You are on page 1of 11

Phytomedicine Plus 2 (2022) 100343

Contents lists available at ScienceDirect

Phytomedicine Plus
journal homepage: www.sciencedirect.com/journal/phytomedicine-plus

Anti-Parkinsonian effect of Mucuna pruriens and Ursolic acid on GSK3β/


Calcium signaling in neuroprotection against
Rotenone-induced Parkinsonism
Walia Zahra , Hareram Birla , Saumitra Sen Singh , Aaina Singh Rathore , Hagera Dilnashin ,
Richa Singh , Priyanka Kumari Keshri, Shekhar Singh , Surya Pratap Singh *
Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, (U.P.), 221005 India

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

Keywords: Background: Exposure of environmental toxin is linked to the onset of Parkinson’s disease (PD), especially in
Parkinson’ disease majority of sporadic cases. One of such toxins, namely Rotenone has been widely investigated to produce the
Ca2+ level Parkinsonian model in laboratories. The toxin causes the death of the dopaminergic neurons, aggregation of
GSK-3β
α-Synuclein, oxidative stress, neuroinflammation and initiate other pathways leading to PD pathogenesis.
Rotenone
Mucuna pruriens
Glycogen synthase kinase-3 (GSK-3) is a pleiotropic serine/threonine protein kinase found in eukaryotes. One of
Ursolic acid the two isoforms of the enzyme; GSK-3β plays an important role in regulating the pathogenesis of neurode­
generative diseases, including PD. Whereas, Calcium (Ca2+) is found to regulate important cellular activities in
all eukaryotic cells. However, an increased Ca2+ concentration is seen during ageing and the progression of
neurodegenerative diseases. Thus, maintaining the Ca2+ homoeostasis is very crucial for proper cell function.
Methodology: Hence, behavioural tests were performed to assess the motor abnormalities; immunohistochemical
and western blot analyses to identify the alteration in the protein expression; and mitochondrial complexes and
antioxidants assay to detect the extent of mitochondrial dysfunction in Rotenone-induced Parkinsonian mouse
model. Alternatively, the effect of Mucuna pruriens (Mp) and Ursolic acid (UA) in the PD mouse model was also
observed.
Results: Increased Ca2+level, and activation of GSK-3β was observed in the Rotenone-intoxicated mouse model of
PD. The extract of Mp and UA, on the other hand shows neuroprotection through their anti-oxidative and anti-
inflammatory properties. However, their role in maintaining the Ca2+ level and GSK-3β signaling is not yet
observed in PD. So, our study deals with the downregulated activity of GSK-3β and Ca2+ level upon the
administration of Mp extract and UA, thereby providing neuroprotection to the PD mouse model.
Conclusion: Thus, this study deals to explore further the neuroprotective activity of Mp and UA through GSK-3β/
Calcium signaling by ameliorating mitochondrial dysfunction mediated apoptosis and inhibiting the over­
expression of α-Synuclein in Rotenone-induced PD mouse model.

selective and progressive loss of the dopaminergic neurons and forma­


tion of inclusions called Lewy bodies (LBs) in substantia nigra pars
Introduction compacta of midbrain (Ruiz et al., 2011). Dopaminergic neuronal loss is
often associated with Oxidative stress, overexpression and aggregation
The neurodegenerative disorder Parkinson’s disease (PD), affects of α-Synuclein, Neuroinflammation and Mitochondrial dysfunction; the
mostly elderlies and is associated with ageing. It is characterized clini­ key players for neurodegeneration in PD that induces dopaminergic
cally by bradykinesia, rigidity, tremor and stooped posture (Dexter and neuronal loss and reduction in the level of dopamine (DA) ( Huang et al.,
Jenner, 2013; Kalia and Lang, 2015). Non-motor symptoms, such as 2020; Martinez et al., 2017; Moon and Paek, 2015; Morris and Berk,
emotional disturbances, stress, olfactory impairments, sleep disorders, 2015; Hu et al., 2019; Tolleson and Fang 2013).
and autonomic dysfunctions, are also related with the pathogenesis of GSK-3β is one of the isoforms of GSK-3, a serine/threonine kinase
PD (Kalia and Lang, 2015). The pathology of the disease involves the

* Corresponding author.
E-mail address: suryasinghbhu16@gmail.com (S.P. Singh).

https://doi.org/10.1016/j.phyplu.2022.100343
Received 15 February 2022; Received in revised form 30 August 2022; Accepted 6 September 2022
Available online 7 September 2022
2667-0313/© 2022 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-
nc-nd/4.0/).
W. Zahra et al. Phytomedicine Plus 2 (2022) 100343

inflammatory mediators, such as microglia and inflammatory molecules


List of abbreviations in SNpc of brain (Rai et al., 2017; Zahra et al., 2020). Alteration in the
Ca2+ level also plays major role in the PD pathogenesis (Surmeier et al.,
Ca2+ Calcium; 2012). GSK-3β activation and increased Ca2+level is also interlinked
GSK-3β Glycogen synthase kinase-3β; with each other which further potentiates the formation of the α-Synu­
mPTP Mitochondrial permeability transition pore; clein aggregates (Li et al., 2014). Since GSK-3β signaling pathway plays
Mp Mucuna pruriens; prominent role in the PD pathogenesis, serving as a central point for
PD Parkinson’s disease; mediating and accelerating different pathological events; can be tar­
ROS Reactive oxygen species; geted for therapeutic management of PD pathogenesis (Fig. 1).
SEM standard error of mean; Cure for PD is still unknown and treatment with L-DOPA, DA receptor
TH Tyrosine hydroxylase; agonists and deep brain stimulation (DBS) surgery, only manages the
UA Ursolic acid clinical symptoms, and rarely alleviate loss of the dopaminergic neurons
(Iarkov et al., 2020; Rizek et al., 2016). Thus, identifying new neuro­
protectants that reduce neuronal loss is of great significance for the
treatment of PD (Birla et al., 2019; Singh et al., 2018; Rai et al., 2020;
found in mammals. The regulatory role of GSK-3β in stress-related Rai et al., 2021a; Rai et al., 2021b). The neuroprotective property of
mitochondrial dysfunction is seen in the neuronal cells (Li et al., Mucuna pruriens (Mp) in PD has been widely studied (Cilia et al., 2017;
2014). Other pathogenic markers of PD such as oxidative stress, in­ Rai et al., 2017, 2020; Yadav et al., 2013; Yadav et al., 2014, 2016). But,
flammatory response, apoptosis and aggregation of α-Synuclein, are also the mechanism through which it mediates its anti-Parkinsonian effect is
facilitated by the activation of GSK-3β (Li et al., 2014; Wang et al., yet to be explored. The anti-inflammatory and anti-oxidative studies of
2007). Whereas, GSK-3β inhibition, suppresses different pathogenic Mp has been seen in the animal models of PD (Uchegbu et al., 2016; Rai
events in PD, thereby promoting the survival of the dopaminergic neu­ et al., 2017; Yadav et al., 2013). Moreover, the Reverse-Phase High
rons (Singh et al., 2020; Wang et al., 2007; Yuskaitis and Jope, 2009). performance liquid chromatography (RP-HPLC) data have shown the
Administration of the neurotoxin Rotenone, causes the death of the presence of L-DOPA and Ursolic acid (UA) in the aqueous extract of Mp,
dopaminergic neurons from SNpc, in animal models of PD (Cannon in the study conducted in our lab (Yadav et al., 2013; Rai et al., 2020).
et al., 2009; Zahra et al., 2020). Activation of GSK-3β, α-Synuclein UA is natural triterpenoid carboxylic acid found in variety of plants,
overexpression and increased Calcium (Ca2+) level in the dopaminergic studied for its neuroprotective activity against PD and is also capable of
neurons is mediated by Rotenone-neurotoxicity (Rcom-H’cheo-­ crossing the blood-brain barrier (Habtemariam, 2019; Zahra et al., 2020;
Gauthier et al., 2016; Zahra et al., 2020). Also, being the inhibitor of Rai et al., 2019). Although, both Mp and UA have been studied for their
Complex I of the mitochondrial Electron Transport System (ETS), it anti-oxidative and anti-inflammatory properties in PD, but their role in
persuades mitochondrial dysfunction, in PD pathogenesis. Complex I maintaining the Ca2+ level, inhibition of mitochondrial dysfunction and
impairment is also related to dopaminergic neuronal loss in idiopathic α-Synuclein overexpression, and rescuing dopaminergic neurons from
PD patients (Li et al., 2014). Other pathological contributor for the apoptosis; by inhibiting the activation of GSK-3β has not yet been
neurodegeneration in PD is the increased inflammatory response in studied. Thus, this study aims to explore the neuroprotective potential of
dopaminergic neurons (Monahan et al., 2008). Post-mortem studies, Mp and UA by investigating their effect on GSK-3β/ Ca2+ signaling in the
along with the studies including animal models of PD (also by Rotenone-intoxicated mouse model of PD.
Rotenone-intoxication), has revealed the presence of activated

