Skip to main content
Top
Published in: BMC Anesthesiology 1/2015

Open Access 01-12-2015 | Research article

Melatonin prevents morphine-induced hyperalgesia and tolerance in rats: role of protein kinase C and N-methyl-D-aspartate receptors

Authors: Li Song, Chaoran Wu, Yunxia Zuo

Published in: BMC Anesthesiology | Issue 1/2015

Login to get access

Abstract

Background

Morphine-induced hyperalgesia and tolerance significantly limits its clinical use in relieving acute and chronic pain. Melatonin, a pineal gland neurohormone, has been shown to participate in certain neuropsychopharmacological actions. The present study investigated the effect of melatonin on morphine-induced hyperalgesia and tolerance and possible involvement of protein kinase C (PKC)/N-methyl-D-aspartate (NMDA) pathway in melatonin-mediated.

Methods

Experiments were performed on adult, male Sprague–Dawley rats. Melatonin (10 mg/kg, intraperitoneal, i.p.) or saline was administrated 10 min after morphine injection (10 mg/kg, subcutaneous, s.c.) each day for consecutive 14 days. Withdrawal threshold of the hindpaw to mechanical and thermal stimulation was measured before any drug administration and one hour after melatonin or saline on each designated test day. On the 15th day, thermal withdrawal was measured after s.c. morphine (20 mg/kg), but not melatonin, and morphine tolerance was measured and expressed by MPAE% (percent of maximal possible anti-nociceptive effect) of morphine. Levels of expression of protein kinase C gamma (PKCγ) and NMDA receptor subtype NR1 in spinal cord were detected by Western blotting.

Results

The mechanical withdrawal threshold and thermal withdrawal latency decreased and shortened significantly (i.e., threshold decreased) in rats that received morphine treatment for two weeks compared with that in rats receiving saline. This morphine-induced mechanical and thermal hyperalgesia were greatly attenuated by co-administration of morphine with melatonin. The MPAE% representing morphine analgesic effect was reduced approximately 60% in rats that received morphine treatment. However, following the treatment of morphine with melatonin, the MPAE% was reduced only about 30%, comparing with those that received saline treatment as control. Administration of morphine alone resulted in significantly increased expression of PKCγ and NR1 proteins in the spinal cord. These increased levels of expression of PKCγ and NR1 were significantly inhibited by co-administration of morphine with melatonin.

