ArticlePDF AvailableLiterature Review

Rodent Models and Contemporary Molecular Techniques: Notable Feats yet Incomplete Explanations of Parkinson’s Disease Pathogenesis

Authors:

Abstract

Rodent models and molecular tools, mainly omics and RNA interference, have been rigorously used to decode the intangible etiology and pathogenesis of Parkinson's disease (PD). Although convention of contemporary molecular techniques and multiple rodent models paved imperative leads in deciphering the role of putative causative factors and sequential events leading to PD, complete and clear-cut mechanisms of pathogenesis are still hard to pin down. The current article reviews the implications and pros and cons of rodent models and molecular tools in understanding the molecular and cellular bases of PD pathogenesis based on the existing literature. Probable rationales for short of comprehensive leads and future possibilities in spite of the extensive applications of molecular tools and rodent models have also been discussed.
Rodent Models and Contemporary Molecular Techniques:
Notable Feats yet Incomplete Explanations of Parkinsons
Disease Pathogenesis
Sharawan Yadav &Anubhuti Dixit &Sonal Agrawal &
Ashish Singh &Garima Srivastava &Anand Kumar
Singh &Pramod Kumar Srivastava &Om Prakash &
Mahendra Pratap Singh
Received: 14 May 2012 /Accepted: 13 June 2012
#Springer Science+Business Media, LLC 2012
Abstract Rodent models and molecular tools, mainly
omics and RNA interference, have been rigorously used to
decode the intangible etiology and pathogenesis of Parkin-
sons disease (PD). Although convention of contemporary
molecular techniques and multiple rodent models paved
imperative leads in deciphering the role of putative causa-
tive factors and sequential events leading to PD, complete
and clear-cut mechanisms of pathogenesis are still hard to
pin down. The current article reviews the implications and
pros and cons of rodent models and molecular tools in
understanding the molecular and cellular bases of PD path-
ogenesis based on the existing literature. Probable rationales
for short of comprehensive leads and future possibilities in
spite of the extensive applications of molecular tools and
rodent models have also been discussed.
Keywords Parkinsons disease .Rodent models .
Genomics .Transcriptomics .Proteomics .RNA interference
Introduction
James Parkinson offered the first landmark portrayal on
shaking palsy; however, the name Parkinsonsdisease
(PD) was given by Jean-Martin Charcot [1,2]. PD is
recognized as the most common progressive, baffling, and
devastating neurodegenerative disorder in the elderly after
the Alzheimer disease [3,4]. This movement disorder is
distinguished by the selective degeneration of the nigros-
triatal dopaminergic neurons, accumulation of cytoplasmic
protein aggregates and onset of phenotypic features, such as
resting tremor, rigidity and bradykinesia, etc., leading to loss
of control over the movement [36]. The degeneration of
selective neurons is accountable for the decreased dopamine
level in the striatum that ultimately results in the clinical
manifestations [6]. Although an early diagnosis is dreadfully
difficult, physical and clinical examinations and sympto-
matic features are used to diagnose the patients after consid-
erable dopaminergic neurodegeneration and manifestation of
noticeable complications [7]. Moreover, the comprehensive
explanations of pathogenesis and permanent cure are not yet
established, and therapeutic and surgical procedures offer
provisional aids [7,8].
One of the most commonly accepted notions for the onset
of symptomatic features of PD is the resultant interplay of
the environmental factors, increased age, and genetic sus-
ceptibility of an individual [3,5,9]. Administration of 6-
hydroxydopamine (6-OHDA), 1-methyl-4-phenyl-1,2,3,6-
tetrahydropyridine (MPTP), reserpine, methamphetamine,
rotenone, maneb, zinc, manganese, paraquat, and cyper-
methrin in rodents develop many symptomatic features
mimicking PD [914]. These chemicals either alone or in
combination inhibit mitochondrial function resulting in
depleted energy metabolism, free radical generation, and
neuroinflammation leading to programmed cell death and
selective neurodegeneration [1519]. Some environmentally
relevant chemicals directly cross the bloodbrain barrier
(BBB) and enter the brain owing to their lipophilic nature,
such as MPTP and rotenone. Others, those are hydrophilic in
Authors Sharawan Yadav and Anubhuti Dixit contributed equally to
this work.
S. Yadav :A. Dixit :S. Agrawal :A. Singh :G. Srivastava :
A. K. Singh :M. P. Singh (*)
CSIR-Indian Institute of Toxicology Research (CSIR-IITR),
M.G. Marg, Post Box 80, Lucknow -226 001, Uttar Pradesh, India
e-mail: singhmahendrapratap@rediffmail.com
P. K. Srivastava :O. Prakash
Banaras Hindu University,
Varanasi -221 005, Uttar Pradesh, India
Mol Neurobiol
DOI 10.1007/s12035-012-8291-8
nature, require either specific transporters, such as paraquat
that requires a neutral amino acid transporter or needs to be
imported directly in the target tissue, such as 6-OHDA [9,14].
Environmental factors could contribute notably, if the genetic
factors clutch them appropriately, as every synthetic heroin
(which contains MPTP) user does not develop PD-like symp-
toms. Pesticides and other environmental chemicals, when
enter the body and subsequently in the brain, get converted
by the phase I and II xenobiotic metabolizing enzymes either to
more or less toxic intermediary metabolites. The bioconversion
is directly proportional to the catalytic activity, protein expres-
sion, and DNA sequence ofthe coding and non-coding regions
of a gene, which encode the enzyme [20]. Twin analysis,
omics, and RNA interference (RNAi) further supported the
genetic bases in a few cases and deciphered autosomal pattern
of inheritance of many PARK genes [5,2123]. Omics and
RNAi also deduced many indefinable aspects, which include
the identification of molecular fingerprints and molecular
explanations of PD pathogenesis [7,22]. Although amalgama-
tion of modern techniques and multiple rodent models were
expected to offer novel clues to disease pathogenesis and
molecular biomarkers, clinical and phenotypic symptoms are
still used as the gold standard to diagnose PD [7]. Despite
noteworthy and extensive endeavors made by rodent models
and molecular tools to pinpoint the biochemical, clinical,
pathological, epidemiological, and molecular bases of disease
pathogenesis, the complete molecular machinery of PD patho-
genesis is still mysterious [21,22,24].
Salient characteristics, achievements, and limitations of
various rodent models and/or molecular tools employed for
understanding PD pathogenesis have been comprehensively
reviewed elsewhere [35,10,21,22,2427]. This article
updates the contributions made by imperative rodent mod-
els, omics, and RNAi approaches altogether based on the
information available in the literature along with the reasons
why such sincere efforts could not yet embrace the desired
success. Attempts are also made to portray likely explan-
ations for lack of comprehensive translation of the informa-
tion generated from rodents to humans.
Contributory Factors of PD
Since most of the rodent models are based on the alleged
causative factors, it is worthwhile to discuss about the undeni-
able and suspected contributory factors of sporadic PD.
Undoubtedly, aging is the main perpetrator, as it has been
found to increase the incidences of PD in humans. It is
estimated that approximately 1 % of the population of 50
60 years of age may develop PD, and the incidences may go
up further in elderly individuals [5]. The effect of age is
reflected even in the experimental rodent models, as the aged
rats have been found to be more susceptible to a chemical that
induces the nigrostriatal dopaminergic neurodegeneration
[13]. Environmental factors that include pesticides exposure,
rural living, well water drinking, and head trauma could be
other contributory factors, as evidenced from epidemiological
studies and/or rodent experimentations [35].
Appearance of the disease warning signs in an early age
has been limited; however, onset of the disease in the
individuals below 50 years of age gave a notion that genetic
factors could play a critical role in PD pathogenesis. Now,
the familial PD is well recognized by the twin analyses and
case studies [5]. Several genes have been mapped and are
suspected to manipulate PD pathogenesis in the genetically
susceptible persons owing to point mutations and inappro-
priate epigenetic regulation [2830]. Despite the fact that
the majority of the patients do not possess familial history,
absence of cardinal differences between the sporadic and
familial forms and the lack of symptoms in a few patients
carrying defective genes, point out the influence of herit-
able factors on the age of onset, particularly when the
appropriate environmental conditions are available [5,24,
30,31].
Rodent Models
Many chemicals, which include pesticides and metals, turn
out to be inseparable parts of the environment, and their
involvement in PD pathogenesis could be incredible. A few
pesticides and metals may lead to PD in humans and also
reproduce PD-like symptoms in the exposed experimental
rodents [5,25,32,33]. Salient characteristics and pros and
cons of a few extensively used rodent models are described
below.
6-OHDA
Even after several decades of history of its use for develop-
ing PD-like features in rats, 6-OHDA is still widely used for
the same purpose [21]. The severity of the symptoms pro-
duced by 6-OHDA depends on the site of its administration
in the substantia nigra and the extent of the lesions [32]. The
6-OHDA model provided new insights to understand PD
pathogenesis and validated pre-existing information gath-
ered from sporadic cases and also from other rodent models.
6-OHDA offered multifaceted confirmations of the degen-
eration of cell bodies of dopaminergic neurons in the sub-
stantia nigra and fibers in the striatum [34], the fundamental
features of sporadic PD in humans. Mitochondrial dysfunc-
tion has been widely accepted as the priming event leading
to oxidative stress and thereby the nigrostriatal dopaminer-
gic neurodegeneration. 6-OHDA mimics sporadic PD in the
sense that it inhibits mitochondrial electron transport chain
complex I (nicotinamide adenine dinucleotide-ubiquinone
Mol Neurobiol
reductase), generates free radicals, and induces programmed
cell death [4].
6-OHDA induces neurodegeneration by multiple means.
Oxidative stress is critical in PD pathogenesis whether it is
generated by the mitochondrial complex I inhibition or owing
to other means. Although growth arrest and DNA damage-
induced gene 153 is also reported to act as a neuronal cell
death mediator, a null mutation of the gene results in reduction
of apoptosis in the 6-OHDA model [35]. Similarly, free rad-
icals produced owing to 6-OHDA exposure causes several
changes in dopaminergic neurons that mediate neurodegener-
ation. Fos expression also mediates some of the abnormal
sensory circuits of neurodegeneration in 6-OHDA-treated rats
[36]. Proteases, such as caspases, are known to regulate apop-
totic machinery and induce toxicity in dopaminergic neurons.
Caspase-3 mediated proteolytic cleavage and activation of
protein kinase C-δis reported to be decisive in dopaminergic
neurodegeneration and cleavage resistant form of the protein
is found to protect against apoptosis in 6-OHDA-treated
rodents [37]. 6-OHDA augments unfolded protein stress,
causes cytochrome-c release and upregulates pro-apoptotic
proteins, which include caspases and p53 [38]. 6-OHDA
downregulates tumorous imaginal discs 1 protein, which ham-
pers functional and structural compensation and exacerbates
neurodegeneration [39]. Moreover, the 6-OHDA model high-
lighted the fact that auto-oxidation of dopamine into free
radicals may also lead to PD-like features [32].
6-OHDA neither induces the formation of the Lewy
bodies nor produces the similar degree of phenotypic symp-
toms in all experimental rodents [24,32]. Owing to inability
of 6-OHDA to cross the BBB and directly enter the brain
[4], the nigrostriatal administration is required, which is tricky
and cumbersome. Due to lack of its environmental occurrence
and direct human exposure, and inconvenient delivery in brain
the 6-OHDA model could not be considered very ideal for
extrapolation of the results to humans and is now becoming
less popular as compared with its popularity in the past [24].
MPTP
MPTP itself is not as toxic as its intermediary metabolite, 1-
methyl-4-phenylpyridinium cation (MPP
+
), which is highly
reactive and severely neurotoxic [4,16,25,40]. Monoamine
oxidase B (MAO-B) of the astrocytes synthesizes MPP
+
from
MPTP [4,25,41]. The organic cation transporter (Oct)-3
present in the astrocytes (glial cells) and dopamine transporter
(DAT) localized on neurons regulate the entry of MPP
+
into
dopaminergic neurons (Fig. 1)[42]. The MPTP rodent model
supported the notion that the mitochondrion is an epicenter of
PD pathogenesis [40]. Although it is not yet clear whether
mitochondrial depolarization or free radical generation after
the mitochondrial complex I inhibition is the most critical event,
the MPTP model elucidated the contributions made by the
nicotinamide adenine dinucleotide phosphate (NADPH) oxi-
dase, microglial cells, neuronal inflammation, and secondary
signaling molecules, including p38 mitogen activated protein
kinase, c-Jun N-terminal kinase (JNK) and tumor suppressor
protein 53, in PD pathogenesis [10,25,43]. MPTP triggers the
activation of JNK, which is regulated by GST Pi protein via
proteinprotein interactions [44]. Although the role of free
radicals and microglial activation in the MPTP model is estab-
lished, a recent study has shown that MPTP-induced dopami-
nergic neurodegeneration primarily depends on the free radical
outburst and activation of NADPH oxidase in dopaminergic
neurons,and the activationof microglial NADPHoxidase takes
place at later stage [45]. Furthermore, theMPTP model showed
that phosphorylated Akt/protein kinase B depletion hinders
with the normal functioning of cell survival [46].
PINK 1 and DJ-1, two known neuroprotective molecules,
modulate the dopaminergic sensitivity towards MPTP-
induced toxicity [24]. This assumption gave a conduit of
how genetic factors play important roles even in toxin-
induced disease pathogenesis. It is validated by a recent
study in which loss of PARK 7 (DJ-1), a cellular target, is
found to be associated with the modulation of MPTP-
induced PD phenotype. In this study, a cell permeable Tat-
DJ-1 protein is reported to protect dopaminergic neurode-
generation by reducing the effects produced by MPTP-
mediated oxidative stress [47]. MPTP model elucidated the
inputs of peroxisome proliferator-activated receptor γin
dopaminergic neuroprotection and treatment outcomes
[48]. Moreover, the MPTP model deciphered the roles of
heat shock proteins (HSPs), such as HSP1b, in the regula-
tion of neurodegeneration, as knocking down the HSP1b
gene increases the vulnerability of dopaminergic neurons
[49]. Neurotrophic factors and apolipoprotein could encoun-
ter PD pathogenesis and help in the repairing of dopaminer-
gic neurons, as their expressions are increased in MPTP-
induced rodent models [50,51]. The MPTP model also
improved our awareness about the contribution of environ-
mental factors, the mechanisms of PD pathogenesis and
therapeutic strategy to encounter the disease.
MPTP develops an acute rodent model; low doses and
long-term exposures may help in reproducing chronic PD
phenotype [52]. Despite species to species variation,
absence of slow and progressive degeneration and distinct
Lewy body formation, the MPTP model is widely used to
understand pathogenesis and to appraise the efficacy of anti-
PD chemical entities [25,40,41,52].
Paraquat and Maneb
Initially, 1,1-dimethyl-4,4-bipyridinium dichloride (para-
quat), which has a structural similarity to MPP
+
and produ-
ces neurotoxicity, was assessed for neurodegenerative
potential in the experimental rodents [53,54]. Paraquat
Mol Neurobiol
enhances alpha (α)-synuclein-induced disruption of mem-
brane integrity and increases the conductance, as a result of
increased oxidative stress [55]. Later on, a fungicide, man-
ganese ethylene bis-dithiocarbamate (maneb) was also
tested for its potential in experimental animals either alone
or with paraquat [15]. Structural and functional anomalies in
the endoplasmic reticulum (ER) and mitochondrion are
found to be associated with PD pathogenesis. Paraquat
slows down mitochondrial complex I activity and augments
the microglial activation and free radical production through
NADPH oxidase, while maneb reduces mitochondrial com-
plex III activity and both of them together or individually
may lead to oxidative stress, DNA damage, defective energy
metabolism, and cellular apoptosis (Fig. 1)[4,10,15,25,
56]. Roles of ER and mitochondrion in PD pathogenesis are
also validated in the paraquat alone induced dopaminergic
neurodegeneration model in a study. ER stress and mito-
chondrial dysfunction were found to trigger caspase-12
activation, hydrogen peroxide release, and PARK 13
(HtrA2/Omi) activation; however, minocycline, a microglial
activation inhibitor, encountered such alterations [57].
