You are on page 1of 14

Send Orders for Reprints to reprints@benthamscience.

net
CNS & Neurological Disorders - Drug Targets, 2020, 19, 000-000 1

RESEARCH ARTICLE

Neuroprotection of Rotenone-Induced Parkinsonism by Ursolic Acid in PD


Mouse Model

Walia Zahra1, Sachchida Nand Rai2, Hareram Birla1, Saumitra Sen Singh1, Aaina Singh Rathore1,
Hagera Dilnashin1, Richa Singh1, Chetan Keswani1, Rakesh K. Singh1 and Surya Pratap Singh1,*

1
Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221005, India; 2Centre of Bio-
technology, University of Allahabad, Prayagraj-211002, India

Abstract: Background: Parkinson’s disease (PD) is characterized by both motor and non-motor
symptoms. The presynaptic neuronal protein, α-Synuclein, plays a pivotal role in PD pathogenesis and
is associated with both genetic and sporadic origin of the disease. Ursolic Acid (UA) is a well-known
bioactive compound found in various medicinal plants, widely studied for its anti-inflammatory and
antioxidant activities.
Objective: In this research article, the neuroprotective potential of UA has been further explored in the
Rotenone-induced mouse model of PD.
Methods: To investigate our hypothesis, we have divided mice into 4 different groups, control, drug
ARTICLE HISTORY only control, Rotenone-intoxicated group, and Rotenone-intoxicated mice treated with UA. After the
completion of dosing, behavioral parameters were estimated. Then mice from each group were sacri-
ficed and the brains were isolated. Further, the biochemical tests were assayed to check the balance
Received: April 24, 2020 between the oxidative stress and endogenous anti-oxidants; and TH (Tyrosine Hydroxylase),
Revised: June 29, 2020 α-Synuclein, Akt (Serine-threonine protein kinase), ERK (Extracellular signal-regulated kinase) and
Accepted: July 01, 2020
inflammatory parameters like Nuclear factor-κB (NF-κB) and Tumor Necrosis Factor- α (TNF-α) were
DOI: assessed using Immunohistochemistry (IHC). Western blotting was also done to check the expressions
10.2174/1871527319666200812224457
of TH and α-Synuclein. Moreover, the expression levels of PD related genes like α-Synuclein,
β-Synuclein, Interleukin-1β (IL-1β), and Interleukin-10 (IL-10) were assessed by using Real-time
PCR.
Results: The results obtained in our study suggested that UA significantly reduced the overexpression
of α-Synuclein and regulated the phosphorylation of survival-related kinases (Akt and ERK) apart
from alleviating the behavioral abnormalities and protecting the dopaminergic neurons from oxidative
stress and neuroinflammation.
Conclusion: Thus, our study shows the neuroprotective potential of UA, which can further be ex-
plored for possible clinical intervention.

Keywords: Parkinson’s disease, Rotenone, α-Synuclein, neuroinflammation, oxidative stress, ursolic acid.

1. INTRODUCTION are found in the form of intracellular inclusions known as


Lewy Bodies (LBs) and Lewy Neurites (LNs), which are
PD is the second most common neurodegenerative disor-
der, clinically detected by several motor symptoms such as also predominantly considered as the neuropathological
bradykinesia, rigidity, resting tremors, and postural instabil- hallmarks of PD. The fact that elevated levels of LBs and
ity that might be originated sporadically or have a familial LNs in the basal ganglia concomitantly lead to increased
origin. Different genetic and environmental factors are sup- degeneration of Dopaminergic (DA) neurons in-vivo strong-
posed to be responsible for the pathobiogenesis of PD [1]. In ly puts forward that the overexpression of α-Synuclein is one
Substantia Nigra pars compacta (SNpc) of basal ganglia, of the main pathological hallmarks of PD [2-4]. Oxidative
primarily insoluble aggregates of ubiquitin and α-Synuclein stress and neuroinflammation play a crucial role in the pro-
gression of PD. In response to neuroinflammation, microglia
get activated, and - in turn - produce and release neurotoxic
*Address correspondence to this author at the Department of Biochemistry, factors such as Tumor Necrosis Factor-α (TNF-α) and Inter-
Institute of Science, Banaras Hindu University, Varanasi-221005, India;
Tel: 0542-6701543; E-mail: suryasinghbhu16@gmail.com leukin-1β (IL-1β). Some experimental studies have reported

1871-5273/20 $65.00+.00 © 2020 Bentham Science Publishers


2 CNS & Neurological Disorders - Drug Targets, 2020, Vol. 19, No. 0 Zahra et al.

that suppression of pro-inflammatory mediators and free technology (Santa Cruz, CA, United States), and the primary
radical generation, along with the inhibition of α-Synuclein antibodies for α-Synuclein (ab191692), TNF-α (ab1793),
aggregation, could be achieved by exploiting the naturally Akt (ab81283), ERK (ab76165) and NF-κB (ab16502) were
occurring compounds from medicinal herbs [5, 6]. Also, procured from Abcam Life Science, Biogenuix Medsystems
evidences from several studies suggest that abnormality in Pvt. Ltd. (New Delhi, India). Secondary antibodies for
the function of different protein kinases; such as the kinases western blot were mouse anti-rabbit IgG-HRP (Horseradish
of the Phosphoinositide 3-Kinase (PI3K) and Mitogen- peroxidase) (sc-2357) and Goat anti-mouse (GeNei-
Activated Protein Kinase (MAPK) contribute to PD’s etiolo- 62114068001A), and secondary fluorescent-tagged antibod-
gy [7, 8]. ERK and Akt are the respective modulators of ies for IHC (such as Cy2 conjugated and Cy3 conjugated for
MAPK and PI3K pathways [8-11]. Overexpression of α- immunohistochemistry) were purchased from Merck Milli-
Synuclein is also linked with suppression in the phosphoryla- pore and Chemicon, respectively.
tion and of these molecules in different studies [12-14]. There-
fore, by understanding how the activities of these enzymes get 2.2. Experimental Animals
changed during the disease pathogenesis would allow us to Adult Swiss albino male mice weighing 25 ± 5g were
modulate these signaling pathways for PD therapy. provided by an animal research facility of the Institute of
Rotenone is a widely used neurotoxin for generating an- Medical Sciences, Banaras Hindu University, Varanasi, In-
imal models of chronic PD and is being frequently used to dia, to conduct the experiment. Mice were housed in clean
screen potential compounds that might be used therapeutical- polypropylene cages in a regulated air-conditioned environ-
ly to improve the symptoms of PD [15, 16]. Medicinal plants ment on 12 h light-dark cycles with access to standard mice
are being used as an alternative drug therapy because of hav- pellet diet and water. All efforts were made to minimize the
ing minimal side-effects [17-23]. Ursolic Acid (UA) is found suffering of the animals, and all the animal experiments were
as a component of medicinal herbs and ornamental species. performed in accordance with the experimental practices and
It is also found in fruits like prunes, apples, blueberries, and standards approved by the Animal Ethics Committee of Ba-
cranberries [24, 25], with their amounts ranging between naras Hindu University, Varanasi, India.
0.15 to 1.8 g/100 g dry matter [26]. Moreover, UA has been
reported as a potent anti-oxidative agent in 1-methyl-4- 2.3. Experimental Design
phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mice After the mice were acclimatized to the laboratory condi-
model of PD [27]. The compound is capable of crossing the tions, they were divided into 4 groups with 8 animals in each
blood-brain barrier [28] and various reports have demon- group. Group-I was used as a control group. Group-II was
strated the neuroprotective effects of UA through its anti- drug only control: mice were orally administered with UA
oxidative and anti-inflammatory properties in different ani- (25mg/kg bwt/day) for 42 days. Group-III was given Rote-
mal models [27, 29-31]. Therefore, the keystone of our study none (2 mg/kg bwt/day) subcutaneously for 35 days. Lastly,
was to explore further the neuroprotective potential of UA in Group-IV was first orally-administered with UA for a week,
Rotenone-induced Parkinsonian mice model. then simultaneous administration of UA and Rotenone-
Regardless of various studies already being conducted intoxication was done for the next 35 days, similar to that of
based on the anti-oxidative and neuroprotective properties of Group-III. Dosing of UA was done according to Rai et al.,
UA, the present study demonstrates the potential of UA 2016, with slight modifications [27].
against nigrostriatal degeneration of DA neurons by the inhi-
bition of α-Synuclein-mediated neurotoxicity in Rotenone- 2.4. Behavioral Assessment
induced mouse model of PD.
After the completion of the administration of the dosages,
2. MATERIALS AND METHODS behavioral parameters were performed at room temperature
without any outside interference and their motor discrepan-
2.1. Reagents and Antibodies cies were studied. Various tests were performed for the as-
Normal goat serum (NGS), UA, and Rotenone were pro- sessment, including rotarod, narrow beam walking, and
cured from Sigma-Aldrich (St. Louis, MO, United States). hanging tests.
Bovine serum albumin (BSA), ammonium chloride, potassium
chloride, acetic acid, reduced nicotinamide adenine dinucleo- 2.4.1. Rotarod Test
tide phosphate (NADPH), disodium hydrogen phosphate, and The mice were trained for three consecutive days at a
sodium dihydrogen phosphate were purchased from Sisco fixed speed (15 rpm) and then subjected to analysis. The
Research Laboratories (SRL, Mumbai, India). The protein performance time was recorded for a maximum of 5 min,
estimation kit by Bradford GeNeiTM, potassium dichromate while mice ran on the rod. Each mouse was subjected to four
and hydrogen peroxide (H2O2) were bought from Merck trials, and the average time was recorded [32]. Similarly,
(Darmstadt, Germany), and thiobarbituric acid (TBA), 1,4- after the completion of the experiments, the rotarod test was
diazabicyclo[2.2.2]octane (DABCO), sodium dodecyl sul- again performed and the mean of their falling latency was
phate (SDS), and Griess reagent were obtained from HiMe- calculated.
dia (Mumbai, India). Paraformaldehyde and sodium nitrite
were bought from Lobachemie, India. Protease inhibitor and
2.4.2. Narrow Beam Walking Test
RNase inhibitor were procured from Sigma Aldrich and Ge-
nei, respectively. Primary antibodies for β-actin (SC-47778) The purpose of performing this test was to assess the
and TH (SC-25269) were purchased from Santa Cruz, Bio- mouse’s motor coordination, where the mouse needed to stay
Neuroprotection of Rotenone-Induced Parkinsonism by Ursolic Acid CNS & Neurological Disorders - Drug Targets, 2020, Vol. 19, No. 0 3

