21.12.2013 Views

Intracerebroventricular administration of riluzole prevents morphine ...

Intracerebroventricular administration of riluzole prevents morphine ...

Intracerebroventricular administration of riluzole prevents morphine ...

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

Pharmacological Reports<br />

2010, 62, 664–673<br />

ISSN 1734-1140<br />

Copyright © 2010<br />

by Institute <strong>of</strong> Pharmacology<br />

Polish Academy <strong>of</strong> Sciences<br />

<strong>Intracerebroventricular</strong> <strong>administration</strong> <strong>of</strong> <strong>riluzole</strong><br />

<strong>prevents</strong> <strong>morphine</strong>-induced apoptosis in the<br />

lumbar region <strong>of</strong> the rat spinal cord<br />

Kambiz Hassanzadeh 1,4 , Bohlool Habibi-asl 2,4 , Leila Roshangar 5 ,<br />

Mahboob Nemati 2,6 , Masood Ansarin 4 , Safar Farajnia 3,4<br />

Department <strong>of</strong> Physiology and Pharmacology, Faculty <strong>of</strong> Medicine, Kurdistan University <strong>of</strong> Medical Sciences,<br />

Pasdaran Street, Sanandaj, Iran, Zip Code: 66177–13446<br />

Department <strong>of</strong> Pharmacology and Toxicology, ! Biotechnology Research Center, " Drug Applied Research Center,<br />

# $ Department <strong>of</strong> Anatomy and Histology, Faculty <strong>of</strong> Medicine, Department <strong>of</strong> Drug and Food Control, Faculty <strong>of</strong><br />

Pharmacy, Tabriz University <strong>of</strong> Medical Sciences, Tabriz, Iran<br />

Correspondence: Safar Farajnia, e-mail: farajnias@tbzmed.ac.ir<br />

Abstract:<br />

Opiates are the most effective drugs for pain relief. However, the repeated use <strong>of</strong> opiates induces tolerance to their analgesic effects.<br />

It has been shown that this <strong>morphine</strong>-induced tolerance is associated with apoptosis in the central nervous system. The aim <strong>of</strong> this<br />

study is to evaluate the effects <strong>of</strong> intracerebroventricular (icv) <strong>administration</strong> <strong>of</strong> <strong>riluzole</strong>, an anti-glutamatergic drug, on <strong>morphine</strong>induced<br />

apoptosis in the lumbar region <strong>of</strong> the rat spinal cord. Animals were given daily injections <strong>of</strong> <strong>morphine</strong> and vehicle, <strong>morphine</strong><br />

and <strong>riluzole</strong>, or <strong>riluzole</strong> alone. Nociception was assessed using a hot plate apparatus, and apoptosis was assessed using the in situ terminal<br />

deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling (TUNEL) method. The levels <strong>of</strong> anti-apoptotic factors<br />

Bcl-2 and HSP 70 and the pro-apoptotic agent caspase-3 were evaluated using immunoblotting. The glutamate concentration in the<br />

lumbar spinal cord was measured with high performance liquid chromatography (HPLC). The results indicate that the icv <strong>administration</strong><br />

<strong>of</strong> <strong>riluzole</strong> attenuated <strong>morphine</strong> tolerance and reduced the number <strong>of</strong> TUNEL positive cells. Immunoblotting revealed that<br />

the levels <strong>of</strong> the selected anti-apoptotic agents were greater in the treatment groups compared to the controls. Furthermore, the results<br />

demonstrated that the <strong>administration</strong> <strong>of</strong> <strong>riluzole</strong> can attenuate the <strong>morphine</strong>-induced elevation <strong>of</strong> glutamate in the lumbar spinal<br />

cord. In conclusion, icv <strong>administration</strong> <strong>of</strong> <strong>riluzole</strong> attenuated <strong>morphine</strong>-induced tolerance to analgesia and apoptosis in addition to<br />

preventing the <strong>morphine</strong>-induced increase <strong>of</strong> glutamate in the lumbar spinal cord <strong>of</strong> rats.<br />

Key words:<br />

apoptosis, lumbar spinal cord, <strong>morphine</strong>, <strong>riluzole</strong>, tolerance<br />

Introduction<br />

The neurobiological mechanisms underlying the development<br />

<strong>of</strong> opiate tolerance are multifaceted and<br />

only partially understood. Glutamate and N-methyl-<br />

D-aspartate receptors (NMDARs) play an important<br />

role in different forms <strong>of</strong> behavioral and neural plasticity,<br />

including tolerance, sensitization, and physical<br />

dependence, that arise from long-term treatment with<br />

different drugs <strong>of</strong> abuse [17, 22]. Numerous studies<br />

have demonstrated that the <strong>administration</strong> <strong>of</strong> a variety<br />

664 Pharmacological Reports, 2010, 62, 664–673


Riluzole <strong>prevents</strong> <strong>morphine</strong>-induced apoptosis in the rat spinal cord<br />

Kambiz Hassanzadeh et al.<br />

<strong>of</strong> NMDA receptor antagonists can inhibit the development<br />

<strong>of</strong> opiate tolerance and dependence [2, 12, 13,<br />

37–39]. It has also been shown that the activation <strong>of</strong><br />

NMDARs can lead to neurotoxicity under many circumstances<br />

[30, 32]. For instance, peripheral nerve<br />

injury has been shown to activate spinal cord<br />

NMDARs, which results in intractable neuropathic<br />

pain and neuronal cell death via apoptosis [23, 41]. In<br />

addition, it has been shown in vivo that neuronal<br />

apoptosis occurs in the dorsal horn <strong>of</strong> the rat spinal<br />

cord after chronic <strong>morphine</strong> treatment [24]. Mao et al.<br />

demonstrated that prolonged <strong>morphine</strong> <strong>administration</strong><br />

induced an upregulation <strong>of</strong> pro-apoptotic elements,<br />

such as Bax and caspase-3, and a downregulation <strong>of</strong><br />

the anti-apoptotic oncoprotein Bcl-2 in the dorsal<br />

horn <strong>of</strong> the spinal cord. Importantly, the upregulation<br />

<strong>of</strong> caspase-3 and Bax was inhibited when <strong>morphine</strong><br />

was co-administered with the noncompetitive NMDAR<br />

antagonist MK-801, thereby supporting a link between<br />

NMDAR activation and the intracellular changes<br />

<strong>of</strong> caspase-3 and Bax in response to a prolonged <strong>morphine</strong><br />

<strong>administration</strong> [24]. Interestingly, we recently<br />

demonstrated that both systemic and intracerebroventricular<br />

(icv) <strong>administration</strong> <strong>of</strong> <strong>riluzole</strong> – 2-amino-<br />

