FROM:
Nutrition Reviews 2012 (Jul); 70 (7): 373–386 ~ FULL TEXT
Chao J, Leung Y, Wang M, Chang RC.
Laboratory of Neurodegenerative Diseases,
Department of Anatomy,
LKS Faculty of Medicine,
The University of Hong Kong,
Pokfulam, Hong Kong SAR, China.
Parkinson's disease (PD) is the second most common aging-related disorder in the world, after Alzheimer's disease. It is characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta and other parts of the brain, leading to motor impairment, cognitive impairment, and dementia. Current treatment methods, such as L-dopa therapy, are focused only on relieving symptoms and delaying progression of the disease. To date, there is no known cure for PD, making prevention of PD as important as ever. More than a decade of research has revealed a number of major risk factors, including oxidative stress and mitochondrial dysfunction. Moreover, numerous nutraceuticals have been found to target and attenuate these risk factors, thereby preventing or delaying the progression of PD. These nutraceuticals include vitamins C, D, E, coenzyme Q10, creatine, unsaturated fatty acids, sulfur-containing compounds, polyphenols, stilbenes, and phytoestrogens. This review examines the role of nutraceuticals in the prevention or delay of PD as well as the mechanisms of action of nutraceuticals and their potential applications as therapeutic agents, either alone or in combination with current treatment methods.
From the Full-Text Article:
INTRODUCTION
Parkinson’s disease (PD) is regarded as the second most
prevalent aging-related neurodegenerative disorder after
Alzheimer’s disease (AD), affecting approximately
0.017% of people between the ages of 50 and 59 years,
with a median onset age of around 60 years. Aging is
undoubtedly amajor risk factor of PD, as the incidence of
PD jumps to approximately 0.093% in people between the
ages of 70 and 79 years. In people over 70 years of age, the
number of men diagnosed with PD is about 1.5 times
higher than that of women. [1]
Environmental toxins and exposure to pesticides
have also been reported to contribute to PD morbidity.
Examples of environmental toxins include 1-methyl-4-
phenyl-1,2,3,6-tetrahydropyridine (MPTP), toluene,
carbon disulfide, and cyanide, [2] while examples of pesticides
include paraquat, organophosphates, and rotenone. [3]
MPTP exposure has been found to induce both loss of
dopaminergic neurons and clinical parkinsonism. Similarly,
rotenone and paraquat, when applied to experimental
animals, have been found to induce loss of
dopaminergic neurons and typical parkinsonism. [4] As
potential etiological factors of PD, environmental
chemicals may interact with gene expression and modulate
expression of mutated genes in humans.Thus, genetic
analysis of patients with PD has become an important
research theme in PD.
Table 1
|
A small percentage of PD cases are attributed to
single gene defects. Thus far, genetic studies have identified
more than 10 genes that cause familial PD. The functions
of these genes, along with associated clinical
features, are listed in Table 1. [5, 6]
PATHOLOGICAL FACTORS ASSOCIATED WITH PARKINSON'S DISEASE
PD is defined pathologically by the progressive loss of
dopaminergic neurons in the substantia nigra (SN) pars
compacta accompanied by the presence of intracellular
Lewy bodies.Parkinsonism, along with parkinsonian syndrome,
should be distinguished from PD.Parkinsonism is
a term that refers only to the clinical symptoms of PD,
such as the occurrence of tremors and dementia,but bears
no implication of disease mechanism, while PD refers to
the pathology described above. The exact mechanisms of
PD are not yet fully understood, although several factors,
including protein misfolding, oxidative stress, and mitochondrial
dysfunction, have been reported.
Numerous studies indicate a causal role of misfolded
proteins such as beta-amyloid and a-synuclein inAD and
PD, as misfolding results in accumulation of the protein
either extra- or intracellularly. [7] If chaperones fail to
restore misfolded proteins, the ubiquitin proteasome
system and autophagy will subsequently clear the misfolded
proteins. [8] Both mutation and aggregation of
a-synuclein can cause parkinsonism, but mutation of
a-synuclein is rarely found in patients with sporadic PD. [9]
Unique biochemical features of the SN render it
more vulnerable to oxidative stress when compared with
other parts of the brain. In particular, the SN has a
uniquely high iron content.Dopamine can be oxidized by
monoamine oxidase B (MAO-B) to form hydroxyl free
radicals in the presence of ferrous iron. The combined
effects result in enormous amounts of hydroxyl free radicals,
leading to severe damage of the dopaminergic
neurons of the SN. These pathological events suggest that
oxidative stress is the most important pathological factor
for the initiation and progression of PD. [10]
Indeed, the SNs of PD patients have been found to
show elevated levels of oxidative stress.Youdim and Riederer [11]
reported that lipid-peroxidation-promoting substances
such as ferrous iron are found in high levels in the
SN of postmortem PD brain, concomitant with decreased
levels of antioxidants. Similar results were obtained by a
study investigating the level of hydroxynonenal adducts,
which are products of lipid peroxidation. [12] Elevated levels
of lipid peroxidation have been found in the SN region of
the brain and in erythrocytes from blood samples of PD
patients. [13]
Numerous studies suggest that impairment of mitochondrial
function is also involved in the pathogenesis of
PD.Mitochondrial function is closely related to oxidative
stress because mitochondria produce ATP by oxidative
phosphorylation. During ATP production, mitochondria
may produce superoxide radicals as by-products. Defects
of the electron transport chain can result in the failure
of energy metabolism, increased free-radical-mediated
damage, and activation of downstream cell death
pathways. [14–16] In the early 1980s, contamination of MPTP
in heroin was found to cause parkinsonism in drug abusers. [17] Since then, MPTP has been used extensively to
