메뉴 건너뛰기




Volumn 338, Issue , 2016, Pages 160-182

Opioid-induced hyperalgesia: Cellular and molecular mechanisms

Author keywords

morphine; mu opioid receptor; neuroinflammation; opiates; opioid induced hyperalgesia; pain

Indexed keywords

ADENOSINE TRIPHOSPHATE; BETA ARRESTIN 2; BRAIN DERIVED NEUROTROPHIC FACTOR; CHEMOKINE RECEPTOR CX3CR1; CHEMOKINE RECEPTOR CXCR2; CHEMOKINE RECEPTOR CXCR4; CXCL1 CHEMOKINE; EPHRIN B RECEPTOR; EPHRIN RECEPTOR; INTERLEUKIN 1BETA; INTERLEUKIN 8; MACROPHAGE INFLAMMATORY PROTEIN 1ALPHA; MAMMALIAN TARGET OF RAPAMYCIN; MITOGEN ACTIVATED PROTEIN KINASE 1; MITOGEN ACTIVATED PROTEIN KINASE 3; MONOCYTE CHEMOTACTIC PROTEIN 1; MU OPIATE RECEPTOR; N METHYL DEXTRO ASPARTIC ACID RECEPTOR 1; POTASSIUM CHLORIDE COTRANSPORTER 2; PURINERGIC P2X4 RECEPTOR; RANTES; SEROTONIN 3 RECEPTOR; STROMAL CELL DERIVED FACTOR 1; TACHYKININ NK1 RECEPTOR; TACHYKININ RECEPTOR; TOLL LIKE RECEPTOR 4; TRANSIENT RECEPTOR POTENTIAL CHANNEL M8; TUMOR NECROSIS FACTOR; UNCLASSIFIED DRUG; UNINDEXED DRUG; VANILLOID RECEPTOR 1; NARCOTIC ANALGESIC AGENT;

EID: 84994000020     PISSN: 03064522     EISSN: 18737544     Source Type: Journal    
DOI: 10.1016/j.neuroscience.2016.06.029     Document Type: Review
Times cited : (302)

References (264)
  • 1
    • 84926476027 scopus 로고    scopus 로고
    • Amitriptyline, minocycline and maropitant reduce the sevoflurane minimum alveolar concentration and potentiate remifentanil but do not prevent acute opioid tolerance and hyperalgesia in the rat: a randomised laboratory study
    • Aguado, D., Abreu, M., Benito, J., Garcia-Fernandez, J., Gomez de Segura, I.A., Amitriptyline, minocycline and maropitant reduce the sevoflurane minimum alveolar concentration and potentiate remifentanil but do not prevent acute opioid tolerance and hyperalgesia in the rat: a randomised laboratory study. Eur J Anaesthesiol 32 (2015), 248–254.
    • (2015) Eur J Anaesthesiol , vol.32 , pp. 248-254
    • Aguado, D.1    Abreu, M.2    Benito, J.3    Garcia-Fernandez, J.4    Gomez de Segura, I.A.5
  • 2
    • 0034659718 scopus 로고    scopus 로고
    • Chronic hypersensitivity for inflammatory nociceptor sensitization mediated by the epsilon isozyme of protein kinase C
    • Aley, K.O., Messing, R.O., Mochly-Rosen, D., Levine, J.D., Chronic hypersensitivity for inflammatory nociceptor sensitization mediated by the epsilon isozyme of protein kinase C. J Neurosci 20 (2000), 4680–4685.
    • (2000) J Neurosci , vol.20 , pp. 4680-4685
    • Aley, K.O.1    Messing, R.O.2    Mochly-Rosen, D.3    Levine, J.D.4
  • 3
    • 84920702096 scopus 로고    scopus 로고
    • Role of C-fibers in pain and morphine induced analgesia/hyperalgesia in rats
    • Alizadeh, Z., Fereidoni, M., Behnam-Rassouli, M., Hosseini, S., Role of C-fibers in pain and morphine induced analgesia/hyperalgesia in rats. Iran J Neurol 13 (2014), 19–27.
    • (2014) Iran J Neurol , vol.13 , pp. 19-27
    • Alizadeh, Z.1    Fereidoni, M.2    Behnam-Rassouli, M.3    Hosseini, S.4
  • 4
    • 0037251441 scopus 로고    scopus 로고
    • Relationships among morphine metabolism, pain and side effects during long-term treatment: an update
    • Andersen, G., Christrup, L., Sjogren, P., Relationships among morphine metabolism, pain and side effects during long-term treatment: an update. J Pain Symptom Manage 25 (2003), 74–91.
    • (2003) J Pain Symptom Manage , vol.25 , pp. 74-91
    • Andersen, G.1    Christrup, L.2    Sjogren, P.3
  • 5
    • 33645509700 scopus 로고    scopus 로고
    • Opioid-induced hyperalgesia: a qualitative systematic review
    • Angst, M.S., Clark, J.D., Opioid-induced hyperalgesia: a qualitative systematic review. Anesthesiology 104 (2006), 570–587.
    • (2006) Anesthesiology , vol.104 , pp. 570-587
    • Angst, M.S.1    Clark, J.D.2
  • 6
    • 55749090067 scopus 로고    scopus 로고
    • Numbers, densities, and colocalization of AMPA- and NMDA-type glutamate receptors at individual synapses in the superficial spinal dorsal horn of rats
    • Antal, M., Fukazawa, Y., Eordogh, M., Muszil, D., Molnar, E., Itakura, M., Takahashi, M., Shigemoto, R., Numbers, densities, and colocalization of AMPA- and NMDA-type glutamate receptors at individual synapses in the superficial spinal dorsal horn of rats. J Neurosci 28 (2008), 9692–9701.
    • (2008) J Neurosci , vol.28 , pp. 9692-9701
    • Antal, M.1    Fukazawa, Y.2    Eordogh, M.3    Muszil, D.4    Molnar, E.5    Itakura, M.6    Takahashi, M.7    Shigemoto, R.8
  • 7
    • 84941247747 scopus 로고    scopus 로고
    • Repeated mu-opioid exposure induces a novel form of the hyperalgesic priming model for transition to chronic pain
    • Araldi, D., Ferrari, L.F., Levine, J.D., Repeated mu-opioid exposure induces a novel form of the hyperalgesic priming model for transition to chronic pain. J Neurosci 35 (2015), 12502–12517.
    • (2015) J Neurosci , vol.35 , pp. 12502-12517
    • Araldi, D.1    Ferrari, L.F.2    Levine, J.D.3
  • 8
    • 84897413716 scopus 로고    scopus 로고
    • C-Fos activation in the periaqueductal gray following acute morphine-3beta-D-glucuronide or morphine administration
    • Arout, C.A., Caldwell, M., McCloskey, D.P., Kest, B., C-Fos activation in the periaqueductal gray following acute morphine-3beta-D-glucuronide or morphine administration. Physiol Behav 130 (2014), 28–33.
    • (2014) Physiol Behav , vol.130 , pp. 28-33
    • Arout, C.A.1    Caldwell, M.2    McCloskey, D.P.3    Kest, B.4
  • 9
    • 84929617004 scopus 로고    scopus 로고
    • Spinal and supraspinal N-methyl-D-aspartate and melanocortin-1 receptors contribute to a qualitative sex difference in morphine-induced hyperalgesia
    • Arout, C.A., Caldwell, M., Rossi, G., Kest, B., Spinal and supraspinal N-methyl-D-aspartate and melanocortin-1 receptors contribute to a qualitative sex difference in morphine-induced hyperalgesia. Physiol Behav 147 (2015), 364–372.
    • (2015) Physiol Behav , vol.147 , pp. 364-372
    • Arout, C.A.1    Caldwell, M.2    Rossi, G.3    Kest, B.4
  • 10
    • 84938082269 scopus 로고    scopus 로고
    • Targeting opioid-induced hyperalgesia in clinical treatment: neurobiological considerations
    • Arout, C.A., Edens, E., Petrakis, I.L., Sofuoglu, M., Targeting opioid-induced hyperalgesia in clinical treatment: neurobiological considerations. CNS Drugs 29 (2015), 465–486.
    • (2015) CNS Drugs , vol.29 , pp. 465-486
    • Arout, C.A.1    Edens, E.2    Petrakis, I.L.3    Sofuoglu, M.4
  • 12
    • 84914160082 scopus 로고    scopus 로고
    • Toll-like receptor 4-mediated nuclear factor-kappaB activation in spinal cord contributes to chronic morphine-induced analgesic tolerance and hyperalgesia in rats
    • Bai, L., Zhai, C., Han, K., Li, Z., Qian, J., Jing, Y., Zhang, W., Xu, J.T., Toll-like receptor 4-mediated nuclear factor-kappaB activation in spinal cord contributes to chronic morphine-induced analgesic tolerance and hyperalgesia in rats. Neurosci Bull 30 (2014), 936–948.
    • (2014) Neurosci Bull , vol.30 , pp. 936-948
    • Bai, L.1    Zhai, C.2    Han, K.3    Li, Z.4    Qian, J.5    Jing, Y.6    Zhang, W.7    Xu, J.T.8
  • 13
    • 79959523287 scopus 로고    scopus 로고
    • Pregabalin suppresses spinal neuronal hyperexcitability and visceral hypersensitivity in the absence of peripheral pathophysiology
    • Bannister, K., Sikandar, S., Bauer, C.S., Dolphin, A.C., Porreca, F., Dickenson, A.H., Pregabalin suppresses spinal neuronal hyperexcitability and visceral hypersensitivity in the absence of peripheral pathophysiology. Anesthesiology 115 (2011), 144–152.
    • (2011) Anesthesiology , vol.115 , pp. 144-152
    • Bannister, K.1    Sikandar, S.2    Bauer, C.S.3    Dolphin, A.C.4    Porreca, F.5    Dickenson, A.H.6
  • 14
    • 70349845440 scopus 로고    scopus 로고
    • Cellular and molecular mechanisms of pain
    • Basbaum, A.I., Bautista, D.M., Scherrer, G., Julius, D., Cellular and molecular mechanisms of pain. Cell 139 (2009), 267–284.
    • (2009) Cell , vol.139 , pp. 267-284
    • Basbaum, A.I.1    Bautista, D.M.2    Scherrer, G.3    Julius, D.4
  • 15
    • 84925765046 scopus 로고    scopus 로고
    • Endogenous regulation of inflammatory pain by T-cell-derived opioids: when friend turns to foe
    • Basso, L., Boue, J., Bourreille, A., Dietrich, G., Endogenous regulation of inflammatory pain by T-cell-derived opioids: when friend turns to foe. Inflamm Bowel Dis 20 (2014), 1870–1877.
    • (2014) Inflamm Bowel Dis , vol.20 , pp. 1870-1877
    • Basso, L.1    Boue, J.2    Bourreille, A.3    Dietrich, G.4
  • 17
    • 84926409714 scopus 로고    scopus 로고
    • Implication of delta opioid receptor subtype 2 but not delta opioid receptor subtype 1 in the development of morphine analgesic tolerance in a rat model of chronic inflammatory pain
    • Beaudry, H., Gendron, L., Moron, J.A., Implication of delta opioid receptor subtype 2 but not delta opioid receptor subtype 1 in the development of morphine analgesic tolerance in a rat model of chronic inflammatory pain. Eur J Neurosci 41 (2015), 901–907.
    • (2015) Eur J Neurosci , vol.41 , pp. 901-907
    • Beaudry, H.1    Gendron, L.2    Moron, J.A.3
  • 18
    • 26444578942 scopus 로고    scopus 로고
    • Low dose of S+-ketamine prevents long-term potentiation in pain pathways under strong opioid analgesia in the rat spinal cord in vivo
    • Benrath, J., Brechtel, C., Stark, J., Sandkuhler, J., Low dose of S+-ketamine prevents long-term potentiation in pain pathways under strong opioid analgesia in the rat spinal cord in vivo. Br J Anaesth 95 (2005), 518–523.
    • (2005) Br J Anaesth , vol.95 , pp. 518-523
    • Benrath, J.1    Brechtel, C.2    Stark, J.3    Sandkuhler, J.4
  • 19
    • 84862826216 scopus 로고    scopus 로고
    • Acute morphine activates satellite glial cells and up-regulates IL-1beta in dorsal root ganglia in mice via matrix metalloprotease-9
    • Berta, T., Liu, T., Liu, Y.C., Xu, Z.Z., Ji, R.R., 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, 18.
    • (2012) Mol Pain , vol.8 , pp. 18
    • Berta, T.1    Liu, T.2    Liu, Y.C.3    Xu, Z.Z.4    Ji, R.R.5
  • 20
    • 84880623225 scopus 로고    scopus 로고
    • Tissue plasminogen activator contributes to morphine tolerance and induces mechanical allodynia via astrocytic IL-1beta and ERK signaling in the spinal cord of mice
    • Berta, T., Liu, Y.C., Xu, Z.Z., Ji, R.R., Tissue plasminogen activator contributes to morphine tolerance and induces mechanical allodynia via astrocytic IL-1beta and ERK signaling in the spinal cord of mice. Neuroscience 247 (2013), 376–385.
    • (2013) Neuroscience , vol.247 , pp. 376-385
    • Berta, T.1    Liu, Y.C.2    Xu, Z.Z.3    Ji, R.R.4
  • 21
    • 0025307494 scopus 로고
    • Pre- and postsynaptic distribution of mu, delta and kappa opioid receptors in the superficial layers of the cervical dorsal horn of the rat spinal cord
    • Besse, D., Lombard, M.C., Zajac, J.M., Roques, B.P., Besson, J.M., Pre- and postsynaptic distribution of mu, delta and kappa opioid receptors in the superficial layers of the cervical dorsal horn of the rat spinal cord. Brain Res 521 (1990), 15–22.
