Skip to main content
Oxidative Medicine and Cellular Longevity logoLink to Oxidative Medicine and Cellular Longevity
. 2019 Feb 20;2019:9426867. doi: 10.1155/2019/9426867

Benefits of Vitamins in the Treatment of Parkinson's Disease

Xiuzhen Zhao 1, Ming Zhang 2, Chunxiao Li 3, Xue Jiang 1, Yana Su 1, Ying Zhang 1,
PMCID: PMC6402202  PMID: 30915197

Abstract

Parkinson's disease (PD) is the second most common neurodegenerative disease in the elderly, which is clinically characterized by bradykinesia, resting tremor, abnormal posture balance, and hypermyotonia. Currently, the pathogenic mechanism of PD remains unclear. Numerous clinical studies as well as animal and cell experiments have found a certain relationship between the vitamin family and PD. The antioxidant properties of vitamins and their biological functions of regulating gene expression may be beneficial for the treatment of PD. Current clinical evidence indicates that proper supplementation of various vitamins can reduce the incidence of PD in the general population and improve the clinical symptoms of patients with PD; nevertheless, the safety of regular vitamin supplements still needs to be highlighted. Vitamin supplementation may be an effective adjuvant treatment for PD. In this review, we summarized the biological correlations between vitamins and PD as well as the underlying pathophysiological mechanisms. Additionally, we elaborated the therapeutic potentials of vitamins for PD.

1. Introduction

Parkinson's disease (PD) is the second most common neurodegenerative disorder following Alzheimer's disease. Clinically, PD is characterized by resting tremor, hypermyotonia, postural instability, and bradykinesia [1]. Additionally, patients with PD can also manifest with nonmotor symptoms, such as cognitive decline, olfactory dysfunction, constipation, sleep disorders, and autonomic symptoms [2], and these nonmotor symptoms usually occur prior to the onset of motor symptoms [3]. PD severely affects the quality of life of the individual with the disease and also creates a great burden on the caregivers. The typical pathological hallmark of PD is degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and eosinophilic inclusion bodies (Lewy bodies) in the remaining neurons, which is the major contributor to the deficiency of dopamine in the basal ganglia [4, 5]. The exact pathogenetic mechanisms of PD is not yet fully understood. Current theories regard PD as a multifactorial disease, involving various genetic and environmental factors, among which mitochondrial dysfunction and oxidative stress play an important role in the pathogenesis and development of PD [6, 7]. The treatment for PD is challenging, and the existing therapeutic strategies can only relieve clinical symptoms but fail to control the progression of PD.

Vitamins are natural bioactive products with antioxidant properties, which are necessities for maintaining the normal functions of human organisms. Essential vitamins cannot be endogenously synthesized in the organism and therefore must be obtained through the diet. Clinically, vitamin deficiency is quite common, especially in infants and elderly. Vitamins are generally divided into fat-soluble variants (vitamins A, D, E, and K) and water-soluble variants (vitamins B and C). The former mainly bind to cellular nuclear receptors and affect the expression of specific genes [8]. The latter mainly constitute a cofactor for the enzyme, affecting the enzymatic activity [9].

Numerous clinical studies as well as animal and cell experiments have found a certain relationship between the vitamin family and PD [10]. The antioxidant properties of vitamins and their biological functions of regulating gene expression may be beneficial for the treatment of PD. Current clinical evidence indicates that proper supplementation of various vitamins can reduce the incidence of PD in the general population and improve the clinical symptoms of patients with PD; nevertheless, the safety of regular vitamin supplements still needs to be highlighted. Vitamin supplementation may represent an effective adjuvant treatment for PD. In this review, we summarized the biological correlations between vitamins and PD as well as the underlying pathophysiological mechanisms. Additionally, we elaborated the therapeutic potentials of vitamins for PD.

2. The Pathogenesis of Oxidative Stress in PD

Oxidative stress refers to the imbalance between the oxidation system and antioxidant system, resulting in excessive accumulation of oxidative substances, such as reactive oxygen species (ROS) and reactive nitrogen species (RNS) [11]. ROS include superoxide anion radical (O2), hydroxyl radical (OH), and hydrogen peroxide (H2O2); RNS include nitric oxide (NO), nitrogen dioxide (NO2), and peroxynitrite (ONOO). The antioxidant system mainly consists of two subtypes: (1) enzymatic antioxidant system, including superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-Px), and (2) nonenzymatic antioxidant system, including vitamin C, vitamin E, glutathione, melatonin, alpha-lipoic acid, carotenoids, and trace elements copper, zinc, and selenium.

Oxidative stress plays an important physiological role in the organism. For example, phagocytic cells kill pathogenic microorganisms, participate in detoxification and enzymatic reactions, and synthesize some essential biologically active substances. Meanwhile, it can as well cause damage to the body, such as cell membrane destruction, protein denaturation, and nucleic acid changes.

There is increasing evidence that oxidative stress represents a pathophysiological characteristic of PD, and the production of reactive oxygen species can result in neuronal death [12, 13]. The mitochondrial respiratory chain is regarded as the major source of ROS [14]. Additionally, previous studies have found that mitochondrial dysfunction exists in the substantia nigra of patients with PD [15]. Reduced glutathione (GSH) can enhance the production of ROS and RNS [16], and oxidation of dopamine and dopamine metabolites such as 3,4-dihydroxyphenylacetic acid (DOPAC) can inhibit the activity of complex I [17]. These findings indicate that the downstream metabolites of dopamine may make dopamine neurons more susceptible. Moreover, iron accumulation in the substantia nigra is common in patients with PD, leading to overproduction of hydrogen peroxide and molecular oxygen in the Fenton reaction from Fe2+ to Fe3+; hydrogen peroxide generates a highly toxic hydroxyl radical through the Haber-Weiss reaction in the presence of Fe2+, which causes severe oxidative damage to the cellular components [18]. From the above, the oxidant stress is closely associated with the pathogenesis of PD. Oxidative stress can cause neuronal loss through some underlying intracellular damage, such as protein aggregation, mitochondrial dysfunction, and DNA rupture. Therefore, antioxidant damage has become a potential target for the treatment of PD.

3. Vitamin B and PD

The B family of vitamins is water-soluble, which includes thiamine (vitamin B1), riboflavin (vitamin B2), niacin (vitamin B3), pantothenic acid (vitamin B5), pyridoxine (vitamin B6), biotin (vitamin B7), folate (vitamin B9), and cobalamin (vitamin B12) [19]. These vitamins play an important role as enzyme cofactors in multiple biochemical pathways in all tissues, such as regulating metabolism, improving the function of the immune system and nervous system, and promoting cell growth and division [20].

Almost all of these B family vitamins are essential variants dependent on diet supply, except niacin which can also be synthesized from tryptophan. Vitamin B deficiencies are frequent in the children, elderly, vegetarians, pregnant women, and patients with gastrointestinal diseases. Recently, the association with vitamin B and PD is getting more and more attention. Herein, we use vitamin B3 as a representative to discuss the relationship between vitamin B and PD.

3.1. Vitamin B3

Nicotinamide is the active form of niacin, and it is the precursor of coenzymes NADH and NADPH, which are essential for over 200 enzymatic reactions in the organism, especially the production of adenosine triphosphate (ATP). Meat, fish, and wheat are generally rich in nicotinamide, while vegetables have a low nicotinamide content [21]. Deficiency of nicotinamide/niacin can lead to pellagra, causing dermatitis, diarrhea, and depression [22]. Nicotinamide has neuroprotective and antioxidant functions at low doses but exhibits neurotoxicity, especially dopaminergic toxicity, at high doses [23]. Fukushima also suggests that excessive nicotinamide is related to the development of PD [24]; excessive nicotinamide can induce overproduction of 1-methylnicotinamide (MNA), which is increased in patients with PD [25]. In an in vitro study, Griffin et al. found that low-dose nicotinamide (10 mM) has a significant effect on inducing differentiation from embryonic stem cells into neurons; however, higher doses (>20 mM) of nicotinamide induce cytotoxicity and cell death [26]. The definitive protective dose of vitamin B3 still needs further researches.

3.2. Possible Neuroprotective Mechanisms of Vitamin B3 in PD

Firstly, numerous studies have demonstrated that mitochondrial dysfunction and cellular energy failure are pathophysiological features of PD. Nicotinamide participates in the biosynthesis of nicotinamide adenine dinucleotide (NAD; oxidized form: NAD+; reduced form: NADH) via various metabolic pathways [27]. NADH is an essential cofactor assisting the tetrahydrobiopterin functioning in tyrosine hydroxylase, which can hydroxylate tyrosine and produce dopamine; NADH deficiency is common in PD [28].

Secondly, NADH is indispensable for the physiological function of mitochondrial complex I in ATP synthesis, and the corresponding dysfunction is involved in PD patients and animal models [15, 29, 30]. Nicotinamide mononucleotide (NMN) constitutes one of the key precursors of NAD+. In previous in vitro studies, the scholars have established a cellular model of PD using rotenone-treated PC12 cells, and they found the NMN (0.1 mM or 1 mM) treatment was associated with a significantly higher survival rate in the rotenone-treated (0.5 μm) PC12 cells. NMN is assumed to enhance the intracellular levels of NAD+ and ATP in the cellular model of PD [31].

In addition, nicotinamide can act a neuroprotective role by inhibiting the oxidative stress. In 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine- (MPTP-) induced mouse models of PD, nicotinamide (500 mg/kg) was injected before subacute (30 mg/kg/d for 5 days) MPTP administration. This study showed that cotreatment with MPTP and nicotinamide significantly improved the locomotor activity compared to single-agent treatment with MPTP. Nicotinamide administration significantly attenuated the MPTP-induced dopamine depletion (47.11 ± 21.06 vs. 12.77 ± 8.06). Meanwhile, nicotinamide pretreatment markedly inhibited MPTP-induced lactate dehydrogenase (LDH) and NOS activities, which prevented the oxidative stress and alleviated the oxidative damage.

