ArticlePDF Available

A Flavonoid-Rich Extract of Mandarin Juice Counteracts 6-OHDA-Induced Oxidative Stress in SH-SY5Y Cells and Modulates Parkinson-Related Genes

Authors:

Abstract and Figures

Parkinson’s disease (PD) is a degenerative disorder of the nervous system due to unceasing impairment of dopaminergic neurons situated in the substantia nigra. At present, anti-PD drugs acting on dopamine receptors are mainly symptomatic and have only very limited neuroprotective effects, whereas drugs slowing down neurodegeneration of dopaminergic neurons and deterioration of clinical symptoms are not yet available. Given that, the development of more valuable pharmacological strategies is highly demanded. Comprehensive research on innovative neuroprotective drugs has proven that anti-inflammatory and antioxidant molecules from food sources may prevent and/or counteract neurodegenerative diseases, such as PD. The present study was aimed at the evaluation the protective effect of mandarin juice extract (MJe) against 6-hydroxydopamine (6-OHDA)-induced SH-SY5Y human neuroblastoma cell death. Treatment of differentiated SH-SY5Y cells with 6-OHDA brought cell death, and specifically, apoptosis, which was significantly inhibited by the preincubation with MJe through caspase 3 blockage and the modulation of p53, Bax, and Bcl-2 genes. In addition, it showed antioxidant properties in abiotic models as well as in vitro, where it reduced both reactive oxygen and nitrogen species induced by 6-OHDA, along with restored mitochondrial membrane potential, and prevented the oxidative DNA damage evoked by 6-OHDA. Furthermore, MJe restored the impaired balance of SNCA, LRRK2, PINK1, parkin, and DJ-1 gene levels, PD-related factors, caused by 6-OHDA oxidative stress. Overall, these results indicate that MJe exerts neuroprotective effects against 6-OHDA-induced cell death in SH-SY5Y cells by mechanisms involving both the specific interaction with intracellular pathways and its antioxidant capability. Our study suggests a novel possible strategy to prevent and/or ameliorate neurodegenerative diseases, such as PD.
Content may be subject to copyright.
antioxidants
Article
A Flavonoid-Rich Extract of Mandarin Juice Counteracts
6-OHDA-Induced Oxidative Stress in SH-SY5Y Cells and
Modulates Parkinson-Related Genes
Santa Cirmi 1,2 , Alessandro Maugeri 2, Giovanni Enrico Lombardo 2, *, Caterina Russo 2,3, Laura Musumeci 2,
Sebastiano Gangemi 4, Gioacchino Calapai 5, Davide Barreca 2and Michele Navarra 2, *


Citation: Cirmi, S.; Maugeri, A.;
Lombardo, G.E.; Russo, C.;
Musumeci, L.; Gangemi, S.; Calapai,
G.; Barreca, D.; Navarra, M. A
Flavonoid-Rich Extract of Mandarin
Juice Counteracts 6-OHDA-Induced
Oxidative Stress in SH-SY5Y Cells
and Modulates Parkinson-Related
Genes. Antioxidants 2021,10, 539.
https://doi.org/10.3390/
antiox10040539
Academic Editor: Domenico Nuzzo
Received: 24 February 2021
Accepted: 26 March 2021
Published: 30 March 2021
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2021 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
1Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy;
santa.cirmi@uniba.it
2Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina,
98168 Messina, Italy; amaugeri@unime.it (A.M.); carusso@unime.it (C.R.); laura.musumeci@unime.it (L.M.);
davide.barreca@unime.it (D.B.)
3Fondazione “Prof. Antonio Imbesi”, 98123 Messina, Italy
4Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy;
sebastiano.gangemi@unime.it
5Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina,
98125 Messina, Italy; gioacchino.calapai@unime.it
*Correspondence: gelombardo@unime.it (G.E.L.); mnavarra@unime.it (M.N.)
Abstract:
Parkinson’s disease (PD) is a degenerative disorder of the nervous system due to unceasing
impairment of dopaminergic neurons situated in the substantia nigra. At present, anti-PD drugs
acting on dopamine receptors are mainly symptomatic and have only very limited neuroprotective
effects, whereas drugs slowing down neurodegeneration of dopaminergic neurons and deterioration
of clinical symptoms are not yet available. Given that, the development of more valuable pharmaco-
logical strategies is highly demanded. Comprehensive research on innovative neuroprotective drugs
has proven that anti-inflammatory and antioxidant molecules from food sources may prevent and/or
counteract neurodegenerative diseases, such as PD. The present study was aimed at the evaluation
the protective effect of mandarin juice extract (MJe) against 6-hydroxydopamine (6-OHDA)-induced
SH-SY5Y human neuroblastoma cell death. Treatment of differentiated SH-SY5Y cells with 6-OHDA
brought cell death, and specifically, apoptosis, which was significantly inhibited by the preincubation
with MJe through caspase 3 blockage and the modulation of p53, Bax, and Bcl-2 genes. In addition, it
showed antioxidant properties in abiotic models as well as
in vitro
, where it reduced both reactive
oxygen and nitrogen species induced by 6-OHDA, along with restored mitochondrial membrane
potential, and prevented the oxidative DNA damage evoked by 6-OHDA. Furthermore, MJe restored
the impaired balance of SNCA, LRRK2, PINK1, parkin, and DJ-1 gene levels, PD-related factors,
caused by 6-OHDA oxidative stress. Overall, these results indicate that MJe exerts neuroprotective
effects against 6-OHDA-induced cell death in SH-SY5Y cells by mechanisms involving both the
specific interaction with intracellular pathways and its antioxidant capability. Our study suggests a
novel possible strategy to prevent and/or ameliorate neurodegenerative diseases, such as PD.
Keywords:
neurodegenerative diseases; Parkinson’s disease; mandarin juice; Citrus reticulata; 6-
OHDA; neuroprotection; SH-SY5Y; oxidative stress; natural products
1. Introduction
Parkinson’s disease (PD) is an aging-related neurodegenerative disease (ND) whose
characteristics are progressive aggregation of
α
-synuclein in surviving neurons and se-
lective death of dopaminergic neurons in the substantia nigra, particularly its pars com-
pacta [
1
]. PD shows tremor at rest, rigidity, bradykinesia, slowed movements, and postural
instability. The Global Burden of Diseases 2016 has gathered the evidence on neurological
Antioxidants 2021,10, 539. https://doi.org/10.3390/antiox10040539 https://www.mdpi.com/journal/antioxidants
Antioxidants 2021,10, 539 2 of 16
disorders, showing that PD had an incidence of 1 million cases worldwide, of which more
than 150,000 in the EU, and PD-related deaths were more than 340,000 globally, with 60,000
just in the EU [
2
]. Although the actual triggers of PD remain undefined, the link between
oxidative stress, mitochondrial impairment, protein misprocessing, and genetic variation is
pivotal in the pathogenesis of the disease [3].
In the unceasing search of innovative neuroprotective drugs, anti-inflammatory and
antioxidant molecules from dietary sources have been suggested to prevent and/or coun-
teract NDs, such as PD [
4
6
]. In this context, nutraceuticals along with food supplements
have been proven to provide neuroprotective effects in several experimental models, and
their use as substitute to synthetic drugs or in combination with them is supported by
their ability to hinder oxidative stress as well as to interact with intracellular pathways
involved in NDs [
4
,
7
,
8
]. These properties might be ascribed to the presence of polyphenolic
compounds, abundantly present in fruits, vegetables, cereals, and beverages. The main
class of polyphenols is flavonoids, whose major dietary sources are fruits, especially citrus,
vegetables, tea, coffee, and red wine. They possess a remarkable spectrum of biological
activities, such as antioxidant, free radical scavenging, metal ion chelating, vasoprotective,
hepatoprotective, anti-cancer, anti-infective and anti-inflammatory [913].
Based on this, we investigated whether a flavonoid-rich extract from Citrus reticulata
Blanco (mandarin; MJe) protects differentiated SH-SY5Y cells from 6-hydroxydopamine
(6-OHDA)-induced neurotoxicity and explored its mechanism of action.
2. Materials and Methods
2.1. Drug
Citrus reticulata Blanco fruits were harvested from crops located in Sicily (Italy). The
flavonoid fraction of mandarin juice (MJe) was provided by the company “Agrumaria
Corleone” (Palermo, Italy). The extract was obtained by passing the mandarin juice through
columns equipped with flavonoid-adsorbent resins, which were then eluted. The extract
was centrifuged for 15 min at 6000 rpm and then spray-dried. One hundred milliliters of
MJe yielded 109 mg of dry extract. Aliquots of MJe were stored at
20
C and defrosted
just before use.
2.2. Chemical Characterization of MJe
2.2.1. Reagents and Standard Solutions
HPLC-grade acetonitrile and DMSO were obtained by Sigma-Aldrich (St. Louis, MO,
USA). Vicenin-2, hesperidin, eriocitrin, narirutin, sinensetin, nobiletin, and tangeretin
were supplied from Extrasynthèse (Genay, France). Lucenin-2 4
0
-methylether and orientin
4
0
-methyl ether were separated from Citrus limetta and Citrus bergamia [
14
,
15
] and used
as standards. The Iso-Disc P-34, 3 mm diameter PTFE membrane (0.45
µ
m pore size) was
from Supelco (Bellefonte, PA, USA). All the other reagents and chemicals employed in this
study were of analytical grade and were purchased from Sigma (St. Louis, MO, USA).
2.2.2. Sample Preparation
A solution of DMSO/H
2
O (1:1) was added to the lyophilized powder to obtain
a final concentration of (10.0 mg/mL), and the mixture was centrifuged for 5 min at
3200 rpm. The supernatant liquid was filtered through an Iso-Disc P-34, 3 mm diameter
PTFE membrane with a 0.45
µ
m pore size (Supelco, Bellefonte, PA, USA) and utilized for
HPLC-DAD separation.
2.2.3. RP-DAD-HPLC Separation and Identification
Reverse phase-diode array detector-high performance liquid chromatography (RP-
DAD-HPLC) was performed using a Shimadzu system (Shimadzu Ltd., Canby, OR, USA),
consisting of a LC-10AD pump system, a vacuum degasser, a quaternary solvent mixing,
an SPD-M10A diode array detector, and a Rheodyne 7725i injector. The separation of
each compound was performed on a 250
×
4.6 mm i.d., 5 mm Discovery C18 column,
Antioxidants 2021,10, 539 3 of 16
supplied by Supelco (Bellefonte, PA, USA), equipped with a 20
×
4.0 mm guard column.
The column was placed in a column oven set at 30
C. The injection loop was 20
µ
L,
and the flow rate was 1.0 mL/min. The mobile phase consisted of a linear gradient of
acetonitrile in H
2
O as follows: 5–20% (0–15 min), 20–30% (15–20 min), 30–50% (20–30 min),
50–100% (30–35 min), 100% (35–40 min), 100–5% (40–50 min), and 5% (50–60 min). UV
spectra were recorded between 200 and 600 nm, and simultaneous detection by diode array
was performed at 278, 310, and 325 nm. Each sample was tested three times and gave
superimposable chromatograms. Peak identification was performed by matching retention
time and UV spectra against reference compounds and spiking the samples with pure
reference compounds. The calibration lines were obtained using known concentrations
of pure compounds and selected to match the concentration present in the tested sample.
Quantitative analysis was carried out by integration of the areas of the peaks from the
chromatogram at 280 and 325 nm for flavanone and flavone derivatives, respectively. The
calibration curves were constructed, and linear regression equations were obtained by
plotting the ratios of compounds’ peak areas to the peak areas of the external standard,
against the known concentrations of pure compounds.
2.2.4. Acid Hydrolysis
Hydrolysis was performed on MJe according to already published procedure [
16
].
Briefly, 10 mL of HCl (6 M) in a methanol (25 mL)/water (10 mL) solution was added to
5 mL of MJe to obtain a solution of 1.2 M HCl in 50% aqueous methanol. As an antioxidant,
we added ascorbic acid (50 mg). After refluxing at 90
C for 20 h under constant stirring, the
solution was cooled at room temperature, the solvents evaporated under reduced pressure,
and the residue suspended in 10 mL water/DMF (1:1). The mixture was filtered through
an Iso-Disc P-34 membrane and analyzed by HPLC.
2.3. Cell Culture and Treatment
Experiments were carried out using the SH-SY5Y human neuroblastoma cell line orig-
inally from ATCC (Rockville, MD, USA). Their differentiation was performed with 10
µ
M
retinoic acid (RA; Sigma–Aldrich, Milan, Italy) in MEM/Ham’s F12 medium supplemented
for 5 days [
17
]. All reagents were from Gibco (Life Technologies, Monza, Italy). The stock
solution of MJe 400 mg/mL was prepared in DMSO that was employed, upon further
dilution in culture medium, to obtain the working concentrations. The same percentages
of DMSO present in these dilutions served as vehicle controls that were tested in each
of the following experiments to confirm that no effect was induced by the solvent (data
not shown).
2.4. Cytotoxicity Assays
Cell viability was evaluated by the 1-(4,5-dimethylthiazol-2-yl)-3,5-diphenylformazan
(MTT) test as reported by Morisi and collaborators [
18
]. Cells were seeded into 96-well
plates at density of 5
×
10
4
cells/well and left to attach overnight. Then, cells were treated
with MJe at various concentrations (from 0.001 to 1 mg/mL) for 1 h. As stressor, 6-OHDA
50
µ
M (Sigma–Aldrich, Milan, Italy) was added for additional 24 h [
17
]. Afterwards, the
plates were centrifuged, supernatants were removed, and fresh media without phenol red
containing 0.5 mg/mL of MTT (Sigma-Aldrich) was added to each well. Plates were put
in the incubator for an additional 4 h. Then, formazan crystals were solubilized in 100
µ
L
HCl/isopropanol 0.1 N. The absorbance was spectrophotometrically quantified through
iMark
microplate reader (Bio-Rad Laboratories, Milan, Italy) at a wavelength of 570 nm.
The viability was determined as percentage of viable cells in treated cultures compared to
those in untreated ones.
Cell death was assessed by the trypan blue (0.4% w/v; TB) exclusion test. Cells were
seeded onto 6-well plates at a density of 1
×
10
4
cells/well for 24 h and then treated
with MJe (0.001–1 mg/mL) for 1 h before exposure to 6-OHDA (50
µ
M) for another 24 h.
Then, cells were detached and stained with trypan blue dye before proceeding with cell
Antioxidants 2021,10, 539 4 of 16
count [
19
,
20
]. Dead cells were reported as percentage of stained (nonviable) vs. total
cells counted.
