine and α-synuclein levels in the striatal region of mice [133]. In a rotenone-induced Drosophila model, saffron methanolic extract and crocin significantly reduced rotenone-induced oxidative stress, mitochondrial dysfunction, and cell death. Furthermore, saffron methanolic extract and crocin can also attenuate acetylcholinesterase (AChE) activity, restore dopamine levels, and delay the onset of locomotor deficits of flies, suggesting their effectiveness to mitigate cholinergic function [134]. Crocin administration attenuates ROT-induced neurotoxicity by increasing tyrosine hydroxylase (TH) and dopamine levels as well as decreasing α-synuclein expression. Mechanistically, crocin showed neuroprotective effects in rotenone-induced PD via activation of PI3K/Akt/mTOR signaling pathway and enhanced the expression of microRNA-7 (miRNA-7) and microRNA-221 (miRNA 221) [135]. Crocetin protects against 6-OHDA-induced neuronal damage and motor injury via increase of antioxidant enzyme activity and dopamine content in the striatum and decreasing TBARS content in the substantia nigra in a model of rat Parkinsonism [136]. Crocin improves MPTP-induced motor impairment and decreases cell death in the substantia nigra [137]. Additionally, crocin has been shown to reduce the nitrite levels in the hippocampus and improve the aversive memory of 6-OHDA rat model of PD [138]. Moreover, crocin inhibits endoplasmic reticulum(ER) stress by inverting the C/EBP homologous protein (CHOP)-Wnt pathway, which significantly attenuated MPP + -induced apoptosis and mitochondrial damage [139]. Depression is a common nonmotor symptom in patients with PD and difficult to treat [140]. Interestingly, crocin can inhibit the spontaneous discharge in dopaminergic neurons in the ventral tegmental area (VTA) by activating mTOR, which subsequently improves the synaptic plasticity in the medial prefrontal cortex (mPFC), and ameliorates depression-like behavior in PD mice [141]. According to evidence, environmental factors, such as pesticides, herbicides, and heavy metals, play a part in induction and progression of PD [142]. Malathion, an organophosphate insecticide that binds to cholinesterase enzymes, can cause Parkinson-like symptoms by activating the apoptotic pathway and increasing α-synuclein expression in the striatum [72]. Malathion induced apoptosis by increasing Bax/bcl2 ratio, expression of cleaved caspase-3 and cleaved caspase-9, and α-synuclein protein and RNA level in rat striatal tissues, wheat administration of crocin significantly reduced the apoptotic pathway, suggesting that crocin ameliorates the neurotoxic effects of malathion through its anti-apoptotic activity [72].

Abnormal aggregation and accumulation of α-synuclein in neural tissues is an important factor in the pathogenesis of PD [143]. It was found that saffron and its main components, crocin-1, crocin-2, and crocetin, dose-dependently inhibited α-synuclein aggregation and dissociated α-synuclein fibrils, with crocetin being the most potent. This effect was observed under a transmission electron microscope, which showed mainly a reduction and shortening of α-synuclein fibrils [144]. Mutations, A53T, A30p, and E46K in the α-synuclein gene, have been found to be associated with rare familial Parkinson. Among them, the E46K mutant protein more effectively promotes α-synuclein aggregation and increases amyloid fiber formation than wild-type and other mutations [145, 146]. Both results from molecular dynamics and docking studies indicate that crocin as a molecular chaperone may prevent the formation of α-synuclein aggregates by binding to the C-terminal and mainly NAC (NAM, ATAF1/2, and CUC2) domain of the E46K α-synuclein, and stabilize the protein by masking the aggregation hotspot, and consequently inhibit amyloid fibril formation in E46K α-synuclein [147]. Additionally, administering dietary saffron and crocetin to the PD transgenic fly models (A30P or G51D-mutated α-synuclein) had a positive effect on α-synuclein aggregation, climbing ability, survival rate, and eye morphological defects [148].

Advertisement

5. Conclusions

In PD cells and animal models, a growing number of studies have demonstrated that carotenoid administration protects dopaminergic neurons through various mechanisms such as quenching ROS, ameliorating mitochondrial damage, upregulating antioxidant enzyme systems, and causing anti-neuroinflammatory effects. Although the protective effects of certain carotenoids on neurons have been extensively studied, their specific mechanisms in vivo need to be further clarified. For example, the mechanisms by which carotenoids inhibit neuroinflammation and activate autophagy have not been thoroughly investigated. Additionally, clinical application studies in PD patients are needed. For example, the correlation between carotenoid intake and disease occurrence may be inferred from comparative studies of races that eat different foods. In terms of the complexity in the pathogenic mechanisms underlying PD, the protection of dopaminergic neurons by dietary carotenoids alone is limited. Thereby, other neuroprotective reagents or therapies that confer synergistic effects in combination with carotenoids in PD will be essential in searching for effective treatments.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

Advertisement

Notes/thanks/other declarations

N/A.

