You are on page 1of 8

Brain Disorders 3 (2021) 100020

Contents lists available at ScienceDirect

Brain Disorders
journal homepage: www.sciencedirect.com/journal/brain-disorders

DJ-1 and Parkinson’s disease


Ross Gibson a, Sanika P. Dalvi b, Prasad S. Dalvi a, *
a
Biology Department, Morosky College of Professions and Sciences, Gannon University, Erie PA 16541, USA
b
The University of Buckingham Medical School, Buckingham, Buckinghamshire, MK18 1EG, UK

A R T I C L E I N F O A B S T R A C T

Keywords: Parkinson’s disease (PD) is characterized by a significant loss in movement, tremors, and impaired posture.
Parkinson’s disease Worldwide prevalence of PD indicates that aging increases the risk of PD development and is more common in
Dopaminergic neurons males than females. Several environmental factors along with potential genetic factors have been found to affect
Substantia nigra
the development of PD. Most common finding in PD is degeneration of the dopamine-producing (dopaminergic)
Oxidative stress
DJ-1 protein
neurons within the nigrostriatal pathway, and neuroinflammation due to increased oxidative stress has been
PARK-7 gene associated to trigger the degeneration of these dopaminergic neurons in substantia nigra. Within the substantia
nigra, dopaminergic neurons have been well known to produce reactive oxygen species (ROS) that cause
overactivation of microglia, which are the major resident immune cells in the brain. The pro-inflammatory
cytokines produced by hyperactive microglia lead to significant neuroinflammation and degeneration of neu­
rons. Mutations in several genes also play a large role in the overaccumulation of ROS within these neurons.
Among such genes, mutations in the protein DJ-1 encoded by PARK-7 gene cause autosomal recessive forms of
PD. Several animal studies have explored DJ-1 as a possible therapeutic target for PD and neurodegeneration,
and therefore, human clinical studies that would represent an important step in unravelling the potential of DJ-1
as an ideal target for therapeutic intervention are warranted. This review attempts to describe functions and
mutations of DJ-1 and its possible roles in the pathogenesis of PD.

1. ntroduction increasingly found to play important role in its pathophysiology [3].


Often, PD is diagnosed specifically through symptom recognition and
1.1. Parkinson’s disease neuronal as well as physical examination due to present lack of a reliable
test or combination of tests that diagnose PD accurately [4]. Worldwide
Parkinson’s disease (PD) is one of the most common neurodegener­ prevalence of PD varies depending on the study; however, in the US and
ative disorders mainly associated with a significant loss in movement Europe it is estimated that between 100 and 300 per 100,000 individuals
(bradykinesia), tremors, and impaired posture. There are many different below the age of 65 develop PD, and the prevalence increases to around
types of “parkinsonisms”, a generalized term described for a group of 950 per 100,000 individuals above the age of 65. The prevalence in
disorders that exhibit these movement defects (Table 1). Generally, China and South America is similar to Europe and the US; however,
these parkinsonisms are classified as degenerative and nondegenerative Africa sees a much smaller prevalence [5]. Through these prevalence
types with the difference being that degenerative disorders are caused data, conclusions can be made that PD seems to increase in risk in
by some progressive neurological degenerative process in contrast to conjunction with age and globally varies by region. It is also found to be
nondegenerative types where neuronal genetic defects play a more more common in males than in females as 134 in 100,000 males develop
prominent role. Most of these parkinsonisms carry the same or similar PD compared to only 41 in 100,000 females [6]. Potential genetic factors
symptoms with differences mainly in histological, molecular, and with many environmental factors, such as, cigarette smoking and
neurochemical findings [1]. It is important to note that the type of increased exposure to pesticides increase risk of development of PD,
parkinsonism is quite challenging to diagnose and most often is deter­ whereas consumption of coffee and tea potentially decrease probability
mined post-mortem [2]. PD is the most common cause of parkinsonism of PD development [7].
and is seen as a degenerative disorder; however, genetic properties are

* Corresponding author at: Biology Department, Morosky College of Professions and Sciences, Gannon University, Erie, PA, 16541, USA.
E-mail address: dalvi001@gannon.edu (P.S. Dalvi).

https://doi.org/10.1016/j.dscb.2021.100020
Received 28 June 2021; Received in revised form 9 August 2021; Accepted 10 August 2021
Available online 12 August 2021
2666-4593/© 2021 The Author(s). Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
R. Gibson et al. Brain Disorders 3 (2021) 100020

