Skip to main content
Top
Published in: Current Pain and Headache Reports 2/2010

01-04-2010

The Effect of Morphine on Glial Cells as a Potential Therapeutic Target for Pharmacological Development of Analgesic Drugs

Authors: Haroon Hameed, Mariam Hameed, Paul J. Christo

Published in: Current Pain and Headache Reports | Issue 2/2010

Login to get access

Abstract

Opioids have played a critical role in achieving pain relief in both modern and ancient medicine. Yet, their clinical use can be limited secondary to unwanted side effects such as tolerance, dependence, reward, and behavioral changes. Identification of glial-mediated mechanisms inducing opioid side effects include cytokine receptors, κ-opioid receptors, N-methyl-D-aspartate receptors, and the recently elucidated Toll-like receptors. Newer agents targeting these receptors such as AV411, MK-801, AV333, and SLC022, and older agents used outside the United States or for other disease conditions, such as minocycline, pentoxifylline, and UV50488H, all show varied but promising profiles for providing significant relief from opioid side effects, while simultaneously potentiating opioid analgesia.
Literature
1.
go back to reference •• Hutchinson MR, Bland ST, Johnson KW, et al.: Opioid-induced glial activation: mechanisms of activation and implications for opioid analgesia, dependence, and reward. ScientificWorldJournal 2007, 7:98–111. This article discusses TLRs and modifying agents and their relevance to the future of the management of pain. It also includes detailed information related to the research regarding naloxone in the management of opioid side effects. •• Hutchinson MR, Bland ST, Johnson KW, et al.: Opioid-induced glial activation: mechanisms of activation and implications for opioid analgesia, dependence, and reward. ScientificWorldJournal 2007, 7:98–111. This article discusses TLRs and modifying agents and their relevance to the future of the management of pain. It also includes detailed information related to the research regarding naloxone in the management of opioid side effects.
2.
go back to reference Coyle DE: Partial peripheral nerve injury leads to activation of astroglia and microglia which parallels the development of allodynic behavior. Glia 1998, 23:75–83.CrossRefPubMed Coyle DE: Partial peripheral nerve injury leads to activation of astroglia and microglia which parallels the development of allodynic behavior. Glia 1998, 23:75–83.CrossRefPubMed
3.
go back to reference McMahon SB (2002) Neuropathic pain mechanisms. In: Giamberardino MA (ed) Pain 2002—an updated review. Seattle: IASP Press, 155–164. McMahon SB (2002) Neuropathic pain mechanisms. In: Giamberardino MA (ed) Pain 2002—an updated review. Seattle: IASP Press, 155–164.
4.
go back to reference Nakajima K, Kohsaka S: Functional roles of microglia in the brain. Neurosci Res 1993, 17:187–203.CrossRefPubMed Nakajima K, Kohsaka S: Functional roles of microglia in the brain. Neurosci Res 1993, 17:187–203.CrossRefPubMed
5.
go back to reference Milligan ED, Watkins LR: Pathological and protective roles of glia in chronic pain. Nat Rev Neurosci 2009, 10:23–36.CrossRefPubMed Milligan ED, Watkins LR: Pathological and protective roles of glia in chronic pain. Nat Rev Neurosci 2009, 10:23–36.CrossRefPubMed
6.
go back to reference Halassa MM, Fellin T, Haydon PG: The tripartite synapse: roles for gliotransmission in health and disease. Trends Mol Med 2007, 13:54–63.CrossRefPubMed Halassa MM, Fellin T, Haydon PG: The tripartite synapse: roles for gliotransmission in health and disease. Trends Mol Med 2007, 13:54–63.CrossRefPubMed
7.
go back to reference Pocock JM, Kettenmann H: Neurotransmitter receptors on microglia. Trends Neurosci 2007, 30:527–535.CrossRefPubMed Pocock JM, Kettenmann H: Neurotransmitter receptors on microglia. Trends Neurosci 2007, 30:527–535.CrossRefPubMed
8.
go back to reference Cao H, Zhang YQ: Spinal glial activation contributes to pathological pain states. Neurosci Biobehav Rev 2008, 32:972–983.CrossRefPubMed Cao H, Zhang YQ: Spinal glial activation contributes to pathological pain states. Neurosci Biobehav Rev 2008, 32:972–983.CrossRefPubMed
9.
