You are on page 1of 13

Journal of the American College of Nutrition

ISSN: 0731-5724 (Print) 1541-1087 (Online) Journal homepage: https://www.tandfonline.com/loi/uacn20

Alzheimer’s Disease and Parkinson’s Disease: A


Nutritional Toxicology Perspective of the Impact
of Oxidative Stress, Mitochondrial Dysfunction,
Nutrigenomics and Environmental Chemicals

Aayushi Agnihotri & Okezie I. Aruoma

To cite this article: Aayushi Agnihotri & Okezie I. Aruoma (2020) Alzheimer’s Disease and
Parkinson’s Disease: A Nutritional Toxicology Perspective of the Impact of Oxidative Stress,
Mitochondrial Dysfunction, Nutrigenomics and Environmental Chemicals, Journal of the American
College of Nutrition, 39:1, 16-27, DOI: 10.1080/07315724.2019.1683379

To link to this article: https://doi.org/10.1080/07315724.2019.1683379

Published online: 12 Dec 2019. Submit your article to this journal

Article views: 1901 View related articles

View Crossmark data Citing articles: 6 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=uacn20
JOURNAL OF THE AMERICAN COLLEGE OF NUTRITION
2020, VOL. 39, NO. 1, 16–27
https://doi.org/10.1080/07315724.2019.1683379

Alzheimer’s Disease and Parkinson’s Disease: A Nutritional Toxicology


Perspective of the Impact of Oxidative Stress, Mitochondrial Dysfunction,
Nutrigenomics and Environmental Chemicals
Aayushi Agnihotria and Okezie I. Aruomab
a
School of Medicine, The University of Manchester, Manchester, UK; bDepartment of Chemistry and Biochemistry, College of Natural and
Social Sciences, California State University Los Angeles, Los Angeles, California, USA

ABSTRACT KEYWORDS
Introduction: Alzheimer’s disease is primarily a dementia-related disorder from progressive cogni- Mitochondrial Dysfunction;
tive deterioration and memory impairment, while Parkinson’s disease is primarily a movement dis- Neuropsychiatric disorders;
order illness having movement disorder symptoms, bradykinesia (slowness of movements), metabolic disturbances;
Alzheimer’s disease;
hypokinesia (reduction of movement amplitude), and akinesia (absence of normal unconscious Parkinson’s disease;
movements) along with muscle rigidity and tremor at rest. While aging is the main risk factor, epi- Oxidative stress; Nutritional
demiological evidence suggests that the exposure to environmental toxicants, mainly pesticides, management;
metals and solvents could increase the risk of developing neurodegenerative conditions. Nutrigenomics;
Oxidative stress in neurodegenerative diseases: Mitochondria function impacts cell respiratory Nutraceuticals;
processes, metabolism, energy production, intracellular signaling, free radical production, and Environmental toxicants;
apoptosis. In neurodegenerative diseases, mitochondrial dysfunction is associated with a compro- Pesticides and
mised energy production, impaired calcium buffering, activation of proteases and phospholipases, neurodegeneration.
and increased oxidative stress. Oxidative stress induced microglial cells activation, protein aggrega- Xenobiotics and metabolism
tion, neuroinflammation and mitochondrial dysfunction lead to neuronal deaths in these disorders.
Role of nutrition: Neurodegenerative disease is not curable, but treatment is available to manage the
symptoms and slow down the disease progression. The drugs for treating these diseases only reduce
the cognitive impairment and behavioral problems, but do not stop the progression of neurodegener-
ation. Healthy diet, lifestyle improvement and nutraceuticals targeting of oxidative stress, inflamma-
tion, abnormal mitochondrial dynamics and the mitochondrial interaction with abnormal disease-
related proteins and assessment of impact of environmental contaminants including occupational
exposures to pesticides, can be a promising approach in the treatment of neurodegenerative diseases.
Conclusion: These innovations can be benchmarked on firm understanding of nutrigenomics and
the personalized management of individuals at risk.

Introduction disabling and irreversible disease has two recognizable hall-


marks, the first, familial or of early onset, is directly related
Neurodegenerative diseases are a group of chronic disabling
to specific gene mutations in the amyloid precursor protein
disorders, characterized by the gradual and selective loss of
(APP) and presenilin (PSEN) 1 and 2 genes, both related to
neurons in the central or peripheral nervous system. The
the amyloid-beta (Ab) peptide synthesis perpetuated by the
major age-related neurodegenerative diseases such as
Alzheimer’s disease (AD) and Parkinson’s disease (PD) can neurofibrillary tangles (NFT). The second hypothesis sug-
give rise to dementia, which is a leading cause of disability gests that the hyperphosphorylation of the Tau protein and
worldwide and represents a major public health burden with its subsequent deposition as NFTs is the ultimate responsible
spiraling healthcare costs (1,2). AD is primarily a dementia- for the disease. According to Chin-Chan, Navarro-Yepes, &
related disorder from progressive cognitive deterioration and Quintanilla-Vega (3), the amyloid cascade hypothesis estab-
memory impairment, while PD is primarily a movement dis- lishes that Ab aggregation initiates the brain damage leading
order illness having three major kinesia or movement dis- to memory loss and to AD. There is no cure for AD, and
order symptoms, bradykinesia (slowness of movements), therapeutic treatments are basically to ameliorate the symp-
hypokinesia (reduction of movement amplitude), and akin- toms. Therefore, an early and opportune diagnosis is indis-
esia (absence of normal unconscious movements) along with pensable to slow the progression of the disease. Currently
muscle rigidity and tremor at rest. Alzheimer’s disease is the (4), the determination of Tau and Ab levels in blood and
major form of dementia in the elderly and possibly contrib- CSF are broadly used for the diagnosis of AD, and several
utes to 60–70% of cases worldwide. (2). This progressive, medical tools are also used to confirm the diagnosis

CONTACT Okezie I. Aruoma oaruoma@calstatela.edu Department of Chemistry and Biochemistry, College of Natural and Social Sciences, California State
University Los Angeles, 5151 State University Drive, Los Angeles, CA 90032, USA.
Color versions of one or more of the figures in the article can be found online at www.tandfonline.com/uacn.
ß 2019 American College of Nutrition
JOURNAL OF THE AMERICAN COLLEGE OF NUTRITION 17

Figure 1. Innate Immune response and Tau pathology: a vicious circle. Hyperphosphorylated pathological Tau species can be secreted extracellularly, explaining
the progressive spread of tauopathy (A). Therefore, it promotes microglial activation/reactive astrocytes which release cytokines or neurotoxic inflammatory mole-
cules including IL1b or TNFa (B). By a modulation of Tau kinases (p38, cdk5 … ), glial activation enhances Tau pathology, self-perpetuating the detrimental circle
(C). Microglia may be involved in Tau propagation by releasing exosomal Tau once pathological Tau phagocytosed (D). (Adapted from Laurent, C., Buee, L., Blum, D.
(2018). Tau and neuroinflammation: What impact for Alzheimer’s disease and Tauopathies? Biomedical Journal, 41, 21–33).

