You are on page 1of 16

Journal of Molecular Neuroscience

https://doi.org/10.1007/s12031-018-1181-4

Antioxidant and Anti-Apoptotic Activity of Octadecaneuropeptide


Against 6-OHDA Toxicity in Cultured Rat Astrocytes
Hadhemi Kaddour 1,2,3 & Yosra Hamdi 1 & Fatma Amri 1 & Seyma Bahdoudi 1,4 & Ibtissem Bouannee 1 & Jérôme Leprince 4,5 &
Sami Zekri 6 & Hubert Vaudry 4,5 & Marie-Christine Tonon 4 & David Vaudry 4,5 & Mohamed Amri 1 & Sana Mezghani 1 &
Olfa Masmoudi-Kouki 1

Received: 25 July 2018 / Accepted: 21 September 2018


# Springer Science+Business Media, LLC, part of Springer Nature 2018

Abstract
Oxidative stress, associated with various neurodegenerative diseases, promotes ROS generation, impairs cellular antioxidant
defenses, and finally, triggers both neurons and astroglial cell death by apoptosis. Astrocytes specifically synthesize and release
endozepines, a family of regulatory peptides, including the octadecaneuropeptide (ODN). We have previously reported that ODN
acts as a potent neuroprotective agent that prevents 6-OHDA-induced apoptotic neuronal death. The purpose of the present study
was to investigate the potential glioprotective effect of ODN on 6-OHDA-induced oxidative stress and cell death in cultured rat
astrocytes. Incubation of astrocytes with graded concentrations of ODN (10−14 to 10−8 M) inhibited 6-OHDA-evoked cell death
in a concentration- and time-dependent manner. In addition, ODN prevented the decrease of mitochondrial activity and caspase-3
activation induced by 6-OHDA. 6-OHDA-treated cells also exhibited enhanced levels of ROS associated with a generation of
H2O2 and O2°-, and a reduction of both superoxide dismutase (SOD) and catalase (CAT) activities. Co-treatment of astrocytes
with low concentrations of ODN dose-dependently blocked 6-OHDA-evoked production of ROS and inhibition of antioxidant
enzyme activities. Concomitantly, ODN stimulated Mn-SOD, CAT, glutathione peroxidase-1, and sulfiredoxin-1 gene transcrip-
tion and rescued 6-OHDA-associated reduced expression of endogenous antioxidant enzymes. Taken together, these data
indicate that, in rat astrocytes, ODN exerts anti-apoptotic and anti-oxidative activities, and hence prevents 6-OHDA-induced
oxidative assault and cell death. ODN is thus a potential candidate to delay neuronal damages in various pathological conditions
involving oxidative neurodegeneration.

Keywords Astrocytes . Octadecaneuropeptide . Oxidative stress . Cell death . 6-hydroxydopamine . Neuroprotection

Hadhemi Kaddour and Yosra Hamdi contributed equally to this study.


Abbreviations
* Olfa Masmoudi-Kouki 6-OHDA 6-hydroxydopamine
olfa.masmoudi@fst.utm.tn CNS Central nervous system
DBI Diazepam-binding inhibitor
1
University Tunis El Manar, Faculty of Sciences of Tunis, LR18ES03, DCFH 2′,7′-dichlorodihydrofluorescein
Laboratory of Neurophysiology, Cellular Physiopathology and
JC-1 5,5″,6,6″-tetrachloro-1,1″,3,3″-
Biomelcules Valorisation, 2092 Tunis, Tunisia
2
tetraethylbenzimidazolylcarbocyanine iodide
CIRB, CNRS UMR 7241/INSERM U1050, PSL University, Labex
DHE Dihydroethidium
MemoLife, Collège de France, 11 place Marcelin Berthelot,
75231 Paris, France DHR123 Dihydrorhodamine 123
3 FDA Fluorescein diacetate
Imagine Institute and Center of Psychiatry and Neuroscience,
Université Paris Descartes, 102-108 rue de la Santé, Gpx Glutathione peroxidase
75014 Paris, France H2O2 Hydrogen peroxide
4
UNIROUEN, Inserm U1239, Laboratory of Neuronal and LDH Lactate dehydrogenase
Neuroendocrine Communication and Differentiation, Normandie NBT Nitrotetrazolium blue chloride
Univ, 76000 Rouen, France O2°− Superoxide anions
5
UNIROUEN, Regional Cell Imaging Platform of Normandy ODN Octadecaneuropeptide
(PRIMACEN), Normandie Univ, 76000 Rouen, France PD Parkinson’s disease
6
USCR Transmission Electron Microscopy, Faculty of Medicine, ROS Reactive oxygen species
University Tunis El Manar, Tunis, Tunisia
J Mol Neurosci

SOD Superoxide dismutase 2010). Although activated astrocytes were traditionally


Srxn Sulfiredoxin thought to impede neuronal regeneration by forming glial
scars (Lev et al. 2013; Pekny et al. 2016), growing evidence
indicates that reactive astrocytes contribute to functional re-
Introduction covery after brain injuries (L’ Episcopo et al. 2010;
L’Episcopo et al. 2011). Concurrently, analysis of postmortem
Astrocytes have long been considered as just providing tro- PD brains has revealed a higher neuronal loss within areas
phic support to neurons, but their importance in many func- where astrocytes are less abundant (Gu et al. 2010; Mirza
tions, such as neurotransmission, cell signaling, inflammation, et al. 2000). Moreover, it has been shown that, in rodents,
and synapse modulation is now well established (Dallerac and injection of 6-OHDA within the nigrostriatal pathway pro-
Rouach 2016; Dzyubenko et al. 2016; Garzon et al. 2016; Liu duces a loss of astrocytes associated with the degeneration
and Chopp 2016). Currently, the involvement of astrocyte of dopaminergic neurons (Espinosa-Oliva et al. 2014).
dysfunction in the pathophysiology of neurological disorders, Indeed, astroglial cells are a source of neurotrophic factors
including neurodegenerative diseases and stroke, is highlight- that can protect remaining neurons against oxidative stress
ed (Garzon et al. 2016; Pekny et al. 2016). Parkinson’s disease by detoxifying ROS through antioxidant enzymes and ROS
(PD) is a neurodegenerative disorder that is primarily charac- scavenger molecules (Chen et al. 2005; Lin et al. 2016).
terized by a progressive loss of dopaminergic neurons within Although glial cells are equipped with high levels of protec-
the substantia nigra pars compacta (SNpc) (Michel et al. 2013; tive and antioxidant compounds, astrocyte cell death has been
Toulorge et al. 2016). One of the widely used models of PD reported in human and animal models of brain injuries (Ishii
involves treatment with 6-hydroxydopamine (6-OHDA), a et al. 2017; Liu et al. 2016). As loss of glial cells may strongly
neurotoxin that selectively damages catecholaminergic neu- affect neuronal survival, protection of astrocytes from oxida-
rons both in vivo and in vitro (Blesa and Przedborski 2014). tive insults appears essential to maintain brain functions.
In vitro experiments show that the sensitivity of neurons to 6- The octadecaneuropeptide (ODN) is a peptide generated
OHDA is mainly associated with an auto-oxidation-mediated through the proteolytic cleavage of the 86-amino acid precur-
overproduction of reactive oxygen species (ROS), such as sor diazepam-binding inhibitor (DBI) (Guidotti et al. 1983),
hydrogen peroxide (H2O2), superoxide anion (O2°-), and hy- which is exclusively expressed in astroglial cells in the central
droxyl radical (OH°) (Han et al. 2014; Soto-Otero et al. 2000). nervous system of mammals (Tonon et al. 2006; Tonon et al.
The main cellular defense mechanism to cope with oxidative 2013). DBI and its derived peptides, including ODN, belong
stress is based on antioxidant molecules and enzymes, i.e., to the endozepine family. The primary structure of ODN has
glutathione, catalase, superoxide dismutases (SODs), glutathi- been strongly preserved during evolution, suggesting that this
one peroxidase (Gpx), and sulfiredoxin1 (Srxn1), which can peptide exerts a wide range of biological functions. In fact,
prevent cell oxidative stress damage (Afshin-Majd et al. 2017; ODN is involved in the control of food intake, sleep, aggres-
Kiasalari et al. 2016; Zhou et al. 2015a), and thus play a major siveness, and anxiety (Tonon et al. 2013). At the cellular level,
neuroprotective role against the deleterious effects of 6- numerous data indicate that ODN acts both as an autocrine
OHDA. Treatment of neuroblastoma cell lines with and paracrine factor modulating proliferation and differentia-
astrocyte-conditioned medium, significantly increases the re- tion of astrocytes and neurons (Alfonso et al. 2012; Dumitru
sistance of neurons to oxidative stress induced by 6-OHDA et al. 2017). In addition, ODN exerts a potent protective effect
(Murakami et al. 2018). It has been shown that the reduction against the deleterious action of oxidative stress in cultured
of neuronal cell death induced by astrocyte-conditioned me- astrocytes (Ghouili et al. 2018; Hamdi et al. 2011; Hamdi et al.
dium is associated with a stimulation of glutathione peroxi- 2012) and neurons (Kaddour et al. 2013). More recently, the
dase expression, an increase of glutathione levels and activa- deficiency of the ODN precursor, in an in vivo model of PD,
tion of the production of neuroprotective factors (Gardaneh was reported to increase sensitivity of dopaminergic neurons
et al. 2011; Ghouili et al. 2018; Sandhu et al. 2009). These in SNpc and ventral tegmental area (VTA) to MPTP neuro-
data highlight the role of astrocyte-derived factors to maintain toxicity (Bahdoudi et al. 2018). Thus, ODN may be an endog-
cerebral antioxidant competences and to prevent neuronal enous peptide with neuroprotective and antioxidant properties
damages. on astrocytes.
Activation of glia, which is a common pathological feature It is well established that 6-OHDA provokes cell apoptosis
of multiple neurodegenerative conditions (Burda et al. 2016; through the production of ROS, leading to apoptosis and ac-
Lev et al. 2013; Osborn et al. 2016), is characterized by a tivation of caspase-3 in both neurons and glial cells
series of metabolic and morphological changes of astrocytes, (Hernandez-Baltazar et al. 2013; Mladenovic et al. 2004).
and is notably observed in the substantia nigra and cerebral Since, the gliopeptide ODN protects granule neurons against
cortex of human PD brains as well as in 6-OHDA-treated apoptosis induced by 6-OHDA exposure (Kaddour et al.
animals, a model of PD (Gomide et al. 2005; Wachter et al. 2013), we hypothesized that ODN may also protect astrocytes
J Mol Neurosci