Sirtuins (SIRTs) are NAD+-dependent protein deacetylases involved in vital biological processes [32]. Recently, sirtuin 5 (SIRT5) has received considerable attention. Liu et al. investigated the role of SIRT5 in MPTP-induced mouse PD models [33]. They found that deletion of SIRT5 exacerbated motor deficits, nigrostriatal dopaminergic degeneration in the compact part of substantia nigra (SNc), and mitochondrial antioxidant activities in the PD models. These findings provide new insight into the therapeutic strategies for PD. However, the protective effects of nicotinamide are still controversial, and further researches are needed to clarify the biological function of vitamin B3 in PD.

3.3. Clinical Studies regarding Vitamin B3 in PD

Current existing clinical studies have shown that a high-niacin diet can reduce the risk of PD [34, 35]. A previous case report also demonstrated that oral niacin (500 mg twice daily for three months) significantly improved rigidity and bradykinesia in a patient with idiopathic PD, though the original purpose was to treat hypertriglyceridemia; after the cessation of oral niacin due to obvious adverse effects (unacceptable nightmares and skin rash), the symptoms of rigidity and bradykinesia relapsed [36]. However, other studies failed to notice the remarkable clinical efficacy [37, 38]. Therefore, more clinical observations are warranted to verify the efficacy as well as side effects of niacin in PD.

4. Vitamin C and PD

Vitamin C (ascorbic acid) is another water-soluble, essential vitamin, which is widely distributed in various tissues. This nutriment is abundant in vegetables, fresh fruits, and animal livers. Vitamin C contains two molecular subforms in organisms: the reduced form (ascorbic acid (AA)) and the oxidized form (dehydroascorbic acid (DHA)). Deficiency of vitamin C is common, especially in children and elderly. A long-term lack of vitamin C can cause scurvy. Vitamin C is very important for the physiological function of the nervous system and the antioxidant function by inhibiting oxidative stress, reducing lipid peroxidation, and scavenging free radicals [39]. Moreover, it is also involved in many nonoxidative stress processes, such as synthesis of collagen, cholesterol, carnitine, catecholamines, amino acids, and some peptide hormones [40, 41].

Dopamine metabolism can produce oxidative stress products, which in return induce accumulation of abnormal proteins in PD [42]. Vitamin C has potentials for the treatment of PD considering the following reasons. Firstly, vitamin C is mainly distributed in areas that are rich in neurons [43, 44]. Secondly, vitamin C can be transported to the brain by SVCT2 (vitamin C transporter type 2) [45], and DHA can be transported to the brain by GLUT1 (glucose transporter type 1) and GLUT3 (glucose transporter type 3) [46].

4.1. Possible Neuroprotective Mechanisms of Vitamin C in PD

There is evidence that ascorbic acid can protect against both levodopa toxicity and the MPTP neurotoxicity [47, 48]. Vitamin C can increase the production of dihydroxyphenylalanine (DOPA). Seitz et al. noted overproduction of DOPA in a dose-dependent manner after incubation of the human neuroblastoma cell line SK-N-SH with ascorbic acid (100-500 mM) for 2 hours. Additionally, the gene expression of tyrosine hydroxylase increased threefold after incubation with ascorbic acid (200 mM) for 5days. The scholars speculated that ascorbic acid may be effective in the treatment of early-stage PD [49].

Vitamin C can improve the absorption of levodopa in elderly PD patients with a poor levodopa bioavailability [50]. Previous studies showed that ascorbic acid can reduce the levodopa dosage under the premise of equal efficacy [51]. Combination of anti-PD drugs and vitamin C may be more effective for alleviating the symptoms of PD.

Vitamin C is essential for the brain development. A study showed that ascorbic acid treatment can promote a 10-fold increase of dopaminergic differentiation in CNS precursor cells derived from the E12 rat mesencephalon [52]. Soon after, another in vitro study also reported that AA can stimulate the CNS precursor cells differentiating into CNS neurons and glia [53]. Recently, He et al. proposed that vitamin C can greatly enhance the embryonic midbrain neural stem cells differentiating into midbrain dopaminergic neurons in vitro. Vitamin C induces the gain of 5-hydroxymethylcytosine (5HMC) and loss of H3K27m3 in dopaminergic phenotype gene promoters, which are catalysed by ten-eleven translocation 1 methylcytosine dioxygenase 1 (TET1) and histone H3K27 demethylase (JMJD3), respectively. However, subsequent TET1 and JMJD3 knockdown/inhibition experiments did not show this effect of vitamin C, and the epigenetic role of vitamin C may be associated with the midbrain dopaminergic neuron development [54, 55].

4.2. Clinical Studies regarding Vitamin C in PD

Although vitamin C has many potential positive effects on PD, the serum level of vitamin C in patients with PD remains controversial [56, 57]. Noteworthily, the vitamin C level in lymphocytes has been found significantly lower in patients with severe PD [58]. Theoretically, vitamin C supplementation may be beneficial for the treatment of PD. A cohort study involving 1036 patients with PD supported this hypothesis, which found that dietary vitamin C intake significantly reduced the risk of PD, but this effect is invalid for a 4-year-lag analysis [59]. Controversially, many studies did not support that vitamin C supplementation can reduce the risk of PD [10, 60, 61]. We speculate this contradiction may be related to the timing of vitamin application.

5. Vitamin E and PD

Vitamin E is a fat-soluble vitamin with high antioxidant properties. Natural vitamin E includes two subgroups: tocopherols and tocotrienols; and they can further be divided into four lipophilic molecules, respectively: α-, β-, γ-, and δ-tocopherol (αT, βT, γT, and δT) and α-, β-, γ-, and δ-tocotrienol (αTE, βTE, γTE, and δTE). The major difference between tocopherols and tocotrienols is the side chain. Tocopherols have a saturated phytyl tail, while tocotrienols possess an unsaturated isoprenoid side chain [62]. Because of this unsaturated side chain, the tocotrienol is superior to the tocopherol as an antioxidant by increasing the molecular mobility through lipid membranes and by accepting electrons readily. Overt vitamin E deficiency is relatively rare, mainly in infants and premature babies.

In addition to its potent antioxidant capacity, vitamin E is involved in many physiological processes such as immune function [63], cognitive function, physical performance [64, 65], and regulation of gene expression. In humans, deficiency of vitamin E is clinically characterized by peripheral neuropathy, ataxia, and anemia [66, 67].

5.1. Possible Neuroprotective Mechanisms of Vitamin E in PD

Unilateral 6-hydroxydopamine (6-OHDA) injections into the striatum can cause circling behaviours and biochemical abnormalities in rats. Cadet et al. found that pretreatment with either D-alpha-tocopherol or all-racemic-alpha-tocopherol significantly attenuated these pathological changes [68]. Roghani and Behzadi [69] and Sharma and Nehru [70] also demonstrated the similar phenomenon in 6-OHDA-induced PD models and in rotenone-induced PD models, respectively. However, some studies have shown that vitamin E did not completely protect dopaminergic neurons from MPTP-mediated damage in PD models [71, 72]. The protective effects of vitamin E may be achieved through preventing oxidative stress in cells and inhibiting apoptosis. Moreover, one study has found that tocotrienol participates not only in antioxidant stress but also in estrogen receptor beta (ERβ) signal transduction [73]. Then, Nakaso's team demonstrated a protective effect of vitamin E via this signaling pathway. Firstly, they reported that γ-tocotrienol/δ-tocotrienol exerts neuroprotective effects through the ERβ-PI3K/Akt signaling pathways in SH-SY5Y cells by resisting 1-methyl-4-phenylpyridiniumion- (MPP+-) induced toxicity [74]. Secondly, they verified this mechanism in a mouse model of PD. Meanwhile, they found δ-tocotrienol administration can reduce the loss of dopaminergic neurons in the substantia nigra and ER inhibitors can attenuate this neuroprotective effect [75]. These findings indicate vitamin E may be potential therapeutic agents for PD.

5.2. Clinical Studies regarding Vitamin E in PD

The DATATOP (Deprenyl and Tocopherol Antioxidative Therapy of Parkinsonism) experiment is a multicentre-controlled clinical trial to investigate the long-term efficacy of treatment with deprenyl and/or copherol (vitamin E) and to explore whether it is possible to extend the time before the application of levodopa treatment. At 28 US and Canadian sites, 800 eligible patients with untreated early-stage PD were enrolled in DATATOP and randomized to four groups: (1) deprenyl 10 mg/d, (2) copherol 2000 IU/d, (3) placebo-controlled, and (4) deprenyl 10 mg/d and copherol 2000 IU/d. Deprenyl can delay the development of functional disorders, delay the application of levodopa, and improve motor symptoms, but vitamin E is disappointing [76]. Similarly, another two population-based studies also did not find the association between vitamin E intake and risk of PD [10, 77].

However, a large community-based study showed that high intake of dietary vitamin E (10 mg/day) may reduce the occurrence of PD [78]. Another pilot trail suggests that long-term treatment with vitamin E may delay the use of levodopa in patients with PD [79]. Further research is needed to verify these results.

Although there seems to be no difference in the level of alpha-tocopherol (vitamin E) in serum, cerebrospinal fluid, and brains between PD and normal controls [8082], there is evidence showing that high-dose vitamin E (2000 IU/day) can significantly elevate the vitamin E level in cerebrospinal fluid [83]. At present, the protective mechanism of vitamin E in PD is still unclear and may be related to the strong antioxidant effect of vitamin E. Further research is needed to determine whether vitamin E can be used as a potential treatment for PD.