Fig. 1. Rotenone-induced GSK‑3β signaling cascade in PD pathogenesis.

2
W. Zahra et al. Phytomedicine Plus 2 (2022) 100343

Chemical and reagents Behavioural test

Reagents and Antibodies. Paraformaldehyde, Glycylglycine buffer, Behavioural tests were done at day time (between 10AM- 12 noon) to
trichloroacetic acid, sodium fluoride, and Tris buffer were purchased assess motor deficits in the Rotenone-intoxicated mice; and how Mp and
from LobaChemie, India. Normal goat serum (NGS), DABCO, 5,5-dithio­ UA ameliorated this neurotoxic effect in parkinsonian mouse model. The
bis 2-nitrobenzoic acid (DTNB), Rotenone and Ursolic acid were bought tests were performed for five consecutive days after the completion of
from Sigma-Aldrich (St. Louis, MO, United States). Bovine serum al­ the dosing and include the rotarod test, pole test, catalepsy test, hanging
bumin (BSA), Mannitol, oxidized cytochrome c, sodium dihydrogen test and traction test.
phosphatedigitonin, disodium hydrogen phosphate, potassium chloride,
reduced nicotinamide adenine dinucleotide phosphate (NADPH), and Catalepsy test
ammonium chloride were purchased from Sisco Research Laboratories
(SRL, Mumbai, India). Sodium carbonate and Sucrose were obtained Muscle stiffness in mice is observed by performing the Catalepsy test.
from Merck (Darmstadt, Germany). EGTA and sodium dodecyl sulphate In this test, the diseased mice are unable to change its posture, in which
(SDS) were purchased from HiMedia (Mumbai, India). Primary anti­ they are imposed. The animals were made to stand on a wooden plat­
bodies for Akt (ab8805), p-Akt (ab81283), caspase-3 (ab214430), Iba-1 form with their hind limb and their forepaws were supported on the
(ab153696), α-Synuclein (ab191692), Bcl-2 (GR99542–4) and calcium ground (Singh et al., 2020). Cataleptic intensity was measured as the
detection kit (ab102505) were acquired from Abcam Life Science, Bio­ mouse moved its hind limbs from a wooden platform. The test was
genuix Medsystems Pvt. Ltd. (New Delhi, India), and primary antibodies performed for 180 s for each mouse of each group.
for GSK3β (sc-81,462), p-GSK3β (sc-373,800), TH (sc-25,269), β-actin
(sc-47,778), and Bax (sc-6236) were purchased from Santa Cruz Pole test
Biotechnology (Santa Cruz, CA, United States).
Pole test was performed to assess bradykinesia in the Parkinsonian
mouse model (Hu et al., 2018). The animals were placed on the top of
Experimental animals pole having a rough surface (52 cm in height and 10 mm in diameter).
The time taken by the mouse to step down the pole (T-turn) was further
Male Swiss albino mice (adult; 25 ± 5 g) were used in the experi­ recorded.
mental groups and were obtained from the Institute of Medical Sciences’
animal facility, Banaras Hindu University, Varanasi (India). The exper­ Traction test
imental animals were kept under proper laboratory condition; in clean
polypropylene cages with constant light-dark cycles of 12 h; and pro­ This test was done to assess the muscle strength and equilibrium in
vided with standard diet pellet and water ad libitum until the dosing was the experimental animals (Hu et al., 2018). Forelimbs of the mouse were
completed. Experiment was conducted according to the Animal Ethics supported on a horizontal bar in this test and traction score was recorded
Committee’s guidelines of Banaras Hindu University, Varanasi, India. on the basis of their hind limb placement. The lowest score was given to
mice which were severely impaired. While the score was increased as the
Plant extract preparation mouse placed one or both hind limbs on the bar.

Seed powder of Mp was procured from the Ayurveda Pharmacy, Rotarod test
Institute of Medical Sciences, Banaras Hindu University, Varanasi, India.
The extract was later prepared by following the method of Uhegbu et al. This test was also done to assess the equilibrium in the mouse model
(2005) and Rai et al., (2017), by using distilled water as solvent. Briefly, of PD. Mice from each group were placed on a rotating beam (speed 5
200 mL of distilled water (sterile) was used for soaking 20 g of Mp seed rpm). The experiment was performed for a maximum duration of 5 min.
powder. It was left overnight after 6 min of stirring, for proper mix-up. The time for which the animal stayed on the rotarod was noted down
The solution was further filtered using Whatman No. A-1, the other day. (Manna et al., 2006).
The filtered solution was then dried in rotary vacuum evaporator below
40◦ C, under reduced pressure. The extract thus obtained was adminis­ Hanging test
tered to the respective experimental group.
The test was performed by using a horizontal grid on which the
animals were placed, and the grid was turned upside down. Hanging
Animal dosing time was recoded and the experiment was repeated thrice (Rai et al.,
2016).
Mice were randomly divided into four groups, viz.; Control, Rote­
none, Rotenone+Mp, and Rotenone+UA (n = 10/group). Dosing to the Mitochondrial parameter analysis
animals were done in accordance with our previous studies with some
modifications (Rai et al., 2017, 2019; Zahra et al., 2020). The Control Isolation of mitochondria
group animals received normal saline orally and subcutaneously, once
daily for 42 days. The Rotenone-intoxicated group was administered Mitochondrial pellet was isolated from the mouse brain, by differ­
with Rotenone (2 mg/kg bwt/day) subcutaneously for 35 days. While ential centrifugation (Singh et al., 2020). Homogenization of the
the Rotenone+Mp, and Rotenone+UA groups were first nigrostriatal region of midbarin was done using a buffer prepared by
orally-administered with Mp (100 mg/kg bwt/day) and UA (25 mg/kg mixing 225 mM mannitol, 5 mM HEPES, 75 mM sucrose, 1 mM ethylene
bwt/day), respectively for a week, then simultaneous administration of glycol tetra acetic acid (EGTA), and 1 mg/ml BSA (pH 7.4). The ho­
Mp and UA was done with Rotenone-intoxication for the next 35 days, in mogenate obtained was further centrifuged at 2000 g for 30 min at 4 ◦ C.
their respective groups. The behavioural parameters were analysed after The supernatant obtained after the centrifugation was further centri­
the completion of dosing within the next four days. Animals were further fuged at 12,000 g for 10 min. The pellet obtained after second centri­
sacrificed for brain isolation, to perform mitochondrial complexes and fugation constituted the mitochondria and synaptosomes and was
antioxidants assay, Western blotting, calcium level assay and immuno­ dissolved in homogenizing buffer containing digitonin (0.02%). The
histochemical analysis. mixture was again set to centrifugation for 10 min at 12,000 g to obtain a