induce neurotoxic experimental PD. The active form of
MPTP is metabolized in the mitochondria of astrocytes
in the SN to form MPP+ and is then transferred to inhibit
complex I of the electron transport chain in neurons,
leading to ATP depletion and accumulation of reactive
oxygen species (ROS). [18] Elevated mitochondrial ROS
levels result in mitochondrial DNA mutations, proteins/
lipids perturbation and can further affect redox signaling
pathways. [19] Another widely used neurotoxin, 6-OHDA,
induces pathological events similar to those seen in
experimental PD. [20] Numerous studies have shown that
food components and nutritional substances can prevent
or delay the progression of PD by protecting mitochondrial
function. [21] This further supports the role of mitochondrial
impairment as a major pathological factor in
PD. [22–24]
TREATMENT FOR PATIENTS WITH PARKINSON'S DISEASE
Although PD was first diagnosed almost two centuries
ago, a cure has yet to be found. Current treatments are
mainly categorized into symptom-relieving drugs and
surgical treatments. L-dopa, dopamine agonists (pramipexole,
bromocriptine, pergolide, ropinirole, piribedil,
cabergoline, apomorphine, and lisuride) and MAO-B
inhibitors (selegiline and rasagiline) are examples of
symptom-relieving drugs, while deep brain stimulation,
implantation of embryonic dopaminergic cells, and gene
therapy have been applied as surgical treatments for PD
patients. These treatments only aim to improve the
quality of life by attenuating motor or nonmotor symptoms
of PD. As the global population ages, the need to
develop a disease-modifying drug for PD is becoming
increasingly urgent.
Existing PD treatments have undesirable effects. For
example, L-dopa, a commonly used symptom-relieving
drug for PD, has various side effects because 95–99% of it
is metabolized to dopamine in the body in places other
than the dopaminergic neurons in the SN.For this reason,
dopa decarboxylase inhibitors (e.g., carbidopa and
benserazide) and COMT enzyme inhibitors (e.g., tolcapone
and entacapone) are prescribed in combination
with L-lopa to enhance its effect. Discontinuous delivery
of L-dopa has been another limitation of the treatment.
Novel delivery methods of L-dopa seek to overcome this,
such as an intravenous infusion delivery approach and a
transdermal delivery system, both of which have been
applied in clinical settings for the past two decades. [5, 6]
POTENTIAL NEUROPROTECTIVE EFFECTS OF NUTRACEUTICALS
Combining the words “nutrition” and “pharmaceutical,”
the word“nutraceuticals” refers to foods or food products
that reasonable clinical evidence suggests may provide
health and medical benefits, including for prevention and
treatment of disease. Such products may be categorized as
dietary supplements, specific diets, herbal products, or
processed foods such as cereals, soups, and beverages.
Dietary supplements can be extracts or concentrates and
are found in many forms, including tablets, capsules,
liquids, and powders. Vitamins, minerals, herbs, or isolated
bioactive compounds are only a few examples of
dietary ingredients in the products. Functional foods are
designed as enriched foods close to their natural state,
providing an alternative to dietary supplements manufactured
in liquid or capsule form.
It is generally accepted that neuroprotection prevents
neurons from succumbing to damages by different
insults. Nutraceuticals can provide neuroprotection via a
wide range of proposed mechanisms, such as scavenging
of free radicals and ROS, chelation of iron, modulation of
cell-signaling pathways, and inhibition of inflammation. [25]
Neuroprotection can prevent and impede the progression
of PD as well as the loss of dopaminergic neurons. In the following section, the neuroprotective effects of selected dietary supplements and functional foods are reviewed
and discussed. In addition, several relevant therapeutic
effects are evaluated.
ANTIOXIDANT VITAMIN SUPPLEMENTS (VITAMINS C AND E)
Antioxidant vitamin supplements such as vitamin C,
vitamin E (or tocopherol), and beta-carotene are
common forms of nutraceuticals. [26] A cross-sectional
study found that vitamin E supplements are popular in
PD patients, while epidemiological studies have shown
that consuming foods rich in vitamins C and E are associated
with a lower risk of developing PD. [27] However, it
should be noted that these studies are not specific to
individual antioxidant nutrients; rather, it is the foods
rich in these nutrients that are studied.
Potential Neuroprotective Effects
An early study has suggested a protective effect of these
two antioxidative vitamins on PD patients.28 In an openlabel
trial, high doses of vitamins C and E were administered
to patients in the early stage of PD. It was found that
patients who took antioxidant vitamins had a 2.5- to
3-year delay in receiving L-dopa treatment compared
with those of Dr. CM Tanner, who did not treat patients
with vitamins, as reported by Fahn. [28] Treatment was
delayed from 40 months to 72 6.5 months for those PD
patients who started taking the vitamins before 54 years
of age, and from 24 months to 63 3.9 months for those
who started the vitamins after 54 years of age. Although
the placebo effect might be at play here, the delay of onset
of parkinsonism was remarkably significant. Another
report showed that vitamin C at 10 mM can reduce neurotoxicity
elicited by dopamine metabolism. [28]
An important double-blind and placebo-controlled
clinical study, the Deprenyl and Tocopherol Antioxidative
Therapy of Parkinsonism (DATATOP) by the
Parkinson Study Group, showed that vitamin E supplementation
was not able to delay the need for introducing
L-dopa therapy. [30] However, as pointed out in a commentary
for this study, the trial did not exclude the possibility
that nutritional supplements may delay progression of
PD by preventing loss of dopaminergic neurons. [31] A contradicting
report showed that 9.8 IU/day of vitamin E
intake from the diet may be beneficial. [32] A meta-analysis
produced similar results, showing that dietary intake of
vitamin E in moderate amounts may be neuroprotective.
