Skip to main content
Top
Published in: Molecular Pain 1/2012

Open Access 01-12-2012 | Research

Acute morphine activates satellite glial cells and up-regulates IL-1β in dorsal root ganglia in mice via matrix metalloprotease-9

Authors: Temugin Berta, Tong Liu, Yen-Chin Liu, Zhen-Zhong Xu, Ru-Rong Ji

Published in: Molecular Pain | Issue 1/2012

Login to get access

Abstract

Background

Activation of spinal cord glial cells such as microglia and astrocytes has been shown to regulate chronic opioid-induced antinociceptive tolerance and hyperalgesia, due to spinal up-regulation of the proinflammatory cytokines such as interleukin-1 beta (IL-1β). Matrix metalloprotease-9 (MMP-9) has been implicated in IL-1β activation in neuropathic pain. However, it is unclear whether acute opioid treatment can activate glial cells in the peripheral nervous system. We examined acute morphine-induced activation of satellite glial cells (SGCs) and up-regulation of IL-1β in dorsal root ganglia (DRGs), and further investigated the involvement of MMP-9 in these opioid-induced peripheral changes.

Results

Subcutaneous morphine injection (10 mg/kg) induced robust peripheral glial responses, as evidenced by increased GFAP expression in DRGs but not in spinal cords. The acute morphine-induced GFAP expression is transient, peaking at 2 h and declining after 3 h. Acute morphine treatment also increased IL-1β immunoreactivity in SGCs and IL-1β activation in DRGs. MMP-9 and GFAP are expressed in DRG neurons and SGCs, respectively. Confocal analysis revealed a close proximity of MMP-9 and GFAP immunostaining. Importantly, morphine-induced DRG up-regulation of GFAP expression and IL-1β activation was abolished after Mmp9 deletion or naloxone pre-treatment. Finally, intrathecal injections of IL-1β-selective siRNA not only reduced DRG IL-1β expression but also prolonged acute morphine-induced analgesia.

Conclusions

Acute morphine induces opioid receptors- and MMP-9-dependent up-regulation of GFAP expression and IL-1β activation in SGCs of DRGs. MMP-9 could mask and shorten morphine analgesia via peripheral neuron-glial interactions. Targeting peripheral glial activation might prolong acute opioid analgesia.
Appendix
Available only for authorised users
Literature
1.
go back to reference Tsuda M, Kuboyama K, Inoue T, Nagata K, Tozaki-Saitoh H, Inoue K: Behavioral phenotypes of mice lacking purinergic P2X4 receptors in acute and chronic pain assays. Mol Pain 2009, 5: 28. 10.1186/1744-8069-5-28PubMedCentralCrossRefPubMed Tsuda M, Kuboyama K, Inoue T, Nagata K, Tozaki-Saitoh H, Inoue K: Behavioral phenotypes of mice lacking purinergic P2X4 receptors in acute and chronic pain assays. Mol Pain 2009, 5: 28. 10.1186/1744-8069-5-28PubMedCentralCrossRefPubMed
2.
5.
go back to reference McMahon SB, Malcangio M: Current challenges in glia-pain biology. Neuron 2009, 64: 46–54. 10.1016/j.neuron.2009.09.033CrossRefPubMed McMahon SB, Malcangio M: Current challenges in glia-pain biology. Neuron 2009, 64: 46–54. 10.1016/j.neuron.2009.09.033CrossRefPubMed
7.
go back to reference Katsura H: Activation of Src-family kinases in spinal microglia contributes to mechanical hypersensitivity after nerve injury. 2006. Katsura H: Activation of Src-family kinases in spinal microglia contributes to mechanical hypersensitivity after nerve injury. 2006.
8.
go back to reference Song P, Zhao ZQ: The involvement of glial cells in the development of morphine tolerance. Neurosci Res 2001, 39: 281–286. 10.1016/S0168-0102(00)00226-1CrossRefPubMed Song P, Zhao ZQ: The involvement of glial cells in the development of morphine tolerance. Neurosci Res 2001, 39: 281–286. 10.1016/S0168-0102(00)00226-1CrossRefPubMed
9.
go back to reference Raghavendra V, Rutkowski MD, DeLeo JA: The role of spinal neuroimmune activation in morphine tolerance/hyperalgesia in neuropathic and sham-operated rats. J Neurosci 2002, 22: 9980–9989.PubMed Raghavendra V, Rutkowski MD, DeLeo JA: The role of spinal neuroimmune activation in morphine tolerance/hyperalgesia in neuropathic and sham-operated rats. J Neurosci 2002, 22: 9980–9989.PubMed
10.
go back to reference Wen YR, Tan PH, Cheng JK, Liu YC, Ji RR: Microglia: a promising target for treating neuropathic and postoperative pain, and morphine tolerance. J Formos Med Assoc 2011, 110: 487–494. 10.1016/S0929-6646(11)60074-0PubMedCentralCrossRefPubMed Wen YR, Tan PH, Cheng JK, Liu YC, Ji RR: Microglia: a promising target for treating neuropathic and postoperative pain, and morphine tolerance. J Formos Med Assoc 2011, 110: 487–494. 10.1016/S0929-6646(11)60074-0PubMedCentralCrossRefPubMed
11.