6. Vitamin D and PD

Vitamin D, a steroid hormone, is crucial for calcium homeostasis and skeletal health. This nutriment mainly includes two forms: vitamin D2 and vitamin D3; the latter is endogenously produced when skin is exposed to UV-B rays from the sun. Both of the above forms are inactive, and they are transformed into the active form 1,25-dihydroxy vitamin D3 (1,25-(OH)2-D3) after being hydroxylated twice [84, 85]. 1,25-(OH)2-D3 were secreted into the blood system by the kidney, having a direct effect on gene regulation by binding to the nuclear vitamin D receptor (VDR) [86, 87]. Vitamin D deficiency is prevalent at all ages, especially in elderly. Vitamin D not only regulates the calcium homeostasis and skeletal health but also regulates the physiological and pathological processes, such as cell proliferation and differentiation, immunomodulatory, and antioxidative stress [8890]. Children with a lack of vitamin D may suffer from rickets, and adults may develop osteomalacia. Additionally, vitamin D deficiency is also associated with cardiovascular diseases, muscle weakness, diabetes mellitus, cancers, and multiple sclerosis [91]. The relationship between vitamin D and PD has gradually attracted attention [92].

6.1. Possible Neuroprotective Mechanisms of Vitamin D in PD

VDR belongs to the intranuclear receptor superfamily, composing of eight coding exons and three alternative 5′ noncoding exons, spanning over 105 kb, on chromosome 12 [93]. The most widely studied biallelic polymorphic sites are BsmI, TaqI, ApaI, and FokI. Substantial researches have been carried out to explore the relationship between these allelic variations and PD. Kim et al. detected VDR gene BsmI polymorphisms in over 300 Korean individuals (85 PD and 231 controls). The frequency of VDR genotype bb was significantly increased in the PD patients (84.7%) than that in the controls (72.7%). The bb genotype was more common in PD patients with postural instability and gait difficulty than in the PD patients with tremor (94.3% vs. 75.6%) [94]. A meta-analysis showed that VDR BsmI and FokI polymorphisms were associated with the risk of PD [95], and VDR FokI genotype was associated with the severity and cognitive decline of PD [96, 97]. Muscular and motor impairments, which can seriously affect the motor behaviour, were found in the VDR-knockout mice [98], indicating that vitamin D may be involved in the pathogenesis of PD.

Glial cell line-derived neurotrophic factor (GDNF) is a protein that is essential for the maintenance and survival of dopaminergic neurons and can inhibit microglial activation [99]. Many animal studies showed that 1,25-(OH)2-D3 could enhance the endogenous GDNF expression in vitro and in vivo and inhibit the glial cell activation to protect dopaminergic neurons from immune inflammation [100102].

Vitamin D3 can protect dopaminergic neurons against 6-hydroxydopamine-mediated neurotoxicity and improve the motor performance in the 6-hydroxydopamine-induced PD rat [103]. It may be related to vitamin D's properties of inhibiting oxidative stress and decreasing the production of reactive oxygen species and free radicals [104]. In addition, endothelial dysfunction may be associated with low vitamin D levels in patients with PD [105]. The definitive correlations between vitamin D and PD require more researches.

6.2. Clinical Studies regarding Vitamin D in PD

Substantial epidemiological and clinical studies suggest that vitamin D has a positive effect on PD. In a cohort study, over 7000 Finnish's serum samples were collected for measuring the 25-hydroxy vitamin D level, and meanwhile, the occurrence of PD was instigated over a 30-year follow-up period. The results showed that individuals with higher serum vitamin D concentrations had a lower risk of PD [106]. Evatt et al. also noted consistent findings [107].

As mentioned above, vitamin D3 can be endogenously synthesized upon sunlight exposure in the skin. In a large case-control study of Danish men, involving 3819 PD patients and 19,282 controls, the scholars proposed that men working outdoors have a lower risk of PD [108]. Another nationwide ecologic study in France also suggests that vitamin D levels are negatively correlated with the risk of PD, but this result needs taking ages into account [109]. Wang et al. not only demonstrated a positive correlation between serum 25-hydroxy vitamin D and sunlight exposure but also noted that lower serum levels of 25-hydroxy vitamin D and sunlight exposure can increase the risk of PD [110].

Furthermore, PD patients with lower 25-hydroxy vitamin D levels may exhibit more severe symptoms compared with normal controls [111, 112]. Unsurprisingly, a randomized, double-blind, placebo-controlled trial found that vitamin D3 supplementation (1200 IU/day for 12 months) significantly prevented the deterioration of PD [113].

7. Conclusion

In summary, vitamins may play a protective role in PD. Among the fat-soluble vitamins, we briefly summarized the effects of vitamin E and vitamin D. At present, although many studies have shown that vitamin E supplementation can reduce the risk of Parkinson's disease (Table 1), the DATATOP study has showed that vitamin E supplementation is ineffective in Parkinson's disease (Table 2). Many noninterventional studies found that the high levels of serum vitamin D can reduce the risk of PD (Table 1), and several clinical intervention trials also proposed that vitamin D supplementation can attenuate the deterioration of the Parkinson's disease and reduce the occurrence of fractures in patients with PD (Table 2). Among the water-soluble vitamins, we elaborated the functions of vitamin B3 and vitamin C. There is still a paucity of clinical evidence for determining the pros and cons of vitamin B3 in PD (Table 2). Vitamin C is vital to the human organism, and it can improve levodopa absorption in elderly PD patients (Table 2); current epidemiological evidence is still insufficient to establish a correlation between the serum level or dietary intakes of vitamin C and the risk of PD (Table 1). Although there have been many researches on the relationship between vitamins and PD (Tables 13), there is still lack of a clinical intervention trial explicitly confirming that vitamin supplementation can reduce the incidence of PD and prevent the progression of the disease. Moreover, the individual physical and chemical properties, absorption rate, and bioavailability of vitamins may affect the efficacy. Further studies are still needed to clarify the potentials of vitamins for the treatment of PD.

Table 1.

The other clinical study of vitamins and Parkinson's disease.

Vitamins Authors Type of study Patients/controls Conclusions
Vitamin B3 Abbott et al. [37] A Honolulu-Asia Aging Study in Japanese-American Total 8006 and observed 137 PD Niacin has no obvious relationship with clinical PD
Johnson et al. [38] A case-control study in US 126/432 Niacin has no relationship with PD
Fall et al. [34] A case-control study in Sweden 113/263 High-niacin diet can reduce the risk of PD
Hellenbrand et al. [35] A case-control study in German 342/342 PD patients with lower intake of niacin than controls
Vitamin C Yang et al. [61] A prospective study in Sweden Total 84,774 and observed 1329 PD cases Intake of vitamin C has a negative correlation with PD risk in women at borderline significance (P = 0.04)
Hughes et al. [59] A prospective study in American Total 129,422 and observed 1036 PD cases Intake of vitamin C has no relationship with PD risk
Ide et al. [58] A case-case study in Japan 62 PD The severe PD patients with significantly lower lymphocyte vitamin C levels (P < 0.01)
Miyake et al. [60] A case-control study in Japan 249/368 Intake of vitamin C has no relationship with PD risk
Zhang et al. [10] A prospective study in US Total 124,221 and observed 371 PD cases Intake of vitamin C has no relationship with PD risk
Férnandez-Calle et al. [56] A case-control study in Spain 63/63 Vitamin C has no relationship with PD
King et al. [127] A case-control study in United States 27/16 Vitamin C was higher in PD groups
Vitamin E Yang et al. [61] A prospective study in Sweden Total 84,774 and observed 1329 PD cases Dietary intake of vitamin E has negative correlation with the incidence of PD in women (P = 0.02)
Hughes et al. [59] A prospective study in American Total 129,422 and observed 1036 PD cases Vitamin E has no relationship with PD risk
Miyake et al. [60] A case-control study in Japan 249/368 Vitamin E significantly reduced the risk of PD
Zhang et al. [10] A prospective study in US Total 124,221 and observed 371 PD cases Intaking foods containing more vitamin E can reduce the risk of Parkinson's disease
Molina et al. [82] A case-control study in Spain 34/47 CSF and serum vitamin E levels have no difference between two groups
de Rijk et al. [78] A cross-sectional study in Netherlands 5342 individuals including 31 PD cases Intaking 10 mg dietary vitamin E daily may reduce the risk of PD
Logroscino et al. [77] A case-control study in USA 110/287 Vitamins A, C, and E were not associated with PD
Férnandez-Calle et al. [80] A case-control study in Spain 42/42 Serum levels of alpha-tocopherol (vitamin E) have no difference between two groups
Vitamin D Kim et al. [128] A prospective, observational study in Korea 39 PD cases The level of vitamin D might impact the olfactory dysfunction in PD
Sleeman et al. [112] A prospective observational study in England 145/94 Serum 25(OH)D concentrations are often lower in PD patients than controls and relate to the severity of motor symptoms
Wang et al. [110] A case-control study in China 201/199 The serum 25(OH)D and sunlight exposure inversely correlated with PD occurrence
Shrestha et al. [129] A prospective study in USA Total 12,762 participants and observed 67 PD cases This study did not suggest that the vitamin D can reduce the risk of PD
Liu and Zhang [111] A case-control study in China 229/120 The 25(OH) D levels may be inversely associated with the PD severity
Lin et al. [130] A case-control study in China 700/792 They have not found the associations between the genetic variants of VDR and PD occurrence
Zhu et al. [131] A case-control study in China 209/210 Outdoor activity and total vitamin D intake may reduce the risk of PD
Petersen et al. [132] A case-control study in Denmark 121/235 They have not found the association between PD and vitamin D polymorphisms and/or 25(OH)D levels
Török et al. [133] A case-control study in Hungary 100/109 It showed the association between the FokI C allele and PD
Lv et al. [134] A case-control study in China 483/498 The study did not support the relationship between VDR gene and PD
Peterson et al. [135] A cross-sectional, observational study in USA 40 PD Serum vitamin D levels are inversely related to the severity of Parkinson's disease and play an important role in balance of PD
Suzuki et al. [96] A prospective cohort study in Japan 137 PD The 25-hydroxyvitamin D levels and the vitamin D receptor FokI CC genotype may be associated with the severity of the PD
Kenborg et al. [108] A case-control study in Denmark 3819/19,282 This study supports that working outdoors can reduce the risk of PD
Evatt et al. [136] A survey study in USA 199 PD (from DATATOP) Vitamin D insufficiency is very common in early PD patients
Miyake et al. [137] A case-control study in Japan 249/368 The study showed that vitamin D was not related to the PD
Knekt et al. [106] The Mini-Finland Health Survey in Finland Total 3173 and observed 50 PD cases The serum vitamin D concentrations were inversely correlated with the risk of PD
Kim et al. [94] A case-control study in Korea 85/231 Vitamin D receptor gene polymorphism was associated with the PD

Table 2.