Table 1 nigrosomes of SNr and over time the degeneration across the substantia
Classification of parkinsonism nigra increases [18].
Category Type Etiology
Nondegenerative Toxic Continued exposure to Manganese (Mn) 1.3. Factors involved in pathogenesis of PD
parkinsonism causes motor-related rigidity [1, 8]
Nondegenerative Vascular Secondary parkinsonism instigated from
Oxidative stress is caused by accumulation of reactive oxygen species
parkinsonism cerebrovascular disease such as stroke
and hypertension [1, 9] (ROS) through cellular metabolism and the diminished capability of
Nondegenerative Drug-induced Primarily related to prolonged usage of these cells to safely detoxify the oxygen radical end products in meta­
parkinsonism antipsychotic drugs, can be caused by bolism. ROS can cause a multitude of detrimental effects within cells, as
other drugs such as antiepileptic drugs their free radical properties can harm cell membrane function, alter DNA
[1, 10]
structure, and induce lipid peroxidation [19]. Dopaminergic neurons are
Degenerative Lewy body Atypical α-Synuclein protein build-ups in
dementia the brain [1, 11] specifically vulnerable to the accumulation of ROS through high iron
Degenerative Multiple system α-Synuclein protein collects in glial cells, content, increased number of mitochondria, and polyunsaturated fatty
atrophy particularly oligodendrocytes (myelin acids. Furthermore, oxidative stress can also lead to activation of glial
producing cells) causing demyelination
cells, leading to an inflammatory response [20]. The inflammatory
[1, 12]
Degenerative Progressive Tau protein lesions [1, 13]
response generated following the excess accumulation of ROS in the
supranuclear palsy neurons is known as neuroinflammation and has been proposed to
Degenerative Parkinson’s Degeneration of neurons in substantia trigger the actual degeneration of neurons (Fig. 2). Within the substantia
disease nigra and nigrostriatal pathway [14]; nigra, high concentration of activated microglia which are the major
however, genetic factors are also
resident immune cells in the brain and low density of astrocytes that
involved [3]
provide neuroprotective mechanisms by detoxification of ROS
contribute to an inflammatory state, leading to slow degeneration of the
1.2. Brain regions involved in the development of PD neurons [21]. In response to the threat of the increased ROS and the
resulting neuroinflammation, there are several mechanisms within the
PD most commonly sees degeneration of the dopamine-producing dopaminergic neurons to prevent increases of oxidative stress, such as,
neuronal cells known as dopaminergic neurons within the nigrostriatal complex III, Vitamin E, and glutathione [22, 23]. While the antioxidant
pathway (Fig. 1A), the pathway connecting the substantia nigra to the action of these molecules varies greatly depending on the mechanism,
striatum, which is the largest structure of the basal ganglia of the brain they all similarly deactivate or neutralize free oxygen radicals after their
[14]. The dopaminergic neurons, especially concentrated within the production within the mitochondria [24]. Although the antioxidant
substantia nigra, are specifically known to generate high levels of mechanisms are present within the neurons, the exact causative mech­
oxidative stress [15]. The substantia nigra lies within the midbrain and anism of neuronal degeneration in PD patients remains unknown.
functions predominantly to assist in muscle movement through release Relatively recently, protein deglycase DJ-1, also known as Parkinson
of dopamine [16]. There are two parts in the substantia nigra that differ disease protein 7 that is encoded by PARK-7 gene, was discovered to be
in both structure and function (Fig. 1B). The pars compacta of the sub­ one of the main genes that, if mutated, can be responsible for the
stantia nigra (SNc) lies dorsal and is made up of clusters of dopaminergic degeneration of these neurons [25]. Much remains to be studied about
neurons, and the pars reticulata of the substantia nigra (SNr) lies ventral the role of DJ-1 in the development of PD. In this review, we will analyze
and is composed of mostly gamma aminobutyric acid (GABA) neurons the connection between neuroinflammation and oxidative stress within
that provide inhibitory control to the local dopaminergic neurons of the PD, and the impact of the DJ-1 protein mutation and its dysfunction on
region [17]. Within the substantia nigra there are clusters of dopami­ the pathogenesis of PD.
nergic neurons called nigrosomes, and in PD, there seems to be heavy
degeneration within a nigrosome of the SNc that spreads to other

A Nigrostriatal B
pathway Superior
Colliculus
Cerebral
aqueduct

Basal Pars Ventral Pars


Ganglia Compacta Tegmental Reculata
Substana
Nigra Area

Fig. 1. (A) A schematic diagram of the intact nigrostriatal pathway showing ascending axons of dopaminergic neurons originating in the substantia nigra of the
ventral midbrain and innervating the basal ganglia (red dotted circle), (B) Transverse section through the superior colliculus of the midbrain emphasizing the pars
compacta and pars reticulata of the substantia nigra.

2
R. Gibson et al. Brain Disorders 3 (2021) 100020

A B

↑ Oxidave stress
↑ Neuroinflammaon
↓ Dopaminergic neurons
↓ Innervaon of basal ganglia
↓ Dopamine level in basal ganglia

Fig. 2. Pathogenesis of Parkinson’s disease: (A) a schematic diagram of the intact nigrostriatal pathway of dopaminergic neurons originating in the substantia nigra
and innervating the basal ganglia (red dotted circle), (B) a comparative schematic diagram of the progressive degeneration of the dopaminergic neurons (represented
by black dotted lines) that results in a loss of dopaminergic innervation of the basal ganglia and consequently leads to pathogenesis of Parkinson’s disease.

2. PD and Neuroinflammation a combination of all the actions of the glial cells [26]. There are three
types of glial cells within the CNS, microglia, astrocytes, and oligoden­
Neuroinflammation is the generic action of the innate immune sys­ drocytes. Microglial cells provide most of the initial response, as they
tem to eliminate pathogens, cell debris, and misfolded proteins in neu­ begin in a resting state continuously scanning the brain for any abnor­
rons. This inflammatory response of the central nervous system (CNS) is malities. When an abnormality is found through recognition of an

Fig. 3. The process of an inactive microglia activating and releasing pro-inflammatory cytokines to cause neuroinflammation that leads to degenerate a healthy
neuron. IFN-γ, interferon-γ; TNF-α, tumor necrosis factor-α; IL-1α, interleukin-1α; ROS, reactive oxygen species