go back to reference Johnston IN, Milligan ED, Wieseler-Frank J, Frank MG, Zapata V, Campisi J, et al.: A role for proinflammatory cytokines and fractalkine in analgesia, tolerance, and subsequent pain facilitation induced by chronic intrathecal morphine. J Neurosci 2004, 24: 7353–7365. 10.1523/JNEUROSCI.1850-04.2004CrossRefPubMed Johnston IN, Milligan ED, Wieseler-Frank J, Frank MG, Zapata V, Campisi J, et al.: A role for proinflammatory cytokines and fractalkine in analgesia, tolerance, and subsequent pain facilitation induced by chronic intrathecal morphine. J Neurosci 2004, 24: 7353–7365. 10.1523/JNEUROSCI.1850-04.2004CrossRefPubMed
12.
go back to reference Watkins LR, Hutchinson MR, Johnston IN, Maier SF: Glia: novel counter-regulators of opioid analgesia. Trends Neurosci 2005, 28: 661–669. 10.1016/j.tins.2005.10.001CrossRefPubMed Watkins LR, Hutchinson MR, Johnston IN, Maier SF: Glia: novel counter-regulators of opioid analgesia. Trends Neurosci 2005, 28: 661–669. 10.1016/j.tins.2005.10.001CrossRefPubMed
13.
go back to reference DeLeo JA, Tanga FY, Tawfik VL: Neuroimmune activation and neuroinflammation in chronic pain and opioid tolerance/hyperalgesia. Neuroscientist 2004, 10: 40–52. 10.1177/1073858403259950CrossRefPubMed DeLeo JA, Tanga FY, Tawfik VL: Neuroimmune activation and neuroinflammation in chronic pain and opioid tolerance/hyperalgesia. Neuroscientist 2004, 10: 40–52. 10.1177/1073858403259950CrossRefPubMed
14.
15.
go back to reference Kawasaki Y, Zhang L, Cheng JK, Ji RR: Cytokine mechanisms of central sensitization: distinct and overlapping role of interleukin-1beta, interleukin-6, and tumor necrosis factor-alpha in regulating synaptic and neuronal activity in the superficial spinal cord. J Neurosci 2008, 28: 5189–5194. 10.1523/JNEUROSCI.3338-07.2008PubMedCentralCrossRefPubMed Kawasaki Y, Zhang L, Cheng JK, Ji RR: Cytokine mechanisms of central sensitization: distinct and overlapping role of interleukin-1beta, interleukin-6, and tumor necrosis factor-alpha in regulating synaptic and neuronal activity in the superficial spinal cord. J Neurosci 2008, 28: 5189–5194. 10.1523/JNEUROSCI.3338-07.2008PubMedCentralCrossRefPubMed
16.
go back to reference Li J, Xie W, Zhang JM, Baccei ML: Peripheral nerve injury sensitizes neonatal dorsal horn neurons to tumor necrosis factor-alpha. Mol Pain 2009, 5: 10. 10.1186/1744-8069-5-10PubMedCentralCrossRefPubMed Li J, Xie W, Zhang JM, Baccei ML: Peripheral nerve injury sensitizes neonatal dorsal horn neurons to tumor necrosis factor-alpha. Mol Pain 2009, 5: 10. 10.1186/1744-8069-5-10PubMedCentralCrossRefPubMed
17.
go back to reference Zhang L, Berta T, Xu ZZ, Liu T, Park JY, Ji RR: TNF-alpha contributes to spinal cord synaptic plasticity and inflammatory pain: distinct role of TNF receptor subtypes 1 and 2. Pain 2011, 152: 419–427. 10.1016/j.pain.2010.11.014PubMedCentralCrossRefPubMed Zhang L, Berta T, Xu ZZ, Liu T, Park JY, Ji RR: TNF-alpha contributes to spinal cord synaptic plasticity and inflammatory pain: distinct role of TNF receptor subtypes 1 and 2. Pain 2011, 152: 419–427. 10.1016/j.pain.2010.11.014PubMedCentralCrossRefPubMed
18.
go back to reference Hutchinson MR, Coats BD, Lewis SS, Zhang Y, Sprunger DB, Rezvani N, et al.: Proinflammatory cytokines oppose opioid-induced acute and chronic analgesia. Brain Behav Immun 2008, 22: 1178–1189. 10.1016/j.bbi.2008.05.004PubMedCentralCrossRefPubMed Hutchinson MR, Coats BD, Lewis SS, Zhang Y, Sprunger DB, Rezvani N, et al.: Proinflammatory cytokines oppose opioid-induced acute and chronic analgesia. Brain Behav Immun 2008, 22: 1178–1189. 10.1016/j.bbi.2008.05.004PubMedCentralCrossRefPubMed
19.
go back to reference Bessler H, Shavit Y, Mayburd E, Smirnov G, Beilin B: Postoperative pain, morphine consumption, and genetic polymorphism of IL-1beta and IL-1 receptor antagonist. Neurosci Lett 2006, 404: 154–158. 10.1016/j.neulet.2006.05.030CrossRefPubMed Bessler H, Shavit Y, Mayburd E, Smirnov G, Beilin B: Postoperative pain, morphine consumption, and genetic polymorphism of IL-1beta and IL-1 receptor antagonist. Neurosci Lett 2006, 404: 154–158. 10.1016/j.neulet.2006.05.030CrossRefPubMed
20.