including the medical history, mental status tests, and evalu- elevated levels of tau phosphorylation in AD patients
ations of the brain structure and function with neuroimag- (reviewed by 5).
ing techniques. Reference (5) has reviewed the concept of Mitochondria play important roles in cell respiratory
Tau and neuroinflammation exploring the impact this has processes, metabolism, energy production, intracellular sig-
on AD. As can be seen in Figure 1, this is a very complex naling, free radical production, and apoptosis. In neurodege-
process, more so because knowledge on AD can only be nerative diseases, mitochondrial dysfunction is associated
derived from postmortem samples. Such postmortem obser- with a compromised energy production, impaired calcium
vation of AD brains reveals a characteristic distribution pat- buffering, activation of proteases and phospholipases, and
tern of NFT lesions in the disease course, starting in the increased oxidative stress (6–8). Increased level of oxidative
transentorhinal cortex and progressively affecting the hippo- stress and accumulation of mitochondrial DNA mutations
campus, temporal cortex and polymodal association areas. resulting in mitochondrial dysfunction plays an important
The modalities of the propagation of Tau pathology have role in the aging process and the pathogenesis of many neu-
been investigated. Experimental transfer of brain homoge- rodegenerative diseases (9,10). Impaired calcium influx, dis-
nates containing pathologic Tau from transgenic P301S mice sipation of mitochondrial membrane potential, accumulation
promotes the accumulation of NFT in wild-type recipient of mutant proteins in mitochondria, increased accumulation
animals in a stereotypical and time-dependent fashion. Tau of mtDNA deficiencies, and deficiencies in mitochondrial
spreading occurs at distance of the injection site and con- oxidative phosphorylation are significant cellular changes in
cerns area connected anatomically instead of affecting prox- late-onset neurodegenerative diseases (11). Reference (12)
imal structures. Among the diverse modifications of tau, suggested that mitochondrial proteases in the inner mem-
phosphorylation induces the most critical change of tau brane perform the quality control surveillance by having
leading to tau aggregation. Full-length tau protein has 80 chaperone-like activities and degrading the misfolded pro-
serine and threonine residues and 5 tyrosine residues. In an teins and play a crucial role in the pathogenesis of many
intact cell, tau is constantly phosphorylated and dephos- neurodegenerative diseases. The m-AAA protease is a key
phorylated for the regulation of microtubule assembly. Once modulator of mitochondrial quality control system. Loss of
the balance is disrupted, tau is highly phosphorylated. The m-AAA protease promotes neurodegeneration through vari-
additional phosphates disrupt the charge balance between ous pathways, including decreased mitochondrial protein
tau and microtubules, and tau dissociates from microtu- synthesis due to impaired maturation of Mrpl32, accumula-
bules. Therefore, hyper-phosphorylation is the critical event tion of misfolded proteins, defective Opa1-mediated mito-
in the initiation of tau pathology. Tau phosphorylation is chondrial fusion, abnormal mitochondrial transport and
tightly regulated by kinases or phosphatases. Among diverse reduced assembly and stability of complexes I and III (12).
enzymes, GSK3b is the most effective tau kinase in the AD is the most common cause of dementia, characterized
human brain. Increased GSK3b activity is directly linked to by extracellular accumulation of amyloid-b (Ab) peptides in
18 A. AGNIHOTRI AND O. I. ARUOMA

the form of senile plaques and intracellular aggregation of Due to its structural similarity to MPP (the active metabolite
microtubules binding protein (tau) as tangles in the brain in of MPTP), it was thought to be toxic to dopaminergic neu-
the gray matter (13). The interaction between amyloid-b rons and thus might be related to PD. Paraquat can be taken
and hyperphosphorylated tau aggregates causes metabolic up into dopaminergic terminals by the dopamine transport
decline in the limbic structures such as the cingulate, orbito- and organic cation transporter 3 and have been reported to
frontal, and medial and basal temporal cortices resulting in cause cellular toxicity by oxidative stress. This may occur
imminent neurodegeneration (14). Substantial changes in through the cellular redox cycling generating superoxide
mitochondrial size and number in vulnerable neurons sug- radical by the oxidation of NADPH, which in turn impairs
gest a potential alteration in mitochondrial dynamics in AD the restauration of GSH levels and thus the activity of sev-
(15). Several studies revealed an increased expression of fis- eral antioxidant systems. Rotenone, an OP insecticide has
sion-related genes such as Drp1 and Fis1, and a decreased also been associated with an increased risk of PD. Rotenone
expression of fusion-related genes such as Mfn1, Mfn2 and can freely cross the blood–brain barrier and is a well-estab-
Opa1, leading to mitochondrial fragmentation in AD lished mitochondrial toxin that specifically inhibits the com-
patients. Overexpression of Drp1 interacts with Ab and plex I (NADH–dehydrogenase) of the electron transport
hyperphosphorylated tau proteins leading to mitochondrial chain leading to ATP depletion, energy failure and mito-
fragmentation and neuronal damage (16–18). chondrial ROS production, which in turn induces cyto-
PD is the second common age-related neurodegenerative chrome C release and apoptotic cell death. Rotenone
disease, which causes dementia in about 80% of people dur- treatment in animal models can reproduce features of PD
ing the disease (1). Parkinson’s disease dementia (PDD) and such as bradykinesia, postural instability and/or rigidity,
Dementia with Lewy bodies (DLB) are the two separate enti- reduces the tyrosine hydroxylase-positive neurons in the
ties of the aging population characterized by the similar substance nigra, induces a loss of striatal dopamine, and the
morphological features such as a-synuclein accumulation accumulation of a-syn and poly-ubiquitin positive aggregates
(Lewy bodies) in cortical and subcortical regions, but differ- in remaining dopaminergic neurons. Since rotenone recapit-
ent clinical profile at onset (19). Postmortem examination ulates several mechanisms of PD pathogenesis, this pesticide
reveals more pronounced cortical atrophy, and elevated cor- is currently used as a toxicological model to study the
tical and limbic Lewy pathologies in DLB as compared to underlying mechanisms on the PD development. Reference
PDD (19). The overexpression of wild type a-synuclein pro- (3) reviewed the use of insecticides as depicted in Figure 2.
motes neuronal death by inducing Drp-1 dependent mito- Their paper indicated that despite the widespread use of
chondrial fragmentation (20). Interestingly, about 90% of organophosphates (OP) insecticides such as malathion,
PD cases are sporadic and cannot be attributed only to gen- methyl parathion, chlorpyriphos and diazinon, not many
etic factors, which suggests that PD have a multifactorial eti- studies have evaluated the association between specific OP
ology. In addition to the aging, which is the main risk factor and PD risk. Solvents are widespread used due to their com-
for PD, epidemiological evidence suggests that the exposure mercial applications, including metal degreasing, dry clean-
to environmental toxicants, mainly pesticides, metals and ing, and as ingredients of paint thinners and detergents.
solvents could increase the risk of developing PD, and fac- Some solvents are lipophilic and thus easily absorbed by the
tors such as tobacco consumption can protect against PD central and peripheral nervous system tissues. There are iso-
development, as depicted in Figure 2 based on several toxi- lated cases of acute Parkinsonism associated with large solv-
cological data at molecular level and reviewed in (as ent exposures such as in workers exposed to n-hexane, and
reviewed in 3). The hypothesis that pesticide exposures may toluene, among others. There is no consistent evidence of
be related to PD development was prompted by the discov- the association of solvent exposure and PD.
ery that intravenous injection of 1-methy l-4 pheny l-1, 2, 3, Trichloroethylene (TCE) is one of the specific solvents that
6-tetrahydropyridine (MPTP), a by-product of the synthesis has been investigated in detail. Some clinical case reports
of heroin, developed a Parkinson syndrome clinically indis- have reported the onset of PD in workers exposed to TCE
tinguishable from PD and subsequent findings showed that through chronic inhalation and dermal exposure by han-
MPTP selectively damaged dopaminergic neurons in the dling TCE, suggesting a potential link between the exposure
substantia nigra (21,22). Environmental factors with similar to TCE and PD. The two major uses of trichloroethylene are
toxicological profiles have received attention as potential as a solvent to remove grease from metal parts and as a
risk factors for PD and AD (3). Dieldrin may cross the chemical that is used to make other chemicals, especially the
blood–brain barrier and remains in lipid-rich tissues such as refrigerant, HFC-134a. Trichloroethylene has also been used
the brain, and it has been shown that it is selectively toxic as an extraction solvent for greases, oils, fats, waxes, and
to dopaminergic neurons and could induce several of the tars; by the textile processing industry to scour cotton, wool,
pathologic mechanisms of PD including the depletion of and other fabrics; in dry cleaning operations; and as a com-
brain dopamine levels, increased ROS in nigral dopamin- ponent of adhesives, lubricants, paints, varnishes, paint
ergic neurons, inhibition of mitochondrial oxidative phos- strippers, pesticides, and cold metal cleaners. As there is no
phorylation that lead to a reduction of cellular ATP consistent evidence from either the toxicological or epide-
production, alteration of the mitochondrial membrane miologic perspective that any specific solvent or class of sol-
potential and cytochrome C release in animal models. vents is a cause of PD, toxicological research that addresses
Paraquat is a quaternary nitrogen herbicide used worldwide. mechanisms of nigral damage from TCE and its metabolites,
JOURNAL OF THE AMERICAN COLLEGE OF NUTRITION 19