2.5. Detection of Apoptotic Cell Death and Caspase-3 Enzymatic Activity
The protective effect of MJe against 6-OHDA-induced cell death was analyzed by
fluorescence-activated cell sorting (FACS), using the Annexin V-fluorescein isothiocyanate
(FITC)/propidium iodide (PI) staining [
21
], a method employed to discriminate the living
status of cells.
SH-SY5Y were seeded similarly to TB assay, but pretreated for 1 h with MJe (0.1
and 0.5 mg/mL) and then exposed to 50
µ
M 6-OHDA for another 24 h. Next, cells were
collected and processed as already reported [
21
]. Finally, samples were run on a Novocyte
2000 cytofluorimeter (ACEA Biosciences Inc., San Diego, CA, USA).
Caspase 3 enzymatic activity was measured using a commercial kit (AbCam, Cam-
bridge, UK). SH-SY5Y cells were differentiated in 100 mm petri dishes (1.5
×
10
6
cells) and
then treated for 6 h, as explained above. Then, according to the manufacturer’s instructions,
the analysis was carried out on cell lysates [
17
]. Absorbance was measured at 405 nm by a
microplate spectrophotometer.
2.6. Real-Time PCR Analysis
For the evaluation of MJe treatment on the expression of genes encoding for regulatory
proteins of apoptosis, SH-SY5Y cells were seeded and treated as for the apoptosis evaluation.
Afterwards, total RNA of treated or untreated SH-SY5Y cells was extracted and reverse
transcribed following procedures of Curròand collaborators [
22
]. Messenger RNA levels
were analyzed by quantitative real-time PCR, using SYBR green as a fluorescent probe.
The analysis was carried out on a 7300 Real-Time PCR System (Applied Biosystems).
β
-
Actin was used as housekeeping control, and a standard dissociation stage was included
to assess primer specificity. Data were collected and analysed using the 2
∆∆CT
relative
quantification method [
22
]. Values are presented as fold change relative to untreated cells.
The primer sequences used for real-time PCR are listed in Table 1.
Table 1. Oligonucleotide primers used for real-time PCR.
Gene Product NCBI Reference Sequence Primer Sequence
p53 NM_000546.6 Forward: 50-GTGTGGAGTATTTGGATGAC-30
Reverse: 50-ATGTAGTTGTAGTGGATGGT-30
Bax NM_138764.5 Forward: 50-GGACGAACTGGACAGTAACATGG-30
Reverse: 50-GCAAAGTAGAAAAGGGCGACAAC-30
Bcl-2 NM_000657.3 Forward: 50-ATCGCCCTGTGGATGACTGAG-30
Reverse: 50-CAGCCAGGAGAAATCAAACAGAGG-30
SNCA NM_000345.4 Forward: 5’-TGACAAATGTTGGAGGAGCA-3’
Reverse: 5’-TGTCAGGATCCACAGGCATA-3’
LRRK2 NM_198578.4 Forward: 5’-TCAGCTTGTTGTTGGACAGC-3’
Reverse: 5’-ACTGCGTGAGGAAGCTCATT-3’
PINK1 NM_032409.3 Forward: 5’-ACGTTCAGTTACGGGAGTGG-3’
Reverse: 5’-GGCTAGTCAGGAGGGAAACC-3’
DJ-1 NM_007262.5 Forward: 5’-GGGTGCAGGCTTGTAAACAT-3’
Reverse: 5’-GGACAAATGACCACATCACG-3’
PARK2 NM_004562.3 Forward: 5’-CTGACACCAGCATCTTCCAG-3’
Reverse: 5’-CCAGTCATTCCTCAGCTCCT-3’
β-Actin NM_001101.5 Forward: 50-TTGTTACAGGAAGTCCCTTGCC-30
Reverse: 50-ATGCTATCACCTCCCCTGTGTG-30
Antioxidants 2021,10, 539 5 of 16
2.7. Evaluation of MJe Anti-Oxidant Activity through Abiotic Assays
The total phenolic content of MJe was evaluated through the Folin–Ciocalteu assay and
its total antioxidant activity through the oxygen radical absorbance capacity (ORAC) assay,
following Ferlazzo and co-workers [
23
]. The 2,2-Diphenyl-1-picrylhydrazyl (DPPH
) was
employed to test the radical scavenging activity of our extract, in accordance to Lombardo
and co-workers [
24
]. The reducing power of MJe was determined according to Ferlazzo
and collaborators through the potassium ferricyanide reducing power assay [
25
]. All tests
were repeated three times, and the results are expressed as means
±
SEM. As standards,
we employed gallic acid, Trolox, and ascorbic acid, in relation to the assay performed.
2.8. Measurement of Glutathione, Catalase and Superoxide Dismutase Activity
Catalase (CAT) and superoxide dismutase (SOD) activities and glutathione (GSH)
content were measured using commercial assay kits (AbCam). SH-SY5Y cells were plated
at density of 5
×
10
5
cells/well in 6-well plates and were treated with MJe 24 h later
at a concentration 0.1 and 0.5 mg/mL for 1 h. Then, 50
µ
M of 6-OHDA was added for
additional 24 h. Then, the assays were conducted according to the manufacturer’s protocols
on cell lysates. The absorbance was recorded by an iMark
microplate reader (Bio-Rad
Laboratories) at 450 nm for SOD, 570 nm for CAT, and 405 nm for GSH [25,26].
2.9. Determination of Reactive Oxygen Species (ROS) and Mitochondrial Membrane
Potential (∆Ψm)
Reactive oxygen species (ROS) and mitochondrial membrane potential (
∆Ψ
m) were
measured fluorometrically, as oxidative stress-related biomarkers. In both assays, cells
were seeded on 96-well plates (5
×
10
4
cells/well) and treated as explained for caspase 3
activity (see Section 2.5).
ROS were quantified using probe 2
0
,7
0
-dichlorodihydrofluorescein diacetate (DCFH-
DA) 25
µ
M (Sigma-Aldrich) [
23
] while variation of
∆Ψ
m were estimated by measuring
Rhodamine 123 incorporation (R123; Sigma-Aldrich), as reported by [
25
]. The fluorescence
was recorded by a microplate reader (POLARstar Omega, BMG Labtech, Ortenberg, Ger-
many) at 485 nm excitation and 535 nm emission for DCFH-DA and 488 nm excitation and
525 nm emission for R123.
2.10. Cytofluorimetric Evaluation of 8-oxo-dG
Oxidative DNA damage was assessed as levels of 8-Oxo-2’-deoxyguanosine (8-oxo-
dG) employing the FITC-labelled avidin probe, which is highly affine to 8-hydroxyguanine
(8-OH-Gua), being structurally similar to biotin. Briefly, cells were permeabilized with
methanol at
20
C for 15 min and incubated with avidin-FITC conjugate (0.2
µ
M) at
37
C for 1 h. The fluorescence was recorded cytofluorimetrically with Novocyte 2000
cytofluorimeter at 495 nm excitation and 520 nm emission (ACEA Biosciences Inc.) [23].
2.11. Determination of NO Accumulation in SH-SY5Y Culture Supernatant
The production of nitric oxide (NO) was assayed by a colorimetric commercial kit
(Sigma-Aldrich). In a 6-well plate, 5
×
10
5
cells/well were preincubated with MJe for 1 h,
and then treated with 50
µ
M 6-OHDA for 24 h. Supernatants were collected and processed
as described by Ferlazzo and colleagues [
17
]. Absorbance was recorded at a wavelength of
540 nm by an iMark™ microplate reader (Bio-Rad Laboratories).
2.12. Statistical Analyses
One-way analysis of variance (ANOVA) was used to interpret the data. Multiple
comparisons of the means of the groups were performed using the Tukey–Kramer test
(SigmaPlot Version 12.0, Systat Software, San Jose, CA, USA).
Antioxidants 2021,10, 539 6 of 16
3. Results
3.1. Chromatographic Analysis of MJe
The identification of the compounds present in the lyophilized powder was performed
by RP-DAD-HPLC. The preliminary inspection of chromatographic separation at 280 and
325 nm let us easily identify and discriminate the presence of flavanone and flavone basic
skeletons in the lyophilized powder. Both classes of flavonoids showed maxima of absorp-
tion (called band II) in the 240–280 nm range, but only flavones had a further well-defined
absorption maximum in the 300–380 nm range (called band I). The comparison, together
with inspection of UV spectra recorded in correspondence to each chromatographic peak,
showed that compounds 1–3 and 7–9 possessed a flavone skeleton, whereas 4–6 belonged
to the flavanone class (Figure 1). Moreover, the inspection of chromatogram recorded after
treatment with aqueous HCl (data not shown) indicated that compounds 1–3 were resistant
to acidic hydrolysis, whereas the remaining compounds were not resistant, suggesting the
presence of C-linked saccharide moieties in the former, while the latter were O-glycoside
compounds. Taking into consideration the retention time, UV spectra and spiking the
samples with pure reference compounds, the main peaks of the chromatogram have been
identified as vicenin-2 (1), lucenin-2 4
0
-methyl ether (2), orientin4
0
methylether (3), erioc-
itrin (4), narirutin (5), hesperidin (6), sinensetin (7), tangeretin (8), and nobiletin (9). The
flavonoid profile identified after the separation of compounds present in the lyophilized
powder was dominated by the presence of the flavanone hesperidin, which was by far the
most abundant component (about 353.6 mg/g), followed by a significant amount of di-
C-glucosyl flavone (6,8-di-C-glucosyl-apigenin, about 55.2 mg/g) and another rutinoside
(narirutin), although in smaller amount (about 47.8 mg/g). The quantitative analysis of all
the identified flavonoids is depicted in Table 2.
Antioxidants 2021, 10, x FOR PEER REVIEW 6 of 16
3. Results
3.1. Chromatographic Analysis of MJe
The identification of the compounds present in the lyophilized powder was per-
formed by RP-DAD-HPLC. The preliminary inspection of chromatographic separation at
280 and 325 nm let us easily identify and discriminate the presence of flavanone and fla-
vone basic skeletons in the lyophilized powder. Both classes of flavonoids showed max-
ima of absorption (called band II) in the 240–280 nm range, but only flavones had a further
well-defined absorption maximum in the 300–380 nm range (called band I). The compar-
ison, together with inspection of UV spectra recorded in correspondence to each chroma-
tographic peak, showed that compounds 1–3 and 7–9 possessed a flavone skeleton,
whereas 4–6 belonged to the flavanone class (Figure 1). Moreover, the inspection of chro-
matogram recorded after treatment with aqueous HCl (data not shown) indicated that
compounds 1–3 were resistant to acidic hydrolysis, whereas the remaining compounds
were not resistant, suggesting the presence of C-linked saccharide moieties in the former,
while the latter were O-glycoside compounds. Taking into consideration the retention
time, UV spectra and spiking the samples with pure reference compounds, the main peaks
of the chromatogram have been identified as vicenin-2 (1), lucenin-2 4-methyl ether (2),
orientin4methylether (3), eriocitrin (4), narirutin (5), hesperidin (6), sinensetin (7), tange-
retin (8), and nobiletin (9). The flavonoid profile identified after the separation of com-
pounds present in the lyophilized powder was dominated by the presence of the fla-
vanone hesperidin, which was by far the most abundant component (about 353.6 mg/g),
followed by a significant amount of di-C-glucosyl flavone (6,8-di-C-glucosyl-apigenin,
about 55.2 mg/g) and another rutinoside (narirutin), although in smaller amount (about
47.8 mg/g). The quantitative analysis of all the identified flavonoids is depicted in Table
2.
Figure 1. Reverse phase-diode array detector-high performance liquid chromatography (RP-
HPLC-DAD) separation of the compounds present in the lyophilized powder registered at 280 (A)
Figure 1.
Reverse phase-diode array detector-high performance liquid chromatography (RP-HPLC-
DAD) separation of the compounds present in the lyophilized powder registered at 280 (
A
) and
325 nm (
B
). Vicenin-2 (1), lucenin-2 4
0
-methyl ether (2), orientin4
0
methylether (3), eriocitrin (4),
narirutin (5), hesperidin (6), sinensetin (7), tangeretin (8), nobiletin (9). Peak identification was
performed by matching retention time and UV spectra against commercially available reference
compounds and spiking the samples with pure reference compounds.
Antioxidants 2021,10, 539 7 of 16
Table 2. Quantitative determination of the identified compounds.
mg/g (Dried Extract)
Peak Compounds Mean SD
1 Vicenin-2 55.2 2.5
2 Lucenin-2 40-methyl ether 21.5 1.7
3 Orientin40methylether 1.4 0.21
4 Eriocitrin 8.2 1.6
5 Narirutin 47.8 1.9
6 Hesperidin 353.6 20.5
7 Sinensetin 2.0 0.3
8 Tangeretin 6.4 0.5
9 Nobiletin 17.3 1.4
3.2. MJe Prevents 6-OHDA Induced SH-SY5Y Cell Death
To evaluate the potential neuroprotective effect of MJe, SH-SY5Y cells were preincu-
bated with MJe and then exposed to 6-OHDA before the assessment of cell viability. The
incubation of SH-SY5Y cells with 6-OHDA reduced cell viability by 37
±
5.1% compared to
that of control cells (p< 0.001; Figure 2A), whereas pretreatment with MJe at concentration
of 0.1, 0.5 and 1 mg/mL restored cell viability up to 77
±
6.1, 83
±
6.9, and
85 ±7.9%
,
respectively (p< 0.05 and p< 0.01 vs. 6-OHDA-injured cells; Figure 2A). Data of TB
assay followed the same pattern of those of MTT test (Figure 2B). In particular, 6-OHDA
increased cell death by 28
±
2.5%, whereas 0.01, 0.1, 0.5, and 1 mg/mL MJe maintained it
to 23.1 ±1.3, 19.4 ±1.2, 15.9 ±2.6, and 14.4 ±2.0%, respectively (p< 0.05 and p< 0.01 vs.
6-OHDA-exposed cells; Figure 2B).
Antioxidants 2021, 10, x FOR PEER REVIEW 7 of 16
and 325 nm (B). Vicenin-2 (1), lucenin-2 4-methyl ether (2), orientin4methylether (3), eriocitrin (4),
narirutin (5), hesperidin (6), sinensetin (7), tangeretin (8), nobiletin (9). Peak identification was
performed by matching retention time and UV spectra against commercially available reference
compounds and spiking the samples with pure reference compounds.