upright and walk across an elevated narrow beam to a safe enzyme activity was done in n moles/min/mg protein. By
platform. Primarily, the mice were unaware/untrained for this using its substrate NADH, the activity of SOD was evaluated
test. Hence, they were trained to walk on a stationary narrow [39]. Ultimately, the absorbance was read at 560 nm against
wooden beam placed 100 cm above the floor (L100 cm × W1 reagent blank. The difference between reference and experi-
cm). The experiment was repeated thrice and the time taken mental OD of the sample gave the inhibition of Nitro Blue
by the mice to cross over the beam was recorded [33]. Tetrazolium chloride (NBT) reduction by an enzyme source.
From the enzyme source, the protein was also estimated.
2.4.3. Hanging Test Under the assay conditions, the unit of SOD enzyme activity
was defined as the amount of enzyme required to inhibit the
On a horizontal grid, each mouse was placed and allowed optical density at 560 nm of NBT reduction by 50% in 1
to hold it firmly before inverting it so that the mouse hung min. The activity of SOD was expressed as unit per milli-
upside down until it lost its grip and fell. The time of hang- gram of protein.
ing was recorded, and the experiment was repeated thrice
[34].
2.6. Immunohistochemical Staining
2.5. Biochemical Parameters For the histopathological assessment, mice from each
group were anesthetized with diethyl ether, perfused intra-
The mice were sacrificed by cervical dislocation, fol- cardially initially with 0.9% saline (chilled) followed by 4%
lowed by decapitation with minimum pain. The brains were paraformaldehyde (chilled) prepared in 0.1 M phosphate-
dissected out quickly on dry ice and were stored at -80oC buffered saline (PBS), pH 7.4, and then decapitated. Brains
until further use. The SNpc was procured [35] and further were extracted and post-fixed in 10% paraformaldehyde
homogenized in KCl buffer (Tris–HCl 10 mM, NaCl 140 overnight and later cryoprotected in 30% sucrose solution.
mM, KCl 300 mM, Ethylenediaminetetraacetic acid 1 mM, Immunohistochemical staining of TH, α-Synuclein, Akt-1,
Triton-X 100 0.5%) at pH 8.0, supplemented with protease ERK1/2, TNF-α and NF-κB was performed in SNpc using
and phosphatase inhibitors. The acquired tissue homogenates the standard procedure [40]. Coronal sections of 20 µm
were then subjected to centrifugation at 12,000 × g for 20 thickness passing through SNpc were cut using a cryomicro-
min at 4oC to obtain a supernatant. The specific antioxidant tome (Leica, Wetzlar, Germany). The sections were rinsed
activities of the enzyme were measured for Superoxide Dis- thrice for 10 min each with 0.01 M PBS (pH 7.4), and then
mutase (SOD) and catalase, along with determining the re- blocked with 10% NGS in PBS 0.3% Triton-X 100 and 1%
duced Glutathione content, levels of lipid peroxidation, and BSA in phosphate-buffered saline with Triton X-100
nitrite from the supernatant. (PBST), as blocking reagent for about 1 h. The sections were
further incubated with primary antibody (polyclonal anti-
2.5.1. Estimation of Lipid Peroxidation and Nitrite Levels mouse antibody against TH in 1:1000 dilution, polyclonal
The method used by Ohkawa et al., [36] was incorpo- anti-rabbit α-Synuclein in 1:1500 dilution, monoclonal anti-
rated with few modifications to test tissue lipid peroxidation. mouse TNF-α in 1:700 dilution, monoclonal anti-rabbit
Foremost, 10% tissue homogenate was added to 10% SDS pAkt1 (1:1000), polyclonal anti-rabbit antibody pERK1/2
and then mixed with 20% acetic acid. Subsequently, the re- (1:1000), polyclonal anti rabbit NF-κB p65 antibody in
action mixture was kept in a boiling water bath for 1 h after 1:1000 dilution) for 16 h at 4oC. Further, in order to remove
adding 0.8% TBA. Later, when the mixture was cooled and unbound primary antibodies, rinsing in PBS and 1% BSA-
centrifuged to extract out supernatant, the absorbance was PBS was done twice for each well. It was then incubated with
recorded at 532 nm in reference to control. Peroxidation of Cy2-conjugated secondary antibody (for anti-mice primary)
lipids was measured in terms of micromoles of malondialde- and Cy3-conjugated secondary antibody (for anti-rabbit pri-
hyde (MDA) released per milligram protein. The level of mary), prepared in 1% BSA-PBS, for 1 h at room temperature.
nitrite synthesis was assessed according to the standard pro- Then, sections were rinsed thrice with 1% BSA-PBS at 3-min
cedures with slight modifications [37]. The 10% tissue ho- interval, and 4’,6-diamidino-2-phenylindole (DAPI) was add-
mogenate was taken and mixed with ammonium chloride ed (1 mg/ml). Ultimately, sections were rinsed thrice with
and Griess reagent. The solution was allowed to stand for PBS and then mounted on slides using polyvinyl alcohol
half an hour at 37oC, and then the absorbance was taken at mounting medium with DABCO anti-fading (Fluka analyti-
540 nm. The nitrite content was calculated using a standard cal). Visualisation was performed under Nikon fluorescent
curve for sodium nitrite (10-100 mM) in terms of mi- microscope (Thermo Fisher Scientific). Percentage area val-
cromoles per milliliter. ue and cell counting were determined by using Image J soft-
ware 1.52a (NIH, United States).
2.5.2. Estimation of Antioxidant Levels
2.7. Real-Time Polymerase Chain Reaction (PCR) Analy-
Reduced GSH content in the brain homogenate was de- sis
termined using the method described by Moron et al., (1979)
and values were expressed as µg/mg protein [38]. The rate of Total RNA from the frozen tissues was extracted using
decomposition of hydrogen peroxide (substrate) was exam- the TRIzol RNA isolation reagent (Invitrogen, Carlsbad, CA,
ined using a spectrophotometer to assess the catalase activity United States) according to the manufacturer’s instructions.
[35]. To do this, the striatal tissue was mixed with potassium The RNA yield was quantified on Nanodrop 1000, and the
dichromate and acetic acid (1:3) in a boiling water bath for RNA purity was determined based on the A260/A280 ratio.
10 min, and OD was taken at 570 nm. Measurement of the For cDNA preparation, reverse transcription was carried out
4 CNS & Neurological Disorders - Drug Targets, 2020, Vol. 19, No. 0 Zahra et al.

using 2 mg total RNA (kept equal for each amplification) test, whereas the fold changes of mRNA were analyzed by
and 20U M-MLV reverse transcriptase (Fermentas, Germa- Student’s two-tailed t-test through Graph Pad Prism 5.0
ny), 1 X reverse transcriptase (RT) buffer, 20mM dNTPs software. The results are expressed as the means ± SEM, and
(New England Biolabs, United States), 20 U RNasin (Fer- p-values less than 0.05 (p < 0.05) were considered statistical-
mentas, Germany), 0.1 M dithiothreitol (DTT) with dieth- ly significant.
ylpyrocarbonate (DEPC)-treated water, and 100 ng of ran-
dom hexamers (Fermentas, Germany). Gene expression pro- 3. RESULTS
file analysis was done on the ABI7500 Fast system. Glycer-
aldehyde 3-phosphate dehydrogenase (GAPDH) was taken 3.1. Behavioural Studies
as an endogenous control. PCR was operated according to
3.1.1. Effect of UA on Motor Coordination
the previously reported studies, with few modifications. The
reactions were operated by using 10 ml of a real-time mix It was seen that, compared to control mice, the duration
containing 5 ml of SYBER green master mix (Applied Bio- of Rotenone-intoxicated mice in the hanging test was signifi-
system), 1 ml cDNA, 2 ml nuclease-free water, 0.5 ml each cantly poorer (p<0.0001; time reduced to 0.58-fold (Rote-
of forward and reverse primers, and 1 ml RNase inhibitor. none) as compared to control). On the other hand, the UA
The samples were initially incubated at 50oC for 2 min, fol- treatment given to the Rotenone group significantly in-
lowed by denaturation at 95oC for 10 min. This was pro- creased the hanging time (p<0.001; time was increased to
gressed by 40 cycles at 95oC for 15 s, 60oC for 1 min, and 0.63 fold (Rotenone+UA) as compared to Rotenone), as
72oC for 40 s [41]. The abundance or decline of mRNA was compared to the hanging time Rotenone-intoxicated group
normalized to the geometric average of endogenous control (Fig. 1A). It was observed that the time duration of Rote-
GAPDH for ΔCt. The fold change was calculated using 2-Δ Δ
Ct none-intoxicated mice on rotarod significantly reduced,
method and reported as fold change unit.
compared to control (p<0.0001; time reduced to 0.5-fold
(Rotenone) as compared to control). On the other hand, Ro-
2.8. Western Blot Analysis
tenone-intoxicated mice that were given UA treatment stayed
Western blot analysis was done according to the method significantly longer on the rotarod than Rotenone mice
described by Gupta et al. in 2010 with slight modifications (p<0.001; time was increased to about 0.6 fold (Rote-
[42]. Primary antibody dilutions included TH (1:4000), α- none+UA) as compared to Rotenone) (Fig. 1B).
Synuclein (1:1000) and β-actin (1:500). The blots obtained
were developed by using 3,3’-Diaminobenzidine (DAB) and Moreover, in narrow beam walking test, rotenone-
H2O2 as substrates. The relative band density was calculated induced mice showed a higher increase in time duration in
with respect to the expression of β-actin, and the fold change comparison with control mice (p<0.0001; time increased to
indicated the resultant expressions of proteins. 2.6 folds (Rotenone) as compared to control). However, the
UA-treated rotenone-induced group showed a significant
2.9. Statistical Analysis reduction in the narrow beam walking time, as compared to
One-way analysis of variance (ANOVA) was used to Rotenone mice (p<0.001; time was reduced to 0.76 fold (Ro-
analyze the data by applying the Student-Newman-Keuls tenone+UA) as compared to control) (Fig. 1C).