The clinical intervention trial of vitamins and Parkinson's disease.

Vitamins Authors Patients Treatment Conclusions
Vitamin C Nagayama et al. [50] 67 elderly PD patients 200 mg ascorbic acid Ascorbic acid can improve levodopa absorption in elderly PD patients
Vitamin E Parkinson Study Group (DATATOP study) [76] 800 untreated and early PD patients Deprenyl 10 mg/d and/or tocopherol (vitamin E) 2000 IU/d There was no effect of tocopherol on PD
Parkinson Study Group (DATATOP study) [124] 800 untreated and early PD patients Deprenyl 10 mg/d and/or tocopherol (vitamin E) 2000 IU/d Alpha-tocopherol did not improve clinical features in patients with Parkinson's disease
Vatassery et al. (DATATOP study) [83] n = 18 (vitamin E group)/n = 5 (placebo group) Tocopherol (vitamin E) 2000 IU/d Treatment with vitamin E significantly increased the alpha-tocopherol concentrations in cerebrospinal fluid
Taghizadeh et al. [125] n = 30 (vitamin E group)/n = 30 (placebo group) 1000 mg omega-3 fatty acids plus 400 IU vitamin or placebo Omega-3 and vitamin E cosupplementation in PD patients improved UPDRS compared with the placebo
Vitamin D Suzuki et al. [113] n = 56 (vitamin D3 group)/n = 58 placebo group) Vitamin D3 1200 IU/d or placebo for 12 months Vitamin D3 prevented the deterioration of the PD and especially patients with FokI TT genes
Sato et al. [126] n = 43 (vitamin D group)/n = 43 (placebo group) 1α(OH)D3μg/d or placebo for 12 months 1alpha-hydroxyvitamin D3 supplements can reduce the risk of hip and other nonvertebral fractures in PD patients

Table 3.

The basic study of vitamins in PD.

Vitamin Authors Object of study Treatment Conclusions
Vitamin B3 (nicotinamide) Lu et al. [31] Rotenone-PC12 cells NMN (0.1 mM, 1.0 mM, 5 mM, and 10 mM) coculture Attenuated apoptosis and improved energy metabolism
Xu et al. [114] MPTP-C57BL/6 mice 500 mg/kg/day for 5 days i.p. Nicotinamide can alleviate MPTP-induced damage to dopaminergic neurons through antioxidant stress
Jia et al. [115] MPP(+)-SK-N-MC human neuroblastoma cells and alpha-synuclein transgenic Drosophila PD model Nicotinamide concentration (21, 51, 101, 301, and 501 mg/L and 3, 15, 30, and 60 mg/100 g) High doses of nicotinamide can reduce oxidative stress and improve mitochondrial function
Anderson et al. [116] MPTP-adult male C57Bl/6 mice Nicotinamide (125, 250, or 500 mg/kg i.p.) Recovered the striatal DA levels and SNc neurons after accepting nicotinamide
Vitamin C (ascorbic acid) Khan et al. [117] PD Drosophila model L-Ascorbic acid (AA, 11.35 × 10−5 M, 22.71 × 10−5 M, 45.42 × 10−5 M, and 68.13 × 10−5 M for 21 days) Except 11.35 × 10−5 M, other concentrations of AA attenuated the loss of climbing ability of PD model flies in a dose-dependent manner
Yan et al. [52] Mesencephalic precursors from the E12 rat Ascorbic acid (0.1 μM, 1 μM, 10 μM, 100 μM, and 1 mM) Ascorbic acid promoted the dopaminergic differentiation
Seitz et al. [49] Human neuroblastoma cell line SK-N-SH Short-term incubation (100–500 μM for 2 h) and long-term incubation (200 μM for 5 days) Ascorbic acid increased the DOPA production and tyrosine hydroxylase gene expression
Pardo et al. [47] Human neuroblastoma cell NB69 10−3 M ascorbic acid or 23 and 115 × 10−3 M alpha-tocopherol Ascorbic acid prevents the levodopa toxicity and quinone formation, but alpha-tocopherol did not
Sershen et al. [48] MPTP-BALB/cBy mice Ascorbic acid 100 mg/kg i.p. Ascorbic acid may protect against the MPTP neurotoxicity
Vitamin E Nakaso et al. [75] MPTP-C57BL/6 mice δ-Tocotrienol (100 μg/kg for 4 days, p.o.) δ-Tocotrienol administration inhibited the loss of dopaminergic neurons and improved the motor performance
Sharma and Nehru [70] Rotenone-Sprague-Dawley rats Vitamin E (100 IU/kg/day for 35 days i.m.) Vitamin E administration significantly improved locomotor activity and increased the dopamine level, GSH, and SOD
Ortiz et al. [118] MPTP-C57BL/6 mice Vitamin E (50 mg/kg/day p.o.) Vitamin E administration has decreased the COX-2 activity, LPO, and nitrite/nitrate level
Pasbakhsh et al. [119] 6-OHDA-rat Alpha-tocopherol acid succinate (24 IU/kg, i.m.) Vitamin E treatment can protect locus coeruleus neurons in the PD model
Roghani and Behzadi [69] 6-OHDA – Sprague-Dawley rats d-α-Tocopheryl acid succinate (24 I.U./kg, i.m.) Vitamin E treatment improved the rotational behaviour and prevented the reduction of tyrosine hydroxylase-immunoreactive cells
Vitamin D Lima et al. [120] 6-OHDA-Wistar rats 1,25-(OH)2D3 (1 μg/kg for 7 days or for 14 days, p.o.) Vitamin D can protect the dopaminergic neurons by its anti-inflammatory and antioxidant properties
Calvello et al. [121] MPTP-male C57BL/6 N mice Vitamin D (1 μg/kg for 10 days, i.g.) Vitamin D administration attenuates neuroinflammation and dopaminergic neurodegeneration
Li et al. [122] MPTP-C57BL/6 mice Calcitriol (0.2, 1, and 5 μg/kg/day for 7 days p.o.) Calcitriol can significantly attenuate the neurotoxicity induced by MPTP
Jang et al. [104] Rotenone-SH-SY5Y cells Calcitriol (0.0 μM, 0.63 μM, 1.25 μM, 2.5 μM, 5 μM, and 10 μM) 1,25-Dyhydroxyvitamin D3 can induce the autophagy to protect against the rotenone-induced neurotoxicity
Cass et al. [123] 6-OHDA-male Fischer-344 rats Calcitriol (0.3 or 1.0 μg/kg/day for 8 days, i.h.) Calcitriol can promote functional recovery of dopaminergic neurons and release of dopamine
Sanchez et al. [100] 6-OHDA-Sprague-Dawley rats 1,25(OH)(2)D(3) (1 μg/mL/kg/day for 7 days i.p.) 1,25(OH)(2)D(3) treatment increased the GDNF protein expression and partially restored TH expression
Kim et al. [102] 6-OHDA Sprague-Dawley rats and MPTP-C57BL/6 mice 1,25-(OH)2D3 (1 μg/mL at 1 mL/kg/day for 7 days, i.p.) 1,25-(OH)2D3 can inhibit the microglial activation and protect against nigrostriatal degeneration

LPO: lipid peroxides; COX-2: ciclooxigenase-2; TH: tyrosine hydroxylase; i.p.: intraperitoneal; i.m.: intramuscular; i.g.: intragastrical; i.h.: hypodermic injection; p.o.: peros.

Acknowledgments

This work was supported by the crosswise project from the Ministry of Science and Technology of Jilin Province (Grant No. 3R2168713428) and the Natural Science Foundation of Jilin Province (Grant No. 20180101154JC).

Conflicts of Interest

The authors have no conflicts of interest to declare.

Authors' Contributions

Xiuzhen Zhao and Ming Zhang equally contributed to this study.