    • (1990) Brain Res , vol.521 , pp. 15-22
    • Besse, D.1    Lombard, M.C.2    Zajac, J.M.3    Roques, B.P.4    Besson, J.M.5
  • 22
    • 84855723461 scopus 로고    scopus 로고
    • Contribution of G inhibitory protein alpha subunits in paradoxical hyperalgesia elicited by exceedingly low doses of morphine in mice
    • Bianchi, E., Galeotti, N., Menicacci, C., Ghelardini, C., Contribution of G inhibitory protein alpha subunits in paradoxical hyperalgesia elicited by exceedingly low doses of morphine in mice. Life Sci 89 (2011), 918–925.
    • (2011) Life Sci , vol.89 , pp. 918-925
    • Bianchi, E.1    Galeotti, N.2    Menicacci, C.3    Ghelardini, C.4
  • 23
    • 70349205440 scopus 로고    scopus 로고
    • Supraspinal Gbetagamma-dependent stimulation of PLCbeta originating from G inhibitory protein-mu opioid receptor-coupling is necessary for morphine induced acute hyperalgesia
    • Bianchi, E., Norcini, M., Smrcka, A., Ghelardini, C., Supraspinal Gbetagamma-dependent stimulation of PLCbeta originating from G inhibitory protein-mu opioid receptor-coupling is necessary for morphine induced acute hyperalgesia. J Neurochem 111 (2009), 171–180.
    • (2009) J Neurochem , vol.111 , pp. 171-180
    • Bianchi, E.1    Norcini, M.2    Smrcka, A.3    Ghelardini, C.4
  • 25
    • 77953479985 scopus 로고    scopus 로고
    • Sex differences in opioid analgesia, hyperalgesia, tolerance and withdrawal: central mechanisms of action and roles of gonadal hormones
    • Bodnar, R.J., Kest, B., Sex differences in opioid analgesia, hyperalgesia, tolerance and withdrawal: central mechanisms of action and roles of gonadal hormones. Horm Behav 58 (2010), 72–81.
    • (2010) Horm Behav , vol.58 , pp. 72-81
    • Bodnar, R.J.1    Kest, B.2
  • 26
    • 0034619796 scopus 로고    scopus 로고
    • Mu-opioid receptor desensitization by beta-arrestin-2 determines morphine tolerance but not dependence
    • Bohn, L.M., Gainetdinov, R.R., Lin, F.T., Lefkowitz, R.J., Caron, M.G., Mu-opioid receptor desensitization by beta-arrestin-2 determines morphine tolerance but not dependence. Nature 408 (2000), 720–723.
    • (2000) Nature , vol.408 , pp. 720-723
    • Bohn, L.M.1    Gainetdinov, R.R.2    Lin, F.T.3    Lefkowitz, R.J.4    Caron, M.G.5
  • 27
    • 33645231857 scopus 로고    scopus 로고
    • Survey of chronic pain in Europe: prevalence, impact on daily life, and treatment
    • Breivik, H., Collett, B., Ventafridda, V., Cohen, R., Gallacher, D., Survey of chronic pain in Europe: prevalence, impact on daily life, and treatment. Eur J Pain 10 (2006), 287–333.
    • (2006) Eur J Pain , vol.10 , pp. 287-333
    • Breivik, H.1    Collett, B.2    Ventafridda, V.3    Cohen, R.4    Gallacher, D.5
  • 28
    • 84890852892 scopus 로고    scopus 로고
    • The individual and societal burden of chronic pain in Europe: the case for strategic prioritisation and action to improve knowledge and availability of appropriate care
    • Breivik, H., Eisenberg, E., O'Brien, T., The individual and societal burden of chronic pain in Europe: the case for strategic prioritisation and action to improve knowledge and availability of appropriate care. BMC Public Health, 13, 2013, 1229.
    • (2013) BMC Public Health , vol.13 , pp. 1229
    • Breivik, H.1    Eisenberg, E.2    O'Brien, T.3
  • 30
    • 84862334333 scopus 로고    scopus 로고
    • The role of the immune system in the generation of neuropathic pain
    • Calvo, M., Dawes, J.M., Bennett, D.L., The role of the immune system in the generation of neuropathic pain. Lancet Neurol 11 (2012), 629–642.
    • (2012) Lancet Neurol , vol.11 , pp. 629-642
    • Calvo, M.1    Dawes, J.M.2    Bennett, D.L.3
  • 33
    • 0345035480 scopus 로고    scopus 로고
    • Evidence for opiate-activated NMDA processes masking opiate analgesia in rats
    • Celerier, E., Laulin, J., Larcher, A., Le Moal, M., Simonnet, G., Evidence for opiate-activated NMDA processes masking opiate analgesia in rats. Brain Res 847 (1999), 18–25.
    • (1999) Brain Res , vol.847 , pp. 18-25
    • Celerier, E.1    Laulin, J.2    Larcher, A.3    Le Moal, M.4    Simonnet, G.5
  • 34
    • 0035370642 scopus 로고    scopus 로고
    • Progressive enhancement of delayed hyperalgesia induced by repeated heroin administration: a sensitization process
    • Celerier, E., Laulin, J.P., Corcuff, J.B., Le Moal, M., Simonnet, G., Progressive enhancement of delayed hyperalgesia induced by repeated heroin administration: a sensitization process. J Neurosci 21 (2001), 4074–4080.
    • (2001) J Neurosci , vol.21 , pp. 4074-4080
    • Celerier, E.1    Laulin, J.P.2    Corcuff, J.B.3    Le Moal, M.4    Simonnet, G.5
  • 36
    • 0347480442 scopus 로고    scopus 로고
    • Prevention of fentanyl-induced delayed pronociceptive effects in mice lacking the protein kinase Cgamma gene
    • Celerier, E., Simonnet, G., Maldonado, R., Prevention of fentanyl-induced delayed pronociceptive effects in mice lacking the protein kinase Cgamma gene. Neuropharmacology 46 (2004), 264–272.
    • (2004) Neuropharmacology , vol.46 , pp. 264-272
    • Celerier, E.1    Simonnet, G.2    Maldonado, R.3
  • 38
    • 84962511720 scopus 로고    scopus 로고
    • Demethylation regulation of BDNF gene expression in dorsal root ganglion neurons is implicated in opioid-induced pain hypersensitivity in rats
    • Chao, Y.C., Xie, F., Li, X., Guo, R., Yang, N., Zhang, C., Shi, R., Guan, Y., Yue, Y., Wang, Y., Demethylation regulation of BDNF gene expression in dorsal root ganglion neurons is implicated in opioid-induced pain hypersensitivity in rats. Neurochem Int, 2016.
    • (2016) Neurochem Int
    • Chao, Y.C.1    Xie, F.2    Li, X.3    Guo, R.4    Yang, N.5    Zhang, C.6    Shi, R.7    Guan, Y.8    Yue, Y.9    Wang, Y.10
  • 39
    • 0025895334 scopus 로고
    • Sustained potentiation of NMDA receptor-mediated glutamate responses through activation of protein kinase C by a mu opioid
    • Chen, L., Huang, L.Y., Sustained potentiation of NMDA receptor-mediated glutamate responses through activation of protein kinase C by a mu opioid. Neuron 7 (1991), 319–326.
    • (1991) Neuron , vol.7 , pp. 319-326
    • Chen, L.1    Huang, L.Y.2
  • 41
    • 45949105581 scopus 로고    scopus 로고
    • Activation of TRPV1 contributes to morphine tolerance: involvement of the mitogen-activated protein kinase signaling pathway
    • Chen, Y., Geis, C., Sommer, C., Activation of TRPV1 contributes to morphine tolerance: involvement of the mitogen-activated protein kinase signaling pathway. J Neurosci 28 (2008), 5836–5845.
    • (2008) J Neurosci , vol.28 , pp. 5836-5845
    • Chen, Y.1    Geis, C.2    Sommer, C.3
  • 42
    • 49649102696 scopus 로고    scopus 로고
    • Opioid-induced hyperalgesia in humans: molecular mechanisms and clinical considerations
    • Chu, L.F., Angst, M.S., Clark, D., Opioid-induced hyperalgesia in humans: molecular mechanisms and clinical considerations. Clin J Pain 24 (2008), 479–496.
    • (2008) Clin J Pain , vol.24 , pp. 479-496
    • Chu, L.F.1    Angst, M.S.2    Clark, D.3
  • 43
    • 30744442773 scopus 로고    scopus 로고
    • Opioid tolerance and hyperalgesia in chronic pain patients after one month of oral morphine therapy: a preliminary prospective study
    • Chu, L.F., Clark, D.J., Angst, M.S., Opioid tolerance and hyperalgesia in chronic pain patients after one month of oral morphine therapy: a preliminary prospective study. J Pain 7 (2006), 43–48.
    • (2006) J Pain , vol.7 , pp. 43-48
    • Chu, L.F.1    Clark, D.J.2    Angst, M.S.3
  • 44
    • 84863717526 scopus 로고    scopus 로고
    • Analgesic tolerance without demonstrable opioid-induced hyperalgesia: a double-blinded, randomized, placebo-controlled trial of sustained-release morphine for treatment of chronic nonradicular low-back pain
    • Chu, L.F., D'Arcy, N., Brady, C., Zamora, A.K., Young, C.A., Kim, J.E., Clemenson, A.M., Angst, M.S., Clark, J.D., Analgesic tolerance without demonstrable opioid-induced hyperalgesia: a double-blinded, randomized, placebo-controlled trial of sustained-release morphine for treatment of chronic nonradicular low-back pain. Pain 153 (2012), 1583–1592.
    • (2012) Pain , vol.153 , pp. 1583-1592
    • Chu, L.F.1    D'Arcy, N.2    Brady, C.3    Zamora, A.K.4    Young, C.A.5    Kim, J.E.6    Clemenson, A.M.7    Angst, M.S.8    Clark, J.D.9
  • 45
    • 78650713562 scopus 로고    scopus 로고
    • The endogenous opioid system is not involved in modulation of opioid-induced hyperalgesia
    • Chu, L.F., Dairmont, J., Zamora, A.K., Young, C.A., Angst, M.S., The endogenous opioid system is not involved in modulation of opioid-induced hyperalgesia. J Pain 12 (2011), 108–115.
    • (2011) J Pain , vol.12 , pp. 108-115
    • Chu, L.F.1    Dairmont, J.2    Zamora, A.K.3    Young, C.A.4    Angst, M.S.5
  • 47
    • 84859421578 scopus 로고    scopus 로고
    • Hyperalgesia in heroin dependent patients and the effects of opioid substitution therapy
    • Compton, P., Canamar, C.P., Hillhouse, M., Ling, W., Hyperalgesia in heroin dependent patients and the effects of opioid substitution therapy. J Pain 13 (2012), 401–409.
    • (2012) J Pain , vol.13 , pp. 401-409
    • Compton, P.1    Canamar, C.P.2    Hillhouse, M.3    Ling, W.4
  • 48
    • 0035400798 scopus 로고    scopus 로고
    • Pain intolerance in opioid-maintained former opiate addicts: effect of long-acting maintenance agent
    • Compton, P., Charuvastra, V.C., Ling, W., Pain intolerance in opioid-maintained former opiate addicts: effect of long-acting maintenance agent. Drug Alcohol Depend 63 (2001), 139–146.
    • (2001) Drug Alcohol Depend , vol.63 , pp. 139-146
    • Compton, P.1    Charuvastra, V.C.2    Ling, W.3
  • 52
    • 0042860080 scopus 로고    scopus 로고
    • Trans-synaptic shift in anion gradient in spinal lamina I neurons as a mechanism of neuropathic pain
    • Coull, J.A., Boudreau, D., Bachand, K., Prescott, S.A., Nault, F., Sik, A., De Koninck, P., De Koninck, Y., Trans-synaptic shift in anion gradient in spinal lamina I neurons as a mechanism of neuropathic pain. Nature 424 (2003), 938–942.
    • (2003) Nature , vol.424 , pp. 938-942
    • Coull, J.A.1    Boudreau, D.2    Bachand, K.3    Prescott, S.A.4    Nault, F.5    Sik, A.6    De Koninck, P.7    De Koninck, Y.8
  • 53
    • 0346221268 scopus 로고    scopus 로고
    • Neuraminidase inhibitor, oseltamivir blocks GM1 ganglioside-regulated excitatory opioid receptor-mediated hyperalgesia, enhances opioid analgesia and attenuates tolerance in mice
    • Crain, S.M., Shen, K.F., Neuraminidase inhibitor, oseltamivir blocks GM1 ganglioside-regulated excitatory opioid receptor-mediated hyperalgesia, enhances opioid analgesia and attenuates tolerance in mice. Brain Res 995 (2004), 260–266.
    • (2004) Brain Res , vol.995 , pp. 260-266
    • Crain, S.M.1    Shen, K.F.2
  • 54
    • 77950604262 scopus 로고    scopus 로고
    • Adverse effects of chronic opioid therapy for chronic musculoskeletal pain
    • Crofford, L.J., Adverse effects of chronic opioid therapy for chronic musculoskeletal pain. Nat Rev Rheumatol 6 (2010), 191–197.
    • (2010) Nat Rev Rheumatol , vol.6 , pp. 191-197
    • Crofford, L.J.1
  • 55
    • 39649102928 scopus 로고    scopus 로고
    • Side effects of ketamine in the long-term treatment of neuropathic pain
    • Cvrcek, P., Side effects of ketamine in the long-term treatment of neuropathic pain. Pain Med 9 (2008), 253–257.
    • (2008) Pain Med , vol.9 , pp. 253-257
    • Cvrcek, P.1
  • 56
    • 0033635736 scopus 로고    scopus 로고
    • EphB receptors interact with NMDA receptors and regulate excitatory synapse formation
    • Dalva, M.B., Takasu, M.A., Lin, M.Z., Shamah, S.M., Hu, L., Gale, N.W., Greenberg, M.E., EphB receptors interact with NMDA receptors and regulate excitatory synapse formation. Cell 103 (2000), 945–956.