Toxicant responsive genes, such as cytochrome P450
(CYP/Cyp) 2D6, play critical roles in PD pathogenesis and
treatment outcomes. The expression of Cyp2d22, a human
CYP2D6 ortholog in the mice, is increased in maneb and/or
paraquat-induced PD phenotypes, showing the role of detox-
ification machinery in the disease pathogenesis [20]. Toxicity
of pesticide-induced PD depends on the metabolic conversion
of pesticides in to too high or too low toxic species. The
conversion of paraquat dication to paraquat cation and sub-
sequent transport to dopaminergic neurons mainly depend on
Oct-3 and DAT [58]. Maneb and paraquat induce apoptosis
through Bak-dependent pathway when administered alone;
however, on combined systemic exposure, they follow the
Bax-dependent apoptotic pathway [59]. Although it is known
that males are often at the higher risk for PD as compared with
females, supportive evidence from an animal experimentation
is provided in a recent study in which the effect of paraquat
was monitored. Paraquat administration was found to increase
the brain-derived neurotrophic factor (BDNF) expression in
the hippocampus of female rats, which was responsible for
reduced susceptibility of the females towards the nigrostriatal
NADH-
ubiquinone
oxidoreductase
Succina te -
ubiquinone
oxidoreductase
Ubiquinol -
cytochrome c
redu cta s e
cytochrome
coxidase
6-OHDA
PARAQUAT MANEB
DOPAMINERGIC NEURON
MPTP
MITOCHONDRIA
BBB
MPP+
MAO-B
F1Fo-ATP
syntha se
ATP
MAIN PROS
AND CONS
Specific to
injected site
Direct
administration
to the target site
is required, as it
can not cross the
BBB
MAIN PROS AND
CONS
Mimics most of the
features of sporadic PD
but can not induce
Lewy body f ormation
An acute mod el,
however, alteration in
dose and treatment
schedule can make this
suitable as chron ic
MAIN PROS AND
CONS
Induces slow and
progressive
degeneration in the
mice
Reported toxic in
rats, if admin ister ed
for longer duration
Lipoph ilic
DAT
MPP+
6-OHDA
6-OHDA
CYPERMETHRIN
Hydrophilic
Lipophilic
Neutral amino
acid transporter
PQ
+2
METHAMPHETAMINE
ROS
Lipophilic
PQ
+
Astroc yte
ROTENONE
MAIN PROS
AND CONS
Forms Lewy
bodies
Non specific
neurodegenerat
ion
Lipophilic
Rotenone
Maneb
MAIN PROS AND
CONS
Causes slow and
progressive
dopaminergic
neurodegeneration in
rats
Mechanism is not yet
completely known
MAIN PROS AND
CONS
Degeneration of the
striatal nerve terminal s
only
Not suitable for
mechani st ic
understanding
OCT-3
Lipophilic
DA
Syna ptic
vesicle
Extracellular DA
DAT
Metham phetamine
autooxidation
PQ
+
Microglial
activation
Cypermethrin
Hydrophilic
MPTP
-
-
-
-
-
Needs direct target-
specific a dministration
Microglia
OCT-3
Fig. 1 Routes of entry of the major PD-inducing chemicals in the
brain, their enzymatic/non-enzymatic conversion into active radicals in
glial cells, subsequent entry into dopaminergic neurons, and the inhib-
itory effects produced by them at the level of mitochondrial complexes
or dopamine auto-oxidation [4,10,24,25,42,58,68]. 6-OHDA,
MAO-B, Oct-3, DAT, PQ
2+
,PQ
+
, MPTP, MPP
+
, NADH, DA, ATP,
BBB, and ROS abbreviate 6-hydroxydopamine, monoamine transporter
B, organic cation transporter-3, dopamine transporter, paraquat dication,
paraquat cation, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, 1-
methyl-4-phenylpyridinium cation, reduced nicotinamide adenine
dinucleotide, dopamine, adenosine triphosphate, bloodbrain barrier,
and reactive oxygen species, respectively
Mol Neurobiol
dopaminergic neurodegeneration as compared with males, as
decreased BDNF expression was noted in males [60].
Despite mild to severe toxicity of paraquat and/or maneb
administrations in rodents after prolonged exposure [9,61],
the use of their combination in rodents is widely accepted
and offers two main advantagesinevitable human expo-
sure that makes them environmentally relevant and ability of
progressive and slow neurodegeneration [61]. Epidemiolog-
ical studies have also shown the direct relevance of maneb
and paraquat co-exposure to humans [25,62].
Rotenone
Like other animal models, the rotenone model also exhibits
mitochondrial complex I inhibition and induction of oxida-
tive stress and apoptosis [4]. The release of cytochrome c is
found to be independent of caspase activation in rotenone-
induced dopaminergic neurodegeneration. This phenome-
non exists even in the presence of cytoplasmic cytochrome
c release after mitochondrial complex I inhibition [63].
Matrix metalloproteinase-3 (MMP-3) is also implicated in
dopaminergic neurodegeneration induced by rotenone,
which is regulated by multiple mechanisms. MMP-3
requires activation from proMMP-3 by an intracellular ser-
ine protease. Under stress, HtrA2/Omi, a mitochondrial
serine protease, translocates into the cytosol, causes MMP-
3 activation, and triggers apoptosis in dopaminergic cells
[64].
Although rotenone readily enters the brain, inhibits mito-
chondrial complex I, generates free radicals, exhibits Lewy
bodies formation and other anatomical and phenotypic
symptoms of PD (Fig. 1)[65], it is not as popular as MPTP
or 6-OHDA, owing to its non-specificity, high mortality
rate, and severity of the lesions [9,10,41].
Other Pesticides
Epidemiological studies showed the contribution of a few
other pesticides in increased incidences of PD [5,33,66,
67]. Several classes of pesticides, which include, pyrethroids,
dithiocarbamates, organochlorines, and organophosphates
have been reported to induce PD-like symptoms in the exper-
imental rodents [68,69]. As these pesticides are commonly
used globally, therefore, could be of major health concern.
Dieldrin and cypermethrin induce neurodegeneration in the
adult experimental animals after prolonged exposure [13,69].
Postnatal pre-exposure of cypermethrin is also found to
enhance the susceptibility of the animals, when re-exposed
upon adulthood [13]. Despite the fact that little is known about
the mechanism of cypermethrin-induced neurodegeneration,
the neuronal loss is specific to dopaminergic neurons of the
nigrostriatal pathway [68]. Moreover, its effect on microglial
activation is known, but prolonged opening of ion channels or
mitochondrial dysfunction could possibly be the most impor-
tant events involved therein [19,68].
Methamphetamine
Methamphetamine produces dopamine depletion leading to
temporary or permanent disturbance in the dopaminergic
system after chronic or intermittent exposure and has estab-
lished the role of growth factors, particularly glial derived
neurotrophic factor [10,70]. Methamphetamine selectively
reduces phasic, but not tonic, dopaminergic signaling in the
striatum [71]. Methamphetamine-induced response is found
to be age dependent in the primates owing to age-related
changes in the neurotrophic capacity of the striatal dopa-
mine system [72]; however, in rodents, such information has
not yet been reported. As methamphetamine depletes dop-
amine level in the striatum, the effects of such drugs have
also been studied in the exposed humans to assess the actual
risk. Drug users are found to have a high risk for developing
PD as compared with unexposed individuals [73].
Methamphetamine-induced neurotoxicity also involves the
striatal vasoconstriction leading to hypoxia and dopamine
reduction in the exposed individuals [74].
In general, the methamphetamine model lacks many fun-
damental features of PD and is considered as a dopamine
depletion model rather than a true PD model.
Metals
Apart from chemicals, several metals, including iron, lead,
zinc, and manganese have been found to be associated with
PD pathogenesis, as abnormal metal exposures or accumu-
lations have been associated with the increased incidences
of PD in humans or PD-like pathology in experimental
animals [12,75,76]. Metal accumulation may lead to
PDcame into existence, when the postmortem brain of
PD patients was seen to possess high level of metals [75].
Epidemiological studies revealed that zinc, lead, and iron
are increased in the substantia nigra of PD patients, whereas
the copper level is decreased. Like pesticides, metals induce
free radical biosynthesis leading to the degeneration of
dopaminergic neurons of the nigrostriatal pathway [77].
Metals catalyze the formation of free radicals either by
Fentons reaction or directly by reacting with macromole-
cules, which lead to the generation of fatty acid radicals and
4-hydroxynonenal causing DNA damage and apoptosis.
Additionally, the metals, such as cadmium, arsenic, and lead
bind to sulphydryl group of proteins and lead to depletion of
glutathione [76]. Zinc, another metal, also induces oxidative
stress via the activation of NADPH oxidase and depletion of
glutathione, which in turn activate the apoptotic machinery
leading to dopaminergic neurodegeneration similar to para-
quat [11,78]. Although metal-based rodent models have not
Mol Neurobiol
yet been fully understood, the contributions of metals in
increasing oxidative stress and reducing antioxidant
defense system cannot be straightforwardly ignored
[11,76].
Like other chemical-induced PD models, metal models
lack many fundamental features of sporadic disease. How-
ever, more exposure time is required to develop a few
cardinal features of the disease in rodents like that of spora-
dic PD in humans.
Some Other Toxins
In addition to the above-mentioned toxins, many chemicals
contribute to PD pathogenesis as reported in epidemiolog-
ical investigations and/or from animal experimentations.
Majority of such chemicals inhibit the mitochondrial com-
plex I or elicit neurotoxicity by multiple mechanisms
[7981]. Trichloroethylene, a complex I inhibitor, is
reported to cause PD-like features in humans upon exposure
and also elicit motor impairment in experimental animals.
Its oral exposure to experimental animals for 6 weeks selec-
tively inhibits complex I and leads to the nigrostriatal dop-
aminergic neurodegeneration [79]. Annonacin, another
mitochondrial complex I inhibitor, causes the nigrostriatal
dopaminergic neurodegeneration by impairing the energy
metabolism [81]. Several bacterial neurotoxins, from which
humans and aquatic animals are exposed, could also elicit
PD-like features. β-Methylamino-L-alanine, a cyanobacte-
rial neurotoxin, elicits a few features quite similar to PD
[80]. Isoquinoline derivatives, such as tetrahydroisoquino-
line, elicit PD-like features in the animals possibly because
of its structural similarity with MPTP [82,83]. Similarly,
haloperidol, a dopamine D2 receptor blocker; epoxomixin, a
proteasome inhibitor; and 3,4-dihydroxyphenylacetalde-
hyde, a dopamine metabolite, also cause some or the other
characteristic feature of PD [8486]. Such agents although
not yet very widely studied, could be tested across multiple
studies either alone or in combination with established and
widely studied toxins for developing better rodent models to
understand PD pathogenesis and to assess the efficacy of
neuroprotective agents.
Genetic Models
The role of genetic factors becomes vital when the mutated
gene carrier is exposed to pesticides and heavy metals. The
genetic theory of PD came into existence from the studies
related to close relatives and twins who were diagnosed with
an early onset of the disease [5]. The inheritance of genes,
critical for an early onset of the disease, follows either
autosomal dominant or autosomal recessive pattern [87].
Early onset PD-related genes are mapped in the specific
chromosomal locations, together designated as the PARK
loci [88,89]. α-Synuclein protein is encoded by PARK 1
and 4, Parkin by PARK 2, ubiquitin carboxyl terminal
hydrolase (UCH) L-1 by PARK 5, PTEN-induced kinase 1
(PINK1) by PARK 6, DJ-1 by PARK 7, leucine-rich repeat
kinase 2 (LRRK2) by PARK 8, ATP13A2 by PARK 9 and
Omi/HtrA2 by PARK 13 genes [88,89]. Moreover, many
other genes of elusive nature or function have also been
mapped as PARK 3, PARK 10, PARK 11, and PARK 12
[8789]. Apart from PARK genes, synphilin 1, paired like
homeodomain transcription factor 3 (Pitx 3) and nuclear
receptor-related factor 1 genes, etc., have also been mapped
at various chromosomal locations and could possibly influ-
ence the genetics of the disease [28,29].
Genetic models proved to be quite useful in under-
standing familial PD pathogenesis. Owing to absence of
most of the critical phenotypic symptoms altogether and
the roles of all the genes involved in PD make little
sense of genetic models in understanding the sporadic
PD.
Dual Models
Several rodent models of the genetically linked PD have
been developed by knocking down the critical gene(s)
responsible for familial forms of the disease [65,90,91].
By and large, the genetic models provided evidence-based
proofs of the molecular mechanisms elucidated by the
toxins-induced rodent models [24,65,90]. Recently, dual/
fusion/combinational/two-hit rodent models have been
generated to test the hypothesis that the environmental
factors mainly act on the genetically susceptible individuals,
as apt environmental factors and suitable genetic makeup
together could be decisive for an early onset of PD [92].
Dual rodent models are mainly based on the principle of the
combination of two contributory factors for creation of a
new animal category to study the multifactorial etiology and
to reiterate the major cardinal disease symptoms [24,92,
93]. Generation of such model is an animated tool to com-
prehend PD in the conditions, which maximally imitate
idiopathic PD [92,94]. For example, lipopolysaccharide
(LPS) induces several neuroinflammatory molecules that
include nitric oxide, cytokines, and interferons, while α-
synuclein dysfunction is associated with its abnormal
accumulation, but low-dose of LPS and dysfunctional α-
synuclein together may cause all the above-mentioned
events and significantly mimic idiopathic PD features [24,
94].
Since dual models could be chronic and progressive in
nature as far as dopaminergic neurodegeneration is con-
cerned, they prove to be quite useful to investigate the
mechanistic and therapeutic aspects of idiopathic PD [24,
94]. Fusion models need to be developed across laboratories
all over the world employing major causative gene(s) and
Mol Neurobiol
toxin(s) for wider acceptance and real implication in under-
standing sporadic PD.
Advantages and Limitations of Rodent Models
Over Other Models
Despite the fact that nonhuman primate models proved to be
very useful in understanding a few decisive aspects of PD
pathogenesis, rodents have many advantages over primates
owing to several reasons. Convenient availability, housing,
maintenance, handling, and plausible use of a large number
of experimental animals per set for generation of much
reliable data, make rodents the preferred choice over pri-
mates [95]. Additionally, it is possible to test multiple toxins
alone as well as in combination to assess their neurodegen-
erative potential in the limited time. Although nonhuman
primates are close to humans, generation to generation stud-
ies to assess the role of a particular gene [96], or the effect of
toxins could be easily performed in rodents within the short
span as compared with nonhuman primates. Nonetheless,
the short life span of rodents does not always reflect an
advantage, as the sporadic disease in humans generally
appear very late and after long-term exposure to environ-
mental factors. Owing to such advantages and disadvan-
tages, many suspected toxins have been widely tested to
check neurodegenerative potential in rodents, but limited
studies are available with primates [32,97]. Although lower
animal models that include, Drosophila, zebra fish, nemat-
odes have been continuously used to reveal the familial PD
and genetic bases of onset and progression of the disease
[98], successful application of such models for more than
90 % of the cases of the disease, which are sporadic in
nature, is difficult. Furthermore, the genetic makeup of
lower animals does not share a noticeable resemblance with
humans, if a comparison is made with rodents or primates
[99], leading to a possibility that data obtained from the
toxicological response to a PD toxin or pharmacological
response of anti-PD compounds could vary significantly
and data extrapolation in humans would become more
difficult.