3
R. Gibson et al. Brain Disorders 3 (2021) 100020

Fig. 4. Chemical pathway of the degradation of dopamine leading to the generation of reactive oxygen species and the adverse effects of oxidative stress. MAO,
monoamine oxidase enzyme; DOPAC, 3,4-Dihydroxyphenylacetic acid; O2-, superoxide ion; NO▪, nitric oxide radical; ONOO-, peroxynitrite; ▪OH, hydroxyl radical

activating signal, which is usually a cytokine, the microglia undergo a the symptoms of PD, implying the relevance of neuroinflammation
shape change, ultimately impacting their function. The activated during PD pathogenesis [36]. These indications support that neuro­
microglia can perform a multitude of active responses, such as, phago­ inflammation within the CNS most likely contributes to the degenera­
cytic action that forms a major portion of its response, synaptic stripping tion of neurons associated with pathogenesis of PD (Fig. 3).
which is a process of ingesting synaptic terminals to decrease sympa­
thetic input, and controlling the electrolyte efflux to regulate neuronal 3. Neuroinflammatory response to oxidative stress
metabolism and activation [27, 28]. The astrocytes control a mostly
passive response through amplification of microglial signals, but they Many studies have analyzed the relationship between oxidative
can also exhibit more of an active response via regulation of the meta­ stress and neuroinflammation. Due to the damaging nature of ROS, these
bolism of neurons through control of ion channels and alterations of radicals are often utilized as a modulator of inflammatory response.
neurotransmitter release [29]. Most frequently, neuroinflammation is Cytokines IL-1β, TNF-α, IL-2, IL-6, and IFN-γ have all been seen to be
associated with demyelination of the axon connections within the CNS; significantly expressed in post-mortem brain analyses of PD patients [37].
however, oligodendrocytes handle the remyelination of these connec­ In PD and other diseases associated with excessive accumulation of ROS,
tions during inflammatory response as the precursors of oligodendrocyte such as fibrosis and diabetes, the cytokine transforming growth factor-β
are activated by the presence of other inflammatory cells that results in has shown to cause an increase in production of ROS, specifically
the generation of more oligodendrocytes and the subsequent remyeli­ through increasing expression of an enzyme called nicotinamide
nation [30, 31]. adenine dinucleotide phosphate oxidase (NOX)-4 [38, 39]. The other
In PD, neuroinflammation is specifically thought to be one of the forms of the NOX enzyme are also believed to be involved in the ROS
main causes for the degeneration of the dopaminergic neurons in the generation in humans [40]. The resulting overproduction of ROS creates
SNc and the nigrostriatal pathway. The pro-inflammatory cytokines are a vicious cycle eventually leading to a significant increase in neuro­
produced by overactive microglia leading to significant neuro­ inflammatory response due to microglial activation (Fig. 3). Most
inflammation [32]. There is an indication that within the sensitive re­ significantly, microglial activation in PD patients has been associated to
gions of the SNc, expression of pro-inflammatory cytokines, such as, the increase in ROS production around the Lewy bodies which are
interleukin (IL)-1α, tumor necrosis factor-α (TNF-α), and interferon-γ clumps of proteins that form in specific areas of the brain, in particular,
(IFN-γ) increases [33], whereas antioxidant expression, such as, gluta­ within the substantia nigra [41]. Whether the increase in ROS directly
thione, decreases [34]. Investigations of the P2Y6R receptor, a receptor stimulates the microglia or is mediated through increasing
located on the microglia specifically expressed during phagocytic acti­ pro-inflammatory cytokine production is debated; however, the relation
vation, an activated state of the microglia, has been found to be present between the microglial activation and ROS concentrations in develop­
in higher concentrations in PD patients than healthy individuals sug­ ment of neuroinflammation is uncontested.
gesting a neuroinflammatory connection to PD [35]. Therefore, the
P2Y6R receptor is considered for its use as a biomarker for diagnosis of 4. Degeneration of dopaminergic neurons
PD. Furthermore, the use of anti-inflammatory drugs, in particular,
non-steroidal anti-inflammatory drugs, seems to reduce or even prevent Dopaminergic neurons have been well known to produce ROS within

