AJA Asian Journal of Anesthesiology

Advancing, Capability, Improving lives

Research Paper
Volume 54, Issue 2, Pages 65-69
KambizHassanzadeh 1 , Bashir Khodadadi 2 , Mohammad Raman Moloudi 1.3 , Hassan Amini 1 , Mohammad Reza Rahmani 4 , Esmael Izadpanah 1
4707 Views


Abstract

Objective

Tolerance to the analgesic effect is the main side effect of chronic administration of opioids. Several drugs have been studied to try to find agents to prevent the development of this phenomenon. In the present study we aimed to evaluate the effect of thalidomide on morphine-induced tolerance to the analgesic effect.

Methods

Groups of male rats were randomly rendered and received daily morphine in combination with thalidomide vehicle or thalidomide (2.5 mg/kg, 5 mg/kg, or 10 mg/kg, intraperitoneally). Nociception was measured using the plantar test apparatus. Latency time was recorded when the animal reacted to the light stimulus; licking or raising its hind paw. Treatments and evaluations continued until completion of tolerance to the analgesic effect of morphine.

Results

Our findings indicated that tolerance was achieved following 11 days of morphine administration, while thalidomide postponed the day of tolerance completion for 4 days (2.5 mg/kg and 5 mg/kg thalidomide) or 10 days (10 mg/kg thalidomide). Moreover, thalidomide prevented the morphine-induced shift to the right of the ED50 in the dose–response curve.

Conclusion

It was concluded that thalidomide attenuated the morphine-induced tolerance to the analgesic effect.

Keywords

analgesia ; morphine tolerance ; thalidomide;


1. Introduction

Tolerance to the analgesic effect of opioids is one of the major side effects associated with their long-term administration. This phenomenon limits the beneficial therapeutic use of opioids. Many attempts have been made to find agents that can prevent this adverse effect.

In recent years, inflammatory mediators have been reported to play a key role in pathways of opioid-tolerance induction.123 Morphine augments the secretion of inflammatory cytokines such as tumor necrosis factor alpha (TNF-α), interleukin 1 beta (IL-1β), and interleukin 6 through glial cell activation.4 These inflammatory products enhance the neuronal excitability and sensitize the pain transmission neurons.56 TNF-α may have a pivotal role in the genesis of mechanical inflammatory hyperalgesia in rats.7 It augments glutamate neurotoxicity, upregulates the expression of the NMDA receptors and enhances the glutamatergic transmission.58

Several lines of evidence indicate the benefit of agents that interfere with the inflammatory signaling pathways as a strategic approach to attenuate development of morphine tolerance.910 It has been shown recently that neuroprotective and anti-inflammatory agents, such as ketamine, minocycline, riluzole, donepezil, and pioglitazone, prevented morphine-induced tolerance and apoptosis in the rat central nervous system.1112131415 Researchers indicated that etanercept, a TNF-α inhibitor, restored the antinociceptive effect of morphine by inhibiting the spinal proinflammatory cytokine (e.g., TNF-α, IL-1β, and IL-6) expression in the morphine-tolerant rats.9

There is evidence indicating that thalidomide inhibits the production of human monocyte TNF-α and alveolar macrophages.1617 Thalidomide, a glutamic acid derivative, was approved in 1998 by the US Food and Drug Administration for erythema nodosum leprosum and in 2006 for multiple myeloma.18 This drug was a common over-the-counter sedative and antiemetic until 1961, when it was withdrawn because of teratogenicity.19 In 1965 an accidental discovery of its immunomodulatory effects was made in erythema nodosum leprosum patients.2021 Today thalidomide and its analogs have shown efficacy against a wide variety of diseases, including inflammation and cancer.22 Thalidomide exerts its inhibitory action on TNF-α by enhancing mRNA degradation.23 In addition, it was reported to inhibit inflammatory hyperalgesia in rats and the writhing nociceptive response in mice, possibly due to inhibition of TNF-α production.7

The evidence from these studies prompted us to investigate the effect of thalidomide on morphine-induced tolerance to the analgesic effect.