6-(trifluoromethoxy)benzothiazole, can attenuate the<br />

<strong>morphine</strong>-induced tolerance to analgesic effects [14,<br />

15]. Riluzole is an anti-glutamatergic agent that indirectly<br />

interferes with glutamate responses without interacting<br />

with any known binding site on NMDA, kainate,<br />

or AMPA glutamate receptors [8]. It has been<br />

shown in both in vivo and in vitro models that <strong>riluzole</strong><br />

confers neuroprotection during both spinal cord and<br />

cortical injury/ischemia [16, 21], a beneficial effect in<br />

various neurodegenerative diseases [1, 9]. In this study<br />

we investigate the effect <strong>of</strong> <strong>riluzole</strong> on <strong>morphine</strong>induced<br />

apoptosis in the lumbar section <strong>of</strong> the rat spinal<br />

cord after <strong>morphine</strong> tolerance.<br />

Guide for the Care and Use <strong>of</strong> Laboratory Animals<br />

(National Institutes <strong>of</strong> Health Publication No. 85–23,<br />

revised 1985) and were approved by the research and<br />

ethics committee <strong>of</strong> the Tabriz University <strong>of</strong> Medical<br />

Sciences.<br />

<strong>Intracerebroventricular</strong> cannula implantation<br />

Rats were anesthetized with an intraperitoneal (ip) injection<br />

<strong>of</strong> sodium pentobarbital (50 mg/kg) (Merck,<br />

Germany) and stereotaxically implanted with a 23-<br />

gauge stainless steel guide cannula into the lateral<br />

cerebral ventricle (–0.8 mm posterior, –1.3 mm midline<br />

to lateral and 3.5 mm ventral with respect to the<br />

bregma) [28]. A 30-gauge stainless steel dummy cannula<br />

was placed into the guide cannula to maintain the<br />

patency. After surgery, the animals were given a 7-day<br />

recovery period. During recovery, animals were habituated<br />

to the testing protocol, including being transferred<br />

to the experimental environment, handled,<br />

weighed, and restrained on the test platform for 1 min<br />

with a gentle removal and replacement <strong>of</strong> the dummy<br />

cannula twice daily. The animals were also habituated<br />

to the hot plate apparatus.<br />

Verification <strong>of</strong> cannula placement<br />

At the end <strong>of</strong> all behavioral experiments, methylene<br />

blue solution (5 µl, icv) was injected into the animals,<br />

which were then euthanized with pentobarbital and<br />

decapitated. The brain <strong>of</strong> each animal was removed<br />

and sliced along the coronal plane to verify placement<br />

<strong>of</strong> the guide cannula by distribution <strong>of</strong> the methylene<br />

blue in the ventricles. Only data from those animals<br />

that displayed a uniform distribution <strong>of</strong> methylene<br />

blue in the ventricles were considered for statistical<br />

analysis. In total, six animals were discarded due to<br />

incorrect placement <strong>of</strong> the guide cannula.<br />

Materials and Methods<br />

Animals<br />

Male Wistar rats weighing 250–300 g were used. The<br />

animals were housed in a temperature-controlled environment<br />

(24 ± 0.5°C) and kept on a 12 h light/dark<br />

cycle (light on 08:00) with free access to food and water.<br />

All experiments were in accordance with the<br />

Drug treatment<br />

Morphine sulfate (Darupakhsh, Iran) (10 mg/kg, ip)<br />

was dissolved in sterile 0.9% normal saline and injected<br />

using 1-ml insulin syringes. Riluzole (Sigma-<br />

Aldrich, Inc.) (20, 40, and 80 µg/10 µl) was dissolved<br />

with 1% Tween 80 in sterile 0.9% normal saline and<br />

infused (icv) with a Hamilton syringe. In addition,<br />

two groups <strong>of</strong> animals were given either vehicle or <strong>riluzole</strong><br />

(80 µg/10 µl) alone without <strong>morphine</strong>. The volume<br />

<strong>of</strong> infusion was 10 µl given at a rate <strong>of</strong> 10 µl/min.<br />

Pharmacological Reports, 2010, 62, 664–673 665


Induction <strong>of</strong> tolerance to the analgesic effect <strong>of</strong><br />

<strong>morphine</strong><br />

Nociception was assessed with a hot plate apparatus.<br />

Animals were placed on a hot plate (55 ± 0.5°C) [10]<br />

and the latency until the rat licked its hind paw was<br />

recorded. A cut<strong>of</strong>f time <strong>of</strong> 40 s was imposed to prevent<br />

tissue damage [29]. Hot plate response latencies<br />

(s) are expressed as a percentage <strong>of</strong> the maximal possible<br />

effect (%MPE) using the equation below:<br />

Post - drug latency – Baseline latency<br />

% MPE =<br />

× 100<br />

Cut<strong>of</strong>f value – Baseline latency<br />

The baseline latency (BL) was determined once per<br />

day for each rat prior to the daily injection <strong>of</strong> <strong>morphine</strong><br />

(10 mg/kg). Morphine was then injected, and<br />

20 min later the animals were given either <strong>riluzole</strong> or<br />

vehicle. The post-drug latency (LT) was measured after<br />

10 min, a total <strong>of</strong> 30 min after the original <strong>morphine</strong><br />

injection. The percent maximal possible effect<br />

(%MPE) was then calculated for that day. The experiments<br />

were repeated until there was no significant difference<br />

in %MPE between the vehicle- or drug-treated<br />

groups and the saline group.<br />

Tissue preparation<br />

For the in situ TUNEL assay, different groups <strong>of</strong> animals<br />

(n = 6) were given the same treatment regimen<br />

as the behavioral groups. On the ninth day (one day<br />

after tolerance was observed in the control group), the<br />

animals were euthanized with pentobarbital 2 h after<br />

the last dose <strong>of</strong> vehicle or <strong>riluzole</strong>, and their lumbar<br />

spinal cords were immediately enucleated and fixed<br />

in 10% (w/v) formaldehyde (Merck, Germany) with<br />

the pH adjusted to 7.0 with NaOH. The tissue was<br />

then embedded in paraffin. In order to evaluate the<br />

possible association between <strong>morphine</strong> tolerance and<br />

apoptosis, one group was given <strong>morphine</strong> and <strong>riluzole</strong><br />

at a dose <strong>of</strong> 80 µg/10 µl, which was the most effective<br />

dose for preventing <strong>morphine</strong> tolerance in behavioral<br />

studies. On the 14 th day, one day after the onset <strong>of</strong> behavioral<br />