3
W. Zahra et al. Phytomedicine Plus 2 (2022) 100343

crude mitochondrial fraction. This was further washed with the ho­ Calcium level determination
mogenizing buffer without BSA and EGTA twice, and the resuspension of
fraction was done in phosphate buffer (50 mM, pH 7.4). Proteins in all The level of calcium in the nigrostriatal region of the midbrain was
the samples were assayed by using 20 μg proteins, through the Bradford assayed through colorimetric calcium detection kit (Abcam). According
assay. to the kit instructions, the standards and the samples were prepared. 50
mg of the nigrostriatal tissue was resuspended in 500 μL of Calcium
Complex I-III assay Assay Buffer after washing in cold PBS solution. Homogenization of
tissue was done manually on ice, using a tissue grinder. Homogenates
The method of Sood et al. was incorporated for assessing complexes was set to centrifugation at 5000 rpm 5 min at 4 ◦ C, and supernatant
I-III activities, in our study (Sood et al., 2011). In this assay, cytochrome were collected. 90 μL of the Chromogenic Reagent, 50 μL of sample, and
c was reduced upon the catalytic oxidation of NADH to NAD+. NADH 60 μL of Calcium Assay Buffer was used as a reaction mixture which was
(6mMin 2mMglycylglycine buffer), glycylglycine buffer (0.2 M, pH 8.5), incubated for 10 min in dark. absorbence was further taken at 575 nm
and cytochrome c (10.5 mM) was used for the preparation of the reac­ and the concentration was determined using standard curve (Kumar
tion mixture. It was further added with 20 μg mitochondrial protein and et al., 2019).
change in the absorbence was recorded for 2 min at 550 nm. The
extinction coefficient for NADH at 340 nm is 6.22/mM/cm. The unit Immunohistochemistry (IHC)
nmol NADH oxidized/min/mg protein was used to express the activity
of the enzymes. Perfusion of Mice from each group was done using chilled 0.9% sa­
line solution and 4% paraformaldehyde solution (prepared in 0.1 M
Complex IV assay phosphate buffered saline, pH 7.4), intracardially. Whereas, 10% para­
formaldehyde was used for overnight postfixing of brains; immersed
The oxidation of reduced cytochrome c was assayed at 550 nm to later into 30% sucrose, until used further. Immunohistochemical stain­
detect the Complex IV activity (Sood et al., 2011). Potassium ferricya­ ing of TH and α-Synuclein was done using standard protocol (Gorbatyuk
nide, 10 mM phosphate buffer (pH 7.4), reduced cytochrome c and 20 μg et al., 2008). The cryotome (Leica, Wetzlar, Germany) was used to cut
of mitochondrial protein was used as a reaction mixture. The reduced 15 μm thick Coronal brain sections, which were washed further with
Cytochrome c was prepared by adding few crystals of sodium borohy­ 0.01 M PBS (pH 7.4) twice for10 min. Sections were then blocked for 1 h,
dride in the solution of oxidized cytochrome c (10 mg/ml). Change in first with 10% NGS in PBST and then 1% BSA-PBST. They were rinsed
the absorbance was observed for about 3 min and the complex IV ac­ thereafter with PBS and incubated with primary antibodies for TH (1:
tivity was calculated in nmol cytochrome c oxidized/min/mg of protein. 1000) and α-Synuclein (1:1500) at 4 ◦ C for 16 h. The secondary anti­
bodies viz.; TRITC-conjugated (anti-rabbit) and FITC-conjugated
Complex V assay (anti-mice), diluted in 1% BSA-PBS, were further incubated with the
respective primary-antibody incubated tissue sections for 2 h at room
In the Mitochondrial ATPase test, the amount of inorganic phos­ temperature. At each step, the washing of sections was done using PBS
phorus was measured in the assay that was liberated on ATP to ADP and 1% BSA-PBS and PBS, respectively. Mounting of sections on slides
hydrolysis (Singh et al., 2020). ATPase buffer (5 mM ATP, 2 mM MgCl2, was further done with polyvinyl alcohol with DABCO. The fluorescent
and 50 mM Tris–HCl, pH 8.5) and 20 μg of mitochondrial protein was microscope (Nikon, Thermo Fisher Scientific) was used to acquire the
incubated for 5 min at 30 ◦ C. The reaction mixture was then spun for 10 images, further processed using ImageJ software (NIH, United States).
min at 3000 g, after the addition of 10% TCA to it. The results were Results are expressed as % area and number of TH-positive neurons for
expressed as nmol inorganic phosphate (Pi) liberated/min/mg protein. the immunohistochemistry of α-Synuclein and TH respectively.

Mitochondrial GSH Western Blot (WB) analysis

Mitochondrial protein sample (20 μg) suspended in phosphate buffer RIPA buffer was used for homogenizing the nigrostriatal region of
was used to estimate the reduced Mitochondrial GSH. Centrifugation of the mouse brain. After 2 h of incubation of the homogenates at 4 ◦ C, they
all the samples were done after the addition of 25% trichloroacetic acid were centrifuged at 12,000 rpm for 30 min at 4 ◦ C. Supernatant from
(TCA) at 1500 g. 5,5-dithiobis 2-nitrobenzoic acid (DTNB) was added to each sample was collected and Bradford assay was performed for protein
the supernatant obtained which reacts with thiol groups to form 2-nitro- concentration quantification. 50 μg protein from each sample was
5mercapto benzoic acid. absorbance was recorded at 412 nm. Also, loaded in the wells of polyacrylamide gels, for electrophoresis. There­
standard curve was plotted using commercially available GSH and the after, PVDF membranes were used for transblotting of proteins; incu­
unit mmol of GSH/g mitochondrial protein was used to express the bated overnight further with primary antibodies of TH (1:4000),
result (Khanna and Nehru, 2007). α-Synuclein (1:1000), Bax (1:1000), Bcl-2 (1:800), caspase-3 (1:1000),
Iba1 (1:1000), p-Akt (1:1000), Akt (1:1000), p-GSK3β (1:1000), GSK3β
Manganese SOD (1:1000), and β-actin (1:1000). After washing with TBST and TBS, the
membranes were further incubated with the horseradish peroxidase-
To assess selectively activity of manganese superoxide dismutase (HRP-) conjugated secondary antibody for 2 h at room temperature.
(Mn-SOD), addition of hydrogen peroxide is done that inhibits Cu/Zn- Blots were visualized using the Enhanced chemiluminescence (ECL)
SOD, so as to distinguish the enzyme’s activity from that of Cu/Zn- system; relative density for each band was calculated with respect to that
SOD. EDTA (0.1 mM), sodium carbonate (50 mM), Triton-X (0.6%), of β-actin. Expression of relative density was done using Quantity One
and NBT (90 mM) and hydroxylamine hydrochloride were used to software (Windows, Bio-Rad).
prepare the reaction mixture (Khanna and Nehru, 2007). The reaction
progresses as the photooxidation of Hydroxylamine hydrochloride oc­ Statistical analysis
curs to produce superoxide which further reduces nitroblue tetrazolium
(NBT) in the reaction medium, so as the reaction was inhibited by SOD. Oneway analysis of variance (ANOVA) using the Stu­
Activity of SOD was expressed in units by taking the reading at 560 nm dent–Newman–Keuls test and by Student’s two-tailed t-test using
for 3 min and one enzymatic unit of SOD defines the amount of enzyme GraphPad Prism software was done to analyse the data obtained in the
required for 50% inhibition. study. Results are expressed as the mean ± SEM. p values < 0.05 were