High intake of vitamin C in the form of a supplement was
not significantly protective, with no association found
between vitamin C intake and risk of PD. [33]
Mechanisms of Action
Antioxidant vitamins have a putative role in reducing the
oxidative damage in SN dopaminergic neurons in progressive
disease. [34] Vitamin C has been proven in vitro to
be a major free-radical scavenger in the cytosol, while
tocopherols act as a major lipid-soluble antioxidant to
prevent lipid peroxidation in membranes. Both vitamins
also act in a synergistic manner whereby vitamin C can
reduce oxidized vitamin E to restore its antioxidative
function. [35] Thus, supplemental vitamins can be useful in
prevention or in delaying progression of PD by reducing
oxidative stress.
VITAMIN D
Potential Neuroprotective Effects
In 2007,Newmark and Newmark [36] proposed that vitamin
D deficiency had a significant role in the development
and progression of PD. Vitamin D has been found to
attenuate 6-OHDA-induced and MPP+-induced neurotoxicity,
while vitamin D receptor knockout mice show
motor defect.Moreover, the levels of vitamin-D-binding
protein have been proposed as one of the biomarkers
for PD. [37–39]
It has been debated that vitamin D inadequacy in PD
patients is a result of reduced physical activity and exposure
to sunlight, rather than a causal factor in PD progression.
However, the results of a recent longitudinal
study by Knekt et al. [40] oppose this view.A large sample of
Finnish adults aged 30 years or older was selected from
1978 to 1980, and blood serum samples were examined.
Occurrences of PD were recorded in a 29-year follow-up
period. In 2002, serum levels of vitaminDweremeasured,
and results showed that subjects with higher serum
vitamin D levels had a significantly lower risk of developing
PD. [40] These data suggest that vitamin D levels
could be used as a predictive indicator of PD risk.
Mechanisms of Action
The SN is one of the regions in the brain containing high
levels of vitamin D receptors and 1a-hydroxylase, [40] the
enzyme responsible for the biological activation of
vitamin D. Hence, vitamin D may be involved in a
number of signaling pathways, and several mechanisms
may be responsible for the neuroprotective effects of
vitamin D.
In animal studies, vitamin D was found to upregulate
glial cell line-derived neurotrophic factor levels. [37] Glial
cell line-derived neurotrophic factor has been shown to
be antiparkinsonian in animal and in vitro studies. It can promote the outgrowth of dopaminergic axons in striatal
neurons in a region-specific manner and can even
rescue SN neurons from 6-OHDA toxicity.41 In addition,
vitamin D can increase glutathione levels, regulate
calcium homeostasis, exert anti-apoptotic and immunomodulatory
effects, reduce nitric oxide synthase, and
regulate dopamine levels. [42, 43]
COENZYME Q10
Potential Neuroprotective Effects
Figure 1
|
Coenzyme Q10 (CoQ10 or ubiquinone) is a popular commercially
available dietary supplement (Figure 1). It has
been recognized as a neuroprotective agent in the prevention
and treatment of PD. [44] CoQ10 has been demonstrated
to prevent the loss of dopaminergic neurons in
MPTP-induced neurotoxicity and parkinsonism. [45, 46] In a
placebo-controlled, randomized, double-blind study
involving 80 patients with early-stage PD, patients in the
treatment group were found to have less disability, as
evaluated for over 16 months using the Unified Parkinson
Disease Rating Scale. It should be noted that the effects of
CoQ10 were dose dependent. The group receiving
1,200 mg/day, which was the highest dose among the different
groups, exhibited a 44% reduction in functional
decline compared with the placebo group. [47] In another
study, amild symptomatic benefit was observed using the
Farnsworth-Munsell 100 Hue test. The authors suggested
that an oral supplement of CoQ10 could achieve a moderate
beneficial effect, but not a great neuroprotective effect. [48] From these reports, there is no conclusion about
whether the effect of CoQ10 on PD is neuroprotective or
merely symptom relieving.
Mechanisms of Action
CoQ10 is a fat-soluble and vitamin-like quinone found
abundantly in liver and the brain. [49] CoQ10 is particularly
relevant to mitochondrial dysfunction because of its
unique electron-accepting property, which allows it to
bridge mitochondrial complex I with other complexes.
CoQ10 plays an important role in maintaining proper
transfer of electrons in the electron transport chain of
mitochondria and, thus, in the production of ATP as well.
As a result, CoQ10 has a protective effect on dopaminergic
neurons in the SN. In addition, it is a potent antioxidant
and can exert its antioxidant effect by reducing the oxidized
form of alpha-tocopherol, [50] which is important in
the prevention of lipid peroxidation.