Fig. (1). Effect of UA on behavioral parameters. (A) A significant improvement in hanging time was observed upon UA administration in
Rotenone intoxicated group. Hanging time in Rotenone-induced Parkinsonian mice was observed to be comparatively reduced as compared
to control. (B) Rotenone-intoxicated mice group has stayed on the rotarod for a lesser time interval as compared to that of the control group.
UA administered-Rotenone group has stayed significantly more on the rotarod as compared to the untreated Rotenone-induced PD mice
group. (C) As compared to control, narrow beam walking time was found to be comparatively increased in Rotenone-intoxicated mice.
While UA treatment has reduced the walking time in the Rotenone-intoxicated group, as shown in comparison with untreated Rotenone-
intoxicated mice. Data are expressed in terms of mean ± SEM (*p < 0.01, **p < 0.001, ***p < 0.0001, n = 8). UA, Ursolic acid; SEM, stand-
ard error of mean. (A higher resolution/colour version of this figure is available in the electronic copy of the article).
Neuroprotection of Rotenone-Induced Parkinsonism by Ursolic Acid CNS & Neurological Disorders - Drug Targets, 2020, Vol. 19, No. 0 5

Fig. (2). Estimation of Catalase, MDA, SOD, GSH and nitrite in SNpc of mice. Rotenone intoxication to the mice has significantly reduced
the antioxidants; Catalase (A), SOD (C) and GSH (D) and increased the level of oxidative stress mediators MDA (B) and Nitrite (E) in com-
parison to control group. Whereas, UA administration to the Rotenone-induced PD mice group has concomitantly increased the level of
Catalase (A), SOD (C) and GSH (D) and decreased the level of MDA (B) and Nitrite (E) as compared to the Rotenone-intoxicated group.
Values are expressed as mean ± SEM (*p < 0.01, **p < 0.001, ***p < 0.0001, n = 4). SOD, Superoxide dismutase; MDA, malondialdehyde;
GSH, glutathione; SEM, standard error of mean. (A higher resolution/colour version of this figure is available in the electronic copy of the
article).

3.2. Biochemical Studies icantly reduced (p<0.0001), while GSH levels (Fig. 2D) sig-
nificantly enhanced (p<0.0001), after the administration of
3.2.1. Effect of UA on Catalase, SOD and Nitrite
UA to the Rotenone-intoxicated group.
The results revealed that in comparison to the control
group, the Rotenone-intoxicated mice showed a significant 3.3. Effect of UA on the Expression of TH, α-Synuclein
reduction in the activities of Catalase (Fig. 2A) and SOD and TNF-α in SNpc
(Fig. 2C) (p<0.0001), and a significant increase in the nitrite
In comparison to the control group mice, in the rotenone-
content (Fig. 2E) (p<0.0001). Although, as compared to the
intoxicated group, the expression of α-Synuclein (p<0.0001;
Rotenone-treated group, UA-treated rotenone-induced mice
percentage area increased from about 2% (Control) to 6.5%
showed increased activity of catalase (p<0.01) (Fig. 2A),
SOD (p<0.001) (Fig. 2C), and decreased nitrite level (Rotenone)) (Fig. 3A) and TNF-α (p<0.001; percentage area
increased from about 4% (Control) to 7.5% (Rotenone))
(p<0.0001) (Fig. 2E).
(Fig. 3C) was found to be significantly enhanced. Although
on treatment with UA (Rotenone+UA), a fall in the expres-
3.2.2. Effect of UA on MDA and Glutathione (GSH) Con- sion of α-Synuclein (p<0.001; percentage area reduced from
tent
about 6.5% (Rotenone) to 3% (Rotenone+UA)) (Fig. 3A)
Significant elevation (Fig. 2B) in the level of lipid perox- and TNF-α (p<0.01; percentage area reduced from about 7.5%
idation product, MDA, was seen in the Rotenone-intoxicated (Rotenone) to 5% (Rotenone+UA)) (Fig. 3C) was noticed in
group as compared to the control group (p<0.0001). On the comparison with Rotenone-intoxicated mice. Furthermore, in
other hand, GSH level showed a significant decline (Fig. 2D) comparison to control group, Rotenone-intoxication reduced
in the Rotenone-intoxicated group, when compared to the the number TH-positive neurons (p<0.001; number of TH-
control group (p<0.0001). However, compared to the Rote- positive neurons reduced from about 200 (Control) to 125
none group, MDA level (Fig. 2B) was observed to be signif- (Rotenone)) (Fig. 3B), whereas, in comparison to Rotenone
6 CNS & Neurological Disorders - Drug Targets, 2020, Vol. 19, No. 0 Zahra et al.

Fig. (3). Immunofluorescence expression of α-Synuclein, TH and TNF-α in SNpc of Control, Drug only Control (Control+UA), Rotenone
and Rotenone+UA mice groups by using Image J Software at 20x magnification. Rotenone intoxication has led to a significantly increased
expression of α-Synuclein (A) and TNF-α (C) and reduced immunoreactivity of TH (B) as compared to the control group. While, UA admin-
istration in PD mice has significantly alleviated the expression of α-Synuclein (A) and TNF-α (C) and enhanced the immunoreactivity of TH
(B) as compared to Rotenone-intoxicated mice. Values are expressed as mean ± SEM of percentage area (*p < 0.01, **p < 0.001, ***p <
0.0001, n = 4). TNF-α, tumor necrosis factor alpha; TH, Tyrosine hydroxylase; SNpc, substantia nigra pars compacta; UA, Ursolic acid. The
arrows represent the expression of the respective antibodies in SNpc. Scale bar represents the degree of magnification of the image (For 20X=
100µm). (A higher resolution/colour version of this figure is available in the electronic copy of the article).

group, the UA treatment group showed significantly increased none-intoxicated mice occurred (p<0.001; Integrated Fluo-
numbers of TH-positive neurons in SNpc of mice brain rescence Value (IFV) increased from about 3 to 12) (Fig. 5B)
(p<0.001; number of TH-positive neurons increased from as a result of Rotenone intoxication. On the other hand, UA
about 125 (Rotenone) to 180 (Rotenone+UA)) (Fig. 3B). treatment (p<0.01; Integrated Fluorescence Value (IFV) re-
duced from about 12 to 7.5) (Fig. 5C) significantly inhibited
3.4. Effect of UA on the Expression of p-Akt-1 and p- the nuclear translocation of NF-κB in Rotenone-intoxicated
ERK1/2 mice.
The intoxication of Rotenone in mouse has led to the
3.6. Real-Time PCR Analysis: Effect of UA on mRNA
decreased phosphorylation of Akt (p<0.0001; percentage
Expression of Proinflammatory Cytokine, IL-1 β;
area reduced from about 14% (Control) to 6% (Rotenone))
Anti-inflammatory Cytokine, IL-10; α-Synuclein and
(Fig. 4A) and ERK1/2 (p<0.0001; percentage area reduced
β-Synuclein
from about 15% (Control) to 7% (Rotenone)) (Fig. 4B) when
compared with the control group, whereas, treatment of UA When compared to control group, the mRNA expressions
to the PD mouse model has shown the elevated level of of α-Synuclein (p<0.0001) (Fig. 6A) and the pro-inflammatory
phosphorylation for both Akt (p<0.0001; percentage area cytokine, IL-1β (p<0.01) (Fig. 6C) were found to be signifi-
increased from about 6% (Rotenone) to 8% (Rotenone+UA)) cantly elevated, while the mRNA expressions of β-Synuclein
(Fig. 4A) and ERK1/2 (p<0.0001; percentage area increased (p<0.01) (Fig. 6B) and IL-10 (p<0.001) (Fig. 6D) were com-
from about 7% (Rotenone) to 9% (Rotenone+UA)) (Fig. 4B) paratively reduced in Rotenone-intoxicated group. UA ad-
as compared to the disease group. ministration to the Rotenone-induced Parkinsonian mice
group, on the other hand, had significantly reduced mRNA
3.5. Effect of UA on the Nuclear Translocation of NF-κB levels of α-Synuclein (p<0.0001) (Fig. 6A) and IL-1β
(p65) in SNpc (p<0.0001) (Fig. 6C) and significantly enhanced mRNA
When compared to the control group (Fig. 5A), it was levels of β-Synuclein (p<0.0001) (Fig. 6B) and IL-10
observed that the nuclear translocation of NF-κB in Rote- (p<0.0001) (Fig. 6D).
Neuroprotection of Rotenone-Induced Parkinsonism by Ursolic Acid CNS & Neurological Disorders - Drug Targets, 2020, Vol. 19, No. 0 7