4
W. Zahra et al. Phytomedicine Plus 2 (2022) 100343

considered statistically significant. was shortened on the administration of Mp (p < 0.001; Fig. 2(B)) and UA
(p < 0.001; Fig. 2(B)), in their respective Rotenone-intoxicated groups;
Results suggesting that bradykinesia induced by Rotenone-toxicity was allevi­
ated by Mp and UA treatment. The hind limb score of Rotenone-
Mp and UA ameliorated the behavioural deficits in Parkinsonian mouse intoxicated group was lowered in traction test ((p < 0.001); Fig. 2(C)),
model in comparison to control. However, pre-treatment with Mp (p < 0.01
Fig. 2(C)) and UA (p < 0.01 Fig. 2(C)) increased the traction score in
Different behavioural tests were performed to assess the behavioural their respective PD groups when compared to the diseased group. While
abnormalities in our study. Rotenone-intoxication in our study has in hanging test, the time was reduced for the Rotenone-intoxicated
induced Catalepsy in the PD mouse model (p < 0.0001; Fig. 2(A)) as group (p < 0.0001; Fig. 2(D)) in comparison to control. On the other
compared the control group. While the mice treated with Mp and UA in hand, Mp and UA treatment given to the Rotenone group significantly
the respective Rotenone-intoxicated groups showed little latency time improved the hanging time (p < 0.0001; Fig. 2(D)), as compared to the
(p < 0.0001; Fig. 2(A)) than the Rotenone-intoxicated group. Prolonged hanging time in Rotenone-intoxicated group. Rotenone-intoxicated mice
time (T-turn) was taken by the mice of Rotenone-intoxicated group (p < fall early in comparison to control group (p < 0.0001; Fig. 2(E)), in the
0.0001; Fig. 2(B)), when compared to control. Whereas, the duration rotarod test. On the other hand, by the administration of Mp and UA to

Fig. 2. Mp and UA alleviated the behavioural abnormalities in Rotenone-intoxicated PD mouse. Values are represented in the form of mean ± SEM (n = 10), *p <
0.01, **p < 0.001 & ***p < 0.0001. Abbreviations: UA, Ursolic acid; Mp, Mucuna pruriens; SEM, standard error of mean.

5
W. Zahra et al. Phytomedicine Plus 2 (2022) 100343

their respected Rotenone-treated groups have increased the time on the (p < 0.0001 for both Mp and UA) (Fig. 4(B)) significantly, in their
rotarod (p < 0.0001; Fig. 2(E)). Thus, results have suggested that Mp and respective PD groups.
UA improved the neurobehavioral lesion in the Rotenone-intoxicated PD
mouse model. Mp and UA attenuated the increased calcium level in nigrostriatal tissue of
PD mouse
Mp and UA improved Mitochondrial ETS impairment in rotenone-
intoxicated mice The level of Calcium (Fig. 5) was found significantly higher in the
nigrostriatal tissue of PD mouse as compared to control (p < 0.0001).
ATP in mitochondria, is produced by proper functioning of five While the neuroprotection provided by Mp and UA have significantly
enzyme complexes embedded in the inner mitochondrial membrane. reduced (p < 0.0001 for both Mp and UA) the Calcium level in their
Mitochondrial dysfunction led to the catastrophic consequences, haz­ respective Rotenone-intoxicated groups (Fig. 5).
ardous to the functionality of cell. Therefore, the effect of Mp and UA
was observed on the mitochondrial dysfunction caused by Rotenone- Effect of Mp and UA on TH and α-Synuclein
intoxication in our study. The complex I inhibitor, Rotenone was
observed to alter the activities of other complexes of ETS too, in our The immunohistochemistry of the SNpc region of the Rotenone-
study. As shown in Fig. 3, the effects of Mp and UA treatment has intoxicated mouse has revealed a decline in TH immunoreactivity (p
reversed the toxic effect of Rotenone on complex I-III (Fig. 3(A)), IV < 0.0001; Fig. 6(A)) indicating the loss of TH-positive neurons, and
(Fig. 3(B)), and V activities (Fig. 3(C)) in PD mouse model. Attenuated reduced protein expression (p < 0.0001; Fig. 7(A)) as compared to
activities of complexes I-III (p < 0.001), complex IV (p < 0.001), and control. While Mp and UA treatment has enhanced the TH immunore­
complex V (p < 0.0001) was observed in Rotenone-intoxicated PD mice activity (p < 0.0001 for Mp and p < 0.001 for UA; Fig. 6(A)) and the
as compared to that of Control. Whereas, neuroprotection by Mp and UA protein expression (p < 0.0001 for both Mp and UA; Fig. 7(A)) in their
to the Rotenone-treated animals helped improving the activities of respective Rotenone-intoxicated groups. On the other hand, the over­
complexes I-III (p < 0.01 for Mp treatment and p < 0.001 for UA expression of α-Synuclein was observed in the Rotenone-intoxicated as
treatment), complex IV (p < 0.001 for Mp treatment and p < 0.01 for UA suggested by the increased immunoreactivity (p < 0.0001; Fig. 6(B)) and
treatment), and complex V (p < 0.0001 for both Mp and UA treatment), expression (p < 0.0001; Fig. 7(B)) of the protein. Whereas, the treatment
when compared with Rotenone-intoxicated mice. with Mp and UA has significantly reduced both the immunoreactivity (p
< 0.0001 for Mp and p < 0.001 for UA; Fig. 6(B)) and expression (p <
Mitochondrial glutathione and superoxide dismutase level analysis 0.0001 for Mp and p < 0.001 for UA; Fig. 7(B)) of α-Synuclein in their
respective PD groups.
Oxidative stress leading to mitochondrial dysfunction results in the
depletion of reduced mtGSH and impaired Mn-SOD activity (Figs. 4(A) Mp and UA inhibited the rotenone-induced activation of mitochondrial
and 4(B)). In the Rotenone treated group, the attenuated level of mtGSH dysfunction mediated apoptosis in nigrostriatal tissue of mice
(p < 0.0001) and decline in the Mn-SOD activity (p < 0.0001) was
observed as compared to control in our study. On the other hand, Mp Expression of different apoptotic markers, namely, Bax, Bcl2 and
and UA reduced the oxidative stress and maintained the proper func­ cleaved Caspase 3 were investigated in our study to analyse the effect of
tionality of ETS, leading to an increment in the level of mtGSH (p < Mp and UA on PD mouse model (Figs. 7(D)–7(E)). Bax/Bcl-2 ratio (p <
0.0001 for Mp and p < 0.001 for UA) (Fig. 4(A)) and activity of Mn-SOD 0.0001) was seen to be increased considerably upon Rotenone-

Fig. 3. Mp and UA improved the mitochondrial complexes activities in Rotenone-mouse brain. Values are represented in the form of mean ± SEM (n = 5), *p < 0.01,
**p < 0.001 & ***p < 0.0001. Abbreviations: UA, Ursolic acid; Mp, Mucuna pruriens; SEM, standard error of mean.