go back to reference Ji RR, Gereau RW 4th, Malcangio M, Strichartz GR: MAP kinase and pain. Brain Res Rev 2009, 60:135–148.CrossRefPubMed Ji RR, Gereau RW 4th, Malcangio M, Strichartz GR: MAP kinase and pain. Brain Res Rev 2009, 60:135–148.CrossRefPubMed
10.
go back to reference Watkins LR, Maier SF: The pain of being sick: implications of immune-to-brain communication for understanding pain. Annu Rev Psychol 2000, 51:29–57.CrossRefPubMed Watkins LR, Maier SF: The pain of being sick: implications of immune-to-brain communication for understanding pain. Annu Rev Psychol 2000, 51:29–57.CrossRefPubMed
11.
go back to reference Minghetti L, Levi G: Microglia as effector cells in brain damage and repair: focus on prostanoids and nitric oxide. Prog Neurobiol 1998, 54:99–125.CrossRefPubMed Minghetti L, Levi G: Microglia as effector cells in brain damage and repair: focus on prostanoids and nitric oxide. Prog Neurobiol 1998, 54:99–125.CrossRefPubMed
12.
go back to reference Hutchinson MR, Coats BD, Lewis SS, et al.: Proinflammatory cytokines oppose opioid-induced acute and chronic analgesia. Brain Behav Immun 2008, 22:1178–1189.CrossRefPubMed Hutchinson MR, Coats BD, Lewis SS, et al.: Proinflammatory cytokines oppose opioid-induced acute and chronic analgesia. Brain Behav Immun 2008, 22:1178–1189.CrossRefPubMed
13.
go back to reference Malcangio M, Bowery NG, Flower RJ, Perretti M: Effect of interleukin-1 on the release of substance P from rat isolated spinal cord. Eur J Pharmacol 1996, 299:113–118.CrossRefPubMed Malcangio M, Bowery NG, Flower RJ, Perretti M: Effect of interleukin-1 on the release of substance P from rat isolated spinal cord. Eur J Pharmacol 1996, 299:113–118.CrossRefPubMed
14.
go back to reference •• Mika J: Modulation of microglia can attenuate neuropathic pain symptoms and enhance morphine effectiveness. Pharmacol Rep 2008, 60:297–307. This review discusses the roles of various cytokine-related cascades in addition to other TLR-related mechanisms in the generation of nonopioid receptor-mediated opioid effects and the agents that may be employed to modify them. •• Mika J: Modulation of microglia can attenuate neuropathic pain symptoms and enhance morphine effectiveness. Pharmacol Rep 2008, 60:297–307. This review discusses the roles of various cytokine-related cascades in addition to other TLR-related mechanisms in the generation of nonopioid receptor-mediated opioid effects and the agents that may be employed to modify them.
15.
go back to reference Obreja O, Rathee PK, Lips KS, et al.: IL-1 potentiates heat-activated currents in rat sensory neurons: involvement of IL-1RI, tyrosine kinase, and protein kinase C. FASEB J 2002, 16:1497–1503.CrossRefPubMed Obreja O, Rathee PK, Lips KS, et al.: IL-1 potentiates heat-activated currents in rat sensory neurons: involvement of IL-1RI, tyrosine kinase, and protein kinase C. FASEB J 2002, 16:1497–1503.CrossRefPubMed
16.
go back to reference Flatters SJ, Fox AJ, Dickenson AH: Spinal interleukin-6 (IL-6) inhibits nociceptive transmission following neuropathy. Brain Res 2003, 984:54–62.CrossRefPubMed Flatters SJ, Fox AJ, Dickenson AH: Spinal interleukin-6 (IL-6) inhibits nociceptive transmission following neuropathy. Brain Res 2003, 984:54–62.CrossRefPubMed
17.
go back to reference Moore KW, O’Garra A, de Waal Malefyt R, et al.: Interleukin-10. Annu Rev Immunol 1993, 11:165–190CrossRefPubMed Moore KW, O’Garra A, de Waal Malefyt R, et al.: Interleukin-10. Annu Rev Immunol 1993, 11:165–190CrossRefPubMed
19.