References

  • 1.Gibb W. R., Lees A. J. The relevance of the Lewy body to the pathogenesis of idiopathic Parkinson’s disease. Journal of Neurology, Neurosurgery, and Psychiatry. 1988;51(6):745–752. doi: 10.1136/jnnp.51.6.745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Khoo T. K., Yarnall A. J., Duncan G. W., et al. The spectrum of nonmotor symptoms in early Parkinson disease. Neurology. 2013;80(3):276–281. doi: 10.1212/WNL.0b013e31827deb74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Postuma R. B., Aarsland D., Barone P., et al. Identifying prodromal Parkinson’s disease: pre-motor disorders in Parkinson’s disease. Movement Disorders. 2012;27(5):617–626. doi: 10.1002/mds.24996. [DOI] [PubMed] [Google Scholar]
  • 4.Goswami P., Joshi N., Singh S. Neurodegenerative signaling factors and mechanisms in Parkinson’s pathology. Toxicology In Vitro. 2017;43:104–112. doi: 10.1016/j.tiv.2017.06.008. [DOI] [PubMed] [Google Scholar]
  • 5.Spillantini M. G., Schmidt M. L., Lee V. M. Y., Trojanowski J. Q., Jakes R., Goedert M. α-Synuclein in Lewy bodies. Nature. 1997;388(6645):839–840. doi: 10.1038/42166. [DOI] [PubMed] [Google Scholar]
  • 6.Hattingen E., Magerkurth J., Pilatus U., et al. Phosphorus and proton magnetic resonance spectroscopy demonstrates mitochondrial dysfunction in early and advanced Parkinson’s disease. Brain. 2009;132(12):3285–3297. doi: 10.1093/brain/awp293. [DOI] [PubMed] [Google Scholar]
  • 7.Parker W. D., Jr, Parks J. K., Swerdlow R. H. Complex I deficiency in Parkinson’s disease frontal cortex. Brain Research. 2008;1189:215–218. doi: 10.1016/j.brainres.2007.10.061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Sánchez-Hernández D., Anderson G. H., Poon A. N., et al. Maternal fat-soluble vitamins, brain development, and regulation of feeding behavior: an overview of research. Nutrition Research. 2016;36(10):1045–1054. doi: 10.1016/j.nutres.2016.09.009. [DOI] [PubMed] [Google Scholar]
  • 9.Chawla J., Kvarnberg D. Hydrosoluble vitamins. Handbook of Clinical Neurology. 2014;120:891–914. doi: 10.1016/B978-0-7020-4087-0.00059-0. [DOI] [PubMed] [Google Scholar]
  • 10.Zhang S. M., Hernan M. A., Chen H., Spiegelman D., Willett W. C., Ascherio A. Intakes of vitamins E and C, carotenoids, vitamin supplements, and PD risk. Neurology. 2002;59(8):1161–1169. doi: 10.1212/01.WNL.0000028688.75881.12. [DOI] [PubMed] [Google Scholar]
  • 11.Sies H., Berndt C., Jones D. P. Oxidative stress. Annual Review of Biochemistry. 2017;86(1):715–748. doi: 10.1146/annurev-biochem-061516-045037. [DOI] [PubMed] [Google Scholar]
  • 12.Kalia L. V., Lang A. E. Parkinson’s disease. The Lancet. 2015;386(9996):896–912. doi: 10.1016/S0140-6736(14)61393-3. [DOI] [PubMed] [Google Scholar]
  • 13.Blesa J., Trigo-Damas I., Quiroga-Varela A., Jackson-Lewis V. R. Oxidative stress and Parkinson’s disease. Frontiers in Neuroanatomy. 2015;9:p. 91. doi: 10.3389/fnana.2015.00091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Murphy M. P. How mitochondria produce reactive oxygen species. The Biochemical Journal. 2009;417(1):1–13. doi: 10.1042/BJ20081386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Schapira A. H., Cooper J. M., Dexter D., Jenner P., Clark J. B., Marsden C. D. Mitochondrial complex I deficiency in Parkinson’s disease. The Lancet. 1989;333(8649):p. 1269. doi: 10.1016/s0140-6736(89)92366-0. [DOI] [PubMed] [Google Scholar]
  • 16.Sian J., Dexter D. T., Lees A. J., et al. Alterations in glutathione levels in Parkinson’s disease and other neurodegenerative disorders affecting basal ganglia. Annals of Neurology. 1994;36(3):348–355. doi: 10.1002/ana.410360305. [DOI] [PubMed] [Google Scholar]
  • 17.Gluck M. R., Zeevalk G. D. Inhibition of brain mitochondrial respiration by dopamine and its metabolites: implications for Parkinson’s disease and catecholamine-associated diseases. Journal of Neurochemistry. 2004;91(4):788–795. doi: 10.1111/j.1471-4159.2004.02747.x. [DOI] [PubMed] [Google Scholar]
  • 18.Hare D. J., Double K. L. Iron and dopamine: a toxic couple. Brain. 2016;139(4):1026–1035. doi: 10.1093/brain/aww022. [DOI] [PubMed] [Google Scholar]
  • 19.Sauberlich H. E. Implications of nutritional status on human biochemistry, physiology, and health. Clinical Biochemistry. 1984;17(2):132–142. doi: 10.1016/S0009-9120(84)90344-8. [DOI] [PubMed] [Google Scholar]
  • 20.Mikkelsen K., Stojanovska L., Tangalakis K., Bosevski M., Apostolopoulos V. Cognitive decline: a vitamin B perspective. Maturitas. 2016;93:108–113. doi: 10.1016/j.maturitas.2016.08.001. [DOI] [PubMed] [Google Scholar]
  • 21.Gehring W. Nicotinic acid/niacinamide and the skin. Journal of Cosmetic Dermatology. 2004;3(2):88–93. doi: 10.1111/j.1473-2130.2004.00115.x. [DOI] [PubMed] [Google Scholar]
  • 22.Surjana D., Damian D. L. Nicotinamide in dermatology and photoprotection. Skinmed. 2011;9(6):360–365. [PubMed] [Google Scholar]
  • 23.Williams A., Ramsden D. Nicotinamide: a double edged sword. Parkinsonism & Related Disorders. 2005;11(7):413–420. doi: 10.1016/j.parkreldis.2005.05.011. [DOI] [PubMed] [Google Scholar]
  • 24.Fukushima T. Niacin metabolism and Parkinson’s disease. Environmental Health and Preventive Medicine. 2005;10(1):3–8. doi: 10.1265/ehpm.10.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Aoyama K., Matsubara K., Kondo M., et al. Nicotinamide-N-methyltransferase is higher in the lumbar cerebrospinal fluid of patients with Parkinson’s disease. Neuroscience Letters. 2001;298(1):78–80. doi: 10.1016/S0304-3940(00)01723-7. [DOI] [PubMed] [Google Scholar]
  • 26.Griffin S. M., Pickard M. R., Orme R. P., Hawkins C. P., Fricker R. A. Nicotinamide promotes neuronal differentiation of mouse embryonic stem cells in vitro. Neuroreport. 2013;24(18):1041–1046. doi: 10.1097/WNR.0000000000000071. [DOI] [PubMed] [Google Scholar]
  • 27.Imai S. Nicotinamide phosphoribosyltransferase (Nampt): a link between NAD biology, metabolism, and diseases. Current Pharmaceutical Design. 2009;15(1):20–28. doi: 10.2174/138161209787185814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Pearl S. M., Antion M. D., Stanwood G. D., Jaumotte J. D., Kapatos G., Zigmond M. J. Effects of NADH on dopamine release in rat striatum. Synapse. 2000;36(2):95–101. doi: 10.1002/(SICI)1098-2396(200005)36:2&#x0003c;95::AID-SYN2&#x0003e;3.0.CO;2-U. [DOI] [PubMed] [Google Scholar]
  • 29.Mizuno Y., Ohta S., Tanaka M., et al. Deficiencies in complex I subunits of the respiratory chain in Parkinson’s disease. Biochemical and Biophysical Research Communications. 1989;163(3):1450–1455. doi: 10.1016/0006-291X(89)91141-8. [DOI] [PubMed] [Google Scholar]
  • 30.Nicklas W. J., Vyas I., Heikkila R. E. Inhibition of NADH-linked oxidation in brain mitochondria by 1-methyl-4-phenyl-pyridine, a metabolite of the neurotoxin, 1-methyl-4-phenyl-1, 2, 5, 6-tetrahydropyridine. Life Sciences. 1985;36(26):2503–2508. doi: 10.1016/0024-3205(85)90146-8. [DOI] [PubMed] [Google Scholar]
  • 31.Lu L., Tang L., Wei W., et al. Nicotinamide mononucleotide improves energy activity and survival rate in an in vitro model of Parkinson’s disease. Experimental and Therapeutic Medicine. 2014;8(3):943–950. doi: 10.3892/etm.2014.1842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Yang L., Ma X., He Y., et al. Sirtuin 5: a review of structure, known inhibitors and clues for developing new inhibitors. Science China Life Sciences. 2017;60(3):249–256. doi: 10.1007/s11427-016-0060-7. [DOI] [PubMed] [Google Scholar]
  • 33.Liu L., Peritore C., Ginsberg J., Shih J., Arun S., Donmez G. Protective role of SIRT5 against motor deficit and dopaminergic degeneration in MPTP-induced mice model of Parkinson’s disease. Behavioural Brain Research. 2015;281:215–221. doi: 10.1016/j.bbr.2014.12.035. [DOI] [PubMed] [Google Scholar]
  • 34.Fall P. A., Fredrikson M., Axelson O., Granérus A. K. Nutritional and occupational factors influencing the risk of Parkinson’s disease: a case-control study in southeastern Sweden. Movement Disorders. 1999;14(1):28–37. doi: 10.1002/1531-8257(199901)14:1&#x0003c;28::AID-MDS1007&#x0003e;3.0.CO;2-O. [DOI] [PubMed] [Google Scholar]
  • 35.Hellenbrand W., Boeing H., Robra B. P., et al. Diet and Parkinson’s disease. II: a possible role for the past intake of specific nutrients. Results from a self-administered food-frequency questionnaire in a case-control study. Neurology. 1996;47(3):644–650. doi: 10.1212/WNL.47.3.644. [DOI] [PubMed] [Google Scholar]
  • 36.Alisky J. M. Niacin improved rigidity and bradykinesia in a Parkinson’s disease patient but also caused unacceptable nightmares and skin rash—a case report. Nutritional Neuroscience. 2005;8(5-6):327–329. doi: 10.1080/10284150500484638. [DOI] [PubMed] [Google Scholar]