    • (2000) Cell , vol.103 , pp. 945-956
    • Dalva, M.B.1    Takasu, M.A.2    Lin, M.Z.3    Shamah, S.M.4    Hu, L.5    Gale, N.W.6    Greenberg, M.E.7
  • 57
    • 84857476249 scopus 로고    scopus 로고
    • Mechanisms of rapid opioid receptor desensitization, resensitization and tolerance in brain neurons
    • Dang, V.C., Christie, M.J., Mechanisms of rapid opioid receptor desensitization, resensitization and tolerance in brain neurons. Br J Pharmacol 165 (2012), 1704–1716.
    • (2012) Br J Pharmacol , vol.165 , pp. 1704-1716
    • Dang, V.C.1    Christie, M.J.2
  • 58
    • 0032571156 scopus 로고    scopus 로고
    • Differential release of cholecystokinin by morphine in rat spinal cord
    • de Araujo, Lucas G., Alster, P., Brodin, E., Wiesenfeld-Hallin, Z., Differential release of cholecystokinin by morphine in rat spinal cord. Neurosci Lett 245 (1998), 13–16.
    • (1998) Neurosci Lett , vol.245 , pp. 13-16
    • de Araujo, L.G.1    Alster, P.2    Brodin, E.3    Wiesenfeld-Hallin, Z.4
  • 60
    • 33646043440 scopus 로고    scopus 로고
    • The tetrapartite synapse: path to CNS sensitization and chronic pain
    • De Leo, J.A., Tawfik, V.L., LaCroix-Fralish, M.L., The tetrapartite synapse: path to CNS sensitization and chronic pain. Pain 122 (2006), 17–21.
    • (2006) Pain , vol.122 , pp. 17-21
    • De Leo, J.A.1    Tawfik, V.L.2    LaCroix-Fralish, M.L.3
  • 61
    • 84893754701 scopus 로고    scopus 로고
    • Pain vulnerability: a neurobiological perspective
    • Denk, F., McMahon, S.B., Tracey, I., Pain vulnerability: a neurobiological perspective. Nat Neurosci 17 (2014), 192–200.
    • (2014) Nat Neurosci , vol.17 , pp. 192-200
    • Denk, F.1    McMahon, S.B.2    Tracey, I.3
  • 62
    • 0037561201 scopus 로고    scopus 로고
    • Normal sensitivity to acute pain, but increased inflammatory hyperalgesia in mice lacking the nociceptin precursor polypeptide or the nociceptin receptor
    • Depner, U.B., Reinscheid, R.K., Takeshima, H., Brune, K., Zeilhofer, H.U., Normal sensitivity to acute pain, but increased inflammatory hyperalgesia in mice lacking the nociceptin precursor polypeptide or the nociceptin receptor. Eur J Neurosci 17 (2003), 2381–2387.
    • (2003) Eur J Neurosci , vol.17 , pp. 2381-2387
    • Depner, U.B.1    Reinscheid, R.K.2    Takeshima, H.3    Brune, K.4    Zeilhofer, H.U.5
  • 64
    • 78650732061 scopus 로고    scopus 로고
    • Epigenetics in pain and analgesia: an imminent research field
    • Doehring, A., Geisslinger, G., Lotsch, J., Epigenetics in pain and analgesia: an imminent research field. Eur J Pain 15 (2011), 11–16.
    • (2011) Eur J Pain , vol.15 , pp. 11-16
    • Doehring, A.1    Geisslinger, G.2    Lotsch, J.3
  • 65
    • 84871639310 scopus 로고    scopus 로고
    • Chronic opioid use is associated with increased DNA methylation correlating with increased clinical pain
    • Doehring, A., Oertel, B.G., Sittl, R., Lotsch, J., Chronic opioid use is associated with increased DNA methylation correlating with increased clinical pain. Pain 154 (2013), 15–23.
    • (2013) Pain , vol.154 , pp. 15-23
    • Doehring, A.1    Oertel, B.G.2    Sittl, R.3    Lotsch, J.4
  • 66
    • 67649658025 scopus 로고    scopus 로고
    • Differential mediation of descending pain facilitation and inhibition by spinal 5HT-3 and 5HT-7 receptors
    • Dogrul, A., Ossipov, M.H., Porreca, F., Differential mediation of descending pain facilitation and inhibition by spinal 5HT-3 and 5HT-7 receptors. Brain Res 1280 (2009), 52–59.
    • (2009) Brain Res , vol.1280 , pp. 52-59
    • Dogrul, A.1    Ossipov, M.H.2    Porreca, F.3
  • 68
    • 29244468283 scopus 로고    scopus 로고
    • Evidence of a role for descending serotonergic facilitation in a rat model of cancer-induced bone pain
    • Donovan-Rodriguez, T., Urch, C.E., Dickenson, A.H., Evidence of a role for descending serotonergic facilitation in a rat model of cancer-induced bone pain. Neurosci Lett 393 (2006), 237–242.
    • (2006) Neurosci Lett , vol.393 , pp. 237-242
    • Donovan-Rodriguez, T.1    Urch, C.E.2    Dickenson, A.H.3
  • 69
    • 0025641576 scopus 로고
    • The cholecystokinin receptor antagonist devazepide enhances morphine-induced analgesia but not morphine-induced respiratory depression in the squirrel monkey
    • Dourish, C.T., O'Neill, M.F., Schaffer, L.W., Siegl, P.K., Iversen, S.D., The cholecystokinin receptor antagonist devazepide enhances morphine-induced analgesia but not morphine-induced respiratory depression in the squirrel monkey. J Pharmacol Exp Ther 255 (1990), 1158–1165.
    • (1990) J Pharmacol Exp Ther , vol.255 , pp. 1158-1165
    • Dourish, C.T.1    O'Neill, M.F.2    Schaffer, L.W.3    Siegl, P.K.4    Iversen, S.D.5
  • 70
    • 67650410076 scopus 로고    scopus 로고
    • Induction of synaptic long-term potentiation after opioid withdrawal
    • Drdla, R., Gassner, M., Gingl, E., Sandkuhler, J., Induction of synaptic long-term potentiation after opioid withdrawal. Science (New York, NY) 325 (2009), 207–210.
    • (2009) Science (New York, NY) , vol.325 , pp. 207-210
    • Drdla, R.1    Gassner, M.2    Gingl, E.3    Sandkuhler, J.4
  • 73
    • 0029040497 scopus 로고
    • The effect of intrinsic efficacy on opioid tolerance
    • Duttaroy, A., Yoburn, B.C., The effect of intrinsic efficacy on opioid tolerance. Anesthesiology 82 (1995), 1226–1236.
    • (1995) Anesthesiology , vol.82 , pp. 1226-1236
    • Duttaroy, A.1    Yoburn, B.C.2
  • 74
    • 84884913853 scopus 로고    scopus 로고
    • Blockade of Toll-like receptor 4 attenuates morphine tolerance and facilitates the pain relieving properties of morphine
    • Eidson, L.N., Murphy, A.Z., Blockade of Toll-like receptor 4 attenuates morphine tolerance and facilitates the pain relieving properties of morphine. J Neurosci 33 (2013), 15952–15963.
    • (2013) J Neurosci , vol.33 , pp. 15952-15963
    • Eidson, L.N.1    Murphy, A.Z.2
  • 75
    • 84884478870 scopus 로고    scopus 로고
    • Persistent peripheral inflammation attenuates morphine-induced periaqueductal gray glial cell activation and analgesic tolerance in the male rat
    • Eidson, L.N., Murphy, A.Z., Persistent peripheral inflammation attenuates morphine-induced periaqueductal gray glial cell activation and analgesic tolerance in the male rat. J Pain 14 (2013), 393–404.
    • (2013) J Pain , vol.14 , pp. 393-404
    • Eidson, L.N.1    Murphy, A.Z.2
  • 76
    • 84921628250 scopus 로고    scopus 로고
    • Assessment of morphine-induced hyperalgesia and analgesic tolerance in mice using thermal and mechanical nociceptive modalities
    • Elhabazi, K., Ayachi, S., Ilien, B., Simonin, F., Assessment of morphine-induced hyperalgesia and analgesic tolerance in mice using thermal and mechanical nociceptive modalities. J Vis Exp: JoVE, 2014, e51264.
    • (2014) J Vis Exp: JoVE , pp. e51264
    • Elhabazi, K.1    Ayachi, S.2    Ilien, B.3    Simonin, F.4
  • 78
    • 84949309174 scopus 로고    scopus 로고
    • Impact of chronic morphine on delta opioid receptor-expressing neurons in the mouse hippocampus
    • Erbs, E., Faget, L., Ceredig, R.A., Matifas, A., Vonesch, J.L., Kieffer, B.L., Massotte, D., Impact of chronic morphine on delta opioid receptor-expressing neurons in the mouse hippocampus. Neuroscience 313 (2016), 46–56.
    • (2016) Neuroscience , vol.313 , pp. 46-56
    • Erbs, E.1    Faget, L.2    Ceredig, R.A.3    Matifas, A.4    Vonesch, J.L.5    Kieffer, B.L.6    Massotte, D.7
  • 79
    • 0020681585 scopus 로고
    • Evidence for the neuropeptide cholecystokinin as an antagonist of opiate analgesia
    • Faris, P.L., Komisaruk, B.R., Watkins, L.R., Mayer, D.J., Evidence for the neuropeptide cholecystokinin as an antagonist of opiate analgesia. Science (New York, NY) 219 (1983), 310–312.
    • (1983) Science (New York, NY) , vol.219 , pp. 310-312
    • Faris, P.L.1    Komisaruk, B.R.2    Watkins, L.R.3    Mayer, D.J.4
  • 81
    • 68349127240 scopus 로고    scopus 로고
    • Do opioids induce hyperalgesia in humans? An evidence-based structured review
    • Fishbain, D.A., Cole, B., Lewis, J.E., Gao, J., Rosomoff, R.S., Do opioids induce hyperalgesia in humans? An evidence-based structured review. Pain Med 10 (2009), 829–839.
    • (2009) Pain Med , vol.10 , pp. 829-839
    • Fishbain, D.A.1    Cole, B.2    Lewis, J.E.3    Gao, J.4    Rosomoff, R.S.5
  • 82
    • 84901052663 scopus 로고    scopus 로고
    • Opioid-induced hyperalgesia in patients after surgery: a systematic review and a meta-analysis
    • Fletcher, D., Martinez, V., Opioid-induced hyperalgesia in patients after surgery: a systematic review and a meta-analysis. Br J Anaesth 112 (2014), 991–1004.
    • (2014) Br J Anaesth , vol.112 , pp. 991-1004
    • Fletcher, D.1    Martinez, V.2
  • 83
    • 0030029425 scopus 로고    scopus 로고
    • Effects of memantine and MK-801 on NMDA-induced currents in cultured neurones and on synaptic transmission and LTP in area CA1 of rat hippocampal slices
    • Frankiewicz, T., Potier, B., Bashir, Z.I., Collingridge, G.L., Parsons, C.G., Effects of memantine and MK-801 on NMDA-induced currents in cultured neurones and on synaptic transmission and LTP in area CA1 of rat hippocampal slices. Br J Pharmacol 117 (1996), 689–697.
    • (1996) Br J Pharmacol , vol.117 , pp. 689-697
    • Frankiewicz, T.1    Potier, B.2    Bashir, Z.I.3    Collingridge, G.L.4    Parsons, C.G.5
  • 84
    • 79955467534 scopus 로고    scopus 로고
    • Distinct pharmacological properties of morphine metabolites at G(i)-protein and beta-arrestin signaling pathways activated by the human mu-opioid receptor
    • Frolich, N., Dees, C., Paetz, C., Ren, X., Lohse, M.J., Nikolaev, V.O., Zenk, M.H., Distinct pharmacological properties of morphine metabolites at G(i)-protein and beta-arrestin signaling pathways activated by the human mu-opioid receptor. Biochem Pharmacol 81 (2011), 1248–1254.
    • (2011) Biochem Pharmacol , vol.81 , pp. 1248-1254
    • Frolich, N.1    Dees, C.2    Paetz, C.3    Ren, X.4    Lohse, M.J.5    Nikolaev, V.O.6    Zenk, M.H.7
  • 85
    • 84920141912 scopus 로고    scopus 로고
    • Heteromers of mu-delta opioid receptors: new pharmacology and novel therapeutic possibilities
    • Fujita, W., Gomes, I., Devi, L.A., Heteromers of mu-delta opioid receptors: new pharmacology and novel therapeutic possibilities. Br J Pharmacol 172 (2015), 375–387.
    • (2015) Br J Pharmacol , vol.172 , pp. 375-387
    • Fujita, W.1    Gomes, I.2    Devi, L.A.3
  • 86
    • 84874112399 scopus 로고    scopus 로고
    • Microglial activation involved in morphine tolerance is not mediated by toll-like receptor 4
    • Fukagawa, H., Koyama, T., Kakuyama, M., Fukuda, K., Microglial activation involved in morphine tolerance is not mediated by toll-like receptor 4. J Anesth 27 (2013), 93–97.
    • (2013) J Anesth , vol.27 , pp. 93-97
    • Fukagawa, H.1    Koyama, T.2    Kakuyama, M.3    Fukuda, K.4
  • 87
    • 33745700151 scopus 로고    scopus 로고
    • Signaling pathway of morphine induced acute thermal hyperalgesia in mice
    • Galeotti, N., Stefano, G.B., Guarna, M., Bianchi, E., Ghelardini, C., Signaling pathway of morphine induced acute thermal hyperalgesia in mice. Pain 123 (2006), 294–305.
    • (2006) Pain , vol.123 , pp. 294-305
    • Galeotti, N.1    Stefano, G.B.2    Guarna, M.3    Bianchi, E.4    Ghelardini, C.5
  • 88
    • 84884187234 scopus 로고    scopus 로고
    • Opiate-induced analgesia: contributions from mu, delta and kappa opioid receptors mouse mutants
    • Gaveriaux-Ruff, C., Opiate-induced analgesia: contributions from mu, delta and kappa opioid receptors mouse mutants. Curr Pharm Des 19 (2013), 7373–7381.