No Model Is Absolutely Close to Ideal to Understand PD
Pathogenesis
None of the rodent models developed so far can be said to
be perfectly ideal since an ideal animal model should be an
indicator of the multifactorial etiology and reproduce slow,
progressive, and exposure-dependent onset of the pheno-
typic, behavioral, biochemical, and anatomical impairments
along with the secondary changes associated with sporadic
PD [24]. The major achievements of rodent models have
been the confirmation of the contributory roles of pesticides
and heavy metals in PD pathogenesis. Rodent models also
validated the contributory roles of environmental factors,
which were reported from the epidemiological studies [5,
11,25]. Rodent models have shown that susceptibility to
environmental neurotoxins is age dependent, as in sporadic
PD. The postnatal exposure enhances the vulnerability upon
adulthood in the rats [13] also validates the theory that age is
the most important contributory factor. Although rodent
models gave a validation of the major contributory factors
reported through epidemiological or clinical investigations,
rodent models fail to offer the role of newer contributory
factors or all the culprits of PD [100,101].
Chemically induced rodent models established the role of
mitochondrial dysfunction, defective ubiquitin proteasomal
pathway and energy metabolism, apoptosis, neuroinflamma-
tion, microglial activation, dopamine autoxidation and oxi-
dative stress in the pathogenesis of PD [4,10,101]. Many
chemicals used to mimic PD symptoms in rodents lead to α-
synuclein aggregation (but lack of defined Lewy body for-
mation, except in rotenone) and subsequent cell death owing
to their ability to inhibit proteasomal and mitochondrial
functions. The results obtained from rodent models substan-
tially mimic the results obtained from the clinical samples
and postmortem brain of PD patients [101]. Owing to such
similarities, rodent models are also used to test the thera-
peutic efficacy of several drugs and natural products and
also to design new modes of therapy for the disease [52].
Despite all possible efforts, none of the current models
completely mimics sporadic PD [100,102]. The difference
in the life span from humans could also contribute to the
absence of cardinal pathogenic features of sporadic PD,
such as Lewy body formation in rodents. Furthermore, lack
of a neuromelanin and distinct regulatory pattern of tyrosine
hydroxylase (TH) makes rodent models less reliable [101,
102]. Although immunohistochemical and biochemical
observations exhibited the similar pattern of results, the
degree of differences is still enormous not only between
sporadic and chemically induced PD but also among various
rodent models [100]. A few of them develop PD symptoms
after acute exposure contrary to the chronic progression of
sporadic and familial forms of PD in the humans [68,100,
102]. A few others though chronic in nature, are either not
yet established unequivocally or reported from specific lab-
oratories or do not produce the cardinal features of PD
(Fig. 1)[9]. For example, rotenone is nonspecific, MPTP
mainly leads to rapid neurodegeneration, and paraquat may
lead to death of many experimental animals at the concen-
tration and time of exposure, which induce PD phenotype in
mice. The development of defined Lewy body, one of the
basic hallmarks of sporadic PD, is reported only in a
rotenone-induced rodent model [25,32]. Moreover,
reserpine and amphetamine do not produce significant
Mol Neurobiol
degeneration of dopaminergic neurons and pesticides, and
metals possess one or the other drawbacks detailed above
[52,65,100]. Dual rodent models are relevant to PD patho-
genesis and could be more appropriate for assessing the
efficacy of therapeutic agents [92]; however, developing
such models in experimental rodents is difficult, cumber-
some, and cost-consuming.
While it seems quite difficult to translate findings of
rodent models in humans, as most of rodent models possess
different chromosomal localization of a few identified
causative genes with significant degree of differences in
behavior, environment and anatomy of the brain and
responses towards a toxin as compared with humans, it is
expected that after the advancement of newer tools and
development of an ideal animal model, it would become
easier [100102]. Most of the neuroprotective agents, which
succeeded in preclinical investigations in rodents, could not
be successfully translated into clinical interventions. For
proper translation of the mechanistic observations and
extrapolation of rodent data to humans, it is essential to
develop and validate newer rodent models that could help
to overcome the disparity and drawbacks in connection with
the current animal models.
Omics and RNAi in Understanding PD Pathology
Omics tools, such as genomics, proteomics and transcrip-
tomics, are large-scale technologies used to generate a pleth-
ora of information based on genetic variations and global
expression profiles of genes and proteins and to identify the
differentially expressed transcripts/proteins. Omics-driven
information provided clues to identify the individuals at
high risk, develop molecular fingerprints for diagnosis as
well as for discrimination of various stages of PD patho-
genesis (Fig. 2)[3,26,103,104].
Single Nucleotide Polymorphisms
Genome-wide association studies of several single nucleotide
polymorphisms (SNPs) with PD-linked genes are reported
[105]. For example, SNPs in various forms of synuclein (SNC-
A, SNC-B, and SNC-G), Parkin, UCHL1, PINK1, DJ1,
LRRK2,ATP13A2, and Omi/HtrA2 genes and their association
with PD are recently established [105107]. Polymorphism in
mitochondrial DNA-encoded complex I gene validated the role
of mitochondria in PD pathogenesis [108]. As most of such
studies are performed in humans rather than rodents, such
GENOMICS: Facts
Identificat ion of SNPs
in PARK genes -
SNCA, UCHL1, DJ-1
and LRR K2, etc.
Evidence to genetic
basis of diseases
Inconsistency in
results owing t o small
sample size and
environmental and life
style factors
PROTEOMICS:Facts
Establishes the role of pathogeni c
modifications of proteins in PD related genes ,
proteasomal proteins and peroxiredoxins
Provid es many prot ein-prot ein in teract ions ,
post-translational modifications and
diff erential protein expression pat terns
Lack of proper tool t o indentify membrane
bound and high and low abundant proteins
Reproducibility of experiments is a tricky
issue
TRANSCRIPTOMICS:Facts
Establishes the roles of ILs,
cyclins an d G STs
Val id at e s rol e s of mu lt i p l e
pathways in PD
Needs quantitat ive tools for
valid atio n
Presen ce of f alse po sitive a nd
negative transcripts
Short life of most of the
transcripts
RNAi: Facts
Explores early events and
validates the roles of
suspected genes
Helps in designing
therapeutic strategy by
targeting desired RNA
Off-targeting and
undesired immune
responses are the major
pitfalls
GENOMICS: Facts
Ident ification of SNPs of
mitochondrial genes
suspected to part icipate in
PD pathogenesis
Provides evidence that
mitochondrion is the
epicenter of PD
Interpretation of the
results with absolute
cert ainty is dif ficu lt due to
sample size
TRANSCRIPTOMICS: Facts
Establishes the roles of
mitochondrial genes in PD
pathogenesis
Investigates mitochondrial
gene network and their
interactions
Lacks fully reprod ucible
patterns across various studies
owing to systemic and global
variations
PROTEOMICS: Facts
Esta blishes the ro les of mito chond rial prot eins, suc h
as NADH-ubiquinone oxidoreductase and DJ-1
mainly in the stria tum
Investigates mitoch ondrial protein expression
patterns and their interactions
Yields specific but not absolutely reproducible
patterns because of multiple reasons leading to
biological variations, mainly in the substantia nigra
Extremely diff icult to isolate pure mitochond rial
fraction
RNAi: Facts
Validate s the
roles of PINK
1and Parkin
Substitutes gene
knockouts
Difficult to
target the brain
NEURON
RNA
PROTEIN
Exogenous
siRNA
miRNA
Mitochondria
Nucleus
RNA
DNA
mRNA
PROTEIN
mtDNA
Golgi bod y
Endoplasmic
reticulum
Fig. 2 Contribution of genomics, transcriptomics, proteomics, and RNAi in understanding PD pathogenesis at cellular and organellar levels and
their pros and cons in understanding PD pathogenesis [3,5,21,22,26,27,106108,116,137]
Mol Neurobiol
investigations are not being discussed in detail. In summary,
conflicting reports are available in literature even in the same
population. The results of a few investigations revealed the
association of SNPs of the selected genes with PD; however,
others have shown lack of such associations [105,107].
Transcriptomics
The differential transcription profiling has been used to
simultaneously assess the role of many transcripts in PD
pathogenesis [109]. Several groups of investigators working
in this area have generated enormous information employ-
ing various rodent models. The main biological pathways
involved in PD pathogenesis, which could get consensus
among the studies, are neurotransmission, dopamine metab-
olism, biodegradation and transportation, oxidative stress,
mitochondrial function, energy metabolism, neuroinflam-
mation, protein accumulation and degradation, and apopto-
sis (Table 1)[110115]. Several studies concentrated on the
role of transcripts derived from nuclear origin; however,
studies are also available in which the role of the mitochon-
drial system is assessed. A study based on mitochondrial
transcripts profiling has shown specific susceptibility of the
striatum for oxidative phosphorylation deficiency [116].
Transcriptional profiling of brain and blood showed an
alteration in SNC-A as a critical pathogenic feature [117].
Microarray has been extensively used to assess the changes
in gene expression patterns in most of toxins-induced rodent
models, except a few models that were recently reported
[118122]. Microarray data established the roles of many
genes that are associated with familial and toxins-induced
PD. For example, Parkin and α-synuclein are reported to
play critical roles in the regulation of ubiquitin proteasomal
pathway and subsequently in neurodegeneration and neuro-
protection [110]. Although many studies have also been
conducted using human sample, which can provide direct
information about the alteration in the gene expression at the
later stages of PD [109,110], such information is not dis-
cussed in detail as the main purpose of the article is to
discuss rodent models. Transcriptomics can even be done
with the human samples as well, since the mRNA of the
postmortem brain can be stable up to 36 h after death [123].
Proteomics
Classical proteomics and modern quantitative proteomics
in combination with bioinformatics are used to analyze
the expression levels and posttranslational changes in
the proteins of the tissues and biological fluids [124,
125]. Nigrostriatal proteomics starting from the postmor-
tem brain up to animal models not only transformed the
understanding of the genetic basis of the disease but
also provided information to develop therapeutic
strategies to encounter PD by identifying specific targets
[126]. Posttranslational modification-based molecular finger-
prints could be identified and protein localization and trans-
location can be recognized by employing proteomic tools
[127129]. Furthermore, proteomics provided the clues to
PD pathogenesis by generating novel information or by vali-
dating the traditional assumptions and established the roles of
mitochondrial dysfunction, oxidative stress, kinase and
autophagy modulators, and disturbances in protein aggrega-
tion and degradation [130134]. Proteomic approaches,
applied to various toxins-induced rodent models, demonstra-
ted that the alteration in the proteins related to mitochondrial
complex I, neuronal cytoskeleton, and ubiquitin proteasome
system (UPS) was common among them (Table 2)[19,114,
135137].
Cellular or quantitative proteomicsof blood and cerebrospi-
nal fluid (CSF) is being used to identify, if any suitable bio-
marker exists for diagnosis of the disease [138]. Blood is
consideredto be the best sample because it is not onlyobtained
using a less invasive tool but also is the most dynamic tissue of
the body [139]. CSF is also an attractive and ideal body fluid to
search for PD biomarkers [140,141]. By proteomics-based
approaches, neuromelanin, mortalin, DJ-1, haptoglobin deriv-
atives, truncated globin, aggregated serum amyloid P compo-
nent, and ion channel proteins have been identified as
differentially regulated proteins in the blood or CSF of PD
patients [21,135,140,142145]. Many proteins that include,
α-synuclein and UCH L-1, are dysregulated in patients and are
expected to help in understanding and solving a few mysteries
of PD pathogenesis [146,147]. Proteomic approaches con-
firmed that α-synuclein undergoes posttranslational modifica-
tion, a triggering event for neurotoxicity [146].
RNAi
Silencing of autosomal dominant genes, which play critical
roles in PD pathogenesis could act as novel therapeutic
targets for treating PD. RNAi, used to silence the selected
gene expression, is projected as a substitute for the gene
knockout approach to study the complex molecular and
biochemical interactions within the pathways known to
involve in PD pathogenesis [148]. As mutation in the genes
belonging to PARK loci, oxidative stress, dysfunctional
xenobiotic metabolizing machinery, inflammation, autoph-
agy, and apoptosis [4,22,88] are associated with PD path-
ogenesis, therefore, direct or indirect modulators of such
genes could be astonishing targets of RNAi-based studies
[149,150]. Small interfering RNAs (siRNAs) and micro
RNAs (miRNAs) have made substantial input towards under-
standing the early events implicated in the functioning of
dopaminergic neurons. The siRNA knockdown studies eluci-
dated that the expression of homeoprotein LIM homeobox
transcription factor 1 α(Lmx1 α) is the prerequisite for
Mol Neurobiol
neuronal progenitors to determine a dopaminergic fate [22,
151]. Similarly, the mutual interaction between miRNA-133b
and Pitx 3 is reported to regulate the differentiation and
survival of the midbrain dopaminergic neurons [152]. The
tiny non-coding RNAs carry out localized control of gene
expression in the neurons and any anomaly in it may lead to
PD [153]. Tiny non-coding RNAs play a fundamental role in
neurodegenerative diseases in rodents, as evidenced by the
complete deficiency of a particular miRNA expression during
neuronal loss [154]. Downregulations of miRNA-34b and
miRNA-34c in PD brains hypothesize the involvement of
these tiny non-coding molecules in mitochondrial dysfunction
[155]. Furthermore, a few miRNAs are also identified to
regulate the synthesis of neurotransmitter substance P by the
tachykinin (TA C 1 )gene[156]. Small non-coding RNAs val-
idated the existing knowledge regarding various genes, such
Table 1 Details of some transcriptomics studies conducted employing rodent models and list of genes and pathways involved in PD pathogenesis
Serial
no.