6
W. Zahra et al. Phytomedicine Plus 2 (2022) 100343

Fig. 4. Mp and UA mediated enhancement of mitochondrial antioxidants in Rotenone-treated PD mouse model. Values are represented in the form of mean ± SEM
(n = 5), *p < 0.01, **p < 0.001 & ***p < 0.0001. Abbreviations: UA, Ursolic acid; Mp, Mucuna pruriens; SEM, standard error of mean.

neuroprotection against PD. Rotenone-intoxication in our study has


significantly reduced the p-Akt/Akt ratio (p < 0.0001; Fig. 7(G)) and p-
GSK3β/GSK3β ratio (p < 0.0001; Fig. 7(H)), when compared with the
control group, whereas the treatment of Mp and UA has increased the p-
Akt/Akt ratio (p<0.0001 for both Mp and UA; Fig. 7(G)) and p-GSK3β/
GSK3β ratio (p < 0.001 for both Mp and UA; Fig. 7(H)) in their respective
Parkinsonian Mouse groups. Furthermore, Rotenone-intoxication has
resulted in the microglial activation as suggested by the enhanced
expression of Iba1 (p < 0.0001; Fig. 7(F)) as compared to control. But the
treatment of Mp and UA has significantly reduced (p < 0.001 for Mp and
p < 0.0001 for UA; Fig. 7(F)) the expression of Iba1 in their respective
PD mouse groups, suggesting the attenuated immune response.

Discussion

Neurodegenerative diseases including PD, show their pathology


through interrelated pathogenic processes, viz., aggregation of proteins,
dysfunction of cell signaling, and ultimately the death of the neurons
(Iarkov et al., 2020; Kaur et al., 2019). Increased expression of α-Syn­
Fig. 5. Rotenone-intoxicity elevates the calcium level within the nigrostriatal uclein, both at transcriptional and translational level has been observed
region of the Parkinsonian mouse brain as compared to control. While neuro­ in the case of PD (Yuan et al., 2015). Thus, inhibition of α-Synuclein
protection by Mp and UA has reversed the toxicity and lessened the calcium overexpression and aggregation, in addition to the amelioration of other
level in both the Mp and UA treated Rotenone-intoxicated groups. Values are hallmarks of PD, is important to stop the progressive loss of the dopa­
represented in the form of mean ± SEM (n = 5), *p < 0.01, **p < 0.001 & ***p minergic neurons in the SNpc of midbrain. Also, PD therapies used
< 0.0001. Abbreviations: UA, Ursolic acid; Mp, Mucuna pruriens; SEM, stan­ nowadays are only symptomatic and identification of effective neuro­
dard error of mean. protectants having minimal side effects, is necessary (Iarkov et al.,
2020). Mp and UA, in this framework, have been extensively considered
intoxication whereas Mp and UA-treatment has reduced the same Bax/ for their anti-oxidative and anti-inflammatory properties, reported in
Bcl-2 ratio (p < 0.0001) in their respective groups, signifying the anti­ many studies (Rai et al., 2016, 2017; Yadav et al., 2013; Zahra et al.,
apoptotic property of Mp and UA in the parkinsonian mouse model 2020). The present study is aimed to explore further the neuroprotective
(Fig. 7(E)). Also, cleaved caspase-3 (p < 0.0001) was elevated in the activities of Mp and its constituent compound UA in ameliorating the
nigrostriatal tissue of PD mice whereas it was significantly attenuated on mitochondrial dysfunction, overexpression of α-synuclein, and dopa­
the administration of Mp (p < 0.01) and UA (p < 0.01) to the Rotenone- minergic neuronal loss through GSK-3β/Ca2+ signaling in
intoxicated group (Fig. 7(D)). Rotenone-induced mouse model of PD. This study also aims to check
whether, UA other than L-DOPA found in the aqueous extract of Mp is
Mp and UA reduced the activation of microglia by ameliorating the also responsible for its neuroprotective activity or not. Accordingly, the
dysregulation of kinases, Akt and GSK3β neurobehavior deficits by Rotenone-intoxication in our study, were
examined by performing the Catalepsy test, Hanging test, Pole test,
The phosphorylation of GSK3β by Akt plays important role in the

7
W. Zahra et al. Phytomedicine Plus 2 (2022) 100343

Fig. 6. Immunohistochemistry of TH and α-Synuclein in SNpc


of Control, Rotenone, Rotenone+Mp and Rotenone+UA mice
groups by using Image J Software at 20x magnification.
Rotenone intoxicity has reduced the TH immunoreactivity (A)
and increased the expression of α-Synuclein (B) in Rotenone-
treated group in comparison with control. While, neuro­
protection by Mp and UA has significantly improved the TH
immunoreactivity (A) and downregulated the expression of
α-Synuclein (B) in their respective groups as compared to
Rotenone-intoxicated group. Values are expressed as mean ±
SEM of percentage area (*p < 0.01, **p < 0.001, ***p <
0.0001, n = 3). Scale bar represents the degree of magnifi­
cation of the image (For 20X= 100 µm). Abbreviations: TH,
Tyrosine hydroxylase; SNpc, substantia nigra pars compacta;
Mp, Mucuna pruriens; UA, Ursolic acid.

Fig. 7. Relative expression of TH, α-Synuclein, pGSK-3β, p-Akt, Bcl-2, Bax, Caspase-3, and Iba1 in nigrostriatal region of different mouse groups. Values are expressed
as mean ± SEM (**p < 0.001, ***p < 0.0001). Abbreviations: Mp, Mucuna pruriens; UA, Ursolic acid; TH, Tyrosine hydroxylase; SEM, standard error of mean.

Rotarod test, and Traction test. Rotenone-intoxicity induced the and associated pathological pathways in Rotenone-induced mouse
behavioural impairments in the PD group, which were reversed upon the model of PD (Fig. 7).
neuroprotection offered by Mp and UA (Fig. 2), in consistent with pre­ Increment in the expression α-synuclein in transgenic mice (Duka
vious studies (Zahra et al., 2020). et al., 2009), also elevates the activation of GSK-3β, indicating, that the
Overexpressed and activated GSK-3β has also been observed in re­ activation of GSK-3β promotes accumulation and overexpression of
gions related with PD pathology, in the post-mortem brains (Golpich α-synuclein and vice-versa (Yuan et al., 2015). Thus, alteration in the
et al., 2015; Yuan et al., 2015). Dysregulation of PI3K/Akt signaling is activity of GSK-3β can protect against the debilitating conditions of PD
related to PD pathology and reduced phospho-Akt/total Akt ratio is (Singh et al., 2020; Yuan et al., 2015; Zhao et al., 2012). It has been
observed in the dopaminergic neurons of post-mortem PD brains demonstrated that neurotoxicity with MPTP, 6-OHDA and Rotenone
(Malagelada et al., 2008; Timmons et al., 2009). As Akt negatively enhanced the activation of GSK-3β (Hernandez-Baltazar et al., 2013;
regulates the activation of GSK-3β by phosphorylating the latter at Ser9, Singh et al., 2020). Moreover, reduced Ser9 and an elevated Tyr216
thus the activation of PI3K/Akt pathways might be investigated for phosphorylation resulted in the overexpression of α-Synuclein while the
confirming neuroprotection against PD (Zhang et al., 2011). Our study inhibition of GSK-3β abrogated the α-Synuclein mediated neurotoxicity
too suggested that activation of Akt by Mp and UA, deactivated GSK-3β (Gassowska et al., 2014). Altogether, by modulation of GSK-3β activity,