2. Materials and methods

2.1. Drugs

Morphine sulfate was purchased from Temad Company (Tehran, Iran). It was dissolved in normal saline and injected using 1-mL insulin syringes. Thalidomide (Sigma-Aldrich Inc., Chemie GmbH, Germany) was dissolved in vehicle (dimethyl sulfoxide + saline, 4:1) and injected intraperitoneally (i.p.). All the solutions were freshly prepared on the day of the experiment.

2.2. Animals

Male Wistar rats (n = 88) weighing 250–300 g were purchased from the Razi Institute (Tehran, Iran). The animals were kept in temperature-controlled conditions (25 ± 2˚C) and standard cages (four rats per cage), on a 12-h light/dark cycle with free access to food and water ad libitum. They were randomly divided into several experimental groups, eight animals per group. In order to minimize the nonspecific stress response, animals were habituated to the testing environment, including transfer to the experimental laboratory, weighing, and handling. All the experiments were in accordance with the Guide for the Care and Use of Laboratory Animals (National Institutes of Health Publication No. 85–23, revised 1985), and were approved by the research and ethics committee of Kurdistan University of Medical Sciences.

2.3. Experimental details

The experimental groups are described in Table 1.

2.3.1. Model of tolerance induction

In order to induce a tolerance to the analgesic effect, morphine (10 mg/kg, i.p.) was injected daily according to our previous study.24 It was injected daily 30 minutes after the thalidomide administration.

2.3.2. Assessment of nociception

The nociception was assessed using a plantar test apparatus (IITC Inc. Life Science, Los Angeles, USA) (Hargreaves method).25 Rats were placed on a glass plate on the plantar test apparatus and a noxious heat source was held directly under the hind paw. As soon as it was started, the device supplied a continuous beam stimulus to the paw and the withdrawal reflex was produced. The latency time between exposure to the radiant and the paw withdrawal was measured as the analgesia. For each animal the average for three measurements of the baseline paw-withdrawal latency was determined as the baseline latency.

The intensity of the light was adjusted so that the baseline latencies were 8–10 seconds, with a cut-off time of 20 seconds in order to avoid tissue damage. Two measurements of the latency were averaged for each hind paw in each test session. Maximal possible effect (%MPE) using the following equation was measured for the latency-withdrawal response for each rat:

%MPE = [postdrug latency (s) − baseline latency (s)] / [cut-off value (s) − baseline latency (s)] × 100

It is worth noting that the baseline latency was determined once per day for each rat before the daily injection of morphine (10 mg/kg). Later, the drugs or their vehicles were injected. Thirty minutes after drug/vehicle administration, morphine was injected. Finally, 30 minutes after morphine administration, the postdrug latency was measured. Moreover, the baseline and the latency time were registered daily and the %MPE was calculated. The experiments were continued until there was no significant difference in the %MPE between the vehicle- or the drug-treated groups (the tolerant animals) and the vehicle-received group.26

2.3.3. Total analgesic effect assessment

In order to assess the total analgesic effect in different groups, the area under the curve (AUC) for %MPE against the time was calculated. This analysis allows a comparison of the effects from different analgesic tests. The AUC was calculated from the observed values using the trapezoidal rule.

2.3.4. Analgesic dose–response curves

The dose–response curve was plotted for each group for assessment of tolerance induction. Rats received the thalidomide vehicle or the morphine + thalidomide vehicle or morphine + thalidomide once a day for 11 days. On Day 12 (1 day after the morphine-tolerance completion in the control group), the logarithmic doses of the morphine (1 mg/kg, 10 mg/kg, 100 mg/kg, i.p.) were injected to create the analgesic dose–response curve. In these curves, the morphine antinociceptive 50% effective dose (ED50) values for each group were derived using the linear regression method.27

2.4. Thalidomide analgesic effect assay

In order to find the possible analgesic effect of thalidomide, animals received only the most effective dose of the thalidomide (10 mg/kg, i.p.) or its vehicle. Another test was done 60 minutes later (similar to treatment in assessment of nociception section) to verify whether the observed effect of thalidomide was not simply because of additive mechanism.