Toxin
(rodent
model)
Tissue Affected
pathways
Differential gene expression level References
1. 6-OHDA
(rat)
Striatum Neurotrophic
factors and
neurotransmitter
release
Increases neurotensin, neuromedin U receptor, Finkel
BiskisJinkins murine osteosarcoma viral oncogene
cellular homolog, cyclooxygenase-2, follistatin, neu-
romedin U, platelet-derived growth factor-D, orphan
nuclear receptor-1 and TAC 2 expressions
[115]
Reduces TAC 1 expression
2. MPTP (rat) Striatum Cell growth,
differentiation,
regeneration
and survival
Upregulates cerebellin 1 precursor protein, galanin,
nerve growth factor receptor, and signal transducer and
activator of transcription 4 expressions
[118]
Downregulates ciliary neurotrophic factor expression
3. MPTP
(mouse)
Nigro-
striatum
Mitochondrial
dysfunction,
oxidative stress,
and apoptosis
Augments cathepsin D and UCH-14 expressions [114]
Attenuates ATP synthase protein 8, glutathione S-
transferase (GST) mu-5 (Gstm5), and NADHubiqui-
none oxidoreductase expressions
4. MPTP
(mouse)
Striatum Inflammatory responses,
cytokine and mammalian
target of rapamycin-signaling
pathways, activation of astro-
cytes and cellular stress
Increases heme oxygenase 1 metallothionein 2,
uncoupling protein 2, growth arrest and DNA-damage-
inducible-beta, nuclear factor of kappa light polypep-
tide gene enhancer inhibitor, FBJ murine osteosarcoma
viral oncogene homolog B, DNA-damage-inducible
transcript 4, CD9 antigen and heparin-binding epider-
mal growth factor-like growth factor expressions
[119]
Reduces retinoid X receptor gamma and paired box gene
8 expressions
5. Maneb and
paraquat
(mouse)
Striatum Electron transport,
lipid metabolism,
cell cycle, oxido-reductase
activity, ubiquitin
proteosomal system,
and apoptosis
Upregulates ubiquitin C and programmed cell death 10
expressions
[111]
Downregulates cytochrome c oxidase subunit VI a
polypeptide 1, diazepam binding inhibitor, growth
arrest specific 5, superoxide dismutase 1, and clusterin
expressions
6. MPTP
(mouse)
Substantia
nigra
Apoptosis, proteasomal system,
and energy metabolism
Increases lactate dehydrogenase 2 B chain, fibroblast
growth factor 1, cytochrome P450 family 4 subfamily
V polypeptide 3, RAS-like estrogen-regulated growth-
inhibitor, and protein tyrosine phosphatase receptor
type Z polypeptide 1 expressions
[120]
Reduces lipoprotein lipase, cadherin 2, G protein-
coupled receptor 83, neuropilin, cysteine-rich motor
neuron 1 and growth hormone receptor expressions
7. MPTP
(mouse)
Nigro-
striatum
Inflammation, oxidative stress,
glutamate toxicity, cell cycle,
and cell death processes
Increases interleukin (IL)-1b, IL-10, nuclear factor kappa
B (NF-kB) p65, N-methyl D-aspartate (NMDA)
adenosine A
2A
receptor (A
2A
-R), cyclin B2, tumor
necrosis factor (TNF) αand parkin expressions
[121]
8. MPTP
(mouse)
Nigro-
striatum
Cell cycle, oxidative stress,
inflammatory processes,
glutamate signaling, and
neuronal differentiation
Increases Bax membrane isoform α, IL-2 receptor
gamma, TNF-βand IL-1βexpressions
[122]
Reduces G2/M-specific cyclin B2, proliferation-
associated protein 1, cytochrome P450 1A1, GST- A,
inhibitor-kB αsubunit and NF-kB p65 expressions
Mol Neurobiol
as α-synuclein, TH, Parkin, PINK1 and LRRK2, which are
implicated in PD pathogenesis and assisted to gain newer
insights [149,157159]. Thus, siRNA-mediated depletion of
the major players involved in PD pathology and progression
may help in designing a new therapeutic strategy for PD.
Additionally, miRNA mimetic or expression vector could be
used to restore or overexpress miRNAs of interest [22,160,
161].
Omics and RNAi in PD: Noteworthy Validation
but Incomplete Applications
Genomic (SNP-based) studies have a number of limitations,
including sample size, life style factors, data interpretation,
and varying levels of environmental exposure. Indeed,
genomics offered an initiative to assess the association of
SNPs in a larger scale at a rapid rate employing a huge
sample size and predicted the possible association of many
genes in PD pathogenesis (Fig. 2)[162]. The major disad-
vantage is variation among studies even within the same
population.
Evidences about the dysregulation of genes, associated
with the neurotransmission, synaptic function, oxidative
stress, mitochondrial function and energy production, pro-
tein misfolding and aggregation, UPS dysfunction, autoph-
agy and apoptosis, etc., are known, however, the microarray
data yielded inconsistent results across multiple studies
[110113]. Despite validation with secondary quantitative
tools, transcriptomics of the nigrostriatal tissues could not
help in projecting the role of infrequently expressed genes in
PD pathogenesis [111113]. One of the major reasons could
be the loss of a substantial number of cell bodies of the
dopaminergic neurons in the substantia nigra and related
Table 2 Details of a few proteomics studies conducted employing various rodent models and list of proteins and pathways involved in PD
pathogenesis
Serial
no.
Toxin
(rodent model)
Used tissue Affected pathways Differential protein expression level References
1. 6-OHDA (rat) Substantia nigra Mitochondrial function Increases prohibitin and complex I 30-kDa subunit
expressions
[135]
2. 6-OHDA (rat) Striatum Energy metabolism,
calcium homeostasis,
antioxidation, and
cytoskeletal
Reduces calreticulin and calmodulin expressions [136]
Increases peroxiredoxin 2, mitochondrial complex
I and III expressions
3. MPTP (mouse) Mitochondrial
fraction of the
striatum
UPS Attenuates 19S proteasome
ATPase Rpt6 expression
[132]
Augments α-synuclein expression
4. MPTP (mouse) Striatum,
cortex,
cerebellum
Dopamine signaling,
mitochondrial function,
UPS, calcium signaling
Upregulates ubiquitin-specific protease expression [133]
Downregulates cytochrome c oxidase subunit Vic,
vacuolar ATP synthase subunit F, and TH
expressions
5. MPTP (mouse) Mitochondrial
fraction of the
substantia
nigra
Mitochondrial function
and oxidative stress
signaling
Increases DJ-1 expression [137]
Reduces complex I expression
6. MPTP and
methampheta-
mine (mouse)
Striatum Mitochondrial function,
oxidative stress,
signaling, and UPS
Upregulates cytochrome c1, calpain-2,
and ubiquitin ligases expressions
[114]
Downregulates glutathione peroxidase-4
and GST-M5, F- and V-type ATPase
and proteasome subunit protein expressions
7. Maneb and
paraquat (mouse)
Striatum Neurotransmitter release
and glycolysis
Decreases complexin I, α-enolase,
and glia maturation factor-βexpressions
[166]
8. Cypermethrin
(rat)
Striatum and
substantia
nigra
Mitochondrial function,
neurotransmitter release,
and cytoskeletal
assembly
Reduces stathmin, nicotinamide adenine
dinucleotide isocitrate dehydrogenase
α-subunit, prohibitin, ubiquitin conjugating
enzyme, nicotinamide adenine dinucleotide
dehydrogenase 24 k chain precursor, heat shock
protein (Hsp)-70, and synaptosomal associated
protein-25 expressions
[19]
Increases phosphoethanolamine binding protein-1,
mitogen-activated protein kinase activated
kinase-5, Hsp-60, and α-internexin intermediate
filament expressions
Mol Neurobiol
fibers in the striatum, which are used for data generation and
subsequent analysis. Many genes expressed at low levels
and in a particular cell type make the implication of global
study of the gene expression pattern questionable [163].
Therefore, the gene expression patterns of the striatum or
substantia nigra, the ideal tissues used for microarray assess-
ment of rodents with PD phenotype, need an error-free
approach. The disagreement could also be due to the use
of multiple arrays (mitochondrial genes specific, apoptotic
genes specific, cytoskeletal genes specific, etc.), varying
labeling procedures (direct or specific indirect methods),
variable hybridization patterns (time of hybridization, types,
and compositions of hybridization and washing buffers),
and difference in the number and type of replicates (with
or without dye swapping/use of single dye or two different
dyes for controls and experimental sets) [26]. Therefore,
microarray data generated and analyzed from the rodents
across multiple studies would offer invaluable insights, if all
such variables are kept unchanged. Furthermore, the RNA
sample of the striatum or substantia nigra represents RNAs
of multiple neuronal origins (such as dopaminergic, seroto-
ninergic, glutaminergic, cholinergic, etc.); therefore, the
representation of the RNAs of the dopaminergic neurons
are compromised, which makes it complicated to distinguish
[26]. Employing a microarray technique, several novel tran-
scripts involved in PD pathogenesis, and treatment out-
comes are identified [109]. But the transcripts getting
upregulated or downregulated do not necessarily mimic
the level of protein expressions, which are the actual regu-
lators of neurodegeneration, as many genes are transcribed
but not translated [27,114]. Similarly, microarray profiling
of blood offered important clues to assess the role of specific
genes in PD pathogenesis [164]; however, in blood
microarray patterns, several regulating factors could be
compromised. Overall, the identification of causative and
modulatory genes improved our understanding of the under-
lying etiology and mechanisms of pathogenesis, prevention
and cure are perhaps at the same stage as those were before
the application of these tools in PD research. A microarray
technique is not more than a semiquantitative tool even after
considering all the available corrective measures and vali-
dation of data with real-time PCR or other secondary tools is
mandatory [27,111]. Despite significant contribution of
microarray tool in assessing the putative mechanism of the
disease pathogenesis, the technology still fails to provide
accurate and complete mechanism of sporadic PD. Overall,
genomic approaches generated huge amounts of data, the
main drawbacks remain to be elusive etiology and patho-
genesis, as little success could be achieved in translating the
information.
Several groups of investigators have identified a few
differentially expressed proteins, which could not be tested
across multiple laboratories due to lack of expensive tools
and requirement of expert technical skills [124]. As
observed in the clinical investigation [165], reproducibility
across multiple studies could have been a major concern
even in the rodents due to lesser sample size and varying
conditions of animals storage and maintenance as well
underlying experimental procedures [9,21]. One of the most
important reasons of failure is the lack of standard exper-
imental procedures for the collection, storage and process-
ing of samples, and lack of common strategies across
various studies for the removal of the highly abundant
proteins [21,142,166]. Proteomics has not yet delivered
an expected breakthrough in PD diagnosis and identification
of potential and efficacious counteractive measures [21].
The major reasons have been the huge discrepancy in the
translation of rodent proteome data to human and most
importantly difference in the expression patterns of the
identified proteins from one rodent stock to another [102,
167]. The difficulty is more critical in rodents where the
variation could be observed from a rodent to another one as
well as between the inbred and outbred stocks of the same
rodent. Owing to the BBB, many proteins are not able to
enter the blood stream [168] and the proteins identified to
date are the outcomes of pathogenesis rather than the trig-
gering response and therefore cannot serve the purpose of a
genuine biomarker [21]. In summary, proteomic approaches
identified hundreds and hundreds of proteins that are differ-
entially expressed but have not yet satisfactorily met the
goal of developing biomarkers or molecular fingerprints
suitable for real application in clinics.
RNAi elucidates mainly the role of already suspected
genes/proteins in PD and mainly acts as a validation tool
[22]. RNAi could be very successful for monogenic domi-
nant genetic disorders, as only one gene is responsible for
the overall consequence of that disease. But for PD, which
has a multifactorial etiology and elusive pathogenesis, only
combinational approaches, such as silencing of multiple
targets, could possibly help in developing therapy. Similarly,
the drugs developed from the synthetic tiny non-coding
RNA molecules need to be tested for their toxicity, toler-
ability, and undesired off-target effects before they are used
for clinical interventions [22]. Tiny non-coding RNA-
mediated interference elicits immune responses and produ-
ces pro-inflammatory cytokines and interferons that may
affect the disease progression [169171], which tip off a
caution for its use in treating PD. This technique may get
desirable success beyond laboratory only when the unde-
sired immune responses could be defeated. RNAi tools are
still in their infancy and need to be continuously exploited to
gain certain novel information regarding elusive aspects of
PD pathophysiology.
The exact translation of omics and RNAi-generated data
employing rodent models is not possible because of many
primary reasons. The total number of genes and transcripts,
Mol Neurobiol
translated proteins, and posttranslational modifications
varies from rodent to rodent and from rodent to human
[27,102]. Although these sophisticated tools gave the role
of multiple genes/proteins/transcripts and validate the role
of the most important ones (Fig. 2), lack of consensus
among the results obtained from many studies and their
subsequent translation without fail in humans are limited
owing to lack of an ideal rodent model [102,167]. Many
proteins do not perform the similar functions across multiple
species/genus [102]. Apart from such limitations, there are
many additional hurdles for the application of sophisticated
tools in understanding the disease pathogenesis. Such stud-
ies need to be performed in the multiple rodent models and
postmortem human brains at extensive scale across laborato-
ries, globally. But the drawbacks of the recent molecular tools
are lack of cheap consumables, need of high technical exper-
tise, and requirement of highly specialized and costly equip-
ments [7]. Many more questions are still unresolved even with
the availability of these sophisticated techniques as the rela-
tionship between protein aggregation and the molecular events
leading to neurodegeneration has not yet been clarified [131].
Future Possibilities
Despite extensive efforts, rodent models and molecular tools
could not identify the realistic biomarkers and still face
barricading to pick up the fingerprints for an early diagnosis
and unbiased assessment of treatment outcomes. Although
lower animal models are beneficial to understand the role of
genetic factors in PD, development of only an appropriate
rodent or primate model could help in understanding the
pathogenesis by employing modern molecular tools and
RNAi. Success of a rodent or nonhuman primate model is
purely dependent on the availability of epidemiological
information across multiple populations, followed by appro-
priate statistical analyses. If everything goes all right, an
ideal rodent or nonhuman primate model can be developed.
Once all genuine causative factors would be identified, an
authentic dual model may be developed to understand the
precise pathology of sporadic PD. A combinational
approach of employing an ideal model with multifaceted
omics and RNAi could be expected to offer the pragmatic
mechanistic understanding of PD pathogenesis and success-
fully be translated to sporadic PD. Theoretically, the dual or
interactive model approach would offer the best prospect to
understand sporadic PD pathogenesis; however, practical
conquest lies in the development of a suitable combination
approach.
Acknowledgments We sincerely thank the Council of Scientific and
Industrial Research (CSIR), New Delhi, India/University Grants Com-
mission, New Delhi, India for providing research fellowships to
Sharawan Yadav, Sonal Agrawal, Garima Srivastava, Anand Kumar
Singh, and Anubhuti Dixit. The CSIR-IITR communication number of
the article is 3012.
Conflicts of Interest None declared.
References
1. Parkinson J (2002) An essay on the shaking palsy 1817. J Neuro-
psychiatry Clin Neurosci 14:223236
2. Goetz CG (1986) Charcot on Parkinsons disease. Mov Disord
1:2732
3. Singh MP, Patel S, Dikshit M, Gupta YK (2006) Contribution of
genomics and proteomics in understanding the role of modifying
factors in Parkinsons disease. Indian J Biochem Biophys 43:6981
4. Miller RL, James-Kracke M, Sun GY, Sun AY (2009) Oxidative
and inflammatory pathways in Parkinsons disease. Neurochem
Res 34:5565
5. Wirdefeldt K, Adami HO, Cole P, Trichopoulos D, Mandel J
(2011) Epidemiology and etiology of Parkinsonsdisease:a
review of the evidence. Eur J Epidemiol 26:S1S58
6. Klockgether T (2004) Parkinsons disease: clinical aspects. Cell
Tissue Res 318:115120
7. Morgan JC, Mehta SH, Sethi KD (2010) Biomarkers in Parkin-
sons disease. Curr Neurol Neurosci Rep 10:423430
8. Ebin J (1951) Surgical treatment of Parkinsonism: indications and
results. Bull N Y Acad Med 27:653678
9. Cicchetti F, Drouin-Ouellet J, Gross RE (2009) Environmental
toxins and Parkinsons disease: what have we learned from
pesticide-induced animal models? Trends Pharmacol Sci
30:475483
10. Singhal NK, Srivastava G, Agrawal S, Jain SK, Singh MP (2012)
Melatonin as a neuroprotective agent in the rodent models of
Parkinsons disease: is it all set to irrefutable clinical translation?