4
R. Gibson et al. Brain Disorders 3 (2021) 100020

Table 2 against oxidative damage at least to some degree; however, the exact
Overview of the most common PD genes mechanism in which the protein performs this function is still debated.
PD Gene Chromosomal Position The leading suggestion of the DJ-1 protein function is that it may act as a
Protein deglycase DJ-1 (PARK7) Chromosome 1p36 [52] chaperone that assists in refolding damaged proteins, folding of new
PARK2 Chromosome 6q26 [53] proteins, and/or delivering damaged proteins to proteasomes [59].
PARK8 Chromosome 12p11.2-q13.1 [54]
Within PD rodent experimental models, dopamine producing neurons in
α-Synuclein (SNCA) Chromosome 4q21-q23 [55]
PTEN-induced kinase 1 (PINK1) Chromosome 1p35-36 [53] substantia nigra seem to show high rates of transcription of the PARK7
Leucine-rich repeat kinase 2 (LRRK2) Chromosome 12q12 [53] gene, which relates to the high amounts of ROS produced by these
dopaminergic neurons [60].
The PARK7 gene is present on chromosome 1, and mutations in the
the substantia nigra. Dopamine exists in these neuronal cells within gene are related to an autosomal recessive variant of PD. Firstly, mu­
synaptic vesicles for eventual release into the synaptic cleft to contribute tations in the Cys-106 residue have been observed to prevent cell growth
to its neurotransmitter function. When dopamine concentrations within in high oxidative stress environments [61]; however, this mutation has
the neurons are in excess, they are redistributed into the cytosol. Within not been tested in association with PD yet and warrants further inves­
the cytosol, the dopamine stability decreases significantly and triggers tigation. Next, frameshift mutations in exon 2, which is the first coding
degradation [42]. During this degradation of dopamine, the actions of exon, and exon 7 have been found to cause a complete loss of functional
the monoamine oxidase enzyme produce cytotoxic ROS in the DJ-1 protein in an analysis of 107 early-onset PD patients who were less
manufacturing of H2O2 and 3,4-Dihydroxyphenylacetic acid, or the than 50 years old at the time of diagnosis [62]. The specific DJ-1 mu­
spontaneous autoxidation of dopamine produces superoxide (O2-) ion tations show differences based on ethnic origin as well. As an example, a
and quinones [43] (Fig. 4). The O2- from the autoxidation specifically 14bp deletion placing the centromeric border within an intron (intron 5)
can react with nitric oxide radicals (NO▪) to create peroxynitrite was seen in Dutch/Italian patients but was not present in any other
(ONOO-). ONOO- is a very reactive molecule and can cause damage ethnicity [63]. In eastern Indian populations and potentially present in
within the cell in two distinct ways [44]. Firstly, ONOO- stimulates the Western Europeans as well, an Arg98Gln variant of the DJ-1 protein has
nitration and the guanine oxidation of DNA, eventually creating a hy­ been present in PD patients [64]. Mutations of the gene itself seem to
droxyl radical (▪OH) which results in DNA strand breakage [45]. Sec­ show significant variance between most patients; however, the muta­
ondly, the ONOO- can act as a very potent oxidant, specifically targeting tions that affect protein function substantially affect survival of cells in
thiol groups in lipids and proteins. The oxidation of these thiols results in oxidative environments [65]. Mutations within the DJ-1 protein have
the production of disulfide bonds that alters the structure of these been seen to cause an apparent loss in the function of the chaperone
molecules and concurrently disrupts any apparent function [45, 46] action and/or the ability to protect the cells from oxidative stress,
(Fig. 4). By analyzing the breaking down of cytosolic dopamine into potentially leading to PD [66]. There are a multitude of specific muta­
ROS, it can be concluded that an excess of dopamine production within tions within DJ-1 protein that can be associated with PD, some more
these neurons could be the cause of neurodegeneration. clearly linked to PD pathogenesis than others [67].
The pathological development of Lewy bodies due to accumulation
of α-Synuclein [47], and the mitochondrial dysfunction resulting in ROS 6. Current therapies and treatments for PD
accumulation also seem to be prominent theories on the degeneration of
the dopaminergic neurons. Under pathological conditions, α-Synuclein There have been many treatments tested to improve PD progression.
monomers develop toxic α-Synuclein oligomers, fibrils and Currently, the main drugs are focused on controlling the symptoms of
micro-aggregates, which are considered the most toxic species of the disorder and are prescribed based on the stages of the disorder.
α-Synuclein, and these toxic fibril aggregates trigger the synapse Levodopa and carbidopa, or a combination of both, are used to treat the
deconstruction followed by neurodegeneration [11, 48, 49]. However, it symptoms; however, they induce some long-term consequences related
is still unclear why the degeneration is selective for the dopamine pro­ to heart rhythm, so dopamine agonists can be given in its place [68].
ducing neurons in the SNc. While collectively there is plentiful evidence One notable drug tested is Bruceine D, which has been found in multiple
of a correlation between α-Synuclein accumulation and SNc neuron studies to reduce oxidative stress and decrease inflammatory response
death [50], the mechanism at which the α-Synuclein specifically targets by improving an antioxidant system regulated by E2-related factor 2
the SNc neurons is unclear [51]. The mitochondrial dysfunction related (Nrf2) [69]. The natural medicine derived from Crocus Sativus, an herb
to the ROS accumulation within the neurons seems to be a slightly more found in countries like Iran and India, and commonly known as saffron,
reliable outlook in referencing the specificity of the neurodegeneration. has shown effectiveness in suppressing inflammatory cytokines and
Normally there is a multitude of mechanisms that counteract for the ROS. Many compounds have been derived from saffron and have shown
increased ROS concentrations; however, mutations in several genes play some promise in mice studies related to neuronal injuries [68, 70].
a large role in the over-accumulation of ROS within these neurons While early on in its research, some suggestions have been made for
(Table 2). Among such genes, mutations in PARK-7 gene lead to pro­ investigation of brain-gut peptides like glucagon-like peptide 1, pitui­
duction of a non-functional deglycase DJ-1 protein that prevents it from tary adenylate cyclase-activating peptide, nesfatin-1, and ghrelin due to
protecting dopaminergic neurons against oxidative damage [25]. their neuroprotective effects on the PD symptoms [71]. Stem cell re­
placements have shown some promise in reference to neuronal diseases;
5. DJ-1 action and mutations however, in vivo applications have not demonstrated as much success.
Cell transplantation therapies exhibit much promise on the effects of PD
The PD patients have an autosomal recessive variant of the PARK7 due to the direct loss of SNc neurons being one of the main patho­
gene; however, sometimes an autosomal dominant variation of the physiological features of PD. Currently, fetal ventral mesencephalic
leucine-rich repeat kinase 2 (LRRK2) or α-Synuclein (SNCA) genes can (FVM) tissue transplantation using 6-9 week old aborted human embryo
contribute to the symptoms of this disorder [56]. The PARK7 shows the most promise, as majority of studies investigating FVM grafts
gene-encoded DJ-1 protein is highly expressed in cells with high energy show significant reduction in typical PD symptoms while similarly
demand and increased oxidative stress environment, as it is observed to improving dopamine release [72-74].
be overexpressed in most human cancers and similarly expressed in the
testis where low expression of DJ-1 is associated with infertility [57, 58]. 7. Diagnostic and therapeutic potency of DJ-1 in PD
These findings potentially suggest that the predominant action of DJ-1 is
against oxidative stress and that the overexpression of DJ-1 protects The accuracy of the clinical diagnosis in the early stages of PD is still