tolerance in this group, the lumbar spinal<br />

cords <strong>of</strong> the rats were removed 2 h after the last dose<br />

<strong>of</strong> vehicle or treatment and prepared as described<br />

above.<br />

Detection <strong>of</strong> apoptotic cells<br />

After the tissue was fixed and embedded into the paraffin,<br />

3 µm sections were cut with a microtome (Leitz,<br />

Germany) and the TUNEL assay was carried out using<br />

the in situ Cell Death Detection kit (Roche Applied<br />

Science Cat # 11 684 817 910) according to the<br />

manufacturer’s instructions. Briefly, the paraffin was<br />

removed and the sections were rehydrated and pretreated<br />

with proteinase K (Roche, Germany) for<br />

30 min at 37°C. The sections were then exposed to the<br />

TUNEL reaction mixture containing terminal deoxynucleotidyl<br />

transferase and nucleotides, including<br />

fluorescein isothiocyanate-labeled dUTP (37°C). After<br />

1 h, the preparations were incubated for 30 min at<br />

37°C with a peroxidase conjugated anti-fluorescein<br />

antibody. Finally, the reaction product was visualized<br />

by incubation for 15 min at room temperature with<br />

3,3-diaminobenzidine tetrahydrochloride (DAB) substrate,<br />

followed by counterstaining with methylene<br />

blue. Apoptotic cells throughout the tissue section<br />

were intensely stained (brown) by the TUNEL treatment<br />

and analyzed with a Zeiss Axiovert 100 light<br />

microscope (100× objective) in 30 fields.<br />

Western blot analysis <strong>of</strong> Bcl-2, HSP 70 and<br />

caspase-3<br />

For western blotting, rats (n = 6) were rapidly (< 1 min)<br />

sacrificed, and lumbar spinal cord segments were removed,<br />

cleaned, and frozen in liquid nitrogen. Subsequently,<br />

tissue samples were homogenized in lysis<br />

buffer (500 mM Tris-HCl, pH 7.4, 150 mM NaCl,<br />

EDTA 0.5 mM, n-octyl--D-glucopyranoside 1.5% w/v)<br />

containing a complete Protease Inhibitor Cocktail<br />

(Roche Cat # 04 693 132 001). The protein quantification<br />

for each loading lane was estimated by an absorbance<br />

protein assay (280 nm). Lysates (20 µg protein)<br />

were resolved with 12% SDS-PAGE for Bcl2<br />

and HSP 70 and 15% SDS-PAGE for caspase-3. Next,<br />

the lysates were transferred to a polyvinylidene<br />

difluoride (PVDF) membrane (Millipore, Bedford,<br />

MA), blocked with 5% milk, and incubated overnight<br />

at 4°C with the following primary antibodies: caspase-3<br />

(18–20 kDa), Abcam, Cat # ab2302; Bcl-2 (26–29 kDa),<br />

Abcam, Cat # ab16904; HSP 70 (70 kDa), Abcam, Cat<br />

# ab6535; and -actin (42 kDa), Abcam, Cat # 8226.<br />

Subsequently, the PVDF membranes were incubated<br />

for 1 h at room temperature with an HRP-conjugated<br />

secondary antibody. The blots were then visualized<br />

666 Pharmacological Reports, 2010, 62, 664–673


Riluzole <strong>prevents</strong> <strong>morphine</strong>-induced apoptosis in the rat spinal cord<br />

Kambiz Hassanzadeh et al.<br />

with the enhanced chemiluminiscence (ECL) detection<br />

kit/system for 1 min and exposed to Hyperfilm<br />

(Roche) for 30 s to 5 min. Finally, the developed films<br />

were scanned, and the density <strong>of</strong> the immunoreactive<br />

bands was measured using Image J s<strong>of</strong>tware and normalized<br />

to the bands <strong>of</strong> the internal control (-actin<br />

was the loading control). Differences in the image<br />

density were compared by a one-way analysis <strong>of</strong> variance<br />

(ANOVA) (multiple groups) followed by Tukey’s<br />

test.<br />

High performance liquid chromatography<br />

(HPLC) analysis <strong>of</strong> neurotransmitter glutamate<br />

To quantify the amount <strong>of</strong> glutamate, HPLC was employed.<br />

Tissue samples from rats (n = 6) were homogenized<br />

and protein determination was performed.<br />

Subsequently, the homogenates were spun at 4°C for 10<br />

min at 10,000 × g. The chromatograph was a KNAUER<br />

(Berlin, Germany) HPLC instrument. The system included<br />

a quaternary pump, a RF-551 fluorescence detector<br />

(FLD), and an auto sampler (Spark, Triatlon),<br />

which was controlled by Chromgate s<strong>of</strong>tware. The<br />

analytical column used was a reversed-phase Hypersil<br />

ODS column (250 × 4.0 mm, 5 µm particle size). For<br />

the chromatographic separation, the mobile phases<br />

consisted <strong>of</strong> 8% acetonitrile in 12.5 mM phosphate<br />

buffer at pH 7.2, with an o-phthalaldehyde (OPA) derivative<br />

<strong>of</strong> glutamate eluted at a gradient flow rate <strong>of</strong><br />

1 ml/min for 8 min and 2 ml/min for 12 min. The detection<br />

was carried out at 330 nm and 460 nm as the<br />

excitation and emission wavelengths, respectively.<br />

Glutamic acid was used as the standard. Solutions <strong>of</strong><br />

0.75, 1.5, 3, 6 and 12 µg/ml glutamic acid were injected<br />

into the HPLC instrument and the resulting<br />

calibration curve was plotted. A 2.5% solution <strong>of</strong> supernatant<br />

in distilled water was used for the derivatization<br />

and determination <strong>of</strong> glutamate. The level <strong>of</strong><br />

glutamate was calculated by comparing peak areas<br />

with those <strong>of</strong> standards, and the values are expressed<br />

as µmol/100 mg protein [26].<br />

Data analysis<br />

Data obtained from the hot plate tests are expressed as<br />

the mean %MPE ± SEM. The histological data from<br />

the lumbar spinal cord sections were averaged and are<br />

expressed as the mean ± SEM. Data from the western<br />

blots are expressed as % <strong>of</strong> the control, and data from<br />

the HPLC are expressed as the mean ± SEM. The Student’s<br />

t-test was used to compare the means <strong>of</strong> two<br />

groups and a one-way analysis <strong>of</strong> variance (ANOVA)<br />

followed by Tukey’s test was used to compare the<br />

means <strong>of</strong> multiple treatment groups; p-values <strong>of</strong> less<br />

than 0.05 were considered to be significant.<br />

Fig. 1. Effect <strong>of</strong> daily icv injection <strong>of</strong> <strong>riluzole</strong> (20, 40 and 80 µg/10 µl) on the tolerance to the analgesic effect <strong>of</strong> <strong>morphine</strong>. Each bar represents<br />

the mean %MPE ± SEM for 8 rats. The Student’s t-test was used to compare the statistical differences between each treatment group and the<br />

saline group. A one-way ANOVA followed by Tukey’s test was used to analyze the statistical significance between treatments and the control<br />

group.Ap-value <strong>of</strong> less than 0.05 was considered significant for all analyses. *p< 0.05; ** p < 0.01; *** p < 0.001 when compared to the saline<br />

group. #p < 0.001 when compared to the control. The arrow represents the onset <strong>of</strong> <strong>morphine</strong> tolerance. M = <strong>morphine</strong>, Rilu = <strong>riluzole</strong><br />

