Publication Cover
Neurological Research
A Journal of Progress in Neurosurgery, Neurology and Neurosciences
Volume 41, 2019 - Issue 6
262
Views
6
CrossRef citations to date
0
Altmetric
Original Research Paper

COX-2 contributed to the remifentanil-induced hyperalgesia related to ephrinB/EphB signaling

, , , &
Pages 519-527 | Received 23 Sep 2018, Accepted 03 Feb 2019, Published online: 13 Feb 2019

References

  • Angst MS, Clark JD. Opioid-induced hyperalgesia: a qualitative systematic review. Anesthesiology. 2006;104:570–587.
  • Chen L, Malarick C, Seefeld L, et al. Altered quantitative sensory testing outcome in subjects with opioid therapy. Pain. 2009;143:65–70.
  • Egan TD, Lemmens HJ, Fiset P, et al. The pharmacokinetics of the new short-acting opioid remifentanil (GI87084B) in healthy adult male volunteers. Anesthesiology. 1993;79:881–892.
  • Celerier E, Rivat C, Jun Y, et al. Long-lasting hyperalgesia induced by fentanyl in rats: preventive effect of ketamine. Anesthesiology. 2000;92:465–472.
  • Derrode N, Lebrun F, Levron JC, et al. Influence of peroperative opioid on postoperative pain after major abdominal surgery: sufentanil TCI versus remifentanil TCI. A randomized, controlled study. Br J Anaesth. 2003;91:842–849.
  • Ma JF, Huang ZL, Li J, et al. Cohort study of remifentanil-induced hyperalgesia in postoperative patients. Zhonghua yi xue za zhi. 2011;91:977–979.
  • Xia WS, Peng YN, Tang LH, et al. Spinal ephrinB/EphB signalling contributed to remifentanil-induced hyperalgesia via NMDA receptor. Eur J pain. 2014;18:1231–1239.
  • Feldman PL, James MK, Brackeen MF, et al. Design, synthesis, and pharmacological evaluation of ultrashort- to long-acting opioid analgetics. J Med Chem. 1991;34:2202–2208.
  • Thompson JP, Rowbotham DJ. Remifentanil–an opioid for the 21st century. Br J Anaesth. 1996;76:341–343.
  • Trescot AM, Datta S, Lee M, et al. Opioid pharmacology. Pain Physician. 2008;11:S133–S153.
  • Drdla R, Gassner M, Gingl E, et al. Induction of synaptic long-term potentiation after opioid withdrawal. Science. 2009;325:207–210.
  • Koppert W, Sittl R, Scheuber K, et al. Differential modulation of remifentanil-induced analgesia and postinfusion hyperalgesia by S-ketamine and clonidine in humans. Anesthesiology. 2003;99:152–159.
  • Rivat C, Laulin JP, Corcuff JB, et al. Fentanyl enhancement of carrageenan-induced long-lasting hyperalgesia in rats: prevention by the N-methyl-D-aspartate receptor antagonist ketamine. Anesthesiology. 2002;96:381–391.
  • Song XJ, Zheng JH, Cao JL, et al. EphrinB-EphB receptor signaling contributes to neuropathic pain by regulating neural excitability and spinal synaptic plasticity in rats. Pain. 2008;139:168–180.
  • Ruan JP, Zhang HX, Lu XF, et al. EphrinBs/EphBs signaling is involved in modulation of spinal nociceptive processing through a mitogen-activated protein kinases-dependent mechanism. Anesthesiology. 2010;112:1234–1249.
  • Lenz H, Raeder J, Draegni T, et al. Effects of COX inhibition on experimental pain and hyperalgesia during and after remifentanil infusion in humans. Pain. 2011;152:1289–1297.
  • Troster A, Sittl R, Singler B, et al. Modulation of remifentanil-induced analgesia and postinfusion hyperalgesia by parecoxib in humans. Anesthesiology. 2006;105:1016–1023.
  • Bezzi P, Carmignoto G, Pasti L, et al. Prostaglandins stimulate calcium-dependent glutamate release in astrocytes. Nature. 1998;391:281–285.
  • O’Rielly DD, Loomis CW. Increased expression of cyclooxygenase and nitric oxide isoforms, and exaggerated sensitivity to prostaglandin E2, in the rat lumbar spinal cord 3 days after L5-L6 spinal nerve ligation. Anesthesiology. 2006;104:328–337.
  • Malmberg AB, Yaksh TL. Hyperalgesia mediated by spinal glutamate or substance P receptor blocked by spinal cyclooxygenase inhibition. Science. 1992;257:1276–1279.
  • Yu LN, Zhou XL, Yu J, et al. PI3K contributed to modulation of spinal nociceptive information related to ephrinBs/EphBs. PloS one. 2012;7:e40930.
  • Ji RR, Strichartz G. Cell signaling and the genesis of neuropathic pain. Sci STKE. 2004;2004:reE14.
  • Simonetti M, Hagenston AM, Vardeh D, et al. Nuclear calcium signaling in spinal neurons drives a genomic program required for persistent inflammatory pain. Neuron. 2013;77:43–57.
  • Karim F, Wang CC, Gereau R. Metabotropic glutamate receptor subtypes 1 and 5 are activators of extracellular signal-regulated kinase signaling required for inflammatory pain in mice. J Neurosci. 2001;21:3771–3779.
  • Samad TA, Moore KA, Sapirstein A, et al. Interleukin-1beta-mediated induction of Cox-2 in the CNS contributes to inflammatory pain hypersensitivity. Nature. 2001;410:471–475.