5
R. Gibson et al. Brain Disorders 3 (2021) 100020

Fig. 5. Schematic diagram summarizing the roles of normal and mutant DJ-1 protein in health and pathogenesis of Parkinson’s disease (PD), and proposed diagnostic
and therapeutic potency of DJ-1 in PD.

limited. Functional neuroimaging techniques are helpful in patients with oxidative damage. The loss of function or even reduced function of DJ-1
first signs of parkinsonism, but are expensive and not broadly available. trigger the onset of increased oxidative stress leading to pathogenesis of
Therefore, a priority for the scientific community is the detection of PD PD. Experimental rodent PD models have demonstrated that reversal of
pathology at early stages of the disease. In this regard, a recent study excessive oxidation of DJ-1 can restore its neuroprotective activity
found that the ratio of neural-derived exosomal DJ-1 levels to total DJ-1 against neurodegeneration due to overaccumulation of ROS in PD, and
was increased in PD patients versus controls, and a positive correlation inhibition of overoxidation of PD-1, and thereby its inactivation, can be
was found between levels of DJ-1 and α-Synuclein in plasma neural- a therapeutic target for the oxidative stress-related diseases such as PD.
derived exosomes, thus indicating a potential for exosomal DJ-1 as a Furthermore, detection of DJ-1 in the cerebrospinal fluid and plasma
PD biomarker [75]. Another study found that detection of DJ-1 in the exosome can be used as a promising biomarker for early detection and
cerebrospinal fluid can be used as a biomarker for PD [76], however, this diagnosis for PD. Overall, future studies should be directed to establish
potential of DJ-1 as a biomarker is still unclear, and larger clinical trials DJ-1 as a biomarker and to utilize DJ-1 targeting compounds that either
are needed to fully validate DJ-1 as a reliable PD biomarker. stabilize the active DJ-1 form or enhance DJ-1 activity to restore neu­
DJ-1 has been found to be therapeutically effective for animal roprotection and reverse neurodegeneration in PD. Furthermore, clin­
models of neurodegenerative disorders [77]. In some of these studies, rat ical studies using human cohorts to explore the potential of DJ-1 as a
PD models were tested for the efficacy of recombinant wild type DJ-1 for biomarker and therapeutic target for PD are warranted (Fig. 5).
protection of dopaminergic neurons [78, 79]. Other studies adopted a
novel strategy for obtaining DJ-1-mediated dopaminergic neuronal Declaration of Competing Interests
protection by preventing its overoxidation and thereby its inactivation
[80]. By using brain penetrant molecules able to interact with the The authors declare that they have no known competing financial
Cys106 region of DJ-1 that help DJ-1 maintain its active form, it was interests or personal relationships that could have appeared to influence
found that these DJ-1 interactors prevented dopaminergic neuronal the work reported in this paper.
death and restored normal locomotor function in rodent models of PD
[81, 82]. These and other animal studies have explored DJ-1 as a Acknowledgements
possible therapeutic target for PD and neurodegeneration, however,
such studies using human cohorts are currently missing and need to be This research did not receive any specific grant from funding
undertaken that would represent an important step in unravelling the agencies in the public, commercial, or not-for-profit sectors. The authors
potential of DJ-1 as an ideal target for therapeutic intervention. wish to thank the Biology Department of Gannon University for its
financial support of the publication of this article.
8. Conclusion and perspective
References
PD most commonly results due to degeneration of the dopaminergic
neurons within the substantia nigra and nigrostriatal pathway. Genetics [1] D.W. Dickson, Parkinson’s disease and parkinsonism: neuropathology, Cold Spring
Harb. Perspect. Med. 2 (8) (2012), a009258.
has been found to play an important role in pathophysiology of PD by [2] D. Toulorge, A.H. Schapira, R. Hajj, Molecular changes in the postmortem
causing neuroinflammation and degeneration of the dopaminergic parkinsonian brain, J. Neurochem. (2016) 27–58, 139 Suppl 1.
neurons in the SNc and the nigrostriatal pathway. Dopaminergic neu­ [3] C. Klein, A. Westenberger, Genetics of Parkinson’s disease. Cold Spring Harb.
Perspect. Med. 2 (1) (2012), a008888.
rons within the substantia nigra actively produce ROS, however, several [4] J. Jankovic, Parkinson’s disease: clinical features and diagnosis, J. Neurol.
protective mechanisms counteract for the increased oxidative stress Neurosurg. Psychiatry 79 (4) (2008) 368–376.
among which DJ-1 protein encoded by PARK-7 gene has been found to [5] K. Wirdefeldt, et al., Epidemiology and etiology of Parkinson’s disease: a review of
the evidence, Eur. J. Epidemiol. (2011) S1–58, 26 Suppl 1.
have a specific protective role against oxidative stress-induced neuro­ [6] T. Pringsheim, et al., The prevalence of Parkinson’s disease: a systematic review
inflammation and neuronal cell death. However, mutations in PARK-7 and meta-analysis, Mov. Disord. 29 (13) (2014) 1583–1590.
gene lead to excessive oxidation and resulting inactivation of DJ-1 [7] A. Delamarre, W.G. Meissner, Epidemiology, environmental risk factors and
genetics of Parkinson’s disease, Presse Med. 46 (2017) 175–181, 2 Pt 1.
protein that prevents it from protecting dopaminergic neurons against