go back to reference Kawasaki Y, Xu ZZ, Wang X, Park JY, Zhuang ZY, Tan PH, et al.: Distinct roles of matrix metalloproteases in the early- and late-phase development of neuropathic pain. Nat Med 2008, 14: 331–336. 10.1038/nm1723PubMedCentralCrossRefPubMed Kawasaki Y, Xu ZZ, Wang X, Park JY, Zhuang ZY, Tan PH, et al.: Distinct roles of matrix metalloproteases in the early- and late-phase development of neuropathic pain. Nat Med 2008, 14: 331–336. 10.1038/nm1723PubMedCentralCrossRefPubMed
21.
go back to reference Chattopadhyay S, Shubayev VI: MMP-9 controls Schwann cell proliferation and phenotypic remodeling via IGF-1 and ErbB receptor-mediated activation of MEK/ERK pathway. Glia 2009, 57: 1316–1325. 10.1002/glia.20851PubMedCentralCrossRefPubMed Chattopadhyay S, Shubayev VI: MMP-9 controls Schwann cell proliferation and phenotypic remodeling via IGF-1 and ErbB receptor-mediated activation of MEK/ERK pathway. Glia 2009, 57: 1316–1325. 10.1002/glia.20851PubMedCentralCrossRefPubMed
22.
go back to reference Hanani M: Satellite glial cells in sensory ganglia: from form to function. Brain Res Brain Res Rev 2005, 48: 457–476.CrossRefPubMed Hanani M: Satellite glial cells in sensory ganglia: from form to function. Brain Res Brain Res Rev 2005, 48: 457–476.CrossRefPubMed
23.
go back to reference Xie W, Strong JA, Zhang JM: Early blockade of injured primary sensory afferents reduces glial cell activation in two rat neuropathic pain models. Neuroscience 2009, 160: 847–857. 10.1016/j.neuroscience.2009.03.016PubMedCentralCrossRefPubMed Xie W, Strong JA, Zhang JM: Early blockade of injured primary sensory afferents reduces glial cell activation in two rat neuropathic pain models. Neuroscience 2009, 160: 847–857. 10.1016/j.neuroscience.2009.03.016PubMedCentralCrossRefPubMed
24.
go back to reference Dublin P, Hanani M: Satellite glial cells in sensory ganglia: their possible contribution to inflammatory pain. Brain Behav Immun 2007, 21: 592–598. 10.1016/j.bbi.2006.11.011CrossRefPubMed Dublin P, Hanani M: Satellite glial cells in sensory ganglia: their possible contribution to inflammatory pain. Brain Behav Immun 2007, 21: 592–598. 10.1016/j.bbi.2006.11.011CrossRefPubMed
25.
go back to reference Dubovy P, Klusakova I, Svizenska I, Brazda V: Satellite glial cells express IL-6 and corresponding signal-transducing receptors in the dorsal root ganglia of rat neuropathic pain model. Neuron Glia Biol 2010, 6: 73–83. 10.1017/S1740925X10000074CrossRefPubMed Dubovy P, Klusakova I, Svizenska I, Brazda V: Satellite glial cells express IL-6 and corresponding signal-transducing receptors in the dorsal root ganglia of rat neuropathic pain model. Neuron Glia Biol 2010, 6: 73–83. 10.1017/S1740925X10000074CrossRefPubMed
26.
go back to reference Liang L, Wang Z, Lu N, Yang J, Zhang Y, Zhao Z: Involvement of nerve injury and activation of peripheral glial cells in tetanic sciatic stimulation-induced persistent pain in rats. J Neurosci Res 2010, 88: 2899–2910.PubMed Liang L, Wang Z, Lu N, Yang J, Zhang Y, Zhao Z: Involvement of nerve injury and activation of peripheral glial cells in tetanic sciatic stimulation-induced persistent pain in rats. J Neurosci Res 2010, 88: 2899–2910.PubMed
27.
go back to reference Zhang H, Mei X, Zhang P, Ma C, White FA, Donnelly DF, et al.: Altered functional properties of satellite glial cells in compressed spinal ganglia. Glia 2009, 57: 1588–1599. 10.1002/glia.20872PubMedCentralCrossRefPubMed Zhang H, Mei X, Zhang P, Ma C, White FA, Donnelly DF, et al.: Altered functional properties of satellite glial cells in compressed spinal ganglia. Glia 2009, 57: 1588–1599. 10.1002/glia.20872PubMedCentralCrossRefPubMed
28.
go back to reference Takeda M, Tanimoto T, Kadoi J, Nasu M, Takahashi M, Kitagawa J, et al.: Enhanced excitability of nociceptive trigeminal ganglion neurons by satellite glial cytokine following peripheral inflammation. Pain 2007, 129: 155–166. 10.1016/j.pain.2006.10.007CrossRefPubMed Takeda M, Tanimoto T, Kadoi J, Nasu M, Takahashi M, Kitagawa J, et al.: Enhanced excitability of nociceptive trigeminal ganglion neurons by satellite glial cytokine following peripheral inflammation. Pain 2007, 129: 155–166. 10.1016/j.pain.2006.10.007CrossRefPubMed
29.