Conclusions

Our findings demonstrate that melatonin have potential to attenuate repetitive morphine-induced hyperalgesia and tolerance, possibly by inhibiting PKCγ and NR1 activities in the spinal cord.
Literature
1.
go back to reference Somogyi AA, Barratt DT, Coller JK: Pharmacogenetics of opioids.Clin Pharmacol Ther 2007, 1:429–44.CrossRef Somogyi AA, Barratt DT, Coller JK: Pharmacogenetics of opioids.Clin Pharmacol Ther 2007, 1:429–44.CrossRef
2.
go back to reference Hutchinson MR, Shavit Y, Grace PM, Rice KC, Maier SF, Watkins LR: Exploring the neuroimmunopharmacology of opioids: an integrative review of mechanisms of central immune signaling and their implications for opioid analgesia[J].Pharmacol Rev 2011, 63:772–810. 10.1124/pr.110.004135CrossRefPubMedPubMedCentral Hutchinson MR, Shavit Y, Grace PM, Rice KC, Maier SF, Watkins LR: Exploring the neuroimmunopharmacology of opioids: an integrative review of mechanisms of central immune signaling and their implications for opioid analgesia[J].Pharmacol Rev 2011, 63:772–810. 10.1124/pr.110.004135CrossRefPubMedPubMedCentral
3.
go back to reference Xin W, Chun W, Ling L, Wei W: Role of melatonin in the prevention of morphine-induced hyperalgesia and spinal glial activation in rats: protein kinase C pathway involved.Int J Neurosci 2012, 122:154–63. 10.3109/00207454.2011.635828CrossRefPubMed Xin W, Chun W, Ling L, Wei W: Role of melatonin in the prevention of morphine-induced hyperalgesia and spinal glial activation in rats: protein kinase C pathway involved.Int J Neurosci 2012, 122:154–63. 10.3109/00207454.2011.635828CrossRefPubMed
4.
go back to reference Mao J, Price DD, Mayer DJ: Experimental mononeuropathy reduces the antinociceptive effects of morphine: implications for common intracellular mechanisms involved in morphine tolerance and neuropathic pain.Pain 1995, 61:353–64. 10.1016/0304-3959(95)00022-KCrossRefPubMed Mao J, Price DD, Mayer DJ: Experimental mononeuropathy reduces the antinociceptive effects of morphine: implications for common intracellular mechanisms involved in morphine tolerance and neuropathic pain.Pain 1995, 61:353–64. 10.1016/0304-3959(95)00022-KCrossRefPubMed
5.
go back to reference Mao J, Price DD, Mayer DJ: Mechanisms of hyperalgesia and morphine tolerance: a current view of their possible interactions.Pain 1995, 62:259–74. 10.1016/0304-3959(95)00073-2CrossRefPubMed Mao J, Price DD, Mayer DJ: Mechanisms of hyperalgesia and morphine tolerance: a current view of their possible interactions.Pain 1995, 62:259–74. 10.1016/0304-3959(95)00073-2CrossRefPubMed
6.
go back to reference Mayer DJ, Mao J, Holt J, Price DD: Cellular mechanisms of neuropathic pain, morphine tolerance, and their interactions.Proc Natl Acad Sci U S A 1999, 96:7731–6. 10.1073/pnas.96.14.7731CrossRefPubMedPubMedCentral Mayer DJ, Mao J, Holt J, Price DD: Cellular mechanisms of neuropathic pain, morphine tolerance, and their interactions.Proc Natl Acad Sci U S A 1999, 96:7731–6. 10.1073/pnas.96.14.7731CrossRefPubMedPubMedCentral
7.
go back to reference Lim G, Wang S, Zeng Q, Sung B, Yang L, Mao J: Expression of spinal NMDA receptor and PKCgamma after chronic morphine is regulated by spinal glucocorticoid receptor.J Neurosci 2005, 25:11145–54. 10.1523/JNEUROSCI.3768-05.2005CrossRefPubMed Lim G, Wang S, Zeng Q, Sung B, Yang L, Mao J: Expression of spinal NMDA receptor and PKCgamma after chronic morphine is regulated by spinal glucocorticoid receptor.J Neurosci 2005, 25:11145–54. 10.1523/JNEUROSCI.3768-05.2005CrossRefPubMed
8.