Mol Neurobiol 45:186199
11. Kumar A, Ahmad I, Shukla S, Singh BK, Patel DK, Pandey HP,
Singh C (2010) Effect of zinc and paraquat co-exposure on
neurodegeneration: modulation of oxidative stress and expression
of metallothioneins, toxicant responsive and transporter genes in
rats. Free Radic Res 44:950965
12. Khalid M, Aoun RA, Mathews TA (2011) Altered striatal dopa-
mine release following a sub-acute exposure to manganese. J
Neurosci Methods 202:182191
13. Singh AK, Tiwari MN, Upadhyay G, Patel DK, Singh D, Prakash
O, Singh MP (2012) Long-term exposure to cypermethrin indu-
ces nigrostriatal dopaminergic neurodegeneration in adult rats:
postnatal exposure enhances the susceptibility during adulthood.
Neurobiol Aging 33:404415
14. Betarbet R, Sherer TB, Greenamyre JT (2002) Animal models of
Parkinsons disease. Bioessays 24:308318
15. Thiruchelvam M, Brockel BJ, Richfield EK, Baggs RB, Cory-
Slechta DA (2000) Potentiated and preferential effects of com-
bined paraquat and maneb on nigrostriatal dopamine systems:
environmental risk factors for Parkinsons disease? Brain Res
873:225234
16. Langston JW, Ballard P, Tetrud JW, Irwin I (1983) Chronic
parkinsonism in humans due to a product of meperidine-analog
synthesis. Science 219:979980
17. Patel S, Singh V, Kumar A, Gupta YK, Singh MP (2006) Status of
antioxidant defense system and expression of toxicant responsive
genesin striatum of maneband paraquat-induced Parkinsonsdisease
phenotype in mouse: mechanism of neurodegeneration. Brain Res
108:918
Mol Neurobiol
18. Gorell JM, Johnson CC, Rybicki BA, Peterson EL, Kortsha GX,
Brown GG, Richardson RJ (1999) Occupational exposure to
manganese, copper, lead, iron, mercury and zinc and the risk of
Parkinsons disease. Neurotoxicology 20:239247
19. Singh AK, Tiwari MN, Dixit A, Upadhyay G, Patel DK, Singh D,
Prakash O, Singh MP (2011) Nigrostriatal proteomics of
cypermethrin-induced dopaminergic neurodegeneration: micro-
glial activation dependent and independent regulations. Toxicol
Sci 122:526538
20. Srivastava G, Dixit A, Yadav S, Patel DK, Prakash O, Singh MP
(2012) Resveratrol potentiates cytochrome P450 2d22-mediated
neuroprotection in maneb- and paraquat-induced parkinsonism in
the mouse. Free Radic Biol Med 52:12941306
21. Srivastava G, Singh K, Tiwari MN, Singh MP (2010) Proteomics
in Parkinsons disease: current trends, translational snags and
future possibilities. Expert Rev Proteomics 7:127139
22. Srivastava G, Dixit A, Prakash O, Singh MP (2011) Tiny non-
coding RNAs in Parkinsons disease: Implications, expectations
and hypes. Neurochem Int 59:759769
23. Klein C, Schlossmacher MG (2007) Parkinson disease, 10 years
after its genetic revolution: multiple clues to a complex disorder.
Neurology 69:20932104
24. Gao HM, Hong JS (2011) Geneenvironment interactions: key to
unravelingthemysteryofParkinsonsdisease.ProgNeurobiol94:119
25. Uversky VN (2004) Neurotoxicant-induced animal models of
Parkinsons disease: understanding the role of rotenone, maneb
and paraquat in neurodegeneration. Cell Tissue Res 318:225241
26. Ginsberg SD, Mirnics K (2006) Functional genomic methodolo-
gies. Prog Brain Res 158:1540
27. Shilling PD, Kelsoe JR (2002) Functional genomics approaches
to understanding brain disorders. Pharmacogenomics 3:3145
28. Vargas RH, Ornelas LF, González IL, Escovar JR, Zurita M,
Reynaud E (2011) Synphilin suppresses α-synuclein neurotoxicity
in a ParkinsonsdiseaseDrosophila model. Genesis 49:392402
29. Ramsden DB, Parsons RB, Ho SL, Waring RH (2001) The aetiol-
ogy of idiopathic Parkinsons disease. Mol Pathol 54:369380
30. Migliore L, Coppede F (2009) Environmental-induced oxidative
stress in neurodegenerative disorders and aging. Mutat Res
674:7384
31. Wood-Kaczmar A, Gandhi S, Wood NW (2006) Understanding
the molecular causes of Parkinsons disease. Trends Mol Med
12:521528
32. Bove J, Prou D, Perier C, Przedborski S (2005) Toxin-induced
models of Parkinsons disease. NeuroRx 2:484494
33. Ascherio A, Chen H, Weisskopf MG, OReilly E, McCullough ML,
Calle EE, Schwarzschild MA, Thun MJ (2006) Pesticide exposure
and risk for Parkinsonsdisease. Ann Neurol 60:197203
34. He Y, Appel S, Le W (2001) Minocycline inhibits microglial
activation and protects nigral cells after 6-hydroxydopamine
injection into mouse striatum. Brain Res 909:187193
35. Silva RM, Ries V, Oo TF, Yarygina O,Jackson-Lewis V, Ryu EJ, Lu
PD, Marciniak SJ, Ron D, Przedborski S, Kholodilov N, Greene
LA, Burke RE (2005) CHOP/GADD153 is a mediator of apoptotic
death in substantia nigra dopamine neurons in an in vivo neurotoxin
model of parkinsonism. J Neurochem 95:974986
36. Reyes S, Mitrofanis J (2008) Patterns of FOS expression in the
spinal cord and periaqueductal grey matter of 6OHDA-lesioned
rats. Int J Neurosci 118:10531079
37. Latchoumycandane C, Anantharam V, Jin H, Kanthasamy A,
Kanthasamy A (2011) Dopaminergic neurotoxicant (2011) 6-
OHDA induces oxidative damage through proteolytic activation
of PKCδin cell culture and animal models of Parkinsons disease.
Toxicol Appl Pharmacol 256:314323
38. Bernstein AI, Garrison SP, Zambetti GP, OMalley KL (2011) 6-
OHDA generated ROS induces DNA damage and p53- and
PUMA-dependent cell death. Mol Neurodegener 6:2
39. Proft J, Faraji J, Robbins JC, Zucchi FC, Zhao X, Metz GA,
Braun JE (2011) Identification of bilateral changes in TID1
expression in the 6-OHDA rat model of Parkinsons disease.
PLoS One 6:e26045
40. Przedborski S, Vila M (2003) The 1-methyl-4-phenyl-1,2,3,6-
tetrahydropyridine mouse model: a tool to explore the patho-
genesis of Parkinsons disease. Ann N Y Acad Sci 991:189198
41. Ross CA, Smith WW (2007) Gene-environment interactions in
Parkinsons disease. Parkinsonism Relat Disord 13:S309S315
42. Cui M, Aras R, Christian WV, Rappold PM, Hatwar M, Panza J,
Jackson-Lewis V, Javitch JA, Ballatori N, Przedborski S, Tieu K
(2009) The organic cation transporter-3 is a pivotal modulator of
neurodegeneration in the nigrostriatal dopaminergic pathway.
Proc Natl Acad Sci U S A 106:80438048
43. Karunakaran S, Saeed U, Mishra M, Valli RK, Joshi SD, Meka
DP, Seth P, Ravindranath V (2008) Selective activation of p38
mitogen-activated protein kinase in dopaminergic neurons of
substantia nigra leads to nuclear translocation of p53 in 1-
methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated mice. J Neu-
rosci 28:1250012509
44. Castro-Caldas M, Carvalho AN, Rodrigues E, Henderson C, Wolf
CR, Gama MJ (2012) Glutathione S-transferase pi mediates MPTP-
induced c-Jun N-terminal kinase activation in the nigrostriatal path-
way. Mol Neurobiol. doi:10.1007/s12035-012-8266-9
45. Zawada WM, Banninger GP, Thornton J, Marriott B, Cantu D,
Rachubinski AL, Das M, Griffin WS, Jones SM (2011) Generation
of reactive oxygen species in 1-methyl-4-phenylpyridinium (MPP
+) treated dopaminergic neurons occurs as an NADPH oxidase-
dependent two-wave cascade. J Neuroinflammation 8:129
46. Durgadoss L, Nidadavolu P, Valli RK, Saeed U, Mishra M, Seth
P, Ravindranath V (2012) Redox modification of Akt mediated
by the dopaminergic neurotoxin MPTP, in mouse midbrain, leads
to down-regulation of pAkt. FASEB J 26:14731483
47. Jeong HJ, Kim DW, Woo SJ, Kim HR, Kim SM, Jo HS, Park M,
Kim DS, Kwon OS, Hwang IK, Han KH, Park J, Eum WS, Choi
SY (2012) Transduced Tat-DJ-1 protein protects against oxida-
tive stress-induced SH-SY5Y cell death and Parkinson disease in
a mouse model. Mol Cells. doi:10.1007/s10059-012-2255-8
48. Martin HL, Mounsey RB, Mustafa S, Sathe K, Teismann P
(2012) Pharmacological manipulation of peroxisome
proliferator-activated receptor γ(PPARγ) reveals a role for
anti-oxidant protection in a model of Parkinsons disease. Exp
Neurol 235:528538
49. Park HK, Cho AR, Lee SC, Ban JY (2012) MPTP-induced model
of Parkinsons disease in heat shock protein 70.1 knockout mice.
Mol Med Report 5:14651468
50. Domenger D, Dea D, Theroux L, Moquin L, Gratton A, Poirier J
(2012) The MPTP neurotoxic lesion model of Parkinsons disease
activates the apolipoprotein E cascade in the mouse brain. Exp
Neurol 233:513522
51. Airavaara M, Harvey BK, Voutilainen MH, Shen H, Chou J,
Lindholm P, Lindahl M, Tuominen RK, Saarma M, Wang Y,
Hoffer B (2011) CDNF protects the nigrostriatal dopamine sys-
tem and promotes recovery after MPTP treatment in mice. Cell
Transplant. doi:10.3727/096368911X600948
52. Bezard E, Przedborski S (2011) A tale on animal models of
Parkinsons disease. Mov Disord 26:9931002
53. LoPachin RM, Gavin T (2008) Response to Paraquat: the red
herring of Parkinsons disease research. Toxicol Sci 103:219221
54. Shimizu K, Ohtaki K, Matsubara K, Aoyama K, Uezono T, Saito
O, Suno M, Ogawa K, Hayase N, Kimura K, Shiono H (2001)
Carrier-mediated processes in bloodbrain barrier penetration and
neural uptake of paraquat. Brain Res 906:135142
55. Feng LR, Maguire-Zeiss KA (2011) Dopamine and paraquat
enhance α-synuclein-induced alterations in membrane conduc-
tance. Neurotox Res 20:387401
Mol Neurobiol
56. Miller RL, Sun GY, Sun AY (2007) Cytotoxicity of paraquat in
microglial cells: involvement of PKCdelta- and ERK1/2-dependent
NADPH oxidase. Brain Res 1167:129139
57. Huang CL, Lee YC, Yang YC, Kuo TY, Huang NK (2012)
Minocycline prevents paraquat-induced cell death through
attenuating endoplasmic reticulum stress and mitochondrial dys-
function. Toxicol Lett 209:203210
58. Rappold PM, Cui M, Chesser AS, Tibbett J, Grima JC, Duan L,
SenN,JavitchJA,TieuK(2011) Paraquat neurotoxicity is
mediated by the dopamine transporter and organic cation
transporter-3. Proc Natl Acad Sci U S A 108:2076620771
59. Fei Q, Ethell DW (2008) Maneb potentiates paraquat neurotox-
icity by inducing key Bcl-2 family members. J Neurochem
105:20912097
60. Litteljohn D, Nelson E, Bethune C, Hayley S (2011) The effects
of paraquat on regional brain neurotransmitter activity, hippo-
campal BDNF and behavioural function in female mice. Neurosci
Lett 502:186191
61. Kachroo A, Irizarry MC, Schwarzschild MA (2010) Caffeine
protects against combined paraquat and maneb-induced dopami-
nergic neuron degeneration. Exp Neurol 223:657661
62. Wang A, Costello S, Cockburn M, Zhang X, Bronstein J, Ritz B
(2011) Parkinsons disease risk from ambient exposure to pesti-
cides. Eur J Epidemiol 26:547555
63. Kang H, Han BS, Kim SJ, Oh YJ (2012) Mechanisms to prevent
caspase activation in rotenone-induced dopaminergic neurode-
generation: role of ATP depletion and procaspase-9 degradation.