6
R. Gibson et al. Brain Disorders 3 (2021) 100020

[8] T.R. Guilarte, K.K. Gonzales, Manganese-Induced Parkinsonism Is Not Idiopathic [44] M.J. LaVoie, T.G. Hastings, Peroxynitrite- and nitrite-induced oxidation of
Parkinson’s Disease: Environmental and Genetic Evidence, Toxicol. Sci. 146 (2) dopamine: implications for nitric oxide in dopaminergic cell loss, J. Neurochem. 73
(2015) 204–212. (6) (1999) 2546–2554.
[9] B. Thanvi, N. Lo, T. Robinson, Vascular parkinsonism–an important cause of [45] S. Bartesaghi, R. Radi, Fundamentals on the biochemistry of peroxynitrite and
parkinsonism in older people, Age Ageing 34 (2) (2005) 114–119. protein tyrosine nitration, Redox. Biol. 14 (2018) 618–625.
[10] H.-W. Shin, S.J. Chung, Drug-induced parkinsonism, J. Clin. Neurol. (Seoul, Korea) [46] R. Radi, et al., Peroxynitrite oxidation of sulfhydryls. The cytotoxic potential of
8 (1) (2012) 15–21. superoxide and nitric oxide, J. Biol. Chem. 266 (7) (1991) 4244–4250.
[11] M.L. Kramer, W.J. Schulz-Schaeffer, Presynaptic alpha-synuclein aggregates, not [47] J.Q. Trojanowski, V.M. Lee, Aggregation of neurofilament and alpha-synuclein
Lewy bodies, cause neurodegeneration in dementia with Lewy bodies, J. Neurosci. proteins in Lewy bodies: implications for the pathogenesis of Parkinson disease and
27 (6) (2007) 1405–1410. Lewy body dementia, Arch. Neurol. 55 (2) (1998) 151–152.
[12] J.A. Palma, L. Norcliffe-Kaufmann, H. Kaufmann, Diagnosis of multiple system [48] I.C. Brás, M. Xylaki, T.F. Outeiro, Mechanisms of alpha-synuclein toxicity: An
atrophy, Auton. Neurosci. 211 (2018) 15–25. update and outlook, Prog. Brain Res. 252 (2020) 91–129.
[13] D.G. Coughlin, I. Litvan, Progressive supranuclear palsy: advances in diagnosis and [49] J.C. Bridi, F. Hirth, Mechanisms of α-Synuclein Induced Synaptopathy in
management, Parkinsonism Relat. Disord. 73 (2020) 105–116. Parkinson’s Disease, Front. Neurosci. 12 (80) (2018).
[14] D. Huang, et al., Dynamic Changes in the Nigrostriatal Pathway in the MPTP Mouse [50] P. Brundin, R. Melki, Prying into the Prion Hypothesis for Parkinson’s Disease,
Model of Parkinson’s Disease, Parkinsons Dis. (2017), 9349487, 2017. J. Neurosci. 37 (41) (2017) 9808–9818.
[15] P. Jenner, Oxidative stress in Parkinson’s disease, Ann. Neurol. (2003) S26–S36, 53 [51] N. Uemura, et al., Inoculation of alpha-synuclein preformed fibrils into the mouse
Suppl 3discussion S36-8. gastrointestinal tract induces Lewy body-like aggregates in the brainstem via the
[16] Y. Zhang, et al., Anatomical and functional organization of the human substantia vagus nerve, Mol. Neurodegener. 13 (1) (2018) 21.
nigra and its connections, Elife (2017) 6. [52] C.M. van Duijn, et al., Park7, a novel locus for autosomal recessive early-onset
[17] C. Bouarab, B. Thompson, A.M. Polter, VTA GABA Neurons at the Interface of parkinsonism, on chromosome 1p36, Am. J. Hum. Genet. 69 (3) (2001) 629–634.
Stress and Reward, Front. Neural Circuits 13 (2019) 78. [53] K. Nuytemans, et al., Genetic etiology of Parkinson disease associated with
[18] Z. Cheng, et al., Imaging the Nigrosome 1 in the substantia nigra using mutations in the SNCA, PARK2, PINK1, PARK7, and LRRK2 genes: a mutation
susceptibility weighted imaging and quantitative susceptibility mapping: An update, Hum. Mutat. 31 (7) (2010) 763–780.
application to Parkinson’s disease, Neuroimage Clin. 25 (2020), 102103. [54] M. Funayama, et al., A new locus for Parkinson’s disease (PARK8) maps to
[19] G. Pizzino, et al., Oxidative Stress: harms and Benefits for Human Health, Oxid. chromosome 12p11.2-q13.1, Ann. Neurol. 51 (3) (2002) 296–301.
Med. Cell Longev (2017), 8416763, 2017. [55] M.H. Polymeropoulos, et al., Mapping of a gene for Parkinson’s disease to
[20] J.M. Taylor, B.S. Main, P.J. Crack, Neuroinflammation and oxidative stress: co- chromosome 4q21-q23, Science 274 (5290) (1996) 1197–1199.
conspirators in the pathology of Parkinson’s disease, Neurochem. Int. 62 (5) (2013) [56] K. Nuytemans, et al., Genetic etiology of Parkinson disease associated with
803–819. mutations in the SNCA, PARK2, PINK1, PARK7, and LRRK2 genes: a mutation