go back to reference Highfield DA, Grant S: Ng-nitro-L-arginine, an NOS inhibitor, reduces tolerance to morphine in the rat locus coeruleus.Synapse 1998, 29:233–9. 10.1002/(SICI)1098-2396(199807)29:3<233::AID-SYN5>3.0.CO;2-7CrossRefPubMed Highfield DA, Grant S: Ng-nitro-L-arginine, an NOS inhibitor, reduces tolerance to morphine in the rat locus coeruleus.Synapse 1998, 29:233–9. 10.1002/(SICI)1098-2396(199807)29:3<233::AID-SYN5>3.0.CO;2-7CrossRefPubMed
9.
go back to reference Mao J, Price DD, Mayer DJ: Thermal hyperalgesia in association with the development of morphine tolerance in rats: roles of excitatory amino acid receptors and protein kinase C.J Neurosci 1994, 14:2301–12.PubMed Mao J, Price DD, Mayer DJ: Thermal hyperalgesia in association with the development of morphine tolerance in rats: roles of excitatory amino acid receptors and protein kinase C.J Neurosci 1994, 14:2301–12.PubMed
10.
go back to reference Mayer DJ, Mao J, Price DD: The development of morphine tolerance and dependence is associated with translocation of protein kinase C.Pain 1995, 61:365–74. 10.1016/0304-3959(95)00023-LCrossRefPubMed Mayer DJ, Mao J, Price DD: The development of morphine tolerance and dependence is associated with translocation of protein kinase C.Pain 1995, 61:365–74. 10.1016/0304-3959(95)00023-LCrossRefPubMed
11.
go back to reference Mao J, Sung B, Ji RR, Lim G: Chronic morphine induces downregulation of spinal glutamate transporters: implications in morphine tolerance and abnormal pain sensitivity.J Neurosci 2002, 22:8312–23.PubMed Mao J, Sung B, Ji RR, Lim G: Chronic morphine induces downregulation of spinal glutamate transporters: implications in morphine tolerance and abnormal pain sensitivity.J Neurosci 2002, 22:8312–23.PubMed
12.
go back to reference Bailey CP, Smith FL, Kelly E, Dewey W, LHenderson G: How important is protein kinase C in mu-opioid receptor desensitization and morphine tolerance?Trends Pharmacol Sci 2006, 27:558–65. 10.1016/j.tips.2006.09.006CrossRefPubMed Bailey CP, Smith FL, Kelly E, Dewey W, LHenderson G: How important is protein kinase C in mu-opioid receptor desensitization and morphine tolerance?Trends Pharmacol Sci 2006, 27:558–65. 10.1016/j.tips.2006.09.006CrossRefPubMed
13.
go back to reference Xu T, Jiang W, Du D, Xu Y, Hu Q, Shen Q: Role of spinal metabotropic glutamate receptor subtype 5 in the development of tolerance to morphine-induced antinociception in rat.Neurosci Lett 2007, 420:155–9. 10.1016/j.neulet.2007.04.065CrossRefPubMed Xu T, Jiang W, Du D, Xu Y, Hu Q, Shen Q: Role of spinal metabotropic glutamate receptor subtype 5 in the development of tolerance to morphine-induced antinociception in rat.Neurosci Lett 2007, 420:155–9. 10.1016/j.neulet.2007.04.065CrossRefPubMed
14.
go back to reference Xu T, Jiang W, Du D, Xu Y, Zhou Q, Pan X, et al.: Inhibition of MPEP on the development of morphine antinociceptive tolerance and the biosynthesis of neuronal nitric oxide synthase in rat spinal cord.Neurosci Lett 2008, 436:214–8. 10.1016/j.neulet.2008.03.028CrossRefPubMed Xu T, Jiang W, Du D, Xu Y, Zhou Q, Pan X, et al.: Inhibition of MPEP on the development of morphine antinociceptive tolerance and the biosynthesis of neuronal nitric oxide synthase in rat spinal cord.Neurosci Lett 2008, 436:214–8. 10.1016/j.neulet.2008.03.028CrossRefPubMed
15.
go back to reference Song L, Wang S, Zuo Y, Chen L, Martyn JA, Mao J: Midazolam exacerbates morphine tolerance and morphine-induced hyperactive behaviors in young rats with burn injury.Brain Res 2014, 1564:52–61.CrossRefPubMedPubMedCentral Song L, Wang S, Zuo Y, Chen L, Martyn JA, Mao J: Midazolam exacerbates morphine tolerance and morphine-induced hyperactive behaviors in young rats with burn injury.Brain Res 2014, 1564:52–61.CrossRefPubMedPubMedCentral
16.