CREATINE
Potential Neuroprotective Effects
Creatine has also been investigated for its possible role in
the treatment and prevention of PD (Figure 1). In a study
using MPTP in a PD mouse model, a diet supplement
of 1% creatine reduced loss of dopaminergic neurons in
the SN.51A placebo-controlled and randomized pilot trial
for a 2-year period showed that creatine can improve
mood and reduce the dosages required for dopaminereplacement
therapy in the treated group. [52]
Mechanisms of action
Creatine is considered to be neuroprotective due to its
ability to counter ATP depletion by increasing intracellular
phosphocreatine levels. [51] Phosphocreatine is a key
player in the maintenance of ATP levels, which in turn
are important in synaptic activity and skeletal muscle
functions. [53, 54]
UNSATURATED FATTY ACIDS
Potential Neuroprotective Effects
While unsaturated fatty acids were reported to reduce the
risk of developing PD, [55] results from past epidemiological
and retrospective studies were inconsistent. To study the
relationship, a prospective study was conducted in two
cohorts, the Health Professional Follow-up Study and the
Nurses’ Health Study. [56] The authors concluded that if saturated fatty acids are replaced by polyunsaturated fatty
acids (PUFAs), the risk of developing PD may be reduced.
In another large prospective population-based cohort
study, the Rotterdam Study, the authors investigated the
relationship between dietary unsaturated fatty acids and
the risk of developing PD. [55] In contrast to the previous
study, they showed no relationship between the level of
saturated fatty acids and the risk of developing PD. In
addition to the above studies, the results of a recent investigation
on omega-3 PUFAs suggest a neuroprotective
effect of omega-3 PUFAs against dopamine loss and an
inhibitory effect against the formation of dihydroxyphenylacetic
acid in MPTP-induced parkinsonism in mice. [57]
This positive result should encourage future studies on
the possible mechanism of PUFAs.
Mechanisms of Action
PUFAs such as linoleic acid, alpha-linolenic acid, and
docosahexaenoic acid can be components of cell membrane
and precursors of signaling molecules. [58] Some of
these PUFAs cannot be synthesized in the human body
and must be obtained from food.Monounsaturated fatty
acids (MUFAs) can also reduce cholesterol and triacylglycerides
in plasma. [59] Impaired brain function is strongly
associated with deficiency of MUFAs and PUFAs. Endogenous
cannabinoids derived from MUFAs are important
modulators for dopaminergic neurons in the basal ganglia. [60] A report has shown that fatty acid composition in
the brain is highly correlated with the intake of dietary
fatty acids. [61] All these facts justify further study of the
relationship between the intake of unsaturated fatty acids
and the risk of developing PD.
NATURAL SOURCES OF L-DOPA
Potential Neuroprotective Effects
To date, natural L-dopa has been found in several plants
belonging to Mucuna genus, such as Mucuna pruriens
(velvet bean or mucuna, the seeds of which, in 1937, were
found to contain L-dopa), Stizolobium deeringianum, and
Vicia faba (broad bean, in which L-dopa was identified in
1913). M. pruriens (called “atmagupta” in India) is a
climbing legume endemic in tropical regions that include
India and Central and South America. The plant has been
documented in Ayurvedic medicine to treat a neurological
disorder bearing symptoms similar to those of PD and
up to 10% of the plant’s volume is L-dopa. [62] In recent
years, velvet bean seed extract has been used for the treatment
of PD in India. [63]
Several open-label studies with sample sizes ranging
between 18 and 60 patients prescribed mucuna seed
powder extract at mean doses of 45 g/day (containing
about 1,500 mg L-dopa). Significant improvements in
parkinsonism were reported and better tolerability
was found compared with standard L-dopa treatment
alone. [64–66] In a recent double-blind study involving eight
PD patients, the anti-Parkinsonian effect, tolerability, and
L-dopa pharmacokinetic profile were compared between
the mucuna seed formulation and the commercial
L-dopa. [67] The results showed that the effects of 30 g of
M. pruriens formulation were superior to those of the
standard single doses of 200/50 mg L-dopa/carbidopa.
The bean powder enabled a more rapid onset of action in
patients and had a slightly longer duration of therapeutic
response. Moreover, severe dyskinesia or peripheral
dopaminergic adverse events were not found in the
mucuna-treated patients. It is suggested that the mucuna
formulation may have greater bioavailability, perhaps as a
result of synergistic properties of different compounds in
the seed extract.
Mechanisms of Action
Most in vitro studies on natural L-dopa sources focus on
mucuna. In 2004, Manyam et al. [68] showed that mucuna
seed powder contained significant amounts of two neuroprotective
agents, namely nicotine adenine dinucleotide
(NADH) and CoQ10. Both agents protect neurons
against 6-OHDA toxicity by counteracting the inhibition
of mitochondrial complex I activity.NADHis also known
to increase dopamine levels via the upregulation of
tyrosine hydrolase.
Mucuna seed powder has also been found to protect
neurons against plasmid DNA and genomic DNA
damage caused by a combination of L-dopa and divalent
copper ions. [69, 70] Mucuna seed powder protects neurons
against this type of damage by chelating the divalent
copper ions present, preventing them from interacting
with L-dopa to produce
POLYPHENOLIC COMPOUNDS
Polyphenolic compounds, or polyphenols, are products of
secondary plant metabolism and are widely distributed in
the plant kingdom. Polyphenolic compounds refer to a
range of substances that possess an aromatic ring bearing
more than one hydroxyl group.More than 8,000 phenolic
structures have been identified. Polyphenols are generally
divided into hydrolyzable tannins (gallic acid esters of
glucose and other sugars) and phenylpropanoids, such as
lignins, flavonoids, and condensed tannins.