Pharmacological Reports, 2010, 62, 664–673 667


Results<br />

Riluzole attenuated <strong>morphine</strong>-induced<br />

tolerance to the analgesic effect<br />

As shown in Figure 1, the daily <strong>administration</strong> <strong>of</strong> <strong>morphine</strong><br />

(10 mg/kg, ip) for 8 days produced tolerance to<br />

the antinociceptive effects in the control group that received<br />

<strong>morphine</strong> (10 mg/kg, ip) and 1% Tween 80 in<br />

0.9% saline (10 µl/rat, icv). Daily <strong>administration</strong> <strong>of</strong><br />

80 µg/10 µl <strong>riluzole</strong> significantly delayed this <strong>morphine</strong><br />

tolerance and shifted the onset <strong>of</strong> tolerance from<br />

day 8 in the control group to day 13 (p < 0.001), whereas<br />

lower concentrations <strong>of</strong> <strong>riluzole</strong> (20 and 40 µg/10 µl) did<br />

not delay the <strong>morphine</strong> tolerance significantly.<br />

TUNEL staining<br />

The TUNEL method was used to identify apoptotic<br />

cells. In the control group (<strong>morphine</strong>, ip and 1%<br />

Tween 80 in 0.9% normal saline, icv), the number <strong>of</strong><br />

TUNEL-positive cells in the lumbar spinal cord was<br />

significantly higher than vehicle-treated animals (Fig.<br />

2, p < 0.01), indicating an increased background level<br />

<strong>of</strong> apoptotic activity in the <strong>morphine</strong>-treated animals.<br />

The average number <strong>of</strong> TUNEL positive cells was<br />

significantly reduced in the lumbar spinal cord <strong>of</strong> the<br />

<strong>riluzole</strong>-treated groups compared to those <strong>of</strong> the control<br />

group. There was a significant difference in the<br />

number <strong>of</strong> TUNEL-positive cells between animals<br />

that received vehicle or <strong>riluzole</strong> without <strong>morphine</strong><br />

versus those that received <strong>morphine</strong> in the control<br />

group (p < 0.05). Animals treated with the highest<br />

dose <strong>of</strong> <strong>riluzole</strong> (80 µg/10 µl) had fewer TUNELpositive<br />

cells than those that received lower doses.<br />

Changes in the Bcl-2, HSP 70 and caspase-3<br />

content <strong>of</strong> lumbar spinal cord<br />

Western blot analysis showed that after the onset <strong>of</strong><br />

<strong>morphine</strong> tolerance, the anti-apoptotic factors, Bcl-2<br />

(p < 0.01) and HSP 70 (p < 0.001), decreased significantly,<br />

whereas the amount <strong>of</strong> pro-apoptotic protein,<br />

caspase-3, was increased (p < 0.05) (Figs. 3–5). There<br />

were no significant differences in the amount <strong>of</strong> Bcl-2<br />

and caspase-3 between control and after tolerance<br />

(AT) groups. Furthermore, the results indicate that the<br />

amount <strong>of</strong> HSP 70 in the AT group was greater than<br />

that <strong>of</strong> the controls. The <strong>administration</strong> <strong>of</strong> <strong>riluzole</strong><br />

(80 µg/10 µl, icv) with <strong>morphine</strong> (ip) for 9 days induced<br />

a significant up-regulation <strong>of</strong> Bcl-2 (p < 0.05),<br />

whereas the 40 and 80 µg/10 µl doses <strong>of</strong> <strong>riluzole</strong> significantly<br />

increased the amount <strong>of</strong> HSP 70 (p < 0.001).<br />

However, these doses did not affect the level <strong>of</strong><br />

caspase-3 in the lumbar spinal cord, as shown in the<br />

corresponding western blots <strong>of</strong> Figure 5.<br />

Effect <strong>of</strong> <strong>riluzole</strong> on glutamate concentration in<br />

the lumbar spinal cord <strong>of</strong> <strong>morphine</strong> treated rats<br />

As shown in Table 1, chronic <strong>administration</strong> <strong>of</strong> <strong>morphine</strong><br />

(10 mg/kg, ip) for 9 days induced a significant<br />

increase in the glutamate concentration in the lumbar<br />

spinal cord <strong>of</strong> <strong>morphine</strong> tolerant rats (p < 0.001). These<br />

findings show that the <strong>administration</strong> <strong>of</strong> <strong>riluzole</strong> (40<br />

and 80 µg/10 µl) after <strong>morphine</strong> attenuated the increase<br />

<strong>of</strong> the glutamate concentration in the related<br />

group (p < 0.01).<br />

Discussion<br />

The cellular mechanisms underlying the development<br />

<strong>of</strong> <strong>morphine</strong> tolerance remain controversial. Previous<br />

studies have indicated that certain addictive drugs,<br />

Tab. 1. Concentration <strong>of</strong> glutamate in the lumbar spinal cord <strong>of</strong> rats<br />

Treatment<br />

Glutamate concentration<br />

[µmol/100 mg protein]<br />

S + 1% Tween 80 138.50 ± 6.34***<br />

M + 1% Tween 80 216.71 ± 8.26<br />

M + Rilu (20 µg/10 µl/rat) 190.09 ± 8.71*<br />

M + Rilu (40 µg/10 µl/rat) 179.37 ± 5.11**<br />

M + Rilu (80 µg/10 µl/rat) 183.80 ± 5.27**<br />

Rilu (80 µg/10 µl/rat) 187.92 ± 5.45**<br />

Each datum represents the mean + SEM. One-way ANOVA followed<br />

by Tukey’s test was used to analyze the statistical significance.<br />

A p- value < 0.05 was considered significant; * p < 0.05, ** p < 0.01,<br />

*** p < 0.001 when compared to control (M + 1% Tween 80). S = saline,<br />

M = <strong>morphine</strong>, Rilu = <strong>riluzole</strong><br />

668 Pharmacological Reports, 2010, 62, 664–673


Riluzole <strong>prevents</strong> <strong>morphine</strong>-induced apoptosis in the rat spinal cord<br />