go back to reference Sawada M, Suzumura A, Hosoya H, et al.: Interleukin-10 inhibits both production of cytokines and expression of cytokine receptors in microglia. J Neurochem 1999, 72:1466–1471.CrossRefPubMed Sawada M, Suzumura A, Hosoya H, et al.: Interleukin-10 inhibits both production of cytokines and expression of cytokine receptors in microglia. J Neurochem 1999, 72:1466–1471.CrossRefPubMed
20.
go back to reference Huber TS, Gaines GC, Welborn MB 3rd, et al.: Anticytokine therapies for acute inflammation and the systemic inflammatory response syndrome: IL-10 and ischemia/reperfusion injury as a new paradigm. Shock 2000, 13:425–434.PubMed Huber TS, Gaines GC, Welborn MB 3rd, et al.: Anticytokine therapies for acute inflammation and the systemic inflammatory response syndrome: IL-10 and ischemia/reperfusion injury as a new paradigm. Shock 2000, 13:425–434.PubMed
21.
go back to reference Merrill JE, Benveniste EN: Cytokines in inflammatory brain lesions: helpful and harmful. Trends Neurosci 1996, 19:331–338.CrossRefPubMed Merrill JE, Benveniste EN: Cytokines in inflammatory brain lesions: helpful and harmful. Trends Neurosci 1996, 19:331–338.CrossRefPubMed
22.
go back to reference Fontaine V, Mohand-Said S, Hanoteau N, et al.: Neurodegenerative and neuroprotective effects of tumor necrosis factor (TNF) in retinal ischemia: opposite roles of TNF receptor 1 and TNF receptor 2. J Neurosci 2002, 22:RC216.PubMed Fontaine V, Mohand-Said S, Hanoteau N, et al.: Neurodegenerative and neuroprotective effects of tumor necrosis factor (TNF) in retinal ischemia: opposite roles of TNF receptor 1 and TNF receptor 2. J Neurosci 2002, 22:RC216.PubMed
23.
go back to reference Tanga FY, Nutile-McMenemy N, Deleo JA: The CNS role of Toll-like receptor 4 in innate neuroimmunity and painful neuropathy. Proc Natl Acad Sci U S A 2005, 102: 5856–5861.CrossRefPubMed Tanga FY, Nutile-McMenemy N, Deleo JA: The CNS role of Toll-like receptor 4 in innate neuroimmunity and painful neuropathy. Proc Natl Acad Sci U S A 2005, 102: 5856–5861.CrossRefPubMed
24.
go back to reference Kim D, Kim MA, Cho IH, et al.: A critical role of toll-like receptor 2 in nerve injury-induced spinal cord glial cell activation and pain hypersensitivity. J Biol Chem 2007, 282:14975–14983.CrossRefPubMed Kim D, Kim MA, Cho IH, et al.: A critical role of toll-like receptor 2 in nerve injury-induced spinal cord glial cell activation and pain hypersensitivity. J Biol Chem 2007, 282:14975–14983.CrossRefPubMed
25.
go back to reference Muzio M, Mantovani A: Toll-like receptors (TLRs) signalling and expression pattern. J Endotoxin Res 2001, 7:297–300.PubMed Muzio M, Mantovani A: Toll-like receptors (TLRs) signalling and expression pattern. J Endotoxin Res 2001, 7:297–300.PubMed
26.
go back to reference •• Guo LH, Schluesener HJ: The innate immunity of the central nervous system in chronic pain: the role of Toll-like receptors. Cell Mol Life Sci 2007, 64:1128–1136. This article offers a detailed discussion on TLRs and their role in the development of various types of pain. •• Guo LH, Schluesener HJ: The innate immunity of the central nervous system in chronic pain: the role of Toll-like receptors. Cell Mol Life Sci 2007, 64:1128–1136. This article offers a detailed discussion on TLRs and their role in the development of various types of pain.
28.
go back to reference •• Watkins LR, Hutchinson MR, Rice KC, Maier SF: The “toll” of opioid-induced glial activation: improving the clinical efficacy of opioids by targeting glia. Trends Pharmacol Sci 2009, 30:581–591. This is a very recent review of TLR receptors and the current foci of interest in the management of dependence, reward, and other opioid-related side effects. •• Watkins LR, Hutchinson MR, Rice KC, Maier SF: The “toll” of opioid-induced glial activation: improving the clinical efficacy of opioids by targeting glia. Trends Pharmacol Sci 2009, 30:581–591. This is a very recent review of TLR receptors and the current foci of interest in the management of dependence, reward, and other opioid-related side effects.