References

  1. 1. Kilzheimer A et al. The challenge and opportunity to diagnose Parkinson’s disease in midlife. Frontiers in Neurology. 2019;10:1328
  2. 2. Jadiya P et al. Sir-2.1 modulates ‘calorie-restriction-mediated’ prevention of neurodegeneration in Caenorhabditis elegans: Implications for Parkinson’s disease. Biochemical and Biophysical Research Communications. 2011;413(2):306-310
  3. 3. Gupta S, Benzeroual K. Neuroprotective effects of antioxidants, Idebenone and Ferulic acid, in MPTP/MPP+ intoxicated PC12 cells as a model of Parkinson’s disease. The FASEB Journal. 2013;27(S1):1175.7-1175.7
  4. 4. Takaichi S. Carotenoids in algae: Distributions, biosyntheses and functions. Marine Drugs. 2011;9(6):1101-1118
  5. 5. Britton G. Structure and properties of carotenoids in relation to function. The FASEB Journal. 1995;9(15):1551-1558
  6. 6. Bhosale P, Bernstein PS. Microbial xanthophylls. Applied Microbiology and Biotechnology. 2005;68(4):445-455
  7. 7. Augustynska D et al. Influence of polar and nonpolar carotenoids on structural and adhesive properties of model membranes. Chemico-Biological Interactions. 2015;239:19-25
  8. 8. Widomska J, Zareba M, Subczynski WK. Can xanthophyll-membrane interactions explain their selective presence in the retina and brain? Food. 2016;5(1):7
  9. 9. Britton G, Liaaen-Jensen SV,Pfander H. Functions of Intact Carotenoids. Berlin, Germany: Springer Verlag. 2008. DOI: 10.1007/978-3-7643-7499-0(Chapter 10): p. 189-212
  10. 10. Lu S, Li L. Carotenoid metabolism: Biosynthesis, regulation, and beyond. Journal of Integrative Plant Biology. 2008;50(7):778-785
  11. 11. Maida JM, Mathers K, Alley CL. Pediatric ophthalmology in the developing world. Current Opinion in Ophthalmology. 2008;19(5):403-408
  12. 12. Fraser PD, Bramley PM. The biosynthesis and nutritional uses of carotenoids. Progress in Lipid Research. 2004;43(3):228-265
  13. 13. Krinsky NI, Johnson EJ. Carotenoid actions and their relation to health and disease. Molecular Aspects of Medicine. 2005;26(6):459-516
  14. 14. Carpentier S, Knaus M, Suh M. Associations between lutein, zeaxanthin, and age-related macular degeneration: An overview. Critical Reviews in Food Science and Nutrition. 2009;49(4):313-326
  15. 15. Group, AR et al. The age-related eye disease study 2 (AREDS2): Study design and baseline characteristics (AREDS2 report number 1). Ophthalmology. 2012;119(11):2282-2289
  16. 16. Etminan M, Takkouche B, Caamano-Isorna F. The role of tomato products and lycopene in the prevention of prostate cancer: A meta-analysis of observational studies. Cancer Epidemiology, Biomarkers & Prevention. 2004;13(3):340-345
  17. 17. Fiedor J, Burda K. Potential role of carotenoids as antioxidants in human health and disease. Nutrients. 2014;6(2):466-488
  18. 18. Chen T et al. ROS-mediated mitochondrial dysfunction and ER stress contribute to compression-induced neuronal injury. Neuroscience. 2019;416:268-280
  19. 19. Brawn K, Fridovich I. DNA strand scission by enzymically generated oxygen radicals. Archives of Biochemistry and Biophysics. 1981;206(2):414-419
  20. 20. Burton GW, Ingold KU. Beta-carotene: An unusual type of lipid antioxidant. Science. 1984;224(4649):569-573
  21. 21. Weitberg AB et al. Effects of antioxidants on oxidant-induced sister chromatid exchange formation. The Journal of Clinical Investigation. 1985;75(6):1835-1841
  22. 22. Kellogg EW 3rd, Fridovich I. Superoxide, hydrogen peroxide, and singlet oxygen in lipid peroxidation by a xanthine oxidase system. The Journal of Biological Chemistry. 1975;250(22):8812-8817
  23. 23. Hira S et al. Beta-carotene: A natural compound improves cognitive impairment and oxidative stress in a mouse model of Streptozotocin-induced Alzheimer’s disease. Biomolecules. 2019;9(9):441
  24. 24. Shen XC, Qian ZY. Effects of crocetin on antioxidant enzymatic activities in cardiac hypertrophy induced by norepinephrine in rats. Pharmazie. 2006;61(4):348-352