Apoptosis 17:449462
64. Shin EJ, Kim EM, Lee JA, Rhim H, Hwang O (2012) Matrix
metalloproteinase-3 is activated by HtrA2/Omi in dopaminergic
cells: relevance to Parkinsons disease. Neurochem Int 60:249256
65. Meredith GE, Sonsalla PK, Chesselet MF (2008) Animal models of
Parkinsons disease progression. Acta Neuropathol 115:385398
66. Ritz BR, Manthripragada AD, Costello S, Lincoln SJ, Farrer MJ,
Cockburn M, Bronstein J (2009) Dopamine transporter genetic
variants and pesticides in Parkinsons disease. Environ Health Per-
spect 117:964969
67. Moretto A, Colosio C (2011) Biochemical and toxicological
evidence of neurological effects of pesticides: the example of
Parkinsons disease. Neurotoxicology 32:383391
68. Singh AK, Tiwari MN, Prakash O, Singh MP (2012) A current
review of cypermethrin-induced neurotoxicity and nigrostriatal
dopaminergic neurodegeneration. Curr Neuropharmacol 10:6471
69. Sharma H, Zhang P, Barber DS, Liu B (2010) Organochlorine
pesticides dieldrin and lindane induce cooperative toxicity in
dopaminergic neurons: role of oxidative stress. Neurotoxicology
31:215222
70. Cass W (1996) GDNF selectively protects dopamine neurons
over serotonin neurons against the neurotoxic effects of metham-
phetamine. J Neurosci 16:81328139
71. Howard CD, Keefe KA, Garris PA, Daberkow DP (2011) Meth-
amphetamine neurotoxicity decreases phasic, but not tonic, dopa-
minergic signaling in the rat striatum. J Neurochem 118:668676
72. Morrow BA, Roth RH, Redmond DE, Elsworth JD (2011) Impact
of methamphetamine on dopamine neurons in primates is
dependent on age: implications for development of Parkinsons
disease. Neuroscience 189:277285
73. Callaghan RC, Cunningham JK, Sykes J, Kish SJ (2012)
IncreasedriskofParkinsons disease in individuals hospital-
ized with conditions related to the use of methamphetamine
or other amphetamine-type drugs. Drug Alcohol Depend
120:3540
74. Kousik SM, Graves SM, Napier TC, Zhao C, Carvey PM
(2011) Methamphetamine-induced vascular changes lead to
striatal hypoxia and dopamine reduction. Neuroreport
22:923928
75. Brar S, Henderson D, Schenck J, Zimmerman EA (2009) Iron
accumulation in the substantia nigra of patients with Alzheimer
disease and parkinsonism. Arch Neurol 66:371374
76. Jomova K, Valko M (2011) Advances in metal-induced oxidative
stress and human disease. Toxicology 283:6587
77. Kienzl E, Puchinger L, Jellinger K, Linert W, Stachelberger H,
Jameson RF (1995) The role of transition metals in the patho-
genesis of Parkinsons disease. J Neurol Sci 134:6978
78. Kumar A, Singh BK, Ahmad I, Shukla S, Patel DK, Srivastava G,
Kumar V, Pandey HP, Singh C (2012) Involvement of NADPH
oxidase and glutathione in zinc-induced dopaminergic neurode-
generation in rats: similarity with paraquat neurotoxicity. Brain
Res 1438:4864
79. Gash DM, Rutland K, Hudson NL, Sullivan PG, Bing G, Cass
WA, Pandya JD, Liu M, Choi DY, Hunter RL, Gerhardt GA,
Smith CD, Slevin JT, Prince TS (2008) Trichloroethylene: par-
kinsonism and complex 1 mitochondrial neurotoxicity. Ann Neu-
rol 63:184192
80. Holtcamp W (2012) The emerging science of BMAA: do cyano-
bacteria contribute to neurodegenerative disease? Environ Health
Perspect 120:A110A116
81. Lannuzel A, Michel PP, Höglinger GU, Champy P, Jousset A,
Medja F, Lombès A, Darios F, Gleye C, Laurens A, Hocquemiller
R, Hirsch EC, Ruberg M (2003) The mitochondrial complex I
inhibitor annonacin is toxic to mesencephalic dopaminergic neu-
rons by impairment of energy metabolism. Neuroscience
121:287296
82. Storch A, Ott S, Hwang YI, Ortmann R, Hein A, Frenzel S,
Matsubara K, Ohta S, Wolf HU, Schwarz J (2002) Selective
dopaminergic neurotoxicity of isoquinoline derivatives related
to Parkinsons disease: studies using heterologous expression
systems of the dopamine transporter. Biochem Pharmacol
63:909920
83. Ohta S, Tachikawa O, Makino Y, Tasaki Y, Hirobe M (1990)
Metabolism and brain accumulation of tetrahydroisoquinoline
(TIQ) a possible parkinsonism inducing substance, in an animal
model of a poor debrisoquine metabolizer. Life Sci 46:599605
84. Mohajjel Nayebi AA, Sheidaei H (2010) Buspirone improves
haloperidol-induced Parkinson disease in mice through 5-HT
(1A) recaptors. Daru 18:4145
85. McNaught KS, Perl DP, Brownell AL, Olanow CW (2004) Sys-
temic exposure to proteasome inhibitors causes a progressive
model of Parkinsons disease. Ann Neurol 56:149162
86. Panneton WM, Kumar VB, Gan Q, Burke WJ, Galvin JE (2010)
The neurotoxicity of DOPAL: behavioral and stereological evi-
dence for its role in Parkinson disease pathogenesis. PLoS One 5:
e15251
87. Gasser T (2007) Update on the genetics of Parkinsons disease.
Mov Disord 17:S343S350
88. Bekris LM, Mata IF, Zabetian CP (2010) The genetics of Parkin-
son disease. J Geriatr Psychiatry Neurol 23:228242
89. Fujioka S, Wszolek ZK (2012) Update on genetics of parkinson-
ism. Neurodegener Dis 10:257260
90. Terzioglu M, Galter D (2008) Parkinsons disease: genetic versus
toxin-induced rodent models. FEBS J 275:13841391
91. Hisahara S, Shimohama S (2010) Toxin-induced and genetic
animal models of Parkinsons disease. Parkinsons Dis
2011:951709
92. Manning-Bog AB, Langston JW (2007) Model fusion, the next
phase in developing animal models for Parkinsonsdisease. Neu-
rotox Res 11:219240
93. Boger HA, Granholm AC, McGinty JF, Middaugh LD (2010) A
dual-hit animal model for age-related Parkinsonism. Prog Neuro-
biol 90:217229
94. Gao HM, Zhang F, Zhou H, Kam W, Wilson B, Hong JS (2011)
Neuroinflammation and α-synuclein dysfunction potentiate each
Mol Neurobiol
other, drivingchronic progression of neurodegeneration in a
mouse model of Parkinsons disease. Environ Health Perspect
119:807814
95. Jakowec MW, Petzinger GM (2004) 1-methyl-4-phenyl-1,2,3,6-
tetrahydropyridine-lesioned model of Parkinsons disease, with
emphasis on mice and nonhuman primates. Comp Med 54:497513
96. Zhu XR, Maskri L, Herold C, Bader V, Stichel CC, Güntürkün O,
Lübbert H (2007) Non-motor behavioural impairments in parkin-
deficient mice. Eur J Neurosci 26:19021911
97. Emborg ME (2007) Nonhuman primate models of Parkinsons
disease. ILAR J 48:339355
98. Duty S, Jenner P (2011) Animal models of Parkinsons disease: a
source of novel treatments and clues to the cause of the disease.
Br J Pharmacol 164:13571391
99. Pienaar IS, Götz J, Feany MB (2010) Parkinsonsdisease:
insights from non-traditional model organisms. Prog Neurobiol
92:558571
100. Antony PM, Diederich NJ, Balling R (2011) Parkinsons disease
mouse models in translational research. Mamm Genome 22:401
419
101. Chesselet MF, Richter F (2011) Modelling of Parkinsons disease
in mice. Lancet Neurol 10:11081118
102. Potashkin JA, Blume SR, Runkle NK (2010) Limitations of
animal models of Parkinsons disease. Parkinsons Dis
2011:658083
103. Mellick GD, Silburn PA, Sutherland GT, Siebert GA (2010)
Exploiting the potential of molecular profiling in Parkinsons
disease: current practice and future probabilities. Expert Rev
Mol Diagn 10:10351050
104. Miller RM, Federoff HJ (2006) Microarrays in Parkinsons dis-
ease: a systematic approach. NeuroRx 3:319326
105. Chang XL, Mao XY, Li HH, Zhang JH, Li NN, Burgunder JM,
Peng R, Tan EK (2011) Association of GWAS loci with PD in
China. Am J Med Genet B Neuropsychiatr Genet 156B:334339
106. Biernacka JM, Armasu SM, Cunningham JM, Eric Ahlskog J,
Chung SJ, Maraganore DM (2011) Do interactions between
SNCA, MAPT, and LRRK2 genes contribute to Parkinsons
disease susceptibility? Parkinsonism Relat Disord 17:730736
107. Chung SJ, Armasu SM, Biernacka JM, Lesnick TG, Rider DN,
Lincoln SJ, Ortolaza AI, Farrer MJ, Cunningham JM, Rocca WA,
Maraganore DM (2011) Common variants in PARK loci and
related genes and Parkinsons disease. Mov Disord 26:280288
108. Autere J, Moilanen JS, Finnila S, Soininen H, Mannermaa A,
Hartikainen P, Hallikainen M, Majamaa K (2004) Mitochondrial
DNA polymorphisms as risk factors for Parkinsons disease and
Parkinsons disease dementia. Hum Genet 115:2935
109. Simunovic F, Yi M, Wang Y, Macey L, Brown LT, Krichevsky
AM, Andersen SL, Stephens RM, Benes FM, Sonntag KC (2009)
Gene expression profiling of substantia nigra dopamine neurons:
further insights into Parkinsons disease pathology. Brain
132:17951809
110. Grunblatt E, Mandel S, Jacob-Hirsch J, Zeligson S, Amariglo N,
Rechavi G, Li J, Ravid R, Roggendorf W, Riederer P, Youdim
MB (2004) Gene expression profiling of parkinsonian substantia
nigra pars compacta alterations in ubiquitin-proteasome, heat
shock protein, iron and oxidative stress regulated proteins, cell
adhesion/cellular matrix and vesicle trafficking genes. J Neural
Transm 111:15431573
111. Patel S, Singh K, SinghS, Singh MP (2008) Gene expression profiles
of mouse striatum in control and maneb+ paraquat-induced Parkin-
sons disease phenotype: validation of differentially expressedenergy
metabolizing transcripts. Mol Biotechnol 40:5968
112. Duke DC, Moran LB, Kalaitzakis ME, Deprez M, Dexter DT,
Pearce RK, Graeber MB (2006) Transcriptome analysis reveals
link between proteasomal and mitochondrial pathways in Parkin-
sons disease. Neurogenetics 7:139148
113. Miller RM, Kiser GL, Kaysser-Kranich TM (2006) Robust dys-
regulation of gene expression in substantia nigra and striatum in
Parkinsons disease. Neurobiol Dis 21:305313
114. Chin MH, Qian WJ, Wang H, Petyuk VA, Bloom JS, Sforza DM,
Laćan G, Liu D, Khan AH, Cantor RM, Bigelow DJ, Melega WP,
Camp DG 2nd, Smith RD, Smith DJ (2008) Mitochondrial dys-
function, oxidative stress, and apoptosis revealed by proteomic
and transcriptomic analyses of the striata in two mouse models of
Parkinsons disease. J Proteome Res 7:666677
115. Cadet JL, Brannock C, Krasnova IN, Ladenheim B, McCoy MT,
Chou J, Lehrmann E, Wood WH, Becker KG, Wang Y (2010)
Methamphetamine-induced dopamine-independent alterations in
striatal gene expression in the 6-hydroxydopamine hemiparkin-
sonian rats. PLoS One 5:e15643
116. Pickrell AM, Fukui H, Wang X, Pinto M, Moraes CT (2011) The
striatum is highly susceptible to mitochondrial oxidative phos-
phorylation dysfunctions. J Neurosci 631:98959904
117. Grünblatt E (2012) Parkinsons disease: molecular risk factors.
Parkinsonism Relat Disord 18(Suppl 1):S45S48
118. Wernicke C, Hellmann J, Zieba B, Kuter K, Ossowska K, Frenzel
M, Dencher NA, Rommelspacher H (2010) 9-Methyl-beta-carbo-
line has restorative effects in an animal model of Parkinsons
disease. Pharmacol Rep 62:3553
119. Pattarini R, Rong Y, Qu C, Morgan JI (2008) Distinct mecha-
nisms of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyrimidine resist-
ance revealed by transcriptome mapping in mouse striatum.
Neuroscience 155:11741194
120. Chung CY, Seo H, Sonntag KC, Brooks A, Lin L, Isacson O
(2005) Cell type-specific gene expression of midbrain dopami-
nergic neurons reveals molecules involved in their vulnerability
and protection. Hum Mol Genet 14:17091725
121. Mandel S, Grunblatt E, Maor G, Youdim MB (2002) Early and
late gene changes in MPTP mice model of Parkinsons disease
employing cDNA microarray. Neurochem Res 27:12311243
122. Grunblatt E, Mandel S, Maor G, Youdim MB (2001) Gene
expression analysis in N-methyl-4-phenyl-1, 2, 3, 6-
tetrahydropyridine mice model of Parkinsons disease using
cDNA microarray: effect of R-apomorphine. J Neurochem
78:112
123. Tomita H, Vawter MP, Walsh DM, Evans SJ, Choudary PV, Li J,
Overman KM, Atz ME, Myers RM, Jones EG, Watson SJ, Akil
H, Bunney WE Jr (2004) Effect of agonal and postmortem factors
on gene expression profile: quality control in microarray analyses
of postmortem human brain. Biol Psychiatry 55:346352
124. Mischak H, Apweiler R, Banks RE, Conaway M, Coon J,
Dominiczak A, Ehrich JH, Fliser D, Girolami M, Hermjakob H,
Hochstrasser D, Jankowski J, Julian BA, Kolch W, Massy ZA,
Neusuess C, Novak J, Peter K, Rossing K, Schanstra J, Semmes
OJ, Theodorescu D, Thongboonkerd V, Weissinger EM, Van Eyk
JE, Yamamoto T (2007) Clinical proteomics: a need to define the
field and to begin to set adequate standards. Proteomics Clin
Appl 1:148156
125. Zhang J, Keene CD, Pan C, Montine KS, Montine TJ (2008)
Proteomics of human neurodegenerative diseases. J Neuropathol
Exp Neurol 67:923932
126. Pienaar IS, Daniels WM, Gotz J (2008) Neuroproteomics as a
promising tool in Parkinsons disease research. J Neural Transm
115:14131430
127. Butler GS, Overall CM (2009) Proteomic identification of multi-
tasking proteins in unexpected locations complicates drug target-
ing. Nat Rev Drug Discov 8:935948
128. Farley AR, Link AJ (2009) Identification and quantification of
protein posttranslational modifications. Methods Enzymol
463:725763
129. Hwang H, Zhang J, Chung KA, Leverenz JB, Zabetian CP,
Peskind ER, Jankovic J, Su Z, Hancock AM, Pan C, Montine
Mol Neurobiol
TJ, Pan S, Nutt J, Albin R, Gearing M, Beyer RP, Shi M, Zhang J
(2010) Glycoproteomics in neurodegenerative diseases. Mass
Spectrom Rev 29:79125
130. Zhang J, Goodlett DR (2004) Proteomic approach to studying
Parkinsons disease. Mol Neurobiol 29:271288
131. Robinson PA (2010) Understanding the molecular basis of Par-
kinsons disease, identification of biomarkers and routes to ther-
apy. Expert Rev Proteomics 7:565578
132. Liu B, Shi Q, Ma S, Feng N, Li J, Wang L, Wang X (2008)
Striatal 19S Rpt6 deficit is related to alpha-synuclein accumula-
tion in MPTP-treated mice. Biochem Biophys Res Commun
376:277282
133. Zhang X, Zhou JY, Chin MH, Schepmoes AA, Petyuk VA, Weitz
KK, Petritis BO, Monroe ME, Camp DG, Wood SA, Melega WP,
Bigelow DJ, Smith DJ, Qian WJ, Smith RD (2010) Region-
specific protein abundance changes in the brain of MPTP-
induced Parkinsons disease mouse model. J Proteome Res
9:14961509
134. Jin J, Hulette C, Wang Y (2006) Proteomic identification of a
stress protein, mortalin/mthsp70/GRP75: relevance to Parkinson
disease. Mol Cell Proteomics 5:11931204
135. Park B, Yang J, Yun N, Choe KM, Jin BK, Oh YJ (2010)
Proteomic analysis of expression and protein interactions in a 6-
hydroxydopamine-induced rat brain lesion model. Neurochem Int
57:1632
136. Lessner G, Schmitt O, Haas SJ, Mikkat S, Kreutzer M, Wree A,
Glocker MO (2010) Differential proteome of the striatum from
hemiparkinsonian rats displays vivid structural remodeling pro-
cesses. J Proteome Res 9:46714687
137. Jin J, Meredith GE, Chen L, Zhou Y, Xu J, Shie FS, Lockhart P,
Zhang J (2005) Quantitative proteomic analysis of mitochondrial
proteins: relevance to Lewy body formation and Parkinsons
disease. Brain Res Mol Brain Res 134:119138
138. Shi M, Caudle WM, Zhang J (2009) Biomarker discovery in
neurodegenerative diseases: a proteomic approach. Neurobiol
Dis 35:157164
139. Goldknopf IL (2008) Blood-based proteomics for personalized
medicine: examples from neurodegenerative disease. Expert Rev
Proteomics 5:18
140. Sinha A, Srivastava N, Singh S, Singh AK, Bhushan S, Shukla R,
Singh MP (2009) Identification of differentially displayed pro-
teins in cerebrospinal fluid of Parkinsons disease patients: a
proteomic approach. Clin Chim Acta 400:1420
141. van Dijk KD, Teunissen CE, Drukarch B, Jimenez CR,
Groenewegen HJ, Berendse HW, van de Berg WD (2010) Diag-
nostic cerebrospinal fluid biomarkers for Parkinsons disease: a
pathogenetically based approach. Neurobiol Dis 39:229241
142. Tribl F, Marcus K, Bringmann G, Meyer HE, Gerlach M, Riederer P
(2006) Proteomics of the human brain: sub-proteomes might hold
the key to handle brain complexity. J Neural Transm 113:1041
1054
143. Sinha A, Patel S, Singh MP, Shukla R (2007) Blood proteome
profiling in case controls and Parkinsons disease patients in
Indian population. Clin Chim Acta 380:232234
144. Chen HM, Lin CY, Wang V (2011) Amyloid P component as a
plasma marker for Parkinsons disease identified by a proteomic
approach. Clin Biochem 44:377385
145. Waragai M, Wei J, Fujita M, Nakai M, Ho GJ, Masliah E, Akatsu
H, Yamada T, Hashimoto M (2006) Increased level of DJ-1 in the
cerebrospinal fluids of sporadic Parkinsons disease. Biochem
Biophys Res Commun 345:967972
146. Muntané G, Dalfó E, Martinez A, Ferrer I (2008) Phosphoryla-
tion of tau and alpha-synuclein in synaptic-enriched fractions of
the frontal cortex in Alzheimers disease, and in Parkinsons
disease and related alpha-synucleinopathies. Neuroscience
152:913923
147. Choi J, Levey AI, Weintraub ST, Rees HD, Gearing M, Chin LS,
Li L (2004) Oxidative modifications and down-regulation of
ubiquitin carboxyl-terminal hydrolase L1 associated with idio-
pathic ParkinsonsandAlzheimers diseases. J Biol Chem
279:1325613264
148. Salahpour A, Medvedev IO, Beaulieu JM, Gainetdinov RR,
Caron MG (2007) Local knockdown of genes in the brain using
small interfering RNA: a phenotypic comparison with knockout
animals. Biol Psychiatry 61:6569
149. Ko HS, Bailey R, Smith WW, Liu Z, Shin JH, Lee YI, Zhang YJ,
Jiang H, Ross CA, Moore DJ, Patterson C, Petrucelli L, Dawson
TM, Dawson VL (2009) CHIP regulates leucine-rich repeat
kinase-2 ubiquitination, degradation, and toxicity. Proc Natl Acad
Sci U S A 106:28972902
150. Ko HS, Lee Y, Shin JH, Karuppagounder SS, Gadad BS, Koleske
AJ, Pletnikova O, Troncoso JC, Dawson VL, Dawson TM (2010)
Phosphorylation by the c-Abl protein tyrosine kinase inhibits
parkins ubiquitination and protective function. Proc Natl Acad
Sci U S A 107:1669116696
151. Cai J, Donaldson A, Yang M, German MS, Enikolopov G,
Iacovitti L (2009) The role of Lmx1a in the differentiation of
human embryonic stem cells into midbrain dopamine neurons in
culture and after transplantation into a Parkinsons disease model.