against 6-OHDA oxidative insult. Therefore, the purpose of glutamine, 1‰ insulin, 5 mM HEPES, 0.4% glucose, and
the present study was to examine the potential protective ac- 1% of the antibiotic-antimycotic solution. The tissues were
tion of ODN against 6-OHDA-induced astrocyte cell death dissociated mechanically with a syringe equipped with a 1-
and to investigate the effects of the peptide on some oxidative mm gauge needle, and filtered through a 100-μm sieve
stress-related parameters. (Falcon, Franklin Lakes, NJ, USA). Dissociated cells were
resuspended in culture medium supplemented with 10%
FBS, plated in 75-cm2 flasks (Greiner Bio-one GmbH,
Materials and Methods Frickenhausen, Germany) and incubated at 37 °C in a 5%
CO2/95% air. When cultures were confluent, astrocytes were
Animals isolated by shaking overnight the flasks with an orbital agita-
tor. Adhesive cells were detached by trypsination and
Wistar rats (Pasteur Institute, Tunis, Tunisia, and Charles preplated for 5 min to discard contaminating microglial cells.
River Laboratories, St Germain sur l’Arbresle, France) were Then, the non-adherent astrocytes were harvested and plated
kept in a temperature-controlled room (21 ± 1 °C) under an on 35-mm Petri dishes or 24-well plates at a density of 42,000
established photoperiod (lights on from 7:00 am to 7:00 pm) cells/cm2. After 5 days (DIV5), more than 98% of the cells
with free access to food and water. Experiments were per- were labeled with antibodies against glial fibrillary acidic pro-
formed in accordance with the American Veterinary Medical tein (Douiri et al. 2016). All experiments were performed on
Association. Approval for these experiments was obtained 5- to 7-day-old secondary cultures.
from the Medical Ethical Committee For the Care and Use
of Laboratory Animals of Pasteur Institute of Tunis Cell Cytotoxicity Measurement
(Approval No. FST/LNFP/Pro152012).
Cultured astrocytes seeded into 24-well plates were incubated
Chemicals at 37 °C with fresh serum-free culture medium in the absence
or presence of test substances. At the end of the incubation,
Dulbecco’s modified Eagle’s medium (DMEM), D(+)-glu- membrane integrity was assessed as a function of the amount
cose, F-12, L-glutamine, foetal bovine serum (FBS), N-2- of cytoplasmic LDH released into the medium with a LDH
hydroxyethylpiperazine-N-2-ethane sulfonic acid buffer solu- assay kit, according to the manufacturer’s instructions. LDH
tion (HEPES), antibiotic-antimycotic solution, and trypsin- activity was measured at 340 nm with a spectrophotometric
EDTA were purchased from Gibco (Invitrogen, Grand Island microplate reader (Bio-Rad Laboratories, Philadelphia, USA).
NY, USA). 6-OHDA, fluorescein diacetate-acetoxymethyl es- The results were expressed as a percentage of total LDH re-
ter (FDA-AM), bovine serum albumin (BSA), lease after cell lysis with 1% Triton X-100 in saline phosphate
nitrotetrazolium blue chloride (NBT), Triton X-100, bovine buffer (PBS, 0.1 M, pH 7.4).
liver catalase, DL-epinephrine, dimethylsulfoxyde (DMSO),
dihydrorhodamine 123 (DHR123), lactate dehydrogenase Cell Survival Measurement
(LDH) assay kit, and insulin were obtained from Sigma-
Aldrich (St. Louis, MO, USA). 5-6-chloromethyl-2′,7′- Cultured astrocytes seeded into 24-well plates were incubated
dichlorodihydrofluorescein diacetate, acetyl ester (CM- at 37 °C with fresh serum-free culture medium in the absence
H2DCFDA), dihydroethidium (DHE), and 5,5″,6,6″- or presence of test substances. Cell survival was quantified by
tetrachloro-1,1″,3,3″-tetraethylbenzimidazolylcarbocyanine measuring FDA in cultured astrocytes. At the end of the incu-
iodide (JC-1) were from Molecular Probes (Eugene, Oregon, bation, cells were incubated in the dark with FDA-AM (15 μg/
USA). The Apo-ONE homogeneous caspase-3/7 assay kit mL, 8 min, 37 °C), rinsed twice with PBS (0.1 M, pH 7.4), and
was purchased from Promega (Charbonnières, France). Rat lysed with a 10 mM Tris–HCl solution containing 1% sodium
ODN (QATVGDVNTDRPGLLDLK) was synthesized by dodecyl sulfate (SDS). Fluorescence intensity (λ excitation =
using the standard fluorenylmethyloxycarbonyl (Fmoc) pro- 485 nm and λ emission = 538 nm) was measured with a
cedure, as previously described (Leprince et al. 2001). FL800TBI fluorescence microplate reader (Bio-Tek
Instruments, Winooski, VT, USA).
Secondary Culture of Cortical Rat Astrocytes
Measurement of Mitochondrial Activity
Secondary cultures of rat cortical astrocytes were prepared
from newborn Wistar rats of both sexes as previously de- Cultured astrocytes seeded into 24-well plates were incubated
scribed (Douiri et al., 2016) with minor modifications. at 37 °C with fresh serum-free culture medium in the absence
Briefly, cerebral hemispheres were collected in DMEM/F12 or presence of test substances. Mitochondrial membrane po-
(2:1; v/v) culture medium supplemented with 2 mM tential was quantified using the JC-1 probe. At the end of the
J Mol Neurosci

experiments, astrocytes were treated for 15 min with the JC-1 Intracellular Superoxide Measurement
probe (10 μg/mL) and then washed twice with PBS.
Fluorescence intensity was measured with a FL800TBI fluo- Superoxide levels were detected by measuring the fluores-
rescence microplate reader and expressed as a ratio of the cence of ethidium, which derived from the oxidation of the
fluorescence emission at 590 nm (orange, intact mitochondrial non-fluorescent compound dihydroethidium (DHE).
membrane potential) versus 530 nm (green, collapsed mito- Astrocytes were incubated at 37 °C with fresh serum-free
chondrial membrane potential). medium in the absence or presence of 6-OHDA with or with-
out ODN. At the end of the incubation, cells were incubated
Caspase-3 Activity Measurement for 20 min in fresh medium containing 2 μM DHE at 37 °C
and then washed twice with PBS. Ethidium fluorescence (λ
Cultured astrocytes seeded into 24-well plates were incubated excitation = 520 nm and λ emission = 610 nm) was measured
at 37 °C with fresh serum-free culture medium in the absence with a FL800TBI microplate reader, and data were analyzed
or presence of test substances. At the end of the experiments, by Gen5 Data Analysis Software.
cells were washed twice with PBS and caspase-3 activity was
measured with a caspase-3 assay system (Apo-ONE Respiratory Burst Assay
Homogeneous Caspase-3/7 kit, Promega). Cells were resus-
pended in DMEM (100 μL) mixed with 1X caspase assay The formation of the O2°−, produced by respiratory burst, was
buffer containing 25 μM caspase-3 substrate. Caspase-3 ac- detected by the reduction of nitroblue tetrazolium (NBT) to
tivity was calculated from the slope of the fluorescence mea- dark-blue formazan deposits. Astrocytes were incubated at
sured every 15 min for 3 h with excitation at 485 nm and 37 °C with fresh serum-free medium in the absence or pres-
emission at 530 nm, and expressed as a percentage of the ence of 6-OHDA with or without ODN. At the end of the
control. incubation, cells were washed once with PBS at 37 °C, and
then incubated for 2 h in the dark with a reaction mixture
Intracellular Reactive Oxygen Species Measurement containing NBT (1 mg/mL) and BSA (1 mg/mL). To visualize
the formation of blue crystals, cells were examined and im-
ROS were detected by measuring the fluorescence of 2′,7′- ages were acquired with an eclipse E-600 microscope (Nikon,
dichlorodihydrofluorescein (DCF) resulting from the Champigny-sur-Marne, France) equipped with a 3 CCD Sony
deacetylation and oxidation of the non-fluorescent compound DXC950 camera interfaced with the Visiolab computerized
DCFH2-DA. Cells seeded into 24-well plates were exposed to program (Biocom, Les Ulis, France).
6-OHDA with or without ODN, incubated with 10 μM cell-
permeant DCFH2DA in serum-free loading medium for Measurement of Antioxidant Enzyme Activities
30 min at 37 °C and then washed twice with PBS. DCF fluo-
rescence (λ excitation = 485 nm and λ emission = 538 nm) Cultured cells seeded into 35-mm Petri dishes were incubated
was measured with a FL800TBI fluorescence microplate read- at 37 °C for 72 h with fresh serum-free medium in the absence
er, and data were analyzed by Gen5 Data Analysis Software. or presence of test substances. At the end of the incubation,
cells were washed twice with PBS and total cellular proteins
Intracellular Peroxide Measurement were extracted by using the lysis buffer containing 50 mM
Tris–HCl (pH 8), 10 mM EDTA, 100 μM phenylmethyl-
H 2 O 2 and/or OH ° generation were detected by using sulfonylfluoride, and 1% Triton X-100. The homogenate
DHR123, which is a cell-permeable non-fluorescent com- was centrifuged (16,000 g, 4 °C, 20 min), and the cellular
pound that is oxidized by cellular peroxides to fluorescent extract contained in the supernatant was stored at − 20°C until
rhodamine 123 (Rh 123) and exhibits green fluorescence. enzyme activity determinations.
Astrocytes were incubated at 37 °C with fresh serum-free SOD activity was measured using a spectrophotometric
medium in the absence or presence of 6-OHDA with or with- assay, which consists in measuring epinephrine autoxidation
out ODN. At the end of the incubation, cultures were washed induced by superoxide anion. Samples, prepared as described
twice with PBS, to prevent oxidation of DHR123 by perox- above, were incubated for 3 min with a mixture containing
ides in the medium, and incubated at 37 °C for 15 min in fresh bovine catalase (0.4 U/μL), DL-epinephrine (5 mg/mL), and
medium containing 6 μM DHR123, and then washed twice Na2CO3/NaHCO3 buffer (62.5 mM, pH 10.2). The oxidation
with PBS. Rh 123 fluorescence (λ excitation = 505 nm and λ of epinephrine was measured at 480 nm with a Bio-Rad spec-
emission = 529 nm) was measured with a FL800TBI micro- trophotometric microplate reader.
plate reader, and data were analyzed by Gen5 Data Analysis Catalase activity was determined on the basis of the de-
Software. crease of H2O2. Samples, prepared as described above, were
J Mol Neurosci

mixed with 30 mM H2O2 in PBS. The disappearance of H2O2 Results


was measured at 240 nm for 180 s at 30-s intervals.
Glioprotective Effect of ODN Against 6-OHDA-Induced
Quantitative PCR Analysis Astroglial Cell Death