Figure 2. Molecular mechanisms altered by environmental factors related to increased Parkinson’s disease risk. Exposure to environmental toxicants mainly pesti-
cides, metals and solvents may lead to the selective loss of dopaminergic neurons on the substantia nigra pars comp acta (SNpc) through the dysregulation/alter-
ation of the molecular mechanisms implicated on PD development such as mitochondrial dysfunction, impairment of protein quality pathways, microglia activation
and inflammation, which converge in the production of oxidative stress as the main factor in PD. (Copyright # 2015 Chin-Chan, Navarro-Yepes and Quintanilla-
Vega. Chin-Chan M, Navarro-Yepes J, Quintanilla-Vega B. Environmental pollutants as risk factors for neurodegenerative disorders: Alzheimer and Parkinson diseases.
Front Cell Neurosci. 9: 124, 2015).

with exposure routes and doses relevant to human expo- (32) appear to have a safer alternative treatment option to
sures, is recommended (3, 23). mitigate the neurodegenerative processes. Recent researches
Understanding the pathophysiology of neurodegeneration (33) show that the natural therapeutics with anti-oxidative
may improve the efforts to prevent and treat dementia in and anti-inflammatory properties, including caffeine, trigonel-
aged population. Today, no neurodegenerative is curable, and line, shogaol, curcumin, resveratrol, baicalein, wogonin, ginse-
the treatment is available to manage the symptoms and slow nosides, tanshinones, withanolides, picrosides, parthenolide,
down the disease progression (24). Acetylcholinesterase inhib- cannabinoids, Devil’s claw and white willow bark may play a
itors are used mainly to alleviate cognitive impairment and crucial role in protecting neurons due to their potential neu-
behavioral problems in both AD and PD, but display heavy roprotective, anti-inflammatory, antioxidant, anti-amyloido-
side effects such as nausea, vomiting, diarrhea, dizziness, con- genic and anticholinesterase activities (33,34).
fusion, and cardiac arrhythmias (25,26). Although target-bases
therapies such as stem cells (27), neuroprotective drugs (28),
Oxidative stress in neurodegenerative diseases
hormonal replacement therapy (29), neurotrophic factors (30)
and regulators of mRNA processing into disease-causing Several studies (35) have demonstrated the role of chronic
mutant proteins (31) have been developed, neutraceuticals oxidative stress in a common pathogenesis of age-related
20 A. AGNIHOTRI AND O. I. ARUOMA

neurodegenerative diseases such as AD and PD, and an anti- (DHA) in neuroprotection and reducing the risk of cogni-
oxidant therapy for the prevention and treatment of these tive decline (49). In addition to a healthy diet, cooking
diseases. Oxidative stress can induce mitochondrial DNA methods should also be considered. The cooking procedures
mutations, depress the mitochondrial respiratory chain func- can modify the original dietary ingredients, not only by
tions, alter membrane permeability, disturb Ca2þ homeosta- destroying the nutrients, but also by generating potential
sis, and damage mitochondrial antioxidant system (6,36,37). toxic compounds such as advanced glycation end products
A sustained increased production of ROS in mitochondria (AGEs) that contribute to the ageing process and neurode-
promotes calcium uptake by oxidizing thiol groups in pro- generative diseases (50). Advanced glycation end products
teins and initiates calcium overloaded-induced cell death (AGEs) play a crucial role in neurodegeneration by down
that might contribute to the pathogenesis of various neuro- regulating thioredoxin, which as an anti-oxidative and anti-
degenerative diseases (38). A classic mitochondrial free rad- apoptotic protein accelerates neuronal cells apoptosis (51).
ical theory of aging suggests that reactive oxygen species Recent studies (52–54) demonstrate the association
generated due to mitochondrial DNA (mtDNA) mutations between the Mediterranean-style diet (MsD) and age-related
cause persistent DNA damage leading to more mutations neurodegenerative diseases especially Alzheimer’s disease.
and give rise to further mitochondrial dysfunction and neu- According to the “Global Strategy on Diet, Physical Activity
rodegeneration (39,40). and Health”, MsD can prevent many diseases including neu-
Over production of reactive oxygen species (ROS) and/or
rodegenerative diseases and improve the quality of life (55).
a defect in the antioxidants systems lead to neuronal loss in
MsD includes plenty of plant-based foods (such as fruits,
majority of the neurodegenerative diseases. Recently, a
vegetables, whole grains, legumes and nuts), a moderate to
superoxide anion (O2-) generating enzyme i.e. NADPH oxi-
high intake of fish and sea food, a low to moderate intake
dase 2 (Nox2) has been considered as a major source of age-
of poultry, eggs, cheese and yogurt, low consumption of red
ing-related vascular endothelial oxidative damage in
neurodegenerative diseases (41). Under normal metabolic meat, and a regular intake of red wine during the meals. A
conditions, mitochondria convert about 1-5% oxygen into systemic review of the thirty-one population-based studies
ROS mainly by complex I (NADH dehydrogenase) and by (52) from Australia, China, France, Greece, Scotland,
complex III (cytochrome oxidoreductase) (42). Once gener- Spain, Sweden, and USA shows that MsD has the beneficial
ated, superoxide O2.- is dismutated to H2O2 by superoxide effects on cognitive function and reduces the risk of
dismutase, and afterwards, H2O2 is fully reduced to water by Alzheimer’s disease and dementia. Similarly, another review
GPX (43). The damage and inactivation of Fe-S clusters of of twenty-four observational-type studies (prospective and
electron transport chain complexes (I, II, and III) (ETC) retrospective) by (54) from Australia, Canada, Finland,
and Krebs cycle aconitase due to acute ROS exposure can France, Japan, Norway and USA summarizes the impact of
shut down the energy production in mitochondria leading MsD on cognition and in Alzheimer’s disease in human
to cell death (44). Neurons are extremely dependent on the clinical trials. Reference (54) further add that the bioactive
ETC complexes of mitochondria, and impaired functions of components of MsD have both neuroprotective and neuro-
these complexes are related to several neurodegenerative dis- rescue effects not only through antioxidant activity but also
eases including AD and PD (45). Reduced ETC complex I, due to anti-inflammatory, and anti-amyloidogenic activities.
III and IV activities have been found in platelets, lympho- A recent population-based cohort study (56) of older adults
cytes and postmortem brain tissue of patients with in Greece suggests that the adherence to MsD is associated
Alzheimer’s disease, however major change association with with a lower probability of prodromal Parkinson’s disease in
complex IV (46). In Parkinson’s disease, a mild deficiency of elderly people.
complex I and NADH dehydrogenase has been demon-
strated in substantia nigra, platelets and the skeletal
muscles (47). Components of mediterranean-style diet and their
impact on neurodegeneration

Role of nutrition in neurodegenerative diseases Vegetables and fruits

Recently, the role of diet and nutrition in preventing or An adequate intake of vegetable and fruits particularly flavo-
delaying the progression of neurodegeneration in aged noids and carotenoids prevent or delays the age-related cog-
population has received a great amount of attention. nitive decline and reduces the risk of developing
Reference (48) reported that consumption of diet rich in neurodegenerative diseases (57). These beneficial effects are
antioxidants (such as fruits, nuts, vegetables and spices), not only due to three major antioxidant vitamins such as
reduced caloric intake by consuming less carbohydrate and a-tocopherol (vitamin E), ascorbic acid (vitamin C), or
low consumption of alcohol may lower age-relative cognitive b-carotene (vitamin A), but also because of having a large
decline and lessen the risk of developing neurodegenerative amount of non-vitamin antioxidants such as polyphenols
disease. Emerging evidence supports the role of B-vitamins and anthocyanins (58). Researches (59,60) show that some
(vitamin B12, vitamin B6 and riboflavin), vitamin D, folate, specific vegetable and fruits especially green leafy and nuts
polyphenols, and n-3 polyunsaturated fatty acids (PUFAs) – are strongly associated with a reduced rate of age-related
eicosapentaenoic acid (EPA) and docosahexaenoic acid cognitive decline.
JOURNAL OF THE AMERICAN COLLEGE OF NUTRITION 21

Table 1. Summary of nutraceuticals-targeting age-related neurodegenerative diseases.