Table 2. Quantitative determination of the identified compounds.
mg/g (Dried Extract)
Peak Compounds Mean SD
1 Vicenin-2 55.2 2.5
2 Lucenin-2 4-methyl ether 21.5 1.7
3 Orientin4methylether 1.4 0.21
4 Eriocitrin 8.2 1.6
5 Narirutin 47.8 1.9
6 Hesperidin 353.6 20.5
7 Sinensetin 2.0 0.3
8 Tangeretin 6.4 0.5
9 Nobiletin 17.3 1.4
3.2. MJe Prevents 6-OHDA Induced SH-SY5Y Cell Death
To evaluate the potential neuroprotective effect of MJe, SH-SY5Y cells were preincu-
bated with MJe and then exposed to 6-OHDA before the assessment of cell viability. The
incubation of SH-SY5Y cells with 6-OHDA reduced cell viability by 37 ± 5.1% compared
to that of control cells (p < 0.001; Figure 2A), whereas pretreatment with MJe at concentra-
tion of 0.1, 0.5 and 1 mg/mL restored cell viability up to 77 ± 6.1, 83 ± 6.9, and 85 ± 7.9%,
respectively (p < 0.05 and p < 0.01 vs. 6-OHDA-injured cells; Figure 2A). Data of TB assay
followed the same pattern of those of MTT test (Figure 2B). In particular, 6-OHDA in-
creased cell death by 28 ± 2.5%, whereas 0.01, 0.1, 0.5, and 1 mg/mL MJe maintained it to
23.1 ± 1.3, 19.4 ± 1.2, 15.9 ± 2.6, and 14.4 ± 2.0%, respectively (p < 0.05 and p < 0.01 vs. 6-
OHDA-exposed cells; Figure 2B).
Figure 2. Protective effect of mandarin juice extract (MJe) against 6-hydroxydopamine (6-OHDA)-
induced cytotoxicity. Cell viability was detected with the 1-(4,5-dimethylthiazol-2-yl)-3,5-diphe-
nylformazan (MTT) assay (A). The viability variation was calculated as percentage of viable cells
in treated cultures relative to those in untreated ones. Cell death was estimated by the trypan blue
(TB) assay and expressed as percentage of nonviable (blue stained) cells relative to total counted
cells (B). Results are displayed as mean ± standard error of the means (SEM) from three independ-
ent experiments in eight replicates (MTT; N = 24) or in triplicate (TB; N = 9). *** p < 0.001 vs. ctrl; ° p
< 0.05, °° p < 0.01 and °°° p < 0.001 vs. 6-OHDA 50 μM.
3.3. MJe Reduces the Apoptotic Cell Death Induced by 6-OHDA
Figure 2.
Protective effect of mandarin juice extract (MJe) against 6-hydroxydopamine (6-OHDA)-induced cytotoxicity. Cell
viability was detected with the 1-(4,5-dimethylthiazol-2-yl)-3,5-diphenylformazan (MTT) assay (
A
). The viability variation
was calculated as percentage of viable cells in treated cultures relative to those in untreated ones. Cell death was estimated
by the trypan blue (TB) assay and expressed as percentage of nonviable (blue stained) cells relative to total counted cells (
B
).
Results are displayed as mean
±
standard error of the means (SEM) from three independent experiments in eight replicates
(MTT; N = 24) or in triplicate (TB; N = 9). *** p< 0.001 vs. ctrl; p< 0.05, ◦◦ p< 0.01 and p< 0.001 vs. 6-OHDA 50 µM.
3.3. MJe Reduces the Apoptotic Cell Death Induced by 6-OHDA
The cytoprotecting activity elicited by MJe was also evaluated by flow cytometry
through an Annexin V-FITC/PI assay. As shown in Figure 3A,B, the incubation of SH-
SY5Y cells with 6-OHDA for 24 h augmented the percentage of cells in early (8
±
0.3%,
Annexin V+/PI
) and late (28
±
2.0%, Annexin V+/PI+) apoptosis, as well as in necrosis
(
16 ±1.1%,
Annexin V
/PI+). The pretreatment with 0.1 mg/mL MJe for 1 h reduced the
number of cells undergoing apoptosis (4
±
0.3% and 13
±
0.7% of early and late apoptosis,
respectively), likewise, in presence of MJe 0.5 mg/mL, it was further reduced to 5
±
0.2%
and 8
±
0.1%, respectively (Figure 3A,B). The incidence of apoptosis in 6-OHDA-treated
Antioxidants 2021,10, 539 8 of 16
cells was confirmed by the results of the caspase 3 activity assay. The exposure to 6-
OHDA for 6 h increased caspase 3 activity of SH-SY5Y cells compared to that of unexposed
ones by 250
±
15% (p< 0.001; Figure 3C). Pretreatment with MJe (0.1 or 0.5 mg/mL) for
1 h inhibited the activity of caspase 3 induced by 6-OHDA to 140
±
9 and 123
±
10%,
respectively (
p< 0.001
), while MJe alone, at both tested concentrations, had no effect on
this enzymatic activity (Figure 3C).
Antioxidants 2021, 10, x FOR PEER REVIEW 8 of 16
The cytoprotecting activity elicited by MJe was also evaluated by flow cytometry
through an Annexin V-FITC/PI assay. As shown in Figure 3A,B, the incubation of SH-
SY5Y cells with 6-OHDA for 24 h augmented the percentage of cells in early (8 ± 0.3%,
Annexin V+/PI) and late (28 ± 2.0%, Annexin V+/PI+) apoptosis, as well as in necrosis (16
± 1.1%, Annexin V/PI+). The pretreatment with 0.1 mg/mL MJe for 1 h reduced the num-
ber of cells undergoing apoptosis (4 ± 0.3% and 13 ± 0.7% of early and late apoptosis, re-
spectively), likewise, in presence of MJe 0.5 mg/mL, it was further reduced to 5 ± 0.2% and
8 ± 0.1%, respectively (Figure 3A,B). The incidence of apoptosis in 6-OHDA-treated cells
was confirmed by the results of the caspase 3 activity assay. The exposure to 6-OHDA for
6 h increased caspase 3 activity of SH-SY5Y cells compared to that of unexposed ones by
250 ± 15% (p < 0.001; Figure 3C). Pretreatment with MJe (0.1 or 0.5 mg/mL) for 1 h inhibited
the activity of caspase 3 induced by 6-OHDA to 140 ± 9 and 123 ± 10%, respectively (p <
0.001), while MJe alone, at both tested concentrations, had no effect on this enzymatic ac-
tivity (Figure 3C).
Figure 3. MJe mitigated the apoptosis induced by 6-OHDA in SH-SY5Y cells. (A) Evaluation of
apoptosis was performed cytofluorimetrically by the Annexin V/PI test. Representative Annexin V
vs. PI dot plots are shown, where necrotic, late apoptosis, viable cells, and early apoptosis cells are
in Q1, Q2, Q3 and Q4, respectively. (B) The histogram shows the percentage of cells in each quad-
rant, representing the mean ± SEM of three different experiments in triplicate (N = 9). (C) Data of
caspase 3 activity are presented as the mean of three experiments ± SEM in triplicate (N = 9). °°° p <
0.001 vs. control; *** p < 0.001 vs. 6-OHDA 50 μM.
3.4. Protective Effect of MJe on mRNA Levels of Apoptosis-Related Genes Modulated by 6-
OHDA
As shown in Figure 4, the incubation of SH-SY5Y cells with 6-OHDA for 24 h signif-
icantly enhanced the mRNA levels of the proapoptotic B-cell lymphoma 2 (Bcl-2)-associ-
ated X protein (Bax) and p53 genes up to 1.8 ± 0.2- and 2.2 ± 0.3-fold, respectively (p < 0.01
and p < 0.001), along with decreasing those of the antiapoptotic Bcl-2 up to 3 ± 0.9-fold (p
< 0.01). These effects were significantly hampered by pre-exposure to MJe at both 0.1 (1.35
± 0.2-fold and 1.55 ± 0.1-fold down, p < 0.05, for Bax and Bcl-2, and 1.2 ± 0.1-fold, p < 0.01,
for p53) and 0.5 mg/mL concentrations (1.25 ± 0.1-fold, p < 0.05, for Bax and 1.25 ± 0.1-fold
down and 1.4 ± 0.2-fold, p < 0.01 for Bcl-2 and p53) (Figure 4).
Figure 3.
MJe mitigated the apoptosis induced by 6-OHDA in SH-SY5Y cells. (
A
) Evaluation of apoptosis was performed
cytofluorimetrically by the Annexin V/PI test. Representative Annexin V vs. PI dot plots are shown, where necrotic, late
apoptosis, viable cells, and early apoptosis cells are in Q1, Q2, Q3 and Q4, respectively. (
B
) The histogram shows the
percentage of cells in each quadrant, representing the mean
±
SEM of three different experiments in triplicate (N = 9). (
C
)
Data of caspase 3 activity are presented as the mean of three experiments
±
SEM in triplicate (N = 9).
◦◦
p< 0.001 vs.
control; *** p< 0.001 vs. 6-OHDA 50 µM.
3.4. Protective Effect of MJe on mRNA Levels of Apoptosis-Related Genes Modulated by 6-OHDA
As shown in Figure 4, the incubation of SH-SY5Y cells with 6-OHDA for 24 h signifi-
cantly enhanced the mRNA levels of the proapoptotic B-cell lymphoma 2 (Bcl-2)-associated
X protein (Bax) and p53 genes up to 1.8
±
0.2- and 2.2
±
0.3-fold, respectively (p< 0.01
and p< 0.001), along with decreasing those of the antiapoptotic Bcl-2 up to 3
±
0.9-fold
(p< 0.01). These effects were significantly hampered by pre-exposure to MJe at both 0.1
(1.35
±
0.2-fold and 1.55
±
0.1-fold down, p< 0.05, for Bax and Bcl-2, and 1.2
±
0.1-fold,
p< 0.01, for p53) and 0.5 mg/mL concentrations (1.25
±
0.1-fold, p< 0.05, for Bax and
1.25 ±0.1-fold down and 1.4 ±0.2-fold, p< 0.01 for Bcl-2 and p53) (Figure 4).
Antioxidants 2021,10, 539 9 of 16
Antioxidants 2021, 10, x FOR PEER REVIEW 9 of 16
Figure 4. Protective effect of MJe on mRNA levels of apoptosis-related genes modulated by 6-
OHDA. SH-SY5Y cells were pretreated with MJe for 1 h and then exposed to 6-OHDA 50 μM for
additional 24 h and then assessed with real-time PCR. The 2
ΔΔCT
method was employed to calcu-
late the relative quantities of mRNA. Results are expressed as fold change relative to untreated
cells. Data, expressed as mean ± SEM, represent the values obtained in three different sets of ex-
periments in triplicate (N = 9). ** p < 0.01, *** p < 0.001 vs. control; ° p < 0.05, °° p < 0.01 vs. 6-OHDA
50 μM.
3.5. Antioxidant Activity of MJe
The antioxidant and radical scavenging properties of MJe were demonstrated using
a range of tests as described in the Methods section. As shown in Table 3, the total phenolic
content expressed as milligrams of gallic acid equivalents (GAE) per gram of MJe evalu-
ated by FolinCiocalteau method was 117.76 ± 4.8. The valuable antiradical activity of MJe
shown in the DPPH test and expressed as milligrams of Trolox equivalents (TE) per gram
of extract (mgTE/g) was 60.07 ± 4.2, while in the Reducing Power test it was 53.6 ± 2.3
expressed as milligrams of ascorbic acid equivalent (AAE) per gram of MJe (mg AAE/g).
Moreover, the obtained values of 3753.7 ± 221.5 μmol TE/g MJe demonstrated high anti-
oxidant capacity of MJe against peroxyl radicals (Table 3).
Table 3. Antioxidant activity of MJe evaluated by abiotic assays. Results are reported as mean ±
SEM of three experiments performed in triplicate and expressed in standard equivalent/g of dried
extract (N = 9).
Folin-Ciocalteau (mg GAE/g) 117.76 ± 4.8
DPPH (mg TE/g) 60.07 ± 4.2
Reducing Power (mg AAE/g) 53.6 ± 2.3
ORAC (μmol TE/g) 3753.72 ± 221.5
3.6. Effects of MJe on 6-OHDA-Induced SOD and CAT Activities and GSH Content
We evaluated the levels of catalase and superoxide dismutase activities as well as
glutathione content to determine the extent of oxidative stress in the cells. As shown in
Figure 5, the treatment of SH-SY5Y cells with 6-OHDA caused a significant decrease in
the levels of these biomarkers in comparison to those of control cells (62 ± 2.3, 41 ± 1.7, and
53 ± 2.1% for SOD, CAT, and GSH, respectively, p < 0.001). Pretreatment with MJe at both
concentrations of 0.1 (62 ± 2.3, 41 ± 1.7, and 53 ± 2.1% for SOD, CAT, and GSH, respectively,
p < 0.01) and 0.5 mg/mL (88 ± 2.8, 65 ± 2.2, and 79 ± 2.0% for SOD, CAT, and GSH, respec-
tively, p < 0.001) for 1 h significantly augmented the activity of both CAT and SOD, along
with levels of GSH, indicative of a reduction of oxidative stress (Figure 5).
Figure 4.
Protective effect of MJe on mRNA levels of apoptosis-related genes modulated by 6-OHDA.
SH-SY5Y cells were pretreated with MJe for 1 h and then exposed to 6-OHDA 50
µ
M for additional
24 h and then assessed with real-time PCR. The 2
∆∆CT
method was employed to calculate the relative
quantities of mRNA. Results are expressed as fold change relative to untreated cells. Data, expressed
as mean
±
SEM, represent the values obtained in three different sets of experiments in triplicate
(N = 9). ** p< 0.01, *** p< 0.001 vs. control; p< 0.05, p< 0.01 vs. 6-OHDA 50 µM.
3.5. Antioxidant Activity of MJe
The antioxidant and radical scavenging properties of MJe were demonstrated using a
range of tests as described in the Methods section. As shown in Table 3, the total phenolic
content expressed as milligrams of gallic acid equivalents (GAE) per gram of MJe evaluated
by Folin–Ciocalteau method was 117.76
±
4.8. The valuable antiradical activity of MJe
shown in the DPPH test and expressed as milligrams of Trolox equivalents (TE) per gram
of extract (mgTE/g) was 60.07
±
4.2, while in the Reducing Power test it was 53.6
±
2.3
expressed as milligrams of ascorbic acid equivalent (AAE) per gram of MJe (mg AAE/g).
Moreover, the obtained values of 3753.7
±
221.5
µ
mol TE/g MJe demonstrated high
antioxidant capacity of MJe against peroxyl radicals (Table 3).
Table 3.
Antioxidant activity of MJe evaluated by abiotic assays. Results are reported as
mean ±SEM
of three experiments performed in triplicate and expressed in standard equivalent/g of dried extract
(N = 9).