Cultured cells seeded into 35-mm Petri dishes were incubated Administration of 6-OHDA on cultured astrocytes in-
at 37 °C with fresh serum-free medium. At the end of the duced a time- and dose-dependent cell death (Fig. 1a,
incubation, the culture medium was removed and cells b). The concentration of 120 μM 6-OHDA, which killed
were washed twice with PBS (0.1 M, pH 7.4). Total 50% of the cells within 72 h of treatment, was used in
RNA was extracted by using Tri reagent (Sigma, St subsequent experiments. In these conditions, co-
Quentin Fallavier, France) and purified using the incubation of cells with 6-OHDA and graded concentra-
NucleoSpin kit (Macherey-Nagel, Hoerd, France). tions of ODN (10−14 to 10−8 M) provoked mirror pro-
cDNA was synthetized from 3 to 4 μg of total RNA tective effects on astroglial survival and LDH leakage
with ImProm II Promega kit (Promega). Quantitative (Fig. 1c). The glioprotection of ODN was concentration-
RT-PCR was performed on cDNA in the presence of a dependent in the range 10−11 to 10−8 M, and a complete
1× Fast SYBR Green universal PCR Master mix reversal of the toxic effect of 6-OHDA was obtained
(Applied Biosystems, Courtaboeuf, France) containing with subnanomolar concentrations of ODN (Fig. 1c).
dNTPs, MgCl2, SYBR green reporter dye, and Incubation of astrocytes with ODN alone did not affect
AmpliTaq Gold DNA polymerase, with forward and re- cell survival whatever the time or the dose.
verse primers (Table 1), using an ABI Prism 7500
Sequence Detection System (Applied Biosystems). The ODN Exerts its Glioprotective Effect
relative amount of cDNA in each sample was calculated Through the Intrinsic Mitochondrial and Caspase-3
using the comparative quantification cycle (Cq) method Pathway
and expressed as 2−ΔΔCq using GAPDH as an internal
control. As previously reported in different types of cells (Jordan et al.
2004; Perfeito et al. 2013), 6-OHDA inhibited the mitochondrial
Statistical Analysis electron transport chain, measured with the fluorescent
ratiometric probe JC-1 (Fig. 2a, b), and caused caspase-3 acti-
Data are expressed as the mean ± SEM from three indepen- vation (Fig. 2c) in a dose-dependent manner in astrocytes, phe-
dent experiments. Statistical analysis of the data was per- nomena which lead to typical apoptotic cell death. The present
formed by using ANOVA, followed by Bonferroni’s test. A data showed that addition of glioprotective doses of ODN to the
p value of 0.05 or less was considered as statistically incubation medium (10−10 and 10−9 M) totally suppressed the
significant. deleterious effect of 6-OHDA on mitochondrial membrane

Table 1 Sequences of the primers


used for real-time PCR Gene GenBank accession number Sequence
experiments
Mn-SOD NM_017051.2
Forward 5-TGGACAAACCTGAGCCCTAA-3
Reverse 5-GACCCAAAGTCACGCTTGATA-3
CAT NM_012520.2
Forward 5-ATCAGGGATGCCATGTTGTT-3
Reverse 5-GGGTCCTTCAGGTGAGTTTG-3
Gpx1 NM_030826.4
Forward 5-GACATCAGGAGAATGGCAAGA-3
Reverse 5-CACCTCGCACTTCTCAAACA-3
Srxn1 NM_001047858.3
Forward 5-CAACGTGCCAATCGCCGTGC-3
Reverse 5-GGGTCCTCCAGGATCGTGTCCA-3
GAPDH NM_017008.4
Forward 5-CAGCCTCGTCTCATAGACAAGATG-3
Reverse 5-CAATGTCCAACTTTGTCACAAGAGAAA-3
J Mol Neurosci

a 10−9 M ODN totally blocked toxin-induced caspase-3 activation


125 **** 100 (Fig. 2d).
****

LDH release (% of total LDH)


Cell viability (% of control)

100 80
ODN Prevents the Effect of 6-OHDA on Intracellular
* ROS Accumulation
75 * 60
*
It is known that the toxic effect of 6-OHDA is linked to an
50 40
**** over-generation of ROS, such as H2O2, O2°- and OH° (Blum
et al. 2000; Elkon et al. 2004). To examine whether ODN
****
25 20 could block 6-OHDA-induced intracellular ROS accumula-
6-OHDA (120 µM) - + + + + -
tion, astrocytes were first labeled with the CMH2DCFDA
24h 48h 72h 96h
probe, which generates the fluorescent compound DCF upon
b ****
oxidation with ROS. Incubation of cells with graded concen-
100 **** 100 trations of 6-OHDA (10 to 200 μM) for 72 h induced a dose-
dependent increase in DCF fluorescence intensity (Fig. 3a).

LDH release (% of total LDH)


Cell viability (% of control)

**
80 80 For doses of 10−10 and 10−9 M, ODN did not modify DCF
***
fluorescence intensity, but totally abolished the effect of
60 60 120 μM 6-OHDA on DCF formation (Fig. 3b).
*
**** We also examined the effect of ODN on 6-OHDA-induced
40 ****
40 H2O2 formation in astrocytes by measuring DHR123 fluores-
cence intensity. Treatment of astroglial cells with toxic con-
20 20
centrations of 6-OHDA (60 to 200 μM) induced a dose-
dependent increase in DHR123 fluorescence intensity
10 20 30 60 120 200
6-OHDA [µM]
(Fig. 3c). Co-treatment of the cells with ODN (10−10 or
c 10−9 M) totally suppressed the effect of the toxin (10 μM to
100 100
120 μM) on peroxide production (Fig. 3c–d).
LDH release (% of total LDH)

####
Cell viability (% of control)

####
80
#
75 ##

ODN Prevents the Effect of 6-OHDA on Superoxide


**
***
60 Radical Generation
***
50 ****
40 Since large amounts of O2°− are produced by respiratory burst
under a drastic oxidative stress status, we have investigated
25 20 the effect of ODN on 6-OHDA-induced O2°− formation by
-14 -13 -12 -11 -10 -9 -8
astroglial cells. Control astrocytes exhibited very few blue
log M [ODN] precipitates (reflecting reduction of NBT by O2°- production)
Fig. 1 Glioprotective effect of ODN on 6-OHDA-induced astrocyte cell in cell bodies (Fig. 4a). Treatment of astrocytes with 120 μM
death. a Effect of 6-OHDA (120 μM) on astrocyte survival after 24, 48, 6-OHDA resulted in a marked increase of the respiratory burst
72, and 96 h of treatment. b Effect of graded concentrations of 6-OHDA as shown by the labeling of most cell bodies in blue (Fig. 4a).
(10 to 200 μM) on astrocyte survival after 72 h. c Effect of graded In contrast, only few cells exhibited blue labeling after co-
concentrations of ODN (10−14 to 10−8 M) on 6-OHDA (120 μM)-induced
astrocyte cell death after 72 h of treatment. Cell survival (___) was quan- administration of ODN (10−10 M) and 6-OHDA (120 μM)
tified by measuring FDA fluorescence intensity, and the results are (Fig. 4a). Quantitative analysis using the DHE probe indicated
expressed as percentages of the control (empty circle). Cell death (- - -) that incubation of cultured astrocytes with graded concentra-
was determined by measuring LDH activity in culture medium, and the tions of 6-OHDA (10–200 μMM) for 72 h, induced a dose-
results are expressed as percentages of total LDH released from Triton-
lysed cells (gray triangle). Each value is the mean (± SEM) from at least dependent increase in DHE fluorescence intensity (Fig. 4b).
18 different wells from 3 independent cultures. ANOVA followed by Addition of ODN (10−10 M) to the incubation medium totally
Bonferroni’s test: *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001 vs. blocked the effect of moderate concentrations of 6-OHDA
control. #P < 0.05; ##P < 0.01; ####P < 0.0001 vs. 6-OHDA-treated cells (from 30 to 120 μM) and partially reversed the effect of a
higher concentration of 6-OHDA (200 μM) on DHE forma-
potential (Fig. 2b). To further explore the mechanism involved tion (Fig. 4b). In addition, co-treatment of the cells with ODN
in the protective action of ODN, we have monitored its effect on (10−10 or 10−9 M) restored the O2°− levels to the control values
caspase-3 activity. Co-treatment of the cells with 10−10 and in 120 μM 6-OHDA-treated cells (Fig. 4c).
J Mol Neurosci

Control Control
a 6-OHDA
6 OHDA b 6-OHDA
6 OHDA
6 OHDA + ODN
6-OHDA 6-OHDA + ODN
125 # ### ### ### 12
125

ontrrol)
ontrrol)