Stem Cells 27:220229
152. Kim J, Inoue K, Ishii J, Vanti WB, Voronov SV, Murchison E,
Hannon G, Abeliovich A (2007) A MicroRNA feedback circuit
in midbrain dopamine neurons. Science 317:12201224
153. Nelson PT, Wang WX, Rajeev BW (2008) MicroRNAs (miR-
NAs) in neurodegenerative diseases. Brain Pathol 18:130138
154. Hebert SS, De Strooper B (2009) Alterations of the microRNA
network cause neurodegenerative disease. Trends Neurosci
32:199206
155. Miñones-Moyano E, Porta S, Escaramís G, Rabionet R, Iraola S,
Kagerbauer B, Espinosa-Parrilla Y, Ferrer I, Estivill X, Martí E
(2011) MicroRNA profiling of Parkinsons disease brains identi-
fies early downregulation of miR-34b/c which modulate mito-
chondrial function. Hum Mol Genet 20:30673078
156. Greco SJ, Rameshwar P (2007) MicroRNAs regulate synthesis of
the neurotransmitter substance P in human mesenchymal stem
cell-derived neuronal cells. Proc Natl Acad Sci U S A
104:1548415489
157. Ziviani E, Tao RN, Whitworth AJ (2010) Drosophila parkin
requires PINK1 for mitochondrial translocation and ubiquitinates
mitofusin. Proc Natl Acad Sci USA 107:50185023
158. Geisler S, Holmstrom KM, Skuja D, Fiesel FC, Rothfuss OC,
Kahle PJ, Springer W (2010) PINK1/Parkin-mediated mitophagy
is dependent on VDAC1 and p62/SQSTM1. Nat Cell Biol
12:119131
159. Scherzer CR, Grass JA, Liao Z, Pepivani I, Zheng B, Eklund AC,
Ney PA, Ng J, McGoldrick M, Mollenhauer B, Bresnick EH,
Schlossmacher MG (2008) GATA transcription factors directly
regulate the Parkinsons disease-linked gene alpha-synuclein.
Proc Natl Acad Sci U S A 105:1090710912
160. Doxakis E (2010) Post-transcriptional regulation of alpha-
synuclein expression by mir-7 and mir-153. J Biol Chem
285:1272612734
161. Wang G, van der Walt JM, Mayhew G, Li YJ, Zuchner S, Scott
WK, Martin ER, Vance JM (2008) Variation in the miRNA-433
binding site of FGF20 confers risk for Parkinson disease by
overexpression of alpha-synuclein. Am J Hum Genet 82:283289
162. Toda T, Momose Y, Murata M, Tamiya G, Yamamoto M,
Hattori N, Inoko H (2003) Toward identification of suscep-
tibility genes for sporadic Parkinsons disease. J Neurol 250:
III40III43
163. Galvin JE (2004) Neurodegenerative diseases: pathology and the
advantage of single-cell profiling. Neurochem Res 29:10411051
Mol Neurobiol
164. Scherzer CR, Eklund AC, Morse LJ, Liao Z, Locascio JJ, Fefer D,
Schwarzschild MA, Schlossmacher MG, Hauser MA, Vance JM,
Sudarsky LR, Standaert DG, Growdon JH, Jensen RV, Gullans SR
(2007) Molecular markers of early Parkinsons disease based on
gene expression in blood. Proc Natl Acad Sci U S A 104:955960
165. Sinha A, Singh C, Parmar D, Singh MP (2007) Proteomics in
clinical interventions: achievements and limitations in biomarker
development. Life Sci 2080:13451354
166. Patel S, Sinha A, Singh MP (2007) Identification of differentially
expressed proteins in striatum of maneb- and paraquat-induced
Parkinsons disease phenotype in mouse. Neurotoxicol Teratol
29:578585
167. Gerlach M, Riederer P (1996) Animal models of Parkinsons
disease: an empirical comparison with the phenomenology of
the disease in man. J Neural Transm 103:9871041
168. Laterra J, Keep R, Betz LA, Goldstein GW (1999) Bloodbrain
barrier. In: Siegel GJ, Agranoff BW, Albers RW et al (eds) Basic
neurochemistry: molecular, cellular and medical aspects, 6th edn.
Lippincott-Raven, Philadelphia
169. Robbins M, Judge A, Liang L, McClintock K, Yaworski E,
MacLachlan I (2007) 2-O-methyl-modified RNAs act as TLR7
antagonists. Mol Ther 15:16631669
170. Ma Z, Li J, He F, Wilson A, Pitt B, Li S (2005) Cationic lipids
enhance siRNA-mediated interferon response in mice. Biochem
Biophys Res Commun 330:755759
171. Hornung V, Guenthner-Biller M, Bourquin C, Ablasser A, Schlee
M, Uematsu S, Noronha A, Manoharan M, Akira S, de Fougerolles
A, Endres S, Hartmann G (2005) Sequence-specific potent induc-
tion of IFN-alpha by short interfering RNA in plasmacytoid den-
dritic cells through TLR7. Nat Med 11:263270
Mol Neurobiol
... Parkinson's disease (PD) is a complicated and multi-factorial motor disorder of unexplained aetiology. It is mainly associated with movement mutilation and motor impairment owing to selective loss of the nigrostriatal dopamine synthesizing neurons [1][2][3][4][5]. Despite mysterious contributory factors, pesticides have been consistently accused in its pathogenesis [1][2][3][4][5]. ...
... It is mainly associated with movement mutilation and motor impairment owing to selective loss of the nigrostriatal dopamine synthesizing neurons [1][2][3][4][5]. Despite mysterious contributory factors, pesticides have been consistently accused in its pathogenesis [1][2][3][4][5]. Cypermethrin, a commonly used synthetic class II pyrethroid pesticide, is found to be safe at lower doses owing to its short half life. ...
... Mutation in inflammatory genes is found to be associated with PD pathogenesis. Besides, it triggers pro-inflammatory pathways resulting into the microglial activation and free radical generation [5,11,12]. Cypermethrin induces the microglial activation and overexpression of proinflammatory proteins, showing that inflammation could be critical in degeneration of the nigrostriatal dopaminergic neurons [8,10]. However, it is not yet clear whether inflammation is a cause or consequence of cypermethrin-mediated progressive demise of dopamine-producing cells akin to sporadic PD. ...
Article
The present study was undertaken to evaluate the mechanism for antiParkinsonian effect of resveratrol employing 6-hydroxydopamine (6-OHDA) induced experimental model of Parkinson's disease (PD). Resveratrol treatment significantly protects the PD related pathological markers like level of tyrosine hydroxylase, dopamine and apoptotic proteins (Bax and cleaved caspase-3). Disease pathology involves significantly decreased level of dopamine transporter, synaptophysin and postsynaptic density protein 95 (PSD-95) along with augmented level of vesicular monoamine transporter and considerably affected the dendrite arborization. Such affected neuronal communication was significantly restored with resveratrol treatment. Biochemical alterations include the depleted level of glutathione (GSH), mitochondrial complex-I activity with concomitant increased level of lipid peroxidation, nitrite level and calcium levels, which were also significantly inhibited with resveratrol treatment. Altered calcium level induces the endoplasmic reticulum (ER) stress related signalling and phosphorylated Nuclear factor erythroid 2-related factor 2 (Nrf2), and with resveratrol treatment the level of phosphorylated Nrf2 was further increased. The concurrent depleted level of proteasome activity was observed which was attenuated with resveratrol treatment. Proinflammatory cytokines and activated astrocytes were observed which was inhibited with resveratrol treatment. In conclusion, findings suggested that resveratrol exhibits the interference in neuronal communication, oxidative stress, mitochondrial pathophysiology, ER stress, protein degradation mechanism and inflammatory responses and could be utilize in clinics to treat the PD patients.
... Environmental poisons Maneb (MB) and paraquat (PQ) have been used in tests to generate selective damage to dopaminergic neurons, leading to the development of Parkinson's disease (PD). When an ethanolic root extract of Withania somnifera (Ws) was given to a mouse model of Parkinson's disease caused by MB-PQ, it was shown to significantly improve classic Parkinson's disease symptoms like slow movement, less dopamine in the substantia nigra and different types of oxidative damage (Ahmad et al., 2005;Yadav et al., 2012). ...
Article
Full-text available
Natural substances originating from plants have long been used to treat neurodegenerative disorders (NDs). Parkinson’s disease (PD) is a ND. The deterioration and subsequent cognitive impairments of the midbrain nigral dopaminergic neurons distinguish by this characteristic. Various pathogenic mechanisms and critical components have been reported, despite the fact that the origin is unknown, such as protein aggregation, iron buildup, mitochondrial dysfunction, neuroinflammation and oxidative stress. Anti-Parkinson drugs like dopamine (DA) agonists, levodopa, carbidopa, monoamine oxidase type B inhibitors and anticholinergics are used to replace DA in the current treatment model. Surgery is advised in cases where drug therapy is ineffective. Unfortunately, the current conventional treatments for PD have a number of harmful side effects and are expensive. As a result, new therapeutic strategies that control the mechanisms that contribute to neuronal death and dysfunction must be addressed. Natural resources have long been a useful source of possible treatments. PD can be treated with a variety of natural therapies made from medicinal herbs, fruits, and vegetables. In addition to their well-known anti-oxidative and anti-inflammatory capabilities, these natural products also play inhibitory roles in iron buildup, protein misfolding, the maintenance of proteasomal breakdown, mitochondrial homeostasis, and other neuroprotective processes. The goal of this research is to systematically characterize the currently available medications for Parkinson’s and their therapeutic effects, which target diverse pathways. Overall, this analysis looks at the kinds of natural things that could be used in the future to treat PD in new ways or as supplements to existing treatments. We looked at the medicinal plants that can be used to treat PD. The use of natural remedies, especially those derived from plants, to treat PD has been on the rise. This article examines the fundamental characteristics of medicinal plants and the bioactive substances found in them that may be utilized to treat PD.
... In this initial model, which used a high dose of rotenone, there was widespread degeneration beyond the nigrostriatal system, leading to a high mortality rate due to systemic effects, including cardiac, stomach, and liver problems (Cicchetti et al. 2009;Lapointe et al. 2004). This high mortality rate, coupled with the severity of the lesion produced and difficulty in repeating administration in the same animal using a sub-chronic or chronic dosing schedule, made the rotenone model less popular than either the MPTP or 6-OHDA models (Yadav et al. 2012). However, these limitations were overcome by the development of a chronic low-dose regimen, as described in a seminal study by Betarbet et al. (2000). ...
Article
Full-text available
Parkinson’s disease (PD), the second most common neurodegenerative disorder, is characterized by cardinal motor impairments, including akinesia and tremor, as well as by a host of non-motor symptoms, including both autonomic and cognitive dysfunction. PD is associated with a death of nigral dopaminergic neurons, as well as the pathological spread of Lewy bodies, consisting predominantly of the misfolded protein alpha-synuclein. To date, only symptomatic treatments, such as levodopa, are available, and trials aiming to cure the disease, or at least halt its progression, have not been successful. Wong et al. (2019) suggested that the lack of effective therapy against neurodegeneration in PD might be attributed to the fact that the molecular mechanisms standing behind the dopaminergic neuronal vulnerability are still a major scientific challenge. Understanding these molecular mechanisms is critical for developing effective therapy. Thirty-five years ago, Calne and William Langston (1983) raised the question of whether biological or environmental factors precipitate the development of PD. In spite of great advances in technology and medicine, this question still lacks a clear answer. Only 5–15% of PD cases are attributed to a genetic mutation, with the majority of cases classified as idiopathic, which could be linked to exposure to environmental contaminants. Rodent models play a crucial role in understanding the risk factors and pathogenesis of PD. Additionally, well-validated rodent models are critical for driving the preclinical development of clinically translatable treatment options. In this review, we discuss the mechanisms, similarities and differences, as well as advantages and limitations of different neurotoxin-induced rat models of PD. In the second part of this review, we will discuss the potential future of neurotoxin-induced models of PD. Finally, we will briefly demonstrate the crucial role of gene-environment interactions in PD and discuss fusion or dual PD models. We argue that these models have the potential to significantly further our understanding of PD.
... Aging, genetic tendency and exposure to certain exogenous and endogenous chemicals are known to contribute to PD [62,74]. Microbial population alters the metabolism of foreign chemicals that directly enter the gut through oral or nasal route and indirectly via the enterohepatic circulation following dermal absorption through chemical modification. ...