Fig. (4). Immunohistochemical expression of p-Akt-1 and p-ERK1/2 in SNpc of Control, Drug only Control (Control+UA), Rotenone and
Rotenone+UA mice groups by using Image J Software at 20x magnification. Phosphorylation of both Akt-1 (A) and ERK1/2 (B) was de-
creased upon rotenone intoxication in the PD group when compared to the control group. While, UA administration in PD mice has signifi-
cantly elevated the phosphorylation of both the kinases Akt-1 (A) and ERK1/2 (B) as compared to the disease group. Values are expressed as
mean ± SEM of percentage area (*p < 0.01, **p < 0.001, ***p < 0.0001, n = 4). Akt, Serine/Threonine-protein kinase; ERK, Extracellular
signal-related kinase. The arrows represent the expression of the respective antibodies in SNpc. Scale bar represents the degree of magnifica-
tion of the image (For 20X= 100µm). (A higher resolution/colour version of this figure is available in the electronic copy of the article).

Fig. (5). Effect of UA on the nuclear translocation of NF-κB in SNpc with 40x magnifications after immunostaining. Rotenone intoxication
has increased the nuclear translocation of NF-κB (B) in substantia nigra pars compacta (SNpc) in Rotenone as compared to the control group
(A). Although, the nuclear translocation of NF-κB was significantly reduced in Rotenone+UA (C) as compared to the Rotenone-intoxicated
mice group (B). Values are expressed as mean ± SEM of Integrated Fluorescence Value (IFV) (*p < 0.01, **p < 0.001). NF-κB, Nuclear Fac-
tor-κB. The arrows represent the expression of the antibody in SNpc. Scale bar represents the degree of magnification of the image (For 40X=
150µm). (A higher resolution/colour version of this figure is available in the electronic copy of the article).
8 CNS & Neurological Disorders - Drug Targets, 2020, Vol. 19, No. 0 Zahra et al.

Fig. (6). Effect of UA on mRNA expression levels of α-Synuclein, β-Synuclein, IL-1 β and IL-10 in the Rotenone-intoxicated PD mouse
model. The mRNA levels of α-Synuclein (A) and pro-inflammatory cytokine, IL-1 β (C) were significantly elevated and of β-Synuclein (B)
and anti-inflammatory cytokine, IL-10 (D) concomitantly reduced upon Rotenone intoxication as compared to control group. Whereas, UA
treatment after Rotenone-intoxication has significantly attenuated the mRNA levels of α-Synuclein (A) and IL-1 β (C) and increased the
mRNA levels of β-Synuclein (B) and IL-10 (D) as compared to Rotenone-induced PD mouse group. Values in the graph are represented as
mean ± SEM with the level of significance *p < 0.01, **p < 0.001 and ***p < 0.0001. UA, Ursolic acid; IL-1 β, Interleukin-1 β; IL-10, Inter-
lekin-10; SEM, standard error of mean. (A higher resolution/colour version of this figure is available in the electronic copy of the article).

3.7. Western Blot Analysis of TH and α-Synuclein oxidative stress, neuroinflammation; reduced the phosphory-
lation of Akt and ERK and increased the expression of α-
The expression levels of α-Synuclein (17 KDa) and TH Synuclein in the mice model of PD.
(62 KDa) (Fig. 7) were also assessed using the Western blot
analysis in tissue lysates isolate of the SNpc region. An en- Previous studies have implicated the anti-cancerous [49],
hanced α-Synuclein (p<0.0001) (A) and reduced TH anti-oxidative [27, 50] and neuroprotective activities of UA
(p<0.0001) (B) expression levels were seen in Rotenone- [48-51]. In our previous study, we have demonstrated the
intoxicated mice, as compared to the control group. In con- neuroprotective activity of UA in MPTP-induced Parkin-
trast, for treatment with UA, a reduced level of α-Synuclein sonian mice model [27] while our present study further dis-
(p<0.001) (A) and increased expression of TH level cusses the neuroprotective potential of UA in Rotenone-
(p<0.001) (B) were observed when compared to Rotenone- induced PD mouse model by inhibiting the overexpression of
intoxicated mice. α-Synuclein, oxidative stress and neuroinflammation and
altering the phosphorylation of the cell- survival-related ki-
4. DISCUSSION nases (Akt and ERK).
The pesticide Rotenone is widely used by gardeners and The symptoms like the slow movement, postural imbal-
is a potent mitochondrial complex I inhibitor that induces the ance, and resting tremor are commonly observed in patients
accumulation of α-Synuclein [15, 16]. Most of the clinical of PD. These symptoms are attributed to the loss of coordi-
and pathological hallmarks of PD are successfully recapitu- nation between the brain and the muscles due to the depletion
lated by Rotenone-intoxication [43, 44]. Substantial evidence of the neurotransmitter, dopamine. Remarkably, the results of
also indicates the reduced phosphorylation of two important our study have also shown the characteristic behavioural im-
kinases of the PI3K and MAPK pathways, viz. Akt and ERK pairment from Rotenone intoxication, similar to that of the
by Rotenone exposure [45-48]. Our study also reveals that patients of PD [27, 55, 58]. On the other hand, UA administra-
Rotenone has induced motor deficits, DA neuronal death, tion to the Parkinsonian mice significantly alleviated these
Neuroprotection of Rotenone-Induced Parkinsonism by Ursolic Acid CNS & Neurological Disorders - Drug Targets, 2020, Vol. 19, No. 0 9

Fig. (7). Western blot analysis to detect the expression level of α-Synuclein and tyrosine hydroxylase (TH) in the SNpc tissue. Values are
expressed as mean ± SEM (**p < 0.001, ***p < 0.0001). The expression of α-Synuclein (17 KDa) (p<0.0001) (A) was increased and TH (62
KDa) (p<0.0001) (B) was decreased in the Rotenone group as compared to the Control group. While, UA treatment to the Rotenone-induced
Parkinsonian mice model has downregulated the expression of α-Synuclein (p<0.001) (A) and upregulated the expression of TH (p<0.001)
(B) as compared to the Rotenone group. (A higher resolution/colour version of this figure is available in the electronic copy of the article).

symptoms, as shown by the assessment of the different be- Brains of PD patients have been well-documented to show
havioural parameters in our study. alteration in redox system [63-65]. Moreover, an imbalance
in redox potential leads to the decline in GSH levels in the
PD pathogenesis is seen to be triggered by two major
brain tissues of Rotenone-intoxicated mice, as similarly re-
factors, oxidative stress and mitochondrial dysfunction [57].
flected by our study. In contrast, GSH level was significantly
Stressful environment leads to the production of free radi-
restored by the UA treatment in Parkinsonian mice, as shown
cals, which further react with free oxygen molecules present
on membrane lipids to generate peroxy radicals responsible by the results of our study. Also, low levels of anti-oxidant
enzymes such as Catalase and SOD make the brain more
for lipid peroxidation. The diagnostic marker for lipid perox-
prone to oxidative damage. Rotenone exposure leads to the
idation, MDA, is used as a lipid biomarker of PD [58, 59].
significant reduction of SOD and Catalase in the striatum of
As suggested in previous studies, our study shows the ele-
the basal ganglia, as observed in our study. H2O2 produced
vated levels of MDA in the nigrostriatal region of Rotenone
during oxidative stress is responsible for inactivating these
intoxicated mice. The chemical messenger involved in the
pathogenesis of PD is Nitric Oxide (NO), which - in combi- enzymes; thus, their anti-oxidant activity is reduced [66].
However, UA administration to the Rotenone-induced Par-
nation with superoxide anion (O2-) - forms highly toxic Per-
kinsonian mice model has significantly enhanced the anti-
oxynitrite (ONOO-), liable for the blockage of the respiratory
oxidant activity of SOD and Catalase due to its anti-
chain. Besides this, NO is also associated with increased
oxidative and free radical scavenging properties. Hence, the
levels of iron, MDA, and aggravated DNA damage account-
UA in our study has shown its potent role by offering protec-
able for neurodegeneration in PD brains [60-62]. Rotenone-
intoxication concomitantly leads to an increased level of NO, tion against oxidative damage and balancing the redox po-
tential of the cell.
which is also seen in our study. Conversely, UA administra-
tion has attenuated the increased level of NO in the Rote- Experimental evidence suggests the alteration in PI3K
none-induced Parkinsonian mice model. and MAPK signaling pathways linked to the pathobiogenesis
The oxidative damage is prevented by an endogenous of PD [8-10, 67]. These two pathways play an important role
antioxidant defence network, which constitutes enzymes in the proliferation, survival and maintenance of the neurons
such as SOD, catalase, and non-enzymatic molecule (such as [7]. Overexpression of α-Synuclein has been linked with
GSH, which helps in scavenging the oxygen free-radicals). reduced phosphorylation of both Akt and ERK, which are
10 CNS & Neurological Disorders - Drug Targets, 2020, Vol. 19, No. 0 Zahra et al.