Kambiz Hassanzadeh et al.<br />

Fig. 2. A: Tissue sections from rat lumbar<br />

spinal cord were prepared and assayed<br />

with the In Situ Cell Death Detection<br />

Kit, POD. Slides were counterstained<br />

with methylene blue. Apoptotic<br />

cells scattered throughout the tissue<br />

section were intensely stained<br />

(brown) by the TUNEL treatment. Slides<br />

were analyzed with a light microscope<br />

(100´ objective). (a) S + 1% Tween 80,<br />

(b) <strong>morphine</strong> + 1% Tween 80, (c) <strong>morphine</strong><br />

+ <strong>riluzole</strong> (20 µg/10 µl), (d) <strong>morphine</strong><br />

+ <strong>riluzole</strong> (40 µg/10 µl), (e) <strong>morphine</strong><br />

+ <strong>riluzole</strong> (80 µg/10 µl), (f) riluole<br />

(80 µg/10 µl). B: Quantification <strong>of</strong> apoptotic<br />

cells. The data represent the mean<br />

± SEM number <strong>of</strong> apoptotic (TUNELpositive)<br />

cells in 30 fields, which were<br />

counted at a magnfiication <strong>of</strong> 100´ with<br />

a light microscope. A one-way ANOVA<br />

followed by Tukey’s test was used to<br />

analyze the statistical significances.<br />

A p-value <strong>of</strong> less than 0.05 was considered<br />

significant for all analyses. * p<br />

< 0.05; ** p < 0.01; *** p < 0.001 when<br />

compared to the control group (M + 1%<br />

Tween 80). S = saline, M = <strong>morphine</strong>,<br />

Rilu = <strong>riluzole</strong>, AT = after tolerance<br />

Average Number <strong>of</strong><br />

Apoptotic Cells<br />

***<br />

* * **<br />

***<br />

such as <strong>morphine</strong>, can induce apoptosis in cultured<br />

neuronal cell lines and human cells [33, 34]. It has<br />

also been demonstrated that in vivo neuronal apoptosis<br />

occurs in the rat spinal cord dorsal horn after<br />

chronic <strong>morphine</strong> treatment, which has been associated<br />

with the expression <strong>of</strong> activated caspase-3 and<br />

involves the activation <strong>of</strong> the mitogen-activated protein<br />

kinase (MAPK) pathway [24]. In this study we<br />

found that the chronic <strong>administration</strong> <strong>of</strong> <strong>morphine</strong> significantly<br />

increased the amount <strong>of</strong> apoptotic cells in<br />

the lumbar spinal cord. This finding is in accordance<br />

with previous reports, which indicated that prolonged<br />

exposure to <strong>morphine</strong> induces apoptotic cell death in<br />

the dorsal horn regions <strong>of</strong> the spinal cord. Since this<br />

region is critically involved in opiate-mediated analgesia,<br />

it is believed that the apoptosis in this area contributes,<br />

at least in part, to the behavioral manifestation<br />

<strong>of</strong> <strong>morphine</strong> tolerance [24]. We also demonstrated<br />

that the <strong>administration</strong> <strong>of</strong> <strong>riluzole</strong> (80 µg/10 µl) significantly<br />

decreased the development <strong>of</strong> this tolerance by<br />

shifting the onset <strong>of</strong> tolerance from the 8 th day in the<br />

control group to the 13 th day. These results are in line<br />

with our previous study, which indicated that there<br />

was a significant right-shift <strong>of</strong> the dose-response<br />

curve and the ED50 <strong>of</strong> <strong>morphine</strong> for animals that received<br />

<strong>morphine</strong> and <strong>riluzole</strong> (80 µg/10 µl) compared<br />

to those that received <strong>morphine</strong> and saline [14]. Furthermore,<br />

it has been demonstrated that both <strong>morphine</strong><br />

tolerance and the associated neuronal apoptosis<br />

share a common cellular mechanism. The NMDAR is<br />

thought to be important for apoptosis and tolerance<br />

because MK-801 or memantine, the NMDAR antagonists,<br />

were able to block both [18, 24]. Activation <strong>of</strong><br />

NMDARs, on the other hand, initiated intracellular<br />

pathways <strong>of</strong> apoptotic cell death. It has been suggested<br />

that multiple intracellular mechanisms may be<br />

involved in the NMDAR-mediated apoptotic changes.<br />

Our results show that <strong>riluzole</strong>, as an anti-glutamatergic<br />

agent, can prevent <strong>morphine</strong>-induced apoptosis and<br />

significantly attenuate the average number <strong>of</strong> TUNEL<br />

positive cells (p < 0.01). After the development <strong>of</strong> tolerance<br />

in the group that received <strong>morphine</strong> and<br />

80 µg/10 µl <strong>of</strong> <strong>riluzole</strong> an increase in the number <strong>of</strong><br />

apoptotic cells was observed, similar to the one seen<br />

Pharmacological Reports, 2010, 62, 664–673 669


Fig. 3. Effect <strong>of</strong> icv <strong>riluzole</strong> (20, 40 and 80 µg/10 µl)<br />

on <strong>morphine</strong>-induced changes in intracellular<br />

Bcl-2. A: Western blots illustrate down-regulation<br />

<strong>of</strong> the Bcl-2 protein (26kDa) in rats receiving<br />

<strong>morphine</strong> (M + 1% Tween 80) daily for 9 days<br />

compared to the corresponding S + 1% Tween<br />

80 group. Riluzole (80 µg/10 µl) increased the<br />

amount <strong>of</strong> Bcl-2 protein in combination with<br />

<strong>morphine</strong>. B: The statistical analysis showed<br />

differences among the different groups in the<br />

gray density obtained for the western blot<br />

bands <strong>of</strong> Bcl-2 in the lumbar spinal cord.<br />

A one-way ANOVA followed by Tukey’s test was<br />

used to analyze the statistical significances.<br />

A p-value <strong>of</strong> less than 0.05 was considered significant<br />

for all analyses. * p < 0.05, ** p < 0.01<br />

compared to the corresponding control group.<br />

S = saline, M = <strong>morphine</strong>, Rilu = <strong>riluzole</strong>, AT =<br />

after tolerance<br />

Fig. 4. Effect <strong>of</strong> icv <strong>riluzole</strong> (20, 40 and 80 µg/10 µl)<br />