S.N. Nutraceuticals Target mechanism Authors (Years)
1. Curcumin Suppress increased production of intracellular ROS (74,75)
Inhibit pro-inflammatory signaling through NF-jB
Inhibit aggregation of misfolded proteins (b-amyloid, a-synuclein)
2. Anthocyanins (Blue berry extract) Improve beta-amyloid induced neurotoxicity by inhibiting calcium influx and (76)
tau hyperphosphorylation.
Modulate inflammatory genes expression.
3. Coenzyme Q10 supplements Protect mitochondrial impairment and oxidative stress (77)
4. Aged garlic extract (AGE) Modulates Nrf2-mediated signaling as well as other oxidative stress- (78)
related pathways.
5. Astaxanthin (Xanthophyll carotenoid) Attenuates microglial activation and the release of pro-inflammatory cytokines. (79,80)
Promotes hippocampal neurogenesis in dose-dependent manner.
6. Epigallocatechin-3-gallate (Green tea) Reduces oxidative stress, regulates signaling pathways and metal chelation (81,82)
7. Alfa-lipoic acid Displays antioxidant, antiapoptotic, and anti-inflammatory properties (83)
8. Vitamin B supplements Improve cognition by lowering homocysteine levels (84)
9 Lycopene Increases glutathione peroxidase and markedly reduces tau (85)
hyperphosphorylation
Has synergistic antioxidant effect with Vitamin E

Seafood Red wine


Seafood is considered to be a highly nutritious food given Wine is an alcoholic beverage commonly made from fer-
its high protein and low-calorie content and associated with mented grapes. Reference (69) have reported that moderate
numerous health benefits. Researchers (61) are interested in consumption of red wine reduces the risk of many diseases
omega-3-polyunsaturated fatty acids, such as eicosapenta- and protects against various age-related neurodegenerative
enoic acid (EPA) and docosahexsaenoic acid (DHA) con- diseases such as Parkinson’s or Alzheimer’s diseases. The
tained in fish and seafood, which are required to maintain neuroprotective effects of red wine are mainly due to the
the structural integrity of neuronal membranes. These fatty presence of polyphenols such as quercetin, myricetin, cate-
acids may decrease with aging that increases the risk of cog- chins, tannins, anthocyanidins, resveratrol, and ferulic acid
nitive impairment and dementia. (62) reported that con- (70). These polyphenols induce endogenous antioxidant
sumption of seafood at least one serving a week results in Defense mechanism by modulating transcription factors
less decline in cognitive functions in elderly people. A cross- such as the nuclear factor erythroid 2-related factor 2 (Nrf2)
sectional analysis of older adults with brain neuropathology and improve cognitive functions (71).
suggests that moderate seafood consumption is correlated
with lesser Alzheimer disease neuropathology. Importantly,
a prospective cohort study by (63) of 5,988 women over a A nutrigenomics perspective and emerging
follow-up period of 4 years concluded that the relationship nutraceuticals targeting neuroprotection
of seafood to cognition might depend on the seafood types, Nutrigenomics is the study of the relationship among nutri-
not on the total consumption. The consumption of tuna and tion, genome and health that examines how different foods
dark-meat fish once weekly or more is strongly associated interact with our gene and how the people respond to food
with lower cognitive decline (63). based on their genetic distinctions. Evidence suggests the
role of nutrition in the genetics of many neurodegenerative
diseases. Epigenetics suggest the involvement of nutrition
Olive oil
in the pathogenesis of age-related cognitive decline.
The fatty acid composition (mainly oleic acid - a monoun- Nutraceuticals can modulate the specific metabolic pathways
saturated omega-9 fatty acid), vitamins and polyphenols in that affect the development of age-related neurodegenera-
olive oil are responsible for longevity, and beneficial for cog- tive diseases.
nitive deficit in age-related neurodegenerative diseases (64). Nutraceuticals are the fortified food products that not
Olive oil has more than 30 phenolic compounds including only supplement the diet but also assist in treating and/or
the three major variants oleuropein, hydroxytyrosol and preventing diseases and disorders (72). Neutraceuticals pro-
oleocanthal (65). Oleocanthal, a phenolic component of tect neuronal damage by targeting the four major pathways
extra-virgin olive oil, enhances b-amyloid clearance by up- – mitochondrial dysfunction, calcium overload, oxidative
regulating major b-amyloid transport proteins (P-glycopro- stress and inflammation (72). Many neutraceuticals, which
tein and LDL lipoprotein receptor related protein-1) at the have antioxidant effects by scavenging ROS inducing the
blood-brain barrier (BBB) (66), inhibits the aggregation of expression of a variety of cytoprotective and detoxification
tau protein aggregation into neurofibrillary tangles (67), and proteins in the Nrf2/ARE pathway, have been identified as a
thus reduces a risk of Alzheimer’s disease. Oleuropein potential therapeutic target for neurodegenerative diseases
Aglycone, another polyphenol present in olive oil, hinders (73). We have categorized sample nutraceuticals-targeting
b-amyloid and tau proteins aggregation, autophagy neuroprotection based on the available evidences from the
impairment, and neuroinflammation (68). published scientific literature (Table 1).
22 A. AGNIHOTRI AND O. I. ARUOMA

Figure 3. Environmental factors associated with Alzheimer’s disease development through different mechanisms. Several factors including metals, pesticides, nano-
particles, and diet can affect the two targets of AD such as Ab generation and Tau phosphorylation. The figure depicts the molecular targets than can be modified
at different levels following the amyloid hypothesis that ends in Ab senile plaques formation (upper part) or the hyperphosphorylation of Tau protein and its subse-
quent deposition as neurofibrillary tangles (NFTs) (lower part). (Copyright # 2015 Chin-Chan, Navarro-Yepes and Quintanilla-Vega. Chin-Chan M, Navarro-Yepes J,
Quintanilla-Vega B. Environmental pollutants as risk factors for neurodegenerative disorders: Alzheimer and Parkinson diseases. Front Cell Neurosci. 9: 124, 2015).

Influence of the environmental contaminants and organic hazards, exposure to toxic metals (aluminum, cop-
lifestyle on neurodegenerative diseases per), pesticides (organochlorine and organophosphate insec-
ticides), industrial chemicals (flame retardants) and air
Lifestyle factors are crucial in minimizing the risk of devel- pollutants (particulate matter). The association between past
oping age-related neurodegenerative diseases. Continuous occupational exposure to pesticides and low cognitive per-
physical and cognitive activity, a balanced or Mediterranean- formance, together with an increase in the risk of developing
style diet with a high proportion of polyunsaturated fatty Alzheimer’s disease or Parkinson’s disease has been reported
acids, proper treatment of hypertension, control diabetes (88,89) and that this may be related to occupational expos-
and cholesterol, and adequate sleep reduce the risk of devel- ure such as vineyards. The possibility of long-term neuro-
oping dementia later in life (86). Given that dietary supple- logic effects of pesticide use in agricultural settings where
ments may fill a void by providing necessary nutrients for fungicides account for much of the occupational exposure
optimizing health, enhancing micronutrient and vitamin lev- (3,88,89) can be paralleled with long term exposures to other
els may be a more pragmatic approach to address stress and environmental contaminants together with bioaccumulation
improve psychosocial mood than pharmacological interven- over an individual’s lifetime. The reader is referred to the
tions with their associated negative side effects (87). paper by (89) which eminently discussed five categories of
Environmental factors possibly include inorganic and
JOURNAL OF THE AMERICAN COLLEGE OF NUTRITION 23