go back to reference Takeda M, Takahashi M, Nasu M, Matsumoto S: Peripheral inflammation suppresses inward rectifying potassium currents of satellite glial cells in the trigeminal ganglia. Pain 2011, 152: 2147–2156. 10.1016/j.pain.2011.05.023CrossRefPubMed Takeda M, Takahashi M, Nasu M, Matsumoto S: Peripheral inflammation suppresses inward rectifying potassium currents of satellite glial cells in the trigeminal ganglia. Pain 2011, 152: 2147–2156. 10.1016/j.pain.2011.05.023CrossRefPubMed
30.
go back to reference Wilson NM, Jung H, Ripsch MS, Miller RJ, White FA: CXCR4 signaling mediates morphine-induced tactile hyperalgesia. Brain Behav Immun 2011, 25: 565–573. 10.1016/j.bbi.2010.12.014PubMedCentralCrossRefPubMed Wilson NM, Jung H, Ripsch MS, Miller RJ, White FA: CXCR4 signaling mediates morphine-induced tactile hyperalgesia. Brain Behav Immun 2011, 25: 565–573. 10.1016/j.bbi.2010.12.014PubMedCentralCrossRefPubMed
31.
go back to reference Hutchinson MR, Coats BD, Lewis SS, Zhang Y, Sprunger DB, Rezvani N, et al.: Proinflammatory cytokines oppose opioid-induced acute and chronic analgesia. Brain Behav Immun 2008, 22: 1178–1189. 10.1016/j.bbi.2008.05.004PubMedCentralCrossRefPubMed Hutchinson MR, Coats BD, Lewis SS, Zhang Y, Sprunger DB, Rezvani N, et al.: Proinflammatory cytokines oppose opioid-induced acute and chronic analgesia. Brain Behav Immun 2008, 22: 1178–1189. 10.1016/j.bbi.2008.05.004PubMedCentralCrossRefPubMed
32.
go back to reference Kawasaki Y, Xu ZZ, Wang X, Park JY, Zhuang ZY, Tan PH, et al.: Distinct roles of matrix metalloproteases in the early- and late-phase development of neuropathic pain. Nat Med 2008, 14: 331–336. 10.1038/nm1723PubMedCentralCrossRefPubMed Kawasaki Y, Xu ZZ, Wang X, Park JY, Zhuang ZY, Tan PH, et al.: Distinct roles of matrix metalloproteases in the early- and late-phase development of neuropathic pain. Nat Med 2008, 14: 331–336. 10.1038/nm1723PubMedCentralCrossRefPubMed
33.
34.
go back to reference Watkins LR, Hutchinson MR, Johnston IN, Maier SF: Glia: novel counter-regulators of opioid analgesia. Trends Neurosci 2005, 28: 661–669. 10.1016/j.tins.2005.10.001CrossRefPubMed Watkins LR, Hutchinson MR, Johnston IN, Maier SF: Glia: novel counter-regulators of opioid analgesia. Trends Neurosci 2005, 28: 661–669. 10.1016/j.tins.2005.10.001CrossRefPubMed
35.
go back to reference Zhou D, Chen ML, Zhang YQ, Zhao ZQ: Involvement of spinal microglial P2X7 receptor in generation of tolerance to morphine analgesia in rats. J Neurosci 2010, 30: 8042–8047. 10.1523/JNEUROSCI.5377-09.2010CrossRefPubMed Zhou D, Chen ML, Zhang YQ, Zhao ZQ: Involvement of spinal microglial P2X7 receptor in generation of tolerance to morphine analgesia in rats. J Neurosci 2010, 30: 8042–8047. 10.1523/JNEUROSCI.5377-09.2010CrossRefPubMed
36.
go back to reference Horvath RJ, Landry RP, Romero-Sandoval EA, DeLeo JA: Morphine tolerance attenuates the resolution of postoperative pain and enhances spinal microglial p38 and extracellular receptor kinase phosphorylation. Neuroscience 2010, 169: 843–854. 10.1016/j.neuroscience.2010.05.030PubMedCentralCrossRefPubMed Horvath RJ, Landry RP, Romero-Sandoval EA, DeLeo JA: Morphine tolerance attenuates the resolution of postoperative pain and enhances spinal microglial p38 and extracellular receptor kinase phosphorylation. Neuroscience 2010, 169: 843–854. 10.1016/j.neuroscience.2010.05.030PubMedCentralCrossRefPubMed
37.
go back to reference Cui Y, Chen Y, Zhi JL, Guo RX, Feng JQ, Chen PX: Activation of p38 mitogen-activated protein kinase in spinal microglia mediates morphine antinociceptive tolerance. Brain Res 2006, 1069: 235–243. 10.1016/j.brainres.2005.11.066CrossRefPubMed Cui Y, Chen Y, Zhi JL, Guo RX, Feng JQ, Chen PX: Activation of p38 mitogen-activated protein kinase in spinal microglia mediates morphine antinociceptive tolerance. Brain Res 2006, 1069: 235–243. 10.1016/j.brainres.2005.11.066CrossRefPubMed
39.