[21] E.C. Hirsch, S. Hunot, Neuroinflammation in Parkinson’s disease: a target for update, Hum. Mutat. 31 (7) (2010) 763–780.
neuroprotection? Lancet Neurol. 8 (4) (2009) 382–397. [57] D. Jahantigh, A.Hosseinzadeh Colagar, S. Salimi, Genetic polymorphisms and
[22] R.D. Guzy, et al., Mitochondrial complex III is required for hypoxia-induced ROS haplotypes of the DJ-1 gene promoter associated with the susceptibility to male
production and cellular oxygen sensing, Cell Metab. 1 (6) (2005) 401–408. infertility, J. Assist. Reprod. Genet. 34 (12) (2017) 1673–1682.
[23] S. Rizvi, et al., The role of vitamin e in human health and some diseases, Sultan [58] C. Salazar, P. Ruiz-Hincapie, L.M. Ruiz, The Interplay among PINK1/PARKIN/Dj-1
Qaboos Univ. Med. J. 14 (2) (2014) e157–e165. Network during Mitochondrial Quality Control in Cancer Biology: protein
[24] G. Napolitano, et al., Vitamin E Supplementation and Mitochondria in Interaction Analysis, Cells 7 (10) (2018).
Experimental and Functional Hyperthyroidism: a Mini-Review, Nutrients 11 (12) [59] D.J. Moore, et al., Association of DJ-1 and parkin mediated by pathogenic DJ-1
(2019). mutations and oxidative stress, Hum. Mol. Genet. 14 (1) (2005) 71–84.
[25] D.J. Choi, et al., A Parkinson’s disease gene, DJ-1, regulates anti-inflammatory [60] I. Devic, et al., Salivary alpha-synuclein and DJ-1: potential biomarkers for
roles of astrocytes through prostaglandin D2 synthase expression, Neurobiol. Dis. Parkinson’s disease, Brain 134 (Pt 7) (2011) e178.
127 (2019) 482–491. [61] S.Z. Potter, et al., Binding of a single zinc ion to one subunit of copper-zinc
[26] Q.Q. Yang, J.W. Zhou, Neuroinflammation in the central nervous system: superoxide dismutase apoprotein substantially influences the structure and
Symphony of glial cells, Glia 67 (6) (2019) 1017–1035. stability of the entire homodimeric protein, J. Am. Chem. Soc. 129 (15) (2007)
[27] H. Kettenmann, et al., Physiology of microglia, Physiol. Rev. 91 (2) (2011) 4575–4583.
461–553. [62] S. Hague, et al., Early-onset Parkinson’s disease caused by a compound
[28] H. Kettenmann, F. Kirchhoff, A. Verkhratsky, Microglia: new roles for the synaptic heterozygous DJ-1 mutation, Ann. Neurol. 54 (2) (2003) 271–274.
stripper, Neuron 77 (1) (2013) 10–18. [63] V. Bonifati, et al., Mutations in the DJ-1 gene associated with autosomal recessive
[29] S. Liddelow, B. Barres, SnapShot: astrocytes in Health and Disease, Cell 162 (5) early-onset parkinsonism, Science 299 (5604) (2003) 256–259.
(2015) e1 1170–1170. [64] T. Sadhukhan, et al., DJ-1 variants in Indian Parkinson’s disease patients, Dis.
[30] M. Bradl, H. Lassmann, Oligodendrocytes: biology and pathology, Acta Markers 33 (3) (2012) 127–135.
Neuropathol. 119 (1) (2010) 37–53. [65] R.H. Kim, et al., Hypersensitivity of DJ-1-deficient mice to 1-methyl-4-phenyl-
[31] A. Setzu, et al., Inflammation stimulates myelination by transplanted 1,2,3,6-tetrahydropyrindine (MPTP) and oxidative stress, Proc Natl Acad Sci U S A,
oligodendrocyte precursor cells, Glia 54 (4) (2006) 297–303. 102 (14) (2005) 5215–5220.
[32] R.L. Mosley, et al., Neuroinflammation, oxidative stress and the pathogenesis of [66] S. Shendelman, et al., DJ-1 is a redox-dependent molecular chaperone that inhibits
Parkinson’s disease, Clin. Neurosci. Res. 6 (5) (2006) 261–281. alpha-synuclein aggregate formation, PLoS Biol. 2 (11) (2004) e362.
[33] A.M. Depino, et al., Microglial activation with atypical proinflammatory cytokine [67] M. Repici, F. Giorgini, DJ-1 in Parkinson’s Disease: Clinical Insights and
expression in a rat model of Parkinson’s disease, Eur. J. Neurosci. 18 (10) (2003) Therapeutic Perspectives, J. Clin. Med. 8 (9) (2019).
2731–2742. [68] I. Carrera, R. Cacabelos, Current Drugs and Potential Future Neuroprotective
[34] M. Smeyne, R.J. Smeyne, Glutathione metabolism and Parkinson’s disease, Free Compounds for Parkinson’s Disease, Curr. Neuropharmacol. 17 (3) (2019)
Radic. Biol. Med. 62 (2013) 13–25. 295–306.
[35] X. Yang, et al., Microglia P2Y6 receptor is related to Parkinson’s disease through [69] Y. Yang, et al., Bruceine D elevates Nrf2 activation to restrain Parkinson’s disease