29.
go back to reference Kakimura J, Kitamura Y, Takata K, et al.: Microglial activation and amyloid-beta clearance induced by exogenous heat-shock proteins. FASEB J 2002, 16:601–603.PubMed Kakimura J, Kitamura Y, Takata K, et al.: Microglial activation and amyloid-beta clearance induced by exogenous heat-shock proteins. FASEB J 2002, 16:601–603.PubMed
30.
go back to reference Tsuda M, Shigemoto-Mogami Y, Koizumi S, et al.: P2X4 receptors induced in spinal microglia gate tactile allodynia after nerve injury. Nature 2003, 424:778–783.CrossRefPubMed Tsuda M, Shigemoto-Mogami Y, Koizumi S, et al.: P2X4 receptors induced in spinal microglia gate tactile allodynia after nerve injury. Nature 2003, 424:778–783.CrossRefPubMed
31.
go back to reference Horvath RJ, DeLeo JA: Morphine enhances microglial migration through modulation of P2X4 receptor signaling. J Neurosci 2009, 29:998–1005.CrossRefPubMed Horvath RJ, DeLeo JA: Morphine enhances microglial migration through modulation of P2X4 receptor signaling. J Neurosci 2009, 29:998–1005.CrossRefPubMed
32.
go back to reference McMahon SB, Cafferty WB, Marchand F: Immune and glial cell factors as pain mediators and modulators. Exp Neurol 2005, 192:444–462.CrossRefPubMed McMahon SB, Cafferty WB, Marchand F: Immune and glial cell factors as pain mediators and modulators. Exp Neurol 2005, 192:444–462.CrossRefPubMed
33.
go back to reference •• Hutchinson MR, Zhang Y, Shridhar M, et al.: Evidence that opioids may have toll-like receptor 4 and MD-2 effects. Brain Behav Immun 2010, 24:83–95. This paper summarizes recent information regarding the highly central role of TLR-4 receptors with implications to the current developments in the opioid-related tolerance and dependence modifying agents. •• Hutchinson MR, Zhang Y, Shridhar M, et al.: Evidence that opioids may have toll-like receptor 4 and MD-2 effects. Brain Behav Immun 2010, 24:83–95. This paper summarizes recent information regarding the highly central role of TLR-4 receptors with implications to the current developments in the opioid-related tolerance and dependence modifying agents.
34.
go back to reference Juni A, Klein G, Pintar JE, Kest B: Nociception increases during opioid infusion in opioid receptor triple knock-out mice. Neuroscience 2007, 147:439–444.CrossRefPubMed Juni A, Klein G, Pintar JE, Kest B: Nociception increases during opioid infusion in opioid receptor triple knock-out mice. Neuroscience 2007, 147:439–444.CrossRefPubMed
35.
go back to reference Shu H, Hayashida M, Huang W, et al.: The comparison of effects of processed Aconiti tuber, U50488H and MK-801 on the antinociceptive tolerance to morphine. J Ethnopharmacol 2008, 117:158–165.CrossRefPubMed Shu H, Hayashida M, Huang W, et al.: The comparison of effects of processed Aconiti tuber, U50488H and MK-801 on the antinociceptive tolerance to morphine. J Ethnopharmacol 2008, 117:158–165.CrossRefPubMed
36.
go back to reference •• Ueda H, Ueda M: Mechanisms underlying morphine analgesic tolerance and dependence. Front Biosci 2009, 14:5260–5272. This is a highly detailed review of the protein pathways responsible for the development of opioid tolerance and dependence. This review includes discussions on protein kinase receptor phosphorylations, NMDA receptors, neurotrophins, and various other neuropeptides. •• Ueda H, Ueda M: Mechanisms underlying morphine analgesic tolerance and dependence. Front Biosci 2009, 14:5260–5272. This is a highly detailed review of the protein pathways responsible for the development of opioid tolerance and dependence. This review includes discussions on protein kinase receptor phosphorylations, NMDA receptors, neurotrophins, and various other neuropeptides.