Folin-Ciocalteau (mg GAE/g) 117.76 ±4.8
DPPH (mg TE/g) 60.07 ±4.2
Reducing Power (mg AAE/g) 53.6 ±2.3
ORAC (µmol TE/g) 3753.72 ±221.5
3.6. Effects of MJe on 6-OHDA-Induced SOD and CAT Activities and GSH Content
We evaluated the levels of catalase and superoxide dismutase activities as well as
glutathione content to determine the extent of oxidative stress in the cells. As shown in
Figure 5, the treatment of SH-SY5Y cells with 6-OHDA caused a significant decrease in
the levels of these biomarkers in comparison to those of control cells (62
±
2.3, 41
±
1.7,
and 53
±
2.1% for SOD, CAT, and GSH, respectively, p< 0.001). Pretreatment with MJe
at both concentrations of 0.1 (62
±
2.3, 41
±
1.7, and 53
±
2.1% for SOD, CAT, and GSH,
respectively, p< 0.01) and 0.5 mg/mL (88
±
2.8, 65
±
2.2, and 79
±
2.0% for SOD, CAT, and
GSH, respectively, p< 0.001) for 1 h significantly augmented the activity of both CAT and
SOD, along with levels of GSH, indicative of a reduction of oxidative stress (Figure 5).
Antioxidants 2021,10, 539 10 of 16
Antioxidants 2021, 10, x FOR PEER REVIEW 10 of 16
Figure 5. Effect of MJe on biomarkers of 6-OHDA-induced oxidative stress in SH-SY5Y cells. (A)
Effect of MJe on the activity of SOD in 6-OHDA-treated SH-SY5Y cells; (B) Effect of MJe on the
activity of CAT in 6-OHDA-treated SH-SY5Y cells; (C) Effect of MJe on the GSH levels in 6-
OHDA-treated SH-SY5Y cells. Data are showed as the mean ± SEM of three experiments in tripli-
cate (N = 9). *** p < 0.001 vs. ctrl; °° p < 0.01 and °°° p < 0.001 vs. 6-OHDA 50 μM.
3.7. MJe Reduces Oxidative Stress Induced by 6-OHDA
The increase of ROS is an acknowledged characteristic of several NDs, including PD.
Therefore, we measured the intracellular ROS amount through DCFH-DA. As shown in
Figure 6A, the exposure of SH-SY5Y cells to 50 μM 6-OHDA for 6 h caused a significant 2
± 0.15-fold intracellular ROS accumulation compared to that in the controls (p < 0.001;
Figure 6A). Pretreatment with MJe at both 0.1 and 0.5 mg/mL concentrations significantly
counteracted the increase in ROS caused by 6-OHDA, by 51 ± 0.2 and 63 ± 0.1%, respec-
tively (p < 0.001; Figure 6A). Moreover, the exposure of SH-SY5Y cells to 50 μM 6-OHDA
for 6 h significantly influenced the ΔΨm that, in comparison to that in control cells, de-
creased by 39 ± 0.6% (p < 0.01). The reduction of ΔΨm evoked by 6-OHDA was prevented
by MJe 0.1 and 0.5 mg/mL (81.3 ± 8 and 86.7 ± 7% p < 0.05, respectively; Figure 6B).
Figure 6. MJe diminished both generation of reactive oxygen species (ROS) and fall of mitochon-
drial membrane potential (ΔΨm) induced by 6-OHDA. (A) ROS accumulation was measured us-
ing the fluorescent probe DCFH-DA. (B) ΔΨm was assessed using the cationic fluorochrome R123.
Results are reported as percentage of the levels detected in untreated cells. Data are displayed as
the mean ± SEM of three experiments in triplicate (N = 9). °° p < 0.01 and °°° p < 0.001 vs. ctrl; * p <
0.05 and *** p < 0.001 vs. 6-OHDA 50 μM.
3.8. Protective Effects of MJe on 6-OHDA DNA Damage
Levels of 8-oxo-dG were measured to study the efficacy of MJe in containing DNA-
oxidative damage using a FITC-conjugated avidin probe (Figure 7). The exposure of SH-
SY5Y cells to 6-OHDA 50 μM for 24 h induced DNA oxidation of about 50 ± 2% in com-
parison to that in control cells. Pretreatment with 0.1 and 0.5 mg/mL of MJe for 1 h before
the exposure to 6-OHDA 50 μM (24 h) decreased oxidative DNA damage by 27 ± 1 and 33
Figure 5.
Effect of MJe on biomarkers of 6-OHDA-induced oxidative stress in SH-SY5Y cells. (
A
) Effect of MJe on the
activity of SOD in 6-OHDA-treated SH-SY5Y cells; (
B
) Effect of MJe on the activity of CAT in 6-OHDA-treated SH-SY5Y
cells; (
C
) Effect of MJe on the GSH levels in 6-OHDA-treated SH-SY5Y cells. Data are showed as the mean
±
SEM of three
experiments in triplicate (N = 9). *** p< 0.001 vs. ctrl; p< 0.01 and ◦◦ p< 0.001 vs. 6-OHDA 50 µM.
3.7. MJe Reduces Oxidative Stress Induced by 6-OHDA
The increase of ROS is an acknowledged characteristic of several NDs, including PD.
Therefore, we measured the intracellular ROS amount through DCFH-DA. As shown in
Figure 6A, the exposure of SH-SY5Y cells to 50
µ
M 6-OHDA for 6 h caused a significant
2±0.15-fold
intracellular ROS accumulation compared to that in the controls (p< 0.001;
Figure 6A). Pretreatment with MJe at both 0.1 and 0.5 mg/mL concentrations significantly
counteracted the increase in ROS caused by 6-OHDA, by 51
±
0.2 and 63
±
0.1%, respec-
tively (p< 0.001; Figure 6A). Moreover, the exposure of SH-SY5Y cells to 50
µ
M 6-OHDA for
6 h significantly influenced the
∆Ψ
m that, in comparison to that in control cells, decreased
by 39
±
0.6% (p< 0.01). The reduction of
∆Ψ
m evoked by 6-OHDA was prevented by MJe
0.1 and 0.5 mg/mL (81.3 ±8 and 86.7 ±7% p< 0.05, respectively; Figure 6B).
Antioxidants 2021, 10, x FOR PEER REVIEW 10 of 16
Figure 5. Effect of MJe on biomarkers of 6-OHDA-induced oxidative stress in SH-SY5Y cells. (A)
Effect of MJe on the activity of SOD in 6-OHDA-treated SH-SY5Y cells; (B) Effect of MJe on the
activity of CAT in 6-OHDA-treated SH-SY5Y cells; (C) Effect of MJe on the GSH levels in 6-
OHDA-treated SH-SY5Y cells. Data are showed as the mean ± SEM of three experiments in tripli-
cate (N = 9). *** p < 0.001 vs. ctrl; °° p < 0.01 and °°° p < 0.001 vs. 6-OHDA 50 μM.
3.7. MJe Reduces Oxidative Stress Induced by 6-OHDA
The increase of ROS is an acknowledged characteristic of several NDs, including PD.
Therefore, we measured the intracellular ROS amount through DCFH-DA. As shown in
Figure 6A, the exposure of SH-SY5Y cells to 50 μM 6-OHDA for 6 h caused a significant 2
± 0.15-fold intracellular ROS accumulation compared to that in the controls (p < 0.001;
Figure 6A). Pretreatment with MJe at both 0.1 and 0.5 mg/mL concentrations significantly
counteracted the increase in ROS caused by 6-OHDA, by 51 ± 0.2 and 63 ± 0.1%, respec-
tively (p < 0.001; Figure 6A). Moreover, the exposure of SH-SY5Y cells to 50 μM 6-OHDA
for 6 h significantly influenced the ΔΨm that, in comparison to that in control cells, de-
creased by 39 ± 0.6% (p < 0.01). The reduction of ΔΨm evoked by 6-OHDA was prevented
by MJe 0.1 and 0.5 mg/mL (81.3 ± 8 and 86.7 ± 7% p < 0.05, respectively; Figure 6B).
Figure 6. MJe diminished both generation of reactive oxygen species (ROS) and fall of mitochon-
drial membrane potential (ΔΨm) induced by 6-OHDA. (A) ROS accumulation was measured us-
ing the fluorescent probe DCFH-DA. (B) ΔΨm was assessed using the cationic fluorochrome R123.
Results are reported as percentage of the levels detected in untreated cells. Data are displayed as
the mean ± SEM of three experiments in triplicate (N = 9). °° p < 0.01 and °°° p < 0.001 vs. ctrl; * p <
0.05 and *** p < 0.001 vs. 6-OHDA 50 μM.
3.8. Protective Effects of MJe on 6-OHDA DNA Damage
Levels of 8-oxo-dG were measured to study the efficacy of MJe in containing DNA-
oxidative damage using a FITC-conjugated avidin probe (Figure 7). The exposure of SH-
SY5Y cells to 6-OHDA 50 μM for 24 h induced DNA oxidation of about 50 ± 2% in com-
parison to that in control cells. Pretreatment with 0.1 and 0.5 mg/mL of MJe for 1 h before
the exposure to 6-OHDA 50 μM (24 h) decreased oxidative DNA damage by 27 ± 1 and 33
Figure 6. MJe diminished both generation of reactive oxygen species (ROS) and fall of mitochondrial membrane potential
(
∆Ψ
m) induced by 6-OHDA. (
A
) ROS accumulation was measured using the fluorescent probe DCFH-DA. (
B) ∆Ψ
m was
assessed using the cationic fluorochrome R123. Results are reported as percentage of the levels detected in untreated cells.
Data are displayed as the mean
±
SEM of three experiments in triplicate (N = 9).
◦◦
p< 0.01 and
◦◦
p< 0.001 vs. ctrl;
*p< 0.05 and *** p< 0.001 vs. 6-OHDA 50 µM.
3.8. Protective Effects of MJe on 6-OHDA DNA Damage
Levels of 8-oxo-dG were measured to study the efficacy of MJe in containing DNA-
oxidative damage using a FITC-conjugated avidin probe (Figure 7). The exposure of
SH-SY5Y cells to 6-OHDA 50
µ
M for 24 h induced DNA oxidation of about 50
±
2% in
comparison to that in control cells. Pretreatment with 0.1 and 0.5 mg/mL of MJe for 1 h
before the exposure to 6-OHDA 50
µ
M (24 h) decreased oxidative DNA damage by 27
±
1
and 33
±
1% relative to stressed cells, respectively (Figure 7). The treatment with MJe alone
Antioxidants 2021,10, 539 11 of 16
at both concentrations tested did not induce DNA oxidation, since the emission values
approximately overlapped those recorded in control cells (data not shown).
Figure 7.
Protective effects of MJe on DNA oxidative damage induced by 6-OHDA. Levels of 8-oxo-dG are measured as
emission signals of fluorochrome FITC-labelled avidin. The plots are representative of three independent experiments. The
histograms show the percentage
±
SEM of healthy cells (non-fluorescent, M1) and the damaged ones (fluorescent, M2) of
three separate experiments in triplicate (N = 9).
3.9. MJe Reduces the Production of NO in SH-SY5Y Cells
The exposure of differentiated SH-SY5Y cells for 24 h to 6-OHDA brought a 79
±
5%
increase of NO production (p< 0.01; Figure 8) that was hindered by pretreatment with MJe
for 1 h at both concentrations tested (47
±
2.5 and 59
±
2% lower, p< 0.01, compared to
that in 6-OHDA-treated cells; Figure 8), while MJe alone had no effect on NO production
(data not shown).
Antioxidants 2021, 10, x FOR PEER REVIEW 11 of 16
± 1% relative to stressed cells, respectively (Figure 7). The treatment with MJe alone at
both concentrations tested did not induce DNA oxidation, since the emission values ap-
proximately overlapped those recorded in control cells (data not shown).
Figure 7. Protective effects of MJe on DNA oxidative damage induced by 6-OHDA. Levels of 8-
oxo-dG are measured as emission signals of fluorochrome FITC-labelled avidin. The plots are rep-
resentative of three independent experiments. The histograms show the percentage ± SEM of
healthy cells (non-fluorescent, M1) and the damaged ones (fluorescent, M2) of three separate ex-
periments in triplicate (N = 9).
3.9. MJe Reduces the Production of NO in SH-SY5Y Cells
The exposure of differentiated SH-SY5Y cells for 24 h to 6-OHDA brought a 79 ± 5%
increase of NO production (p < 0.01; Figure 8) that was hindered by pretreatment with
MJe for 1 h at both concentrations tested (47 ± 2.5 and 59 ± 2% lower, p < 0.01, compared
to that in 6-OHDA-treated cells; Figure 8), while MJe alone had no effect on NO produc-
tion (data not shown).
Figure 8. MJe prevented the release of NO induced by 6-OHDA. The levels of NO were measured
by a colorimetric assay. Differences in NO production are reported as percentage of NO value
detected in treated cells compared to those found in untreated ones. Results are expressed as
means ± SEM from three independent experiments in triplicate (N = 9). °° p < 0.01 vs. ctrl; ** p <
0.01 vs. 6-OHDA 50 μM.
Figure 8.
MJe prevented the release of NO induced by 6-OHDA. The levels of NO were measured by
a colorimetric assay. Differences in NO production are reported as percentage of NO value detected
in treated cells compared to those found in untreated ones. Results are expressed as means
±
SEM
from three independent experiments in triplicate (N = 9).
◦◦
p< 0.01 vs. ctrl; ** p< 0.01 vs. 6-OHDA
50 µM.
Antioxidants 2021,10, 539 12 of 16
3.10. Protective Effect of MJe on mRNA Levels of Parkinson-Related Genes Modulated
by 6-OHDA
As shown in Figure 9, the exposure of SH-SY5Y cells to 50
µ
M 6-OHDA for 24 h
significantly enhanced the levels of
α
-synuclein (SNCA) and leucine-rich repeat kinase
2 (LRRK2) genes up to 2.9
±
0.2- and 2.3
±
0.1-fold, respectively (p< 0.001), as well as
decreased those of phosphatase and tensin homolog (PTEN)-induced putative kinase 1
(PINK1), DJ-1, and parkin (PARK2) up to 2.2
±
0.15, 1.6
±
0.1 and 3
±
0.04-fold down,
respectively (p< 0.01). SNCA, LRRK2, and PARK2 were significantly modulated by the pre-
exposure to MJe at both 0.1 (1.3
±
0.15-fold down p< 0.01 for SNCA, 1.3
±
0.1-fold down,
p< 0.001 for LRRK2 and 2.7
±
0.1-fold p< 0.05 for PARK2, relative to 6-OHDA-stressed
cells) and 0.5 mg/mL concentrations (1.8
±
0.1-fold down for both SNCA and LRRK2,
and 3.7
±
0.3-fold for PARK2, p< 0.001, respect to 6-OHDA-stressed cells)
(Figure 9)
.