  25. 25. Ochiai T et al. Crocin prevents the death of rat pheochromyctoma (PC-12) cells by its antioxidant effects stronger than those of alpha-tocopherol. Neuroscience Letters. 2004;362(1):61-64
  26. 26. Lorenzo Y et al. The carotenoid beta-cryptoxanthin stimulates the repair of DNA oxidation damage in addition to acting as an antioxidant in human cells. Carcinogenesis. 2009;30(2):308-314
  27. 27. Young AJ, Lowe GM. Antioxidant and prooxidant properties of carotenoids. Archives of Biochemistry and Biophysics. 2001;385(1):20-27
  28. 28. Lowe GM et al. Lycopene and beta-carotene protect against oxidative damage in HT29 cells at low concentrations but rapidly lose this capacity at higher doses. Free Radical Research. 1999;30(2):141-151
  29. 29. Muzandu K et al. Lycopene and beta-carotene ameliorate catechol estrogen-mediated DNA damage. The Japanese Journal of Veterinary Research. 2005;52(4):173-184
  30. 30. Niu T et al. Astaxanthin induces the Nrf2/HO-1 antioxidant pathway in human umbilical vein endothelial cells by generating trace amounts of ROS. Journal of Agricultural and Food Chemistry. 2018;66(6):1551-1559
  31. 31. Gu J et al. Astaxanthin-loaded polymer-lipid hybrid nanoparticles (ATX-LPN): Assessment of potential otoprotective effects. Journal of Nanobiotechnology. 2020;18(1):53
  32. 32. Yuan L et al. Protective effect of astaxanthin against La(2)O(3) nanoparticles induced neurotoxicity by activating PI3K/AKT/Nrf-2 signaling in mice. Food and Chemical Toxicology. 2020;144:111582
  33. 33. Deshmukh P et al. The Keap1-Nrf2 pathway: Promising therapeutic target to counteract ROS-mediated damage in cancers and neurodegenerative diseases. Biophysical Reviews. 2017;9(1):41-56
  34. 34. Nan B et al. Astaxanthine attenuates cisplatin ototoxicity in vitro and protects against cisplatin-induced hearing loss in vivo. Acta Pharmaceutica Sinica B. 2022;12(1):167-181
  35. 35. Xiong K et al. Construction of food-grade pH-sensitive nanoparticles for delivering functional food ingredients. Trends in Food Science & Technology. 2020;96:102-113
  36. 36. Hosaka S et al. Comparative study of antioxidants as quenchers or scavengers of reactive oxygen species based on quenching of MCLA-dependent chemiluminescence. Luminescence. 2005;20(6):419-427
  37. 37. Kaulmann A, Bohn T. Carotenoids, inflammation, and oxidative stress--implications of cellular signaling pathways and relation to chronic disease prevention. Nutrition Research. 2014;34(11):907-929
  38. 38. Cavaillon JM, Annane D. Compartmentalization of the inflammatory response in sepsis and SIRS. Journal of Endotoxin Research. 2006;12(3):151-170
  39. 39. Kang NJ et al. Cocoa procyanidins suppress transformation by inhibiting mitogen-activated protein kinase kinase. The Journal of Biological Chemistry. 2008;283(30):20664-20673
  40. 40. Barnes PJ, Karin M. Nuclear factor-kappaB: A pivotal transcription factor in chronic inflammatory diseases. The New England Journal of Medicine. 1997;336(15):1066-1071
  41. 41. Yeung YT et al. Signaling pathways in inflammation and anti-inflammatory therapies. Current Pharmaceutical Design. 2018;24(14):1449-1484
  42. 42. Sasaki M et al. Neuroprotective effect of an antioxidant, lutein, during retinal inflammation. Investigative Ophthalmology & Visual Science. 2009;50(3):1433-1439
  43. 43. Li SY et al. Anti-inflammatory effects of lutein in retinal ischemic/hypoxic injury: in vivo and in vitro studies. Investigative Ophthalmology & Visual Science. 2012;53(10):5976-5984
  44. 44. Rubin LP et al. Effect of carotenoid supplementation on plasma carotenoids, inflammation and visual development in preterm infants. Journal of Perinatology. 2012;32(6):418-424
  45. 45. Wu W et al. Lutein suppresses inflammatory responses through Nrf2 activation and NF-kappaB inactivation in lipopolysaccharide-stimulated BV-2 microglia. Molecular Nutrition & Food Research. 2015;59(9):1663-1673