37.
go back to reference Xu M, Bruchas MR, Ippolito DL, et al.: Sciatic nerve ligation-induced proliferation of spinal cord astrocytes is mediated by kappa opioid activation of p38 mitogen-activated protein kinase. J Neurosci 2007, 27:2570–2581.CrossRefPubMed Xu M, Bruchas MR, Ippolito DL, et al.: Sciatic nerve ligation-induced proliferation of spinal cord astrocytes is mediated by kappa opioid activation of p38 mitogen-activated protein kinase. J Neurosci 2007, 27:2570–2581.CrossRefPubMed
38.
go back to reference • Ledeboer A, Hutchinson MR, Watkins LR, Johnson KW: Ibudilast (AV-411). A new class therapeutic candidate for neuropathic pain and opioid withdrawal syndromes. Expert Opin Investig Drugs 2007, 16:935–950. This review details the effectiveness of ibudilast in the management of opioid tolerance and reward. • Ledeboer A, Hutchinson MR, Watkins LR, Johnson KW: Ibudilast (AV-411). A new class therapeutic candidate for neuropathic pain and opioid withdrawal syndromes. Expert Opin Investig Drugs 2007, 16:935–950. This review details the effectiveness of ibudilast in the management of opioid tolerance and reward.
39.
go back to reference Kishi Y, Ohta S, Kasuya N, et al.: Ibudilast: a non-selective PDE inhibitor with multiple actions on blood cells and the vascular wall. Cardiovasc Drug Rev 2001, 19:215–225.PubMedCrossRef Kishi Y, Ohta S, Kasuya N, et al.: Ibudilast: a non-selective PDE inhibitor with multiple actions on blood cells and the vascular wall. Cardiovasc Drug Rev 2001, 19:215–225.PubMedCrossRef
40.
go back to reference Fujimoto T, Sakoda S, Fujimura H, Yanagihara T: Ibudilast, a phosphodiesterase inhibitor, ameliorates experimental autoimmune encephalomyelitis in Dark August rats. J Neuroimmunol 1999, 95:35–42.CrossRefPubMed Fujimoto T, Sakoda S, Fujimura H, Yanagihara T: Ibudilast, a phosphodiesterase inhibitor, ameliorates experimental autoimmune encephalomyelitis in Dark August rats. J Neuroimmunol 1999, 95:35–42.CrossRefPubMed
41.
go back to reference Feng J, Misu T, Fujihara K, et al.: Ibudilast, a nonselective phosphodiesterase inhibitor, regulates Th1/Th2 balance and NKT cell subset in multiple sclerosis. Mult Scler 2004, 10:494–498.CrossRefPubMed Feng J, Misu T, Fujihara K, et al.: Ibudilast, a nonselective phosphodiesterase inhibitor, regulates Th1/Th2 balance and NKT cell subset in multiple sclerosis. Mult Scler 2004, 10:494–498.CrossRefPubMed
42.
go back to reference Harris GC, Aston-Jones G: Involvement of D2 dopamine receptors in the nucleus accumbens in the opiate withdrawal syndrome. Nature 1994, 371:155–157.CrossRefPubMed Harris GC, Aston-Jones G: Involvement of D2 dopamine receptors in the nucleus accumbens in the opiate withdrawal syndrome. Nature 1994, 371:155–157.CrossRefPubMed
43.
go back to reference • Bland ST, Hutchinson MR, Maier SF, et al.: The glial activation inhibitor AV411 reduces morphine-induced nucleus accumbens dopamine release. Brain Behav Immun 2009, 23:492–497. This review discusses the mechanism of action of AV411 and its relationship to TLRs. • Bland ST, Hutchinson MR, Maier SF, et al.: The glial activation inhibitor AV411 reduces morphine-induced nucleus accumbens dopamine release. Brain Behav Immun 2009, 23:492–497. This review discusses the mechanism of action of AV411 and its relationship to TLRs.