2.5. Data analysis

All data from the analgesia tests was presented as the mean of %MPE ± SEM for each group (n = 8). The independent t-test or repeated analysis of variance tests followed by the Bonferroni test were applied to analyze the statistical significance in two or multiple comparisons, respectively. In all analyses, p < 0.05 was considered to be significant.

3. Results

3.1. Tolerance to the analgesic effect of morphine

As shown in Figure 1, on Day 11, there was no significant difference between the control group and the saline + thalidomide vehicle-treated animals; therefore, this day was considered as the date for morphine-tolerance completion. Thus it was found that daily administration of morphine (10 mg/kg, i.p.) for 11 days developed tolerance to the analgesic effect in the control group that received morphine + thalidomide vehicle.

Figure 1.
Download full-size image
Figure 1. The analgesic effect of the daily administration of morphine (10 mg/kg, i.p.) in combination with thalidomide vehicle (control group) or thalidomide (2.5 mg/kg, 5 mg/kg, 10 mg/kg, i.p.). The developed tolerance to the analgesic effect of the morphine was completed on Day 11 in the control group. Each point represents the mean of %MPE ± SEM for eight rats. a = p < 0.05 when compared to the control group for the day of tolerance completion. The arrows represent the day of the morphine tolerance completion. Mor = morphine; %MPE = maximal possible effect; Sal = saline; Tal = thalidomide; Veh = vehicle.

3.2. Effect of thalidomide on the morphine-induced tolerance to the analgesic effect

The results reveal that thalidomide (2.5 mg/kg, 5 mg/kg, 10 mg/kg, i.p.) delayed the tolerance completion for 4, 4, and 10 days, respectively (Figure 1). Furthermore, according to Figure 2, the dose–response curve depicts a significant shift to the right in the animals that received morphine + thalidomide vehicle compared with saline-treated animals. Also, thalidomide prevented a shift when it was co-injected with morphine. In this regard, a significant shift to the left in the ED50 was also seen in the morphine + thalidomide (10 mg/kg, i.p.) treated group (69.75 mg) compared to the control animals (95.9 mg).

Figure 2.
Download full-size image
Figure 2. The plantar test latencies and the percent of the %MPE to the logarithmic doses of the morphine (1 mg/kg, 10 mg/kg, 100 mg/kg, i.p.) administered on Day 11. Each point represents the mean ± SEM for eight rats. Different morphine doses were administered on Day 11 showing a significant shift to the right in the dose–response curve and the antinociceptive ED50 values in animals treated with the morphine + thalidomide vehicle compared to those which received the thalidomide vehicle or the morphine + thalidomide (10 mg/kg, i.p.). Mor = morphine; %MPE = maximal possible effect; Sal = saline; Tal = thalidomide; Veh = vehicle.

Furthermore, the results showed that the most effective dose of thalidomide (10 mg/kg, i.p.) had no significant analgesic effect (Figure 3). Data analysis for total analgesic effect revealed that the dose of 10 mg/kg thalidomide had the greatest AUC of the %MPE (2693.7) in comparison with the control group (1627.7), the 2.5 mg/kg thalidomide group (1608.2), or the 5 mg/kg thalidomide group (1724.5) (Figure 4).

Figure 3.
Download full-size image
Figure 3. The percent of the %MPE of thalidomide (10 mg/kg, i.p.) in comparison with thalidomide vehicle. Each point represents the mean of %MPE ± SEM for eight rats. %MPE = maximal possible effect; Tal = Thalidomide; Veh = vehicle.
Figure 4.
Download full-size image
Figure 4. AUC of percentage of %MPE was calculated for each group for 21 days. To calculate the AUC, the trapezoidal rule was used. One-way analysis of variance followed by Tukey's test was used to analyze the differences between the control and treatment groups. In all analyses, p < 0.05 was considered to be significant. * p < 0.001; when compared to the control (Mor + Tal Veh) group. AUC = area under the curve; Mor = morphine; %MPE = maximal possible effect; Sal = saline; Tal = thalidomide; Veh = vehicle.