    • (2013) Curr Pharm Des , vol.19 , pp. 7373-7381
    • Gaveriaux-Ruff, C.1
  • 89
    • 84936093169 scopus 로고    scopus 로고
    • Pronociceptive and antinociceptive effects of buprenorphine in the spinal cord dorsal horn cover a dose range of four orders of magnitude
    • Gerhold, K.J., Drdla-Schutting, R., Honsek, S.D., Forsthuber, L., Sandkuhler, J., Pronociceptive and antinociceptive effects of buprenorphine in the spinal cord dorsal horn cover a dose range of four orders of magnitude. J Neurosci 35 (2015), 9580–9594.
    • (2015) J Neurosci , vol.35 , pp. 9580-9594
    • Gerhold, K.J.1    Drdla-Schutting, R.2    Honsek, S.D.3    Forsthuber, L.4    Sandkuhler, J.5
  • 91
    • 84953750711 scopus 로고    scopus 로고
    • GluN2B N-methyl-D-aspartate receptor and excitatory amino acid transporter 3 are upregulated in primary sensory neurons after 7 days of morphine administration in rats: implication for opiate-induced hyperalgesia
    • Gong, K., Bhargava, A., Jasmin, L., GluN2B N-methyl-D-aspartate receptor and excitatory amino acid transporter 3 are upregulated in primary sensory neurons after 7 days of morphine administration in rats: implication for opiate-induced hyperalgesia. Pain 157 (2016), 147–158.
    • (2016) Pain , vol.157 , pp. 147-158
    • Gong, K.1    Bhargava, A.2    Jasmin, L.3
  • 93
    • 84927911504 scopus 로고    scopus 로고
    • Opioid-induced central immune signaling: implications for opioid analgesia
    • Grace, P.M., Maier, S.F., Watkins, L.R., Opioid-induced central immune signaling: implications for opioid analgesia. Headache 55 (2015), 475–489.
    • (2015) Headache , vol.55 , pp. 475-489
    • Grace, P.M.1    Maier, S.F.2    Watkins, L.R.3
  • 95
    • 84875992702 scopus 로고    scopus 로고
    • Induction of thermal hyperalgesia and synaptic long-term potentiation in the spinal cord lamina I by TNF-alpha and IL-1beta is mediated by glial cells
    • Gruber-Schoffnegger, D., Drdla-Schutting, R., Honigsperger, C., Wunderbaldinger, G., Gassner, M., Sandkuhler, J., Induction of thermal hyperalgesia and synaptic long-term potentiation in the spinal cord lamina I by TNF-alpha and IL-1beta is mediated by glial cells. J Neurosci 33 (2013), 6540–6551.
    • (2013) J Neurosci , vol.33 , pp. 6540-6551
    • Gruber-Schoffnegger, D.1    Drdla-Schutting, R.2    Honigsperger, C.3    Wunderbaldinger, G.4    Gassner, M.5    Sandkuhler, J.6
  • 96
    • 84902957395 scopus 로고    scopus 로고
    • Spinal 5-HT3 receptors mediate descending facilitation and contribute to behavioral hypersensitivity via a reciprocal neuron-glial signaling cascade
    • Guo, W., Miyoshi, K., Dubner, R., Gu, M., Li, M., Liu, J., Yang, J., Zou, S., Ren, K., Noguchi, K., Wei, F., Spinal 5-HT3 receptors mediate descending facilitation and contribute to behavioral hypersensitivity via a reciprocal neuron-glial signaling cascade. Mol Pain, 10, 2014, 35.
    • (2014) Mol Pain , vol.10 , pp. 35
    • Guo, W.1    Miyoshi, K.2    Dubner, R.3    Gu, M.4    Li, M.5    Liu, J.6    Yang, J.7    Zou, S.8    Ren, K.9    Noguchi, K.10    Wei, F.11
  • 97
    • 0034941697 scopus 로고    scopus 로고
    • Morphine-induced in vivo release of spinal cholecystokinin is mediated by delta-opioid receptors–effect of peripheral axotomy
    • Gustafsson, H., Afrah, A.W., Stiller, C.O., Morphine-induced in vivo release of spinal cholecystokinin is mediated by delta-opioid receptors–effect of peripheral axotomy. J Neurochem 78 (2001), 55–63.
    • (2001) J Neurochem , vol.78 , pp. 55-63
    • Gustafsson, H.1    Afrah, A.W.2    Stiller, C.O.3
  • 98
    • 84958529382 scopus 로고    scopus 로고
    • The influence of race and gender on pain management: a systematic literature review
    • Hampton, S.B., Cavalier, J., Langford, R., The influence of race and gender on pain management: a systematic literature review. Pain Manag Nurs 16 (2015), 968–977.
    • (2015) Pain Manag Nurs , vol.16 , pp. 968-977
    • Hampton, S.B.1    Cavalier, J.2    Langford, R.3
  • 99
    • 84948845428 scopus 로고    scopus 로고
    • Further optimization and evaluation of bioavailable, mixed-efficacy mu-opioid receptor (MOR) agonists/delta-opioid receptor (DOR) antagonists: balancing MOR and DOR affinities
    • Harland, A.A., Yeomans, L., Griggs, N.W., Anand, J.P., Pogozheva, I.D., Jutkiewicz, E.M., Traynor, J.R., Mosberg, H.I., Further optimization and evaluation of bioavailable, mixed-efficacy mu-opioid receptor (MOR) agonists/delta-opioid receptor (DOR) antagonists: balancing MOR and DOR affinities. J Med Chem 58 (2015), 8952–8969.
    • (2015) J Med Chem , vol.58 , pp. 8952-8969
    • Harland, A.A.1    Yeomans, L.2    Griggs, N.W.3    Anand, J.P.4    Pogozheva, I.D.5    Jutkiewicz, E.M.6    Traynor, J.R.7    Mosberg, H.I.8
  • 100
    • 42249116012 scopus 로고    scopus 로고
    • Ketamine blocks enhancement of spinal long-term potentiation in chronic opioid treated rats
    • Haugan, F., Rygh, L.J., Tjolsen, A., Ketamine blocks enhancement of spinal long-term potentiation in chronic opioid treated rats. Acta Anaesthesiol Scand 52 (2008), 681–687.
    • (2008) Acta Anaesthesiol Scand , vol.52 , pp. 681-687
    • Haugan, F.1    Rygh, L.J.2    Tjolsen, A.3
  • 101
    • 4043171462 scopus 로고    scopus 로고
    • Upstream and downstream of mTOR
    • Hay, N., Sonenberg, N., Upstream and downstream of mTOR. Genes Dev 18 (2004), 1926–1945.
    • (2004) Genes Dev , vol.18 , pp. 1926-1945
    • Hay, N.1    Sonenberg, N.2
  • 102
    • 84955724633 scopus 로고    scopus 로고
    • Differential opioid tolerance and opioid-induced hyperalgesia: a clinical reality
    • Hayhurst, C.J., Durieux, M.E., Differential opioid tolerance and opioid-induced hyperalgesia: a clinical reality. Anesthesiology 124 (2016), 483–488.
    • (2016) Anesthesiology , vol.124 , pp. 483-488
    • Hayhurst, C.J.1    Durieux, M.E.2
  • 104
    • 78651440933 scopus 로고    scopus 로고
    • Interactions between chemokine and mu-opioid receptors: anatomical findings and electrophysiological studies in the rat periaqueductal grey
    • Heinisch, S., Palma, J., Kirby, L.G., Interactions between chemokine and mu-opioid receptors: anatomical findings and electrophysiological studies in the rat periaqueductal grey. Brain Behav Immun 25 (2011), 360–372.
    • (2011) Brain Behav Immun , vol.25 , pp. 360-372
    • Heinisch, S.1    Palma, J.2    Kirby, L.G.3
  • 105
  • 106
    • 84926464909 scopus 로고    scopus 로고
    • Hyperalgesia induced by low-dose opioid treatment before orthopaedic surgery: an observational case-control study
    • Hina, N., Fletcher, D., Poindessous-Jazat, F., Martinez, V., Hyperalgesia induced by low-dose opioid treatment before orthopaedic surgery: an observational case-control study. Eur J Anaesthesiol 32 (2015), 255–261.
    • (2015) Eur J Anaesthesiol , vol.32 , pp. 255-261
    • Hina, N.1    Fletcher, D.2    Poindessous-Jazat, F.3    Martinez, V.4
  • 107
    • 0028603435 scopus 로고
    • The CCK-B receptor antagonist Cl 988 reverses tolerance to morphine in rats
    • Hoffmann, O., Wiesenfeld-Hallin, Z., The CCK-B receptor antagonist Cl 988 reverses tolerance to morphine in rats. NeuroReport 5 (1994), 2565–2568.
    • (1994) NeuroReport , vol.5 , pp. 2565-2568
    • Hoffmann, O.1    Wiesenfeld-Hallin, Z.2
  • 108
    • 14044278005 scopus 로고    scopus 로고
    • Characterization of morphine-induced hyperalgesia in male and female rats
    • Holtman, J.R. Jr., Wala, E.P., Characterization of morphine-induced hyperalgesia in male and female rats. Pain 114 (2005), 62–70.
    • (2005) Pain , vol.114 , pp. 62-70
    • Holtman, J.R.1    Wala, E.P.2
  • 109
    • 34247528508 scopus 로고    scopus 로고
    • Characterization of the antinociceptive and pronociceptive effects of methadone in rats
    • Holtman, J.R. Jr., Wala, E.P., Characterization of the antinociceptive and pronociceptive effects of methadone in rats. Anesthesiology 106 (2007), 563–571.
    • (2007) Anesthesiology , vol.106 , pp. 563-571
    • Holtman, J.R.1    Wala, E.P.2
  • 110
    • 2942753778 scopus 로고    scopus 로고
    • Sex differences in systemic morphine analgesic tolerance following intrathecal morphine injections
    • Hopkins, E., Rossi, G., Kest, B., Sex differences in systemic morphine analgesic tolerance following intrathecal morphine injections. Brain Res 1014 (2004), 244–246.
    • (2004) Brain Res , vol.1014 , pp. 244-246
    • Hopkins, E.1    Rossi, G.2    Kest, B.3
  • 111
    • 59649089153 scopus 로고    scopus 로고
    • Morphine enhances microglial migration through modulation of P2X4 receptor signaling
    • Horvath, R.J., DeLeo, J.A., Morphine enhances microglial migration through modulation of P2X4 receptor signaling. J Neurosci 29 (2009), 998–1005.
    • (2009) J Neurosci , vol.29 , pp. 998-1005
    • Horvath, R.J.1    DeLeo, J.A.2
  • 112
    • 77954623109 scopus 로고    scopus 로고
    • Inhibition of microglial P2X4 receptors attenuates morphine tolerance, Iba1, GFAP and mu opioid receptor protein expression while enhancing perivascular microglial ED2
    • Horvath, R.J., Romero-Sandoval, E.A., De Leo, J.A., Inhibition of microglial P2X4 receptors attenuates morphine tolerance, Iba1, GFAP and mu opioid receptor protein expression while enhancing perivascular microglial ED2. Pain 150 (2010), 401–413.
    • (2010) Pain , vol.150 , pp. 401-413
    • Horvath, R.J.1    Romero-Sandoval, E.A.2    De Leo, J.A.3
  • 113
    • 0035049085 scopus 로고    scopus 로고
    • Changes in the expression of G protein-coupled receptor kinases and beta-arrestin 2 in rat brain during opioid tolerance and supersensitivity
    • Hurle, M.A., Changes in the expression of G protein-coupled receptor kinases and beta-arrestin 2 in rat brain during opioid tolerance and supersensitivity. J Neurochem 77 (2001), 486–492.
    • (2001) J Neurochem , vol.77 , pp. 486-492
    • Hurle, M.A.1
  • 114
    • 79960687433 scopus 로고    scopus 로고
    • Exploring the neuroimmunopharmacology of opioids: an integrative review of mechanisms of central immune signaling and their implications for opioid analgesia
    • Hutchinson, M.R., Shavit, Y., Grace, P.M., Rice, K.C., Maier, S.F., Watkins, L.R., Exploring the neuroimmunopharmacology of opioids: an integrative review of mechanisms of central immune signaling and their implications for opioid analgesia. Pharmacol Rev 63 (2011), 772–810.
    • (2011) Pharmacol Rev , vol.63 , pp. 772-810
    • Hutchinson, M.R.1    Shavit, Y.2    Grace, P.M.3    Rice, K.C.4    Maier, S.F.5    Watkins, L.R.6
  • 116
    • 84888637560 scopus 로고    scopus 로고
    • Glia and pain: is chronic pain a gliopathy?
    • Ji, R.R., Berta, T., Nedergaard, M., Glia and pain: is chronic pain a gliopathy?. Pain 154:Suppl 1 (2013), S10–S28.
    • (2013) Pain , vol.154 , pp. S10-S28
    • Ji, R.R.1    Berta, T.2    Nedergaard, M.3
  • 117
    • 84903814786 scopus 로고    scopus 로고
    • Emerging targets in neuroinflammation-driven chronic pain
    • Ji, R.R., Xu, Z.Z., Gao, Y.J., Emerging targets in neuroinflammation-driven chronic pain. Nat Rev Drug Discovery 13 (2014), 533–548.
    • (2014) Nat Rev Drug Discovery , vol.13 , pp. 533-548
    • Ji, R.R.1    Xu, Z.Z.2    Gao, Y.J.3
  • 118
    • 33646494493 scopus 로고    scopus 로고
    • Decreased morphine analgesia in rat overexpressing beta-arrestin 2 at periaqueductal gray
    • Jiang, B., Shi, Y., Li, H., Kang, L., Ma, L., Decreased morphine analgesia in rat overexpressing beta-arrestin 2 at periaqueductal gray. Neurosci Lett 400 (2006), 150–153.