  • 37.Abbott R. D., Webster Ross G., White L. R., et al. Environmental, life-style, and physical precursors of clinical Parkinson’s disease: recent findings from the Honolulu-Asia Aging Study. Journal of Neurology. 2003;250(Supplement 3):iii30–iii39. doi: 10.1007/s00415-003-1306-7. [DOI] [PubMed] [Google Scholar]
  • 38.Johnson C. C., Gorell J. M., Rybicki B. A., Sanders K., Peterson E. L. Adult nutrient intake as a risk factor for Parkinson’s disease. International Journal of Epidemiology. 1999;28(6):1102–1109. doi: 10.1093/ije/28.6.1102. [DOI] [PubMed] [Google Scholar]
  • 39.Oudemans-van Straaten H. M., Spoelstra-de Man A. M. E., de Waard M. C. Vitamin C revisited. Critical Care. 2014;18(4):p. 460. doi: 10.1186/s13054-014-0460-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Chatterjee I. B., Majumder A. K., Nandi B. K., Subramanian N. Synthesis and some major functions of vitamin C in animals. Annals of the New York Academy of Sciences. 1975;258:24–47. doi: 10.1111/j.1749-6632.1975.tb29266.x. [DOI] [PubMed] [Google Scholar]
  • 41.Grosso G., Bei R., Mistretta A., et al. Effects of vitamin C on health: a review of evidence. Frontiers in Bioscience. 2013;18:1017–1029. doi: 10.2741/4160. [DOI] [PubMed] [Google Scholar]
  • 42.Belluzzi E., Bisaglia M., Lazzarini E., Tabares L. C., Beltramini M., Bubacco L. Human SOD2 modification by dopamine quinones affects enzymatic activity by promoting its aggregation: possible implications for Parkinson’s disease. PLoS One. 2012;7(6, article e38026) doi: 10.1371/journal.pone.0038026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Mefford I. N., Oke A. F., Adams R. N. Regional distribution of ascorbate in human brain. Brain Research. 1981;212(1):223–226. doi: 10.1016/0006-8993(81)90056-1. [DOI] [PubMed] [Google Scholar]
  • 44.Milby K., Oke A., Adams R. N. Detailed mapping of ascorbate distribution in rat brain. Neuroscience Letters. 1982;28(1):15–20. doi: 10.1016/0304-3940(82)90201-4. [DOI] [PubMed] [Google Scholar]
  • 45.Hansen S. N., Tveden-Nyborg P., Lykkesfeldt J. Does vitamin C deficiency affect cognitive development and function? Nutrients. 2014;6(9):3818–3846. doi: 10.3390/nu6093818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hosoya K., Nakamura G., Akanuma S. I., Tomi M., Tachikawa M. Dehydroascorbic acid uptake and intracellular ascorbic acid accumulation in cultured Müller glial cells (TR-MUL) Neurochemistry International. 2008;52(7):1351–1357. doi: 10.1016/j.neuint.2008.02.001. [DOI] [PubMed] [Google Scholar]
  • 47.Pardo B., Mena M. A., Fahn S., de Yébenes J. G. Ascorbic acid protects against levodopa-induced neurotoxicity on a catecholamine-rich human neuroblastoma cell line. Movement Disorders. 1993;8(3):278–284. doi: 10.1002/mds.870080305. [DOI] [PubMed] [Google Scholar]
  • 48.Sershen H., Reith M. E. A., Hashim A., Lajtha A. Protection against 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine neurotoxicity by the antioxidant ascorbic acid. Neuropharmacology. 1985;24(12):1257–1259. doi: 10.1016/0028-3908(85)90163-7. [DOI] [PubMed] [Google Scholar]
  • 49.Seitz G., Gebhardt S., Beck J. F., et al. Ascorbic acid stimulates DOPA synthesis and tyrosine hydroxylase gene expression in the human neuroblastoma cell line SK-N-SH. Neuroscience Letters. 1998;244(1):33–36. doi: 10.1016/S0304-3940(98)00129-3. [DOI] [PubMed] [Google Scholar]
  • 50.Nagayama H., Hamamoto M., Ueda M., Nito C., Yamaguchi H., Katayama Y. The effect of ascorbic acid on the pharmacokinetics of levodopa in elderly patients with Parkinson disease. Clinical Neuropharmacology. 2004;27(6):270–273. doi: 10.1097/01.wnf.0000150865.21759.bc. [DOI] [PubMed] [Google Scholar]
  • 51.Sacks W., Simpson G. M. Letter: ascorbic acid in levodopa therapy. Lancet. 1975;1(7905):p. 527. [PubMed] [Google Scholar]
  • 52.Yan J., Studer L., McKay R. D. G. Ascorbic acid increases the yield of dopaminergic neurons derived from basic fibroblast growth factor expanded mesencephalic precursors. Journal of Neurochemistry. 2001;76(1):307–311. doi: 10.1046/j.1471-4159.2001.00073.x. [DOI] [PubMed] [Google Scholar]
  • 53.Lee J. Y., Chang M. Y., Park C. H., et al. Ascorbate-induced differentiation of embryonic cortical precursors into neurons and astrocytes. Journal of Neuroscience Research. 2003;73(2):156–165. doi: 10.1002/jnr.10647. [DOI] [PubMed] [Google Scholar]
  • 54.He X. B., Kim M., Kim S. Y., et al. Vitamin C facilitates dopamine neuron differentiation in fetal midbrain through TET1- and JMJD3-dependent epigenetic control manner. Stem Cells. 2015;33(4):1320–1332. doi: 10.1002/stem.1932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Wulansari N., Kim E. H., Sulistio Y. A., Rhee Y. H., Song J. J., Lee S. H. Vitamin C-induced epigenetic modifications in donor NSCs establish midbrain marker expressions critical for cell-based therapy in Parkinson’s disease. Stem Cell Reports. 2017;9(4):1192–1206. doi: 10.1016/j.stemcr.2017.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Férnandez-Calle P., Jiménez-Jiménez F. J., Molina J. A., et al. Serum levels of ascorbic acid (vitamin C) in patients with Parkinson’s disease. Journal of the Neurological Sciences. 1993;118(1):25–28. doi: 10.1016/0022-510X(93)90240-Y. [DOI] [PubMed] [Google Scholar]
  • 57.Sudha K., Rao A. V., Rao S., Rao A. Free radical toxicity and antioxidants in Parkinson’s disease. Neurology India. 2003;51(1):60–62. [PubMed] [Google Scholar]
  • 58.Ide K., Yamada H., Umegaki K., et al. Lymphocyte vitamin C levels as potential biomarker for progression of Parkinson’s disease. Nutrition. 2015;31(2):406–408. doi: 10.1016/j.nut.2014.08.001. [DOI] [PubMed] [Google Scholar]
  • 59.Hughes K. C., Gao X., Kim I. Y., et al. Intake of antioxidant vitamins and risk of Parkinson’s disease. Movement Disorders. 2016;31(12):1909–1914. doi: 10.1002/mds.26819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Miyake Y., Fukushima W., Tanaka K., et al. Dietary intake of antioxidant vitamins and risk of Parkinson’s disease: a case-control study in Japan. European Journal of Neurology. 2011;18(1):106–113. doi: 10.1111/j.1468-1331.2010.03088.x. [DOI] [PubMed] [Google Scholar]
  • 61.Yang F., Wolk A., Håkansson N., Pedersen N. L., Wirdefeldt K. Dietary antioxidants and risk of Parkinson’s disease in two population-based cohorts. Movement Disorders. 2017;32(11):1631–1636. doi: 10.1002/mds.27120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Colombo M. L. An update on vitamin E, tocopherol and tocotrienol-perspectives. Molecules. 2010;15(4):2103–2113. doi: 10.3390/molecules15042103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Beharka A., Redican S., Leka L., Meydani S. N. [22] Vitamin E status and immune function. Methods in Enzymology. 1997;282:247–263. doi: 10.1016/S0076-6879(97)82112-X. [DOI] [PubMed] [Google Scholar]
  • 64.Cesari M., Pahor M., Bartali B., et al. Antioxidants and physical performance in elderly persons: the Invecchiare in Chianti (InCHIANTI) study. The American Journal of Clinical Nutrition. 2004;79(2):289–294. doi: 10.1093/ajcn/79.2.289. [DOI] [PubMed] [Google Scholar]
  • 65.Cherubini A., Martin A., Andres-Lacueva C., et al. Vitamin E levels, cognitive impairment and dementia in older persons: the InCHIANTI study. Neurobiology of Aging. 2005;26(7):987–994. doi: 10.1016/j.neurobiolaging.2004.09.002. [DOI] [PubMed] [Google Scholar]
  • 66.Clarke M. W., Burnett J. R., Croft K. D. Vitamin E in human health and disease. Critical Reviews in Clinical Laboratory Sciences. 2008;45(5):417–450. doi: 10.1080/10408360802118625. [DOI] [PubMed] [Google Scholar]
  • 67.Aparicio J. M., Bélanger-Quintana A., Suárez L., et al. Ataxia with isolated vitamin E deficiency: case report and review of the literature. Journal of Pediatric Gastroenterology and Nutrition. 2001;33(2):206–210. doi: 10.1097/00005176-200108000-00022. [DOI] [PubMed] [Google Scholar]
  • 68.Cadet J. L., Katz M., Jackson-Lewis V., Fahn S. Vitamin E attenuates the toxic effects of intrastriatal injection of 6-hydroxydopamine (6-OHDA) in rats: behavioral and biochemical evidence. Brain Research. 1989;476(1):10–15. doi: 10.1016/0006-8993(89)91530-8. [DOI] [PubMed] [Google Scholar]
  • 69.Roghani M., Behzadi G. Neuroprotective effect of vitamin E on the early model of Parkinson’s disease in rat: behavioral and histochemical evidence. Brain Research. 2001;892(1):211–217. doi: 10.1016/S0006-8993(00)03296-0. [DOI] [PubMed] [Google Scholar]
  • 70.Sharma N., Nehru B. Beneficial effect of vitamin E in rotenone induced model of PD: behavioural, neurochemical and biochemical study. Experimental Neurobiology. 2013;22(3):214–223. doi: 10.5607/en.2013.22.3.214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Perry T. L., Yong V. W., Hansen S., et al. α-Tocopherol and β-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. doi: 10.1016/0022-510X(87)90106-7. [DOI] [PubMed] [Google Scholar]