go back to reference Hanani M, Huang TY, Cherkas PS, Ledda M, Pannese E: Glial cell plasticity in sensory ganglia induced by nerve damage. Neuroscience 2002, 114: 279–283. 10.1016/S0306-4522(02)00279-8CrossRefPubMed Hanani M, Huang TY, Cherkas PS, Ledda M, Pannese E: Glial cell plasticity in sensory ganglia induced by nerve damage. Neuroscience 2002, 114: 279–283. 10.1016/S0306-4522(02)00279-8CrossRefPubMed
40.
go back to reference Dublin P, Hanani M: Satellite glial cells in sensory ganglia: their possible contribution to inflammatory pain. Brain Behav Immun 2007, 21: 592–598. 10.1016/j.bbi.2006.11.011CrossRefPubMed Dublin P, Hanani M: Satellite glial cells in sensory ganglia: their possible contribution to inflammatory pain. Brain Behav Immun 2007, 21: 592–598. 10.1016/j.bbi.2006.11.011CrossRefPubMed
41.
go back to reference Chen Y, Zhang X, Wang C, Li G, Gu Y, Huang LY: Activation of P2X7 receptors in glial satellite cells reduces pain through downregulation of P2X3 receptors in nociceptive neurons. Proc Natl Acad Sci USA 2008, 105: 16773–16778. 10.1073/pnas.0801793105PubMedCentralCrossRefPubMed Chen Y, Zhang X, Wang C, Li G, Gu Y, Huang LY: Activation of P2X7 receptors in glial satellite cells reduces pain through downregulation of P2X3 receptors in nociceptive neurons. Proc Natl Acad Sci USA 2008, 105: 16773–16778. 10.1073/pnas.0801793105PubMedCentralCrossRefPubMed
42.
go back to reference Zhang X, Chen Y, Wang C, Huang LY: Neuronal somatic ATP release triggers neuron-satellite glial cell communication in dorsal root ganglia. Proc Natl Acad Sci USA 2007, 104: 9864–9869. 10.1073/pnas.0611048104PubMedCentralCrossRefPubMed Zhang X, Chen Y, Wang C, Huang LY: Neuronal somatic ATP release triggers neuron-satellite glial cell communication in dorsal root ganglia. Proc Natl Acad Sci USA 2007, 104: 9864–9869. 10.1073/pnas.0611048104PubMedCentralCrossRefPubMed
43.
go back to reference Takeda M, Takahashi M, Matsumoto S: Contribution of the activation of satellite glia in sensory ganglia to pathological pain. Neurosci Biobehav Rev 2009, 33: 784–792. 10.1016/j.neubiorev.2008.12.005CrossRefPubMed Takeda M, Takahashi M, Matsumoto S: Contribution of the activation of satellite glia in sensory ganglia to pathological pain. Neurosci Biobehav Rev 2009, 33: 784–792. 10.1016/j.neubiorev.2008.12.005CrossRefPubMed
44.
go back to reference Hu P, Bembrick AL, Keay KA, McLachlan EM: Immune cell involvement in dorsal root ganglia and spinal cord after chronic constriction or transection of the rat sciatic nerve. Brain Behav Immun 2007, 21: 599–616. 10.1016/j.bbi.2006.10.013CrossRefPubMed Hu P, Bembrick AL, Keay KA, McLachlan EM: Immune cell involvement in dorsal root ganglia and spinal cord after chronic constriction or transection of the rat sciatic nerve. Brain Behav Immun 2007, 21: 599–616. 10.1016/j.bbi.2006.10.013CrossRefPubMed
45.
go back to reference Zhang H, Mei X, Zhang P, Ma C, White FA, Donnelly DF, et al.: Altered functional properties of satellite glial cells in compressed spinal ganglia. Glia 2009, 57: 1588–1599. 10.1002/glia.20872PubMedCentralCrossRefPubMed Zhang H, Mei X, Zhang P, Ma C, White FA, Donnelly DF, et al.: Altered functional properties of satellite glial cells in compressed spinal ganglia. Glia 2009, 57: 1588–1599. 10.1002/glia.20872PubMedCentralCrossRefPubMed
46.
go back to reference Li JY, Xie W, Strong JA, Guo QL, Zhang JM: Mechanical hypersensitivity, sympathetic sprouting, and glial activation are attenuated by local injection of corticosteroid near the lumbar ganglion in a rat model of neuropathic pain. Reg Anesth Pain Med 2011, 36: 56–62. 10.1097/AAP.0b013e318203087fPubMedCentralCrossRefPubMed Li JY, Xie W, Strong JA, Guo QL, Zhang JM: Mechanical hypersensitivity, sympathetic sprouting, and glial activation are attenuated by local injection of corticosteroid near the lumbar ganglion in a rat model of neuropathic pain. Reg Anesth Pain Med 2011, 36: 56–62. 10.1097/AAP.0b013e318203087fPubMedCentralCrossRefPubMed
47.