Concerning PINK-1 and DJ-1 genes, instead, only the highest concentration of MJe was
able to counteract the effect of 6-OHDA (2.0
±
0.1-fold p< 0.05 for PINK1, 1.9
±
0.2-fold
p< 0.01 for DJ-1, relative to 6-OHDA-stressed cells).
Antioxidants 2021, 10, x FOR PEER REVIEW 12 of 16
3.10. Protective Effect of MJe on mRNA Levels of Parkinson-Related Genes Modulated by 6-
OHDA
As shown in Figure 9, the exposure of SH-SY5Y cells to 50 μM 6-OHDA for 24 h
significantly enhanced the levels of α-synuclein (SNCA) and leucine-rich repeat kinase 2
(LRRK2) genes up to 2.9 ± 0.2- and 2.3 ± 0.1-fold, respectively (p < 0.001), as well as de-
creased those of phosphatase and tensin homolog (PTEN)-induced putative kinase 1
(PINK1), DJ-1, and parkin (PARK2) up to 2.2 ± 0.15, 1.6 ± 0.1 and 3 ± 0.04-fold down, re-
spectively (p < 0.01). SNCA, LRRK2, and PARK2 were significantly modulated by the pre-
exposure to MJe at both 0.1 (1.3 ± 0.15-fold down p < 0.01 for SNCA, 1.3 ± 0.1-fold down,
p < 0.001 for LRRK2 and 2.7 ± 0.1-fold p < 0.05 for PARK2, relative to 6-OHDA-stressed
cells) and 0.5 mg/mL concentrations (1.8 ± 0.1-fold down for both SNCA and LRRK2, and
3.7 ± 0.3-fold for PARK2, p < 0.001, respect to 6-OHDA-stressed cells) (Figure 9). Concern-
ing PINK-1 and DJ-1 genes, instead, only the highest concentration of MJe was able to
counteract the effect of 6-OHDA (2.0 ± 0.1-fold p < 0.05 for PINK1, 1.9 ± 0.2-fold p < 0.01
for DJ-1, relative to 6-OHDA-stressed cells).
Figure 9. Modulatory effects of MJe on mRNA levels of Parkinson-related genes modulated by 6-
OHDA. SH-SY5Y cells were pretreated with MJe for 1 h and then exposed to 6-OHDA 50 μM for
additional 24 h. Messenger RNA levels were quantified with real-time PCR, and their relative
quantities were calculated through the 2
ΔΔCT
method. Results are expressed as fold change relative
to untreated cells. Data are expressed as mean ± SEM of three separate experiments in triplicate (N
= 9). * p < 0.05, *** p < 0.001 vs. control; ° p < 0.05, °° p < 0.01 and °°° p < 0.001 vs. 6-OHDA 50 μM.
4. Discussion
Citrus reticulata (mandarin), originated from Southeast China, is present in Europe
under a multitude of varieties and has been studied mainly for its anticancer properties
in different in vitro and in vivo models [10]. In these regards, we showed that MJe induces
antiproliferative activity in three different anaplastic thyroid carcinoma cell lines, block-
ing cell cycle in G2/M phase and inducing autophagy, as well as reducing cell migration
and affecting metalloproteinase activity [27].
To the best of our knowledge, this study is the first to assess the neuroprotective ef-
fect of MJe in 6-OHDA-stressed SH-SY5Y cells, a widely employed cell line to mimic the
cellular PD’s environment [28]. Among the most recognized PD models, we chose the 6-
OHDA, a neurotoxin inducing depletion of dopaminergic neurons in the substantia nigra
pars compacta, given the scientific evidence, both in vitro [29] ad in vivo [30,31], support-
ing its use.
First, we showed that MJe was able to defend cell viability from damage induced by
6-OHDA, as shown by MTT and trypan blue assays. Moreover, we found that our stressor
Figure 9.
Modulatory effects of MJe on mRNA levels of Parkinson-related genes modulated by
6-OHDA. SH-SY5Y cells were pretreated with MJe for 1 h and then exposed to 6-OHDA 50
µ
M
for additional 24 h. Messenger RNA levels were quantified with real-time PCR, and their relative
quantities were calculated through the 2
∆∆CT
method. Results are expressed as fold change relative
to untreated cells. Data are expressed as mean
±
SEM of three separate experiments in triplicate
(N = 9)
. * p< 0.05, *** p< 0.001 vs. control;
p< 0.05,
◦◦
p< 0.01 and
◦◦
p< 0.001 vs. 6-OHDA 50
µ
M.
4. Discussion
Citrus reticulata (mandarin), originated from Southeast China, is present in Europe
under a multitude of varieties and has been studied mainly for its anticancer properties in
different
in vitro
and
in vivo
models [
10
]. In these regards, we showed that MJe induces
antiproliferative activity in three different anaplastic thyroid carcinoma cell lines, blocking
cell cycle in G2/M phase and inducing autophagy, as well as reducing cell migration and
affecting metalloproteinase activity [27].
To the best of our knowledge, this study is the first to assess the neuroprotective effect
of MJe in 6-OHDA-stressed SH-SY5Y cells, a widely employed cell line to mimic the cellular
PD’s environment [
28
]. Among the most recognized PD models, we chose the 6-OHDA,
a neurotoxin inducing depletion of dopaminergic neurons in the substantia nigra pars
compacta, given the scientific evidence, both
in vitro
[
29
] ad
in vivo
[
30
,
31
], supporting
its use.
First, we showed that MJe was able to defend cell viability from damage induced
by 6-OHDA, as shown by MTT and trypan blue assays. Moreover, we found that our
stressor dramatically increased apoptotic events, as clearly demonstrated by Annexin V/PI
staining, an effect that was counteracted by MJe treatment both at 0.1 and 0.5 mg/mL,
though to different extent. Apoptosis is regulated by several factors that can push cells
Antioxidants 2021,10, 539 13 of 16
towards survival or programmed death. It is also acknowledged that increased ROS
levels unleash mitochondrial damage and hence the release of apoptosis inducers [
32
].
Proapoptotic proteins (i.e., Bax and Bad) and antiapoptotic ones (i.e., Bcl-2 and Bcl-XL) are
finely balanced to regulate the fate of each cell in the organism, a stability that comes to
an end in cellular degeneration [
33
]. Moreover, tumor suppressor p53 is sensitive to stress
such as DNA damage and hypoxia, being activated by phosphorylation and acetylation,
hence inducing cell-cycle arrest and apoptosis [
34
]. Accordingly, we evaluated the gene
expression of the abovementioned factors, finding that Bax and p53 were downregulated
by MJe treatment, whereas Bcl-2 was upregulated, in a completely opposite manner of our
stressor 6-OHDA. These clearly suggested that their regulation was crucial in the effect of
MJe in SH-SY5Y cells. Noteworthy, ROS are known to be able to directly activate the cascade
of caspases and hence the whole apoptotic machinery, directly aiming at caspase 3 [
35
].
Therefore, we also evaluated the involvement of this caspase in the protective effect of
MJe, and we witnessed a sharp hindering of its activation compared to the levels observed
in control cells, suggesting the role of caspase 3 as a keystone in the overall protective
mechanism brought by MJe. As is widely known, 6-OHDA increases ROS levels as a direct
result of mitochondrial impairment [
36
], being one of the possible cellular causes of PD,
as well as other neurodegenerative diseases. Consequently, we aimed at evaluating the
ROS levels in SH-SY5Y cells after 6-OHDA stress, finding a robust increase of the species,
which was blocked by MJe treatment. Moreover, our extract ameliorated
ψ
m, impaired
by 6-OHDA injury that, together with the reduction of ROS levels, are clear signals of
mitochondrial protection elicited by our extract. Unbalanced ROS production brings its
effects at the nucleus level, where oxidative damage on DNA gives birth to oxidized base
adducts, among which 8-oxo-dG is a relevant marker of early clinical manifestations of
cognitive impairment [
37
]. Therefore, we evaluated its presence in SH-SY5Y cells after
6-OHDA stress, detecting an increase of the portion of fluorescent cells, and hence carrying
this DNA oxidative adduct, whereas MJe was able to lower this effect.
The antioxidant capacity of MJe was also appreciated through the evaluation of typical
cellular biomarkers involved in this process, namely SOD, CAT, and GSH. The exposure
of SH-SY5Y cells to 6-OHDA decreases cellular antioxidant defense, and flavonoids are
acknowledged for hampering it [
38
]. Here, we found that MJe was able to restore both
SOD and CAT activity, two fundamental enzymes that act together to quench oxygen
radicals, as well as GSH levels. Interestingly, MJe proved to also be a great antioxidant in
abiotic models, where it was able to quench both oxygen (ORAC) and nitrogen (DPPH)
radicals along with reducing ferric ions into ferrous ones, effects likely ascribed to the high
polyphenol content, as assessed by Folin–Ciocalteu assay and in line with previous reports
on Citrus extracts [39,40].
Nitric oxide (NO) is another central signaling molecule whose overproduction is
acknowledged to be the cause of neuronal impairment, typical of neurodegenerative
diseases like PD [
41
]. Notably, MJe decreased the cellular levels of NO that were drastically
increased by 6-OHDA. Furthermore, our extract acted as both antioxidant and reducing
agent in different cell-free models, given its high polyphenolic content, reinforcing the
results we obtained
in vitro
. The etiology of PD is not fully understood yet, but scientific
evidence shows that mutations in SNCA, PINK1, parkin, DJ-1, and LRRK2 genes are at the
basis of familial cases of PD [
42
]. These encode for proteins that are tightly intertwined
in the process of mitophagy and hence in the regulation of neuron viability. After an
oxidative stimulus,
α
-synuclein increases in neurons, forming the so-called Lewy bodies,
agglomerates typical of PD. Overexpression of this protein induces an increase of ROS
at the cellular level, starting a vicious cycle where
α
-synuclein induce ROS and vice
versa. Gain-of-function mutation of LRRK2 gene brings to an increased susceptibility of
neurons to oxidative stress, as widely demonstrated. On the other hand, parkin and its
regulator PINK1 are known to be involved in mitochondrial survival and protection against
ROS, together with DJ-1 that homodimerizes, becoming another antioxidant neuronal
defense [
43
]. In our study, the oxidative stress induced by 6-OHDA brought an expected
Antioxidants 2021,10, 539 14 of 16
sharp increase of both SNCA and LRRK2, an effect that was hampered by MJe treatment.
Conversely, the mitochondrial antioxidant machinery, consisting of PINK1/parkin and
DJ-1, was negatively affected by the stressor. However, MJe was able to restore the levels
of genes encoding the abovementioned proteins and hence improving response against
oxidative stress. Our results are in line with previous reports in which natural products
were able to hamper neuronal oxidative stress elicited by 6-OHDA, lowering SNCA and
LRRK2 expression levels along with increasing those of PINK1/parkin and DJ-1 [44,45].
5. Conclusions
Overall, MJe hampered the oxidative stress induced by 6-OHDA treatment in our
cellular model, contemporarily targeting the mitochondria, nucleus, and cytoplasm, pro-
tecting these compartments by ROS overproduction and increasing the survival rate, along
with blocking apoptotic machinery. From a molecular point of view, MJe restored the gene
expression of factors linked to mitochondrial functionality, acknowledged to be crucial in
PD clinical outcomes, whose balance was impaired by 6-OHDA. Therefore, we suggest the
great validity of MJe in facing oxidative-based diseases, such as PD, that needs to be also
proven in more complex models to corroborate our statements.
Author Contributions:
S.C. performed the experiments, analyzed the data, and drafted the manuscript;
A.M. performed the experiments and drafted the manuscript; G.E.L., C.R. and L.M. supported the ex-
perimental procedures, S.G. and G.C. critically revised the manuscript, D.B. performed the chemical
analysis and critically revised the manuscript; M.N. conceived and designed the experiments as well
as critically revised the manuscript. All authors have read and agreed to the published version of
the manuscript.
Funding:
Research was supported by Grant from Sicily Region (PO FESR Sicilia 2007/2013, CUP
G73F11000050004 to MN, project “MEPRA”, n 133 of Linea d’Intervento 4.1.1.1).
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement: Data is contained within the article.
Conflicts of Interest: The authors declare no conflict of interests.
References
1.
Trist, B.G.; Hare, D.J.; Double, K.L. Oxidative stress in the aging substantia nigra and the etiology of Parkinson’s disease. Aging
Cell 2019,18, e13031. [CrossRef]
2.
Deuschl, G.; Beghi, E.; Fazekas, F.; Varga, T.; Christoforidi, K.A.; Sipido, E.; Bassetti, C.L.; Vos, T.; Feigin, V.L. The burden of
neurological diseases in Europe: An analysis for the Global Burden of Disease Study 2017. Lancet. Public Health
2020
,5, e551–e567.
[CrossRef]
3.
Borrageiro, G.; Haylett, W.; Seedat, S.; Kuivaniemi, H.; Bardien, S. A review of genome-wide transcriptomics studies in Parkinson’s
disease. Eur. J. Neurosci. 2018,47, 1–16. [CrossRef] [PubMed]
4.
Cirmi, S.; Ferlazzo, N.; Lombardo, G.E.; Ventura-Spagnolo, E.; Gangemi, S.; Calapai, G.; Navarra, M. Neurodegenerative Diseases:
Might Citrus Flavonoids Play a Protective Role? Molecules 2016,21, 1312. [CrossRef] [PubMed]
5.
Uddin, M.S.; Al Mamun, A.; Kabir, M.T.; Ahmad, J.; Jeandet, P.; Sarwar, M.S.; Ashraf, G.M.; Aleya, L. Neuroprotective role of
polyphenols against oxidative stress-mediated neurodegeneration. Eur. J. Pharmacol. 2020,886, 173412. [CrossRef] [PubMed]
6.
Maugeri, A.; Cirmi, S.; Minciullo, P.L.; Gangemi, S.; Calapai, G.; Mollace, V.; Navarra, M. Citrus fruits and inflammaging: A
systematic review. Phytochem. Rev. 2019,18, 1025–1049. [CrossRef]
7.
Ferlazzo, N.; Cirmi, S.; Calapai, G.; Ventura-Spagnolo, E.; Gangemi, S.; Navarra, M. Anti-Inflammatory Activity of Citrus
bergamia Derivatives: Where Do We Stand? Molecules 2016,21, 1273. [CrossRef]
8.
Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; International Natural Product Sciences, T.; Supuran, C.T. Natural products in drug
discovery: Advances and opportunities. Nat. Rev. Drug Discov. 2021. [CrossRef] [PubMed]
9.
Cirmi, S.; Ferlazzo, N.; Lombardo, G.E.; Maugeri, A.; Calapai, G.; Gangemi, S.; Navarra, M. Chemopreventive Agents and
Inhibitors of Cancer Hallmarks: May Citrus Offer New Perspectives? Nutrients 2016,8, 698. [CrossRef]
10.
Cirmi, S.; Maugeri, A.; Ferlazzo, N.; Gangemi, S.; Calapai, G.; Schumacher, U.; Navarra, M. Anticancer Potential of Citrus Juices
and Their Extracts: A Systematic Review of Both Preclinical and Clinical Studies. Front. Pharmacol. 2017,8, 420. [CrossRef]
11.
Musumeci, L.; Maugeri, A.; Cirmi, S.; Lombardo, G.E.; Russo, C.; Gangemi, S.; Calapai, G.; Navarra, M. Citrus fruits and their
flavonoids in inflammatory bowel disease: An overview. Nat. Prod. Res. 2020,34, 122–136. [CrossRef] [PubMed]
Antioxidants 2021,10, 539 15 of 16
12.
Maugeri, A.; Ferlazzo, N.; De Luca, L.; Gitto, R.; Navarra, M. The link between the AMPK/SIRT1 axis and a flavonoid-rich extract
of Citrus bergamia juice: A cell-free, in silico, and in vitro study. Phytother. Res. 2019,33, 1805–1814. [CrossRef] [PubMed]
13.
Filocamo, A.; Bisignano, C.; Ferlazzo, N.; Cirmi, S.; Mandalari, G.; Navarra, M.
In vitro
effect of bergamot (Citrus bergamia)
juice against cagA-positive and-negative clinical isolates of Helicobacter pylori. BMC Complementary Altern. Med.
2015
,15, 256.
[CrossRef]
14.
Barreca, D.; Bellocco, E.; Caristi, C.; Leuzzi, U.; Gattuso, G. Flavonoid profile and radical-scavenging activity of Mediterranean
sweet lemon (Citrus limetta Risso) juice. Food Chem. 2011,129, 417–422. [CrossRef] [PubMed]
15.
Gattuso, G.; Barreca, D.; Gargiulli, C.; Leuzzi, U.; Caristi, C. Flavonoid composition of Citrus juices. Molecules
2007
,12, 1641–1673.
[CrossRef]
16.
Hertog, M.G.L.; Hollman, P.C.H.; Venema, D.P. Optimization of a quantitative HPLC determination of potentially anticarcinogenic
flavonoids in vegetables and fruits. J. Agric. Food Chem. 1992,40, 1591–1598. [CrossRef]
17.
Ferlazzo, N.; Cirmi, S.; Maugeri, A.; Russo, C.; Lombardo, G.E.; Gangemi, S.; Calapai, G.; Mollace, V.; Navarra, M. Neuroprotective
Effect of Bergamot Juice in 6-OHDA-Induced SH-SY5Y Cell Death, an In Vitro Model of Parkinson’s Disease. Pharmaceutics
2020
,
12, 326. [CrossRef]
18.
Morisi, R.; Celano, M.; Tosi, E.; Schenone, S.; Navarra, M.; Ferretti, E.; Costante, G.; Durante, C.; Botta, G.; D’Agostino, M.;
et al. Growth inhibition of medullary thyroid carcinoma cells by pyrazolo-pyrimidine derivates. J. Endocrinol. Investig.
2007
,30,
RC31–RC34. [CrossRef]
19.
Corasaniti, M.T.; Bilotta, A.; Strongoli, M.C.; Navarra, M.; Bagetta, G.; Di Renzo, G. HIV-1 coat protein gp120 stimulates
interleukin-1beta secretion from human neuroblastoma cells: Evidence for a role in the mechanism of cell death. Br. J. Pharmacol.
2001,134, 1344–1350. [CrossRef] [PubMed]
20.
Corasaniti, M.T.; Navarra, M.; Catani, M.V.; Melino, G.; Nistico, G.; Finazzi-Agro, A. NMDA and HIV-1 coat protein, GP120,
produce necrotic but not apoptotic cell death in human CHP100 neuroblastoma cultures via a mechanism involving calpain.
Biochem. Biophys. Res. Commun. 1996,229, 299–304. [CrossRef]
21. Cirmi, S.; Ferlazzo, N.; Gugliandolo, A.; Musumeci, L.; Mazzon, E.; Bramanti, A.; Navarra, M. Moringin from Moringa Oleifera
Seeds Inhibits Growth, Arrests Cell-Cycle, and Induces Apoptosis of SH-SY5Y Human Neuroblastoma Cells through the
Modulation of NF-kappaB and Apoptotic Related Factors. Int. J. Mol. Sci. 2019,20, 1930. [CrossRef]
22.
Curro, M.; Risitano, R.; Ferlazzo, N.; Cirmi, S.; Gangemi, C.; Caccamo, D.; Ientile, R.; Navarra, M. Citrus bergamia Juice Extract
Attenuates beta-Amyloid-Induced Pro-Inflammatory Activation of THP-1 Cells Through MAPK and AP-1 Pathways. Sci. Rep.
2016,6, 20809. [CrossRef] [PubMed]
23.
Ferlazzo, N.; Visalli, G.; Smeriglio, A.; Cirmi, S.; Lombardo, G.E.; Campiglia, P.; Di Pietro, A.; Navarra, M. Flavonoid Fraction
of Orange and Bergamot Juices Protect Human Lung Epithelial Cells from Hydrogen Peroxide-Induced Oxidative Stress. Evid.
Based Complement. Altern. Med. ECAM 2015,2015, 957031. [CrossRef] [PubMed]
24.
Lombardo, G.E.; Cirmi, S.; Musumeci, L.; Pergolizzi, S.; Maugeri, A.; Russo, C.; Mannucci, C.; Calapai, G.; Navarra, M.
Mechanisms Underlying the Anti-Inflammatory Activity of Bergamot Essential Oil and Its Antinociceptive Effects. Plants
2020
,9,
704. [CrossRef] [PubMed]
25.
Ferlazzo, E.; Sueri, C.; Elia, M.; D’Agostino, T.; Aguglia, U. Reflex seizures in a patient with Angelman syndrome and trisomy 21.
Neurol. Sci. Off. J. Ital. Neurol. Soc. Ital. Soc. Clin. Neurophysiol. 2016,37, 1373–1374. [CrossRef] [PubMed]
26.
Cai, H.A.; Tao, X.; Zheng, L.J.; Huang, L.; Peng, Y.; Liao, R.Y.; Zhu, Y.M. Ozone alleviates ischemia-reperfusion injury by inhibiting
mitochondrion-mediated apoptosis pathway in SH-SY5Y cells. Cell Biol. Int. 2018. [CrossRef]
27.
Celano, M.; Maggisano, V.; De Rose, R.F.; Bulotta, S.; Maiuolo, J.; Navarra, M.; Russo, D. Flavonoid Fraction of Citrus reticulata
Juice Reduces Proliferation and Migration of Anaplastic Thyroid Carcinoma Cells. Nutr. Cancer
2015
,67, 1183–1190. [CrossRef]
[PubMed]
28.
Xicoy, H.; Wieringa, B.; Martens, G.J. The SH-SY5Y cell line in Parkinson’s disease research: A systematic review. Mol. Neurodegener.
2017,12, 10. [CrossRef]
29.
Tsai, R.T.; Tsai, C.W.; Liu, S.P.; Gao, J.X.; Kuo, Y.H.; Chao, P.M.; Hung, H.S.; Shyu, W.C.; Lin, S.Z.; Fu, R.H. Maackiain Ameliorates
6-Hydroxydopamine and SNCA Pathologies by Modulating the PINK1/Parkin Pathway in Models of Parkinson’s Disease in
Caenorhabditis elegans and the SH-SY5Y Cell Line. Int. J. Mol. Sci. 2020,21, 4455. [CrossRef] [PubMed]
30.
Abidar, S.; Boiangiu, R.S.; Dumitru, G.; Todirascu-Ciornea, E.; Amakran, A.; Cioanca, O.; Hritcu, L.; Nhiri, M. The Aqueous Extract
from Ceratonia siliqua Leaves Protects Against 6-hydroxydopamine in Zebrafish: Understanding the Underlying Mechanism.
Antioxidants 2020,9, 304. [CrossRef]
31.
Zhuang, W.; Cai, M.; Li, W.; Chen, C.; Wang, Y.; Lv, E.; Fu, W. Polyphenols from Toona sinensiss Seeds Alleviate Neuroinflamma-
tion Induced by 6-Hydroxydopamine Through Suppressing p38 MAPK Signaling Pathway in a Rat Model of Parkinson’s Disease.
Neurochem. Res. 2020,45, 2052–2064. [CrossRef] [PubMed]
32.
Redza-Dutordoir, M.; Averill-Bates, D.A. Activation of apoptosis signalling pathways by reactive oxygen species. Biochim. Biophys.
Acta 2016,1863, 2977–2992. [CrossRef] [PubMed]
33.
Knight, T.; Luedtke, D.; Edwards, H.; Taub, J.W.; Ge, Y. A delicate balance—The BCL-2 family and its role in apoptosis, oncogenesis,
and cancer therapeutics. Biochem. Pharmacol. 2019,162, 250–261. [CrossRef]
34.
Wang, X.; Simpson, E.R.; Brown, K.A. p53: Protection against Tumor Growth beyond Effects on Cell Cycle and Apoptosis. Cancer
Res. 2015,75, 5001–5007. [CrossRef]
Antioxidants 2021,10, 539 16 of 16
35.
Izeradjene, K.; Douglas, L.; Tillman, D.M.; Delaney, A.B.; Houghton, J.A. Reactive oxygen species regulate caspase activation
in tumor necrosis factor-related apoptosis-inducing ligand-resistant human colon carcinoma cell lines. Cancer Res.
2005
,65,
7436–7445. [CrossRef]
36.
Guo, S.; Bezard, E.; Zhao, B. Protective effect of green tea polyphenols on the SH-SY5Y cells against 6-OHDA induced apoptosis
through ROS-NO pathway. Free Radic. Biol. Med. 2005,39, 682–695. [CrossRef] [PubMed]
37.
Khan, Z.; Ali, S.A. Oxidative stress-related biomarkers in Parkinson’s disease: A systematic review and meta-analysis. Iran. J.
Neurol. 2018,17, 137–144. [CrossRef]
38.
Kesh, S.; Kannan, R.R.; Balakrishnan, A. Naringenin alleviates 6-hydroxydopamine induced Parkinsonism in SHSY5Y cells and
zebrafish model. Comp. Biochem. Physiol. Toxicol. Pharmacol. CBP 2021,239, 108893. [CrossRef]
39.
Fusco, R.; Cirmi, S.; Gugliandolo, E.; Di Paola, R.; Cuzzocrea, S.; Navarra, M. A flavonoid-rich extract of orange juice reduced
oxidative stress in an experimental model of inflammatory bowel disease. J. Funct. Foods 2017,30, 168–178. [CrossRef]
40.
Marino, A.; Paterniti, I.; Cordaro, M.; Morabito, R.; Campolo, M.; Navarra, M.; Esposito, E.; Cuzzocrea, S. Role of natural
antioxidants and potential use of bergamot in treating rheumatoid arthritis. Pharma Nutr. 2015,3, 53–59. [CrossRef]
41.
Dawson, T.M.; Dawson, V.L. Nitric Oxide Signaling in Neurodegeneration and Cell Death. Adv. Pharmacol.
2018
,82, 57–83.
[CrossRef] [PubMed]
42.
Klein, C.; Westenberger, A. Genetics of Parkinson’s disease. Cold Spring Harb. Perspect. Med.
2012
,2, a008888. [CrossRef]
[PubMed]
43. Chang, K.H.; Chen, C.M. The Role of Oxidative Stress in Parkinson’s Disease. Antioxidants 2020,9, 597. [CrossRef]
44.
Lin, C.Y.; Tsai, C.W.; Tsai, C.W. Carnosic acid protects SH-SY5Y cells against 6-hydroxydopamine-induced cell death through
upregulation of parkin pathway. Neuropharmacology 2016,110, 109–117. [CrossRef] [PubMed]
45.
Yang, G.; Li, J.; Cai, Y.; Yang, Z.; Li, R.; Fu, W. Glycyrrhizic Acid Alleviates 6-Hydroxydopamine and Corticosterone-Induced
Neurotoxicity in SH-SY5Y Cells Through Modulating Autophagy. Neurochem. Res. 2018,43, 1914–1926. [CrossRef] [PubMed]
... The specific mechanism of action behind the neuroprotective effect of the bio-compounds in red cabbage extracts is not easy to determine. However, it has been reported that anthocyanins present in black carrot extracts act by inhibiting ROS-mediated oxidative stress and apoptosis [87], while flavonoids in mandarin juice extracts generate protection against the overproduction of ROS in mitochondria, nucleus, and cytoplasm of the cell, restoring the gene expression of factors linked to mitochondrial functionality [84]. Moreover, phenolic compounds such as quercetin have been reported to decrease apoptosis in neurotoxicity-induced models by modulating the autophagic pathway [88]. ...
... On the other hand, fresh red cabbage extract is a rich source of anthocyanins. Some authors have reported that bio-compounds like polyphenols, flavonoids, and anthocyanins from the natural food matrix, such as grape [80], green tea [81], mulberry [82], morus alba fruit [83], mandarin juice [84], elderberry [85], Brazilian green propolis [86], among others, have a neuroprotective effect associated to protection against oxidative stress or neuroinflammation. The specific mechanism of action behind the neuroprotective effect of the bio-compounds in red cabbage extracts is not easy to determine. ...
... The specific mechanism of action behind the neuroprotective effect of the bio-compounds in red cabbage extracts is not easy to determine. However, it has been reported that anthocyanins present in black carrot extracts act by inhibiting ROSmediated oxidative stress and apoptosis [87], while flavonoids in mandarin juice extracts generate protection against the overproduction of ROS in mitochondria, nucleus, and cytoplasm of the cell, restoring the gene expression of factors linked to mitochondrial functionality [84]. Moreover, phenolic compounds such as quercetin have been reported to decrease apoptosis in neurotoxicity-induced models by modulating the autophagic pathway [88]. ...