  46. 46. Liu T et al. Lutein protects against beta-amyloid peptide-induced oxidative stress in cerebrovascular endothelial cells through modulation of Nrf-2 and NF-kappab. Cell Biology and Toxicology. 2017;33(1):57-67
  47. 47. Nam KN et al. Anti-inflammatory effects of crocin and crocetin in rat brain microglial cells. European Journal of Pharmacology. 2010;648(1-3):110-116
  48. 48. Fakhri S et al. Astaxanthin attenuates neuroinflammation contributed to the neuropathic pain and motor dysfunction following compression spinal cord injury. Brain Research Bulletin. 2018;143:217-224
  49. 49. Masoudi A et al. Anti-inflammatory and antioxidant effects of astaxanthin following spinal cord injury in a rat animal model. Brain Research Bulletin. 2021;177:324-331
  50. 50. Choi SK et al. Effects of astaxanthin on the production of NO and the expression of COX-2 and iNOS in LPS-stimulated BV2 microglial cells. Journal of Microbiology and Biotechnology. 2008;18(12):1990-1996
  51. 51. Zhou X et al. Inhibition of inflammation by astaxanthin alleviates cognition deficits in diabetic mice. Physiology & Behavior. 2015;151:412-420
  52. 52. Yeh PT et al. Astaxanthin inhibits expression of retinal oxidative stress and inflammatory mediators in Streptozotocin-induced diabetic rats. PLoS One. 2016;11(1):e0146438
  53. 53. Zhou XY et al. Depression can be prevented by astaxanthin through inhibition of hippocampal inflammation in diabetic mice. Brain Research. 2017;1657:262-268
  54. 54. Zhao D et al. Anti-Neuroinflammatory effects of Fucoxanthin via inhibition of Akt/NF-kappaB and MAPKs/AP-1 pathways and activation of PKA/CREB pathway in lipopolysaccharide-activated BV-2 microglial cells. Neurochemical Research. 2017;42(2):667-677
  55. 55. Sachdeva AK, Chopra K. Lycopene abrogates Abeta(1-42)-mediated neuroinflammatory cascade in an experimental model of Alzheimer’s disease. The Journal of Nutritional Biochemistry. 2015;26(7):736-744
  56. 56. Zhang F et al. Depression-like behaviors and heme oxygenase-1 are regulated by lycopene in lipopolysaccharide-induced neuroinflammation. Journal of Neuroimmunology. 2016;298:1-8
  57. 57. Zhao B et al. Supplementation of lycopene attenuates oxidative stress induced neuroinflammation and cognitive impairment via Nrf2/NF-kappaB transcriptional pathway. Food and Chemical Toxicology. 2017;109(Pt 1):505-516
  58. 58. Norbury CJ, Hickson ID. Cellular responses to DNA damage. Annual Review of Pharmacology and Toxicology. 2001;41:367-401
  59. 59. Elmore S. Apoptosis: A review of programmed cell death. Toxicologic Pathology. 2007;35(4):495-516
  60. 60. Huang C et al. The secretion from neural stem cells pretreated with lycopene protects against tert-butyl Hydroperoxide-induced neuron oxidative damage. Oxidative Medicine and Cellular Longevity. 2018;2018:5490218
  61. 61. Hwang S, Lim JW, Kim H. Inhibitory effect of lycopene on amyloid-beta-induced apoptosis in neuronal cells. Nutrients. 2017;9(8):883
  62. 62. Soleymaninejad M et al. The effects of lycopene and insulin on histological changes and the expression level of Bcl-2 family genes in the hippocampus of Streptozotocin-induced diabetic rats. Journal Diabetes Research. 2017;2017:4650939
  63. 63. Qu M et al. Lycopene prevents amyloid [Beta]-induced mitochondrial oxidative stress and dysfunctions in cultured rat cortical neurons. Neurochemical Research. 2016;41(6):1354-1364
  64. 64. Feng C et al. Lycopene protects human SH-SY5Y neuroblastoma cells against hydrogen peroxide-induced death via inhibition of oxidative stress and mitochondria-associated apoptotic pathways. Molecular Medicine Reports. 2016;13(5):4205-4214
  65. 65. Qu M et al. Lycopene protects against trimethyltin-induced neurotoxicity in primary cultured rat hippocampal neurons by inhibiting the mitochondrial apoptotic pathway. Neurochemistry International. 2011;59(8):1095-1103