4. Discussion

According to the results, chronic administration of morphine for 11 days developed a tolerance to its analgesic effects. Several efforts have been performed to find the possible mechanisms of morphine tolerance. These evidences indicate that chronic morphine administration leads to elevation of proinflammatory mediators and glial activation in the morphine-tolerant animal's spinal cord; therefore, the analgesic effect of morphine is reduced.8928 TNF-α is a known important proinflammatory cytokine and provides either a neurotoxic or neuroprotective effect in different central nervous system disorders. Furthermore, TNF-α has been proven to correlate with neuronal plasticity via activation of spinal glial cells and escalation of glutamate system transmission.2529

In this regard, Ghavimi et al demonstrated that long-term morphine treatment augmented proinflammatory cytokines, including TNF-α, IL-1b, and IL-6, in the cerebral cortex of rats. They revealed that there was an association between morphine tolerance and an increase in the above-mentioned cytokines.14 Also, Zou et al showed that TNF-α increases glutamate-induced neurotoxicity by inhibiting the glutamate uptake in brain-slice cultures.30 Moreover, TNF-α upregulates the expression of the NMDA receptors and enhances the glutamatergic transmission,58which has a major role in the mechanisms of morphine tolerance.31

Supporting these findings there are documents indicating that agents with anti-inflammatory effects are effective in the attenuation of the antinociceptive tolerance of opioids.243233

In addition, it has been demonstrated that chronic administration of glial inhibitors,283334 or blocking glial proinflammatory mediators' signaling, can significantly attenuate morphine tolerance.69103536 Pentoxifylline and minocycline are examples of such agents that have efficacy on the suppression of glial activation and the attenuation of morphine-induced tolerance in animal models.3233 Therefore, the above strategies may provide adjuvant therapy for morphine tolerance, which would develop the application of opioids in clinical pain management.37

For the first time, the findings of the present study showed that injection of thalidomide (10 mg/kg, i.p.), in combination with morphine, attenuated the development of tolerance by shifting the day of completion of the tolerance to the right.

Additionally, thalidomide (10 mg/kg) in combination with morphine provided a greater analgesic effect in the total analgesia analysis than morphine alone. On the other hand, thalidomide by itself showed no significant analgesic effect; this suggests that thalidomide did not simply enhance the morphine analgesia through an additive mechanism.

Palencia et al showed that thalidomide exerts protective effects against cerebral ischemia/reperfusion damage by attenuating both oxidative stress and inflammatory response. They believed that thalidomide protects neurons from cell death by inhibiting oxidative stress and the expression of proinflammatory cytokines (TNF- α and IL-1β).38

Moreover, thalidomide has potent immunomodulatory and anti-TNF-α properties. It inhibits the chemotaxis and cytokine-induced nuclear factor kappa B transcription factor gene expression.39404142

Our results also showed that thalidomide prevented the shifting of the dose–response curve and the ED50 to the right, the phenomenon occurring in morphine-tolerant group.

Another possible mechanism for morphine tolerance is an increase of nitric oxide or induction of guanylyl cyclase and agents that inhibit nitric oxide synthase expression,43 attenuating the development of morphine tolerance and dependence.44On the other hand, it was shown that thalidomide has a beneficial effect in neuropathic pain by decreasing cytokines (TNF-α) and nitric oxide levels.45 Therefore, thalidomide may provide a promising novel therapeutic approach for management of morphine-induced tolerance to the analgesic effect.

5. Conclusion

According to the results it was concluded that thalidomide prevented morphine-induced tolerance to analgesic effects. This effect might be related to its properties in the inhibition of TNF-α, prevention of microglial activation, and/or glutamate neurotoxicity.

Acknowledgments

The authors thank the Vice Chancellor's Office for Research Affairs of Kurdistan University of Medical Sciences for the grant supporting the present study. The data of this study was obtained from medical student thesis (submitted to the Faulty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran).