go back to reference Martini L, Whistler JL: The role of mu opioid receptor desensitization and endocytosis in morphine tolerance and dependence.Curr Opin Neurobiol 2007, 17:556–64. 10.1016/j.conb.2007.10.004CrossRefPubMed Martini L, Whistler JL: The role of mu opioid receptor desensitization and endocytosis in morphine tolerance and dependence.Curr Opin Neurobiol 2007, 17:556–64. 10.1016/j.conb.2007.10.004CrossRefPubMed
17.
go back to reference Mao J, Price DD, Mayer DJ, Lu J, Hayes RL: Intrathecal MK-801 and local nerve anesthesia synergistically reduce nociceptive behaviors in rats with experimental peripheral mononeuropathy.Brain Res 1992, 576:254–62. 10.1016/0006-8993(92)90688-6CrossRefPubMed Mao J, Price DD, Mayer DJ, Lu J, Hayes RL: Intrathecal MK-801 and local nerve anesthesia synergistically reduce nociceptive behaviors in rats with experimental peripheral mononeuropathy.Brain Res 1992, 576:254–62. 10.1016/0006-8993(92)90688-6CrossRefPubMed
18.
go back to reference Manning BH, Mao J, Frenk H, Price DD, Mayer DJ: Continuous co-administration of dextromethorphan or MK-801 with morphine: attenuation of morphine dependence and naloxone-reversible attenuation of morphine tolerance.Pain 1996, 67:79–88. 10.1016/0304-3959(96)81972-5CrossRefPubMed Manning BH, Mao J, Frenk H, Price DD, Mayer DJ: Continuous co-administration of dextromethorphan or MK-801 with morphine: attenuation of morphine dependence and naloxone-reversible attenuation of morphine tolerance.Pain 1996, 67:79–88. 10.1016/0304-3959(96)81972-5CrossRefPubMed
19.
20.
go back to reference Blask DE, Sauer LA, Dauchy RT, Holowachuk EW, Ruhoff MS, Kopff HS: Melatonin inhibition of cancer growth in vivo involves suppression of tumor fatty acid metabolism via melatonin receptor-mediated signal transduction events.Cancer Res 1999, 59:4693–701.PubMed Blask DE, Sauer LA, Dauchy RT, Holowachuk EW, Ruhoff MS, Kopff HS: Melatonin inhibition of cancer growth in vivo involves suppression of tumor fatty acid metabolism via melatonin receptor-mediated signal transduction events.Cancer Res 1999, 59:4693–701.PubMed
21.
go back to reference Sauer LA, Dauchy RT, Blask DE: Polyunsaturated fatty acids, melatonin, and cancer prevention.Biochem Pharmacol 2001, 61:1455–62. 10.1016/S0006-2952(01)00634-7CrossRefPubMed Sauer LA, Dauchy RT, Blask DE: Polyunsaturated fatty acids, melatonin, and cancer prevention.Biochem Pharmacol 2001, 61:1455–62. 10.1016/S0006-2952(01)00634-7CrossRefPubMed
22.
go back to reference El-Shenawy SM, Abdel-Salam OM, Baiuomy AR, El-Batran S, Arbid MS: Studies on the anti-inflammatory and anti-nociceptive effects of melatonin in the rat.Pharmacol Res 2002, 46:235–43. 10.1016/S1043-6618(02)00094-4CrossRefPubMed El-Shenawy SM, Abdel-Salam OM, Baiuomy AR, El-Batran S, Arbid MS: Studies on the anti-inflammatory and anti-nociceptive effects of melatonin in the rat.Pharmacol Res 2002, 46:235–43. 10.1016/S1043-6618(02)00094-4CrossRefPubMed
23.
go back to reference Zahn PK, Lansmann T, Berger E, Speckmann EJ, Musshoff U: Gene expression and functional characterization of melatonin receptors in the spinal cord of the rat: implications for pain modulation.J Pineal Res 2003, 35:24–31. 10.1034/j.1600-079X.2003.00047.xCrossRefPubMed Zahn PK, Lansmann T, Berger E, Speckmann EJ, Musshoff U: Gene expression and functional characterization of melatonin receptors in the spinal cord of the rat: implications for pain modulation.J Pineal Res 2003, 35:24–31. 10.1034/j.1600-079X.2003.00047.xCrossRefPubMed
24.