    • (2006) Neurosci Lett , vol.400 , pp. 150-153
    • Jiang, B.1    Shi, Y.2    Li, H.3    Kang, L.4    Ma, L.5
  • 119
    • 84936953307 scopus 로고    scopus 로고
    • The optimal choice of medication administration route regarding intravenous, intramuscular, and subcutaneous injection
    • Jin, J.F., Zhu, L.L., Chen, M., Xu, H.M., Wang, H.F., Feng, X.Q., Zhu, X.P., Zhou, Q., The optimal choice of medication administration route regarding intravenous, intramuscular, and subcutaneous injection. Patient Prefer Adherence 9 (2015), 923–942.
    • (2015) Patient Prefer Adherence , vol.9 , pp. 923-942
    • Jin, J.F.1    Zhu, L.L.2    Chen, M.3    Xu, H.M.4    Wang, H.F.5    Feng, X.Q.6    Zhu, X.P.7    Zhou, Q.8
  • 120
    • 78149285273 scopus 로고    scopus 로고
    • The prevalence of chronic pain in United States adults: results of an Internet-based survey
    • Johannes, C.B., Le, T.K., Zhou, X., Johnston, J.A., Dworkin, R.H., The prevalence of chronic pain in United States adults: results of an Internet-based survey. J Pain 11 (2010), 1230–1239.
    • (2010) J Pain , vol.11 , pp. 1230-1239
    • Johannes, C.B.1    Le, T.K.2    Zhou, X.3    Johnston, J.A.4    Dworkin, R.H.5
  • 123
    • 84885827972 scopus 로고    scopus 로고
    • TRP channels and pain
    • Julius, D., TRP channels and pain. Annu Rev Cell Dev Biol 29 (2013), 355–384.
    • (2013) Annu Rev Cell Dev Biol , vol.29 , pp. 355-384
    • Julius, D.1
  • 125
    • 32644468581 scopus 로고    scopus 로고
    • Morphine hyperalgesia in mice is unrelated to opioid activity, analgesia, or tolerance: evidence for multiple diverse hyperalgesic systems
    • Juni, A., Klein, G., Kest, B., Morphine hyperalgesia in mice is unrelated to opioid activity, analgesia, or tolerance: evidence for multiple diverse hyperalgesic systems. Brain Res 1070 (2006), 35–44.
    • (2006) Brain Res , vol.1070 , pp. 35-44
    • Juni, A.1    Klein, G.2    Kest, B.3
  • 126
    • 43949130709 scopus 로고    scopus 로고
    • Sex differences in hyperalgesia during morphine infusion: effect of gonadectomy and estrogen treatment
    • Juni, A., Klein, G., Kowalczyk, B., Ragnauth, A., Kest, B., Sex differences in hyperalgesia during morphine infusion: effect of gonadectomy and estrogen treatment. Neuropharmacology 54 (2008), 1264–1270.
    • (2008) Neuropharmacology , vol.54 , pp. 1264-1270
    • Juni, A.1    Klein, G.2    Kowalczyk, B.3    Ragnauth, A.4    Kest, B.5
  • 127
    • 35948948698 scopus 로고    scopus 로고
    • Central administration of selective melanocortin 4 receptor antagonist HS014 prevents morphine tolerance and withdrawal hyperalgesia
    • Kalange, A.S., Kokare, D.M., Singru, P.S., Upadhya, M.A., Chopde, C.T., Subhedar, N.K., Central administration of selective melanocortin 4 receptor antagonist HS014 prevents morphine tolerance and withdrawal hyperalgesia. Brain Res 1181 (2007), 10–20.
    • (2007) Brain Res , vol.1181 , pp. 10-20
    • Kalange, A.S.1    Kokare, D.M.2    Singru, P.S.3    Upadhya, M.A.4    Chopde, C.T.5    Subhedar, N.K.6
  • 128
    • 33846296546 scopus 로고    scopus 로고
    • Effects of the NMDA-receptor antagonist ketamine on perceptual correlates of long-term potentiation within the nociceptive system
    • Klein, T., Magerl, W., Nickel, U., Hopf, H.C., Sandkuhler, J., Treede, R.D., Effects of the NMDA-receptor antagonist ketamine on perceptual correlates of long-term potentiation within the nociceptive system. Neuropharmacology 52 (2007), 655–661.
    • (2007) Neuropharmacology , vol.52 , pp. 655-661
    • Klein, T.1    Magerl, W.2    Nickel, U.3    Hopf, H.C.4    Sandkuhler, J.5    Treede, R.D.6
  • 129
    • 67650229961 scopus 로고    scopus 로고
    • Mechanism of allodynia evoked by intrathecal morphine-3-glucuronide in mice
    • Komatsu, T., Sakurada, S., Katsuyama, S., Sanai, K., Sakurada, T., Mechanism of allodynia evoked by intrathecal morphine-3-glucuronide in mice. Int Rev Neurobiol 85 (2009), 207–219.
    • (2009) Int Rev Neurobiol , vol.85 , pp. 207-219
    • Komatsu, T.1    Sakurada, S.2    Katsuyama, S.3    Sanai, K.4    Sakurada, T.5
  • 132
    • 0031937382 scopus 로고    scopus 로고
    • Acute tolerance associated with a single opiate administration: involvement of N-methyl-D-aspartate-dependent pain facilitatory systems
    • Larcher, A., Laulin, J.P., Celerier, E., Le Moal, M., Simonnet, G., Acute tolerance associated with a single opiate administration: involvement of N-methyl-D-aspartate-dependent pain facilitatory systems. Neuroscience 84 (1998), 583–589.
    • (1998) Neuroscience , vol.84 , pp. 583-589
    • Larcher, A.1    Laulin, J.P.2    Celerier, E.3    Le Moal, M.4    Simonnet, G.5
  • 133
    • 47549101405 scopus 로고    scopus 로고
    • Oxycodone plus ultra-low-dose naltrexone attenuates neuropathic pain and associated mu-opioid receptor-Gs coupling
    • Largent-Milnes, T.M., Guo, W., Wang, H.Y., Burns, L.H., Vanderah, T.W., Oxycodone plus ultra-low-dose naltrexone attenuates neuropathic pain and associated mu-opioid receptor-Gs coupling. J Pain 9 (2008), 700–713.
    • (2008) J Pain , vol.9 , pp. 700-713
    • Largent-Milnes, T.M.1    Guo, W.2    Wang, H.Y.3    Burns, L.H.4    Vanderah, T.W.5
  • 134
    • 84873173202 scopus 로고    scopus 로고
    • Endogenous morphine and its metabolites in mammals: history, synthesis, localization and perspectives
    • Laux-Biehlmann, A., Mouheiche, J., Veriepe, J., Goumon, Y., Endogenous morphine and its metabolites in mammals: history, synthesis, localization and perspectives. Neuroscience 233 (2013), 95–117.
    • (2013) Neuroscience , vol.233 , pp. 95-117
    • Laux-Biehlmann, A.1    Mouheiche, J.2    Veriepe, J.3    Goumon, Y.4
  • 135
    • 84878117310 scopus 로고    scopus 로고
    • Opioid receptors: toward separation of analgesic from undesirable effects
    • Law, P.Y., Reggio, P.H., Loh, H.H., Opioid receptors: toward separation of analgesic from undesirable effects. Trends Biochem Sci 38 (2013), 275–282.
    • (2013) Trends Biochem Sci , vol.38 , pp. 275-282
    • Law, P.Y.1    Reggio, P.H.2    Loh, H.H.3
  • 137
    • 71549118511 scopus 로고    scopus 로고
    • Evidence that intrathecal morphine-3-glucuronide may cause pain enhancement via toll-like receptor 4/MD-2 and interleukin-1beta
    • Lewis, S.S., Hutchinson, M.R., Rezvani, N., Loram, L.C., Zhang, Y., Maier, S.F., Rice, K.C., Watkins, L.R., Evidence that intrathecal morphine-3-glucuronide may cause pain enhancement via toll-like receptor 4/MD-2 and interleukin-1beta. Neuroscience 165 (2010), 569–583.
    • (2010) Neuroscience , vol.165 , pp. 569-583
    • Lewis, S.S.1    Hutchinson, M.R.2    Rezvani, N.3    Loram, L.C.4    Zhang, Y.5    Maier, S.F.6    Rice, K.C.7    Watkins, L.R.8
  • 138
    • 0035977714 scopus 로고    scopus 로고
    • A murine model of opioid-induced hyperalgesia
    • Li, X., Angst, M.S., Clark, J.D., A murine model of opioid-induced hyperalgesia. Brain Res Mol Brain Res 86 (2001), 56–62.
    • (2001) Brain Res Mol Brain Res , vol.86 , pp. 56-62
    • Li, X.1    Angst, M.S.2    Clark, J.D.3
  • 139
    • 41949126999 scopus 로고    scopus 로고
    • Chronic morphine administration enhances nociceptive sensitivity and local cytokine production after incision
    • Liang, D., Shi, X., Qiao, Y., Angst, M.S., Yeomans, D.C., Clark, J.D., Chronic morphine administration enhances nociceptive sensitivity and local cytokine production after incision. Mol Pain, 4, 2008, 7.
    • (2008) Mol Pain , vol.4 , pp. 7
    • Liang, D.1    Shi, X.2    Qiao, Y.3    Angst, M.S.4    Yeomans, D.C.5    Clark, J.D.6
  • 140
    • 79955484611 scopus 로고    scopus 로고
    • 5-Hydroxytryptamine type 3 receptor modulates opioid-induced hyperalgesia and tolerance in mice
    • Liang, D.Y., Li, X., Clark, J.D., 5-Hydroxytryptamine type 3 receptor modulates opioid-induced hyperalgesia and tolerance in mice. Anesthesiology 114 (2011), 1180–1189.
    • (2011) Anesthesiology , vol.114 , pp. 1180-1189
    • Liang, D.Y.1    Li, X.2    Clark, J.D.3
  • 141
    • 84871879281 scopus 로고    scopus 로고
    • Epigenetic regulation of opioid-induced hyperalgesia, dependence, and tolerance in mice
    • Liang, D.Y., Li, X., Clark, J.D., Epigenetic regulation of opioid-induced hyperalgesia, dependence, and tolerance in mice. J Pain 14 (2013), 36–47.
    • (2013) J Pain , vol.14 , pp. 36-47
    • Liang, D.Y.1    Li, X.2    Clark, J.D.3
  • 143
    • 84907048241 scopus 로고    scopus 로고
    • Epigenetic regulation of spinal cord gene expression controls opioid-induced hyperalgesia
    • Liang, D.Y., Sun, Y., Shi, X.Y., Sahbaie, P., Clark, J.D., Epigenetic regulation of spinal cord gene expression controls opioid-induced hyperalgesia. Mol Pain, 10, 2014, 59.
    • (2014) Mol Pain , vol.10 , pp. 59
    • Liang, D.Y.1    Sun, Y.2    Shi, X.Y.3    Sahbaie, P.4    Clark, J.D.5
  • 145
    • 84924297480 scopus 로고    scopus 로고
    • Role of spinal CXCL1 (GROalpha) in opioid tolerance: a human-to-rodent translational study
    • Lin, C.P., Kang, K.H., Lin, T.H., Wu, M.Y., Liou, H.C., Chuang, W.J., Sun, W.Z., Fu, W.M., Role of spinal CXCL1 (GROalpha) in opioid tolerance: a human-to-rodent translational study. Anesthesiology 122 (2015), 666–676.
    • (2015) Anesthesiology , vol.122 , pp. 666-676
    • Lin, C.P.1    Kang, K.H.2    Lin, T.H.3    Wu, M.Y.4    Liou, H.C.5    Chuang, W.J.6    Sun, W.Z.7    Fu, W.M.8
  • 146
    • 84947232208 scopus 로고    scopus 로고
    • Predicting neuroinflammation in morphine tolerance for tolerance therapy from immunostaining images of rat spinal cord
    • Lin, S.L., Chang, F.L., Ho, S.Y., Charoenkwan, P., Wang, K.W., Huang, H.L., Predicting neuroinflammation in morphine tolerance for tolerance therapy from immunostaining images of rat spinal cord. PLoS ONE, 10, 2015, e0139806.
    • (2015) PLoS ONE , vol.10 , pp. e0139806
    • Lin, S.L.1    Chang, F.L.2    Ho, S.Y.3    Charoenkwan, P.4    Wang, K.W.5    Huang, H.L.6
  • 147
    • 84878794004 scopus 로고    scopus 로고
    • Spinal mitochondrial-derived peroxynitrite enhances neuroimmune activation during morphine hyperalgesia and antinociceptive tolerance
    • Little, J.W., Cuzzocrea, S., Bryant, L., Esposito, E., Doyle, T., Rausaria, S., Neumann, W.L., Salvemini, D., Spinal mitochondrial-derived peroxynitrite enhances neuroimmune activation during morphine hyperalgesia and antinociceptive tolerance. Pain 154 (2013), 978–986.
    • (2013) Pain , vol.154 , pp. 978-986
    • Little, J.W.1    Cuzzocrea, S.2    Bryant, L.3    Esposito, E.4    Doyle, T.5    Rausaria, S.6    Neumann, W.L.7    Salvemini, D.8
  • 148
    • 1842372050 scopus 로고    scopus 로고
    • Characterization of long-term potentiation of C-fiber-evoked potentials in spinal dorsal horn of adult rat: essential role of NK1 and NK2 receptors
    • Liu, X., Sandkuhler, J., Characterization of long-term potentiation of C-fiber-evoked potentials in spinal dorsal horn of adult rat: essential role of NK1 and NK2 receptors. J Neurophysiol 78 (1997), 1973–1982.