  66. 66. Wang R, Lu X, Yu R. Lycopene inhibits epithelial-mesenchymal transition and promotes apoptosis in Oral cancer via PI3K/AKT/m-TOR signal pathway. Drug Design, Development and Therapy. 2020;14:2461-2471
  67. 67. Gloria NF et al. Lycopene and beta-carotene induce cell-cycle arrest and apoptosis in human breast cancer cell lines. Anticancer Research. 2014;34(3):1377-1386
  68. 68. Jeong Y, Lim JW, Kim H. Lycopene inhibits reactive oxygen species-mediated NF-kappaB signaling and induces apoptosis in pancreatic cancer cells. Nutrients. 2019;11(4):762
  69. 69. Satomi Y. Antitumor and cancer-preventative function of Fucoxanthin: A marine carotenoid. Anticancer Research. 2017;37(4):1557-1562
  70. 70. Guo SX et al. Astaxanthin attenuates early acute kidney injury following severe burns in rats by ameliorating oxidative stress and mitochondrial-related apoptosis. Marine Drugs. 2015;13(4):2105-2123
  71. 71. Kavitha K et al. 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. Biochimica et Biophysica Acta. 2013;1830(10):4433-4444
  72. 72. Mohammadzadeh L et al. Crocin protects malathion-induced striatal biochemical deficits by inhibiting apoptosis and increasing alpha-Synuclein in Rats’ striatum. Journal of Molecular Neuroscience. 2022;72(5):983-993
  73. 73. Zhang Y et al. Crocin treatment promotes the oxidative stress and apoptosis in human thyroid cancer cells FTC-133 through the inhibition of STAT/JAK signaling pathway. Journal of Biochemical and Molecular Toxicology. 2021;35(1):e22608
  74. 74. Chen W et al. Lutein attenuated methylglyoxal-induced oxidative damage and apoptosis in PC12 cells via the PI3K/Akt signaling pathway. Journal of Food Biochemistry. 2022;46(12):e14382
  75. 75. Kavalappa YP, Gopal SS, Ponesakki G. Lutein inhibits breast cancer cell growth by suppressing antioxidant and cell survival signals and induces apoptosis. Journal of Cellular Physiology. 2021;236(3):1798-1809
  76. 76. Zhu L, He C. Crocetin alleviates the caerulein-induced apoptosis and inflammation in AR42J cells by activating SIRT1 via NF-kappaB. Journal of Natural Medicines. 2022;76(2):410-418
  77. 77. Peng S, Wu Y, Wang Y. Crocetin suppresses uterine ischemia/reperfusion-induced inflammation and apoptosis through the Nrf-2/HO-1 pathway. Current Molecular Medicine. 2023;23(6):527-535
  78. 78. Moradzadeh M et al. Study of the mechanisms of crocetin-induced differentiation and apoptosis in human acute promyelocytic leukemia cells. Journal of Cellular Biochemistry. 2019;120(2):1943-1957
  79. 79. Choi AM, Ryter SW, Levine B. Autophagy in human health and disease. The New England Journal of Medicine. 2013;368(7):651-662
  80. 80. Shen M et al. Protective effect of astaxanthin on liver fibrosis through modulation of TGF-beta1 expression and autophagy. Mediators of Inflammation. 2014;2014:954502
  81. 81. Li J et al. Astaxanthin pretreatment attenuates hepatic ischemia reperfusion-induced apoptosis and autophagy via the ROS/MAPK pathway in mice. Marine Drugs. 2015;13(6):3368-3387
  82. 82. Cai X et al. Astaxanthin activated the Nrf2/HO-1 pathway to enhance autophagy and inhibit Ferroptosis, ameliorating acetaminophen-induced liver injury. ACS Applied Materials & Interfaces. 2022;14(38):42887-42903
  83. 83. Fung FK, Law BY, Lo AC. Lutein attenuates both apoptosis and autophagy upon cobalt (II) chloride-induced hypoxia in rat Muller cells. PLoS One. 2016;11(12):e0167828
  84. 84. Chang CJ et al. Lutein induces autophagy via Beclin-1 upregulation in IEC-6 rat intestinal epithelial cells. The American Journal of Chinese Medicine. 2017;45(6):1273-1291
  85. 85. Zeng C et al. Crocin-elicited autophagy rescues myocardial ischemia/reperfusion injury via paradoxical mechanisms. The American Journal of Chinese Medicine. 2016;44(3):515-530
  86. 86. Zhang L et al. Fucoxanthin provides neuroprotection in models of traumatic brain injury via the Nrf2-ARE and Nrf2-autophagy pathways. Scientific Reports. 2017;7:46763