on <strong>morphine</strong>-induced changes in intracellular<br />

HSP 70. A: Western blots illustrate the downregulation<br />

<strong>of</strong> the HSP 70 protein in rats receiving<br />

<strong>morphine</strong> (M + 1% Tween 80) daily for 9<br />

days compared to the corresponding S + 1%<br />

Tween 80 group. Riluzole (40, 80 µg/10 µl) increased<br />

the amount <strong>of</strong> HSP 70 protein. B: The<br />

statistical analysis showed differences among<br />

the different groups in the gray density obtained<br />

for the western blot bands <strong>of</strong> HSP 70 in<br />

the lumbar spinal cord. A one-way ANOVA followed<br />

by Tukey’s test was used to analyze the<br />

statistical significances. Ap-value <strong>of</strong> less than<br />

0.05 was considered as significant for all analyses.<br />

*** p < 0.001 compared to the corresponding<br />

control group. S = saline, M = <strong>morphine</strong>,<br />

Rilu = <strong>riluzole</strong>, AT = after tolerance<br />

Fig. 5. Effect <strong>of</strong> icv <strong>riluzole</strong> (20, 40 and 80 µg/10 µl)<br />

on <strong>morphine</strong>-induced changes in intracellular<br />

caspase-3. A: Western blots illustrate up-regulation<br />

<strong>of</strong> the caspase-3 protein (19 kDa) in rats<br />

receiving <strong>morphine</strong> (M + 1% Tween 80) daily for<br />

9 days compared to the corresponding S + 1%<br />

Tween 80 group. Riluzole did not change the<br />

amount <strong>of</strong> caspase-3 protein. B: The statistical<br />

analysis showed differences among the different<br />

groups in the gray density obtained for the<br />

western blot bands <strong>of</strong> caspase-3 in the lumbar<br />

spinal cord. A one-way ANOVA followed by<br />

Tukey’s test was used to analyze the statistical<br />

significances. Ap-value <strong>of</strong> less than 0.05 was<br />

considered as significant for all analyses. * p < 0.05<br />

compared to the corresponding control group.<br />

S = saline, M = <strong>morphine</strong>, Rilu = <strong>riluzole</strong>, AT =<br />

after tolerance<br />

670 Pharmacological Reports, 2010, 62, 664–673


Riluzole <strong>prevents</strong> <strong>morphine</strong>-induced apoptosis in the rat spinal cord<br />

Kambiz Hassanzadeh et al.<br />

in control conditions. This means that by the time behavioral<br />

tolerance was observed in the control and<br />

treatment groups, the apoptotic process had already<br />

developed. Other studies have demonstrated that<br />

chronic treatment with <strong>morphine</strong>, both toleranceeducing<br />

and dependence-educing, is associated with<br />

a differential modulation in two key brain proteins involved<br />

in the regulation <strong>of</strong> the programmed cell<br />

death. Specifically, there is an up-regulation <strong>of</strong> the<br />

pro-apoptotic Fas receptor and the intracellular proapoptotic<br />

elements, such as Bax and caspase-3, and<br />

there is a moderate down-regulation <strong>of</strong> the antiapoptotic<br />

oncoprotein Bcl-2 [5, 24]. The results <strong>of</strong><br />

this study are consistent with others that indicate<br />

a <strong>morphine</strong>-induced decrease in the content <strong>of</strong> Bcl-2.<br />

On the other hand, our results show that 80 µg/10 µl<br />

<strong>of</strong> <strong>riluzole</strong> increases the level <strong>of</strong> Bcl-2 in the lumbar<br />

spinal cord when administered with <strong>morphine</strong>. Previous<br />

studies have demonstrated that Hsp70 proteins<br />

participate in protein folding and transport, the refolding<br />

<strong>of</strong> denatured proteins, and protection from apoptosis<br />

[31]. Hsp70 rescues cells from the apoptotic/necrotic<br />

death that normally occurs after heat shock, exposure<br />

to tumor necrosis factor , oxidative stress,<br />

ceramide, anti-cancer drugs, radiation, or nitric oxide<br />

[4, 11, 25]. Hsp70 was reported to inhibit heat shockinduced<br />

apoptosis downstream <strong>of</strong> cytochrome C release<br />

but upstream <strong>of</strong> caspase-3 activation [20]. Our<br />

results, in agreement with those indicating that <strong>morphine</strong><br />

significantly decreased Hsp70 levels in rat neurons<br />

[7], show that the Hsp 70 content <strong>of</strong> lumbar spinal<br />

cord decreased after chronic <strong>morphine</strong> <strong>administration</strong><br />

in <strong>morphine</strong>-tolerant rats. Furthermore, our results<br />

demonstrate that the <strong>administration</strong> <strong>of</strong> <strong>riluzole</strong><br />

(40 and 80 µg/10 µl) significantly increased the<br />

amount <strong>of</strong> Hsp 70 (p < 0.001). This parallel increase<br />

in two anti-apoptotic factors, Bcl-2 and Hsp 70, could<br />

represent a part <strong>of</strong> the neuroprotective mechanisms <strong>of</strong><br />

this drug. Riluzole did not affect the <strong>morphine</strong>induced<br />

increase in the spinal levels <strong>of</strong> caspase 3.<br />

Nevertheless, this does not rule out the possibility that<br />

<strong>riluzole</strong> can effect caspase 3 activity.<br />

Riluzole is known as a neuroprotective agent. In<br />

cerebrocortical nerve terminals, <strong>riluzole</strong> has been reported<br />

to inhibit glutamate release by reducing the<br />

Ca 2+ influx through P/Q type Ca 2+ channels, and involves<br />

in G protein signaling mechanism sensitive to<br />

pertussis toxin. Importantly, the excessive release and<br />

accumulation <strong>of</strong> glutamate is associated with an increased<br />

level <strong>of</strong> intracellular calcium and plays an important<br />

role in CNS injury and neurodegenerative diseases<br />

[40].<br />

Our results demonstrate that <strong>morphine</strong> increases<br />

the level <strong>of</strong> glutamate in the lumbar spinal cord and<br />

that <strong>riluzole</strong> (40 and 80 µg/10 µl) significantly attenuates<br />

this <strong>morphine</strong>-induced increase (p < 0.01). Although<br />

the measurement <strong>of</strong> the glutamate content in<br />

tissue is not specific enough to indicate the glutamate<br />

induced excitotoxicity, these results are in accordance<br />

with others who reported that chronic <strong>morphine</strong> <strong>administration</strong><br />

can increase glutamate release in the<br />

CNS [3, 17, 22, 37–40] and that high concentrations<br />

<strong>of</strong> <strong>riluzole</strong> inhibit glutamate release. It has also been<br />

shown that <strong>riluzole</strong> can attenuate excitatory amino<br />

acid receptor activation and decrease the excitability<br />

<strong>of</strong> the postsynaptic cell membrane [6]. In addition to<br />

being an anti-glutamatergic agent, <strong>riluzole</strong> also has<br />

antioxidant effects and can protect dopaminergic neurons<br />

against oxidative stress by reducing lipid peroxidation<br />

and ATP consumption [19, 27, 35, 36].<br />

In conclusion, we found that <strong>riluzole</strong> can attenuate<br />

<strong>morphine</strong>-induced tolerance and apoptosis, as well as<br />

decrease the <strong>morphine</strong>-induced elevation in glutamate<br />

concentration in the lumbar spinal cord <strong>of</strong> rats. However,<br />

further studies are required to clarify the mechanisms<br />

underlying the <strong>riluzole</strong> effects on <strong>morphine</strong>induced<br />

tolerance.<br />

Acknowledgment:<br />

This research was supported by Drug Applied Research Center <strong>of</strong><br />