    • (1997) J Neurophysiol , vol.78 , pp. 1973-1982
    • Liu, X.1    Sandkuhler, J.2
  • 149
    • 33749509564 scopus 로고    scopus 로고
    • Serotonergic modulation of spinal sensory circuits
    • Lopez-Garcia, J.A., Serotonergic modulation of spinal sensory circuits. Curr Top Med Chem 6 (2006), 1987–1996.
    • (2006) Curr Top Med Chem , vol.6 , pp. 1987-1996
    • Lopez-Garcia, J.A.1
  • 150
    • 67049098505 scopus 로고    scopus 로고
    • Pleiotropic effects of morphine-6beta-glucuronide
    • Lotsch, J., Pleiotropic effects of morphine-6beta-glucuronide. Anesthesiology 110 (2009), 1209–1210.
    • (2009) Anesthesiology , vol.110 , pp. 1209-1210
    • Lotsch, J.1
  • 151
    • 77954173921 scopus 로고    scopus 로고
    • A critical appraisal of human genotyping for pain therapy
    • Lotsch, J., Geisslinger, G., A critical appraisal of human genotyping for pain therapy. Trends Pharmacol Sci 31 (2010), 312–317.
    • (2010) Trends Pharmacol Sci , vol.31 , pp. 312-317
    • Lotsch, J.1    Geisslinger, G.2
  • 152
    • 84928361592 scopus 로고    scopus 로고
    • Neuron-restrictive silencer factor in periaqueductal gray contributes to remifentanil-induced postoperative hyperalgesia via repression of the mu-opioid receptor
    • Lu, C., Shi, L., Zhang, J., Kong, M., Liu, Y., Zhou, Y., Xu, L., He, J., Ma, Z., Gu, X., Neuron-restrictive silencer factor in periaqueductal gray contributes to remifentanil-induced postoperative hyperalgesia via repression of the mu-opioid receptor. J Neurol Sci 352 (2015), 48–52.
    • (2015) J Neurol Sci , vol.352 , pp. 48-52
    • Lu, C.1    Shi, L.2    Zhang, J.3    Kong, M.4    Liu, Y.5    Zhou, Y.6    Xu, L.7    He, J.8    Ma, Z.9    Gu, X.10
  • 153
    • 84952906981 scopus 로고    scopus 로고
    • CXCL12/CXCR4 axis: an emerging neuromodulator in pathological pain
    • Luo, X., Wang, X., Xia, Z., Chung, S.K., Cheung, C.W., CXCL12/CXCR4 axis: an emerging neuromodulator in pathological pain. Rev Neurosci 27 (2016), 83–92.
    • (2016) Rev Neurosci , vol.27 , pp. 83-92
    • Luo, X.1    Wang, X.2    Xia, Z.3    Chung, S.K.4    Cheung, C.W.5
  • 154
    • 84930669960 scopus 로고    scopus 로고
    • MTOR, a new potential target for chronic pain and opioid-induced tolerance and hyperalgesia
    • Lutz, B.M., Nia, S., Xiong, M., Tao, Y.X., Bekker, A., MTOR, a new potential target for chronic pain and opioid-induced tolerance and hyperalgesia. Mol Pain, 11, 2015, 32.
    • (2015) Mol Pain , vol.11 , pp. 32
    • Lutz, B.M.1    Nia, S.2    Xiong, M.3    Tao, Y.X.4    Bekker, A.5
  • 155
    • 79960015639 scopus 로고    scopus 로고
    • Control of neuropathic pain by immune cells and opioids
    • Machelska, H., Control of neuropathic pain by immune cells and opioids. CNS Neurol Disord: Drug Targets 10 (2011), 559–570.
    • (2011) CNS Neurol Disord: Drug Targets , vol.10 , pp. 559-570
    • Machelska, H.1
  • 156
    • 84920167257 scopus 로고    scopus 로고
    • Different mechanisms of homologous and heterologous mu-opioid receptor phosphorylation
    • Mann, A., Illing, S., Miess, E., Schulz, S., Different mechanisms of homologous and heterologous mu-opioid receptor phosphorylation. Br J Pharmacol 172 (2015), 311–316.
    • (2015) Br J Pharmacol , vol.172 , pp. 311-316
    • Mann, A.1    Illing, S.2    Miess, E.3    Schulz, S.4
  • 157
    • 0028215987 scopus 로고
    • Thermal hyperalgesia in association with the development of morphine tolerance in rats: roles of excitatory amino acid receptors and protein kinase C
    • Mao, J., Price, D.D., Mayer, D.J., Thermal hyperalgesia in association with the development of morphine tolerance in rats: roles of excitatory amino acid receptors and protein kinase C. J Neurosci 14 (1994), 2301–2312.
    • (1994) J Neurosci , vol.14 , pp. 2301-2312
    • Mao, J.1    Price, D.D.2    Mayer, D.J.3
  • 158
    • 0037107189 scopus 로고    scopus 로고
    • Chronic morphine induces downregulation of spinal glutamate transporters: implications in morphine tolerance and abnormal pain sensitivity
    • Mao, J., Sung, B., Ji, R.R., Lim, G., Chronic morphine induces downregulation of spinal glutamate transporters: implications in morphine tolerance and abnormal pain sensitivity. J Neurosci 22 (2002), 8312–8323.
    • (2002) J Neurosci , vol.22 , pp. 8312-8323
    • Mao, J.1    Sung, B.2    Ji, R.R.3    Lim, G.4
  • 159
    • 84961371041 scopus 로고    scopus 로고
    • Sex differences in pain: a tale of two immune cells
    • Mapplebeck, J.C., Beggs, S., Salter, M.W., Sex differences in pain: a tale of two immune cells. Pain 157:Suppl 1 (2016), S2–S6.
    • (2016) Pain , vol.157 , pp. S2-S6
    • Mapplebeck, J.C.1    Beggs, S.2    Salter, M.W.3
  • 160
    • 84920169231 scopus 로고    scopus 로고
    • In vivo opioid receptor heteromerization: where do we stand?
    • Massotte, D., In vivo opioid receptor heteromerization: where do we stand?. Br J Pharmacol 172 (2015), 420–434.
    • (2015) Br J Pharmacol , vol.172 , pp. 420-434
    • Massotte, D.1
  • 162
    • 84901287696 scopus 로고    scopus 로고
    • Toll-like receptor 4 mutant and null mice retain morphine-induced tolerance, hyperalgesia, and physical dependence
    • Mattioli, T.A., Leduc-Pessah, H., Skelhorne-Gross, G., Nicol, C.J., Milne, B., Trang, T., Cahill, C.M., Toll-like receptor 4 mutant and null mice retain morphine-induced tolerance, hyperalgesia, and physical dependence. PLoS ONE, 9, 2014, e97361.
    • (2014) PLoS ONE , vol.9 , pp. e97361
    • Mattioli, T.A.1    Leduc-Pessah, H.2    Skelhorne-Gross, G.3    Nicol, C.J.4    Milne, B.5    Trang, T.6    Cahill, C.M.7
  • 163
    • 84955713783 scopus 로고    scopus 로고
    • Does fentanyl lead to opioid-induced hyperalgesia in healthy volunteers?: A double-blind, randomized, crossover trial
    • Mauermann, E., Filitz, J., Dolder, P., Rentsch, K.M., Bandschapp, O., Ruppen, W., Does fentanyl lead to opioid-induced hyperalgesia in healthy volunteers?: A double-blind, randomized, crossover trial. Anesthesiology 124 (2016), 453–463.
    • (2016) Anesthesiology , vol.124 , pp. 453-463
    • Mauermann, E.1    Filitz, J.2    Dolder, P.3    Rentsch, K.M.4    Bandschapp, O.5    Ruppen, W.6
  • 164
    • 0033529232 scopus 로고    scopus 로고
    • Cellular mechanisms of neuropathic pain, morphine tolerance, and their interactions
    • Mayer, D.J., Mao, J., Holt, J., Price, D.D., Cellular mechanisms of neuropathic pain, morphine tolerance, and their interactions. Proc Natl Acad Sci U S A 96 (1999), 7731–7736.
    • (1999) Proc Natl Acad Sci U S A , vol.96 , pp. 7731-7736
    • Mayer, D.J.1    Mao, J.2    Holt, J.3    Price, D.D.4
  • 165
    • 0032742753 scopus 로고    scopus 로고
    • Pain facilitatory circuits in the mammalian central nervous system: their behavioral significance and role in morphine analgesic tolerance
    • McNally, G.P., Pain facilitatory circuits in the mammalian central nervous system: their behavioral significance and role in morphine analgesic tolerance. Neurosci Biobehav Rev 23 (1999), 1059–1078.
    • (1999) Neurosci Biobehav Rev , vol.23 , pp. 1059-1078
    • McNally, G.P.1
  • 167
  • 168
    • 84959508686 scopus 로고    scopus 로고
    • Opioid switching in cancer pain: from the beginning to nowadays
    • Mercadante, S., Bruera, E., Opioid switching in cancer pain: from the beginning to nowadays. Crit Rev Oncol/Hematol 99 (2016), 241–248.
    • (2016) Crit Rev Oncol/Hematol , vol.99 , pp. 241-248
    • Mercadante, S.1    Bruera, E.2
  • 169
    • 63249090831 scopus 로고    scopus 로고
    • Frequency, indications, outcomes, and predictive factors of opioid switching in an acute palliative care unit
    • Mercadante, S., Ferrera, P., Villari, P., Casuccio, A., Intravaia, G., Mangione, S., Frequency, indications, outcomes, and predictive factors of opioid switching in an acute palliative care unit. J Pain Symptom Manage 37 (2009), 632–641.
    • (2009) J Pain Symptom Manage , vol.37 , pp. 632-641
    • Mercadante, S.1    Ferrera, P.2    Villari, P.3    Casuccio, A.4    Intravaia, G.5    Mangione, S.6
  • 170
    • 84884906968 scopus 로고    scopus 로고
    • Morphine mediates a proinflammatory phenotype via mu-opioid receptor-PKCvarepsilon-Akt-ERK1/2 signaling pathway in activated microglial cells
    • Merighi, S., Gessi, S., Varani, K., Fazzi, D., Stefanelli, A., Borea, P.A., Morphine mediates a proinflammatory phenotype via mu-opioid receptor-PKCvarepsilon-Akt-ERK1/2 signaling pathway in activated microglial cells. Biochem Pharmacol 86 (2013), 487–496.
    • (2013) Biochem Pharmacol , vol.86 , pp. 487-496
    • Merighi, S.1    Gessi, S.2    Varani, K.3    Fazzi, D.4    Stefanelli, A.5    Borea, P.A.6
  • 172
    • 84874536688 scopus 로고    scopus 로고
    • Stereo-selective inhibition of spinal morphine tolerance and hyperalgesia by an ultra-low dose of the alpha-2-adrenoceptor antagonist efaroxan
    • Milne, B., Jhamandas, K., Sutak, M., Grenier, P., Cahill, C.M., Stereo-selective inhibition of spinal morphine tolerance and hyperalgesia by an ultra-low dose of the alpha-2-adrenoceptor antagonist efaroxan. Eur J Pharmacol 702 (2013), 227–234.
    • (2013) Eur J Pharmacol , vol.702 , pp. 227-234
    • Milne, B.1    Jhamandas, K.2    Sutak, M.3    Grenier, P.4    Cahill, C.M.5
  • 173
    • 57349111876 scopus 로고    scopus 로고
    • Low doses of alpha 2-adrenoceptor antagonists augment spinal morphine analgesia and inhibit development of acute and chronic tolerance
    • Milne, B., Sutak, M., Cahill, C.M., Jhamandas, K., Low doses of alpha 2-adrenoceptor antagonists augment spinal morphine analgesia and inhibit development of acute and chronic tolerance. Br J Pharmacol 155 (2008), 1264–1278.
    • (2008) Br J Pharmacol , vol.155 , pp. 1264-1278
    • Milne, B.1    Sutak, M.2    Cahill, C.M.3    Jhamandas, K.4
  • 174
    • 84872014582 scopus 로고    scopus 로고
    • Sex differences in pain and pain inhibition: multiple explanations of a controversial phenomenon
    • Mogil, J.S., Sex differences in pain and pain inhibition: multiple explanations of a controversial phenomenon. Nat Rev Neurosci 13 (2012), 859–866.
    • (2012) Nat Rev Neurosci , vol.13 , pp. 859-866
    • Mogil, J.S.1
  • 175
    • 84898438286 scopus 로고    scopus 로고
    • Development of a bioavailable mu opioid receptor (MOPr) agonist, delta opioid receptor (DOPr) antagonist peptide that evokes antinociception without development of acute tolerance
    • Mosberg, H.I., Yeomans, L., Anand, J.P., Porter, V., Sobczyk-Kojiro, K., Traynor, J.R., Jutkiewicz, E.M., Development of a bioavailable mu opioid receptor (MOPr) agonist, delta opioid receptor (DOPr) antagonist peptide that evokes antinociception without development of acute tolerance. J Med Chem 57 (2014), 3148–3153.
    • (2014) J Med Chem , vol.57 , pp. 3148-3153
    • Mosberg, H.I.1    Yeomans, L.2    Anand, J.P.3    Porter, V.4    Sobczyk-Kojiro, K.5    Traynor, J.R.6    Jutkiewicz, E.M.7
  • 176
    • 78650125304 scopus 로고    scopus 로고
    • Opioid-modulating properties of the neuropeptide FF system
    • Mouledous, L., Mollereau, C., Zajac, J.M., Opioid-modulating properties of the neuropeptide FF system. BioFactors 36 (2010), 423–429.
    • (2010) BioFactors , vol.36 , pp. 423-429
    • Mouledous, L.1    Mollereau, C.2    Zajac, J.M.3
  • 178
    • 84879238439 scopus 로고    scopus 로고
    • Role of the mu-opioid receptor in opioid modulation of immune function
    • Ninkovic, J., Roy, S., Role of the mu-opioid receptor in opioid modulation of immune function. Amino Acids 45 (2013), 9–24.