environmental agents, including (i) toxic metals, (ii) insecti- to Pb during development is a good example of an environ-
cides/pesticides, (iii) industrial/commercial pollutants, (iv) mental contaminant as a risk factor to promote neurodege-
antimicrobials and (v) air pollutants, all known or hypothe- nerative diseases such as AD, supporting the hypothesis that
sized to induce or aggravate AD or AD-like progression many adult diseases have a fetal origin. Mercury (Hg) is a
in vitro, in animal models and in human research subjects. heavy metal with a high potential to cause neurotoxicity.
The cognitive disturbances that may persist in occupation- Early studies about Minamata and Iraq disasters led us to
ally exposed subjects, even long after work cessation, may be understand the neurotoxicity of this metal. It is widely
aligned with neuroinflammation and neuropathology paving accepted that Hg disrupts the brain development and produ-
the way for developing AD. While epidemiologic associa- ces cognitive and motor disabilities, and in adults, Hg
tions between environmental contaminant exposures and exposure produces memory loss and cognitive alterations.
AD are still limited, data from many in vitro and animal The ability of Hg to increase Ab levels has been studied
studies have identified toxic effects of environmental con- in vitro and in vivo, and the suggested mechanisms include
taminants at the cellular level, revealing alterations of path- an increased production, a reduced degradation and/or a
ways and metabolisms associated with AD pathogenesis of diminished brain clearance of the peptide. Inorganic arsenic
neuropsychiatric disorders (3,89,90). Neurotoxic metals such (As) is a known neurotoxic metalloid with adverse effects in
as lead, mercury, aluminum, cadmium and arsenic, as well both the neurodevelopment and cognitive function, although
as some pesticides and metal-based nanoparticles have been its effects in elderly have been less studied. A plausible
involved in AD due to their ability to increase beta-amyloid mechanism for the cognitive and memory alterations
(Ab) peptide and the phosphorylation of Tau protein (P- induced by As exposure is by interfering with the amyloid
Tau), causing senile/amyloid plaques and neurofibrillary tan- pathway. The mechanism by which As causes Ab overpro-
gles (NFTs) characteristic of AD. The exposure to lead, duction has not been determined, but As exposure has been
manganese, solvents and some pesticides has been related to associated with brain inflammatory responses and oxidative
hallmarks of PD such as mitochondrial dysfunction, altera- stress, which is in agreement with the inflammatory and oxi-
tions in metal homeostasis and aggregation of proteins such dative hypotheses of AD. Cadmium is another toxic heavy
as a-synuclein (a-syn), which is a key constituent of Lewy
metal associated with neurological alterations including
bodies (LB), a crucial factor in PD pathogenesis (3,89–92).
memory loss and mental retardation which may be linked to
This is an intriguing area for future research, particularly
Ab overproduction. Aluminum is a neurotoxic element
drawing on how nutrition can play a role in modulating
involved in the etiology of neurodegenerative disorders such
cognitive decline in the face of advancing pathophysiology
as AD; however, there is no consistent evidence. Reference
and mitochondrial dysfunction and cognitive decline.
(91) reported that chronic oral Al administration in rats
Individuals have variations in the composition of their gen-
(20 g/day in the food/twice weekly from 6 months of age to
etic characteristics (factored on strategies that embrace test-
the end of their lives) increases the Ab production by rais-
ing for candidate-genes and genome-wide association) that
ing the levels of APP in hippocampal and cortical tissues
will affect the availability of functional proteins which ultim-
ately impacts functional homeostasis and the outcome of (brain regions important for the memory process). The asso-
drug therapy. The brain reflex-receptor mechanism in sig- ciation between chronic pesticide exposure and the preva-
naling for biomarkers and availability of enzymes for metab- lence of dementias, including AD has not been as well
olism is of critical importance here. If this is altered, cellular studied as with other environmental risk factors, and results
homeostasis will be altered. The context of Figure 3 is edu- are often inconsistent. This is mainly because the difficulty
cative. Underlying mechanisms are multiple but can be sum- on getting adequate data on the levels of exposure of indi-
marized as a toxic gain of function including Tau vidual pesticides, which is often indirectly evaluated by
aggregation, release of inflammatory molecules and a loss of structure questionnaires (exposure index). The role of pesti-
physiological functions such as adult neurogenesis, glial cides in alterations observed in cognitive functions and AD
phagocytosis and trophic factor release, Tau-driven axonal has been suggested based on epidemiological studies, but the
transport and RNA/DNA integrity. Also, environmental dis- mechanisms have been poorly explored. Interesting chlor-
ease modifiers (for instance obesity, stress, caffeine con- pyrifos (CPF), an organophosphate insecticide associated
sumption) can act through an alteration of the with cognitive impairment, oxidative stress and neuronal
neuroimmune response and impact upon the vicious circle damage caused a significant increase in Ab levels in the cor-
(3,5,89,90). Environmental factors such as diet (fat-rich), tex and hippocampus, as well as increased memory loss and
heavy metals, biogenic metals and pesticides have been reduced motor activity in Tg2576 mice 6 months after an
involved in AD development due to their ability to disrupt acute subcutaneous administration of 50 mg/Kg of CPF (92).
metabolic pathways involved in the homeostasis of Ab But the work of reference (93) did not find increased Ab
(Figure 3). The following summaries are instructive. Lead levels nor significant changes in memory acquisition in
(Pb) is a heavy metal well known for its neurological toxic Tg2576 mice treated with CPF (25 mg/Kg/twice weekly/
effects, although a direct association with AD development 4 weeks, intragastric) and analyzed 6 months later. This indi-
has not been reported. Pb affects cognitive abilities, intelli- cates that more studies are needed to better understand the
gence, memory, speed processing and motor functions in mechanisms by which organochlorines and organophos-
children, while studies in the elderly are limited. Exposure phates and other insecticides are linked to AD and PD.
24 A. AGNIHOTRI AND O. I. ARUOMA

Paraquat (PQ), is a commonly used herbicide that has neurodegenerative disorders in vitro and in animal models
been suggested to be involved in AD development (Figure but does not adequately demonstrate the neuroprotection
2). One study showed that treatment of wild type and APP efficacy in patients due to the chemical instability of antioxi-
transgenic (Tg2576) mice with PQ (10 mg/Kg/twice a week/ dant. Modifying abnormal mitochondrial dynamics and tar-
3 weeks) produced a significant increase in Ab levels that geting mitochondrial interaction with disease-specific
was associated with mitochondrial oxidative damage in cere- proteins can be the promising strategies to treat neurodege-
bral cortex leading to the impairment of learning and mem- nerative diseases. Diet and lifestyle changes are thought to
ory (4). The amyloid cascade hypothesis of Alzheimer’s slow down the progression of neurodegeneration and cogni-
disease envisages that the initial elevation of amyloid b-pep- tive impairment at a later age. Dietary secondary metabolites
tide (Ab) levels, especially of Ab1-42, is the primary trigger have neuroprotective effects due to their, potential antioxi-
for the neuronal cell death specific to onset of Alzheimer’s dant activities, abilities to suppress inflammation, minimize
disease. There is now substantial evidence that brain amyl- protein aggregation and to modulate several signaling path-
oid levels can be manipulated because of a dynamic equilib- ways. In addition to healthy diet, we should educate the
rium between their synthesis from the amyloid precursor people about food processing (cooking) in order to get max-
protein and their removal by amyloid-degrading enzymes imum benefit from the food nutrients. Further studies are
(ADEs) providing a potential therapeutic strategy. Factors needed to find a define the link of natural bioactive com-
such as lifestyle (antioxidants and exercise) can prevent AD pounds to prevent neurodegenerative diseases due to envir-
development. The brain is particularly vulnerable to oxida- onmental exposure of toxicants and begin understanding
tive stress due to its high glucose-based metabolic rate, low how nutrigenomics can foster the understanding of the indi-
levels of antioxidants, high levels of polyunsaturated fatty vidual variabilities.
acids, and high enzymatic activities related to transition
metals that catalyze the formation of free radicals increasing
the oxidative burden. The identification and validation of Disclosure statement
accurate biomarkers of individual responses to environmen- Dr Okezie I. Aruoma is a member of the Scientific Advisory Board for
tal toxicants, pharmaceutical agent (prescription drugs) or Zurvita, Houston, Texas, USA.
biological secondary metabolites in foods remain prerequis-
ite conditions ascribed to the development of personalized
medicine benchmarked on nutrigenomics, toxicogenomics References
and pharmacogenomics as well as other evolving therapeutic 1. Russell A, Drozdova A, Wang W, Thomas M. The impact of
strategies (94). The sequence variations in the genes for pro- dementia development concurrent with Parkinson’s disease: a
teins involved in xenobiotics (from drugs, environmental new perspective. CNSNDDT. 2014;13:1160–1168. doi:10.2174/
1871527313666140917122739.
toxicants to dietary supplements etc.) as discussed in this
2. World Alzheimer Report. 2015. The global voice on dementia.
paper on disposition can alter the pharmacokinetics of the https://www.alz.co.uk/research/WorldAlzheimerReport2015.pdf
foreign agent, while sequence variations in target genes for 3. Chin-Chan M, Navarro-Yepes J, Quintanilla-Vega B.
pharmacologic actions can change the pharmacodynamics of Environmental pollutants as risk factors for neurodegenerative
the xenobiotics. disorders: Alzheimer and Parkinson diseases. Front Cell
Neurosci. 2015;9:124. doi:10.3389/fncel.2015.00124.
4. Lewczuk P, Mroczko B, Fagan A, Kornhuber J. Biomarkers of
Conclusion Alzheimer’s disease and mild cognitive impairment: a current
perspective. Adv Med Sci. 2015;60(1):76–82. doi:10.1016/j.advms.
Mitochondrial dysfunction and the aggregation of misfolded 2014.11.002.
proteins are the key features of age-related neurodegenera- 5. Laurent C, Buee L, Blum D. Tau and neuroinflammation: What
tive diseases such as AD and PD. The overproduction of impact for Alzheimer’s disease and Tauopathies? Biomed J. 2018;
41(1):21–33. doi:10.1016/j.bj.2018.01.003.
ROS and/or suppression of antioxidant systems in mito-
6. Aruoma OI. Free radicals, oxidative stress, and antioxidants in
chondria can induce severe oxidative stress, which plays an human health and disease. J Amer Oil Chem Soc. 1998;75(2):
important role in the pathogenesis of neurodegeneration. 199–212. doi:10.1007/s11746-998-0032-9.
Maintenance of the major antioxidant systems predomin- 7. Davis RE, Williams M. Mitochondrial function and dysfunction:
antly superoxide dismutase and glutathione peroxidase are an update. J Pharmacol Exp Ther. 2012;342(3):598–607. doi:10.
crucial for normal function of the mitochondria and limits 1124/jpet.112.192104.
8. Z€undorf G, Reiser G. Calcium dysregulation and homeostasis of
the amount of oxidative damage. Modification in the anti- neural calcium in the molecular mechanisms of neurodegenera-
oxidant systems is an attractive therapeutic strategy for the tive diseases provide multiple targets for neuroprotection.
treatment of various neurodegenerative diseases. Vitamin E Antioxid Redox Signal. 2011;14:1275–1288. doi:10.1089/ars.2010.
(a major scavenger of lipid peroxidation in brain), vitamin 3359.
C (an intracellular reducing molecule), and CoQ10 (a coen- 9. Cha MY, Kim DK, Mook-Jung I. The role of mitochondrial
zyme that transfers electrons from complexes I and II to DNA mutation on neurodegenerative diseases. Exp Mol Med.
2015;47:e150. doi:10.1038/emm.2014.122.
complex III, optimizing the proton gradient of the oxidative 10. Udhayabanu T, Manole A, Rajeshwari M, Varalakshmi P,
phosphorylation mechanisms) are commonly studied antiox- Houlden H, Ashokkumar B. Riboflavin responsive mitochondrial
idants for the therapeutic purpose. Although antioxidant dysfunction in neurodegenerative diseases. JCM. 2017;6(5):52.
administration has been found to effective in number of pii:doi:10.3390/jcm6050052.
JOURNAL OF THE AMERICAN COLLEGE OF NUTRITION 25