  • Seybold VS, Jia YP, Abrahams LG. Cyclo-oxygenase-2 contributes to central sensitization in rats with peripheral inflammation. Pain. 2003;105:47–55.
  • Zhou XL, Wang Y, Zhang CJ, et al. COX-2 is required for the modulation of spinal nociceptive information related to ephrinB/EphB signalling. Eur J pain. 2015;19:1277–1287.
  • Craft RM. Modulation of pain by estrogens. Pain. 2007;132(Suppl 1):S3–S12.
  • Mestre C, Pelissier T, Fialip J, et al. A method to perform direct transcutaneous intrathecal injection in rats. J Pharmacol Toxicol Methods. 1994;32:197–200.
  • Criado AB, Gomez E Segura IA. Reduction of isoflurane MAC by fentanyl or remifentanil in rats. Vet Anaesth Analg. 2003;30:250–256.
  • Brennan TJ, Vandermeulen EP, Gebhart GF. Characterization of a rat model of incisional pain. Pain. 1996;64:493–501.
  • Celerier E, Gonzalez JR, Maldonado R, et al. Opioid-induced hyperalgesia in a murine model of postoperative pain: role of nitric oxide generated from the inducible nitric oxide synthase. Anesthesiology. 2006;104:546–555.
  • Cabanero D, Celerier E, Garcia-Nogales P, et al. The pro-nociceptive effects of remifentanil or surgical injury in mice are associated with a decrease in delta-opioid receptor mRNA levels: prevention of the nociceptive response by on-site delivery of enkephalins. Pain. 2009;141:88–96.
  • Hargreaves K, Dubner R, Brown F, et al. A new and sensitive method for measuring thermal nociception in cutaneous hyperalgesia. Pain. 1988;32:77–88.
  • Chaplan SR, Bach FW, Pogrel JW, et al. Quantitative assessment of tactile allodynia in the rat paw. J Neurosci Methods. 1994;53:55–63.
  • Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 1976;72:248–254.
  • Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 2001;25:402–408.
  • Chu LF, Clark DJ, Angst MS. Opioid tolerance and hyperalgesia in chronic pain patients after one month of oral morphine therapy: a preliminary prospective study. J Pain. 2006;7:43–48.
  • Dalva MB, Takasu MA, Lin MZ, et al. EphB receptors interact with NMDA receptors and regulate excitatory synapse formation. Cell. 2000;103:945–956.
  • Chu LF, Angst MS, Clark D. Opioid-induced hyperalgesia in humans: molecular mechanisms and clinical considerations. Clin J Pain. 2008;24:479–496.
  • Ji RR, Baba H, Brenner GJ, et al. Nociceptive-specific activation of ERK in spinal neurons contributes to pain hypersensitivity. Nat Neurosci. 1999;2:1114–1119.
  • Obata K, Yamanaka H, Dai Y, et al. Differential activation of extracellular signal-regulated protein kinase in primary afferent neurons regulates brain-derived neurotrophic factor expression after peripheral inflammation and nerve injury. J Neurosci. 2003;23:4117–4126.
  • Amaya F, Samad TA, Barrett L, et al. Periganglionic inflammation elicits a distally radiating pain hypersensitivity by promoting COX-2 induction in the dorsal root ganglion. Pain. 2009;142:59–67.
  • Buvanendran A, Kroin JS, Berger RA, et al. Upregulation of prostaglandin E2 and interleukins in the central nervous system and peripheral tissue during and after surgery in humans. Anesthesiology. 2006;104:403–410.
  • Kohno T, Wang H, Amaya F, et al. Bradykinin enhances AMPA and NMDA receptor activity in spinal cord dorsal horn neurons by activating multiple kinases to produce pain hypersensitivity. J Neurosci. 2008;28:4533–4540.
  • Ahmadi S, Lippross S, Neuhuber WL, et al. PGE(2) selectively blocks inhibitory glycinergic neurotransmission onto rat superficial dorsal horn neurons. Nat Neurosci. 2002;5:34–40.
  • Harvey RJ, Depner UB, Wassle H, et al. GlyR alpha3: an essential target for spinal PGE2-mediated inflammatory pain sensitization. Science. 2004;304:884–887.
  • Cao DL, Zhang ZJ, Xie RG, et al. Chemokine CXCL1 enhances inflammatory pain and increases NMDA receptor activity and COX-2 expression in spinal cord neurons via activation of CXCR2. Exp Neurol. 2014;261:328–336.
  • Guo YJ, Shi XD, Fu D, et al. Analgesic effects of the COX-2 inhibitor parecoxib on surgical pain through suppression of spinal ERK signaling. Exp Ther Med. 2013;6:275–279.
  • Zhao P, Waxman SG, Hains BC. Extracellular signal-regulated kinase-regulated microglia-neuron signaling by prostaglandin E2 contributes to pain after spinal cord injury. J Neurosci. 2007;27:2357–2368.
  • Zhang X, Xin X, Dong Y, et al. Surgical incision-induced nociception causes cognitive impairment and reduction in synaptic NMDA receptor 2B in mice. J Neurosci. 2013;33:17737–17748.
  • Rivat C, Richebe P, Laboureyras E, et al. Polyamine deficient diet to relieve pain hypersensitivity. Pain. 2008;137:125–137.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.