    • (2013) Amino Acids , vol.45 , pp. 9-24
    • Ninkovic, J.1    Roy, S.2
  • 179
    • 84867998179 scopus 로고    scopus 로고
    • Melanocortin 4 receptor antagonists attenuates morphine antinociceptive tolerance, astroglial activation and cytokines expression in the spinal cord of rat
    • Niu, Z., Ma, J., Chu, H., Zhao, Y., Feng, W., Cheng, Y., Melanocortin 4 receptor antagonists attenuates morphine antinociceptive tolerance, astroglial activation and cytokines expression in the spinal cord of rat. Neurosci Lett 529 (2012), 112–117.
    • (2012) Neurosci Lett , vol.529 , pp. 112-117
    • Niu, Z.1    Ma, J.2    Chu, H.3    Zhao, Y.4    Feng, W.5    Cheng, Y.6
  • 180
    • 84891405428 scopus 로고    scopus 로고
    • Influence of clonidine and ketamine on m-RNA expression in a model of opioid-induced hyperalgesia in mice
    • Ohnesorge, H., Feng, Z., Zitta, K., Steinfath, M., Albrecht, M., Bein, B., Influence of clonidine and ketamine on m-RNA expression in a model of opioid-induced hyperalgesia in mice. PLoS ONE, 8, 2013, e79567.
    • (2013) PLoS ONE , vol.8 , pp. e79567
    • Ohnesorge, H.1    Feng, Z.2    Zitta, K.3    Steinfath, M.4    Albrecht, M.5    Bein, B.6
  • 181
    • 84942919586 scopus 로고    scopus 로고
    • Mu opioid splice variant MOR-1K contributes to the development of opioid-induced hyperalgesia
    • Oladosu, F.A., Conrad, M.S., O'Buckley, S.C., Rashid, N.U., Slade, G.D., Nackley, A.G., Mu opioid splice variant MOR-1K contributes to the development of opioid-induced hyperalgesia. PLoS ONE, 10, 2015, e0135711.
    • (2015) PLoS ONE , vol.10 , pp. e0135711
    • Oladosu, F.A.1    Conrad, M.S.2    O'Buckley, S.C.3    Rashid, N.U.4    Slade, G.D.5    Nackley, A.G.6
  • 182
    • 84927623057 scopus 로고    scopus 로고
    • The role of glia in the spinal cord in neuropathic and inflammatory pain
    • Old, E.A., Clark, A.K., Malcangio, M., The role of glia in the spinal cord in neuropathic and inflammatory pain. Handb Exp Pharmacol 227 (2015), 145–170.
    • (2015) Handb Exp Pharmacol , vol.227 , pp. 145-170
    • Old, E.A.1    Clark, A.K.2    Malcangio, M.3
  • 183
    • 84906074966 scopus 로고    scopus 로고
    • Molecular perspectives for mu/delta opioid receptor heteromers as distinct, functional receptors
    • Ong, E.W., Cahill, C.M., Molecular perspectives for mu/delta opioid receptor heteromers as distinct, functional receptors. Cells 3 (2014), 152–179.
    • (2014) Cells , vol.3 , pp. 152-179
    • Ong, E.W.1    Cahill, C.M.2
  • 184
    • 0037835848 scopus 로고    scopus 로고
    • Induction of pain facilitation by sustained opioid exposure: relationship to opioid antinociceptive tolerance
    • Ossipov, M.H., Lai, J., Vanderah, T.W., Porreca, F., Induction of pain facilitation by sustained opioid exposure: relationship to opioid antinociceptive tolerance. Life Sci 73 (2003), 783–800.
    • (2003) Life Sci , vol.73 , pp. 783-800
    • Ossipov, M.H.1    Lai, J.2    Vanderah, T.W.3    Porreca, F.4
  • 185
    • 0032033683 scopus 로고    scopus 로고
    • Mu-Opposing actions of the kappa-opioid receptor
    • Pan, Z.Z., Mu-Opposing actions of the kappa-opioid receptor. Trends Pharmacol Sci 19 (1998), 94–98.
    • (1998) Trends Pharmacol Sci , vol.19 , pp. 94-98
    • Pan, Z.Z.1
  • 187
    • 84876675044 scopus 로고    scopus 로고
    • Mu opioids and their receptors: evolution of a concept
    • Pasternak, G.W., Pan, Y.X., Mu opioids and their receptors: evolution of a concept. Pharmacol Rev 65 (2013), 1257–1317.
    • (2013) Pharmacol Rev , vol.65 , pp. 1257-1317
    • Pasternak, G.W.1    Pan, Y.X.2
  • 188
    • 84868218099 scopus 로고    scopus 로고
    • Beta-Arrestin-2 desensitizes the transient receptor potential vanilloid 1 (TRPV1) channel
    • Por, E.D., Bierbower, S.M., Berg, K.A., Gomez, R., Akopian, A.N., Wetsel, W.C., Jeske, N.A., Beta-Arrestin-2 desensitizes the transient receptor potential vanilloid 1 (TRPV1) channel. J Biol Chem 287 (2012), 37552–37563.
    • (2012) J Biol Chem , vol.287 , pp. 37552-37563
    • Por, E.D.1    Bierbower, S.M.2    Berg, K.A.3    Gomez, R.4    Akopian, A.N.5    Wetsel, W.C.6    Jeske, N.A.7
  • 189
    • 84960352922 scopus 로고    scopus 로고
    • Examination of sex and minocycline treatment on acute morphine-induced analgesia and inflammatory gene expression along the pain pathway in Sprague-Dawley rats
    • Posillico, C.K., Terasaki, L.S., Bilbo, S.D., Schwarz, J.M., Examination of sex and minocycline treatment on acute morphine-induced analgesia and inflammatory gene expression along the pain pathway in Sprague-Dawley rats. Biol Sex Differ, 6, 2015, 33.
    • (2015) Biol Sex Differ , vol.6 , pp. 33
    • Posillico, C.K.1    Terasaki, L.S.2    Bilbo, S.D.3    Schwarz, J.M.4
  • 190
    • 84921470810 scopus 로고    scopus 로고
    • Chronic morphine-induced microRNA-124 promotes microglial immunosuppression by modulating P65 and TRAF6
    • Qiu, S., Feng, Y., LeSage, G., Zhang, Y., Stuart, C., He, L., Li, Y., Caudle, Y., Peng, Y., Yin, D., Chronic morphine-induced microRNA-124 promotes microglial immunosuppression by modulating P65 and TRAF6. J Immunol 194 (2015), 1021–1030.
    • (2015) J Immunol , vol.194 , pp. 1021-1030
    • Qiu, S.1    Feng, Y.2    LeSage, G.3    Zhang, Y.4    Stuart, C.5    He, L.6    Li, Y.7    Caudle, Y.8    Peng, Y.9    Yin, D.10
  • 191
    • 0037112041 scopus 로고    scopus 로고
    • The role of spinal neuroimmune activation in morphine tolerance/hyperalgesia in neuropathic and sham-operated rats
    • Raghavendra, V., Rutkowski, M.D., DeLeo, J.A., The role of spinal neuroimmune activation in morphine tolerance/hyperalgesia in neuropathic and sham-operated rats. J Neurosci 22 (2002), 9980–9989.
    • (2002) J Neurosci , vol.22 , pp. 9980-9989
    • Raghavendra, V.1    Rutkowski, M.D.2    DeLeo, J.A.3
  • 192
    • 0842286586 scopus 로고    scopus 로고
    • Attenuation of morphine tolerance, withdrawal-induced hyperalgesia, and associated spinal inflammatory immune responses by propentofylline in rats
    • Raghavendra, V., Tanga, F.Y., DeLeo, J.A., Attenuation of morphine tolerance, withdrawal-induced hyperalgesia, and associated spinal inflammatory immune responses by propentofylline in rats. Neuropsychopharmacology 29 (2004), 327–334.
    • (2004) Neuropsychopharmacology , vol.29 , pp. 327-334
    • Raghavendra, V.1    Tanga, F.Y.2    DeLeo, J.A.3
  • 193
    • 82155180464 scopus 로고    scopus 로고
    • Pharmacological treatment of opioid-induced hyperalgesia: a review of the evidence
    • Ramasubbu, C., Gupta, A., Pharmacological treatment of opioid-induced hyperalgesia: a review of the evidence. J Pain Palliat Care Pharmacother 25 (2011), 219–230.
    • (2011) J Pain Palliat Care Pharmacother , vol.25 , pp. 219-230
    • Ramasubbu, C.1    Gupta, A.2
  • 194
    • 84943328434 scopus 로고    scopus 로고
    • Activity-triggered tetrapartite neuron-glial interactions following peripheral injury
    • Ren, K., Dubner, R., Activity-triggered tetrapartite neuron-glial interactions following peripheral injury. Curr Opin Pharmacol 26 (2015), 16–25.
    • (2015) Curr Opin Pharmacol , vol.26 , pp. 16-25
    • Ren, K.1    Dubner, R.2
  • 196
    • 84863727622 scopus 로고    scopus 로고
    • Tolerance and opioid-induced hyperalgesia. Is a divorce imminent?
    • Richebe, P., Cahana, A., Rivat, C., Tolerance and opioid-induced hyperalgesia. Is a divorce imminent?. Pain 153 (2012), 1547–1548.
    • (2012) Pain , vol.153 , pp. 1547-1548
    • Richebe, P.1    Cahana, A.2    Rivat, C.3
  • 197
    • 13244252620 scopus 로고    scopus 로고
    • Ketamine improves the management of exaggerated postoperative pain observed in perioperative fentanyl-treated rats
    • Richebe, P., Rivat, C., Laulin, J.P., Maurette, P., Simonnet, G., Ketamine improves the management of exaggerated postoperative pain observed in perioperative fentanyl-treated rats. Anesthesiology 102 (2005), 421–428.
    • (2005) Anesthesiology , vol.102 , pp. 421-428
    • Richebe, P.1    Rivat, C.2    Laulin, J.P.3    Maurette, P.4    Simonnet, G.5
  • 199
    • 60349107058 scopus 로고    scopus 로고
    • Spinal NK-1 receptor-expressing neurons and descending pathways support fentanyl-induced pain hypersensitivity in a rat model of postoperative pain
    • Rivat, C., Vera-Portocarrero, L.P., Ibrahim, M.M., Mata, H.P., Stagg, N.J., De Felice, M., Porreca, F., Malan, T.P., Spinal NK-1 receptor-expressing neurons and descending pathways support fentanyl-induced pain hypersensitivity in a rat model of postoperative pain. Eur J Neurosci 29 (2009), 727–737.
    • (2009) Eur J Neurosci , vol.29 , pp. 727-737
    • Rivat, C.1    Vera-Portocarrero, L.P.2    Ibrahim, M.M.3    Mata, H.P.4    Stagg, N.J.5    De Felice, M.6    Porreca, F.7    Malan, T.P.8
  • 200
    • 33746812739 scopus 로고    scopus 로고
    • Endogenous nociceptin/orphanin FQ signalling produces opposite spinal antinociceptive and supraspinal pronociceptive effects in the mouse formalin test: pharmacological and genetic evidences
    • Rizzi, A., Nazzaro, C., Marzola, G.G., Zucchini, S., Trapella, C., Guerrini, R., Zeilhofer, H.U., Regoli, D., Calo, G., Endogenous nociceptin/orphanin FQ signalling produces opposite spinal antinociceptive and supraspinal pronociceptive effects in the mouse formalin test: pharmacological and genetic evidences. Pain 124 (2006), 100–108.
    • (2006) Pain , vol.124 , pp. 100-108
    • Rizzi, A.1    Nazzaro, C.2    Marzola, G.G.3    Zucchini, S.4    Trapella, C.5    Guerrini, R.6    Zeilhofer, H.U.7    Regoli, D.8    Calo, G.9
  • 201
    • 0026784599 scopus 로고
    • A review of the role of anti-opioid peptides in morphine tolerance and dependence
    • Rothman, R.B., A review of the role of anti-opioid peptides in morphine tolerance and dependence. Synapse 12 (1992), 129–138.
    • (1992) Synapse , vol.12 , pp. 129-138
    • Rothman, R.B.1
  • 202
    • 84898781739 scopus 로고    scopus 로고
    • Activation of mu opioid receptors sensitizes transient receptor potential vanilloid type 1 (TRPV1) via beta-arrestin-2-mediated cross-talk
    • Rowan, M.P., Bierbower, S.M., Eskander, M.A., Szteyn, K., Por, E.D., Gomez, R., Veldhuis, N., Bunnett, N.W., Jeske, N.A., Activation of mu opioid receptors sensitizes transient receptor potential vanilloid type 1 (TRPV1) via beta-arrestin-2-mediated cross-talk. PLoS ONE, 9, 2014, e93688.
    • (2014) PLoS ONE , vol.9 , pp. e93688
    • Rowan, M.P.1    Bierbower, S.M.2    Eskander, M.A.3    Szteyn, K.4    Por, E.D.5    Gomez, R.6    Veldhuis, N.7    Bunnett, N.W.8    Jeske, N.A.9
  • 203
    • 84905961988 scopus 로고    scopus 로고
    • Beta-arrestin-2-biased agonism of delta opioid receptors sensitizes transient receptor potential vanilloid type 1 (TRPV1) in primary sensory neurons
    • Rowan, M.P., Szteyn, K., Doyle, A.P., Gomez, R., Henry, M.A., Jeske, N.A., Beta-arrestin-2-biased agonism of delta opioid receptors sensitizes transient receptor potential vanilloid type 1 (TRPV1) in primary sensory neurons. Mol Pain, 10, 2014, 50.
    • (2014) Mol Pain , vol.10 , pp. 50
    • Rowan, M.P.1    Szteyn, K.2    Doyle, A.P.3    Gomez, R.4    Henry, M.A.5    Jeske, N.A.6
  • 204
    • 79953080700 scopus 로고    scopus 로고
    • Long-term potentiation in spinal nociceptive pathways as a novel target for pain therapy
    • Ruscheweyh, R., Wilder-Smith, O., Drdla, R., Liu, X.G., Sandkuhler, J., Long-term potentiation in spinal nociceptive pathways as a novel target for pain therapy. Mol Pain, 7, 2011, 20.