  • 72.Heim C., Kolasiewicz W., Kurz T., Sontag K. H. Behavioral alterations after unilateral 6-hydroxydopamine lesions of the striatum. Effect of alpha-tocopherol. Polish Journal of Pharmacology. 2001;53(5):435–448. [PubMed] [Google Scholar]
  • 73.Comitato R., Nesaretnam K., Leoni G., et al. A novel mechanism of natural vitamin E tocotrienol activity: involvement of ERβ signal transduction. American Journal of Physiology. Endocrinology and Metabolism. 2009;297(2):E427–E437. doi: 10.1152/ajpendo.00187.2009. [DOI] [PubMed] [Google Scholar]
  • 74.Nakaso K., Tajima N., Horikoshi Y., et al. The estrogen receptor β-PI3K/Akt pathway mediates the cytoprotective effects of tocotrienol in a cellular Parkinson’s disease model. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease. 2014;1842(9):1303–1312. doi: 10.1016/j.bbadis.2014.04.008. [DOI] [PubMed] [Google Scholar]
  • 75.Nakaso K., Horikoshi Y., Takahashi T., et al. Estrogen receptor-mediated effect of δ-tocotrienol prevents neurotoxicity and motor deficit in the MPTP mouse model of Parkinson’s disease. Neuroscience Letters. 2016;610:117–122. doi: 10.1016/j.neulet.2015.10.062. [DOI] [PubMed] [Google Scholar]
  • 76.DATATOP: a multicenter controlled clinical trial in early Parkinson’s disease: Parkinson Study Group. Archives of Neurology. 1989;46(10):1052–1060. doi: 10.1001/archneur.1989.00520460028009. [DOI] [PubMed] [Google Scholar]
  • 77.Logroscino G., Marder K., Cote L., Tang M. X., Shea S., Mayeux R. Dietary lipids and antioxidants in Parkinson’s disease: a population-based, case-control study. Annals of Neurology. 1996;39(1):89–94. doi: 10.1002/ana.410390113. [DOI] [PubMed] [Google Scholar]
  • 78.de Rijk M. C., Breteler M. M., den Breeijen J., et al. Dietary antioxidants and Parkinson disease. The Rotterdam Study. Archives of Neurology. 1997;54(6):762–765. doi: 10.1001/archneur.1997.00550180070015. [DOI] [PubMed] [Google Scholar]
  • 79.Fahn S. A pilot trial of high-dose alpha-tocopherol and ascorbate in early Parkinson’s disease. Annals of Neurology. 1992;32(S1):S128–S132. doi: 10.1002/ana.410320722. [DOI] [PubMed] [Google Scholar]
  • 80.Fernandez-Calle P., Molina J. A., Jimenez-Jimenez F. J., et al. Serum levels of alpha-tocopherol (vitamin E) in Parkinson’s disease. Neurology. 1992;42(5):1064–1066. doi: 10.1212/WNL.42.5.1064. [DOI] [PubMed] [Google Scholar]
  • 81.Dexter D. T., Jenner P., Ward R. J., et al. α-Tocopherol levels in brain are not altered in Parkinson’s disease. Annals of Neurology. 1992;32(4):591–593. doi: 10.1002/ana.410320420. [DOI] [PubMed] [Google Scholar]
  • 82.Molina J. A., de Bustos F., Jiménez-Jiménez F. J., et al. Cerebrospinal fluid levels of alpha-tocopherol (vitamin E) in Parkinson’s disease. Journal of Neural Transmission. 1997;104(11-12):1287–1293. doi: 10.1007/BF01294729. [DOI] [PubMed] [Google Scholar]
  • 83.Vatassery G. T., Fahn S., Kuskowski M. A. Alpha tocopherol in CSF of subjects taking high-dose vitamin E in the DATATOP study: Parkinson Study Group. Neurology. 1998;50(6):1900–1902. doi: 10.1212/WNL.50.6.1900. [DOI] [PubMed] [Google Scholar]
  • 84.Kulda V. Vitamin D metabolism. Vnitřní Lékařství. 2012;58(5):400–404. [PubMed] [Google Scholar]
  • 85.Christakos S., Dhawan P., Verstuyf A., Verlinden L., Carmeliet G. Vitamin D: metabolism, molecular mechanism of action, and pleiotropic effects. Physiological Reviews. 2016;96(1):365–408. doi: 10.1152/physrev.00014.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Haussler M. R., Whitfield G. K., Kaneko I., et al. Molecular mechanisms of vitamin D action. Calcified Tissue International. 2013;92(2):77–98. doi: 10.1007/s00223-012-9619-0. [DOI] [PubMed] [Google Scholar]
  • 87.Carlberg C., Molnar F. Vitamin D receptor signaling and its therapeutic implications: genome-wide and structural view. Canadian Journal of Physiology and Pharmacology. 2015;93(5):311–318. doi: 10.1139/cjpp-2014-0383. [DOI] [PubMed] [Google Scholar]
  • 88.Samuel S., Sitrin M. D. Vitamin D’s role in cell proliferation and differentiation. Nutrition Reviews. 2008;66(10) Supplement 2:S116–S124. doi: 10.1111/j.1753-4887.2008.00094.x. [DOI] [PubMed] [Google Scholar]
  • 89.Myszka M., Klinger M. The immunomodulatory role of vitamin D. Postȩpy Higieny i Medycyny Doświadczalnej. 2014;68:865–878. doi: 10.5604/17322693.1110168. [DOI] [PubMed] [Google Scholar]
  • 90.Kono K., Fujii H., Nakai K., et al. Anti-oxidative effect of vitamin D analog on incipient vascular lesion in non-obese type 2 diabetic rats. American Journal of Nephrology. 2013;37(2):167–174. doi: 10.1159/000346808. [DOI] [PubMed] [Google Scholar]
  • 91.Sahota O. Understanding vitamin D deficiency. Age and Ageing. 2014;43(5):589–591. doi: 10.1093/ageing/afu104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Newmark H. L., Newmark J. Vitamin D and Parkinson’s disease—a hypothesis. Movement Disorders. 2007;22(4):461–468. doi: 10.1002/mds.21317. [DOI] [PubMed] [Google Scholar]
  • 93.Köstner K., Denzer N., Müller C. S., Klein R., Tilgen W., Reichrath J. The relevance of vitamin D receptor (VDR) gene polymorphisms for cancer: a review of the literature. Anticancer Research. 2009;29(9):3511–3536. [PubMed] [Google Scholar]
  • 94.Kim J. S., Kim Y. I., Song C., et al. Association of vitamin D receptor gene polymorphism and Parkinson’s disease in Koreans. Journal of Korean Medical Science. 2005;20(3):495–498. doi: 10.3346/jkms.2005.20.3.495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Li C., Qi H., Wei S., et al. Vitamin D receptor gene polymorphisms and the risk of Parkinson’s disease. Neurological Sciences. 2015;36(2):247–255. doi: 10.1007/s10072-014-1928-9. [DOI] [PubMed] [Google Scholar]
  • 96.Suzuki M., Yoshioka M., Hashimoto M., et al. 25-hydroxyvitamin D, vitamin D receptor gene polymorphisms, and severity of Parkinson’s disease. Movement Disorders. 2012;27(2):264–271. doi: 10.1002/mds.24016. [DOI] [PubMed] [Google Scholar]
  • 97.Gatto N. M., Paul K. C., Sinsheimer J. S., et al. Vitamin D receptor gene polymorphisms and cognitive decline in Parkinson’s disease. Journal of the Neurological Sciences. 2016;370:100–106. doi: 10.1016/j.jns.2016.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Burne T. H. J., McGrath J. J., Eyles D. W., Mackay-Sim A. Behavioural characterization of vitamin D receptor knockout mice. Behavioural Brain Research. 2005;157(2):299–308. doi: 10.1016/j.bbr.2004.07.008. [DOI] [PubMed] [Google Scholar]
  • 99.Campos F. L., Cristovão A. C., Rocha S. M., Fonseca C. P., Baltazar G. GDNF contributes to oestrogen-mediated protection of midbrain dopaminergic neurones. Journal of Neuroendocrinology. 2012;24(11):1386–1397. doi: 10.1111/j.1365-2826.2012.02348.x. [DOI] [PubMed] [Google Scholar]
  • 100.Sanchez B., Relova J. L., Gallego R., Ben-Batalla I., Perez-Fernandez R. 1, 25-Dihydroxyvitamin D3 administration to 6-hydroxydopamine-lesioned rats increases glial cell line-derived neurotrophic factor and partially restores tyrosine hydroxylase expression in substantia nigra and striatum. Journal of Neuroscience Research. 2009;87(3):723–732. doi: 10.1002/jnr.21878. [DOI] [PubMed] [Google Scholar]
  • 101.Sanchez B., Lopez-Martin E., Segura C., Labandeira-Garcia J. L., Perez-Fernandez R. 1, 25-Dihydroxyvitamin D3 increases striatal GDNF mRNA and protein expression in adult rats. Molecular Brain Research. 2002;108(1-2):143–146. doi: 10.1016/S0169-328X(02)00545-4. [DOI] [PubMed] [Google Scholar]
  • 102.Kim J. S., Ryu S. Y., Yun I., et al. 1α,25-Dihydroxyvitamin D3 protects dopaminergic neurons in rodent models of Parkinson’s disease through inhibition of microglial activation. Journal of Clinical Neurology. 2006;2(4):252–257. doi: 10.3988/jcn.2006.2.4.252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Wang J. Y., Wu J. N., Cherng T. L., et al. Vitamin D3 attenuates 6-hydroxydopamine-induced neurotoxicity in rats. Brain Research. 2001;904(1):67–75. doi: 10.1016/S0006-8993(01)02450-7. [DOI] [PubMed] [Google Scholar]
  • 104.Jang W., Kim H. J., Li H., et al. 1,25-Dyhydroxyvitamin D3 attenuates rotenone-induced neurotoxicity in SH-SY5Y cells through induction of autophagy. Biochemical and Biophysical Research Communications. 2014;451(1):142–147. doi: 10.1016/j.bbrc.2014.07.081. [DOI] [PubMed] [Google Scholar]
  • 105.Yoon J. H., Park D. K., Yong S. W., Hong J. M. Vitamin D deficiency and its relationship with endothelial dysfunction in patients with early Parkinson’s disease. Journal of Neural Transmission (Vienna) 2015;122(12):1685–1691. doi: 10.1007/s00702-015-1452-y. [DOI] [PubMed] [Google Scholar]