11. Golpich M, Amini E, Mohamed Z, Azman-Ali R, Ibrahim NM, 29. Dye RV, Miller KJ, Singer EJ, Levine AJ. Hormone replacement
Ahmadiani A. Mitochondrial dysfunction and biogenesis in therapy and risk for neurodegenerative diseases. Int J
neurodegenerative diseases: Pathogenesis and treatment. CNS Alzheimer’s Dis. 2012;2012:1. doi:10.1155/2012/258454.
Neurosci Ther. 2017;23:5–22. doi:10.1111/cns.12655. 30. Weissmiller AM, Wu C. Current advances in using neurotrophic
12. Langer L. AAA proteases: cellular machines for degrading mem- factors to treat neurodegenerative disorders. Transl
brane proteins. Trends Biochem Sci. 2000;25(5):247–251. doi:10. Neurodegener. 2012;1(1):14. doi:10.1186/2047-9158-1-14.
1016/S0968-0004(99)01541-8. 31. Romano M, Buratti E. Targeting RNA binding proteins involved
13. Scheltens P, Blennow K, Breteler MM, de Strooper B, Frisoni in neurodegeneration. J Biomol Screen. 2013;18(9):967–983. doi:
GB, Salloway S, Van der Flier WM. Alzheimer’s disease. Lancet. 10.1177/1087057113497256.
2016;388(10043):505–517. doi:10.1016/S0140-6736(15)01124-1. 32. Dadhania VP, Trivedi PP, Vikram A, Tripathi DN.
14. Pascoal TA, Mathotaarachchi S, Shin M, Park AY, Mohades S, Nutraceuticals against neurodegeneration: a mechanistic insight.
Benedet AL, Kang MS, Massarweh G, Soucy J-P, Gauthier S, Curr Neuropharmacol. 2016;14:627–640. doi:10.2174/
Rosa-Neto P. Amyloid and tau signatures of brain metabolic 1570159X14666160104142223.
decline in preclinical Alzheimer’s disease. Eur J Nucl Med Mol 33. Parvez MK. Natural or plant products for the treatment of
Imaging. 2018;45(6):1021–1030. doi:10.1007/s00259-018-3933-3. neurological disorders: current knowledge. CDM. 2018;19(5):
15. Cai Q, Tammineni P. Alterations in mitochondrial quality 424–428. doi:10.2174/1389200218666170710190249.
control in Alzheimer’s disease. Front Cell Neurosci. 2016;10:24. 34. Deshpande P, Gogia N, Singh A. Exploring the efficacy of nat-
doi:10.3389/fncel.2016.00024. ural products in alleviating Alzheimer’s disease. Neural Regen
16. Wang X, Su B, Lee HG, Li X, Perry G, Smith MA, Zhu X. Res. 2019;14:1321–1329. doi:10.4103/1673-5374.253509.
Impaired balance of mitochondrial fission and fusion in 35. Kim GH, Kim JE, Rhie SJ, Yoon S. The role of oxidative stress
Alzheimer’s disease. J Neurosci. 2009;29(28):9090–9103. doi:10. in neurodegenerative diseases. Exp Neurobiol. 2015;24(4):
1523/JNEUROSCI.1357-09.2009. 325–340. doi:10.5607/en.2015.24.4.325.
17. Manczak M, Calkins MJ, Reddy PH. Impaired mitochondrial 36. Guo C, Sun L, Chen X, Zhang D. Oxidative stress, mitochondrial
dynamics and abnormal interaction of amyloid beta with mito- damage and neurodegenerative diseases. Neural Regen Res. 2013;
chondrial protein Drp1 in neurons from patients with 8(21):2003–2014.
Alzheimer’s disease: implications for neuronal damage. Human 37. Halliwell B, Gutteridge JM, Cross CE. Free radicals, antioxidants
Mol Genet. 2011;20(13):2495–2509. and human disease: where are we now? J Lab Clin Med. 1992;
18. Manczak M, Reddy PH. Abnormal interaction between the mito- 119(6):598–620.
chondrial fission protein Drp1 and hyperphosphorylated tau in 38. G€orlach A, Bertram K, Hudecova S, Krizanova O. Calcium and
Alzheimer’s disease neurons: implications for mitochondrial dys- ROS: A mutual interplay. Redox Biol. 2015;6:260–271. doi:10.
function and neuronal damage. Human Mol Genet. 2012;21(11): 1016/j.redox.2015.08.010.
2538–2547. doi:10.1093/hmg/dds072. 39. Angelova PR, Abramov AY. Role of mitochondrial ROS in the
19. Jellinger KA, Korczyn AD. Are dementia with Lewy bodies and brain: from physiology to neurodegeneration. FEBS Lett. 2018;
Parkinson’s disease dementia the same disease? BMC Med. 2018; 592(5):692. doi:10.1002/1873-3468.12964.
16(1):34. doi:10.1186/s12916-018-1016-8. 40. Maynard S, Fang EF, Scheibye-Knudsen M, Croteau DL, Bohr
20. Martinez JH, Alaimo A, Gorojod RM, Porte Alcon S, Fuentes F, VA. DNA damage, DNA repair, aging, and neurodegeneration.
Coluccio Leskow F, Kotler ML. Drp-1 dependent mitochondrial Cold Spring Harb Perspect Med. 2015;5(10):a025130. pii: doi:10.
fragmentation and protective autophagy in dopaminergic SH- 1101/cshperspect.a025130.
SY5Y cells overexpressing alpha-synuclein. Mol Cellular 41. Cahill-Smith S, Li JM. Oxidative stress, redox signalling and
Neurosci. 2018;88:107–117. doi:10.1016/j.mcn.2018.01.004. endothelial dysfunction in ageing-related neurodegenerative dis-
21. Langston JW, Ballard PA. Jr. Parkinson’s disease in a chemist eases: a role of NADPH oxidase 2. Br J Clin Pharmacol. 2014;
working with 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine. N 78(3):441–453. doi:10.1111/bcp.12357.
Engl J Med. 1983;309(5):310. doi:10.1056/nejm198308043090511. 42. Ghezzi D, Zeviani M. Assembly factors of human mitochondrial
22. Langston JW, Forno LS, Rebert CS, Irwin I. Selective nigral tox- respiratory chain complexes: physiology and pathophysiology.
icity after systemic administration of 1-methyl-4-phenyl-1,2,5,6- Adv Exper Med Biol. 2012;748:65–106.
tetrahydropyrine (MPTP) in the squirrel monkey. Brain Res. 43. Li X, Fang P, Mai J, Choi ET, Wang H, Yang XF. Targeting
1984;292:390–394. doi:10.1016/0006-8993(84)90777-7. mitochondrial reactive oxygen species as novel therapy for
23. Lock EA, Zhang J, Checkoway H. Solvents and Parkinson dis- inflammatory diseases and cancers. J Hematol Oncol. 2013;6:19.
ease: A systematic review of toxicological and epidemiological doi:10.1186/1756-8722-6-19.
evidence. Toxicol Appl Pharmacol. 2013;266(3):345–355. doi:10. 44. Almeida AM, Bertoncini CR, Borecky J, Souza-Pinto NC,
1016/j.taap.2012.11.016. Vercesi AE. Mitochondrial DNA damage associated with lipid
24. Dur~aes F, Pinto M, Sousa E. Old drugs as new treatments for peroxidation of the mitochondrial membrane induced by Fe2þ-
neurodegenerative diseases. Pharmaceut (Basel). 2018;11(2):44. citrate. Acad Bras Ci^enc. 2006;78(3):505–514. doi:10.1590/S0001-
doi:10.3390/ph11020044. 37652006000300010.
25. Pagano G, Rengo G, Pasqualetti G, Femminella GD, Monzani F, 45. Johri A, Beal M. Mitochondrial dysfunction in neurodegenerative
Ferrara N, Tagliati M. Cholinesterase inhibitors for Parkinson’s diseases. J Pharmacol Exp Ther. 2012;342(3):619–630. doi:10.
disease: a systematic review and meta-analysis. J Neurol 1124/jpet.112.192138.
Neurosurg Psychiatry. 2015;86(7):767–773. doi:10.1136/jnnp- 46. Cardoso SM, Proença MT, Santos S, Santana I, Oliveira CR.
2014-308764. Cytochrome c oxidase is decreased in Alzheimer’s disease plate-
26. Winslow BT, Onysko MK, Stob CM, Hazlewood KA. Treatment lets. Neurobiol Aging. 2004;25(1):105–110. doi:10.1016/s0197-
of Alzheimer disease. Am Family Phys. 2011;12:1403–1412. 4580(03)00033-2.
27. Dantuma E, Merchant S, Sugaya K. Stem cells for the treatment 47. Gatt AP, Duncan OF, Attems J, Francis PT, Ballard CG,
of neurodegenerative diseases. Stem Cell Res Ther. 2010;1:37. Bateman JM. Dementia in Parkinson’s disease is associated with
doi:10.1186/scrt37. enhanced mitochondrial complex I deficiency. Mov Disord.
28. Dunkel P, Chai CL, Sperlagh B, Huleatt PB, Matyus P. Clinical 2016;31(3):352–359. doi:10.1002/mds.26513.
utility of neuroprotective agents in neurodegenerative diseases: 48. Joseph J, Cole G, Head E, Ingram D. Nutrition, brain aging, and
current status of drug development for Alzheimer’s, Parkinson’s neurodegeneration. J Neurosci. 2009;29(41):12795–12801. doi:10.
and Huntington’s diseases, and amyotrophic lateral sclerosis. 1523/JNEUROSCI.3520-09.2009.
Expert Opin Invest Drugs. 2012;9:1267–1308. doi:10.1517/ 49. Moore K, Hughes CF, Ward M, Hoey L, McNulty H. Diet, nutri-
13543784.2012.703178. tion and the ageing brain: current evidence and new directions.
26 A. AGNIHOTRI AND O. I. ARUOMA