8
W. Zahra et al. Phytomedicine Plus 2 (2022) 100343

a potential approach against α-Synucleinopathy can be investigated in apoptosis (Singh et al., 2015). Increased apoptosis indicated by elevated
neurodegenerative conditions including PD. The present study also Bax/Bcl2 ratio and cleaved Caspase-3 expression, was also observed
validates the activation of GSK-3β and overexpression of α-synuclein, in upon Rotenone-intoxication in our study. Whereas, Mp and UA has
consistent with previous studies (Hernandez-Baltazar et al., 2013; Singh reversed these alterations by inhibiting the apoptosis and protecting the
et al., 2020). While neuroprotection provided by Mp and UA has dopaminergic neurons (Fig. 7).
significantly reversed the GSK-3β and α-Synuclein pathology in the Overall, our study suggests that neuroinflammation, mitochondrial
Rotenone-intoxicated PD mouse model (Fig. 6 and 7). dysfunction, apoptosis and loss of the dopaminergic neurons can be
The energy requirement in the CNS is mainly dependant on mito­ reversed by inhibiting the activation of GSK-3β/ α-Synuclein/ Ca2+ pa­
chondria and hence the organelle dysfunction mediates pathogenesis in thology upon neuroprotection with Mp and UA; establishing them as
the neurodegenerative diseases like PD (Hattingen et al., 2009). Rote­ possible therapeutic drugs for clinical interventions in PD therapy.
none being the inhibitor of Complex I, accelerates the formation of ROS
and hence promotes the mitochondrial dysfunction responsible for PD Conclusion
pathogenesis (Kussmaul and Hirst, 2006). Also, GSK-3β is seen to
regulate apoptosis by inhibiting the activity of complex I and associated The present study deals to explore the neuroprotective potential of
ROS production (King et al., 2008). On the other hand, inhibiting the Mp and its constituent compound, UA, in providing neuroprotection
activation of GSK-3β has protected the dopaminergic neurons from against key mediators (α-Synuclein overexpression, neuroinflammation,
Rotenone-induced toxicity by inhibiting the complex I altered Ca2+ level, mitochondrial dysfunction and apoptosis) of neuro­
dysfunction-induced apoptosis in PD (Wang et al., 2007). In this study, degeneration in PD through regulation of GSK-3β/ α-Synuclein/Ca2+
the attenuated activities of complexes I-III, complex IV, and complex V signaling. Collectively, UA other than L-DOPA, is also responsible for the
of ETS was observed upon the activation of GSK-3β through neuroprotective property of the Mp extract and hence both Mp and UA
Rotenone-intoxication. While the neuroprotection provided by Mp and can be used as potential pharmacological candidate for ameliorating
UA has significantly increased the activities of the ETC in the PD mouse neurodegeneration in PD and should be further investigated in pre-
model (Fig. 3). Therefore, it is suggested that activation of GSK-3β clinical studies.
hampered electron transfer in between the complexes, which were
reversed upon the treatment of Mp and UA. Funding
Because of the increased production of ROS and ETC impairment,
excessive consumption of mitochondrial GSH (mtGSH, reduced) and NA
decline in the Mn-SOD is observed. Whereas, GSH helps in protecting the
dopaminergic neurons against the adverse effects of neurotoxins in the
Ethics approval
case of PD (Li et al., 2016; Mani et al., 2018). Our results also potentiate
the neuroprotective activity of Mp and UA, by increasing the level of
The ethical standards of the institution or practice was followed and
mtGSH and activity of Mn-SOD upon ameliorating the adverse effects of
the experiments were performed according to the guidelines for animal
the neurotoxin Rotenone (Fig. 4).
experiments recognized by the Institutional Animal Ethics Committees
Changes in the Ca2+ levels enhance the production of ROS, resulting
(IAECs) of the Laboratory Animal Research (Reg. No. 1802/GO/Re/S/
in the dysfunction of the mitochondria by forming the mitochondrial
15/CPCSEA) at Banaras Hindu University, India.
permeability transition pore (mPTP) opening, and release of cytochrome
c . Oligomerisation of α-Synuclein also enhances the permeability of
CRediT authorship contribution statement
plasma membrane, thereby facilitating Ca2+ influx inside the dopami­
nergic neurons (Schmidt et al., 2012; Tsigelny et al., 2012). GSK-3β
Walia Zahra: Conceptualization, Methodology, Writing – review &
activation and increment in the level of Ca2+ is also linked to PD pa­
editing. Hareram Birla: Visualization, Investigation. Saumitra Sen
thology (Yuan et al., 2015). Our study also suggests that
Singh: Data curation, Writing – original draft. Aaina Singh Rathore:
Rotenone-intoxication leading to activation of GSK-3β and over­
Data curation, Writing – original draft. Hagera Dilnashin: Visualiza­
expression of α-Synuclein has increased the Ca2+ level in the nigros­
tion, Investigation. Richa Singh: Data curation, Writing – original draft.
triatal tissue, which were significantly improved by providing
Priyanka Kumari Keshri: . Shekhar Singh: Visualization, Investiga­
neuroprotection with Mp and UA in Rotenone-intoxicated PD mouse
tion. Surya Pratap Singh: Supervision, Validation.
(Fig. 5).
Various studies have also reported the activation of microglia and
hence neuroinflammation, in the SN of animal models and patients of PD Declaration of Competing Interest
(Hirsch and Hunot, 2009; Ouchi et al., 2009). Additionally, inhibiting
the inflammatory response protects the dopaminergic neurons against The authors declare that they have no known competing financial
neurotoxin-induced cell damage (Lofrumento et al., 2014). Activation of interests or personal relationships that could have appeared to influence
GSK-3β also promotes the microglial activation and associated inflam­ the work reported in this paper.
matory responses (Yuskaitis and Jope, 2009). In our study too, increased
expression of the microglial marker Iba1 is observed in the
References
Rotenone-intoxicated mouse model of PD. While administration of Mp
and UA has ameliorated the inflammatory response as suggested by the Birla, H., Rai, S.N., Singh, S.S., Zahra, W., Rawat, A., Tiwari, N., Singh, S.P., 2019.
reduced expression of Iba1 (Fig. 7). Tinospora cordifolia suppresses neuroinflammation in parkinsonian mouse model.
Neuromol. Med. 21 (1), 42–53.
The expression of the rate-limiting enzyme of the dopamine syn­
Cannon, J.R., Tapias, V., Na, H.M., Honick, A.S., Drolet, R.E., Greenamyre, J.T., 2009.
thesis, i.e., TH is seen to be reduced upon Rotenone-intoxication and A highly reproducible rotenone model of Parkinson’s disease. Neurobiol. Dis. 34 (2),
α-synuclein overexpression. While Mp and UA were found to protect the 279–290.
dopaminergic neurons as suggested by TH expression in our study (Fig. 6 Cilia, R., Laguna, J., Cassani, E., Cereda, E., Pozzi, N.G., Isaias, I.U., et al., 2017. Mucuna
pruriens in Parkinson disease: a double-blind, randomized, controlled, crossover
and 7) (Liu et al., 2008; Zahra et al., 2020). The deregulation in the study. Neurology 89, 432–438.
expression of Bcl-2 family proteins, also potentiates GSK-3β-mediated Dexter, D.T., Jenner, P., 2013. Parkinson disease: from pathology to molecular disease
apoptosis of the neurons (Wang et al., 2013). Activation of Caspase 3 mechanisms. Free Radic. Biol. Med. 62, 132–144.
Duka, T., Duka, V., Joyce, J.N., Sidhu, A., 2009. α-Synuclein contributes to GSK-3β-
and alteration in the expressions of the anti-apoptotic protein, Bcl-2 and catalyzed Tau phosphorylation in Parkinson’s disease models. FASEB J. 23 (9),
proapoptotic protein, Bax is observed during the mitochondrial 2820–2830.