  87. 87. Hald A, Lotharius J. Oxidative stress and inflammation in Parkinson’s disease: Is there a causal link? Experimental Neurology. 2005;193(2):279-290
  88. 88. Sies H, Stahl W, Sundquist AR. Antioxidant functions of vitamins. Vitamins E and C, beta-carotene, and other carotenoids. Annals of the New York Academy of Sciences. 1992;669:7-20
  89. 89. Yang F et al. Dietary antioxidants and risk of Parkinson’s disease in two population-based cohorts. Movement Disorders. 2017;32(11):1631-1636
  90. 90. Agarwal P et al. Dietary antioxidants associated with slower progression of parkinsonian signs in older adults. Nutritional Neuroscience. 2022;25(3):550-557
  91. 91. Kim JH et al. Association of serum carotenoid, retinol, and tocopherol concentrations with the progression of Parkinson’s disease. Nutrition Research and Practice. 2017;11(2):114-120
  92. 92. Ambati RR et al. Astaxanthin: Sources, extraction, stability, biological activities and its commercial applications--a review. Marine Drugs. 2014;12(1):128-152
  93. 93. Ikeda Y et al. Protective effects of astaxanthin on 6-hydroxydopamine-induced apoptosis in human neuroblastoma SH-SY5Y cells. Journal of Neurochemistry. 2008;107(6):1730-1740
  94. 94. Liu X, Osawa T. Astaxanthin protects neuronal cells against oxidative damage and is a potent candidate for brain food. Forum of Nutrition. 2009;61:129-135
  95. 95. Liu X et al. Astaxanthin inhibits reactive oxygen species-mediated cellular toxicity in dopaminergic SH-SY5Y cells via mitochondria-targeted protective mechanism. Brain Research. 2009;1254:18-27
  96. 96. Nonnekes J et al. MPTP-induced parkinsonism: An historical case series. Lancet Neurology. 2018;17(4):300-301
  97. 97. Ye Q et al. Astaxanthin protects against MPP(+)-induced oxidative stress in PC12 cells via the HO-1/NOX2 axis. BMC Neuroscience. 2012;13:156
  98. 98. Ye Q et al. Astaxanthin suppresses MPP(+)-induced oxidative damage in PC12 cells through a Sp1/NR1 signaling pathway. Marine Drugs. 2013;11(4):1019-1034
  99. 99. Lee DH, Kim CS, Lee YJ. Astaxanthin protects against MPTP/MPP+-induced mitochondrial dysfunction and ROS production in vivo and in vitro. Food and Chemical Toxicology. 2011;49(1):271-280
  100. 100. Grimmig B et al. Astaxanthin is neuroprotective in an aged mouse model of Parkinson’s disease. Oncotarget. 2018;9(12):10388-10401
  101. 101. Shen DF et al. Astaxanthin suppresses endoplasmic reticulum stress and protects against neuron damage in Parkinson’s disease by regulating miR-7/SNCA axis. Neuroscience Research. 2021;165:51-60
  102. 102. Kandy SK et al. Astaxanthin protection against neuronal Excitotoxicity via glutamate receptor inhibition and improvement of mitochondrial function. Marine Drugs. 2022;20(10):645
  103. 103. Hussein G et al. Astaxanthin, a carotenoid with potential in human health and nutrition. Journal of Natural Products. 2006;69(3):443-449
  104. 104. Grimmig B et al. Astaxanthin attenuates neurotoxicity in a mouse model of Parkinson’s disease. Functional Foods in Health and Disease. 2017;7(8):562
  105. 105. Wang CC et al. Docosahexaenoic acid-acylated astaxanthin ester exhibits superior performance over non-esterified astaxanthin in preventing behavioral deficits coupled with apoptosis in MPTP-induced mice with Parkinson’s disease. Food & Function. 2020;11(9):8038-8050
  106. 106. McCarty MF, Lerner A. Nutraceuticals targeting generation and oxidant activity of Peroxynitrite may aid prevention and control of Parkinson’s disease. International Journal of Molecular Sciences. 2020;21(10):3624
  107. 107. Rafi MM, Yadav PN, Reyes M. Lycopene inhibits LPS-induced proinflammatory mediator inducible nitric oxide synthase in mouse macrophage cells. Journal of Food Science. 2007;72(1):S069-S074
  108. 108. Khachik F et al. Chemistry, distribution, and metabolism of tomato carotenoids and their impact on human health. Experimental Biology and Medicine (Maywood, N.J.). 2002;227(10):845-851