44.
go back to reference Kest B, Mogil JS, Shamgar BE, et al.: The NMDA receptor antagonist MK-801 protects against the development of morphine tolerance after intrathecal administration. Proc West Pharmacol Soc 1993, 36:307–310.PubMed Kest B, Mogil JS, Shamgar BE, et al.: The NMDA receptor antagonist MK-801 protects against the development of morphine tolerance after intrathecal administration. Proc West Pharmacol Soc 1993, 36:307–310.PubMed
45.
go back to reference Guo RX, Zhang M, Liu W, et al.: NMDA receptors are involved in upstream of the spinal JNK activation in morphine antinociceptive tolerance. Neurosci Lett 2009, 467:95–99.CrossRefPubMed Guo RX, Zhang M, Liu W, et al.: NMDA receptors are involved in upstream of the spinal JNK activation in morphine antinociceptive tolerance. Neurosci Lett 2009, 467:95–99.CrossRefPubMed
46.
go back to reference Morgan MM, Bobeck EN, Ingram SL: Glutamate modulation of antinociception, but not tolerance, produced by morphine microinjection into the periaqueductal gray of the rat. Brain Res 2009, 1295:59–66.CrossRefPubMed Morgan MM, Bobeck EN, Ingram SL: Glutamate modulation of antinociception, but not tolerance, produced by morphine microinjection into the periaqueductal gray of the rat. Brain Res 2009, 1295:59–66.CrossRefPubMed
47.
go back to reference Schmidt AP, Tort AB, Silveira PP, et al.: The NMDA antagonist MK-801 induces hyperalgesia and increases CSF excitatory amino acids in rats: reversal by guanosine. Pharmacol Biochem Behav 2009, 91:549–553.CrossRefPubMed Schmidt AP, Tort AB, Silveira PP, et al.: The NMDA antagonist MK-801 induces hyperalgesia and increases CSF excitatory amino acids in rats: reversal by guanosine. Pharmacol Biochem Behav 2009, 91:549–553.CrossRefPubMed
48.
go back to reference Sweitzer SM, Pahl JL, DeLeo JA: Propentofylline attenuates vincristine-induced peripheral neuropathy in the rat. Neurosci Lett 2006, 400:258–261.CrossRefPubMed Sweitzer SM, Pahl JL, DeLeo JA: Propentofylline attenuates vincristine-induced peripheral neuropathy in the rat. Neurosci Lett 2006, 400:258–261.CrossRefPubMed
49.
go back to reference Gwak YS, Crown ED, Unabia GC, Hulsebosch CE: Propentofylline attenuates allodynia, glial activation and modulates GABAergic tone after spinal cord injury in the rat. Pain 2008, 138:410–422.CrossRefPubMed Gwak YS, Crown ED, Unabia GC, Hulsebosch CE: Propentofylline attenuates allodynia, glial activation and modulates GABAergic tone after spinal cord injury in the rat. Pain 2008, 138:410–422.CrossRefPubMed
50.
go back to reference Narita M, Miyatake M, Narita M, et al.: Direct evidence of astrocytic modulation in the development of rewarding effects induced by drugs of abuse. Neuropsychopharmacology 2006, 31:2476–2488.CrossRefPubMed Narita M, Miyatake M, Narita M, et al.: Direct evidence of astrocytic modulation in the development of rewarding effects induced by drugs of abuse. Neuropsychopharmacology 2006, 31:2476–2488.CrossRefPubMed
51.
go back to reference Mika J, Wawrzczak-Bargiela A, Osikowicz M, et al.: Attenuation of morphine tolerance by minocycline and pentoxifylline in naive and neuropathic mice. Brain Behav Immun 2009, 23:75–84.CrossRefPubMed Mika J, Wawrzczak-Bargiela A, Osikowicz M, et al.: Attenuation of morphine tolerance by minocycline and pentoxifylline in naive and neuropathic mice. Brain Behav Immun 2009, 23:75–84.CrossRefPubMed
52.