go back to reference Wang S, Tian Y, Song L, Lim G, Tan Y, You Z, et al.: Exacerbated mechanical hyperalgesia in rats with genetically predisposed depressive behavior: role of melatonin and NMDA receptors.Pain 2012, 153:2448–57. 10.1016/j.pain.2012.08.016CrossRefPubMedPubMedCentral Wang S, Tian Y, Song L, Lim G, Tan Y, You Z, et al.: Exacerbated mechanical hyperalgesia in rats with genetically predisposed depressive behavior: role of melatonin and NMDA receptors.Pain 2012, 153:2448–57. 10.1016/j.pain.2012.08.016CrossRefPubMedPubMedCentral
25.
go back to reference Zurowski D, Nowak L, Machowska A, Wordliczek J, Thor PJ: Exogenous melatonin abolishes mechanical allodynia but not thermal hyperalgesia in neuropathic pain. The role of the opioid system and benzodiazepine-gabaergic mechanism.J Physiol Pharmacol 2012, 63:641–7.PubMed Zurowski D, Nowak L, Machowska A, Wordliczek J, Thor PJ: Exogenous melatonin abolishes mechanical allodynia but not thermal hyperalgesia in neuropathic pain. The role of the opioid system and benzodiazepine-gabaergic mechanism.J Physiol Pharmacol 2012, 63:641–7.PubMed
26.
go back to reference Raghavendra V, Kulkarni SK: Reversal of morphine tolerance and dependence by melatonin: possible role of central and peripheral benzodiazepine receptors.Brain Res 1999, 834:178–81. 10.1016/S0006-8993(99)01520-6CrossRefPubMed Raghavendra V, Kulkarni SK: Reversal of morphine tolerance and dependence by melatonin: possible role of central and peripheral benzodiazepine receptors.Brain Res 1999, 834:178–81. 10.1016/S0006-8993(99)01520-6CrossRefPubMed
27.
go back to reference Bennett GJ, Xie YK: A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man.Pain 1988, 33:87–107. 10.1016/0304-3959(88)90209-6CrossRefPubMed Bennett GJ, Xie YK: A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man.Pain 1988, 33:87–107. 10.1016/0304-3959(88)90209-6CrossRefPubMed
28.
go back to reference Hargreaves K, Dubner R, Brown F, Flores C, Joris J: A new and sensitive method for measuring thermal nociception in cutaneous hyperalgesia.Pain 1988, 32:77–88. 10.1016/0304-3959(88)90026-7CrossRefPubMed Hargreaves K, Dubner R, Brown F, Flores C, Joris J: A new and sensitive method for measuring thermal nociception in cutaneous hyperalgesia.Pain 1988, 32:77–88. 10.1016/0304-3959(88)90026-7CrossRefPubMed
29.
go back to reference Vanecek J: Cellular mechanisms of melatonin action.Physiol Rev 1998, 78:687–721.PubMed Vanecek J: Cellular mechanisms of melatonin action.Physiol Rev 1998, 78:687–721.PubMed
30.
go back to reference Raghavendra V, Kulkarni SK: Possible mechanisms of action in melatonin reversal of morphine tolerance and dependence in mice.Eur J Pharmacol 2000, 409:279–289. 10.1016/S0014-2999(00)00849-9CrossRefPubMed Raghavendra V, Kulkarni SK: Possible mechanisms of action in melatonin reversal of morphine tolerance and dependence in mice.Eur J Pharmacol 2000, 409:279–289. 10.1016/S0014-2999(00)00849-9CrossRefPubMed
31.
go back to reference Wang S, Zhang L, Lim G, Sung B, Tian Y, Chou CW, et al.: A combined effect of dextromethorphan and melatonin on neuropathic pain behavior in rats.Brain Res 2009, 1288:42–49.CrossRefPubMedPubMedCentral Wang S, Zhang L, Lim G, Sung B, Tian Y, Chou CW, et al.: A combined effect of dextromethorphan and melatonin on neuropathic pain behavior in rats.Brain Res 2009, 1288:42–49.CrossRefPubMedPubMedCentral
Metadata
Title
Melatonin prevents morphine-induced hyperalgesia and tolerance in rats: role of protein kinase C and N-methyl-D-aspartate receptors
Authors
Li Song
Chaoran Wu
Yunxia Zuo
Publication date
01-12-2015
Publisher
BioMed Central
Published in
BMC Anesthesiology / Issue 1/2015
Electronic ISSN: 1471-2253
DOI
https://doi.org/10.1186/1471-2253-15-12

Other articles of this Issue 1/2015

BMC Anesthesiology 1/2015 Go to the issue