References

1
V. Raghavendra, M.D. Rutkowski, J.A. DeLeo
The role of spinal neuroimmune activation in morphine tolerance/hyperalgesia in neuropathic and sham-operated rats
J Neurosci, 22 (22) (2002), pp. 9980-9989
2
I.N. Johnston, E.D. Milligan, J. Wieseler-Frank, M.G. Frank, V. Zapata, J. Campisi, et al.
A role for proinflammatory cytokines and fractalkine in analgesia, tolerance, and subsequent pain facilitation induced by chronic intrathecal morphine
J Neurosci, 24 (33) (2004), pp. 7353-7365
3
M.R. Hutchinson, S.T. Bland, K.W. Johnson, K.C. Rice, S.F. Maier, L.R. Watkins
Opioid-induced glial activation: mechanisms of activation and implications for opioid analgesia, dependence, and reward
Sci World J, 7 (2007), pp. 98-111
4
M.R. Hutchinson, Y. Shavit, P.M. Grace, K.C. Rice, S.F. Maier, L.R. Watkins
Exploring the neuroimmunopharmacology of opioids: an integrative review of mechanisms of central immune signaling and their implications for opioid analgesia
Pharmacol Rev, 63 (3) (2011), pp. 772-810
5
L.R. Watkins, M.R. Hutchinson, I.N. Johnston, S.F. Maier
Glia: novel counter-regulators of opioid analgesia
Trends Neurosci, 28 (12) (2005), pp. 661-669
6
M.R. Hutchinson, B.D. Coats, S.S. Lewis, Y. Zhang, D.B. Sprunger, N. Rezvani, et al.
Proinflammatory cytokines oppose opioid-induced acute and chronic analgesia
Brain Behav Immun, 22 (8) (2008), pp. 1178-1189
7
R.A. Ribeiro, M.L. Vale, S.H. Ferreira, F.Q. Cunha
Analgesic effect of thalidomide on inflammatory pain
Eur J Pharmacol, 391 (1–2) (2000), pp. 97-103
8
C.H. Shen, R.Y. Tsai, Y.H. Tai, S.L. Lin, C.C. Chien, C.S. Wong
Intrathecal etanercept partially restores morphine's antinociception in morphine-tolerant rats via attenuation of the glutamatergic transmission
Anesth Analg, 113 (1) (2011), pp. 184-190
9
C.H. Shen, R.Y. Tsai, M.S. Shih, S.L. Lin, Y.H. Tai, C.C. Chien, et al.
Etanercept restores the antinociceptive effect of morphine and suppresses spinal neuroinflammation in morphine-tolerant rats
Anesth Analg, 112 (2) (2011), pp. 454-459
10
C.H. Liu, C.H. Cherng, S.L. Lin, C.C. Yeh, C.T. Wu, Y.H. Tai, et al.
N-methyl-D-aspartate receptor antagonist MK-801 suppresses glial pro-inflammatory cytokine expression in morphine-tolerant rats
Pharmacol Biochem Behav, 99 (3) (2011), pp. 371-380
11
K. Hassanzadeh, B. Habibi-asl, S. Farajnia, L. Roshangar
Minocycline prevents morphine-induced apoptosis in rat cerebral cortex and lumbar spinal cord: a possible mechanism for attenuating morphine tolerance
Neurotox Res, 19 (4) (2011), pp. 649-659
12
K. Hassanzadeh, B. Habibi-asl, L. Roshangar, M. Nemati, M. Ansarin, S. Farajnia
Intracerebroventricular administration of riluzole prevents morphine-induced apoptosis in the lumbar region of the rat spinal cord
Pharmacol Rep, 62 (4) (2010), pp. 664-673
13
M. Sharifipour, E. Izadpanah, B. Nikkhoo, S. Zare, A. Abdolmaleki, K. Hassanzadeh, et al.
A new pharmacological role for donepezil: attenuation of morphine-induced tolerance and apoptosis in rat central nervous system
J Biomed Sci, 21 (2014), p. 6
CrossRef  
14
H. Ghavimi, M. Charkhpour, S. Ghasemi, M. Mesgari, H. Hamishehkar, K. Hassanzadeh, et al.