####
100 100 ####

( of co
atio (% of co

*
*

m ratio (%
75 *** 75
###
m ra

50 50

30 nm
30 nm

**** ***

0/53
0/53

25 **** 25

590
590

5
5

0 0
ODN (10-10
10M) - - + - + - + - + - + 6-OHDA
O ((120 µM)) - + + + + +
6-OHDA
6 OHDA 6
6-OHDA
OHDA 66-OHDA
OHDA 66-OHDA
OHDA 6 6-OHDA
OHDA -12 -10 -9 -8
(10 µM) (30 µM) (60 µM) (120 µM) (200 µM) log M [ODN]

Control
C t l Control
C t l
6-OHDA 6-OHDA
c 6-OHDA + ODN d 6-OHDA + ODN

# ### #### ###

250 ****
ontrrol)

250
ontrrol))

**** ****
3 activity (% of co
y (% of co

200 200
***
*
##

150 * 150
vity
3 activ

#### ####
100 100
se-3
pase-3

pas

50 50
asp
asp

Ca
Ca

0 0
ODN (10-10M) - - + - + - + - + - + 6-OHDA (120 µM) - + + + + +
6-OHDA 6-OHDA 6-OHDA 6-OHDA 6-OHDA -12 -10 -9 -8
(10 µM) (30 µM) (60 µM) (120 µM) (200 µM) log M [ODN]

Fig. 2 Effect of ODN on 6-OHDA-induced impairment of mitochondrial probe and the ratio of fluorescence emissions 590/530 nm was measured
transmembrane potential and activation of caspase-3 in astrocytes after as an index of mitochondrial activity. Caspase-3 activity was quantified
72 h of treatment. a, c Effect of graded concentrations of 6-OHDA (10 to by measuring the fluorescence of the caspase substrate, Z-DEVD-
200 μM) on mitochondrial transmembrane potential and caspase-3 activ- Rhodamine 110. The results are expressed as percentages of the control.
ity in astrocytes. b, d Effect of graded concentrations of ODN (10−12 to Each value is the mean (± SEM) of at least 12 different wells from 3
10−8 M) on 6-OHDA (120 μM)-induced impairment of mitochondrial independent cultures. ANOVA followed by Bonferroni’s test: *P < 0.05;
***
transmembrane potential and activation of caspase-3 in astrocytes. Cells P < 0.001; **** P < 0.0001 vs. control. # P < 0.05; ## P < 0.01;
###
mitochondrial transmembrane potential was assessed by using the JC-1 P < 0.001; ####P < 0.0001 vs. 6-OHDA-treated cells
J Mol Neurosci

a b Control
250 Control 250 6-OHDA
6 OHDA
****
6O
6-OHDA 6-OHDA + ODN

n
ation
on
ulattio 6-OHDA + ODN
#
200 **** 200 ****
ns

umula
#

ntrrol))
mu

* #
ol)
cum

**

ccu
onttro

150 * # 150 # #

con
#
acc

OS ac
ns # # # #
ns
% of co

o c
Sa

# # #

% of
C lullarr ROS

RO
100

ar R
100

(%
(%

ula
ellu
Ce
50
Cel

50

0 0
ODN (10-10M) - - + - + - + - + - + 6-OHDA - + + + + +

6
6-OHDA
OHDA 10 µM 30 µM 60 µM
M 120 µM
M 200 µM
M (120 µM) -12
12 -10
10 -9 9 -88
log M [ODN]

c d
Control Control
6 OHDA
6-OHDA 6 OHDA
6-OHDA
ntrrol))

2 0
250 250
*** ntrrol))
6-OHDA + ODN 6-OHDA + ODN
con

#
con
o c

200 # 200 ***


% of

o c

**** # #
****
% of
nsity (%

** # #
nsiity (%

#
# # #
150 # 150 #
# # #
#
3 intten

ns ns # # #
#
DHR 123 intten

100 100
R1
DHR 123

50 50
D

0 0
ODN (10-10 M) - - + - + - + - + - + 6 OHDA
6-OHDA - + + + + +
6-OHDA
6 OHDA M
10 µM 30 µM 60 µM
M 120 µM
M 200 µM
M (120 µM) 12 -10
-12 10 -99 -8
8
log M [ODN]
Fig. 3 Effect of ODN on 6-OHDA-induced intracellular generation of DCF and DHR 123 fluorescence, respectively. The results are expressed
ROS and accumulation of H2O2. Cells were incubated for 72 h with as percentages of control. Each value is the mean (± SEM) of at least 12
graded concentrations of 6-OHDA (10 to 200 μM) in the absence or different wells from 3 independent cultures. ANOVA followed by
presence of ODN (10−10 M; a, c). Cells were co-incubated with graded Bonferroni’s test: *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001 vs.
concentrations of ODN (10−12 to 10−8 M) and 6-OHDA (120 μM; b, d). control. #P < 0.05; ##P < 0.01; ###P < 0.001; ns, not statistically different
Cellular ROS and H2O2 generation were quantified by measurement of vs. 6-OHDA-treated cells

ODN Blocks 6-OHDA-Induced Inhibition generated by 6-OHDA, we monitored the activities of


of Antioxidant Enzyme Activities in Cultured the two antioxidant enzymes SOD and catalase in astro-
Astrocytes cytes. Incubation of cultured cells with graded concen-
trations of 6-OHDA (10–200 μM; 72 h) induced a
To further explore the mechanism involved in the pro- dose-dependent decrease of both SOD (Fig. 5a) and
tective action of ODN against the oxidative stress catalase (Fig. 5b) activities. At higher concentrations
J Mol Neurosci

Control
a b
6-OHDA
250 6-OHDA
6 OHDA + ODN

c ntrrol)
Control
200 **** ****

o co
****
***

% of
#
# #

sitty ((%
150 ns # # #
# #

ens
100

nte
E in
DHE
50

DH
6-OHDA
6 OHDA (120 µM)
0
ODN (10-100M) - - + - + - + - + - +
6-OHDA 10 µM 30 µM 60 µM 120 µM 200 µM

c
250
ntrroll)

***
ns
200
con
% of c

#
6 OHDA+ODN (10-10 M)
6-OHDA+ODN #
#
150 #
y ((%

#
#
sity
ens

100
nte
HE in

50
DH
D

0
6-OHDA - + + + + +
(120 µM) -12
12 -10
10 -99 -8
8
log M [ODN]
Fig. 4 Effect of ODN on 6-OHDA-induced superoxide radical produc- 200 μM) in the presence or absence of ODN (10−10 M) (b). Cells were
tion. a Phase-contrast images illustrating the generation of superoxide incubated for 72 h with 120 μM 6-OHDA in the absence or presence of
radicals visualized with the presence of a dark-blue precipitate inside graded concentrations of ODN (10−12 to 10−8 M) (c). The results are
the cells. Cells were incubated for 72 h with medium alone (control) or expressed as percentages of control. Each value is the mean (± SEM) of
with 120 μM 6-OHDA alone or in the presence of ODN (10−10 M). Scale at least 12 different wells from 3 independent experiments. ANOVA
bar, 100 μm. b, c Cellular superoxide radical levels were quantified by followed by the Bonferroni’s test. ***P < 0.001; ****P < 0.0001 vs. con-
measurement of the fluorescence of oxidized dihydroethidium (DHE). trol. #P < 0.05; ###P < 0.001 vs. 6-OHDA-treated cells
Cells were incubated with increasing concentrations of 6-OHDA (10 to

of 6-OHDA tested, ODN (10−10 M) restored SOD and the inhibitory action of 120 μM 6-OHDA on the activity of
catalase activities to control values (Fig. 5a, b). endogenous antioxidant systems SOD (Fig. 5c) and catalase
Similarly, addition of graded concentrations of ODN (Fig. 5d).
(10−12–10−9 M) totally blocked in a dose-dependent manner
J Mol Neurosci

a b
#
200 # 150 #

ntrol))
ntrroll) 6-OHDA ** 6-OHDA
#
#
#
*

on
6-OHDA + ODN 6-OHDA + ODN
*

% off co
con

150 125
( off c

y (%
y (%

vity
100 100
vity

a tiv
ctiv

e act
Daac

alase
50 * 75
SOD

**** **** **** *

Cata
S

Ca
0 50
10 20 30 60 120 200 10 20 30 60 120 200
6 OHDA [µM]
6-OHDA 6 OHDA [µM]
6-OHDA

Control Control
c 6-OHDA d 6 OHDA
6-OHDA
6 OHDA + ODN
6-OHDA 6-OHDA + ODN
#
# #
ol))

100 150 #
ol)

# #
ntro

#
onttro

# #
# #
on

#
% off co
o co

80 125
% of

#
# #
y (%
ctiivity (%

60 100
a tivity

***
**
e act

40 75
D ac

se
SOD

alas
SO

20 50
ata
ca

0 0
6-OHDA
6 OHDA - + + + + + + 6-OHDA
6 OHDA - + + + + + +
(120 µM) -12
12 -11
11 -10
10 -9
9 -8
8 (120 µM) -12
12 -11
11 -10
10 -9
9 -8
8
log
g M [[ODN]] log M [ODN]

Fig. 5 Effect of ODN on modulation of SOD and catalase activities in activity by the decrease of H2O2. The results are expressed as percentages
cultured rat astrocytes. Cells were incubated with increasing of control. Each value is the mean (± SEM) calculated from at least 12
concentrations of 6-OHDA (10–200 μM) in the presence or absence of different dishes from 3 independent cultures. ANOVA followed by the
ODN (10−10 M; a, b). Cells were incubated for 72 h with 120 μM of 6- Bonferroni’s test. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001 vs.
OHDA in the absence or presence of graded concentrations of ODN control. #P < 0.05; ##P < 0.01; ###P < 0.001 different vs. 6-OHDA-treated
(10−12 to 10−8 M; c, d). SOD activity was determined by measurement cells
of epinephrine autoxidation induced by superoxide anion, and catalase