go back to reference Takeda M, Takahashi M, Matsumoto S: Contribution of activated interleukin receptors in trigeminal ganglion neurons to hyperalgesia via satellite glial interleukin-1beta paracrine mechanism. Brain Behav Immun 2008, 22: 1016–1023. 10.1016/j.bbi.2008.03.004CrossRefPubMed Takeda M, Takahashi M, Matsumoto S: Contribution of activated interleukin receptors in trigeminal ganglion neurons to hyperalgesia via satellite glial interleukin-1beta paracrine mechanism. Brain Behav Immun 2008, 22: 1016–1023. 10.1016/j.bbi.2008.03.004CrossRefPubMed
48.
go back to reference Takeda M, Tanimoto T, Kadoi J, Nasu M, Takahashi M, Kitagawa J, et al.: Enhanced excitability of nociceptive trigeminal ganglion neurons by satellite glial cytokine following peripheral inflammation. Pain 2007, 129: 155–166. 10.1016/j.pain.2006.10.007CrossRefPubMed Takeda M, Tanimoto T, Kadoi J, Nasu M, Takahashi M, Kitagawa J, et al.: Enhanced excitability of nociceptive trigeminal ganglion neurons by satellite glial cytokine following peripheral inflammation. Pain 2007, 129: 155–166. 10.1016/j.pain.2006.10.007CrossRefPubMed
49.
go back to reference Copray JC, Mantingh I, Brouwer N, Biber K, Kust BM, Liem RS, et al.: Expression of interleukin-1 beta in rat dorsal root ganglia. J Neuroimmunol 2001, 118: 203–211. 10.1016/S0165-5728(01)00324-1CrossRefPubMed Copray JC, Mantingh I, Brouwer N, Biber K, Kust BM, Liem RS, et al.: Expression of interleukin-1 beta in rat dorsal root ganglia. J Neuroimmunol 2001, 118: 203–211. 10.1016/S0165-5728(01)00324-1CrossRefPubMed
50.
go back to reference Binshtok AM, Wang H, Zimmermann K, Amaya F, Vardeh D, Shi L, et al.: Nociceptors are interleukin-1beta sensors. J Neurosci 2008, 28: 14062–14073. 10.1523/JNEUROSCI.3795-08.2008PubMedCentralCrossRefPubMed Binshtok AM, Wang H, Zimmermann K, Amaya F, Vardeh D, Shi L, et al.: Nociceptors are interleukin-1beta sensors. J Neurosci 2008, 28: 14062–14073. 10.1523/JNEUROSCI.3795-08.2008PubMedCentralCrossRefPubMed
51.
go back to reference Takeda M, Kitagawa J, Takahashi M, Matsumoto S: Activation of interleukin-1beta receptor suppresses the voltage-gated potassium currents in the small-diameter trigeminal ganglion neurons following peripheral inflammation. Pain 2008, 139: 594–602. 10.1016/j.pain.2008.06.015CrossRefPubMed Takeda M, Kitagawa J, Takahashi M, Matsumoto S: Activation of interleukin-1beta receptor suppresses the voltage-gated potassium currents in the small-diameter trigeminal ganglion neurons following peripheral inflammation. Pain 2008, 139: 594–602. 10.1016/j.pain.2008.06.015CrossRefPubMed
52.
go back to reference Hazuda DJ, Strickler J, Kueppers F, Simon PL, Young PR: Processing of precursor interleukin 1 beta and inflammatory disease. J Biol Chem 1990, 265: 6318–6322.PubMed Hazuda DJ, Strickler J, Kueppers F, Simon PL, Young PR: Processing of precursor interleukin 1 beta and inflammatory disease. J Biol Chem 1990, 265: 6318–6322.PubMed
53.
go back to reference Schonbeck U, Mach F, Libby P: Generation of biologically active IL-1 beta by matrix metalloproteinases: a novel caspase-1-independent pathway of IL-1 beta processing. J Immunol 1998, 161: 3340–3346.PubMed Schonbeck U, Mach F, Libby P: Generation of biologically active IL-1 beta by matrix metalloproteinases: a novel caspase-1-independent pathway of IL-1 beta processing. J Immunol 1998, 161: 3340–3346.PubMed
54.
go back to reference Parks WC, Wilson CL, Lopez-Boado YS: Matrix metalloproteinases as modulators of inflammation and innate immunity. Nat Rev Immunol 2004, 4: 617–629. 10.1038/nri1418CrossRefPubMed Parks WC, Wilson CL, Lopez-Boado YS: Matrix metalloproteinases as modulators of inflammation and innate immunity. Nat Rev Immunol 2004, 4: 617–629. 10.1038/nri1418CrossRefPubMed
55.
go back to reference Fantuzzi G, Ku G, Harding MW, Livingston DJ, Sipe JD, Kuida K, et al.: Response to local inflammation of IL-1 beta-converting enzyme- deficient mice. J Immunol 1997, 158: 1818–1824.PubMed Fantuzzi G, Ku G, Harding MW, Livingston DJ, Sipe JD, Kuida K, et al.: Response to local inflammation of IL-1 beta-converting enzyme- deficient mice. J Immunol 1997, 158: 1818–1824.PubMed
56.