Polyphenols can elicit antioxidant, antiinflammatory,
anticarcinogenic, antimutagenic, and antithrombotic effects. [71] The neuroprotective effects of
the major polyphenolic compounds in green tea, black
tea, coffee, curry, and Scutellaria baicalensis, an herb used
in traditional Chinese medicine, are reviewed below.
EGCG in green tea
Potential Neuroprotective Effects. Numerous studies
suggest green tea may confer health benefits due to its
pharmacological and biochemical properties. Epidemiological
studies have shown an inverse relationship
between tea consumption and the risk of developing PD.
There are several experimental studies showing neuroprotective
effects of green tea on MPTP-induced parkinsonism
in mouse models and on cell injury in
pheochromocytoma PC12 cells treated by 6-OHDA. [72]
Many of the beneficial effects of green tea are attributed
to its abundant polyphenol content, mainly the flavans
called catechins (Figure 2). [73] There are numerous
catechins found in green tea, the major ones being
(-)-epicatechin (EC), (-)-epicatechin-3-gallate (ECG),
(-)-epigallocatechin (EGC), and (-)-epigallocatechin-3-
gallate (EGCG). EGCG is the most abundant catechin. [74]
Levites et al. [75] summarized the biological functions of
tea polyphenols and reported the following benefits:
free-radical scavenging and anticarcinogenic, antiinflammatory,
and antiangiogenic effects.
Mechanisms of Action
Different mechanisms have been
proposed for the neuroprotective activity of EGCG in PD.
The study conducted by Levites et al. [75] was the first to
demonstrate the neuroprotective activity of both green
tea extract (0.5 and 1 mg/kg) and EGCG (2 and 10 mg/
kg) on MPTP-induced parkinsonism in animal models. It
is possible that the neuroprotective effects are mediated
by iron-chelating activities and free-radical-scavenging
activities possessed by the catechol group. Since green tea
catechins can pass through the blood-brain barrier, they
can act as both ROS scavengers and iron chelators to clear
the redox active ferrous iron deposited in the SN, reducing
the iron-induced oxidative stress that can lead to neuronal
death.
The putative neuroprotective effects of green tea catechins
also may be mediated via other mechanisms.
Mandel et al. [73] and Levites et al. 76 summarized the
neuroprotective mechanisms of green tea catechins as
regulation of protein kinase C activity and induction
of endogenous antioxidant defense systems. A recent
experimental study using the 6-OHDA rat model of
PD also suggests that green tea catechins protect the
SN dopaminergic neurons through modulation of the
ROS-NO pathway. [77] It appears there is considerable evidence
to support the putative neuroprotective effects of
green tea.Nonetheless, much of the evidence was derived from experimental and animal studies, while evidence
from large prospective studies or case-control studies specific
to green tea catechins rather than to general tea
consumption is limited. In contrast to other reports
showing beneficial effects of green tea, the prospective
cohort study of the Singapore Chinese Health Study [78]
showed no relationship between green tea consumption
and the risk of developing PD if caffeine intake was
excluded. Therefore, more studies of green tea consumption
in humans and the risk of developing PD are required
to verify the possible protective effect of green tea.
Curcuminoids in curry
Potential neuroprotective effects. Curcumin (1,7-bis[4-
hydroxy 3-methoxy phenyl]-1,6-heptadiene-3,5-dione) is
a polyphenolic flavonoid that constitutes approximately
4% of turmeric, which has a long history of use in traditional Asian diets and herbal medicines (Figure 2). Curcumin
is the principal curcuminoid in turmeric. The
other two curcuminoids are desmethoxycurcumin and
bisdesmethoxycurcumin.Curcuminoids, rather than curcumin
alone, are commercially available and are generally
used in experimental studies. The bioactive effects of curcuminoids
have often been attributed to curcumin, as the
curcumin content of curcuminoids reaches up to 80%.
Mechanisms of Action
Like other polyphenolic compounds
such as caffeic acid, EGCG, and resveratrol, curcumin
is well known for its powerful antioxidant
properties. Jagatha et al. [79] reported that curcumin treatment
of mice and of dopaminergic neurons in cell cultures
attenuated oxidative stress by restoring glutathione
levels, thereby protecting neurons against protein oxidation
and preserving mitochondrial complex I activity.