ODN Inhibits 6-OHDA-Evoked Antioxidant Gene of real-time PCR. Consistent with the literature (Khan
Expression et al. 2010; Tuncel et al. 2012), the mRNA levels of
Mn-SOD (− 0.64%, ***P < 0.001), catalase (CAT; −
Since 6-OHDA induces oxidative insults that facilitate 0.46%, **P < 0.01), and glutathione peroxidase (Gpx1,
neurotoxicity, we examined the effect of ODN on the −0.32%, *P < 0.05) were markedly decreased 6 h after
expression of genes implicated in the control of oxida- the beginning of the 6-OHDA treatment (Fig. 6a, c). As
tive metabolism in 6-OHDA-treated astrocytes by means illustrated in Fig. 6a, c, ODN alone, at a concentration
J Mol Neurosci

Control 6-OHDA Control 6-OHDA


a ODN 6-OHDA + ODN b ODN 6-OHDA + ODN
3 ###
3 *** ### ***
mRNA Mn-SOD levels ** ***

mRNA CAT levels


(% of control)
(% of control)
2 2

1 1
*** **

0 0
ODN (10-10M) - + - + ODN (10-10M) - + - +
6-OHDA (120 µM) 6-OHDA (120 µM)

c Control 6-OHDA d Control 6-OHDA


ODN 6-OHDA + ODN ODN 6-OHDA + ODN
###
4 4
*** ***
***
mRNA Srxn1 levels
###
mRNA GPx-1 levels

3 **
(% of control)
3
(% of control)

2 2

1 ** 1 **

0 0
ODN (10-10M) - + - + ODN (10-10M) - + - +

6-OHDA (120 µM) 6-OHDA (120 µM)

Fig. 6 Effect of ODN on 6-OHDA-induced inhibition of Mn-SOD, cat- d) mRNA levels were measured by quantitative RT-PCR. Data were
alase, glutathione peroxidase, and sulfiredoxin-1 mRNA levels in cul- corrected using the GAPDH signal as an internal control and the results
tured rat astrocytes. Cultured astrocytes were incubated for 6 h with are expressed as a percentage of controls. Each value is the mean (± SEM)
medium alone, 10−10 M ODN, 120 μM 6-OHDA, or 120 μM 6-OHDA of at least four different wells from three independent experiments.
plus 10−10 M ODN. Mn-Superoxide dismutase (Mn-SOD; a), catalase ANOVA followed by the Bonferroni’s test. **P < 0.01; ***P < 0.001 vs.
(CAT; b), glutathione peroxidase (Gpx1; c), and sulfiredoxin-1 (Srxn1 control. ###P < 0.001 different vs. 6-OHDA-treated cells

of 10−10 M, provoked a significant increase in Mn-SOD restored Srxn1 mRNA expression above control levels in 6-
(+ 1.75%, ***P < 0.001), CAT (+ 1.33%, ***P < 0.001), OHDA-treated astrocytes (Fig. 6d).
and Gpx-1 (+ 1.82%, ***P < 0.001) and restored Mn-
SOD, CAT, and Gpx1 mRNA levels above control
levels in 6-OHDA-treated astrocytes.
Since sulfiredoxin-1 (Srxn1) is a redox sensor that regulates Discussion
key cellular processes including antioxidant defense and cell
protection (Li et al. 2013; Wu et al. 2017), we examined the It has already been reported that oxidative stress causes apo-
effect of ODN on the expression of Srxn1 gene under oxidative ptosis in various cell types, including neuronal cells (Amri
stress provoked by 6-OHDA. RT-qPCR experiments showed et al. 2017; Ishii et al. 2017; Tronel et al. 2013; Yang et al.
that 6-OHDA (120 μM) induced a decrease of Srxn1 gene 2017), and we have previously shown that ODN protects cer-
transcription (− 0.32%, **P < 0.01). Interestingly, in ODN- ebellar granule neurons against 6-OHDA-induced apoptosis
treated astrocytes a strong stimulation of Srxn1 gene transcrip- (Kaddour et al. 2013). Here, we demonstrate that ODN pro-
tion was observed (+ 2.3%, ***P < 0.001). Furthermore, the tects astroglial cells against 6-OHDA-induced cell death
addition of ODN (10−10 M) to the culture media completely through stimulation of endogenous antioxidant systems
J Mol Neurosci

leading to inhibition of ROS accumulation and caspase-3 suggest that ODN inhibits 6-OHDA-induced astrocyte cell
activation. death through activation of its metabotropic receptor, and
6-OHDA, a neurotoxin classically used to mimic PD that attenuation of ODN action at high doses might in part
in vivo and in vitro, is usually thought to cross the cell mem- be due to desensitization its receptor.
brane through dopamine uptake transporters but has also been It has previously been reported that extracellular auto-
shown to induce apoptosis in various cell types that do not oxidation of 6-OHDA produces a large array of highly reac-
express dopaminergic transporters, such as PC12 cells (Blum tive species such as H2O2, O2°- and OH° which are likely
et al. 2000). In agreement with this, we observed that exposure responsible for part of the toxicity of the molecule (Raicevic
of cultured astrocytes to 6-OHDA concentrations above et al. 2005a; Raicevic et al. 2005b). Nevertheless, it has also
30 μM reduced astrocyte viability in a concentration, and been shown that astroglial cells express catecholamine trans-
time-dependent manner. The resistance of astrocytes to lower porters (Huertas et al. 2015) which can promote cytosol accu-
doses of the toxin can be ascribed to their lower sensitivity to mulation of 6-OHDA and H2O2 generation after deamination
6-OHDA toxicity when compared to neurons (Blum et al. of the toxin by mitochondrial monoamine oxidase.
2000; Blum et al. 2001; Kaddour et al. 2013). This can be Accordingly, intracellular accumulation of ROS i.e. H2O2
explained by the higher antioxidant capacity of astrocytes and O2°- was strongly increased in 6-OHDA-treated astro-
(Baxter and Hardingham 2016; Ferrero-Gutierrez et al. cytes, and administration of ODN in the culture medium at-
2008; Mullett et al. 2013). tenuated in a dose-dependent manner the pro-oxidant action of
The fact that 6-OHDA up to 120 μM did not provoke 6-OHDA. It is widely accepted that overproduction of ROS
cell death within the first 24 h supports the idea that at can cause cell death by multiple mechanisms including dam-
least part of its astrocyte toxicity requires progressive ex- age of mitochondria, leading to activation of mitochondria-
tracellular auto-oxidation of 6-OHDA in the medium and dependent apoptotic pathways (Bahdoudi et al. 2018;
generation of free radicals (Hanrott et al. 2006). Goswami et al. 2016; Masmoudi-Kouki et al. 2011). The pres-
Incubation of cultured astrocytes with subnanomolar con- ent study reveals that ODN blocked the increase of the respi-
centrations of ODN dose-dependently protected cells ration rate and mitochondrial O2°− generation induced by 6-
against 6-OHDA-induced injury, indicating that ODN is OHDA. As a consequence, ODN prevented the effect of 6-
a potent glioprotective agent. While the beneficial effect OHDA on the collapse of the mitochondrial potential and on
of ODN against astrocyte cell death has already been re- the stimulation of caspase-3 activity. These data are consistent
ported (Ghouili et al. 2018; Hamdi et al. 2011; Hamdi with previous studies showing that, in cultured astrocytes and
et al. 2012; Hamdi et al. 2015), this is the first demon- cerebellar neurons, ODN stimulates the expression of the anti-
stration that ODN exerts a glioprotective action against 6- apoptotic gene Bcl-2 and simultaneously blocks 6-OHDA or
OHDA-induced toxicity. Dose-response experiments H2O2-induced expression of the pro-apoptotic gene Bax to
showed that ODN was effective at low concentrations promote cell survival (Hamdi et al. 2012; Kaddour et al.
(in the nanomolar range) while, at higher doses, the pro- 2013). The fact that, in situ, 6-OHDA provokes the expression
tective action of the gliopeptide gradually declined. Such of the active form of caspase-3 in astroglial cells in the SNpc
a bell-shaped concentration-response curve has already (Hernandez-Baltazar et al. 2013), together with the observa-
been observed with ODN on cell proliferation (Gandolfo tion that 6-OHDA-induced upregulation of the Bax to Bcl-2
et al. 1999), neuron protection (Kaddour et al. 2013) and mRNA ratio, provide evidence for the implication of the in-
intracellular calcium mobilization in cultured astrocytes trinsic apoptotic pathway in the gliotoxic action of 6-OHDA.
(Leprince et al. 2001). ODN acts through either a ROS are another key player involved in the oxidative in-
central-type benzodiazepine receptor (CBR) associated sults produced by 6-OHDA, which can impede neuronal sur-
with the GABAA receptor complex (Ferrero et al. 1986) vival (Tong et al. 2018; Tuncel et al. 2012). For instance in
or a metabotropic G protein-coupled receptor positively both the nigrostriatal region and cultured neuronal cells, 6-
coupled to phospholipase C (Patte et al. 1995) and OHDA treatment is associated with alteration of the redox
adenylyl cyclase (Hamdi et al. 2012). It has been reported status with notably glutathione (GSH) levels depletion, in-
that the protective action of ODN against oxidative stress creased protein and lipid oxidation and decreased expression
on astrocytes and cerebellar granules neurons is blocked and activities of antioxidant enzymes (Elkon et al. 2004;
by a selective ODN metabotropic receptor antagonist but Kaddour et al. 2013; Kuruvilla et al. 2013; Tong et al. 2018;
is not affected by a CBR antagonist (Hamdi et al. 2012; Tuncel et al. 2012), In agreement with these data, the present
Hamdi et al. 2015; Kaddour et al. 2013). Consistent with study reveals that ODN totally suppressed the inhibitory effect
this observation, the concentrations of ODN needed to of 6-OHDA on SOD and catalase activities. Furthermore,
prevent the deleterious effects of 6-OHDA on astroglial quantitative PCR analysis indicated that ODN enhanced genic
cells were similar to those which activate the metabotro- expression of endogenous antioxidant enzymes SOD2, cata-
pic receptor (Leprince et al. 2001). Altogether, these data lase, Gpx-1 and Srnx-1 and prevented the 6-OHDA-induced
J Mol Neurosci