  109. 109. Prema A et al. Neuroprotective effect of lycopene against MPTP induced experimental Parkinson’s disease in mice. Neuroscience Letters. 2015;599:12-19
  110. 110. Yi F, He X, Wang D. Lycopene protects against MPP(+)-induced cytotoxicity by maintaining mitochondrial function in SH-SY5Y cells. Neurochemical Research. 2013;38(8):1747-1757
  111. 111. Kaur H, Chauhan S, Sandhir R. Protective effect of lycopene on oxidative stress and cognitive decline in rotenone induced model of Parkinson’s disease. Neurochemical Research. 2011;36(8):1435-1443
  112. 112. Liu CB et al. Effect of lycopene on oxidative stress and behavioral deficits in rotenone induced model of Parkinson’s disease. Zhongguo Ying Yong Sheng Li Xue Za Zhi. 2013;29(4):380-384
  113. 113. Wu LY et al. Dietary beta-carotene and vitamin a and risk of Parkinson disease: A protocol for systematic review and meta-analysis. Medicine (Baltimore). 2022;101(41):e31002
  114. 114. Niu F et al. Vitamin C, vitamin E, beta-carotene and risk of Parkinson’s disease: A systematic review and dose-response meta-analysis of observational studies. Nutritional Neuroscience. 2023:1-13
  115. 115. Molina-Arjona JA et al. Serum pro-oxidant and antioxidant factors and risk of Parkinson’s disease: Population study. Revista de Neurologia. 1999;29(1):12-15
  116. 116. Larumbe Ilundain R et al. Case-control study of markers of oxidative stress and metabolism of blood iron in Parkinson’s disease. Revista Española de Salud Pública. 2001;75(1):43-53
  117. 117. Perry TL et al. Partial protection from the dopaminergic neurotoxin N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine by four different antioxidants in the mouse. Neuroscience Letters. 1985;60(2):109-114
  118. 118. Perry TL et al. Alpha-tocopherol and beta-carotene do not protect marmosets against the dopaminergic neurotoxicity of N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Journal of the Neurological Sciences. 1987;81(2-3):321-331
  119. 119. Chaves NSG et al. Beta-carotene-loaded nanoparticles protect against neuromotor damage, oxidative stress, and dopamine deficits in a model of Parkinson’s disease in drosophila melanogaster. Comparative Biochemistry and Physiology Part C: Toxicology & Pharmacology. 2023;268:109615
  120. 120. Sommerburg O et al. Fruits and vegetables that are sources for lutein and zeaxanthin: The macular pigment in human eyes. The British Journal of Ophthalmology. 1998;82(8):907-910
  121. 121. Fernandes EJ et al. Exposure to lutein-loaded nanoparticles attenuates Parkinson’s model-induced damage in Drosophila melanogaster: Restoration of dopaminergic and cholinergic system and oxidative stress indicators. Chemico-Biological Interactions. 2021;340:109431
  122. 122. Nataraj J et al. Lutein protects dopaminergic neurons against MPTP-induced apoptotic death and motor dysfunction by ameliorating mitochondrial disruption and oxidative stress. Nutritional Neuroscience. 2016;19(6):237-246
  123. 123. Loureno-Lopes C et al. Biological action mechanisms of fucoxanthin extracted from algae for application in food and cosmetic industries. Trends in Food Science & Technology. 2021
  124. 124. Kadekaru T, Toyama H, Yasumoto T. Safety evaluation of Fucoxanthin purified from Undaria pinnatifida. Nippon Shokuhin Kogyo Gakkaishi. 2008;55(6):304-308
  125. 125. Zaragoza MC et al. Toxicity and antioxidant activity in vitro and in vivo of two Fucus vesiculosus extracts. Journal of Agricultural and Food Chemistry. 2008;56(17):7773-7780
  126. 126. Paudel P et al. Characterizing fucoxanthin as a selective dopamine D(3)/D(4) receptor agonist: Relevance to Parkinson’s disease. Chemico-Biological Interactions. 2019;310:108757
  127. 127. Sun G et al. Fucoxanthin attenuates behavior deficits and neuroinflammatory response in 1-methyl-4-phenyl-1,2,3,6 -tetrahydropyridine-induced parkinson’s disease in mice. Pharmacognosy Magazine. 2020;16(67):51