go back to reference Ito A, Mukaiyama A, Itoh Y, Nagase H, Thogersen IB, Enghild JJ, et al.: Degradation of interleukin 1beta by matrix metalloproteinases. J Biol Chem 1996, 271: 14657–14660. 10.1074/jbc.271.25.14657CrossRefPubMed Ito A, Mukaiyama A, Itoh Y, Nagase H, Thogersen IB, Enghild JJ, et al.: Degradation of interleukin 1beta by matrix metalloproteinases. J Biol Chem 1996, 271: 14657–14660. 10.1074/jbc.271.25.14657CrossRefPubMed
57.
go back to reference Chen L, Huang LY: Sustained potentiation of NMDA receptor-mediated glutamate responses through activation of protein kinase C by a mu opioid. Neuron 1991, 7: 319–326. 10.1016/0896-6273(91)90270-ACrossRefPubMed Chen L, Huang LY: Sustained potentiation of NMDA receptor-mediated glutamate responses through activation of protein kinase C by a mu opioid. Neuron 1991, 7: 319–326. 10.1016/0896-6273(91)90270-ACrossRefPubMed
58.
go back to reference Drdla R, Gassner M, Gingl E, Sandkuhler J: Induction of synaptic long-term potentiation after opioid withdrawal. Science 2009, 325: 207–210. 10.1126/science.1171759CrossRefPubMed Drdla R, Gassner M, Gingl E, Sandkuhler J: Induction of synaptic long-term potentiation after opioid withdrawal. Science 2009, 325: 207–210. 10.1126/science.1171759CrossRefPubMed
59.
go back to reference Zhou HY, Chen SR, Chen H, Pan HL: Opioid-induced long-term potentiation in the spinal cord is a presynaptic event. J Neurosci 2010, 30: 4460–4466. 10.1523/JNEUROSCI.5857-09.2010PubMedCentralCrossRefPubMed Zhou HY, Chen SR, Chen H, Pan HL: Opioid-induced long-term potentiation in the spinal cord is a presynaptic event. J Neurosci 2010, 30: 4460–4466. 10.1523/JNEUROSCI.5857-09.2010PubMedCentralCrossRefPubMed
60.
go back to reference Ji RR, Samad TA, Jin SX, Schmoll R, Woolf CJ: p38 MAPK activation by NGF in primary sensory neurons after inflammation increases TRPV1 levels and maintains heat hyperalgesia. Neuron 2002, 36: 57–68. 10.1016/S0896-6273(02)00908-XCrossRefPubMed Ji RR, Samad TA, Jin SX, Schmoll R, Woolf CJ: p38 MAPK activation by NGF in primary sensory neurons after inflammation increases TRPV1 levels and maintains heat hyperalgesia. Neuron 2002, 36: 57–68. 10.1016/S0896-6273(02)00908-XCrossRefPubMed
61.
go back to reference Obata K, Katsura H, Mizushima T, Yamanaka H, Kobayashi K, Dai Y, et al.: TRPA1 induced in sensory neurons contributes to cold hyperalgesia after inflammation and nerve injury. J Clin Invest 2005, 115: 2393–2401. 10.1172/JCI25437PubMedCentralCrossRefPubMed Obata K, Katsura H, Mizushima T, Yamanaka H, Kobayashi K, Dai Y, et al.: TRPA1 induced in sensory neurons contributes to cold hyperalgesia after inflammation and nerve injury. J Clin Invest 2005, 115: 2393–2401. 10.1172/JCI25437PubMedCentralCrossRefPubMed
62.
go back to reference Zhuang ZY, Wen YR, Zhang DR, Borsello T, Bonny C, Strichartz GR, et al.: A peptide c-Jun N-terminal kinase (JNK) inhibitor blocks mechanical allodynia after spinal nerve ligation: respective roles of JNK activation in primary sensory neurons and spinal astrocytes for neuropathic pain development and maintenance. J Neurosci 2006, 26: 3551–3560. 10.1523/JNEUROSCI.5290-05.2006CrossRefPubMed Zhuang ZY, Wen YR, Zhang DR, Borsello T, Bonny C, Strichartz GR, et al.: A peptide c-Jun N-terminal kinase (JNK) inhibitor blocks mechanical allodynia after spinal nerve ligation: respective roles of JNK activation in primary sensory neurons and spinal astrocytes for neuropathic pain development and maintenance. J Neurosci 2006, 26: 3551–3560. 10.1523/JNEUROSCI.5290-05.2006CrossRefPubMed
63.
go back to reference Gold MS, Weinreich D, Kim CS, Wang R, Treanor J, Porreca F, et al.: Redistribution of Na(V)1.8 in uninjured axons enables neuropathic pain. J Neurosci 2003, 23: 158–166.PubMed Gold MS, Weinreich D, Kim CS, Wang R, Treanor J, Porreca F, et al.: Redistribution of Na(V)1.8 in uninjured axons enables neuropathic pain. J Neurosci 2003, 23: 158–166.PubMed
64.