the key modulators of PI3K and MAPK pathways, respec- Synuclein, when it gets overexpressed, hinders the function
tively [12-14]. Moreover, studies have also suggested the of complex I of mitochondria and induces the production of
reduced phosphorylation of Akt and ERK as a result of in- ROS while blocking the respiratory chain. The overexpres-
creased oxidative stress and neuroinflammation [68]. Re- sion of α-Synuclein in Parkinsonian mice model is similar to
searchers have demonstrated the protective effect of natural- that observed clinically [83]. Besides, the brain is remarka-
ly occurred compounds by regulating the phosphorylation of bly more susceptible to oxidative stress in comparison to
these kinases in different neurotoxin-induced model of PD, peripheral organs due to the abundance of polyunsaturated
including Rotenone [8-10, 69]. Thus, we have tried to study fats and moderately lowered anti-oxidant activity [84].
whether the neuroprotective potential of UA is dependent Moreover, there is a bi-directional relationship between oxi-
upon the activation of these kinases (Akt and ERK) or not. dative stress and overexpression of α-Synuclein and oli-
gomerization, as suggested by different studies [16, 85, 86].
Our results have suggested reduced phosphorylation of both
As such, the increased oxidative stress associated with α-
these kinases upon Rotenone-intoxication linked to the over-
Synuclein accumulation triggers the activation of microglial
expression of α-Synuclein; and increased oxidative stress cells. Studies have suggested that α-Synuclein aggregates
and neuroinflammation. At the same time, UA treatment present in extracellular space, either as a result of neuronal
mitigated the effect of Rotenone and offered neuroprotection apoptosis or cellular release, are engulfed by the activated
by alleviating the symptoms of PD by enhancing the phos- microglial cells [87-89]. Thus, because the microglial activa-
phorylation of Akt and ERK in accordance with other studies tion plays an important role in maintaining the brain homeo-
[8, 10, 45, 47]. stasis, its activation due to α-Synuclein aggregation and in-
Neurotoxicity in PD is also influenced by another major creased production of ROS leads to the activation of the
factor, neuroinflammation [70]. Moreover, numerous studies transcription factor NF-κB and production of proinflammato-
implicate the role of NF-κB activation in PD pathogenesis. ry molecules like TNF-α and IL-1β. Also, the phosphoryla-
NF-κB activation is widely seen in the SN of PD patients, as tion of Akt and ERK is reduced by α-Synuclein overexpres-
well as in animal models [71, 72]. NF-κB, on activation, sion, increased oxidative stress and neuroinflammation,
translocates into the nucleus and stimulates the transcription thereby increasing the neurotoxicity [12-14, 90, 91]. Thus,
of various pro-inflammatory cytokines, such as IL-1β, and this gives us evidence that reducing the expression of α-
TNF-α, by the microglia, which further initiates the neuroin- Synuclein might slow the progression of the disease. Simi-
flammatory cascade [74, 75]. In this study, we estimated the larly, in our study, the overexpression of α-Synuclein was
levels of these inflammatory molecules and tried to explore observed in Rotenone-intoxicated mice, while UA treatment
the effects of UA on the inflammatory pathways. Rotenone significantly lowered the overexpression of α-Synuclein. β-
administration led to the activation and translocation of NF- Synuclein expression, on the other hand, is associated with
κB, which subsequently increased the expression of TNF-α counter-inhibiting the expression and aggregation of α-
and IL-1β and decreased level of IL-10 in SNpc, in agree- Synuclein [92]. Also, β-Synuclein shows neuroprotection by
ment with previous studies [76–78]. Interestingly, UA sup- enhancing the phosphorylation of Akt and reducing the α-
plementation to Rotenone-intoxicated mice has significantly Synuclein overexpression [13]. In this regard, we have also
reduced the activation and translocation of NF-κB, and in- checked the mRNA level of β-Synuclein in the Rotenone-
hibited the expression of TNF-α and IL-1β level. In contrast, intoxicated Parkinsonian mice model. The level of β-
the level of IL-10 is increased on UA administration in Par- Synuclein has decreased for Rotenone-intoxication, while
kinsonian mice. IL-10 being an anti-inflammatory cytokine, UA administration has increased the mRNA level of β-
is found to inhibit the production of TNF-α and microglial Synuclein. Thus, our study suggests that the elevated level of
activation, thereby showing neuroprotective effect by pro- β-Synuclein by UA decreases the overexpression and aggre-
tecting the DA neurons. This result also suggests that UA gation of α-Synuclein.
might counteract with the activation of microglia by increas- Overall, the study suggests that Rotenone-induced PD
ing the level of IL-10, thereby controlling the levels of IL-1β phenotype results from the oxidative stress, neuroinflamma-
and TNF-α in accordance with previous studies [79, 80]. tion, degeneration of DA neurons, reduced phosphorylation
The enzyme TH plays a major role in the synthesis of of Akt and ERK and α-Synuclein overexpression. However,
Dopamine and is the characteristic marker of DA neurons. UA has alleviated these factors through its anti-inflammatory
Thus, the activity of this enzyme is directly associated with and anti-oxidant activities.
the functionality of DA neurons in the SN of the brain [15,
79]. The present study also demonstrates the decreased im- CONCLUSION
munoreactivity of TH due to Rotenone intoxication, while Hence, through our current study, we have tried to justify
upon UA treatment, the reduction of TH neurons was found the neuroprotective role of UA in the Rotenone-induced Par-
to be less in the disease group. kinsonian mouse model. Moreover, our study suggests that
Although there are multiple factors responsible for DA there is a relation between oxidative stress, neuroinflamma-
neuronal loss in the case of PD, the formation of intracyto- tion, an aberration in the phosphorylation of Akt and ERK,
plasmic inclusions, oxidative stress, and neuroinflammation over expression of α-Synuclein, and DA neuronal loss in
are major contributors to the disease pathogenesis [44, 81, Parkinsonian mice model (which can be simultaneously tar-
82]. Overexpression of α-Synuclein is the pathological hall- geted by UA). Hence, UA appears to be a potential drug
mark of PD and is associated with other factors such as candidate for preventing neurodegeneration in PD. However,
apoptosis, oxidative stress, and neuroinflammation, and their further research is needed to explore the mechanistic target
interrelation initiates the pathogenic cascade of PD. α- of UA in PD for its therapeutic intervention.
Neuroprotection of Rotenone-Induced Parkinsonism by Ursolic Acid CNS & Neurological Disorders - Drug Targets, 2020, Vol. 19, No. 0 11