Tabriz University <strong>of</strong> Medical Sciences grant.<br />

References:<br />

1. Albo F, Pieri M, Zona C: Modulation <strong>of</strong> AMPA receptors<br />

in spinal motor neurons by the neuroprotective agent <strong>riluzole</strong>.<br />

J Neurosci Res, 2004, 78, 200–207.<br />

2. Asl BH, Hassanzadeh K, Khezri E, Mohammadi S:<br />

Evaluation the effects <strong>of</strong> dextromethorphan and midazolam<br />

on <strong>morphine</strong> induced tolerance and dependence in<br />

mice. Pak J Biol Sci, 2008, 11, 1690–1695.<br />

3. Bobula B, Hess G: Effects <strong>of</strong> <strong>morphine</strong> and methadone<br />

treatments on glutamatergic transmission in rat frontal<br />

cortex. Pharmacol Rep, 2009, 61, 1192–1197.<br />

4. Bellmann K, Jaattela M, Wissing D, Burkart V, Kolb H:<br />

Heat shock protein hsp70 overexpression confers resistance<br />

against nitric oxide. FEBS Lett, 1996, 391, 185–188.<br />

5. Boronat MA, Garcia-Fuster MJ, Garcia-Sevilla JA:<br />

Chronic <strong>morphine</strong> induces up-regulation <strong>of</strong> the proapoptotic<br />

Fas receptor and down-regulation <strong>of</strong> the anti-<br />

Pharmacological Reports, 2010, 62, 664–673 671


apoptotic Bcl-2 oncoprotein in rat brain. Br J Pharmacol,<br />

2001, 134, 1263–1270.<br />

6. Centonze D, Calabresi P, Pisani A, Marinelli S, Marfia<br />

GA, Bernardi G: Electrophysiology <strong>of</strong> the neuroprotective<br />

agent <strong>riluzole</strong> on striatal spiny neurons. Neuropharmacology,<br />

1998, 37, 1063–1070.<br />

7. Chen Q, Cui J, Zhang Y, Yu LC: Prolonged <strong>morphine</strong> application<br />

modulates Bax and Hsp70 levels in primary rat<br />

neurons. Neurosci Lett, 2008, 441, 311–314.<br />

8. Debono MW, Le Guern J, Canton T, Doble A, Pradier L:<br />

Inhibition by <strong>riluzole</strong> <strong>of</strong> electrophysiological responses<br />

mediated by rat kainate and NMDA receptors expressed in<br />

Xenopus oocytes. Eur J Pharmacol, 1993, 235, 283–289.<br />

9. Doble A: The role <strong>of</strong> excitotoxicity in neurodegenerative<br />

disease: implications for therapy. Pharmacol Ther, 1999,<br />

81, 163–221.<br />

10. Eddy NB, Leimbach D: Synthetic analgesics. II. Dithienylbutenyl-<br />

and dithienylbutylamines. J Pharmacol Exp<br />

Ther, 1953, 107, 385–393.<br />

11. Gordon SA, H<strong>of</strong>fman RA, Simmons RL, Ford HR: Induction<br />

<strong>of</strong> heat shock protein 70 protects thymocytes<br />

against radiation-induced apoptosis. Arch Surg, 1997,<br />

132, 1277–1282.<br />

12. Habibi-Asl B, Hassanzadeh, K, Moosazadeh, S: Effects<br />

<strong>of</strong> ketamine and magnesium on <strong>morphine</strong> induced tolerance<br />

and dependence in mice. DARU, 2005, 13, 110–115.<br />

13. Habibi-Asl B, Hassanzadeh K, Vafai H, Mohammadi S:<br />

Development <strong>of</strong> <strong>morphine</strong> induced tolerance and withdrawal<br />

symptoms is attenuated by lamotrigine and magnesium<br />

sulfate in mice. Pak J Biol Sci, 2009, 12, 798–803.<br />

14. Habibi-Asl B, Hassanzadeh K, Charkhpour M: Central<br />

<strong>administration</strong> <strong>of</strong> minocycline and <strong>riluzole</strong> <strong>prevents</strong><br />

<strong>morphine</strong>-induced tolerance in rats. Anesth Analg, 2009,<br />

109, 936–942.<br />

15. Habibi-Asl B, Alimohammadi B, Charkhpour M, Hassanzadeh<br />

K: Evaluation the effects <strong>of</strong> systemic <strong>administration</strong><br />

<strong>of</strong> minocycline and <strong>riluzole</strong> on tolerance to <strong>morphine</strong><br />

analgesic effect in rat. Pharmaceutical Sciences<br />

(Journal <strong>of</strong> Faculty <strong>of</strong> Pharmacy, Tabriz University <strong>of</strong><br />