  • 106.Knekt P., Kilkkinen A., Rissanen H., Marniemi J., Sääksjärvi K., Heliövaara M. Serum vitamin D and the risk of Parkinson disease. Archives of Neurology. 2010;67(7):808–811. doi: 10.1001/archneurol.2010.120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Evatt M. L., Delong M. R., Khazai N., Rosen A., Triche S., Tangpricha V. Prevalence of vitamin d insufficiency in patients with Parkinson disease and Alzheimer disease. Archives of Neurology. 2008;65(10):1348–1352. doi: 10.1001/archneur.65.10.1348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Kenborg L., Lassen C. F., Ritz B., et al. Outdoor work and risk for Parkinson’s disease: a population-based case-control study. Occupational and Environmental Medicine. 2011;68(4):273–278. doi: 10.1136/oem.2010.057448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Kravietz A., Kab S., Wald L., et al. Association of UV radiation with Parkinson disease incidence: a nationwide French ecologic study. Environmental Research. 2017;154:50–56. doi: 10.1016/j.envres.2016.12.008. [DOI] [PubMed] [Google Scholar]
  • 110.Wang J., Yang D., Yu Y., Shao G., Wang Q. Vitamin D and sunlight exposure in newly-diagnosed Parkinson’s disease. Nutrients. 2016;8(3):p. 142. doi: 10.3390/nu8030142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Liu Y., Zhang B. S. Serum 25-hydroxyvitamin D predicts severity in Parkinson’s disease patients. Neurological Sciences. 2014;35(1):67–71. doi: 10.1007/s10072-013-1539-x. [DOI] [PubMed] [Google Scholar]
  • 112.Sleeman I., Aspray T., Lawson R., et al. The role of vitamin D in disease progression in early Parkinson’s disease. Journal of Parkinson's Disease. 2017;7(4):669–675. doi: 10.3233/JPD-171122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Suzuki M., Yoshioka M., Hashimoto M., et al. Randomized, double-blind, placebo-controlled trial of vitamin D supplementation in Parkinson disease. The American Journal of Clinical Nutrition. 2013;97(5):1004–1013. doi: 10.3945/ajcn.112.051664. [DOI] [PubMed] [Google Scholar]
  • 114.Xu J., Xu S. Q., Liang J., Lu Y., Luo J. H., Jin J. H. Protective effect of nicotinamide in a mouse Parkinson’s disease model. Zhejiang Da Xue Xue Bao. Yi Xue Ban. 2012;41(2):146–152. [PubMed] [Google Scholar]
  • 115.Jia H., Li X., Gao H., et al. High doses of nicotinamide prevent oxidative mitochondrial dysfunction in a cellular model and improve motor deficit in a Drosophila model of Parkinson’s disease. Journal of Neuroscience Research. 2008;86(9):2083–2090. doi: 10.1002/jnr.21650. [DOI] [PubMed] [Google Scholar]
  • 116.Anderson D. W., Bradbury K. A., Schneider J. S. Broad neuroprotective profile of nicotinamide in different mouse models of MPTP-induced parkinsonism. The European Journal of Neuroscience. 2008;28(3):610–617. doi: 10.1111/j.1460-9568.2008.06356.x. [DOI] [PubMed] [Google Scholar]
  • 117.Khan S., Jyoti S., Naz F., et al. Effect of L-ascorbic acid on the climbing ability and protein levels in the brain of Drosophila model of Parkinson’s disease. The International Journal of Neuroscience. 2012;122(12):704–709. doi: 10.3109/00207454.2012.709893. [DOI] [PubMed] [Google Scholar]
  • 118.Ortiz G. G., Pacheco-Moisés F. P., Gómez-Rodríguez V. M., González-Renovato E. D., Torres-Sánchez E. D., Ramírez-Anguiano A. C. Fish oil, melatonin and vitamin E attenuates midbrain cyclooxygenase-2 activity and oxidative stress after the administration of 1-methyl-4-phenyl-1, 2, 3, 6- tetrahydropyridine. Metabolic Brain Disease. 2013;28(4):705–709. doi: 10.1007/s11011-013-9416-0. [DOI] [PubMed] [Google Scholar]
  • 119.Pasbakhsh P., Omidi N., Mehrannia K., et al. The protective effect of vitamin E on locus coeruleus in early model of Parkinson’s disease in rat: immunoreactivity evidence. Iranian Biomedical Journal. 2008;12(4):217–222. [PubMed] [Google Scholar]
  • 120.Lima L. A. R., Lopes M. J. P., Costa R. O., et al. Vitamin D protects dopaminergic neurons against neuroinflammation and oxidative stress in hemiparkinsonian rats. Journal of Neuroinflammation. 2018;15(1):p. 249. doi: 10.1186/s12974-018-1266-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Calvello R., Cianciulli A., Nicolardi G., et al. Vitamin D treatment attenuates neuroinflammation and dopaminergic neurodegeneration in an animal model of Parkinson’s disease, shifting M1 to M2 microglia responses. Journal of Neuroimmune Pharmacology. 2017;12(2):327–339. doi: 10.1007/s11481-016-9720-7. [DOI] [PubMed] [Google Scholar]
  • 122.Li H., Jang W., Kim H. J., et al. Biochemical protective effect of 1,25-dihydroxyvitamin D3 through autophagy induction in the MPTP mouse model of Parkinson’s disease. Neuroreport. 2015;26(12):669–674. doi: 10.1097/WNR.0000000000000401. [DOI] [PubMed] [Google Scholar]
  • 123.Cass W. A., Peters L. E., Fletcher A. M., Yurek D. M. Calcitriol promotes augmented dopamine release in the lesioned striatum of 6-hydroxydopamine treated rats. Neurochemical Research. 2014;39(8):1467–1476. doi: 10.1007/s11064-014-1331-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.The Parkinson Study Group. Effects of tocopherol and deprenyl on the progression of disability in early Parkinson’s disease. The New England Journal of Medicine. 1993;328(3):176–183. doi: 10.1056/nejm199301213280305. [DOI] [PubMed] [Google Scholar]
  • 125.Taghizadeh M., Tamtaji O. R., Dadgostar E., et al. The effects of omega-3 fatty acids and vitamin E co-supplementation on clinical and metabolic status in patients with Parkinson’s disease: a randomized, double-blind, placebo-controlled trial. Neurochemistry International. 2017;108:183–189. doi: 10.1016/j.neuint.2017.03.014. [DOI] [PubMed] [Google Scholar]
  • 126.Sato Y., Manabe S., Kuno H., Oizumi K. Amelioration of osteopenia and hypovitaminosis D by 1α-hydroxyvitamin D3 in elderly patients with Parkinson’s disease. Journal of Neurology, Neurosurgery, and Psychiatry. 1999;66(1):64–68. doi: 10.1136/jnnp.66.1.64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.King D., Playfer J. R., Roberts N. B. Concentrations of vitamins A, C and E in elderly patients with Parkinson’s disease. Postgraduate Medical Journal. 1992;68(802):634–637. doi: 10.1136/pgmj.68.802.634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Kim J. E., Oh E., Park J., Youn J., Kim J. S., Jang W. Serum 25-hydroxyvitamin D3 level may be associated with olfactory dysfunction in de novo Parkinson’s disease. Journal of Clinical Neuroscience. 2018;57:131–135. doi: 10.1016/j.jocn.2018.08.003. [DOI] [PubMed] [Google Scholar]
  • 129.Shrestha S., Lutsey P. L., Alonso A., Huang X., Mosley T. H., Jr, Chen H. Serum 25-hydroxyvitamin D concentrations in mid-adulthood and Parkinson’s disease risk. Movement Disorders. 2016;31(7):972–978. doi: 10.1002/mds.26573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Lin C. H., Chen K. H., Chen M. L., Lin H. I., Wu R. M. Vitamin D receptor genetic variants and Parkinson’s disease in a Taiwanese population. Neurobiology of Aging. 2014;35(5):1212.e11–1212.e13. doi: 10.1016/j.neurobiolaging.2013.10.094. [DOI] [PubMed] [Google Scholar]
  • 131.Zhu D., Liu G. Y., Lv Z., Wen S. R., Bi S., Wang W. Z. Inverse associations of outdoor activity and vitamin D intake with the risk of Parkinson’s disease. Journal of Zhejiang University Science B. 2014;15(10):923–927. doi: 10.1631/jzus.B1400005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Petersen M. S., Bech S., Christiansen D. H., Schmedes A. V., Halling J. The role of vitamin D levels and vitamin D receptor polymorphism on Parkinson’s disease in the Faroe Islands. Neuroscience Letters. 2014;561:74–79. doi: 10.1016/j.neulet.2013.12.053. [DOI] [PubMed] [Google Scholar]
  • 133.Török R., Török N., Szalardy L., et al. Association of vitamin D receptor gene polymorphisms and Parkinson’s disease in Hungarians. Neuroscience Letters. 2013;551:70–74. doi: 10.1016/j.neulet.2013.07.014. [DOI] [PubMed] [Google Scholar]
  • 134.Lv Z., Tang B., Sun Q., Yan X., Guo J. Association study between vitamin d receptor gene polymorphisms and patients with Parkinson disease in Chinese Han population. The International Journal of Neuroscience. 2013;123(1):60–64. doi: 10.3109/00207454.2012.726669. [DOI] [PubMed] [Google Scholar]
  • 135.Peterson A. L., Mancini M., Horak F. B. The relationship between balance control and vitamin D in Parkinson’s disease-a pilot study. Movement Disorders. 2013;28(8):1133–1137. doi: 10.1002/mds.25405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Evatt M. L., DeLong M., Kumari M., et al. High prevalence of hypovitaminosis D status in patients with early Parkinson disease. Archives of Neurology. 2011;68(3):314–319. doi: 10.1001/archneurol.2011.30. [DOI] [PubMed] [Google Scholar]
  • 137.Miyake Y., Tanaka K., Fukushima W., et al. Lack of association of dairy food, calcium, and vitamin D intake with the risk of Parkinson’s disease: a case-control study in Japan. Parkinsonism & Related Disorders. 2011;17(2):112–116. doi: 10.1016/j.parkreldis.2010.11.018. [DOI] [PubMed] [Google Scholar]

Articles from Oxidative Medicine and Cellular Longevity are provided here courtesy of Wiley

RESOURCES