53.
go back to reference Li WW, Setzu A, Zhao C, Franklin RJ: Minocycline-mediated inhibition of microglia activation impairs oligodendrocyte progenitor cell responses and remyelination in a non-immune model of demyelination. J Neuroimmunol 2005, 158:58–66.CrossRefPubMed Li WW, Setzu A, Zhao C, Franklin RJ: Minocycline-mediated inhibition of microglia activation impairs oligodendrocyte progenitor cell responses and remyelination in a non-immune model of demyelination. J Neuroimmunol 2005, 158:58–66.CrossRefPubMed
54.
go back to reference Ledeboer A, Sloane EM, Milligan ED, et al.: Minocycline attenuates mechanical allodynia and proinflammatory cytokine expression in rat models of pain facilitation. Pain 2005, 115:71–83.CrossRefPubMed Ledeboer A, Sloane EM, Milligan ED, et al.: Minocycline attenuates mechanical allodynia and proinflammatory cytokine expression in rat models of pain facilitation. Pain 2005, 115:71–83.CrossRefPubMed
55.
go back to reference Zanjani TM, Sabetkasaei M, Mosaffa N, et al.: Suppression of interleukin-6 by minocycline in a rat model of neuropathic pain. Eur J Pharmacol 2006, 538:66–72.CrossRefPubMed Zanjani TM, Sabetkasaei M, Mosaffa N, et al.: Suppression of interleukin-6 by minocycline in a rat model of neuropathic pain. Eur J Pharmacol 2006, 538:66–72.CrossRefPubMed
56.
go back to reference Piao ZG, Cho IH, Park CK, et al.: Activation of glia and microglial p38 MAPK in medullary dorsal horn contributes to tactile hypersensitivity following trigeminal sensory nerve injury. Pain 2006, 121:219–231.CrossRefPubMed Piao ZG, Cho IH, Park CK, et al.: Activation of glia and microglial p38 MAPK in medullary dorsal horn contributes to tactile hypersensitivity following trigeminal sensory nerve injury. Pain 2006, 121:219–231.CrossRefPubMed
57.
go back to reference Cui Y, Liao XX, Liu W, et al.: A novel role of minocycline: attenuating morphine antinociceptive tolerance by inhibition of p38 MAPK in the activated spinal microglia. Brain Behav Immun 2008, 22:114–123.CrossRefPubMed Cui Y, Liao XX, Liu W, et al.: A novel role of minocycline: attenuating morphine antinociceptive tolerance by inhibition of p38 MAPK in the activated spinal microglia. Brain Behav Immun 2008, 22:114–123.CrossRefPubMed
58.
go back to reference Wei T, Sabsovich I, Guo TZ, et al.: Pentoxifylline attenuates nociceptive sensitization and cytokine expression in a tibia fracture rat model of complex regional pain syndrome. Eur J Pain 2009, 13:253–262.CrossRefPubMed Wei T, Sabsovich I, Guo TZ, et al.: Pentoxifylline attenuates nociceptive sensitization and cytokine expression in a tibia fracture rat model of complex regional pain syndrome. Eur J Pain 2009, 13:253–262.CrossRefPubMed
59.
go back to reference Dorazil-Dudzik M, Mika J, Schafer MK, et al.: The effects of local pentoxifylline and propentofylline treatment on formalin-induced pain and tumor necrosis factor-alpha messenger RNA levels in the inflamed tissue of the rat paw. Anesth Analg 2004, 98:1566–1573.CrossRefPubMed Dorazil-Dudzik M, Mika J, Schafer MK, et al.: The effects of local pentoxifylline and propentofylline treatment on formalin-induced pain and tumor necrosis factor-alpha messenger RNA levels in the inflamed tissue of the rat paw. Anesth Analg 2004, 98:1566–1573.CrossRefPubMed
60.
go back to reference Liu J, Feng X, Yu M, et al.: Pentoxifylline attenuates the development of hyperalgesia in a rat model of neuropathic pain. Neurosci Lett 2007, 412:268–272.CrossRefPubMed Liu J, Feng X, Yu M, et al.: Pentoxifylline attenuates the development of hyperalgesia in a rat model of neuropathic pain. Neurosci Lett 2007, 412:268–272.CrossRefPubMed
Metadata
Title
The Effect of Morphine on Glial Cells as a Potential Therapeutic Target for Pharmacological Development of Analgesic Drugs
Authors
Haroon Hameed
Mariam Hameed
Paul J. Christo
Publication date
01-04-2010
Publisher
Current Science Inc.
Published in
Current Pain and Headache Reports / Issue 2/2010
Print ISSN: 1531-3433
Electronic ISSN: 1534-3081
DOI
https://doi.org/10.1007/s11916-010-0093-y

Other articles of this Issue 2/2010

Current Pain and Headache Reports 2/2010 Go to the issue