Proc Nutr Soc. 2018;77(2):152–163. doi:10.1017/ 68. Martorell M, Forman K, Castro N, Cap o X, Tejada S, Sureda A.
S0029665117004177. Potential therapeutic effects of oleuropein aglycone in
50. Abate G, Marziano M, Rungratanawanich W, Memo M, Uberti Alzheimer’s disease. Curr Pharmaceut Biotechnol. 2016;17(11):
D. Nutrition and AGE-ing: Focusing on Alzheimer’s Disease. 994–1001. doi:10.2174/1389201017666160725120656.
Oxid Med Cellular Long. 2017;2017:1. doi:10.1155/2017/7039816. 69. Basli A, Soulet S, Chaher N, Merillon JM, Chibane M, Monti JP,
51. Ren X, Ma H, Qiu Y, Liu B, Qi H, Li Z, Kong H, Kong L. The Richard T. Wine polyphenols: potential agents in neuroprotection.
downregulation of thioredoxin accelerated Neuro2a cell apop- Oxid Med Cellular Long. 2012;2012:1. doi:10.1155/2012/805762.
tosis induced by advanced glycation end product via activating 70. Caruana M, Cauchi R, Vassallo N. Putative role of red wine pol-
several pathways. Neurochem Int. 2015;87:128–135. doi:10.1016/ yphenols against brain pathology in Alzheimer’s and Parkinson’s
j.neuint.2015.06.009. disease. Front Nutr. 2016;3:31.
52. Aridi YS, Walker JL, Wright ORL. The association between the 71. Martınez-Huelamo M, Rodrıguez-Morat o J, Boronat A, de la
Mediterranean dietary pattern and cognitive health: a systematic Torre R. Modulation of Nrf2 by olive oil and wine polyphenols
review. Nutrients. 2017;9(7):674. pii:doi:10.3390/nu9070674. and neuroprotection. Antioxid. 2017;6(4):73. pii:doi:10.3390/
53. Gardener H, Caunca MR. Mediterranean diet in preventing neu- antiox6040073.
rodegenerative diseases. Curr Nutr Rep. 2018;7(1):10–20. doi:10. 72. Nasri H, Baradaran A, Shirzad H, Rafieian-Kopaei M. New con-
1007/s13668-018-0222-5. cepts in nutraceuticals as alternative for pharmaceuticals. Int J
54. Ravi SK, Narasingappa RB, Vincent N. Neuro-nutrients as anti- Prevent Med. 2014;12:1487–1499.
alzheimer’s disease agents: A critical review. Crit Rev Food Sci 73. Calkins MJ, Johnson DA, Townsend JA, Vargas MR, Dowell JA,
Nutrit Food Sci. 2018;58:1–20. doi:10.1080/10408398.2018. Williamson TP, Kraft AD, Lee J-M, Li J, Johnson JA. The Nrf2/
1481012. ARE pathway as a potential therapeutic target in neurodegenera-
55. World Health Organization. 2009. Interventions on diet and tive disease. Antioxid Redox Sign. 2009;11:497–508. doi:10.1089/
physical activity: what works: summary report. http://apps.who. ars.2008.2242.
int/iris/handle/10665/44140 74. Darvesh AS, Carroll RT, Bishayee A, Novotny NA, Geldenhuys
56. Maraki MI, Yannakoulia M, Stamelou M, Stefanis L, WJ, Van der Schyf CJ. Curcumin and neurodegenerative dis-
Xiromerisiou G, Kosmidis MH, Dardiotis E, Hadjigeorgiou GM, eases: a perspective. Expert Opin Invest Drugs. 2012;21(8):
Sakka P, Anastasiou CA, et al. Mediterranean diet adherence is 1123–1140. doi:10.1517/13543784.2012.693479.
related to reduced probability of prodromal Parkinson’s disease. 75. Monroy A, Lithgow GL, Alavez S. Curcumin and neurodegener-
Mov Disord. 2019;34(1):48–57. doi:10.1002/mds.27489. ative diseases. BioFactors. 2013;1:122–132. doi:10.1002/biof.1063.
57. Zieli
nska MA, Białecka A, Pietruszka B, Hamułka J. Vegetables 76. Fuentealba J, Dibarrart AJ, Fuentes-Fuentes MC, Saez-Orellana
F, Qui~ nones K, Guzman L, Perez C, Becerra J, Aguayo LG.
and fruit, as a source of bioactive substances, and impact on
Synaptic failure and adenosine triphosphate imbalance induced
memory and cognitive function of elderly. Postepy Hig Med
by amyloid-b aggregates are prevented by blueberry-enriched
Dosw. 2017;71:267–280. doi:10.5604/01.3001.0010.3812.
polyphenols extract. J Neurosci Res. 2011;89(9):1499–1508. doi:
58. Harasym J, Oledzki R. Effect of fruit and vegetable antioxidants
10.1002/jnr.22679.
on total antioxidant capacity of blood plasma. Nutr. 2014;30(5):
77. Yang X, Zhang Y, Xu H, Luo X, Yu J, Liu J, Chuen-Chung R.
511–517. doi:10.1016/j.nut.2013.08.019.
Neuroprotection of coenzyme Q10 in neurodegenerative diseases.
59. Morris MC, Evans DA, Tangney CC, Bienias JL, Wilson RS.
Curr Topics Med Chem. 2016;16(8):858–866. doi:10.2174/
Associations of vegetable and fruit consumption with age-related
1568026615666150827095252.
cognitive change. Neurology. 2006;67(8):1370–1376. doi:10.1212/
78. Qu Z, Mossine VV, Cui J, Sun GY, Gu Z. Protective effects of
01.wnl.0000240224.38978.d8.
AGE and Its components on neuroinflammation and neurode-
60. Nooyens AC, Bueno-de-Mesquita HB, van Boxtel MP, van
generation. Neuromol Med. 2016;18(3):474–482. doi:10.1007/
Gelder BM, Verhagen H, Verschuren WM. Fruit and vegetable
s12017-016-8410-1.
intake and cognitive decline in middle-aged men and women: 79. Grimmig B, Kim SH, Nash K, Bickford PC, Douglas Shytle R.
the Doetinchem Cohort Study. Br J Nutr. 2011;106(5):752–761. Neuroprotective mechanisms of astaxanthin: a potential
doi:10.1017/S0007114511001024. therapeutic role in preserving cognitive function in age and
61. Hosomi R, Yoshida M, Fukunaga K. Seafood consumption and neurodegeneration. GeroScience. 2017;39(1):19–32. doi:10.1007/
components for health. Global J Health Sci. 2012;4:72–86. doi:10. s11357-017-9958-x.
5539/gjhs.v4n3p72. 80. Yook JS, Okamoto M, Rakwal R, Shibato J, Lee MC, Matsui T,
62. Shinto L. Eating seafood and cognitive decline in older adults. Chang H, Cho JY, Soya H. Astaxanthin supplementation enhan-
Neurology. 2016;86(22):e231–233. doi:10.1212/WNL. ces adult hippocampal neurogenesis and spatial memory in mice.
0000000000002733. Mol Nutr Food Res. 2016;60:589–599. doi:10.1002/mnfr.
63. Kim DH, Grodstein F, Rosner B, Kang JH, Cook NR, Manson 201500634.
JE, Buring JE, Willett WC, Okereke OI. Seafood types and age- 81. Martinez-Perez DA, Jimenez-Del-Rio M, Velez-Pardo C.
related cognitive decline in the Women’s Health Study. J Epigallocatechin-3-gallate protects and prevents paraquat-
Gerontol. Ser A, Biol Sci Med Sci. 2013;68(10):1255–1262. doi: induced oxidative stress and neurodegeneration in knockdown
10.1093/gerona/glt037. dj-1-b Drosophila melanogaster. Neurotox Res. 2018;34(3):
64. Rigacci S, Stefani M. Nutraceutical properties of olive oil poly- 401–416. doi:10.1007/s12640-018-9899-x.
phenols. An itinerary from cultured cells through animal models 82. Xicota L, Rodriguez-Morato J, Dierssen M, de la Torre R.
to humans. IJMS. 2016;17(6):843. pii:doi:10.3390/ijms17060843. Potential Role of (-)-Epigallocatechin-3-Gallate (EGCG) in the
65. Tuck KL, Hayball PJ. Major phenolic compounds in olive oil: secondary prevention of Alzheimer disease. Curr Drug Targets.
metabolism and health effects. J Nutr Biochem. 2002;13(11): 2017;18(2):174–195. doi:10.2174/1389450116666150825113655.
636–644. doi:10.1016/S0955-2863(02)00229-2. 83. Molz P, Schr€ oder N. Potential therapeutic effects of lipoic acid
66. Abuznait AH, Qosa H, Busnena BA, El Sayed KA, Kaddoumi on memory deficits related to aging and neurodegeneration.
A. Olive-oil-derived oleocanthal enhances b-amyloid clearance as Front Pharmacol. 2017;8:849. doi:10.3389/fphar.2017.00849.
a potential neuroprotective mechanism against Alzheimer’s dis- 84. Zhang DM, Ye JX, Mu JS, Cui XP. Efficacy of vitamin B supple-
ease: in vitro and in vivo studies. ACS Chem Neurosci. 2013; mentation on cognition in elderly patients with cognitive-related
4(6):973–982. doi:10.1021/cn400024q. diseases. J Geriatr Psychiatr Neurol. 2017;30(1):50–59.
67. Monti MC, Margarucci L, Riccio R, Casapullo A. Modulation of 85. Yu L, Wang W, Pang W, Xiao Z, Jiang Y, Hong Y. Dietary lyco-
tau protein fibrillization by oleocanthal. J Nat Prod. 2012;75(9): pene supplementation improves cognitive performances in tau
1584–1588. doi:10.1021/np300384h. transgenic mice expressing P301L mutation via inhibiting
JOURNAL OF THE AMERICAN COLLEGE OF NUTRITION 27