decrease in expression of these genes involved in the control in close interaction in the brain, future studies should focus on
of the oxidative stress. The fact that knocking down of DBI, animal models and/or in vitro culture models where the effect
the ODN precursor, results in a decrease in antioxidant en- of ODN can be investigated in an integrated manner on both
zyme expression with an exacerbation of oxidative assaults cell types.
in cultured astrocyte (Ghouili et al. 2018) as well as increased In conclusion, the present study has demonstrated that ex-
sensitivity to MPTP neurotoxicity in PD mouse model posure of cultured astrocytes to 6-OHDA reduces in a dose-
(Bahdoudi et al. 2018), suggests that the gliopeptide ODN and time-dependent manner cell viability, induces a strong
could blunt intracellular ROS accumulation and dampen the increase of ROS production and inhibits both SOD and cata-
oxidative processes through an upregulation of ROS- lase activities. ODN exerts its protective effect upon the dele-
detoxifying enzymes. Overexpression of the transcription fac- terious action of oxidative stress by stimulation of expression
tor NF-E2-related factor (Nrf2), which binds to antioxidant of endogenous antioxidant enzymes (Mn-SOD, CAT, Gpx1
response element (ARE) (Jakel et al. 2007; Johnson et al. and Srnx1) that prevent ROS accumulation, preserve mito-
2008) to induce antioxidant enzyme expression, has been chondrial function and inhibit caspase-3 activation.
found to be protective against oxidative damage and to con- Collectively, these data suggest that astroglia-secreted ODN
tribute to protection of dopaminergic neurons from 6-OHDA- acts as an autocrine and/or paracrine protective agent that may
induced toxicity in vivo and in vitro (Lev et al. 2013; Masaki have therapeutic potential for the treatment of cerebral injuries
et al. 2017). Therefore, in vitro studies have shown that silenc- involving oxidative neurodegeneration.
ing Srxn1 results in decrease of NrF-2/ARE activity and in-
tracellular SOD and glutathione content (Zhou et al. 2015a) as Acknowledgments This work was supported by the France-Tunisia
CMCU-Campus France/PHC Utique 16G0820/34940PK exchange pro-
well as increased sensitivity to ischemia- and H2O2-induced
gram (to Olfa Masmoudi-Kouki and David Vaudry), Alternance scholar-
oxidative damage in astrocytes (Yu et al. 2015; Zhou et al. ship of Tunisian Higher Education Ministry and the Laboratory of
2015a; Zhou et al. 2015b). Altogether, these observations in- Neurophysiology, Cellular Physiopathology and Biomolecules
dicate that upregulation of Srxn1, which might be downstream Valorisation. LR18ES03, INSERM (U1239), the European Regional
Development Fund (PACT-CBS and PHEDERCPG projects), and the
of Nrf2, may be involved in the stimulatory action of ODN on
Region Normandy. Europe gets involved in Normandy with European
antioxidant enzyme gene transcription as a way of defense Regional Development Fund (ERDF). The funders had no role in study
from oxidative assaults. design, data collection and analysis, decision to publish, or preparation of
The importance of astroglial cells in the protection of neu- the manuscript.
rons against oxidative stress in diverse neuropathological con-
ditions such as stroke and Parkinson’s disease is well
established. Several studies have shown that astroglial-
derived peptides, i.e. activity-dependent neurotrophic factor, References
activity-dependent neuroprotective protein and endozepine
Afshin-Majd S, Bashiri K, Kiasalari Z, Baluchnejadmojarad T, Sedaghat
ODN, provide neuroprotection against various neurotoxins R, Roghani M (2017) Acetyl-l-carnitine protects dopaminergic
including 6-OHDA (Dejda et al. 2005; Kaddour et al. 2013; nigrostriatal pathway in 6-hydroxydopamine-induced model of
Masmoudi-Kouki et al. 2007), MPTP (Bahdoudi et al. 2018) Parkinson's disease in the rat. Biomed Pharmacother 89:1–9.
and alcohol (Sari and Gozes 2006). Besides its neuroprotec- https://doi.org/10.1016/j.biopha.2017.02.007
Alfonso J, Le Magueresse C, Zuccotti A, Khodosevich K, Monyer H
tive action, ODN is able to stimulate neurogenesis in the adult
(2012) Diazepam binding inhibitor promotes progenitor prolifera-
mouse brain, and silencing the ODN precursor, DBI, leads to tion in the postnatal SVZ by reducing GABA signaling. Cell Stem
growth arrest and neuro-progenitor cell death (Alfonso et al. Cell 10:76–87. https://doi.org/10.1016/j.stem.2011.11.011
2012; Dumitru et al. 2017), suggesting that ODN plays a Amri F, Ghouili I, Amri M, Carrier A, Masmoudi-Kouki O (2017)
major role during brain development and on neuron-glia in- Neuroglobin protects astroglial cells from hydrogen peroxide-
induced oxidative stress and apoptotic cell death. J Neurochem
teraction. In support of this notion, it has been shown that, in 140:151–169. https://doi.org/10.1111/jnc.13876
the cerebellar cortex, the ODN precursor is exclusively Bahdoudi S, Ghouili I, Hmiden M, do Rego JL, Lefranc B, Leprince J,
expressed by Bergmann glia (Tonon et al. 1990), and alter- Chuquet J, do Rego JC, Marcher AB, Mandrup S, Vaudry H, Tonon
ation of these cells by 6-OHDA treatment reduces DBI ex- MC, Amri M, Masmoudi-Kouki O, Vaudry D (2018)
Neuroprotective effects of the gliopeptide ODN in an in vivo model
pression and disturbs maturation and migration of granule
of Parkinson’s disease. Cell Mol Life Sci 75:2075–2091. https://doi.
neuron precursors (Podkletnova et al. 2001). Moreover, org/10.1007/s00018-017-2727-2
DBI−/− mice are more sensitive to MPTP-induced inflamma- Baxter PS, Hardingham GE (2016) Adaptive regulation of the brain’s
tion, ROS accumulation and oxidative brain injuries antioxidant defences by neurons and astrocytes. Free Radic Biol
(Bahdoudi et al. 2018). Although the mechanisms are not Med 100:147–152. https://doi.org/10.1016/j.freeradbiomed.2016.
06.027
established, these data suggest that, in vivo, ODN acts as an Blesa J, Przedborski S (2014) Parkinson’s disease: animal models and
autocrine and/or paracrine factor important for the protection dopaminergic cell vulnerability. Front Neuroanat 8:155. https://doi.
of both astrocytes and neurons. As astrocytes and neurons are org/10.3389/fnana.2014.00155
J Mol Neurosci