AUTHOR CONTRIBUTIONS http://dx.doi.org/10.1126/science.1067389 PMID: 11823645


[3] Feany MB, Bender WW. A Drosophila model of Parkinson's dis-
WZ: planned and executed the study and wrote the man- ease Nature 2000; 404(6776): 394-8.
uscript. SNR, SSS, ASR, RS and HD: performed the behav- http://dx.doi.org/10.1038/35006074
ioural tests, biochemical tests, and IHC experiments. HB and [4] Masliah, E.; Rockenstein, E.; Veinbergs, I.; Mallory, M.; Hashimo-
to, M.; Takeda, A.; Sagara, Y.; Sisk, A.; Mucke, L. Science (80-.)
CK: helped in designing the experiments, statistical analysis, 2000.
and manuscript writing. RKS: performed the qRT-PCR anal- http://dx.doi.org/10.1038/35006074
ysis. SPS: designed, guided and directed throughout the [5] Bhullar KS, Rupasinghe HP. Polyphenols: multipotent therapeutic
study. agents in neurodegenerative diseases. Oxid Med Cell Longev 2013;
2013(Oct)891748
http://dx.doi.org/10.1155/2013/891748 PMID: 23840922
ETHICS APPROVAL AND CONSENT TO PARTICI- [6] Lee J, Jo DG, Park D, Chung HY, Mattson MP. Adaptive cellular
PATE stress pathways as therapeutic targets of dietary phytochemicals:
focus on the nervous system Pharmacological reviews 2014; 66(3):
All procedures involving animals were performed in ac- 815-68.
cordance with the ethical standards approved by the Animal [7] Dzamko, N.; Zhou, J.; Huang, Y.; Halliday, G. M. Front Mol Neu-
Ethics Committee of Banaras Hindu University, Varanasi, rosci 2014.
India. [8] Cao Q, Qin L, Huang F, et al. Amentoflavone protects dopaminer-
gic neurons in MPTP-induced Parkinson’s disease model mice
through PI3K/Akt and ERK signaling pathways. Toxicol Appl
HUMAN AND ANIMAL RIGHTS Pharmacol 2017; 319: 80-90.
http://dx.doi.org/10.1016/j.taap.2017.01.019 PMID: 28185818
No humans were used in the study that is the basis of this [9] Cheng B, Martinez AA, Morado J, Scofield V, Roberts JL, Maffi
research. All experiments on animals were in agreement with SK. Retinoic acid protects against proteasome inhibition associated
the guidelines for animal experiments established by the Insti- cell death in SH-SY5Y cells via the AKT pathway. Neurochem Int
tutional Animal Ethics Committees (IAECs) of the Laboratory 2013; 62(1): 31-42.
Animal Research (Reg. No. 1802/GO/Re/S/15/CPCSEA) at http://dx.doi.org/10.1016/j.neuint.2012.10.014 PMID: 23142153
[10] Chen WF, Wu L, Du ZR, et al. Neuroprotective properties of icar-
Banaras Hindu University, India. iin in MPTP-induced mouse model of Parkinson’s disease: In-
volvement of PI3K/Akt and MEK/ERK signaling pathways. Phy-
CONSENT FOR PUBLICATION tomedicine 2017; 25: 93-9.
http://dx.doi.org/10.1016/j.phymed.2016.12.017 PMID: 28190476
Not applicable. [11] Rai SN, Dilnashin H, Birla H, et al. The role of PI3K/Akt and ERK
in neurodegenerative disorders. Neurotox Res 2019; 35(3): 775-95.
http://dx.doi.org/10.1007/s12640-019-0003-y PMID: 30707354
AVAILABILITY OF DATA AND MATERIALS [12] Hashimoto M, Takenouchi T, Rockenstein E, Masliah E. α-
Not applicable. synuclein up-regulates expression of caveolin-1 and down-
regulates extracellular signal-regulated kinase activity in B103 neu-
roblastoma cells: role in the pathogenesis of Parkinson’s disease. J
FUNDING Neurochem 2003; 85(6): 1468-79.
http://dx.doi.org/10.1046/j.1471-4159.2003.01791.x
None. PMID: 12787066
[13] Hashimoto M, Bar-On P, Ho G, et al. β-synuclein regulates Akt
CONFLICT OF INTEREST activity in neuronal cells. A possible mechanism for neuroprotec-
tion in Parkinson’s disease. J Biol Chem 2004; 279(22): 23622-9.
The authors declare no conflict of interest, financial or http://dx.doi.org/10.1074/jbc.M313784200 PMID: 15026413
otherwise. [14] Iwata A, Maruyama M, Kanazawa I, Nukina N. α-Synuclein affects
the MAPK pathway and accelerates cell death. J Biol Chem 2001;
276(48): 45320-9.
ACKNOWLEDGEMENTS http://dx.doi.org/10.1074/jbc.M103736200 PMID: 11560921
[15] Ojha S, Javed H, Azimullah S, Abul Khair SB, Haque ME. Neuro-
The authors would also like to acknowledge Ms. Jenan protective potential of ferulic acid in the rotenone model of Parkin-
Husain, Department of Psychology, California State Univer- son’s disease. Drug Des Devel Ther 2015; 9: 5499-510.
sity, Fullerton, USA for assistance in language correction; PMID: 26504373
Mr. Chandra Prakash Patel, Interdisciplinary School of Life [16] Zhang ZN, Zhang JS, Xiang J, et al. Subcutaneous rotenone rat
Sciences (ISLS), BHU for his assistance in the fluorescence model of Parkinson’s disease: Dose exploration study. Brain Res
2017; 1655: 104-13.
microscopy and Mr. Ashok Kumar Yadav, Lab attendant, for http://dx.doi.org/10.1016/j.brainres.2016.11.020 PMID: 27876560
his help in the animal care and other support. The authors [17] Birla H, Rai SN, Singh SS, et al. Tinospora cordifolia suppresses
also would like to acknowledge UGC Dr. D.S. Kothari Post- neuroinflammation in parkinsonian mouse model. Neuromolecular
doctoral scheme for awarding the fellowship to Dr. Sach- Med 2019; 21(1): 42-53.
chida Nand Rai (Ref. No-F.4-2/2006 (BSR)/BL/19-20/0032). http://dx.doi.org/10.1007/s12017-018-08521-7 PMID: 30644041
[18] Fukui M, Choi HJ, Zhu BT. Mechanism for the protective effect of
resveratrol against oxidative stress-induced neuronal death. Free
REFERENCES Radic Biol Med 2010; 49(5): 800-13.
http://dx.doi.org/10.1016/j.freeradbiomed.2010.06.002
[1] Migliore L, Coppedè F. Genetics, environmental factors and the
PMID: 20542495
emerging role of epigenetics in neurodegenerative diseases. Mutat
[19] Keswani C, Bisen K, Singh SP, Singh HB. Traditional knowledge
Res 2009; 667(1-2): 82-97.
and medicinal plants of India in intellectual property landscape.
http://dx.doi.org/10.1016/j.mrfmmm.2008.10.011 PMID: 19026668
Medicinal Plants-International Journal of Phytomedicines and Re-
[2] Auluck PK, Chan HY, Trojanowski JQ, Lee VM, Bonini NM.
lated Industries 2017; 9(1): 1-1.
Chaperone suppression of α-synuclein toxicity in a Drosophila
http://dx.doi.org/10.5958/0975-6892.2017.00001.6
model for Parkinson’s disease. Science 2002; 295(5556): 865-8.
12 CNS & Neurological Disorders - Drug Targets, 2020, Vol. 19, No. 0 Zahra et al.