Medical Sciences) (Persian), 2009, 15, 205–212.<br />

16. Heurteaux C, Laigle C, Blondeau N, Jarretou G, Lazdunski<br />

M: Alpha-linolenic acid and <strong>riluzole</strong> treatment confer<br />

cerebral protection and improve survival after focal brain<br />

ischemia. Neuroscience, 2006, 137, 241–251.<br />

17. Inturrisi CE: Preclinical evidence for a role <strong>of</strong> glutamatergic<br />

systems in opioid tolerance and dependence.<br />

Semin Neurosci, 1997, 9, 110–119.<br />

18. Jantas D, Lasoñ W: Anti-apoptotic effect <strong>of</strong> memantine<br />

against staurosporine- and low-potassium-induced cell<br />

death in cerebellar granule cells: a development-dependent<br />

effect. Pharmacol Rep, 2009, 61, 827–837.<br />

19. Koh JY, Kim DK, Hwang JY, Kim YH, Seo JH: Antioxidative<br />

and proapoptotic effects <strong>of</strong> <strong>riluzole</strong> on cultured<br />

cortical neurons. J Neurochem, 1999, 72, 716–723.<br />

20. Li CY, Lee JS, Ko YG, Kim JI, Seo JS: Heat shock protein<br />

70 inhibits apoptosis downstream <strong>of</strong> cytochrome c<br />

release and upstream <strong>of</strong> caspase-3 activation. J Biol<br />

Chem, 2000, 275, 25665–25671.<br />

21. Lips J, de Haan P, Bodewits P, Vanicky I, Dzoljic M, Jacobs<br />

MJ, Kalkman CJ: Neuroprotective effects <strong>of</strong> <strong>riluzole</strong><br />

and ketamine during transient spinal cord ischemia<br />

in the rabbit. Anesthesiology, 2000, 93, 1303–1311.<br />

22. Mao J: NMDA and opioid receptors: their interactions in<br />

antinociception, tolerance and neuroplasticity. Brain Res<br />

Brain Res Rev, 1999, 30, 289–304.<br />

23. Mao J, Price DD, Zhu J, Lu J, Mayer DJ: The inhibition<br />

<strong>of</strong> nitric oxide-activated poly(ADP-ribose) synthetase attenuates<br />

transsynaptic alteration <strong>of</strong> spinal cord dorsal<br />

horn neurons and neuropathic pain in the rat. Pain, 1997,<br />

72, 355–366.<br />

24. Mao J, Sung B, Ji RR, Lim G: Neuronal apoptosis associated<br />

with <strong>morphine</strong> tolerance: evidence for an opioidinduced<br />

neurotoxic mechanism. J Neurosci, 2002, 22,<br />

7650–7661.<br />

25. Mosser DD, Caron AW, Bourget L, Denis-Larose C,<br />

Massie B: Role <strong>of</strong> the human heat shock protein hsp70 in<br />

protection against stress-induced apoptosis. Mol Cell<br />

Biol, 1997, 17, 5317–5327.<br />

26. Nemati M, Oveisi M R, Abdollahi H, Sabzevari O: Differentiation<br />

<strong>of</strong> bovine and porcine gelatins using principal<br />

component analysis. J Pharm Biomed Anal, 2004,<br />

34, 485–492.<br />

27. Noh KM, Hwang JY, Shin HC, Koh JY: A novel neuroprotective<br />

mechanism <strong>of</strong> <strong>riluzole</strong>: direct inhibition <strong>of</strong><br />

protein kinase C. Neurobiol Dis, 2000, 7, 375–383.<br />

28. Paxinos G, Watson C: The rat brain in stereotaxic coordinates,<br />

4th edn., Academic Press, London, 1998.<br />

29. Porreca F, Mosberg HI, Hurst R, Hruby VJ, Burks TF:<br />

Roles <strong>of</strong> mu, delta and kappa opioid receptors in spinal<br />

and supraspinal mediation <strong>of</strong> gastrointestinal transit effects<br />

and hot plate analgesia in the mouse. J Pharmacol<br />

Exp Ther, 1984, 230, 341–348.<br />

30. Rothman SM, Olney JW: Glutamate and the pathophysiology<br />

<strong>of</strong> hypoxic-ischemic brain damage. Ann Neurol,<br />

1986, 19, 105–111.<br />

31. Soufi FG, Farajnia S, Aslanabadi N, Ahmadiasl N, Alipour<br />

M, Alipour M, Doustar Y, Sheikhzadeh F: Longterm<br />

exercise training affects age-induced changes in<br />

HSP70 and apoptosis in rat heart. Gen Physiol Biophys,<br />

2008, 27, 263–270.<br />

32. Salinska E, Danysz W, Lazarewicz JW: The role <strong>of</strong> excitotoxicity<br />

in neurodegeneration. Folia Neuropathol,<br />

2005, 43, 322–339.<br />

33. Singhal PC, Kapasi AA, Reddy K, Franki N, Gibbons N,<br />

Ding G: Morphine promotes apoptosis in Jurkat cells.<br />

J Leukoc Biol, 1999, 66, 650–658.<br />

34. Singhal PC, Sharma P, Kapasi AA, Reddy K, Franki N,<br />

Gibbons N: Morphine enhances macrophage apoptosis.<br />

J Immunol, 1998, 160, 1886–1893.<br />

35. Storch A, Burkhardt K, Ludolph AC, Schwarz J: Protective<br />

effects <strong>of</strong> <strong>riluzole</strong> on dopamine neurons: involvement<br />

<strong>of</strong> oxidative stress and cellular energy metabolism.<br />

J Neurochem, 2000, 75, 2259–2269.<br />

36. Toklu HZ, Uysal MK, Kabasakal L, Sirvanci S, Ercan F,<br />

Kaya M: The effects <strong>of</strong> <strong>riluzole</strong> on neurological, brain<br />

biochemical, and histological changes in early and late<br />

term <strong>of</strong> sepsis in rats. J Surg Res, 2009, 152, 238–248.<br />

37. Trujillo KA: Effects <strong>of</strong> noncompetitive N-methyl-Daspartate<br />

receptor antagonists on opiate tolerance and<br />

physical dependence. Neuropsychopharmacology, 1995,<br />

13, 301–307.<br />

672 Pharmacological Reports, 2010, 62, 664–673


Riluzole <strong>prevents</strong> <strong>morphine</strong>-induced apoptosis in the rat spinal cord<br />

Kambiz Hassanzadeh et al.<br />

38. Trujillo KA: The neurobiology <strong>of</strong> opiate tolerance, dependence<br />

and sensitization: mechanisms <strong>of</strong> NMDA<br />

receptor-dependent synaptic plasticity. Neurotox Res,<br />

2002, 4, 373–391.<br />

39. Trujillo KA, Akil H: Inhibition <strong>of</strong> <strong>morphine</strong> tolerance<br />

and dependence by the NMDA receptor antagonist<br />

MK-801. Science, 1991, 251, 85–87.<br />

40. Wang SJ, Wang KY, Wang WC: Mechanisms underlying<br />

the <strong>riluzole</strong> inhibition <strong>of</strong> glutamate release from rat cerebral<br />

cortex nerve terminals (synaptosomes). Neuroscience,<br />

2004, 125, 191–201.<br />

41. Whiteside GT, Munglani R: Cell death in the superficial<br />

dorsal horn in a model <strong>of</strong> neuropathic pain. J Neurosci<br />

Res, 2001, 64, 168–173.<br />

Received:<br />

October 20, 2009; in revised form: January 19, 2010.<br />

Pharmacological Reports, 2010, 62, 664–673 673

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!