9
W. Zahra et al. Phytomedicine Plus 2 (2022) 100343

Gąssowska, M., Czapski, G.A., Pająk, B., Cieślik, M., Lenkiewicz, A.M., Adamczyk, A., Rai, S.N., Birla, H., Singh, S.S., Zahra, W., Patil, R.R., Jadhav, J.P., 2017. Mucuna pruriens
2014. Extracellular α-synuclein leads to microtubule destabilization via GSK-3β- protects against MPTP intoxicated neuroinflammation in Parkinson’s disease
dependent Tau phosphorylation in PC12 cells. PLoS ONE 9 (4), e94259. through NF-KB/pAKT signaling pathways. Front Aging Neurosci. 9, 1–14.
Golpich, M., Amini, E., Hemmati, F., Ibrahim, N.M., Rahmani, B., Mohamed, Z., Rai, S.N., Chaturvedi, V.K., Singh, P., Singh, B.K., Singh, M.P., 2020. Mucuna pruriens in
Ahmadiani, A., 2015. Glycogen synthase kinase-3 beta (GSK-3β) signaling: Parkinson’s and in some other diseases: recent advancement and future prospective.
implications for Parkinson’s disease. Pharmacol. Res. 97, 16–26. 3 Biotech 10 (12), 1–11.
Gorbatyuk, O.S., Li, S., Sullivan, L.F., Chen, W., Kondrikova, G., Manfredsson, F.P., Rai, S.N., Singh, P., Varshney, R., Chaturvedi, V.K., Vamanu, E., Singh, M.P., Singh, B.K.,
Muzyczka, N., 2008. The phosphorylation state of Ser-129 in human α-synuclein 2021a. Promising drug targets and associated therapeutic interventions in
determines neurodegeneration in a rat model of Parkinson disease. PNAS 105 (2), Parkinson’s disease. Neural Regen. Res. 16 (9), 1730.
763–768. Rai, S.N., Tiwari, N., Singh, P., Mishra, D., Singh, A.K., Hooshmandi, E., Vamanu, E.,
Habtemariam, S., 2019. Antioxidant and anti-inflammatory mechanisms of Singh, M.P., 2021b. Therapeutic potential of vital transcription factors in
neuroprotection by ursolic acid: addressing brain injury, cerebral ischemia, alzheimer’s and parkinson’s disease with particular emphasis on transcription factor
cognition deficit, anxiety, and depression. Oxid. Med. Cell Longev. 2019. eb mediated autophagy. Front. Neurosci. 15, 777347.
Hattingen, E., Magerkurth, J., Pilatus, U., Mozer, A., Seifried, C., Steinmetz, H., Rai, S.N., Yadav, S.K., Singh, D., Singh, S.P., 2016. Ursolic acid attenuates oxidative
Hilker, R., 2009. Phosphorus and proton magnetic resonance spectroscopy stress in nigrostriatal tissue and improves neurobehavioral activity in MPTP- induced
demonstrates mitochondrial dysfunction in early and advanced Parkinson’s disease. Parkinsonian mouse model. J. Chem. Neuroanat. 71, 41–49.
Brain 132 (12), 3285–3297. Rai, S.N., Zahra, W., Singh, S.S., Birla, H., Keswani, C., Dilnashin, H., Singh, S.P., 2019.
Hernandez-Baltazar, D., Mendoza-Garrido, M.E., Martinez-Fong, D., 2013. Activation of Anti-inflammatory activity of ursolic acid in MPTP-induced parkinsonian mouse
GSK-3β and caspase-3 occurs in nigral dopamine neurons during the development of model. Neurotox. Res. 36 (3), 452–462.
apoptosis activated by a striatal injection of 6-hydroxydopamine. PLoS ONE 8 (8), Rcom-H’cheo-Gauthier, A.N., Osborne, S.L., Meedeniya, A.C., Pountney, D.L., 2016.
e70951. Calcium: alpha-synuclein interactions in alpha-synucleinopathies. Front. Neurosci.
Hirsch, E.C., Hunot, S., 2009. Neuroinflammation in Parkinson’s disease: a target for 10, 570.
neuroprotection? Lancet Neurol. 8 (4), 382–397. Rizek, P., Kumar, N., Jog, M.S., 2016. An update on the diagnosis and treatment of
Hu, D., Sun, X., Liao, X., Zhang, X., Zarabi, S., Schimmer, A., et al., 2019. Alphasynuclein Parkinson disease. CMAJ 188, 1157–1165.
suppresses mitochondrial protease ClpP to trigger mitochondrial oxidative damage Ruiz, P.J.G., Catalan, M.J., Carril, J.F., 2011. Initial motor symptoms of Parkinson
and neurotoxicity. Acta Neuropathol. 137, 939–960. disease. Neurologist 17, S18–S20.
Hu, M., Li, F., Wang, W., 2018. Vitexin protects dopaminergic neurons in MPTP-induced Schmidt, F., Levin, J., Kamp, F., Kretzschmar, H., Giese, A., Bötzel, K., 2012. Single-
Parkinson’s disease through PI3K/Akt signaling pathway. Drug Des. Devel. Ther. 12, channel electrophysiology reveals a distinct and uniform pore complex formed by
565. α-synuclein oligomers in lipid membranes. PLoS ONE 7, e42545.
Huang, Y., Sun, L., Zhu, S., Xu, L., Liu, S., Yuan, C., Wang, X., 2020. Neuroprotection Singh, L., Pushker, N., Saini, N., Sen, S., Sharma, A., Bakhshi, S., Kashyap, S., 2015.
against Parkinson’s disease through the activation of akt/gsk3β signaling pathway Expression of pro-apoptotic Bax and anti-apoptotic Bcl-2 proteins in human
by Tovophyllin A. Front. Neurosci. 14, 723. retinoblastoma. Clin. Experiment. Ophthalmol. 43 (3), 259–267.
Iarkov, A., Barreto, G.E., Grizzell, J.A., Echeverria, V., 2020. Strategies for the treatment Singh, S.S., Rai, S.N., Birla, H., Zahra, W., Kumar, G., Gedda, M.R., Singh, S.P., 2018.
of parkinson’s disease: beyond dopamine. Front. Aging Neurosci. 12, 4. Effect of chlorogenic acid supplementation in MPTP-intoxicated mouse. Front.
Kalia, L.V., Lang, A.E., 2015. Parkinson’s disease. Lancet 386, 896–912. Pharmacol. 9, 757.
Kaur, R., Mehan, S., Singh, S., 2019. Understanding multifactorial architecture of Singh, S.S., Rai, S.N., Birla, H., Zahra, W., Rathore, A.S., Dilnashin, H., Singh, S.P., 2020.
Parkinson’s disease: pathophysiology to management. Neurol. Sci. 40 (1), 13–23. Neuroprotective effect of chlorogenic acid on mitochondrial dysfunction-mediated
Khanna, P., Nehru, B., 2007. Antioxidant enzymatic system in neuronal and glial cells apoptotic death of DA neurons in a Parkinsonian mouse model. Oxid. Med. Cell.
enriched fractions of rat brain after aluminum exposure. Cell. Mol. Neurobiol. 27 (7), Longev. 2020.
959–969. https://doi.org/10.1007/s10571-007-9233-2. Sood, P.K., Nahar, U., Nehru, B., 2011. Curcumin attenuates aluminum-induced
King, T.D., Clodfelder-Miller, B., Barksdale, K.A., Bijur, G.N., 2008. Unregulated oxidative stress and mitochondrial dysfunction in rat brain. Neurotox. Res. 20 (4),
mitochondrial GSK3β activity results in NADH: ubiquinone oxidoreductase 351–361.
deficiency. Neurotox. Res. 14 (4), 367–382. Surmeier, D.J., Guzman, J.N., Sanchez, J., Schumacker, P.T., 2012. Physiological
Kumar, G., Mukherjee, S., Paliwal, P., Singh, S.S., Birla, H., Singh, S.P., Patnaik, R., 2019. phenotype and vulnerability in Parkinson’s disease. Cold Spring Harb. Perspect 2