Article
Full-text available
Parkinson’s disease (PD) is the second most common neurodegenerative disorder, and no efficient therapy able to cure or slow down PD is available. In this study, dehydrated red cabbage was evaluated as a novel source of bio-compounds with neuroprotective capacity. Convective drying was carried out at different temperatures. Total phenolics (TPC), flavonoids (TFC), anthocyanins (TAC), and glucosinolates (TGC) were determined using spectrophotometry, amino acid profile by LC-DAD and fatty acid profile by GC-FID. Phenolic characterization was determined by liquid chromatography-high-resolution mass spectrometry. Cytotoxicity and neuroprotection assays were evaluated in SH-SY5Y human cells, observing the effect on preformed fibrils of α-synuclein. Drying kinetic confirmed a shorter processing time with temperature increase. A high concentration of bio-compounds was observed, especially at 90 °C, with TPC = 1544.04 ± 11.4 mg GAE/100 g, TFC = 690.87 ± 4.0 mg QE/100 g and TGC = 5244.9 ± 260.2 µmol SngE/100 g. TAC degraded with temperature. Glutamic acid and arginine were predominant. Fatty acid profiles were relatively stable and were found to be mostly C18:3n3. The neochlorogenic acid was predominant. The extracts had no cytotoxicity and showed a neuroprotective effect at 24 h testing, which can extend in some cases to 48 h. The present findings underpin the use of red cabbage as a functional food ingredient.
... These findings are in accordance with other studies, which employed 6-OHDA-or MPTP-induced oxidative stress to damage neuronally-derived SH-SY5Y cells. 13,48 Furthermore, in comparison to 2,2'-azobis(2-amidinopropane) dihydrochloride (AAPH) in an ORAC assay, KPS was found to be a moderate ROS generator ( Figure S1). (PPNPs), has been explored as a promising therapy for PD to enhance its accumulation in the blood and brain. ...
Article
Full-text available
Nanomedicine for treating post‐viral infectious disease syndrome is at an emerging stage. Despite promising results from preclinical studies on conventional antioxidants, their clinical translation as a therapy for treating post‐COVID conditions remains challenging. The limitations are due to their low bioavailability, instability, limited transport to the target tissues, and short half‐life, requiring frequent and high doses. Activating the immune system during coronavirus (SARS‐CoV‐2) infection can lead to increased production of reactive oxygen species (ROS), depleted antioxidant reserve, and finally, oxidative stress and neuroinflammation. To tackle this problem, we developed an antioxidant nanotherapy based on lipid (vesicular and cubosomal types) nanoparticles (LNPs) co‐encapsulating ginkgolide B and quercetin. The antioxidant‐loaded nanocarriers were prepared by a self‐assembly method via hydration of a lyophilized mixed thin lipid film. We evaluated the LNPs in a new in vitro model for studying neuronal dysfunction caused by oxidative stress in coronavirus infection. We examined the key downstream signaling pathways that are triggered in response to potassium persulfate (KPS) causing oxidative stress‐mediated neurotoxicity. Treatment of neuronally‐derived cells (SH‐SY5Y) with KPS (50 mM) for 30 min markedly increased mitochondrial dysfunction while depleting the levels of both glutathione peroxidase (GSH‐Px) and tyrosine hydroxylase (TH). This led to the sequential activation of apoptotic and necrotic cell death processes, which corroborates with the crucial implication of the two proteins (GSH‐Px and TH) in the long‐COVID syndrome. Nanomedicine‐mediated treatment with ginkgolide B‐loaded cubosomes and vesicular LNPs showed minimal cytotoxicity and completely attenuated the KPS‐induced cell death process, decreasing apoptosis from 32.6% (KPS) to 19.0% (MO‐GB), 12.8% (MO‐GB‐Quer), 14.8% (DMPC‐PEG‐GB), and 23.6% (DMPC‐PEG‐GB‐Quer) via free radical scavenging and replenished GSH‐Px levels. These findings indicated that GB‐LNPs‐based nanomedicines may protect against KPS‐induced apoptosis by regulating intracellular redox homeostasis.
... In the brains of diabetic untreated rats, STZ-induced diabetes triggered a significant (p < 0.05) increase in pro-inflammatory biomarkers, including interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α), corroborated by several previous literature [60]. Several researchers have put forward the groundbreaking concept by demonstrating that the pro-inflammatory cytokine, TNF-α, could trigger insulin resistance, challenging conventional thinking [61,62]. This revelation led to a paradigm shift, highlighting fat tissue as a source of cytokines and bioactive substances, not limited to TNF-α but also encompassing leptin, IL-6, resistin, MCP-1, PAI-1, and others [63]. ...
Article
Full-text available
Diabetes is a group of medical conditions characterized by the body’s inability to effectively control blood glucose levels, due to either insufficient insulin synthesis in type 1 diabetes or inadequate insulin sensitivity in type 2 diabetes. According to this research, the PI3K/AKT pathway of Ocimum gratissimum leaf flavonoid-rich extracts in streptozotocin-induced diabetic rats was studied. We purchased and used a total of forty (40) male Wistar rats for the study. We divided the animals into five (5) different groups: normal control (Group A), diabetic control (Group B), low dose (150 mg/kg body weight) of Ocimum gratissimum flavonoid-rich leaf extract (LDOGFL) (Group C), high dose (300 mg/kg body weight) of Ocimum gratissimum flavonoid-rich leaf extract (HDOGFL) (Group D), and 200 mg/kg of metformin (MET) (Group E). Streptozotocin induced all groups except Group A, which serves as the normal control group. The experiment lasted for 21 days, following which we sacrificed the animals and harvested their brains for biochemical analysis on the 22nd day. We carried out an analysis that included reduced glutathione (GSH), glutathione transferases (GST), catalase (CAT), glutathione peroxidase (GPx), and superoxide dismutase (SOD), along with GLUT4, MDA, pro-inflammatory cytokines, NO, neurotransmitters, cholinergic enzyme activities, cardiolipin, and the gene expression of PI3K/AKT. The obtained result indicates that the flavonoid-rich extracts of O. gratissimum significantly enhanced the levels of GSH, GST, CAT, GPx, and SOD, as well as GLUT4 and cardiolipin. The levels of GSH, GST, CAT, GPx, and SOD, as well as GLUT4 and cardiolipin, were significantly increased by gratissimum. Moreover, the extracts decrease the levels of MDA, pro-inflammatory cytokines, NO, neurotransmitters, and cholinergic enzyme activities. Additionally, the flavonoid-rich extracts of O. gratissimum significantly improved the AKT and PI3K gene expressions in diabetic rats. gratissimum had their AKT and PI3K gene expressions significantly (p < 0.05) improved. The findings indicate that O. gratissimum leaf flavonoids have the potential to treat diabetes mellitus. O. gratissimum leaf flavonoids possess therapeutic potential in themselves and can be applied in the management of diabetes mellitus. Although further analysis can be carried out in terms of isolating, profiling, or purifying the active compounds present in the plant’s extract.
... Moreover, levels of reactive oxygen or nitrogen species (ROS and RNS, respectively), easily quantifiable by fluorescent probes [32], need to be assessed for a comprehensive view of the inflammatory process due to increased oxidative stress. Finally, also DNA, in terms of 8-oxo-dG, mitochondrial, in terms of membrane potential, and lipid membrane damages, in terms of levels of malondialdehyde (MDA), can be quantified by exploiting several fluorescent probes [33]. ...
Chapter
Natural products have dominated our lives since ancient times. Today, they are an inexhaustible source of new medications for disease treatment. The practice of evaluating bioactive compounds extracted from natural sources has also advanced significantly, prompting a need to understand current methods to identify and evaluate them. This book covers basic scientific aspects of preclinical research on natural products for specific conditions and diseases. These include aging, gynecological disorders, inflammatory disorders, renal disorders and cardiovascular disorders. Each of the 10 book chapters give a structured overview on preclinical methods on the etiology of diseases, natural products as the materials for the bioassays, extract types, concentration of the extracts/compounds for in vitro and in vivo assays, preparation of the test materials, application of the test materials, step-by-step methods and related calculations. The book is intended as a quick reference for natural product researchers, pharmacists and postgraduate students in pharmacognosy. Medical doctors working in preclinical research on natural products will also benefit from the information provided.
... Phytochemicals, such as flavonoids and terpenoids, have been linked to improved cognitive function and brain health. These compounds can protect nerve cells from damage, reduce inflammation in the brain and enhance neural plasticity, potentially benefiting individuals with conditions such as Alzheimer's and Parkinson's disease [43][44][45][46]. Plant compounds, such as soybean isoflavones, can help improve bone health and reduce the risk of osteoporosis. ...
Article
Full-text available
For centuries, plants have been part of human civilisation, serving as food, healing substances and treatments for various diseases. In recent years, advances in biological research have revealed the remarkable potential of plant compounds in the food and medicine sectors. In the food industry, plant extracts are used to improve the safety, nutritional value and shelf life of food products. In addition, antimicrobial phytocompounds have found application in food packaging materials, ensuring inhibition of bacterial growth and prolonging the freshness of perishable goods.
... In a study conducted by Gomez del Rio et al. [8], it was shown that a hyperforin-rich H. perforatum extract prevents dopaminergic neuron loss in experimental animals in a Parkinson's model created with rotenone. Various studies have also shown that 6-OHDA stimulation increases p38 phosphorylation and caspase-3 activation [22,23]. ...
Article
Full-text available
Background and objective: Parkinson’s disease (PD) is the second most common neurodegenerative diseaseafter Alzheimer’s disease. In our study, PD model was created as a result ofexposure to 6-hydroxydopamine (6-OHDA) in SH-SY5Y cells, which is a humanneuroblastoma cell line. The protective effect of Hypericum perforatum on PD wasinvestigated. Materials and methods: Phytochemical analysis of H. perforatum extract was performed. Then, SH-SY5Ycells were differentiated using retinoic acid and then administered 6-OHDAneurotoxin. To determine the protective effects of H. perforatum extract, weinvestigated the changes in the mRNA expression level of caspase-3, totaloxidant status, and antioxidant levels in differentiated SH-SY5Y. Results and conclusion : According to our results, H. perforatum extract contains glycosides, tannins,flavonoids, and carbohydrates as the major secondary metabolites.H. perforatum extract significantly reduced caspase-3 gene expression against6-OHDA toxicity in differentiated SH-SY5Y cells. It was found that total oxidantstatus level increased significantly in the 6-OHDA experimental group comparedwith the control and H. perforatum experimental groups.It was found that H. perforatum extract has an inhibitory effect on caspase-3 geneexpression, which plays an important role in apoptosis. Therfore, H. perforatumextract has been shown to have a therapeutic potential against 6-OHDA toxicity.
Article
Kinnow is a citrus fruit that is extensively cultivated in India, Pakistan and other countries. The production of Kinnow generates a significant amount of waste, including peel, seeds, and pulp. The disposal of this waste is a major environmental issue, as it can lead to pollution and greenhouse gas emissions. Therefore, there is a growing interest in finding ways to valorize Kinnow waste. There are studies which focused on using Kinnow waste for the production of biofuels, biopolymers, and other value-added products. The utilization of Kinnow waste for these purposes can help to reduce the environmental impact of Kinnow production and generate economic benefits for farmers and other stakeholders. The research on Kinnow waste valorization is still in its early stages, but it has the potential to contribute to sustainable development in the citrus industry.
Article
Aim: We purposed to study the neuroprotective effects of Hawthorn berry (crataegus spp.) extract, which is familiar to have antioxidant and anti-inflammatory features, opposite the neurotoxicity led to by 6-OHDA in SH-SY5Y cells. Method: SH-SY5Y cells were treated with Hawthorn berry (25-50-75 and 100 μg/mL) for two hours ago 6-OHDA administration. Cells were exposed to 200 µM 6-OHDA for 24 hours to mimic the in vitro Parkinson's disease model. After one day, cell viability was measured by lactate dehydrogenase and 3-(4,5 dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide analysis. Oxidative stress was evaluated with tumor necrosis factor-α, interleukin-1β, superoxide dismutase, catalase, glutathione, glutathione peroxidase, myeloperoxidase, and malondialdehyde assays. Results: It was found that the viability rate of Hawthorn berry increased depending on the concentration and the cell viability was 94% at the highest concentration (p<0.001). Also, 6-OHDA raised lactate dehydrogenase leakage in SH-SY5Y cells (p<0.001). While 6-OHDA exacerbated oxidative stress by enhancing tumor necrosis factor-α, interleukin-1β, myeloperoxidase, and malondialdehyde (p<0.001), pretreatment with Hawthorn berry alleviated these toxic effects of 6-OHDA through antioxidant capacity by increasing glutathione peroxidase, superoxide dismutase, catalase and glutathione (p<0.05), (p<0.001). In line with all findings, Hawthorn berry attenuated neuronal cell demise in a dose-dependent manner. Conclusion: Considering its neuroprotective role as well as its effects on oxidative stress, Hawthorn berry could be a potential natural bio-medicine to prevent the development of Parkinson's disease.
Article
Introduction: Owing to limited efficient treatment strategies for highly prevalent and distressing Parkinson's disease (PD), an impending need emerged for deciphering new modes and mechanisms for effective management. SH-SY5Y-based in vitro neuronal models have emerged as a new possibility for the elucidation of cellular and molecular processes in the pathogenesis of PD. SH-SY5Y cells are of human origin, adhered to catecholaminergic neuronal attributes, which consequences in imparting wide acceptance and significance to this model over conventional in vitro PD models for high-throughput screening of therapeutics. Areas covered: Herein, the authors review the SH-SY5Y cell line and its value to PD research. The authors also provide the reader with their expert perspectives on how these developments can lead to the development of new impactful therapeutics. Expert opinion: Encouraged by recent research on SH-SY5Y cell lines, it was envisaged that this in vitro model can serve as a primary model for assessing efficacy and toxicity of new therapeutics as well as for nanocarriers' capacity in delivering therapeutic agents across BBB. Considering the proximity with human neuronal environment as in pathogenic PD conditions, SH-SY5Y cell lines vindicated consistency and reproducibility in experimental results. Accordingly, exploitation of this standardized SH-SY5Y cell line can fast-track the drug discovery and development path for novel therapeutics.