Blum D, Torch S, Nissou MF, Benabid AL, Verna JM (2000) protection against oxidative stress: astrocyte-neuron crosstalk rele-
Extracellular toxicity of 6-hydroxydopamine on PC12 cells. vance for neuronal survival. Mol Neurobiol 55:4596–4611. https://
Neurosci Lett 283:193–196 doi.org/10.1007/s12035-017-0630-3
Blum D, Torch S, Nissou MF, Verna JM (2001) 6-hydroxydopamine- Gomide VC, Silveira GA, Chadi G (2005) Transient and widespread
induced nuclear factor-kappa B activation in PC12 cells. Biochem astroglial activation in the brain after a striatal 6-OHDA-induced
Pharmacol 62:473–481 partial lesion of the nigrostriatal system. Int J Neurosci 115:99–117
Burda JE, Bernstein AM, Sofroniew MV (2016) Astrocyte roles in trau- Goswami P, Gupta S, Biswas J, Sharma S, Singh S (2016) Endoplasmic
matic brain injury. Exp Neurol 275(Pt 3):305–315. https://doi.org/ reticulum stress instigates the rotenone induced oxidative apoptotic
10.1016/j.expneurol.2015.03.020 neuronal death: a study in rat brain. Mol Neurobiol 53:5384–5400.
Chen LW, Yung KL, Chan YS (2005) Reactive astrocytes as potential https://doi.org/10.1007/s12035-015-9463-0
manipulation targets in novel cell replacement therapy of Gu XL, Long CX, Sun L, Xie C, Lin X, Cai H (2010) Astrocytic expres-
Parkinson’s disease. Curr Drug Targets 6:821–833 sion of Parkinson's disease-related A53T alpha-synuclein causes
Dallerac G, Rouach N (2016) Astrocytes as new targets to improve cog- neurodegeneration in mice. Mol Brain 3:12. https://doi.org/10.
nitive functions. Prog Neurobiol 144:48–67. https://doi.org/10. 1186/1756-6606-3-12
1016/j.pneurobio.2016.01.003 Guidotti A, Forchetti CM, Corda MG, Konkel D, Bennett CD, Costa E
Dejda A, Sokolowska P, Nowak JZ (2005) Neuroprotective potential of (1983) Isolation, characterization, and purification to homogeneity
three neuropeptides PACAP, VIP and PHI. Pharmacol Rep 57:307– of an endogenous polypeptide with agonistic action on benzodiaze-
320 pine receptors. Proc Natl Acad Sci U S A 80:3531–3535
Douiri S, Bahdoudi S, Hamdi Y, Cubi R, Basille M, Fournier A, Vaudry Hamdi Y, Kaddour H, Vaudry D, Bahdoudi S, Douiri S, Leprince J,
H, Tonon MC, Amri M, Vaudry D, Masmoudi-Kouki O (2016) Castel H, Vaudry H, Tonon MC, Amri M, Masmoudi-Kouki O
Involvement of endogenous antioxidant systems in the protective (2012) The octadecaneuropeptide ODN protects astrocytes against
activity of pituitary adenylate cyclase-activating polypeptide against hydrogen peroxide-induced apoptosis via a PKA/MAPK-dependent
hydrogen peroxide-induced oxidative damages in cultured rat astro- mechanism. PLoS One 7:e42498. https://doi.org/10.1371/journal.
cytes. J Neurochem 137:913–930. https://doi.org/10.1111/jnc.13614 pone.0042498
Dumitru I, Neitz A, Alfonso J, Monyer H (2017) Diazepam binding
Hamdi Y, Kaddour H, Vaudry D, Leprince J, Zarrouk A, Hammami M,
inhibitor promotes stem cell expansion controlling environment- Vaudry H, Tonon MC, Amri M, Masmoudi-Kouki O (2015)
dependent neurogenesis. Neuron 94(125–137):e125. https://doi. Octadecaneuropeptide ODN prevents hydrogen peroxide-induced
org/10.1016/j.neuron.2017.03.003
oxidative damage of biomolecules in cultured rat astrocytes.
Dzyubenko E, Gottschling C, Faissner A (2016) Neuron-glia interactions Peptides 71:56–65. https://doi.org/10.1016/j.peptides.2015.06.010
in neural plasticity: contributions of neural extracellular matrix and
Hamdi Y, Masmoudi-Kouki O, Kaddour H, Belhadj F, Gandolfo P,
perineuronal nets. Neural Plast 2016:5214961. https://doi.org/10.
Vaudry D, Mokni M, Leprince J, Hachem R, Vaudry H, Tonon
1155/2016/5214961
MC, Amri M (2011) Protective effect of the octadecaneuropeptide
Elkon H, Melamed E, Offen D (2004) Oxidative stress, induced by 6-
on hydrogen peroxide-induced oxidative stress and cell death in
hydroxydopamine, reduces proteasome activities in PC12 cells: im-
cultured rat astrocytes. J Neurochem 118:416–428. https://doi.org/
plications for the pathogenesis of Parkinson's disease. J Mol
10.1111/j.1471-4159.2011.07315.x
Neurosci 24:387–400. https://doi.org/10.1385/JMN:24:3:387
Han X, Zhu S, Wang B, Chen L, Li R, Yao W, Qu Z (2014) Antioxidant
Espinosa-Oliva AM, de Pablos RM, Sarmiento M, Villaran RF, Carrillo-
action of 7,8-dihydroxyflavone protects PC12 cells against 6-
Jimenez A, Santiago M, Venero JL, Herrera AJ, Cano J, Machado A
hydroxydopamine-induced cytotoxicity. Neurochem Int 64:18–23.
(2014) Role of dopamine in the recruitment of immune cells to the
https://doi.org/10.1016/j.neuint.2013.10.018
nigro-striatal dopaminergic structures. Neurotoxicology 41:89–101.
https://doi.org/10.1016/j.neuro.2014.01.006 Hanrott K, Gudmunsen L, O'Neill MJ, Wonnacott S (2006) 6-
Ferrero-Gutierrez A, Perez-Gomez A, Novelli A, Fernandez-Sanchez MT hydroxydopamine-induced apoptosis is mediated via extracellular
(2008) Inhibition of protein phosphatases impairs the ability of as- auto-oxidation and caspase 3-dependent activation of protein kinase
trocytes to detoxify hydrogen peroxide. Free Radic Biol Med 44: Cdelta. J Biol Chem 281:5373–5382. https://doi.org/10.1074/jbc.
1806–1816. https://doi.org/10.1016/j.freeradbiomed.2008.01.029 M511560200
Ferrero P, Santi MR, Conti-Tronconi B, Costa E, Guidotti A (1986) Study Hernandez-Baltazar D, Mendoza-Garrido ME, Martinez-Fong D (2013)
of an octadecaneuropeptide derived from diazepam binding inhibi- Activation of GSK-3beta and caspase-3 occurs in nigral dopamine
tor (DBI): biological activity and presence in rat brain. Proc Natl neurons during the development of apoptosis activated by a striatal
Acad Sci U S A 83:827–831 injection of 6-hydroxydopamine. PLoS One 8:e70951. https://doi.
Gandolfo P, Patte C, Thoumas JL, Leprince J, Vaudry H, Tonon MC org/10.1371/journal.pone.0070951
(1999) The endozepine ODN stimulates [3H]thymidine incorpora- Huertas A, Wessinger WD, Kucheryavykh YV, Sanabria P, Eaton MJ,
tion in cultured rat astrocytes. Neuropharmacology 38:725–732 Skatchkov SN, Rojas LV, Maldonado-Martinez G, Inyushin MY
Gardaneh M, Gholami M, Maghsoudi N (2011) Synergy between gluta- (2015) Quinine enhances the behavioral stimulant effect of cocaine
thione peroxidase-1 and astrocytic growth factors suppresses free in mice. Pharmacol Biochem Behav 129:26–33. https://doi.org/10.
radical generation and protects dopaminergic neurons against 6- 1016/j.pbb.2014.11.021
hydroxydopamine. Rejuvenation Res 14:195–204. https://doi.org/ Ishii T, Takanashi Y, Sugita K, Miyazawa M, Yanagihara R, Yasuda K,
10.1089/rej.2010.1080 Onouchi H, Kawabe N, Nakata M, Yamamoto Y, Hartman PS, Ishii
Garzon D, Cabezas R, Vega N, Avila-Rodriguez M, Gonzalez J, Gomez N (2017) Endogenous reactive oxygen species cause astrocyte de-
RM, Echeverria V, Aliev G, Barreto GE (2016) Novel approaches in fects and neuronal dysfunctions in the hippocampus: a new model
astrocyte protection: from experimental methods to computational for aging brain. Aging Cell 16:39–51. https://doi.org/10.1111/acel.
approaches. J Mol Neurosci 58:483–492. https://doi.org/10.1007/ 12523
s12031-016-0719-6 Jakel RJ, Townsend JA, Kraft AD, Johnson JA (2007) Nrf2-mediated
Ghouili I, Bahdoudi S, Morin F, Amri F, Hamdi Y, Coly PM, Walet- protection against 6-hydroxydopamine. Brain Res 1144:192–201.
Balieu ML, Leprince J, Zekri S, Vaudry H, Vaudry D, Castel H, https://doi.org/10.1016/j.brainres.2007.01.131
Amri M, Tonon MC, Masmoudi-Kouki O (2018) Endogenous ex- Johnson JA, Johnson DA, Kraft AD, Calkins MJ, Jakel RJ, Vargas MR,
pression of ODN-related peptides in astrocytes contributes to cell Chen PC (2008) The Nrf2-ARE pathway: an indicator and
J Mol Neurosci