  128. 128. Hu L et al. Neuroprotective role of fucoxanthin against cerebral ischemic/reperfusion injury through activation of Nrf2/HO-1 signaling. Biomedicine & Pharmacotherapy. 2018;106:1484-1489
  129. 129. Wu W et al. Fucoxanthin prevents 6-OHDA-induced neurotoxicity by targeting Keap1. Oxidative Medicine and Cellular Longevity. 2021;2021:6688708
  130. 130. Liu J et al. Fucoxanthin prevents long-term administration l-DOPA-induced neurotoxicity through the ERK/JNK-c-Jun system in 6-OHDA-Lesioned mice and PC12 cells. Marine Drugs. 2022;20(4):245
  131. 131. Nassiri-Asl, Marjan. Neuropharmacology Effects of Saffron (Crocus sativus) and its Active Constituents. Amsterdam, Netherlands: Elsevier Inc.; 2015. pp. 29-39
  132. 132. Hosseinzadeh H et al. Effects of saffron (Crocus sativus L.) and its active constituent, crocin, on recognition and spatial memory after chronic cerebral hypoperfusion in rats. Phytotherapy Research. 2012;26(3):381-386
  133. 133. Rao SV et al. Prophylactic neuroprotective propensity of Crocin, a carotenoid against rotenone induced neurotoxicity in mice: Behavioural and biochemical evidence. Metabolic Brain Disease. 2019;34(5):1341-1353
  134. 134. Rao SV et al. Evidence of neuroprotective effects of saffron and crocin in a drosophila model of parkinsonism. Neurotoxicology. 2016;52:230-242
  135. 135. Salama RM et al. Neuroprotective effect of crocin against rotenone-induced Parkinson’s disease in rats: Interplay between PI3K/Akt/mTOR signaling pathway and enhanced expression of miRNA-7 and miRNA-221. Neuropharmacology. 2020;164:107900
  136. 136. Ahmad AS et al. Neuroprotection by crocetin in a hemi-parkinsonian rat model. Pharmacology, Biochemistry, and Behavior. 2005;81(4):805-813
  137. 137. Haeri P et al. Neuroprotective effect of crocin on substantia nigra in MPTP-induced Parkinson’s disease model of mice. Anatomical Science International. 2019;94(1):119-127
  138. 138. Rajaei Z, Hosseini M, Alaei H. Effects of crocin on brain oxidative damage and aversive memory in a 6-OHDA model of Parkinson’s disease. Arquivos de Neuro-Psiquiatria. 2016;74(9):723-729
  139. 139. Zhang GF, Zhang Y, Zhao G. Crocin protects PC12 cells against MPP(+)-induced injury through inhibition of mitochondrial dysfunction and ER stress. Neurochemistry International. 2015;89:101-110
  140. 140. Reijnders JS et al. A systematic review of prevalence studies of depression in Parkinson’s disease. Movement Disorders. 2008;23(2):183-189 quiz 313
  141. 141. Tang J et al. Crocin reverses depression-like behavior in Parkinson disease mice via VTA-mPFC pathway. Molecular Neurobiology. 2020;57(7):3158-3170
  142. 142. Agim ZS, Cannon JR. Dietary factors in the etiology of Parkinson’s disease. BioMed Research International. 2015;2015:672838
  143. 143. Mehra S, Sahay S, Maji SK. Alpha-Synuclein misfolding and aggregation: Implications in Parkinson’s disease pathogenesis. Biochim Biophys Acta Proteins Proteom. 2019;1867(10):890-908
  144. 144. Inoue E et al. Effects of saffron and its constituents, crocin-1, crocin-2, and crocetin on alpha-synuclein fibrils. Journal of Natural Medicines. 2018;72(1):274-279
  145. 145. Polymeropoulos MH et al. Mutation in the alpha-synuclein gene identified in families with Parkinson’s disease. Science. 1997;276(5321):2045-2047
  146. 146. Zarranz JJ et al. The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia. Annals of Neurology. 2004;55(2):164-173
  147. 147. Ghasemi Tigan M, Ghahghaei A, Lagzian M. In-vitro and in-silico investigation of protective mechanisms of crocin against E46K alpha-synuclein amyloid formation. Molecular Biology Reports. 2019;46(4):4279-4292
  148. 148. Inoue E et al. Saffron ameliorated motor symptoms, short life span and retinal degeneration in Parkinson’s disease fly models. Gene. 2021;799:145811

Written By

Fengjuan Jiao

Submitted: 19 June 2023 Reviewed: 22 June 2023 Published: 18 October 2023