The reduction of 6-OHDA-induced neurotoxicity in
MES 23.5 cells and in a rat model of PD has been attributed
to the antioxidant properties of curcumin. [80] In addition,
curcumin’s direct modulation of 6-OHDA-induced
nuclear factor-kappa B (NF-kB) translocation confers
neuroprotective effects in dopaminergic neuronal cells of
the MES 23.5 cell line. [81]
Curcumin has also been found to exhibit antiinflammatory
properties. In primary cultures of rat mesencephalic
neuronal/glial cells, curcumin inhibited
lipopolysaccharide (LPS)-induced morphological
changes of microglia and dramatically reduced LPSinduced
production of many proinflammatory factors
and their gene expressions. LPS-induced activation of
transcription factors, such as NF-kB and activator
protein-1, were also attenuated by curcumin treatment. [82]
In addition, curcumin has been found to prevent
MPTP/MPP+-induced neurotoxicity in C57BL/6N mice,
SH-SY5Y cells, and PC12 cells by targeting the JNK, the
Bcl-2-mitochondria, and the ROS-iNOS (inducible nitric
oxide synthase) pathways. [83, 84] Systemic administration
of curcumin (80 mg/kg i.p.) and its metabolite tetrahydrocurcumin
(60 mg/kg i.p.) significantly reversed
MPTP-induced depletion of DA and DOPAC (3,4-
dihydroxy phenyl acetic acid) in mice. The authors concluded
that the reversion may be, in part, due to the
inhibition of MAO-B activity by these compounds. [85]
Furthermore, both overexpression and abnormal
accumulation of aggregated alpha-synuclein (AS) have
been found to be closely linked to PD. Recent studies
revealed that curcumin could inhibit aggregation of AS in
cell-free conditions and in a cellular model of A53T-AS
overexpression. [86, 87]
Ortiz-Ortiz et al., [88] however, called for re-evaluation
of the potential of curcumin as a therapeutic agent in
neurodegenerative diseases. In contrast to findings
reported previously by others, Ortiz-Ortiz et al. [89] surprisingly
found that exposure of N27 mesencephalic cells to
10 nM curcumin synergistically enhanced paraquatmediated
apoptosis. A very recent study from the same
group found that exposure of rat mesencephalic cells to
10 nM curcumin induced the expression of LRRK2 in
mRNA and protein levels, although there was no effect on
other PD-related genes like AS and parkin. Overexpression
of LRRK2 is strongly associated with the pathological
inclusions found in PD. Taken together, the findings
for curcumin remain controversial and await further
experimental and clinical studies.
Baicalein
Potential Neuroprotective Effects
Baicalein is a flavonoid
extracted from the root of Scutellaria baicalensis, a traditional
Chinese herb commonly known as Huang Qin.
Baicalein has been shown to be a potent antioxidant in rat
primary neurons (Figure 2). [90] Another study in rats also
showed anti-inflammatory properties of baicalein in
experimental traumatic brain injury. [91] Baicalein was
found to be neuroprotective in several experimental
models of PD, including MPTP-induced neurotoxicity
and 6-OHDA-induced neurotoxicity. [92, 93] It has also been
shown to inhibit fibrillization of AS. [94] In a recent study,
baicalein attenuated depolarization of mitochondria and
proteasome inhibition in PC12 cells induced by the E46K
mutation, an AS mutation linked to familial parkinsonism. [95] The mechanisms underlying the neuroprotective
effects of baicalein, however, remain unclear.
STILBENES
Stilbenes are a class of antioxidants sharing the same
chemical skeleton of a diarylethene, which is a hydrocarbon
consisting of a trans/cis ethene double bond substituted
with a phenyl group on both carbon atoms of the
double bond. The name “stilbene” was derived from the
Greek word “stilbos,” which means “shining.” Many stilbenes
and their derivates (stilbenoids) are naturally
present in plants (dietary fruits or herbs).
Resveratrol
Potential Neuroprotective Effects
The most widely
investigated stilbene is resveratrol (3, 4', 5-transtrihydroxystilbene,
RES, Figure 3), a phytoalexin found in
plants such as grapes, peanuts, berries, and pines. [96] RES is
synthesized in these plants to counteract various environmental
injuries, such as UV irradiation and fungal infection.
RES is reported to be one of the active agents in
Itadori tea, which has been used as a traditional medicine
in China and Japan, mainly for treating heart disease and
stroke. [97] Epidemiological studies reporting the inverse association between moderate consumption of red wine
and the incidence of coronary heart disease have stimulated
investigations on the cardioprotective activity of
RES. [98] In recent years, numerous studies have shown that
RES can protect dopaminergic neurons against toxicity
induced by LPS, DA, or MPP+. [99–101] The neuroprotective
effects of RES have also been reported in 6-OHDAlesioned
rats and in mouse models of MPTP-induced
neuronal loss. [102, 103]
Mechanisms of Action
The underlying mechanisms of
neuroprotection by RES include the inhibition of
NADPH oxidase and the suppression of proinflammatory
genes such as interleukin 1-a and tumor necrosis
factor-a triggered by LPS. [99, 104] Pretreatment with RES
reduced apoptosis in PC12 cells by modulating mRNA
levels and protein expression levels of BAX and Bcl-2 in
vitro. [100] RES may stimulate SIRT1 in 6-OHDA-triggered
SK-N-BE cells, as indicated by the loss of protection in the
presence of the SIRT1 inhibitor sirtinol, a loss that also
occurred when SIRT1 expression was downregulated by
siRNA approach. [105–107] In addition, RES exhibits neuroprotective
effects on MPTP-induced motor coordination
impairment, hydroxyl radical overloading, and neuronal
loss through free-radical-scavenging activity. [102]
Oxyresveratrol
Potential Neuroprotective Effects
Recent studies have
found that RES may not be the most effective neuroprotective
agent. Investigations on the differential
bioactivities of RES and oxyresveratrol (OXY) (2, 32, 4,
52-trans-trihydroxystilbene, Figure 3) have shownOXY to
be a more effective neuroprotective agent. OXY is found
in the heartwood or fruit of Artocarpus heterophyllus,
Artocarpus lakoocha, Artocarpus gomezianus, and Artocarpus
dadah, in the wood or fruit of mulberry trees
(Morus australis,Morus alba L.), in the fruit of Melaleuca
leucadendron, in rhizomes of Smilacis chinae, and in the
Egyptian herb Schoenocaulon officinale.