Pioglitazone prevents morphine antinociceptive tolerance via ameliorating neuroinflammation in rat cerebral cortex
Pharmacol Rep, 67 (1) (2015), pp. 78-84
15
K. Hassanzadeh, B. Habibi-Asl
Effects of ketamine and midazolam on morphine induced dependence and tolerance in mice
DARU, 12 (3) (2004), pp. 101-105
16
E.P. Sampaio, E.N. Sarno, R. Galilly, Z.A. Cohn, G. Kaplan
Thalidomide selectively inhibits tumor necrosis factor alpha production by stimulated human monocytes
J Exp Med, 173 (3) (1991), pp. 699-703
17
J.L. Tavares, A. Wangoo, P. Dilworth, B. Marshall, S. Kotecha, R.J. Shaw
Thalidomide reduces tumour necrosis factor-alpha production by human alveolar macrophages
Respir Med, 91 (1) (1997), pp. 31-39
18
C.S. Yang, C. Kim, R.J. Antaya
Review of thalidomide use in the pediatric population
J Am Acad Dermatol, 72 (4) (2015), pp. 703-711
19
W.G. McBride
Thalidomide embryopathy
Teratology, 16 (1) (1977), pp. 79-82
20
J. Sheskin
The treatment of lepra reaction in lepromatous leprosy. Fifteen years' experience with thalidomide
Int J Dermatol, 19 (6) (1980), pp. 318-322
21
J. Sheskin
Thalidomide in the Treatment of Lepra Reactions
Clin Pharmacol Ther, 6 (1965), pp. 303-306
22
S. Zhou, F. Wang, T.C. Hsieh, J.M. Wu, E. Wu
Thalidomide–a notorious sedative to a wonder anticancer drug
Curr Med Chem, 20 (33) (2013), pp. 4102-4108
23
A.L. Moreira, E.P. Sampaio, A. Zmuidzinas, P. Frindt, K.A. Smith, G. Kaplan
Thalidomide exerts its inhibitory action on tumor necrosis factor alpha by enhancing mRNA degradation
J Exp Med, 177 (6) (1993), pp. 1675-1680
24
F. Ghasemi, A. Moradi, E. Izadpanah, M.R. Moloudi, K. Hassanzadeh, A. Rahimmi
Simvastatin prevents morphine antinociceptive tolerance and withdrawal symptoms in rats
J Formos Med Assoc, 114 (5) (2015), pp. 399-406
25
K. Hargreaves, R. Dubner, F. Brown, C. Flores, J. Joris
A new and sensitive method for measuring thermal nociception in cutaneous hyperalgesia
Pain, 32 (1) (1988), pp. 77-88
26
F.E. D'Amour, D.L. Smith
A method for determining loss of pain sensation
J Pharmacol Exp Ther, 72 (1) (1941), pp. 74-79
27
R.J. McCarthy, J.S. Kroin, K.J. Tuman, R.D. Penn, A.D. Ivankovich
Antinociceptive potentiation and attenuation of tolerance by intrathecal co-infusion of magnesium sulfate and morphine in rats
Anesth Analg, 86 (4) (1998), pp. 830-836
28
V. Raghavendra, F.Y. Tanga, J.A. DeLeo
Attenuation of morphine tolerance, withdrawal-induced hyperalgesia, and associated spinal inflammatory immune responses by propentofylline in rats
Neuropsychopharmacol, 29 (2) (2004), pp. 327-334
29
T. Berta, T. Liu, Y.C. Liu, Z.Z. Xu, R.R. Ji
Acute morphine activates satellite glial cells and up-regulates IL-1beta in dorsal root ganglia in mice via matrix metalloprotease-9
Mol Pain, 8 (2012), p. 18
30
J.Y. Zou, F.T. Crews
TNF alpha potentiates glutamate neurotoxicity by inhibiting glutamate uptake in organotypic brain slice cultures: neuroprotection by NF kappa B inhibition
Brain Res, 1034 (1–2) (2005), pp. 11-24
31
K.A. Trujillo, H. Akil
Inhibition of morphine tolerance and dependence by the NMDA receptor antagonist MK-801