go back to reference Alessandri-Haber N, Dina OA, Chen X, Levine JD: TRPC1 and TRPC6 channels cooperate with TRPV4 to mediate mechanical hyperalgesia and nociceptor sensitization. J Neurosci 2009, 29: 6217–6228. 10.1523/JNEUROSCI.0893-09.2009PubMedCentralCrossRefPubMed Alessandri-Haber N, Dina OA, Chen X, Levine JD: TRPC1 and TRPC6 channels cooperate with TRPV4 to mediate mechanical hyperalgesia and nociceptor sensitization. J Neurosci 2009, 29: 6217–6228. 10.1523/JNEUROSCI.0893-09.2009PubMedCentralCrossRefPubMed
65.
go back to reference Luo MC, Zhang DQ, Ma SW, Huang YY, Shuster SJ, Porreca F, et al.: An efficient intrathecal delivery of small interfering RNA to the spinal cord and peripheral neurons. Mol Pain 2005, 1: 29. 10.1186/1744-8069-1-29PubMedCentralCrossRefPubMed Luo MC, Zhang DQ, Ma SW, Huang YY, Shuster SJ, Porreca F, et al.: An efficient intrathecal delivery of small interfering RNA to the spinal cord and peripheral neurons. Mol Pain 2005, 1: 29. 10.1186/1744-8069-1-29PubMedCentralCrossRefPubMed
66.
go back to reference Mizushima T, Obata K, Yamanaka H, Dai Y, Fukuoka T, Tokunaga A, et al.: Activation of p38 MAPK in primary afferent neurons by noxious stimulation and its involvement in the development of thermal hyperalgesia. Pain 2005, 113: 51–60. 10.1016/j.pain.2004.09.038CrossRefPubMed Mizushima T, Obata K, Yamanaka H, Dai Y, Fukuoka T, Tokunaga A, et al.: Activation of p38 MAPK in primary afferent neurons by noxious stimulation and its involvement in the development of thermal hyperalgesia. Pain 2005, 113: 51–60. 10.1016/j.pain.2004.09.038CrossRefPubMed
67.
go back to reference Stone LS, MacMillan LB, Kitto KF, Limbird LE, Wilcox GL: The alpha2a adrenergic receptor subtype mediates spinal analgesia evoked by alpha2 agonists and is necessary for spinal adrenergic-opioid synergy. J Neurosci 1997, 17: 7157–7165.PubMed Stone LS, MacMillan LB, Kitto KF, Limbird LE, Wilcox GL: The alpha2a adrenergic receptor subtype mediates spinal analgesia evoked by alpha2 agonists and is necessary for spinal adrenergic-opioid synergy. J Neurosci 1997, 17: 7157–7165.PubMed
68.
go back to reference Jin SX, Zhuang ZY, Woolf CJ, Ji RR: p38 mitogen-activated protein kinase is activated after a spinal nerve ligation in spinal cord microglia and dorsal root ganglion neurons and contributes to the generation of neuropathic pain. J Neurosci 2003, 23: 4017–4022.PubMed Jin SX, Zhuang ZY, Woolf CJ, Ji RR: p38 mitogen-activated protein kinase is activated after a spinal nerve ligation in spinal cord microglia and dorsal root ganglion neurons and contributes to the generation of neuropathic pain. J Neurosci 2003, 23: 4017–4022.PubMed
69.
go back to reference Zhuang ZY, Gerner P, Woolf CJ, Ji RR: ERK is sequentially activated in neurons, microglia, and astrocytes by spinal nerve ligation and contributes to mechanical allodynia in this neuropathic pain model. Pain 2005, 114: 149–159. 10.1016/j.pain.2004.12.022CrossRefPubMed Zhuang ZY, Gerner P, Woolf CJ, Ji RR: ERK is sequentially activated in neurons, microglia, and astrocytes by spinal nerve ligation and contributes to mechanical allodynia in this neuropathic pain model. Pain 2005, 114: 149–159. 10.1016/j.pain.2004.12.022CrossRefPubMed
70.
go back to reference Berta T, Poirot O, Pertin M, Ji RR, Kellenberger S, Decosterd I: Transcriptional and functional profiles of voltage-gated Na(+) channels in injured and non-injured DRG neurons in the SNI model of neuropathic pain. Mol Cell Neurosci 2008, 37: 196–208. 10.1016/j.mcn.2007.09.007CrossRefPubMed Berta T, Poirot O, Pertin M, Ji RR, Kellenberger S, Decosterd I: Transcriptional and functional profiles of voltage-gated Na(+) channels in injured and non-injured DRG neurons in the SNI model of neuropathic pain. Mol Cell Neurosci 2008, 37: 196–208. 10.1016/j.mcn.2007.09.007CrossRefPubMed
Metadata
Title
Acute morphine activates satellite glial cells and up-regulates IL-1β in dorsal root ganglia in mice via matrix metalloprotease-9
Authors
Temugin Berta
Tong Liu
Yen-Chin Liu
Zhen-Zhong Xu
Ru-Rong Ji
Publication date
01-12-2012
Publisher
BioMed Central
Published in
Molecular Pain / Issue 1/2012
Electronic ISSN: 1744-8069
DOI
https://doi.org/10.1186/1744-8069-8-18

Other articles of this Issue 1/2012

Molecular Pain 1/2012 Go to the issue