Mechanisms of Action
In vivo and in vitro studies have
shown anti-inflammatory effects of OXY, particularly
OXY isolated from Artocarpus heterophyllus, Artocarpus
dadah, or mulberry wood. [108, 109] OXY can also reduce the
production of beta-amyloid by inhibiting b-secretase 1. [110]
OXY has been demonstrated to protect against 6-OHDAinduced
toxicity in SH-SY5Y cells by reducing the release
of lactate dehydrogenase and caspase-3 specific activity. [111]
Analysis by high-performance liquid chromatography
showed that OXY readily penetrates into neurons,
thereby suppressing the level of intracellular ROS by its
potent free-radical-scavenging activity. OXY was also
found to upregulate SIRT1 levels, indicating that the neuroprotective
properties of SIRT1may be attributable to its
activation. [111]
PHYTOESTROGENS
It has been known that the incidence of PD is lower in
women than in men (using age controls), indicating a
protective effect of estrogen or its derivatives. [112] The incidence
of PD is also lower in premenopausal women
than in postmenopausal women. [113] The neuroprotective
effects of estrogen have been shown in many studies,
including upregulation of Bcl-2 and brain-derived neurotrophic
factor. [114] However, numerous side effects discourage
women from receiving hormone replacement
therapy. Phytoestrogens, obtained through either the diet
or supplements, provide an alternative to traditional
hormone replacement therapy without some of the
reported side effects; this will be discussed in the following
section.
Phytoestrogens are a group of substances that are
found naturally in plants and possess a common chemical
structure similar to that of estradiol. Major food sources
of phytoestrogens include soy products, nuts, and grains. Two types of phytoestrogens are discussed below.
Ginsenoside Rg1
Potential Neuroprotective Effects
Ginsenosides are a class
of molecules extracted from several species of ginseng.
Ginseng has a long history in traditional Chinese medicine,
Indian herbal medicine, and the medicine of other
Asian cultures, and it is well known for its antiaging
effects. Rg1 is a ginsenoside isolated from the root of Panax ginseng. It is one of the relatively well-studied ginsenosides
(Figure 4). In vivo, Rg1 can attenuate 6-OHDA
neurotoxicity, MPTP-induced neurotoxicity, and oxidative
stress. [112, 115] It can also suppress tumor proliferation. [116]
In vitro studies have shown that Rg1 can attenuate rotenone
toxicity. [117]
Mechanisms of Action
Ginsenoside Rg1 has been found
to regulate several signaling pathways, which may explain
its neuroprotective effects. The signaling pathways modulated
by ginsenoside Rg1 include PI3K/Akt, ERK, JNK,
ROS-NFkB, IGF-1 receptor signaling pathways, and
estrogen receptor pathway. [115, 118–120]
In 2005,Chen et al. [115] tested the effects of Rg1 against
MPTP-induced neurotoxicity in mice. Results showed
that Rg1 was able to reduce neuronal loss caused by
MPTP toxicity through two possible mechanisms. First,
Rg1 prevented the reduction of glutathione. Second, Rg1
attenuated phosphorylation of c-Jun, as JNK signaling
can be proapoptotic. [115, 121] A third mechanism was proposed
by Wang et al. [122] in 2009. By iron staining, the
authors showed, in a mouse model of MPTP toxicity, that
elevated iron levels in the SN were linked to neuronal
death. Rg1 prevented this elevation of iron levels by regulating
the expression of iron transport proteins such as
ferroportin 1 and divalent metal transport 1. [122]
Rg1 is also beneficial to the maintenance of mitochondrial
functions. In the presence of rotenone, Rg1
restored depleted mitochondrial membrane potential. [117]
Antiapoptotic effects included inhibition of cytochrome c
release and activation of the PI3K/Akt cell survival
pathway, resulting in enhanced inhibition of Bad protein
expression. [117] Upon blocking the glucocorticoid receptor
with an antagonist, these effects were blocked, indicating
that Rg1 mediates its effects through the glucocorticoid
receptor. [117]
Genistein
Potential Neuroprotective Effects
Soy and peanuts are rich
dietary sources of the phytoestrogen genistein, which has
been found to be the primary circulating soy isoflavone
(Figure 4). [123] In fact, dietary soy is widely used as an alternative to traditional hormonal replacement therapy. In
2007, a study was conducted byAzadbakht et al. [124] to find
the effects of dietary soy on postmenopausal women with
metabolic syndrome. Compared with normal subjects,
the postmenopausal women had reduced plasma levels of
malondialdehyde, an oxidative stress marker. Numerous
studies in rats have shown that treatment with genistein
isolated from plant sources results in similar antioxidative
effects and antiapoptotic effects.
Mechanisms of Action
Many studies have shown that
genistein binds to estrogen receptors in the central
nervous system. The estrogen receptor b has been found
to have a particularly high binding affinity for genistein. [126]
Upon binding to the estrogen receptor, the genisteinreceptor
complex acts as a transcriptional activator to
upregulate antioxidative and antiapoptotic genes. [123, 126]
The antioxidative effects of genistein have been
attributed to its ability to increase the levels of malondialdehyde,
superoxide dismutase, and monoamine oxidase.