Neuroprotective effect of chlorogenic acid in global cerebral ischemia-reperfusion (7), a009290.
rat model. Naunyn Schmiedebergs Arch. Pharmacol. 392 (10), 1293–1309. Timmons, S., Coakley, M.F., Moloney, A.M., O’Neill, C., 2009. Akt signal transduction
Kussmaul, L., Hirst, J., 2006. The mechanism of superoxide production by NADH: dysfunction in Parkinson’s disease. Neurosci. Lett. 467 (1), 30–35.
ubiquinone oxidoreductase (complex I) from bovine heart mitochondria. PNAS 103 Tolleson, C.M., Fang, J.Y., 2013. Advances in the mechanisms of Parkinson’s disease.
(20), 7607–7612. Discov. Med. 15 (80), 61–66.
Li, D.W., Liu, Z.Q., Chen, W., Yao, M., Li, G.R., 2014. Association of glycogen synthase Tsigelny, I.F., Sharikov, Y., Wrasidlo, W., Gonzalez, T., Desplats, P.A., Crews, L.,
kinase‑3β with Parkinson’s disease. Mol. Med. Rep. 9 (6), 2043–2050. Masliah, E., 2012. Role of α-synuclein penetration into the membrane in the
Li, X.H., Dai, C.F., Chen, L., Zhou, W.T., Han, H.L., Dong, Z.F., 2016. 7, 8-dihydroxy­ mechanisms of oligomer pore formation. FEBS J. 279 (6), 1000–1013.
flavone ameliorates motor deficits via suppressing α-synuclein expression and Uchegbu, R.I., Ahuchaogu, A.A., Mbadiugha, C.N., Amanze, K.O., Igara, C.E., Iwu, I.C.,
oxidative stress in the MPTP-induced mouse model of Parkinson’s disease. CNS et al., 2016. Antioxidant, anti-inflammatory and antibacterial activities of the seeds
Neurosci. Ther. 22 (7), 617–624. of Mucuna pruriens (UTILIS). Am. Chem. Sci. J. 13, 1–8.
Liu, D., Jin, L., Wang, H., Zhao, H., Zhao, C., Duan, C., Yang, H., 2008. Silencing Uhegbu, A.F.O., Elekwa, I., Ukoha, C., 2005. Comparative efficacy of crude aqueous
α-synuclein gene expression enhances tyrosine hydroxylase activity in MN9D cells. extract of Mangifera indica, Carica papaya and sulfadoxine pyrimethamine on the
Neurochem. Res. 33 (7), 1401–1409. mice infested with malaria parasite in vivo. Global J. Pure Appl. Sci. 11, 399–401.
Lofrumento, D.D., Nicolardi, G., Cianciulli, A., Nuccio, F.D., Pesa, V.L., Carofiglio, V., Wang, R., Xia, L., Gabrilove, J., Waxman, S., Jing, Y., 2013. Downregulation of Mcl-1
Panaro, M.A., 2014. Neuroprotective effects of resveratrol in an MPTP mouse model through GSK-3β activation contributes to arsenic trioxide-induced apoptosis in acute
of Parkinson’s-like disease: possible role of SOCS-1 in reducing pro-inflammatory myeloid leukemia cells. Leukemia 27 (2), 315–324.
responses. Innate Immun. 20 (3), 249–260. Wang, W., Yang, Y., Ying, C., Li, W., Ruan, H., Zhu, X., Li, M., 2007. Inhibition of
Malagelada, C., Jin, Z.H., Greene, L.A., 2008. RTP801 is induced in Parkinson’s disease glycogen synthase kinase-3β protects dopaminergic neurons from MPTP toxicity.
and mediates neuron death by inhibiting Akt phosphorylation/activation. Neuropharmacology 52 (8), 1678–1684.
J. Neurosci. 28 (53), 14363–14371. Yadav, S.K., Prakash, J., Chouhan, S., Singh, S.P., 2013. Mucuna pruriens seed extract
Mani, S., Sekar, S., Barathidasan, R., Manivasagam, T., Thenmozhi, A.J., Sevanan, M., reduces oxidative stress in nigrostriatal tissue and improves neurobehavioral activity
Sakharkar, M.K., 2018. Naringenin decreases α-synuclein expression and in paraquat-induced Parkinsonian mouse model. Neurochem. Int. 62, 1039–1047.
neuroinflammation in MPTP-induced Parkinson’s disease model in mice. Neurotox. Yadav, S.K., Prakash, J., Chouhan, S., Westfall, S., Verma, M., Singh, T.D., et al., 2014.
Res. 33 (3), 656–670. Comparison of the neuroprotective potential of Mucuna pruriens seed extract with
Manna, S., Bhattacharyya, D., Mandal, T.K., Dey, S., 2006. Neuropharmacological effects estrogen in 1 methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mice
of deltamethrin in rats. J. Vet. Sci. 7 (2), 133–136. model. Neurochem. Int. 65, 1–13.
Martinez, B.A., Petersen, D.A., Gaeta, A.L., Stanley, S.P., Caldwell, G.A., Caldwell, K.A., Yadav, S.K., Rai, S.N., Singh, S.P., 2016. Mucuna pruriens shows neuroprotective effect by
2017. Dysregulation of the mitochondrial unfolded protein response induces non- inhibiting apoptotic pathways of dopaminergic neurons in the paraquat mouse
apoptotic dopaminergic neurodegeneration in C. elegans models of Parkinson’s model of parkinsonism. Eur. J. Pharmaceut. Med. Res. 3, 441–451.
Disease. J. Neurosci. 37, 11085–11100. Yuan, Y.H., Yan, W.F., Sun, J.D., Huang, J.Y., Mu, Z., Chen, N.H., 2015. The molecular
Monahan, A.J., Warren, M., Carvey, P.M., 2008. Neuroinflammation and peripheral mechanism of rotenone-induced α-synuclein aggregation: emphasizing the role of
immune infiltration in Parkinson’s disease: an autoimmune hypothesis. Cell the calcium/GSK3β pathway. Toxicol. Lett. 233 (2), 163–171.
Transplant 17 (4), 363–372. Yuskaitis, C.J., Jope, R.S., 2009. Glycogen synthase kinase-3 regulates microglial
Moon, H.E., Paek, S.H., 2015. Mitochondrial dysfunction in Parkinson’s Disease. Exp. migration, inflammation, and inflammation-induced neurotoxicity. Cell. Signal. 21
Neurobiol. 24, 103–116. (2), 264–273.
Morris, G., Berk, M., 2015. The many roads to mitochondrial dysfunction in
neuroimmune and neuropsychiatric disorders. BMC Med. 13, 68.
Ouchi, Y., Yagi, S., Yokokura, M., Sakamoto, M., 2009. Neuroinflammation in the living
brain of Parkinson’s disease. Parkinsonism Relat D 15, S200–S204.

10
W. Zahra et al. Phytomedicine Plus 2 (2022) 100343

Zahra, W., Rai, S.N., Birla, H., Singh, S.S., Rathore, A.S., Dilnashin, H., Singh, S.P., 2020. Zhao, D.M., Li, N.N., Zhang, J.H., Chang, X.L., Mao, X.Y., Liao, Q., Peng, R., 2012. GSK3β
Neuroprotection of rotenone-induced Parkinsonism by ursolic acid in PD mouse reduces risk of sporadic Parkinson’s disease in ethnic Chinese. Am. J. Med. Genet. B:
model. CNS & Neurological Disorders-Drug Targets 19 (7), 527–540. Neuropsychiatr. Genet. 159 (6), 718–721.
Zhang, H., Mak, S., Cui, W., Li, W., Han, R., Hu, S., Han, Y., 2011. Tacrine (2)–ferulic
acid, a novel multifunctional dimer, attenuates 6-hydroxydopamine-induced
apoptosis in PC12 cells by activating Akt pathway. Neurochem. Int. 59 (7), 981–988.

11

You might also like