modulator of oxidative stress in neurodegeneration. Ann N Y Acad MC, Amri M (2011) Pituitary adenylate cyclase-activating polypep-
Sci 1147:61–69. https://doi.org/10.1196/annals.1427.036 tide protects astroglial cells against oxidative stress-induced apopto-
Jordan J, Galindo MF, Tornero D, Gonzalez-Garcia C, Cena V (2004) sis. J Neurochem 117:403–411. https://doi.org/10.1111/j.1471-
Bcl-x L blocks mitochondrial multiple conductance channel activa- 4159.2011.07185.x
tion and inhibits 6-OHDA-induced death in SH-SY5Y cells. J Masmoudi-Kouki O, Gandolfo P, Castel H, Leprince J, Fournier A, Dejda
Neurochem 89:124–133. https://doi.org/10.1046/j.1471-4159. A, Vaudry H, Tonon MC (2007) Role of PACAP and VIP in
2003.02299.x astroglial functions. Peptides 28:1753–1760. https://doi.org/10.
Kaddour H, Hamdi Y, Vaudry D, Basille M, Desrues L, Leprince J, Castel 1016/j.peptides.2007.05.015
H, Vaudry H, Tonon MC, Amri M, Masmoudi-Kouki O (2013) The Michel PP, Toulorge D, Guerreiro S, Hirsch EC (2013) Specific needs of
octadecaneuropeptide ODN prevents 6-hydroxydopamine-induced dopamine neurons for stimulation in order to survive: implication
apoptosis of cerebellar granule neurons through a PKC-MAPK- for Parkinson disease. FASEB J 27:3414–3423. https://doi.org/10.
dependent pathway. J Neurochem 125:620–633. https://doi.org/10. 1096/fj.12-220418
1111/jnc.12140 Mirza B, Hadberg H, Thomsen P, Moos T (2000) The absence of reactive
Khan MM, Ahmad A, Ishrat T, Khan MB, Hoda MN, Khuwaja G, Raza astrocytosis is indicative of a unique inflammatory process in
SS, Khan A, Javed H, Vaibhav K, Islam F (2010) Resveratrol atten- Parkinson’s disease. Neuroscience 95:425–432
uates 6-hydroxydopamine-induced oxidative damage and dopamine Mladenovic A, Perovic M, Raicevic N, Kanazir S, Rakic L, Ruzdijic S
depletion in rat model of Parkinson’s disease. Brain Res 1328:139– (2004) 6-Hydroxydopamine increases the level of TNFalpha and
151. https://doi.org/10.1016/j.brainres.2010.02.031 bax mRNA in the striatum and induces apoptosis of dopaminergic
Kiasalari Z, Khalili M, Baluchnejadmojarad T, Roghani M (2016) neurons in hemiparkinsonian rats. Brain Res 996:237–245
Protective effect of oral hesperetin againstunilateral striatal 6- Mullett SJ, Di Maio R, Greenamyre JT, Hinkle DA (2013) DJ-1 expres-
hydroxydopamine damage in the rat. Neurochem Res 41:1065– sion modulates astrocyte-mediated protection against neuronal oxi-
1072. https://doi.org/10.1007/s11064-015-1796-6 dative stress. J Mol Neurosci 49:507–511. https://doi.org/10.1007/
Kuruvilla KP, Nandhu MS, Paul J, Paulose CS (2013) Oxidative stress s12031-012-9904-4
mediated neuronal damage in the corpus striatum of 6- Murakami S, Miyazaki I, Asanuma M (2018) Neuroprotective effect of
hydroxydopamine lesioned Parkinson’s rats: neuroprotection by se- fermented papaya preparation by activation of Nrf2 pathway in as-
rotonin, GABA and bone marrow cells supplementation. J Neurol trocytes. Nutr Neurosci 21:176–184. https://doi.org/10.1080/
Sci 331:31–37. https://doi.org/10.1016/j.jns.2013.04.020 1028415X.2016.1253171
L' Episcopo F, Tirolo C, Testa N, Caniglia S, Morale MC, Marchetti B
Osborn LM, Kamphuis W, Wadman WJ, Hol EM (2016) Astrogliosis: an
(2010) Glia as a turning point in the therapeutic strategy of
integral player in the pathogenesis of Alzheimer's disease. Prog
Parkinson’s disease. CNS Neurol Disord Drug Targets 9:349–372
Neurobiol 144:121–141. https://doi.org/10.1016/j.pneurobio.2016.
L'Episcopo F, Tirolo C, Testa N, Caniglia S, Morale MC, Cossetti C,
01.001
D'Adamo P, Zardini E, Andreoni L, Ihekwaba AE, Serra PA,
Patte C, Vaudry H, Desrues L, Gandolfo P, Strijdveen I, Lamacz M,
Franciotta D, Martino G, Pluchino S, Marchetti B (2011) Reactive
To n o n M C ( 1 9 9 5 ) T h e e n d o z e p i n e O D N s t i m u l a t e s
astrocytes and Wnt/beta-catenin signaling link nigrostriatal injury to
polyphosphoinositide metabolism in rat astrocytes. FEBS Lett
repair in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of
362:106–110
Parkinson’s disease. Neurobiol Dis 41:508–527. https://doi.org/10.
1016/j.nbd.2010.10.023 Pekny M, Pekna M, Messing A, Steinhauser C, Lee JM, Parpura V, Hol
Leprince J, Oulyadi H, Vaudry D, Masmoudi O, Gandolfo P, Patte C, EM, Sofroniew MV, Verkhratsky A (2016) Astrocytes: a central
Costentin J, Fauchere JL, Davoust D, Vaudry H, Tonon MC (2001) element in neurological diseases. Acta Neuropathol 131:323–345.
Synthesis, conformational analysis and biological activity of cyclic https://doi.org/10.1007/s00401-015-1513-1
analogs of the octadecaneuropeptide ODN: design of a potent Perfeito R, Cunha-Oliveira T, Rego AC (2013) Reprint of: revisiting
endozepine antagonist. Eur J Biochem 268:6045–6057 oxidative stress and mitochondrial dysfunction in the pathogenesis
Lev N, Barhum Y, Ben-Zur T, Melamed E, Steiner I, Offen D (2013) of Parkinson disease-resemblance to the effect of amphetamine
Knocking out DJ-1 attenuates astrocytes neuroprotection against 6- drugs of abuse. Free Radic Biol Med 62:186–201. https://doi.org/
hydroxydopamine toxicity. J Mol Neurosci 50:542–550. https://doi. 10.1016/j.freeradbiomed.2013.05.042
org/10.1007/s12031-013-9984-9 Podkletnova I, Rothstein JD, Helen P, Alho H (2001) Microglial response
Li Q, Yu S, Wu J, Zou Y, Zhao Y (2013) Sulfiredoxin-1 protects PC12 to the neurotoxicity of 6-hydroxydopamine in neonatal rat cerebel-
cells against oxidative stress induced by hydrogen peroxide. J lum. Int J Dev Neurosci 19:47–52
Neurosci Res 91:861–870. https://doi.org/10.1002/jnr.23218 Raicevic N, Mladenovic A, Perovic M, Harhaji L, Miljkovic D, Trajkovic
Lin CH, Wang CH, Hsu SL, Liao LY, Lin TA, Hsueh CM (2016) V (2005a) Iron protects astrocytes from 6-hydroxydopamine toxic-
Molecular mechanisms responsible for neuron-derived conditioned ity. Neuropharmacology 48:720–731. https://doi.org/10.1016/j.
medium (NCM)-mediated protection of ischemic brain. PLoS One neuropharm.2004.12.003
11:e0146692. https://doi.org/10.1371/journal.pone.0146692 Raicevic N, Mladenovic A, Perovic M, Miljkovic D, Trajkovic V (2005b)
Liu L, Liu L, Shi J, Tan M, Xiong J, Li X, Hu Q, Yi Z, Mao D (2016) The mechanisms of 6-hydroxydopamine-induced astrocyte death.
MicroRNA-34b mediates hippocampal astrocyte apoptosis in a rat Ann N Y Acad Sci 1048:400–405. https://doi.org/10.1196/annals.
model of recurrent seizures. BMC Neurosci 17:56. https://doi.org/ 1342.049
10.1186/s12868-016-0291-6 Sandhu JK, Gardaneh M, Iwasiow R, Lanthier P, Gangaraju S, Ribecco-
Liu Z, Chopp M (2016) Astrocytes, therapeutic targets for neuroprotec- Lutkiewicz M, Tremblay R, Kiuchi K, Sikorska M (2009)
tion and neurorestoration in ischemic stroke. Prog Neurobiol 144: Astrocyte-secreted GDNF and glutathione antioxidant system pro-
103–120. https://doi.org/10.1016/j.pneurobio.2015.09.008 tect neurons against 6OHDA cytotoxicity. Neurobiol Dis 33:405–
Masaki Y, Izumi Y, Matsumura A, Akaike A, Kume T (2017) Protective 414. https://doi.org/10.1016/j.nbd.2008.11.016
effect of Nrf2-ARE activator isolated from green perilla leaves on Sari Y, Gozes I (2006) Brain deficits associated with fetal alcohol expo-
dopaminergic neuronal loss in a Parkinson’s disease model. Eur J sure may be protected, in part, by peptides derived from activity-
Pharmacol 798:26–34. https://doi.org/10.1016/j.ejphar.2017.02.005 dependent neurotrophic factor and activity-dependent neuroprotec-
Masmoudi-Kouki O, Douiri S, Hamdi Y, Kaddour H, Bahdoudi S, tive protein. Brain Res Rev 52:107–118. https://doi.org/10.1016/j.
Vaudry D, Basille M, Leprince J, Fournier A, Vaudry H, Tonon brainresrev.2006.01.004
J Mol Neurosci

Soto-Otero R, Mendez-Alvarez E, Hermida-Ameijeiras A, Munoz-Patino peptide (VIP) against 6-hydroxy dopamine toxicity in the rat corpus
AM, Labandeira-Garcia JL (2000) Autoxidation and neurotoxicity striatum. J Mol Neurosci 46:51–57. https://doi.org/10.1007/s12031-
of 6-hydroxydopamine in the presence of some antioxidants: poten- 011-9618-z
tial implication in relation to the pathogenesis of Parkinson’s dis- Wachter B, Schurger S, Rolinger J, von Ameln-Mayerhofer A, Berg D,
ease. J Neurochem 74:1605–1612 Wagner HJ, Kueppers E (2010) Effect of 6-hydroxydopamine (6-
Tong H, Zhang X, Meng X, Lu L, Mai D, Qu S (2018) Simvastatin OHDA) on proliferation of glial cells in the rat cortex and striatum:
inhibits activation of NADPH oxidase/p38 MAPK pathway and evidence for de-differentiation of resident astrocytes. Cell Tissue
enhances expression of antioxidant protein in Parkinson disease Res 342:147–160. https://doi.org/10.1007/s00441-010-1061-x
models. Front Mol Neurosci 11:165. https://doi.org/10.3389/ Wu J, Chen Y, Yu S, Li L, Zhao X, Li Q, Zhao J, Zhao Y (2017)
fnmol.2018.00165 Neuroprotective effects of sulfiredoxin-1 during cerebral ischemia/
Tonon MC, Desy L, Nicolas P, Vaudry H, Pelletier G (1990) reperfusion oxidative stress injury in rats. Brain Res Bull 132:99–
Immunocytochemical localization of the endogenous benzodiaze- 108. https://doi.org/10.1016/j.brainresbull.2017.05.012
pine ligand octadecaneuropeptide (ODN) in the rat brain. Yang CM, Lin CC, Hsieh HL (2017) High-glucose-derived oxidative
Neuropeptides 15:17–24 stress-dependent heme oxygenase-1 expression from astrocytes con-
Tonon MC, Leprince J, Gandolfo P, Compère V, Pelletier G, Malagon tributes to the neuronal apoptosis. Mol Neurobiol 54:470–483.
MM, Vaudry H (2006) Endozepines.In handbook of biologically https://doi.org/10.1007/s12035-015-9666-4
active peptides,. ed A J Kastin (New York: Elsevier):813–819 Yu S, Wang X, Lei S, Chen X, Liu Y, Zhou Y, Zhou Y, Wu J, Zhao Y
Tonon MC, Leprince J, Morin F, Gandolfo P, Compère V, Pelletier G, (2015) Sulfiredoxin-1 protects primary cultured astrocytes from
Malagon M, Vaudry H (2013) Endozepines. In: Kastin AJ (ed) ischemia-induced damage. Neurochem Int 82:19–27. https://doi.
Handbook of biological active peptides, 2nd edn. Elsevier, Oxford, org/10.1016/j.neuint.2015.01.005
UK, pp 760–765
Zhou Y, Duan S, Zhou Y, Yu S, Wu J, Wu X, Zhao J, Zhao Y (2015a)
Toulorge D, Schapira AH, Hajj R (2016) Molecular changes in the post-
Sulfiredoxin-1 attenuates oxidative stress via Nrf2/ARE pathway
mortem parkinsonian brain. J Neurochem 139 Suppl 1:27–58.
and 2-Cys Prdxs after oxygen-glucose deprivation in astrocytes. J
https://doi.org/10.1111/jnc.13696
Mol Neurosci 55:941–950. https://doi.org/10.1007/s12031-014-
Tronel C, Rochefort GY, Arlicot N, Bodard S, Chalon S, Antier D (2013)
0449-6
Oxidative stress is related to the deleterious effects of heme
oxygenase-1 in an in vivo neuroinflammatory rat model. Oxidative Zhou Y, Zhou Y, Yu S, Wu J, Chen Y, Zhao Y (2015b) Sulfiredoxin-1
Med Cell Longev 2013:264935. https://doi.org/10.1155/2013/ exerts anti-apoptotic and neuroprotective effects against oxidative
264935 stress-induced injury in rat cortical astrocytes following exposure
Tuncel N, Korkmaz OT, Tekin N, Sener E, Akyuz F, Inal M (2012) to oxygen-glucose deprivation and hydrogen peroxide. Int J Mol
Antioxidant and anti-apoptotic activity of vasoactive intestinal Med 36:43–52. https://doi.org/10.3892/ijmm.2015.2205

You might also like