[124, 127] On the other hand,Kaul et al. [128] concluded that
genistein specifically attenuated the generation of ROS,
but not oxidative stress. [128]
They conducted an experiment
testing the effect of genistein on hydrogen-peroxideinduced
cell death in rat mesencephalic dopaminergic
neurons known as N27 cells. While no antioxidative
mechanism was suggested, the authors showed that
genistein acted as a tyrosine kinase inhibitor, thereby
attenuating the activation of protein kinase C gamma and
its downstream proapoptotic effects. [128]
In addition, it has been proposed that genistein may
be able to regulate activity of dopaminergic neurons
because estradiol has been shown to play a role in regulation
of the neurotransmitter in animal studies. [125] A
recent study testing the effects of genistein treatment
prior to intrastriatal 6-OHDA lesions in rats is in line with
this hypothesis. It was found that genistein pretreatment attenuated rotational behavior in rats, a symptom of
parkinsonism. [126]
POTENTIAL APPLICATIONS OF NUTRACEUTICALS IN CURRENT PD THERAPY
The potential benefits of nutraceuticals in PD may extend
from prevention to the delay of disease progression. Furthermore,
dietary supplements or functional foods may
reduce the side effects of current treatments or enhance
the bioavailability of L-dopa.
B vitamins and hyperhomocysteinemia
Numerous studies have demonstrated that treatment with
L-dopa in PD patients induces high levels of homocysteine
(HHcy). Studies show that HHcy is a substantial risk
factor for cardiovascular, cerebrovascular, and peripheral
vascular diseases as well as cognitive impairment and
dementia. [129] L-dopa administered to PD patients is
metabolized to 3-O-methyl-dopa via methlyation by
COMT in peripheral tissues. S-adenosyl-methionine
(SAM) provides the methyl group in the reaction and is
converted to S-adenosyl-homocysteine (SAH) after
donation of the methyl group to L-dopa. Subsequent
metabolic reactions metabolize SAH to HHcy, resulting
in increased levels of HHcy in plasma.130 It is well recognized
that high levels of HHcy can be caused by deficiencies
in any one of the three important B vitamins, namely,
folate, vitamin B12, and vitamin B6, 129 because HHcy can
be catabolized to cysteine by a chain reaction in which
vitamin B6 acts as a cofactor, while methionine synthase,
an enzyme using vitamin B12 as a cofactor, and 5-methyltetrahydrofolate
can also metabolize HHcy to methionine.
[130] Reports have shown that PD patients treated with
L-dopa exhibit higher HHcy levels in plasma, but a significant
reduction in HHcy levels was observed in PD
patients supplemented with folate, vitamin B12, and
vitamin B6. Therefore, supplementation with these vitamins
is important for managing the elevated HHcy levels
in PD patients. [129, 131]
Vitamin C, hydrosoluble fiber, and pharmacokinetics
Although findings about the efficacy of the neuroprotective
effects of vitamin C were inconclusive, vitamin Cmay
improve the efficacy of L-dopa. In a pharmacokinetic
study, vitamin C was found to enhance absorption of
L-dopa in elderly patients with PD. [132] Another study
using water-soluble fiber of Plantago ovata husk showed
that treatment of the plant with L-dopa/carbidopa
benefits PD patients by relieving constipation and
improving the L-dopa profile. [133] These studies suggest that functional foods can help patients via augmentation
with drug therapy.
CONCLUSION
The relationship between diet and disease prevention is
not a new concept. In fact, the basic theory in Chinese
herbal medicine, “medicine and diet share the same
origins,” emphasizes that scientific diet strategy may play
an undeniable role in human health. One after another,
studies have shown the importance of a nutritious diet
and active lifestyle as a healthy aging strategy in the prevention
of most aging-related diseases, such as cancer,
cardiovascular disease, and neurodegenerative diseases.
In fact,many populations worldwide have embraced this
concept for generations and have incorporated various
kinds of nutraceuticals in their diet. Not only should this
concept be encouraged as part of daily living to prevent
disease, it should also be promoted and applied in a clinical
setting.
Nutraceuticals and diet strategies do more than just
improve the quality of life for patients.As discussed,when
applied in combination with L-dopa drug therapy,
B-complex vitamins and vitamin C have positive effects,
including reduced side effects and enhanced absorption
of L-dopa. These nutraceuticals enhance the effect of contemporary
drug therapy and may allow for an attenuated
drug dosage, further reducing any dose-dependent side
effects. There is much potential in the positive synergistic
effects between nutraceuticals and clinical drug therapy.
Hence, instead of identifying the neuroprotective effects
of nutraceuticals alone, future research should focus on
the effects of nutraceuticals in combination with drug
therapy. Furthermore, enhanced drug therapy may be
developed through design and application of co-drugs
linking nutraceuticals and therapeutic drugs, e.g., by
linking stilbene compounds to L-dopa or even by linking
curcuminoids to L-dopa. This strategy of linking nutraceuticals
to drugs may contribute to new drug designs as
well as to more well-designed experimental studies and
clinical trials.
Nutraceuticals, though attractive and beneficial, are
still not the cure for PD. Experimental evidence is too
limited to enable the development of effective drugs from
nutraceuticals. Well-designed and placebo-controlled
human intervention trials are undoubtedly required to
confirm experimental findings. Many of the nutraceuticals
discussed in this review have been shown to be not
only preventative but also therapeutic for PD. Nonetheless,
there are still many unknowns, especially with regard
to the pharmacokinetics and pharmacodynamics of these
nutraceuticals, the effective intake dosage, and the exact
therapeutic target, all of which hinders their usage in a
clinical setting. High-quality research is needed to
promote the entry of more nutraceuticals into therapeutic
usage.