[20] Rai SN, Birla H, Zahra W, Singh SS, Singh SP. Immunomodula- [36] Ohkawa H, Ohishi N, Yagi K. Assay for lipid peroxides in animal
tion of Parkinson’s disease using Mucuna pruriens (Mp). J Chem tissues by thiobarbituric acid reaction. Anal Biochem 1979; 95(2):
Neuroanat 2017; 85: 27-35. 351-8.
http://dx.doi.org/10.1016/j.jchemneu.2017.06.005 http://dx.doi.org/10.1016/0003-2697(79)90738-3 PMID: 36810
PMID: 28642128 [37] Corporation P, Miranda KM, Espey MG, Wink D. Griess Reagent
[21] Rai SN, Birla H, Singh SS, et al. Mucuna pruriens Protects against System Promega Tech Bull 2009.
MPTP Intoxicated Neuroinflammation in Parkinson’s Disease [38] Moron MS, Depierre JW, Mannervik B. Levels of glutathione,
through NF-κB/pAKT Signaling Pathways. Front Aging Neurosci glutathione reductase and glutathione S-transferase activities in rat
2017; 9: 421. lung and liver Biochimica et biophysica acta (BBA)-general sub-
http://dx.doi.org/10.3389/fnagi.2017.00421 PMID: 29311905 jects 1979; 582(1): 67-78.
[22] Birla H, Keswani C, Rai SN, et al. Neuroprotective effects of [39] Misra HP, Fridovich I. The role of superoxide anion in the autoxi-
Withania somnifera in BPA induced-cognitive dysfunction and ox- dation of epinephrine and a simple assay for superoxide dismutase.
idative stress in mice. Behav Brain Funct 2019; 15(1): 9. J Biol Chem 1972; 247(10): 3170-5.
http://dx.doi.org/10.1186/s12993-019-0160-4 PMID: 31064381 PMID: 4623845
[23] Singh SS, Rai SN, Birla H, et al. Neuroprotective Effect of Chloro- [40] Gorbatyuk OS, Li S, Sullivan LF, et al. The phosphorylation state
genic Acid on Mitochondrial Dysfunction-Mediated Apoptotic of Ser-129 in human α-synuclein determines neurodegeneration in
Death of DA Neurons in a Parkinsonian Mouse Model Oxidative a rat model of Parkinson disease. Proc Natl Acad Sci USA 2008;
Medicine and Cellular Longevity 2020. 105(2): 763-8.
http://dx.doi.org/10.1155/2020/6571484 http://dx.doi.org/10.1073/pnas.0711053105 PMID: 18178617
[24] Liu J. Pharmacology of oleanolic acid and ursolic acid. J Eth- [41] Peinnequin A, Mouret C, Birot O, et al. Rat pro-inflammatory
nopharmacol 1995; 49(2): 57-68. cytokine and cytokine related mRNA quantification by real-time
http://dx.doi.org/10.1016/0378-8741(95)90032-2 PMID: 8847885 polymerase chain reaction using SYBR green. BMC Immunol
[25] Neto CC, Amoroso JW, Liberty AM. Anticancer activities of cran- 2004; 5(1): 3.
berry phytochemicals: an update. Mol Nutr Food Res 2008; http://dx.doi.org/10.1186/1471-2172-5-3 PMID: 15040812
52(S1)(Suppl. 1): S18-27. [42] Gupta SP, Patel S, Yadav S, Singh AK, Singh S, Singh MP. In-
http://dx.doi.org/10.1002/mnfr.200700433 PMID: 18504707 volvement of nitric oxide in maneb- and paraquat-induced Parkin-
[26] Yamaguchi H, Noshita T, Kidachi Y, et al. Isolation of ursolic acid son’s disease phenotype in mouse: is there any link with lipid pe-
from apple peels and its specific efficacy as a potent antitumor roxidation? Neurochem Res 2010; 35(8): 1206-13.
agent. J Health Sci 2008; 54(6): 654-60. http://dx.doi.org/10.1007/s11064-010-0176-5 PMID: 20455021
http://dx.doi.org/10.1248/jhs.54.654 [43] Normando EM, Davis BM, De Groef L, et al. The retina as an early
[27] Rai SN, Yadav SK, Singh D, Singh SP. Ursolic acid attenuates biomarker of neurodegeneration in a rotenone-induced model of
oxidative stress in nigrostriatal tissue and improves neurobehavior- Parkinson’s disease: evidence for a neuroprotective effect of rosig-
al activity in MPTP-induced Parkinsonian mouse model. J Chem litazone in the eye and brain. Acta Neuropathol Commun 2016;
Neuroanat 2016; 71: 41-9. 4(1): 86.
http://dx.doi.org/10.1016/j.jchemneu.2015.12.002 http://dx.doi.org/10.1186/s40478-016-0346-z PMID: 27535749
PMID: 26686287 [44] Anusha C, Sumathi T, Joseph LD. Protective role of apigenin on
[28] Habtemariam S. Antioxidant and anti-inflammatory mechanisms of rotenone induced rat model of Parkinson’s disease: Suppression of
neuroprotection by ursolic acid: addressing brain injury, cerebral neuroinflammation and oxidative stress mediated apoptosis. Chem
ischemia, cognition deficit, anxiety, and depression. Oxid Med Cell Biol Interact 2017; 269: 67-79.
Longev 2019; 20198512048 http://dx.doi.org/10.1016/j.cbi.2017.03.016 PMID: 28389404
http://dx.doi.org/10.1155/2019/8512048 PMID: 31223427 [45] Wang H, Dong X, Liu Z, et al. Resveratrol suppresses rotenone‐
[29] Zhu Z, Du S, Ding F, Guo S, Ying G, Yan Z. Ursolic acid attenu- induced neurotoxicity through activation of SIRT1/Akt1 signaling
ates temozolomide resistance in glioblastoma cells by downregulat- pathway. Anat Rec (Hoboken) 2018; 301(6): 1115-25.
ing O(6)-methylguanine-DNA methyltransferase (MGMT) expres- http://dx.doi.org/10.1002/ar.23781 PMID: 29350822
sion. Am J Transl Res 2016; 8(7): 3299-308. [46] Xu Y, Liu C, Chen S, et al. Activation of AMPK and inactivation
PMID: 27508051 of Akt result in suppression of mTOR-mediated S6K1 and 4E-BP1
[30] Lu J, Wu DM, Zheng YL, et al. Ursolic acid attenuates D- pathways leading to neuronal cell death in in vitro models of Par-
galactose-induced inflammatory response in mouse prefrontal cor- kinson’s disease. Cell Signal 2014; 26(8): 1680-9.
tex through inhibiting AGEs/RAGE/NF-κB pathway activation. http://dx.doi.org/10.1016/j.cellsig.2014.04.009 PMID: 24726895
Cereb Cortex 2010; 20(11): 2540-8. [47] Jung SY, Lee KW, Choi SM, Yang EJ. Bee venom protects against
http://dx.doi.org/10.1093/cercor/bhq002 PMID: 20133359 rotenone-induced cell death in NSC34 motor neuron cells toxins
[31] Wang YJ, Lu J, Wu DM, et al. Ursolic acid attenuates lipopolysac- 2015; 7(9): 3715-26.
charide-induced cognitive deficits in mouse brain through sup- [48] Song JX, Choi MY, Wong KC, et al. Baicalein antagonizes rote-
pressing p38/NF-κB mediated inflammatory pathways. Neurobiol none-induced apoptosis in dopaminergic SH-SY5Y cells related to
Learn Mem 2011; 96(2): 156-65. Parkinsonism. Chin Med 2012; 7(1): 1-9.
http://dx.doi.org/10.1016/j.nlm.2011.03.010 PMID: 21496491 http://dx.doi.org/10.1186/1749-8546-7-1 PMID: 22264378
[32] Manna S, Bhattacharyya D, Mandal TK, Dey S. Neuropharmaco- [49] Luo J, Hu YL, Wang H. Ursolic acid inhibits breast cancer growth
logical effects of deltamethrin in rats. J Vet Sci 2006; 7(2): 133-6. by inhibiting proliferation, inducing autophagy and apoptosis, and
http://dx.doi.org/10.4142/jvs.2006.7.2.133 PMID: 16645337 suppressing inflammatory responses via the PI3K/AKT and NF-κB
[33] Pisa M. Regional specialization of motor functions in the rat stria- signaling pathways in vitro. Exp Ther Med 2017; 14(4): 3623-31.
tum: implications for the treatment of parkinsonism. Prog Neuro- http://dx.doi.org/10.3892/etm.2017.4965 PMID: 29042957
psychopharmacol Biol Psychiatry 1988; 12(2-3): 217-24. [50] Wang X, Zhang F, Yang L, et al. Ursolic acid inhibits proliferation
http://dx.doi.org/10.1016/0278-5846(88)90038-3 PMID: 3290995 and induces apoptosis of cancer cells in vitro and in vivo. J Biomed
[34] Mohanasundari M, Srinivasan MS, Sethupathy S, Sabesan M. Biotechnol 2011; 2011419343
Enhanced neuroprotective effect by combination of bromocriptine http://dx.doi.org/10.1155/2011/419343 PMID: 21716649
and Hypericum perforatum extract against MPTP-induced neuro- [51] Wang Y, He Z, Deng S. Ursolic acid reduces the metalloprote-
toxicity in mice. J Neurol Sci 2006; 249(2): 140-4. ase/anti-metalloprotease imbalance in cerebral ischemia and reper-
http://dx.doi.org/10.1016/j.jns.2006.06.018 PMID: 16876826 fusion injury. Drug Des Devel Ther 2016; 10: 1663-74.
[35] Kumar A, Ahmad I, Shukla S, et al. Effect of zinc and paraquat co- http://dx.doi.org/10.2147/DDDT.S103829 PMID: 27274199
exposure on neurodegeneration: Modulation of oxidative stress and [52] Machado DG, Neis VB, Balen GO, et al. Antidepressant-like effect
expression of metallothioneins, toxicant responsive and transporter of ursolic acid isolated from Rosmarinus officinalis L. in mice: ev-
genes in rats. Free Radic Res 2010; 44(8): 950-65. idence for the involvement of the dopaminergic system. Pharmacol
http://dx.doi.org/10.3109/10715762.2010.492832 PMID: 20553223 Biochem Behav 2012; 103(2): 204-11.
http://dx.doi.org/10.1016/j.pbb.2012.08.016 PMID: 22940588
Neuroprotection of Rotenone-Induced Parkinsonism by Ursolic Acid CNS & Neurological Disorders - Drug Targets, 2020, Vol. 19, No. 0 13