Chapter
Parkinson’s disease (PD) is a chronic neurodegenerative disorder (ND) characterised by gradual degradation of dopaminergic neurons in the substantia nigra pars compacta (SNpc) area of the human midbrain, which results in a reduction in dopamine levels. After a certain age, neurodegeneration becomes an extremely important factor in human life. Given its high incidence rate, PD continues to be one of the NDs that represent a serious hazard to adults over 60. Neuroinflammation, oxidative stress, mitochondrial dysfunction, and apoptosis are all recognised as contributing factors to the pathophysiology of PD, despite the fact that the precise origin of the disease is still unknown. There is a need for a highly tailored therapy strategy for PD, as different pharmacological combinations are only available to manage the symptoms. This is where the use of plant-derived natural compounds as a neuroprotective agent has been in the spotlight. With their antioxidant properties, which are thought to help in repairing the damage caused by free radicals, as well as their antiapoptotic and anti-inflammatory properties, flavonoids are one of the main classes of phytochemicals that have demonstrated their effectiveness in acting as a neuroprotective agent for PD. These properties could activate pathways that target mitochondrial dysfunction and induce neurotrophic factors by exhibiting a neuroprotective effect. The scientific literature on the anti-neurodegenerative and neuroprotective functions of phytochemicals is reviewed in this chapter, with a particular emphasis on the potential of flavonoids as natural products for the prevention and treatment of PD.KeywordsPhytochemicalsFlavonoidsParkinson’s diseaseTherapeutics
Article
Full-text available
Natural products and their structural analogues have historically made a major contribution to pharmacotherapy, especially for cancer and infectious diseases. Nevertheless, natural products also present challenges for drug discovery, such as technical barriers to screening, isolation, characterization and optimization, which contributed to a decline in their pursuit by the pharmaceutical industry from the 1990s onwards. In recent years, several technological and scientific developments — including improved analytical tools, genome mining and engineering strategies, and microbial culturing advances — are addressing such challenges and opening up new opportunities. Consequently, interest in natural products as drug leads is being revitalized, particularly for tackling antimicrobial resistance. Here, we summarize recent technological developments that are enabling natural product-based drug discovery, highlight selected applications and discuss key opportunities. Natural products have historically made a major contribution to pharmacotherapy, but also present challenges for drug discovery, such as technical barriers to screening, isolation, characterization and optimization. This Review discusses recent technological developments — including improved analytical tools, genome mining and engineering strategies, and microbial culturing advances — that are enabling a revitalization of natural product-based drug discovery.
Article
Full-text available
Background: Neurological disorders account for a large and increasing health burden worldwide, as shown in the Global Burden of Diseases (GBD) Study 2016. Unpacking how this burden varies regionally and nationally is important to inform public health policy and prevention strategies. The population in the EU is older than that of the WHO European region (western, central, and eastern Europe) and even older than the global population, suggesting that it might be particularly vulnerable to an increasing burden of age-related neurological disorders. We aimed to compare the burden of neurological disorders in the EU between 1990 and 2017 with those of the WHO European region and worldwide. Methods: The burden of neurological disorders was calculated for the year 2017 as incidence, prevalence, mortality, disability-adjusted life-years (DALYs), years of life lost, and years lived with disability for the countries in the EU and the WHO European region, totally and, separately. Diseases analysed were Alzheimer's disease and other dementias, epilepsy, headache (migraine and tension-type headache), multiple sclerosis, Parkinson's disease, brain cancer, motor neuron diseases, neuroinfectious diseases, and stroke. Data are presented as totals and by sex, age, year, location and socio-demographic context, and shown as counts and rates. Findings: In 2017, the total number of DALYs attributable to neurological disorders was 21·0 million (95% uncertainty interval 18·5-23·9) in the EU and 41·1 million (36·7-45·9) in the WHO European region, and the total number of deaths was 1·1 million (1·09-1·14) in the EU and 1·97 million (1·95-2·01) in the WHO European region. In the EU, neurological disorders ranked third after cardiovascular diseases and cancers representing 13·3% (10·3-17·1) of total DALYs and 19·5% (18·0-21·3) of total deaths. Stroke, dementias, and headache were the three commonest causes of DALYs in the EU. Stroke was also the leading cause of DALYs in the WHO European region. During the study period we found a substantial increase in the all-age burden of neurodegenerative diseases, despite a substantial decrease in the rates of stroke and infections. The burden of neurological disorders in Europe was higher in men than in women, peaked in individuals aged 80-84 years, and varied substantially with WHO European region and country. All-age DALYs, deaths, and prevalence of neurological disorders increased in all-age measures, but decreased when using age-standardised measures in all but three countries (Azerbaijan, Turkmenistan, and Uzbekistan). The decrease was mostly attributed to the reduction of premature mortality despite an overall increase in the number of DALYs. Interpretation: Neurological disorders are the third most common cause of disability and premature death in the EU and their prevalence and burden will likely increase with the progressive ageing of the European population. Greater attention to neurological diseases must be paid by health authorities for prevention and care. The data presented here suggest different priorities for health service development and resource allocation in different countries. Funding: European Academy of Neurology.
Article
Full-text available
Parkinson’s disease (PD) is caused by progressive neurodegeneration of dopaminergic (DAergic) neurons with abnormal accumulation of α-synuclein in substantia nigra (SN). Studies have suggested the potential involvement of dopamine, iron, calcium, mitochondria and neuroinflammation in contributing to overwhelmed oxidative stress and neurodegeneration in PD. Function studies on PD-causative mutations of SNCA, PRKN, PINK1, DJ-1, LRRK2, FBXO7 and ATP13A2 further indicate the role of oxidative stress in the pathogenesis of PD. Therefore, it is reasonable that molecules involved in oxidative stress, such as DJ-1, coenzyme Q10, uric acid, 8-hydroxy-2’-deoxyguanosin, homocysteine, retinoic acid/carotenes, vitamin E, glutathione peroxidase, superoxide dismutase, xanthine oxidase and products of lipid peroxidation, could be candidate biomarkers for PD. Applications of antioxidants to modulate oxidative stress could be a strategy in treating PD. Although a number of antioxidants, such as creatine, vitamin E, coenzyme Q10, pioglitazone, melatonin and desferrioxamine, have been tested in clinical trials, none of them have demonstrated conclusive evidence to ameliorate the neurodegeneration in PD patients. Difficulties in clinical studies may be caused by the long-standing progression of neurodegeneration, lack of biomarkers for premotor stage of PD and inadequate drug delivery across blood–brain barrier. Solutions for these challenges will be warranted for future studies with novel antioxidative treatment in PD patients.
Article
Full-text available
The movement disorder Parkinson’s disease (PD) is the second most frequently diagnosed neurodegenerative disease, and is associated with aging, the environment, and genetic factors. The intracellular aggregation of α-synuclein and the loss of dopaminergic neurons in the substantia nigra pars compacta are the pathological hallmark of PD. At present, there is no successful treatment for PD. Maackiain (MK) is a flavonoid extracted from dried roots of Sophora flavescens Aiton. MK has emerged as a novel agent for PD treatment that acts by inhibiting monoamine oxidase B. In this study, we assessed the neuroprotective potential of MK in Caenorhabditis elegans and investigated possible mechanism of this neuroprotection in the human SH-SY5Y cell line. We found that MK significantly reduced dopaminergic neuron damage in 6-hydroxydopamine (6-OHDA)-exposed worms of the BZ555 strain, with corresponding improvements in food-sensing behavior and life-span. In transgenic worms of strain NL5901 treated with 0.25 mM MK, the accumulation of α-synuclein was diminished by 27% (p < 0.01) compared with that in untreated worms. Moreover, in worms and the SH-SY5Y cell line, we confirmed that the mechanism of MK-mediated protection against PD pathology may include blocking apoptosis, enhancing the ubiquitin-proteasome system, and augmenting autophagy by increasing PINK1/parkin expression. The use of small interfering RNA to downregulate parkin expression in vivo and in vitro could reverse the benefits of MK in PD models. MK may have considerable therapeutic applications in PD.
Article
Full-text available
Polyphenols from Toona sinensis seeds (PTSS) have demonstrated anti-inflammatory effects in various diseases, while the anti-neuroinflammatory effects still remain to be investigated. We aimed to investigate the effects of PTSS on Parkinson’s disease and underlying mechanisms using a rat model. We employed 6-hydroxydopamine (6-OHDA) to male Sprague Dawley (SD) rats and PC12 cells to construct the in vivo and vitro models of PD and dopaminergic (DA) neuron injury, respectively. Cell viability was detected by cell counting kit-8 (CCK-8) assay and protein levels of inflammatory mediators and some p38 MAPK pathway molecules were investigated by immunohistochemistry and Western blot analyses. The results showed that 6-OHDA significantly increased protein levels of inflammatory mediators, such as cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), and tumor necrosis factor α (TNF-α), which could be reversed by PTSS through suppressing the p38 MAPK pathway. The anti-inflammatory effects of PTSS were significantly enhanced by the specific p38 inhibitor of SB203580 in vitro. The present work suggests that PTSS can exert anti-inflammatory effects on PD models, which may be attributed to the suppression of p38 MAPK signaling pathway.
Article
Full-text available
Renewed interest in natural products as potential source of drugs led us to investigate on both the anti-inflammatory and anti-nociceptive activity of Citrus bergamia Risso et Poiteau (bergamot) essential oil (BEO). Carrageenan-induced paw edema in rats was used as an experimental model of inflammation. Because of the toxicity of furocoumarins, we performed our study by using the BEO fraction deprived of these compounds (BEO-FF). Treatment with BEO-FF led to a significant inhibition of paw edema induced by a sub-plantar injection of carrageenan. Moreover, histological examination of BEO-FF-treated rat paw biopsies showed a reduction of pathological changes typical of edema. Pre-treatment with BEO-FF significantly reduced interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α levels in the paw homogenates, as well as nitrite/nitrate and prostaglandin E2 (PGE2) content in exudates. In addition, BEO-FF possesses antioxidant properties, as determined by cell-free assays. Furthermore, results of the writhing test showed that BEO-FF elicited a pronounced analgesic response, as demonstrated by a significant inhibition of constrictions in mice receiving acetic acid, with respect to control animals, whereas the results of the hot plate test suggested that the supra-spinal analgesia participates in the anti-nociceptive effect of BEO-FF. Our study indicates that BEO-FF exerts anti-inflammatory and anti-nociceptive effects, and suggests its potential role as an anti-edemigen and analgesic drug.
Article
Full-text available
Ceratonia siliqua L. is a Mediterranean medicinal plant traditionally cultivated for its ethnopharmacological benefits, such as antidiarrheal, antidiabetic, enhance acetylcholine, antioxidant, antiatherosclerotic, and for its possible anti-neurodegenerative potential. The aim of the present study was to evaluate the chemical composition, as well as the cognitive-enhancing, anxiolytic, and antioxidant activities of the aqueous extract from C. siliqua (CsAE) leaves against 6-hydroxydopamine (6-OHDA) zebrafish Parkinson’s disease (PD) model. CsAE (0.1, 0.3, and 1 mg/L) was administered by immersion to zebrafish (Danio rerio) for eight consecutive days and one hour before each behavioral test of each day, while 6-OHDA (250 µM) treatment was supplied one day before the novel tank diving test (NTT). Qualitative and quantitative analyses were performed by the ultra-high-performance liquid chromatography (UHPLC) analysis. The memory performance was evaluated through the NTT and Y-maze tests. Additionally, the in vitro and in vivo antioxidant status and acetylcholinesterase (AChE) activity was also assessed. Our finds demonstrated that CsAE presented positive antioxidant and anti-AChE activities, which contributed to the improvement of cognitive function in the 6-OHDA zebrafish PD model.
Article
Full-text available
Much evidence suggests that both oxidative stress and apoptosis play a key role in the pathogenesis of Parkinson’s disease (PD). The present study aims to evaluate the protective effect of bergamot juice (BJ) against 6-hydroxydopamine (6-OHDA)- or H2O2-induced cell death. Treatment of differentiated SH-SY5Y human neuroblastoma cells with 6-OHDA or H2O2 resulted in cell death that was significantly reduced by the pre-treatment with BJ. The protective effects of BJ seem to correlate with the reduction of intracellular reactive oxygen species and nitric oxide generation caused by 6-OHDA or H2O2. BJ also attenuated mitochondrial dysfunction, caspase-3 activation, imbalance of pro- and anti-apoptotic proteins, MAPKs activation and reduced NF-ĸB nuclear translocation evoked by neurotoxic agents. Additionally, BJ exhibited excellent antioxidant capability in cell-free assays. Collectively, our results suggest that BJ exerts neuroprotective effect through the interplay with specific cell targets and its antioxidant activity, making it worthy of consideration for the management of neurodegenerative diseases.
Article
6-Hydroxydopamine (6-OHDA) is a neurotoxin that inhibits the mitochondrial complex I causing mitochondrial impairment, aetiology of Parkinson's. Naringenin is a flavanone predominantly present in citrus fruits. Due to its high antioxidant and anti-inflammatory potential, it has been widely studied against various disorders. In this study, the neuroprotective effect of naringenin was determined against 6-OHDA induced toxicity with Levodopa (l-DOPA) as the standard. Naringenin reduced 6-OHDA induced oxidative stress biomarker levels such as CAT, GSH, SOD, and ROS. Naringenin rescued 6-OHDA induced reduction of the mitochondrial membrane potential. Treatment with naringenin improved the locomotion of the 6-OHDA treated zebrafish larvae which showed stagnant swimming patterns. Naringenin was also found to downregulate the expression of some Parkinsonian genes such as casp9, lrrk2, and polg and upregulate pink1. These studies attribute to naringenin as a viable molecule to study further for its neuroprotective effects against 6-OHDA induced neurotoxicity and neurodegeneration.
Article
Neurodegenerative diseases (NDs) are characterized by disorders with progressive deterioration of the structure and/or function of neurons. Genetic mutations can lead to many NDs. Nevertheless, neurodegeneration can also take place due to several biological processes. The pathogenesis of several NDs including Alzheimer's (AD), Parkinson's (PD), and Huntington's (HD) diseases are associated with oxidative stress (OS). In order to maintain the normal functions of neurons, lower levels of reactive oxygen species (ROS) and reactive nitrogen species (RNS) are important, since their increased levels can cause neuronal cell death. It has been found that OS-mediated neurodegeneration involves a number of events including mitochondrial dysfunction, Ca2+ overload, and excitotoxicity. A growing number of studies are suggesting the benefit of using polyphenols for the treatment of neurodegenerative disorders. Indeed, in order to treat most of the NDs, synthetic drugs are extensively used which are found to exert side effects in the course of the treatment. There is mounting evidence that researchers have identified several naturally-occurring chemical compounds in plants, which are used for the management of NDs. Overall, polyphenolic phytochemicals are safer in nature and have negligible side effects. In this article, we have focused on the potential efficacy of polyphenols such as epigallocatechin-3-gallate, curcumin, resveratrol, quercetin and methylated polyphenols berberine against the most common neurodegenerative disorders.