Science, 251 (4989) (1991), pp. 85-87
32
H. Hameed, M. Hameed, P.J. Christo
The effect of morphine on glial cells as a potential therapeutic target for pharmacological development of analgesic drugs
Curr Pain Headache Rep, 14 (2) (2010), pp. 96-104
33
M.R. Hutchinson, S.S. Lewis, B.D. Coats, D.A. Skyba, N.Y. Crysdale, D.L. Berkelhammer, et al.
Reduction of opioid withdrawal and potentiation of acute opioid analgesia by systemic AV411 (ibudilast)
Brain Behav Immun, 23 (2) (2009), pp. 240-250
34
J. Mika, A. Wawrzczak-Bargiela, M. Osikowicz, W. Makuch, B. Przewlocka
Attenuation of morphine tolerance by minocycline and pentoxifylline in naive and neuropathic mice
Brain Behav Immun, 23 (1) (2009), pp. 75-84
35
Y. Shavit, G. Wolf, I. Goshen, D. Livshits, R. Yirmiya
Interleukin-1 antagonizes morphine analgesia and underlies morphine tolerance
Pain, 115 (1–2) (2005), pp. 50-59
36
Y. Li, Y. Shu, Q. Ji, J. Liu, X. He, W. Li
Attenuation of morphine analgesic tolerance by rosuvastatin in naive and morphine tolerance rats
Inflammation, 38 (1) (2015), pp. 134-141
37
C.H. Shen, R.Y. Tsai, C.S. Wong
Role of neuroinflammation in morphine tolerance: effect of tumor necrosis factor-alpha
Acta Anaesthesiol Taiwan, 50 (4) (2012), pp. 178-182
38
G. Palencia, J.A. Medrano, A. Ortiz-Plata, D.J. Farfan, J. Sotelo, A. Sanchez, et al.
Anti-apoptotic, anti-oxidant, and anti-inflammatory effects of thalidomide on cerebral ischemia/reperfusion injury in rats
J Neurol Sci, 351 (1–2) (2015), pp. 78-87
39
T.E. Clark, N. Edom, J. Larson, L.J. Lindsey
Thalomid (Thalidomide) capsules: a review of the first 18 months of spontaneous postmarketing adverse event surveillance, including off-label prescribing
Drug Saf, 24 (2) (2001), pp. 87-117
40
J.J. Wu, D.B. Huang, K.R. Pang, S. Hsu, S.K. Tyring
Thalidomide: dermatological indications, mechanisms of action and side-effects
Br J Dermatol, 153 (2) (2005), pp. 254-273
41
W. Rehman, L.M. Arfons, H.M. Lazarus
The rise, fall and subsequent triumph of thalidomide: lessons learned in drug development
Ther Adv Hematol, 2 (5) (2011), pp. 291-308
42
T. Kawai, N. Watanabe, M. Yokoyama, K. Arai, S. Oana, S. Harayama, et al.
Thalidomide attenuates excessive inflammation without interrupting lipopolysaccharide-driven inflammatory cytokine production in chronic granulomatous disease
Clin Immunol, 147 (2) (2013), pp. 122-128
43
N. Durmus, I. Bagcivan, E. Ozdemir, A. Altun, S. Gursoy
Soluble guanylyl cyclase activators increase the expression of tolerance to morphine analgesic effect
Bratisl Lek Listy, 115 (6) (2014), pp. 334-339
44
A.O. Abdel-Zaher, M.G. Mostafa, H.M. Farghly, M.M. Hamdy, G.A. Omran, N.K. Al-Shaibani
Inhibition of brain oxidative stress and inducible nitric oxide synthase expression by thymoquinone attenuates the development of morphine tolerance and dependence in mice
Eur J Pharmacol, 702 (1–3) (2013), pp. 62-70
45
R. Taliyan, P.L. Sharma
Possible mechanism of protective effect of thalidomide in STZ-induced-neuropathic pain behavior in rats
Inflammopharmacology, 20 (2) (2012), pp. 89-97

References

Close