neuroinflammatory process, J. Neuroinflammation 14 (1) (2017) 38. in mice through suppressing oxidative stress and inflammatory response, Biochem.
[36] J.J. Gagne, M.C. Power, Anti-inflammatory drugs and risk of Parkinson disease: a Biophys. Res. Commun. 526 (4) (2020) 1013–1020.
meta-analysis, Neurology 74 (12) (2010) 995–1002. [70] C. Zhang, et al., Neuroprotective effects of safranal in a rat model of traumatic
[37] L.M. Collins, et al., Contributions of central and systemic inflammation to the injury to the spinal cord by anti-apoptotic, anti-inflammatory and edema-
pathophysiology of Parkinson’s disease, Neuropharmacology 62 (7) (2012) attenuating, Tissue Cell 47 (3) (2015) 291–300.
2154–2168. [71] D. Dong, J. Xie, J. Wang, Neuroprotective Effects of Brain-Gut Peptides: a Potential
[38] K. Richter, T. Kietzmann, Reactive oxygen species and fibrosis: further evidence of Therapy for Parkinson’s Disease, Neurosci. Bull. 35 (6) (2019) 1085–1096.
a significant liaison, Cell Tissue Res. 365 (3) (2016) 591–605. [72] D. Yang, et al., Human embryonic stem cell-derived dopaminergic neurons reverse
[39] I. Carmona-Cuenca, et al., Upregulation of the NADPH oxidase NOX4 by TGF-beta functional deficit in parkinsonian rats, Stem Cells 26 (1) (2008) 55–63.
in hepatocytes is required for its pro-apoptotic activity, J. Hepatol. 49 (6) (2008) [73] I. Mendez, et al., Dopamine neurons implanted into people with Parkinson’s
965–976. disease survive without pathology for 14 years, Nat. Med. 14 (5) (2008) 507–509.
[40] A. Samoylenko, et al., Nutritional countermeasures targeting reactive oxygen [74] K.C. Sonntag, et al., Pluripotent stem cell-based therapy for Parkinson’s disease:
species in cancer: from mechanisms to biomarkers and clinical evidence, Antioxid. Current status and future prospects, Prog. Neurobiol. 168 (2018) 1–20.
Redox. Signal. 19 (17) (2013) 2157–2196. [75] Z.H. Zhao, et al., Increased DJ-1 and α-Synuclein in Plasma Neural-Derived
[41] H. Solleiro-Villavicencio, S. Rivas-Arancibia, Effect of Chronic Oxidative Stress on Exosomes as Potential Markers for Parkinson’s Disease, Front. Aging Neurosci. 10
Neuroinflammatory Response Mediated by CD4(+)T Cells in Neurodegenerative (2018) 438.
Diseases, Front. Cell Neurosci. 12 (2018) 114. [76] M.C.T. Dos Santos, et al., Evaluation of cerebrospinal fluid proteins as potential
[42] Y. Schmitz, et al., Amphetamine distorts stimulation-dependent dopamine biomarkers for early stage Parkinson’s disease diagnosis, PLoS One 13 (11) (2018),
overflow: effects on D2 autoreceptors, transporters, and synaptic vesicle stores, e0206536.
J. Neurosci. 21 (16) (2001) 5916–5924. [77] M. Hijioka, et al., DJ-1/PARK7: a new therapeutic target for neurodegenerative
[43] I. Miyazaki, M. Asanuma, Dopaminergic neuron-specific oxidative stress caused by disorders, Biol. Pharm. Bull. 40 (5) (2017) 548–552.
dopamine itself, Acta Med. Okayama 62 (3) (2008) 141–150. [78] M. Inden, et al., PARK7 DJ-1 protects against degeneration of nigral dopaminergic
neurons in Parkinson’s disease rat model, Neurobiol. Dis. 24 (1) (2006) 144–158.

7
R. Gibson et al. Brain Disorders 3 (2021) 100020

[79] S.Y. Sun, C.N. An, X.P. Pu, DJ-1 protein protects dopaminergic neurons against 6- [81] M. Inden, et al., Protection against dopaminergic neurodegeneration in Parkinson’s
OHDA/MG-132-induced neurotoxicity in rats, Brain Res. Bull. 88 (6) (2012) disease-model animals by a modulator of the oxidized form of DJ-1, a wild-type of
609–616. familial Parkinson’s disease-linked PARK7, J. Pharmacol. Sci. 117 (3) (2011)
[80] S. Miyazaki, et al., DJ-1-binding compounds prevent oxidative stress-induced cell 189–203.
death and movement defect in Parkinson’s disease model rats, J. Neurochem. 105 [82] K. Yamane, et al., Oxidative neurodegeneration is prevented by UCP0045037, an
(6) (2008) 2418–2434. allosteric modulator for the reduced form of DJ-1, a wild-type of familial
Parkinson’s disease-linked PARK7, Int. J. Mol. Sci. 10 (11) (2009) 4789–4804.

You might also like