Article
Full-text available
Once recognized as one of the most esoteric diseases of the central nervous system, Parkinson’s disease (PD) is now deemed to be a chronic illness contributed by the central, autonomic and enteric nervous systems. Most likely, an accumulation of α-synuclein in the central and enteric nervous systems is the key that supports this viewpoint. Constipation, one of the non-motor hallmarks in roughly two-third of PD patients, is regulated by the composition of gut bacteria, which is assumed to set off the enteric α-synuclein accrual. Vagus nerve is suggested to direct the signal for α-synuclein over-expression and accumulation to the brain. While trillions of microorganisms reside in the intestinal tract, only one third of the proportion inhabits evenly in all individuals. Existence of an impaired gut-microbe-brain axis consonant with dysbiosis could be an epicenter of this inexplicable disorder. Any alteration in the structure and function of the gastrointestinal tract owing to exposure of endogenous or exogenous chemicals or toxicants could lead to dysbiosis. However, inconsistency in the symptoms even after exposure to same chemical or toxicant in PD patients emphatically creates a conundrum. While the level of a few specific neurotransmitters and metabolites is influenced by microbes, implication of dysbiosis is still debatable. Nevertheless, the scientific literature is overflowing with the remarkable observations supporting the role of dysbiosis in PD. Lack of specificity to differentially diagnose PD with non-PD or PD-plus syndrome, to identify highly precise drug targets and to develop therapeutic stratagems to encounter the disease on the basis of this approach, causes us to be open-minded about the dysbiosis theory. The article reviews the facts supporting gut dysbiosis as the foremost trigger for PD onset along with disagreements.
... Parkinson's disease (PD) is a chronic neurological disorder, phenotypically, biochemically and anatomically identified through motor dysfunction, dopamine depletion and progressive loss of dopaminergic terminals and neurons in the striatum and substantia nigra, respectively Yadav et al. 2012;Singh et al. 2012). The cardinal symptoms of PD are resting tremor, bradykinesia, rigidity and postural instability Dixit et al. 2013). ...
Article
Full-text available
Parkinson’s disease (PD) pathogenesis inevitably involves neuroinflammatory responses attained through contribution of both neuron and glial cells. Investigation done in both experimental models of PD and in samples of PD patients suggested the involvement of both central and peripheral inflammatory responses during PD pathogenesis. Such neuroinflammatory responses could be regulated by neuron-glia interaction which is one of the recently focused areas in the field of disease diagnosis, pathogenesis and therapeutics. Such aggravated neuroinflammatory responses during PD are very well associated with augmented levels of cyclooxygenase (COX). An increased expression of cyclooxygenase (COX) with a concomitant increase in the prostaglandin E2 (PGE2) levels has been observed during PD pathology. Ibuprofen is one of the non-steroidal anti-inflammatory drugs (NSAID) and clinically being used for PD patients. This review focuses on the neuroinflammatory responses during PD pathology as well as the effect of ibuprofen on various disease related signaling factors and mechanisms involving nitrosative stress, neurotransmission, neuronal communication and peroxisome proliferator-activated receptor-γ. Such mechanistic effect of ibuprofen has been mostly reported in experimental models of PD and clinical investigations are still required. Since oxidative neuronal death is one of the major neurodegenerative mechanisms in PD, the antioxidant capacity of ibuprofen along with its antidepressant effects have also been discussed. This review will direct the readers towards fulfilling the existing gaps in the mechanistic aspect of ibuprofen and enhance its clinical relevance in PD therapeutics and probably in other age-related neurodegenerative diseases.
... Revert aid TM minus MuLV reverse transcriptase was used to synthesize cDNA from total RNA as previously described. [23] ...
Article
The neuroprotective effects of an ethanolic extract of Mucuna pruriens (Mp) seeds were evaluated in a Parkinsonian mice model induced by the pesticide paraquat (PQ). PQ induces classical symptoms of Parkinson’s disease (PD) including motor deficits, cellular loss in the substantia nigra (SN) and depletion of dopamine levels. The present study investigates the mechanisms of neuroprotection elicited by Mp, a herb traditionally recognized by the Indian system of medicine, Ayurveda. Co-treatment of PQ-treated mice with Mp significantly enhanced antioxidant activity and decreased oxidant levels. Mp also improved the motor abnormalities observed in PD mice, effectively rescued the levels of dopamine and its metabolites in the SN. HPTLC analysis of the Mp seed extract identified that a large proportion of the extract consisted of L-DOPA and also identified a novel constituent, ursolic acid, present in significant quantity. Mp also facilitated neuroprotection by creating an anti-apoptotic environment indicated by reduced apoptotic (Bax and caspase-3) and increased levels the anti-apoptotic (Bcl2) protein expression, respectively. Altogether, the present study suggests that Mp treatment provides nigrostriatal dopaminergic neuroprotection against PQ induced Parkinsonism by the modulation of oxidative stress and apoptotic machinery possibly accounting for the behavioural effects.
... GW274150, an iNOS inhibitor, reduced microglial activation, cytokine, ROS and NO production, and provided neuroprotection against the 6-OHDA-induced rodent model of PD (see table 2) [167]. Rotenone, used as a toxicant to induce PD in rodents, inhibited mitochondrial complex 1 and induced oxidative stress [168]. Furthermore, it reduced the level of glutathione, which further leads to the increase in nitrite levels and apoptosis [169]. ...
Article
Full-text available
Nitric oxide (NO) is a multifunctional signalling molecule and a neurotransmitter that plays an important role in physiological and pathophysiological processes. In physiological conditions, NO regulates cell survival, differentiation and proliferation of neurons. It also regulates synaptic activity, plasticity and vesicle trafficking. NO affects cellular signalling through protein S-nitrosylation, the NO-mediated posttranslational modification of cysteine thiols (SNO). SNO can affect protein activity, protein-protein interaction and protein localization. Numerous studies have shown that excessive NO and SNO can lead to nitrosative stress in the nervous system, contributing to neuropathology. In this review, we summarize the role of NO and SNO in the progression of neurodevelopmental, psychiatric and neurodegenerative disorders, with special attention to autism spectrum disorder (ASD). Thus, we provide mechanistic insights into the contribution of NO in diverse brain disorders and offers potential and promising therapeutic targets for treatment.
... Hypokinetic rigid syndrome, universally referred to as Parkinson's disease (PD), is a well-known age-related neurodegenerative disorder primarily characterized with motor disability (Olanow 2007;Singh et al. 2006;Schapira 2009;Yadav et al. 2012). Although the tangible contributors of disease have been mysterious, increased age and hereditary and environmental factors are documented as the key perpetrators that accelerate the degeneration of dopamine-producing neurons in the nigrostriatal pathway (Olanow 2007;Schapira 2009;Dauer and Przedborski 2003;Srivastava et al. 2010;Gupta et al. 2014;Rasheed et al. 2017). ...
Article
Full-text available
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) exacerbates mitochondrial impairment and α-synuclein expression leading to Parkinsonism. Impaired mitochondria and over-expressed α-synuclein are degraded and eliminated via macroautophagy and chaperone-mediated autophagy. Owing to multiple properties, silymarin protects from oxidative stress–mediated cellular injury. However, its effect on MPTP-induced changes in autophagy is not yet known. The study aimed to decipher the effect of silymarin on MPTP-induced changes in autophagy. Male mice (20–25 g) were treated with silymarin (intraperitoneally, daily, 40 mg/kg) for 2 weeks. On day 7, a few animals were also administered with MPTP (intraperitoneally, 20 mg/kg, 4 injections at 2-h interval) along with vehicles. Striatal dopamine content was determined. Western blot analysis was done to assess α-synuclein, beclin-1, sequestosome, phosphorylated 5′ adenosine monophosphate–activated protein kinase (p-AMPK), lysosome-associated membrane protein-2 (LAMP-2), heat shock cognate-70 (Hsc-70), LAMP-2A, phosphorylated unc-51-like autophagy activating kinase (p-Ulk1), and phosphorylated mechanistic target of rapamycin (p-mTOR) levels in the nigrostriatal tissue. Silymarin rescued from MPTP-induced increase in beclin-1, sequestosome, p-AMPK, and p-Ulk1 and decrease in LAMP-2, p-mTOR, and LAMP-2A levels. Silymarin defended against MPTP-induced increase in α-synuclein and reduction in dopamine content. The results demonstrate that silymarin protects against MPTP-induced changes in autophagy leading to Parkinsonism.
Article
Full-text available
Maneb, a widely-used dithiocarbamate fungicide, remains in the environment and exerts adverse health effects. Epidemiological evidence shows that maneb exposure is associated with a higher risk of Parkinson’s disease (PD), one of the most common neurodegenerative diseases. However, the molecular mechanisms underlying maneb-induced neurotoxicity remain unclear. Here we investigated the toxic effects and the underlying mechanisms of maneb on the degeneration of dopaminergic cells and α-synuclein in A53T transgenic mice. In SH-SY5Y cells, exposure to maneb reduces cell viability, triggers neuronal apoptosis, induces mitochondrial dysfunction, and generates reactive oxidative species (ROS) in a dose-dependent manner. Furthermore, Western blot analysis found that the mitochondrial apoptosis pathway (Bcl-2, Bax, cytochrome c, activated caspase-3) and the PKA/CREB signaling pathway (PKA, PDE10A, CREB, p-CREB) were changed by maneb both in vitro and in vivo. In addition, the activation of the mitochondrial apoptosis pathway induced by maneb was attenuated by activating PKA. Therefore, these results suggest that the PKA/CREB signaling pathway is involved in maneb-induced apoptosis. This study provides novel insights into maneb-induced neurotoxicity and the underlying mechanisms, which may serve as a guide for further toxicological assessment and standard application of maneb.
Chapter
Genomics comprises several distinct areas of research: transcriptomics, the study of global RNA expression; genotyping, measurement of DNA polymorphisms and mutations; and bioinformatics, the systematic analysis of biological data generated by technologies such as genomics.
Article
Full-text available
Cypermethrin, a class II pyrethroid pesticide, is used to control insects in the household and agricultural fields. Despite beneficial roles, its uncontrolled and repetitive applications lead to unintended effects in non-target organisms. Cypermethrin crosses the blood-brain barrier and induces neurotoxicity and motor deficits. Cypermethrin prolongs the opening of sodium channel, a major site of its action, leading to hyper-excitation of the central nervous system. In addition to sodium channel, cypermethrin modulates chloride, voltage-gated calcium and potassium channels, alters the activity of glutamate and acetylcholine receptors and adenosine triphosphatases and induces DNA damage and oxidative stress in the neuronal cells. Cypermethrin also modulates the level of neurotransmitters, including gamma-aminobutyric acid and dopamine. It is one of the most commonly used pesticides in neurotoxicology research not only because of its variable responses depending upon the doses, time and routes of exposure and strain, age, gender and species of animals used across multiple studies but also owing to its ability to induce the nigrostriatal dopaminergic neurodegeneration. This article describes the effect of acute, chronic, developmental and adulthood exposures to cypermethrin in experimental animals. The article sheds light on cypermethrin-induced changes in the central nervous system, including its contribution in the onset of specific features, which are associated with the nigrostriatal dopaminergic neurodegeneration. Resemblances and dissimilarities of cypermethrin-induced nigrostriatal dopaminergic neurodegeneration with sporadic and chemicals-induced disease models along with its advantages and pitfalls are also discussed.
Article
Recently, we reported specific brain gene expression changes in the chronic MPTP model in the late stage of degeneration, employing cDNA expression array, which indicate a "domino" cascade of events involved in neuronal cell death. In an attempt to elucidate early gene expression profile in the region of the substantia nigra (SN) and the striatum of acute MPTP-treated mice (3-24 h), we elected a restricted number of genes affected by the long-term MPTP treatment, and their expression was examined. Specifically, we detected alterations in the expression of genes implicated in oxidative-stress, inflammatory processes, signal transduction and glutamate toxicity. These pro-toxic genes appear to be compensated by the elevated expression in trophic factors and antioxidant defenses, which are also activated by short exposure to MPTP. The time course of these gene expression changes indicates the importance of investigating the early gene cascade of events occurring prior to late nigrostriatal dopamine neuronal cell death.
Article
The mechanisms underlying Parkinson's disease (PD) and Lewy body (LB) formation, a pathological hallmark of PD, are incompletely understood; however, mitochondrial dysfunction is likely to be at least partially responsible. To study the processes that might be related to nigral neurodegeneration and LB formation, we employed nonbiased quantitative proteomics with isotope-coded affinity tag (ICAT) to compare the mitochondrial protein profiles in the substantia nigra (SN) between controls and mice treated chronically with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a potent mitochondrial toxicant, and an adjuvant, probenecid (prob), for 5 weeks, which produced selective nigrostriatal neurodegeneration with formation of LB-like cytoplasmic inclusions in the remaining nigral neurons. This method identified a total of more than 300 proteins; of these proteins, more than 100 displayed significant changes in relative abundance in the MPTP/prob-treated mice compared to the controls. We validated one of these proteins, DJ-1, whose mutation has been implicated in familial PD, with Western blot analysis, followed by immunohistochemical studies of its distribution in the SN in relation to cytoplasmic inclusions in mice, as well as in classical LBs in PD patients. The results demonstrated that DJ-1 was not only colocalized with alpha-synuclein in dopaminergic neurons but also to cytoplasmic inclusions in mice treated with MPTP/prob. In addition, DJ-1 was present in the halo but not in the core of classical LBs in patients with PD. Our findings suggested that DJ-1 might play an important role in mitochondrial dysfunction, as well as LB formation in PD.
Article
To establish the possible roles of oxidative stress, inflammatory processes and other unknown mechanisms in neurodegeneration, we investigated brain gene alterations in N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mice model of Parkinson's disease using Atlas mouse cDNA expression array membrane. The expression of 51 different genes involved in oxidative stress, inflammation, glutamate and neurotrophic factors pathways as well as in still undefined processes, such as cell cycle regulators and signal transduction molecules, was differentially affected by the treatment. The present study indicates the involvement of an additional cascade of events that might act in parallel to oxidative stress and inflammation to converge eventually into a common pathway leading to neurodegeneration. The attenuation of these gene changes by R-apomorphine, an iron chelator-radical scavenger drug, supports our previous findings in vivo where R-apomorphine was neuroprotective.
Article
DJ-1 is an antioxidant protein whose loss of function by gene mutations has been linked to familial Parkinson’s disease (PD). The main objective of the present study was to determine if this molecule was also involved in the pathogenesis of sporadic PD. For this purpose, quantitative immunoblot assays were performed to evaluate DJ-1 in cerebrospinal fluids (CSF) collected from sporadic PD patients (n=40) and non-PD controls (n=38). The results showed that the CSF DJ-1 levels in PD were significantly higher than those in non-PD controls. Especially, upregulation of CSF DJ-1 in the early stage of PD (Yahr I–II) were distinct compared to those in the advanced stage of PD (Yahr III–IV) and non-PD controls (p
Article
Article
There are major concerns that specific agonal conditions, including coma and hypoxia, might affect ribonucleic acid (RNA) integrity in postmortem brain studies. We report that agonal factors significantly affect RNA integrity and have a major impact on gene expression profiles in microarrays. In contrast to agonal factors, gender, age, and postmortem factors have less effect on gene expression profiles. The Average Correlation Index is proposed as a method for evaluating RNA integrity on the basis of similarity of microarray profiles. Reducing the variance due to agonal factors is critical in investigating small but validated gene expression differences in messenger RNA levels between psychiatric patients and control subjects.