oxidative stress and tau hyperphosphorylation. J Alzheimer’s Dis. 91. Walton JR, Wang MX. APP expression, distribution and accu-
2017;57(2):475–482. doi:10.3233/JAD-161216. mulation are altered by aluminum in a rodent model for
86. Schulz JB, Deuschl G. [Influence of lifestyle on neurodegenera- Alzheimer’s disease. J Inorg Biochem. 2009;103(11):1548–1554.
tive diseases]. Nervenarzt. 2015;86(8):954–959. doi:10.1007/ doi:10.1016/j.jinorgbio.2009.07.027.
s00115-014-4252-y. 92. Salazar JG, Ribes D, Cabre M, Domingo JL, Sanchez-Santed F,
87. Evans M, Antony J, Guthrie N, Bernie Landes B, Aruoma OI. A Colomina MT. Amyloid beta peptide levels increase in brain of
randomized double-blind, placebo controlled, four-arm parallel AbetaPP Swedish mice after exposure to chlorpyrifos. CAR.
study investigating the effect of a broad-spectrum wellness beverage 2011;8:732–740. doi:10.2174/156720511797633197.
on mood state in healthy, moderately stressed adults. J Am Coll 93. Peris-Sampedro F, Salazar JG, Cabre M, Reverte I, Domingo JL,
Nutr. 2018;37(3):234–242. doi:10.1080/07315724.2017.1393356.
Sanchez-Santed F, Colomina MT. Impaired retention in AbPP
88. Baldi I, Lebailly P, Mohammed-Brahim B, Letenneur L,
Swedish mice six months after oral exposure to chlorpyrifos.
Dartigues JF, Brochard P. Neurodegenerative diseases and expos-
ure to pesticides in the elderly. Am J Epidemiol. 2003;157(5): Food Chem Toxicol. 2014;72:289–294. doi:10.1016/j.fct.2014.07.
409–414. doi:10.1093/aje/kwf216. 036.
89. Manivannan Y, Manivannan B, Beach TG, Halden RU. Role of 94. Aruoma OI, Hausman-Cohen S, Pizano J, Schmidt MA, Minich
environmental contaminants in the etiology of Alzheimer’s dis- DM, Joffe Y, Brandhorst S, Evans SJ, Brady DM. Personalized
ease: A review. Curr Alzheimer Res. 2015;12:116–146. nutrition: Translating the science of nutrigenomics into practice:
90. Reddy PH, Reddy TP. Mitochondria as a therapeutic target for Proceedings from the 2018 American College of Nutrition
aging and neurodegenerative diseases. Curr Alzheimer Res. 2011; Meeting. J Am Coll Nutr. 2019;38(4):287–301. doi: 10.1080/
8(4):393–409. 07315724.2019.1582980.

You might also like