[53] Li L, Zhang X, Cui L, et al. Ursolic acid promotes the neuroprotec- http://dx.doi.org/10.1007/s11481-018-9824-3 PMID: 30478761
tion by activating Nrf2 pathway after cerebral ischemia in mice. [69] Greene LA, Levy O, Malagelada C. Akt as a victim, villain and
Brain Res 2013; 1497: 32-9. potential hero in Parkinson’s disease pathophysiology and treat-
http://dx.doi.org/10.1016/j.brainres.2012.12.032 PMID: 23276496 ment. Cell Mol Neurobiol 2011; 31(7): 969-78.
[54] Wilkinson K, Boyd JD, Glicksman M, Moore KJ, El Khoury J. A http://dx.doi.org/10.1007/s10571-011-9671-8 PMID: 21547489
high content drug screen identifies ursolic acid as an inhibitor of [70] Niranjan R. The role of inflammatory and oxidative stress mecha-
amyloid β protein interactions with its receptor CD36. J Biol Chem nisms in the pathogenesis of Parkinson’s disease: focus on astro-
2011; 286(40): 34914-22. cytes. Mol Neurobiol 2014; 49(1): 28-38.
http://dx.doi.org/10.1074/jbc.M111.232116 PMID: 21835916 http://dx.doi.org/10.1007/s12035-013-8483-x PMID: 23783559
[55] Fornai F, Lenzi P, Ferrucci M, et al. Occurrence of neuronal inclu- [71] He Q, Yu W, Wu J, et al. Intranasal LPS-mediated Parkinson’s
sions combined with increased nigral expression of α-synuclein model challenges the pathogenesis of nasal cavity and environmen-
within dopaminergic neurons following treatment with ampheta- tal toxins. PLoS One 2013; 8(11): e78418.
mine derivatives in mice. Brain Res Bull 2005; 65(5): 405-13. http://dx.doi.org/10.1371/journal.pone.0078418 PMID: 24250796
http://dx.doi.org/10.1016/j.brainresbull.2005.02.022 [72] Zhang W, Yan ZF, Gao JH, et al. Role and mechanism of microgli-
PMID: 15833595 al activation in iron-induced selective and progressive dopaminer-
[56] Kowall NW, Hantraye P, Brouillet E, Beal MF, McKee AC, Fer- gic neurodegeneration. Mol Neurobiol 2014; 49(3): 1153-65.
rante RJ. MPTP induces alpha-synuclein aggregation in the sub- http://dx.doi.org/10.1007/s12035-013-8586-4 PMID: 24277523
stantia nigra of baboons. Neuroreport 2000; 11(1): 211-3. [73] Singh SS, Rai SN, Birla H, Zahra W, Rathore AS, Singh SP. NF-
http://dx.doi.org/10.1097/00001756-200001170-00041 κB-mediated neuroinflammation in Parkinson’s disease and poten-
PMID: 10683860 tial therapeutic effect of polyphenols. Neurotox Res 2019; •••: 1-7.
[57] Di Napoli M, Shah IM, Stewart DA. Molecular pathways and ge- PMID: 31823227
netic aspects of Parkinson’s disease: from bench to bedside. Expert [74] Yao K, Zhang L, Zhang Y, Ye P, Zhu N. The flavonoid, fisetin,
Rev Neurother 2007; 7(12): 1693-729. inhibits UV radiation-induced oxidative stress and the activation of
http://dx.doi.org/10.1586/14737175.7.12.1693 PMID: 18052765 NF-kappaB and MAPK signaling in human lens epithelial cells.
[58] Singh R, Kaushik S, Wang Y, et al. Autophagy regulates lipid Mol Vis 2008; 14: 1865-71.
metabolism. Nature 2009; 458(7242): 1131-5. PMID: 18949064
http://dx.doi.org/10.1038/nature07976 PMID: 19339967 [75] Al-Rasheed NM, Al-Rasheed NM, Bassiouni YA, et al. Vitamin D
[59] Shichiri M. The role of lipid peroxidation in neurological disorders. attenuates pro-inflammatory TNF-α cytokine expression by inhibit-
J Clin Biochem Nutr 2014; 54(3): 151-60. ing NF-кB/p65 signaling in hypertrophied rat hearts. J Physiol Bio-
http://dx.doi.org/10.3164/jcbn.14-10 PMID: 24895477 chem 2015; 71(2): 289-99.
[60] Cleeter MW, Cooper JM, Darley-Usmar VM, Moncada S, Schapira http://dx.doi.org/10.1007/s13105-015-0412-1 PMID: 25929726
AH. Reversible inhibition of cytochrome c oxidase, the terminal [76] Litteljohn D, Mangano E, Clarke M, Bobyn J, Moloney K, Hayley
enzyme of the mitochondrial respiratory chain, by nitric oxide. Im- S. Inflammatory mechanisms of neurodegeneration in toxin-based
plications for neurodegenerative diseases. FEBS Lett 1994; 345(1): models of Parkinson’s disease. Parkinsons Dis 2011; •••: 2011.
50-4. http://dx.doi.org/10.4061/2011/713517 PMID: 21234362
http://dx.doi.org/10.1016/0014-5793(94)00424-2 PMID: 8194600 [77] Sherer TB, Betarbet R, Kim JH, Greenamyre JT. Selective micro-
[61] Friend DM, Fricks-Gleason AN, Keefe KA. Is there a role for nitric glial activation in the rat rotenone model of Parkinson’s disease.
oxide in methamphetamine-induced dopamine terminal degenera- Neurosci Lett 2003; 341(2): 87-90.
tion? Neurotox Res 2014; 25(2): 153-60. http://dx.doi.org/10.1016/S0304-3940(03)00172-1
http://dx.doi.org/10.1007/s12640-013-9415-2 PMID: 23918001 PMID: 12686372
[62] Tapias V, Hu X, Luk KC, Sanders LH, Lee VM, Greenamyre JT. [78] Thakur P, Nehru B. Anti-inflammatory properties rather than anti-
Synthetic alpha-synuclein fibrils cause mitochondrial impairment oxidant capability is the major mechanism of neuroprotection by
and selective dopamine neurodegeneration in part via iNOS- sodium salicylate in a chronic rotenone model of Parkinson’s dis-
mediated nitric oxide production. Cell Mol Life Sci 2017; 74(15): ease. Neuroscience 2013; 231: 420-31.
2851-74. http://dx.doi.org/10.1016/j.neuroscience.2012.11.006
http://dx.doi.org/10.1007/s00018-017-2541-x PMID: 28534083 PMID: 23159314
[63] Sutachan JJ, Casas Z, Albarracin SL, et al. Cellular and molecular [79] Singh SS, Rai SN, Birla H, et al. Effect of chlorogenic acid sup-
mechanisms of antioxidants in Parkinson’s disease. Nutr Neurosci plementation in MPTP-intoxicated mouse. Front Pharmacol 2018;
2012; 15(3): 120-6. 9: 757.
http://dx.doi.org/10.1179/1476830511Y.0000000033 http://dx.doi.org/10.3389/fphar.2018.00757 PMID: 30127737
PMID: 22732354 [80] Yan J, Fu Q, Cheng L, et al. Inflammatory response in Parkinson’s
[64] Anderson G, Maes M. Neurodegeneration in Parkinson’s disease: disease (Review). Mol Med Rep 2014; 10(5): 2223-33.
interactions of oxidative stress, tryptophan catabolites and depres- http://dx.doi.org/10.3892/mmr.2014.2563 PMID: 25215472
sion with mitochondria and sirtuins. Mol Neurobiol 2014; 49(2): [81] Johnson ME, Bobrovskaya L. An update on the rotenone models of
771-83. Parkinson’s disease: their ability to reproduce the features of clini-
http://dx.doi.org/10.1007/s12035-013-8554-z PMID: 24085563 cal disease and model gene-environment interactions. Neurotoxi-
[65] Celardo I, Martins LM, Gandhi S. Unravelling mitochondrial cology 2015; 46: 101-16.
pathways to Parkinson’s disease. Br J Pharmacol 2014; 171(8): http://dx.doi.org/10.1016/j.neuro.2014.12.002 PMID: 25514659
1943-57. [82] Jiang XW, Qiao L, Feng XX, et al. Rotenone induces nephrotoxici-
http://dx.doi.org/10.1111/bph.12433 PMID: 24117181 ty in rats: oxidative damage and apoptosis. Toxicol Mech Methods
[66] Pigeolet E, Corbisier P, Houbion A, et al. Glutathione peroxidase, 2017; 27(7): 528-36.
superoxide dismutase, and catalase inactivation by peroxides and http://dx.doi.org/10.1080/15376516.2017.1333553
oxygen derived free radicals. Mech Ageing Dev 1990; 51(3): 283- PMID: 28532211
97. [83] Surmeier DJ. α-Synuclein at the synaptic gate. Neuron 2010; 65(1):
http://dx.doi.org/10.1016/0047-6374(90)90078-T PMID: 2308398 3-4.
[67] Yuan Y, Sun J, Zhao M, et al. Overexpression of α-synuclein http://dx.doi.org/10.1016/j.neuron.2009.12.030 PMID: 20152107
down-regulates BDNF expression. Cell Mol Neurobiol 2010; [84] Sanders LH, Timothy Greenamyre J. Oxidative damage to macro-
30(6): 939-46. molecules in human Parkinson disease and the rotenone model.
http://dx.doi.org/10.1007/s10571-010-9523-y PMID: 20405200 Free Radic Biol Med 2013; 62: 111-20.
[68] Muhammad T, Ali T, Ikram M, Khan A, Alam SI, Kim MO. Mela- http://dx.doi.org/10.1016/j.freeradbiomed.2013.01.003
tonin rescue oxidative stress-mediated neuroinflammation/ PMID: 23328732
neurodegeneration and memory impairment in scopolamine- [85] Michel HE, Tadros MG, Esmat A, Khalifa AE, Abdel-Tawab AM.
induced amnesia mice model. J Neuroimmune Pharmacol 2019; Tetramethylpyrazine ameliorates rotenone-induced Parkinson’s
14(2): 278-94.
14 CNS & Neurological Disorders - Drug Targets, 2020, Vol. 19, No. 0 Zahra et al.

disease in rats: involvement of its anti-inflammatory and anti- http://dx.doi.org/10.1016/j.neurobiolaging.2015.03.015


apoptotic actions. Mol Neurobiol 2017; 54(7): 4866-78. PMID: 25983062
http://dx.doi.org/10.1007/s12035-016-0028-7 PMID: 27514753 [90] Hoenen C, Gustin A, Birck C, et al. Alpha-synuclein proteins pro-
[86] Zhang QS, Heng Y, Yuan YH, Chen NH. Pathological α-synuclein mote pro-inflammatory cascades in microglia: stronger effects of
exacerbates the progression of Parkinson’s disease through micro- the A53T mutant. PLoS One 2016; 11(9)e0162717.
glial activation. Toxicol Lett 2017; 265: 30-7. http://dx.doi.org/10.1371/journal.pone.0162717
http://dx.doi.org/10.1016/j.toxlet.2016.11.002 PMID: 27865851 PMID: 27622765
[87] Goedert M, Spillantini MG, Del Tredici K, Braak H. 100 years of [91] Zhang W, Wang T, Pei Z, et al. Aggregated α-synuclein activates
Lewy pathology. Nat Rev Neurol 2013; 9(1): 13-24. microglia: a process leading to disease progression in Parkinson’s
http://dx.doi.org/10.1038/nrneurol.2012.242 PMID: 23183883 disease. FASEB J 2005; 19(6): 533-42.
[88] Hoffmann A, Ettle B, Bruno A, et al. Alpha-synuclein activates http://dx.doi.org/10.1096/fj.04-2751com PMID: 15791003
BV2 microglia dependent on its aggregation state. Biochem Bio- [92] Wright JA, McHugh PC, Pan S, Cunningham A, Brown DR. Coun-
phys Res Commun 2016; 479(4): 881-6. ter-regulation of alpha- and beta-synuclein expression at the tran-
http://dx.doi.org/10.1016/j.bbrc.2016.09.109 PMID: 27666480 scriptional level. Mol Cell Neurosci 2013; 57: 33-41.
[89] Jiang T, Hoekstra J, Heng X, et al. P2X7 receptor is critical in α- http://dx.doi.org/10.1016/j.mcn.2013.09.002 PMID: 24080388
synuclein--mediated microglial NADPH oxidase activation. Neu-
robiol Aging 2015; 36(7): 2304-18.

DISCLAIMER: The above article has been published in Epub (ahead of print) on the basis of the materials provided by the author. The Edito-
rial Department reserves the right to make minor modifications for further improvement of the manuscript.

You might also like