    • (2011) Mol Pain , vol.7 , pp. 20
    • Ruscheweyh, R.1    Wilder-Smith, O.2    Drdla, R.3    Liu, X.G.4    Sandkuhler, J.5
  • 205
    • 84873943621 scopus 로고    scopus 로고
    • Non-analgesic effects of opioids: mechanisms and potential clinical relevance of opioid-induced immunodepression
    • Sacerdote, P., Franchi, S., Panerai, A.E., Non-analgesic effects of opioids: mechanisms and potential clinical relevance of opioid-induced immunodepression. Curr Pharm Des 18 (2012), 6034–6042.
    • (2012) Curr Pharm Des , vol.18 , pp. 6034-6042
    • Sacerdote, P.1    Franchi, S.2    Panerai, A.E.3
  • 206
    • 85007447747 scopus 로고    scopus 로고
    • Epigenetic regulation of spinal cord gene expression contributes to enhanced postoperative pain and analgesic tolerance subsequent to continuous opioid exposure
    • Sahbaie, P., Liang, D.Y., Shi, X.Y., Sun, Y., Clark, J.D., Epigenetic regulation of spinal cord gene expression contributes to enhanced postoperative pain and analgesic tolerance subsequent to continuous opioid exposure. Mol Pain, 12, 2016.
    • (2016) Mol Pain , vol.12
    • Sahbaie, P.1    Liang, D.Y.2    Shi, X.Y.3    Sun, Y.4    Clark, J.D.5
  • 207
    • 84873410053 scopus 로고    scopus 로고
    • Therapeutic targeting of the ceramide-to-sphingosine 1-phosphate pathway in pain
    • Salvemini, D., Doyle, T., Kress, M., Nicol, G., Therapeutic targeting of the ceramide-to-sphingosine 1-phosphate pathway in pain. Trends Pharmacol Sci 34 (2013), 110–118.
    • (2013) Trends Pharmacol Sci , vol.34 , pp. 110-118
    • Salvemini, D.1    Doyle, T.2    Kress, M.3    Nicol, G.4
  • 209
    • 67349202612 scopus 로고    scopus 로고
    • Models and mechanisms of hyperalgesia and allodynia
    • Sandkuhler, J., Models and mechanisms of hyperalgesia and allodynia. Physiol Rev 89 (2009), 707–758.
    • (2009) Physiol Rev , vol.89 , pp. 707-758
    • Sandkuhler, J.1
  • 210
    • 84904570646 scopus 로고    scopus 로고
    • Regionally selective activation of ERK and JNK in morphine paradoxical hyperalgesia: a step toward improving opioid pain therapy
    • Sanna, M.D., Ghelardini, C., Galeotti, N., Regionally selective activation of ERK and JNK in morphine paradoxical hyperalgesia: a step toward improving opioid pain therapy. Neuropharmacology 86 (2014), 67–77.
    • (2014) Neuropharmacology , vol.86 , pp. 67-77
    • Sanna, M.D.1    Ghelardini, C.2    Galeotti, N.3
  • 211
    • 84936109838 scopus 로고    scopus 로고
    • Activation of JNK pathway in spinal astrocytes contributes to acute ultra-low-dose morphine thermal hyperalgesia
    • Sanna, M.D., Ghelardini, C., Galeotti, N., Activation of JNK pathway in spinal astrocytes contributes to acute ultra-low-dose morphine thermal hyperalgesia. Pain 156 (2015), 1265–1275.
    • (2015) Pain , vol.156 , pp. 1265-1275
    • Sanna, M.D.1    Ghelardini, C.2    Galeotti, N.3
  • 212
    • 84937239474 scopus 로고    scopus 로고
    • Inhibition of spinal ERK1/2-c-JUN signaling pathway counteracts the development of low doses morphine-induced hyperalgesia
    • Sanna, M.D., Mello, T., Ghelardini, C., Galeotti, N., Inhibition of spinal ERK1/2-c-JUN signaling pathway counteracts the development of low doses morphine-induced hyperalgesia. Eur J Pharmacol 764 (2015), 271–277.
    • (2015) Eur J Pharmacol , vol.764 , pp. 271-277
    • Sanna, M.D.1    Mello, T.2    Ghelardini, C.3    Galeotti, N.4
  • 213
    • 13344268990 scopus 로고    scopus 로고
    • Role of intercellular interactions in heterosynaptic long-term depression
    • Scanziani, M., Malenka, R.C., Nicoll, R.A., Role of intercellular interactions in heterosynaptic long-term depression. Nature 380 (1996), 446–450.
    • (1996) Nature , vol.380 , pp. 446-450
    • Scanziani, M.1    Malenka, R.C.2    Nicoll, R.A.3
  • 216
    • 77955154838 scopus 로고    scopus 로고
    • Opioids, pain, the brain, and hyperkatifeia: a framework for the rational use of opioids for pain
    • Shurman, J., Koob, G.F., Gutstein, H.B., Opioids, pain, the brain, and hyperkatifeia: a framework for the rational use of opioids for pain. Pain Med 11 (2010), 1092–1098.
    • (2010) Pain Med , vol.11 , pp. 1092-1098
    • Shurman, J.1    Koob, G.F.2    Gutstein, H.B.3
  • 217
    • 69849096806 scopus 로고    scopus 로고
    • Opioid induced hyperalgesia: clinical implications for the pain practitioner
    • Silverman, S.M., Opioid induced hyperalgesia: clinical implications for the pain practitioner. Pain Physician 12 (2009), 679–684.
    • (2009) Pain Physician , vol.12 , pp. 679-684
    • Silverman, S.M.1
  • 218
    • 33750094911 scopus 로고    scopus 로고
    • Neuropeptide FF receptors as therapeutic targets
    • Simonin, F., Neuropeptide FF receptors as therapeutic targets. Drugs Future 31 (2006), 603–609.
    • (2006) Drugs Future , vol.31 , pp. 603-609
    • Simonin, F.1
  • 220
    • 0032481350 scopus 로고    scopus 로고
    • Disruption of the k-opioid receptor gene in mice enhances sensitivity to chemical visceral pain, impairs pharmacological actions of the selective k-agonist U-50,488H and attenuates morphine withdrawal
    • Simonin, F., Valverde, O., Smadja, S., Slowe, S., Kitchen, I., Dierich, A., Le Meur, M., Roques, B.P., Maldonado, R., Kieffer, B.L., Disruption of the k-opioid receptor gene in mice enhances sensitivity to chemical visceral pain, impairs pharmacological actions of the selective k-agonist U-50,488H and attenuates morphine withdrawal. EMBO J 17 (1998), 886–897.
    • (1998) EMBO J , vol.17 , pp. 886-897
    • Simonin, F.1    Valverde, O.2    Smadja, S.3    Slowe, S.4    Kitchen, I.5    Dierich, A.6    Le Meur, M.7    Roques, B.P.8    Maldonado, R.9    Kieffer, B.L.10
  • 221
    • 85027934270 scopus 로고    scopus 로고
    • Translational potential of naloxone and naltrexone as TLR4 antagonists
    • Skolnick, P., Davis, H., Arnelle, D., Deaver, D., Translational potential of naloxone and naltrexone as TLR4 antagonists. Trends Pharmacol Sci 35 (2014), 431–432.
    • (2014) Trends Pharmacol Sci , vol.35 , pp. 431-432
    • Skolnick, P.1    Davis, H.2    Arnelle, D.3    Deaver, D.4
  • 223
    • 84954483870 scopus 로고    scopus 로고
    • Opioid Receptors
    • Stein, C., Opioid Receptors. Annu Rev Med 67 (2016), 433–451.
    • (2016) Annu Rev Med , vol.67 , pp. 433-451
    • Stein, C.1
  • 224
    • 80053606300 scopus 로고    scopus 로고
    • Modulation of peripheral sensory neurons by the immune system: implications for pain therapy
    • Stein, C., Machelska, H., Modulation of peripheral sensory neurons by the immune system: implications for pain therapy. Pharmacol Rev 63 (2011), 860–881.
    • (2011) Pharmacol Rev , vol.63 , pp. 860-881
    • Stein, C.1    Machelska, H.2
  • 225
    • 84874408845 scopus 로고    scopus 로고
    • Pharmacological characterization of LPS and opioid interactions at the toll-like receptor 4
    • Stevens, C.W., Aravind, S., Das, S., Davis, R.L., Pharmacological characterization of LPS and opioid interactions at the toll-like receptor 4. Br J Pharmacol 168 (2013), 1421–1429.
    • (2013) Br J Pharmacol , vol.168 , pp. 1421-1429
    • Stevens, C.W.1    Aravind, S.2    Das, S.3    Davis, R.L.4
  • 226
    • 34248226218 scopus 로고    scopus 로고
    • Descending serotonergic facilitation of spinal ERK activation and pain behavior
    • Svensson, C.I., Tran, T.K., Fitzsimmons, B., Yaksh, T.L., Hua, X.Y., Descending serotonergic facilitation of spinal ERK activation and pain behavior. FEBS Lett 580 (2006), 6629–6634.
    • (2006) FEBS Lett , vol.580 , pp. 6629-6634
    • Svensson, C.I.1    Tran, T.K.2    Fitzsimmons, B.3    Yaksh, T.L.4    Hua, X.Y.5
  • 228
    • 0037127073 scopus 로고    scopus 로고
    • Modulation of NMDA receptor-dependent calcium influx and gene expression through EphB receptors
    • Takasu, M.A., Dalva, M.B., Zigmond, R.E., Greenberg, M.E., Modulation of NMDA receptor-dependent calcium influx and gene expression through EphB receptors. Science (New York, NY) 295 (2002), 491–495.
    • (2002) Science (New York, NY) , vol.295 , pp. 491-495
    • Takasu, M.A.1    Dalva, M.B.2    Zigmond, R.E.3    Greenberg, M.E.4
  • 229
    • 12144283981 scopus 로고    scopus 로고
    • Morphine-induced chemotaxis and brain-derived neurotrophic factor expression in microglia
    • Takayama, N., Ueda, H., Morphine-induced chemotaxis and brain-derived neurotrophic factor expression in microglia. J Neurosci 25 (2005), 430–435.
    • (2005) J Neurosci , vol.25 , pp. 430-435
    • Takayama, N.1    Ueda, H.2
  • 230
    • 84927606879 scopus 로고    scopus 로고
    • The relationship between opioids and immune signalling in the spinal cord
    • Thomas, J., Mustafa, S., Johnson, J., Nicotra, L., Hutchinson, M., The relationship between opioids and immune signalling in the spinal cord. Handb Exp Pharmacol 227 (2015), 207–238.
    • (2015) Handb Exp Pharmacol , vol.227 , pp. 207-238
    • Thomas, J.1    Mustafa, S.2    Johnson, J.3    Nicotra, L.4    Hutchinson, M.5
  • 231
    • 84960395737 scopus 로고    scopus 로고
    • Nociceptin/orphanin FQ receptor structure, signaling, ligands, functions, and interactions with opioid systems
    • Toll, L., Bruchas, M.R., Calo, G., Cox, B.M., Zaveri, N.T., Nociceptin/orphanin FQ receptor structure, signaling, ligands, functions, and interactions with opioid systems. Pharmacol Rev 68 (2016), 419–457.
    • (2016) Pharmacol Rev , vol.68 , pp. 419-457
    • Toll, L.1    Bruchas, M.R.2    Calo, G.3    Cox, B.M.4    Zaveri, N.T.5
  • 233
    • 63349100852 scopus 로고    scopus 로고
    • Ultra-low dose naloxone restores the antinociceptive effect of morphine in pertussis toxin-treated rats and prevents glutamate transporter downregulation by suppressing the p38 mitogen-activated protein kinase signaling pathway
    • Tsai, R.Y., Tai, Y.H., Tzeng, J.I., Lin, S.L., Shen, C.H., Yang, C.P., Hsin, S.T., Wang, C.B., Wong, C.S., Ultra-low dose naloxone restores the antinociceptive effect of morphine in pertussis toxin-treated rats and prevents glutamate transporter downregulation by suppressing the p38 mitogen-activated protein kinase signaling pathway. Neuroscience 159 (2009), 1244–1256.
    • (2009) Neuroscience , vol.159 , pp. 1244-1256
    • Tsai, R.Y.1    Tai, Y.H.2    Tzeng, J.I.3    Lin, S.L.4    Shen, C.H.5    Yang, C.P.6    Hsin, S.T.7    Wang, C.B.8    Wong, C.S.9
  • 234
  • 235
    • 84940788267 scopus 로고    scopus 로고
    • Histone deacetylase inhibitors relieve morphine resistance in neuropathic pain after peripheral nerve injury
    • Uchida, H., Matsushita, Y., Araki, K., Mukae, T., Ueda, H., Histone deacetylase inhibitors relieve morphine resistance in neuropathic pain after peripheral nerve injury. J Pharmacol Sci 128 (2015), 208–211.
    • (2015) J Pharmacol Sci , vol.128 , pp. 208-211
    • Uchida, H.1    Matsushita, Y.2    Araki, K.3    Mukae, T.4    Ueda, H.5
  • 236
    • 84891372807 scopus 로고    scopus 로고
    • Botulinum toxin A increases analgesic effects of morphine, counters development of morphine tolerance and modulates glia activation and mu opioid receptor expression in neuropathic mice
    • Vacca, V., Marinelli, S., Luvisetto, S., Pavone, F., Botulinum toxin A increases analgesic effects of morphine, counters development of morphine tolerance and modulates glia activation and mu opioid receptor expression in neuropathic mice. Brain Behav Immun 32 (2013), 40–50.
    • (2013) Brain Behav Immun , vol.32 , pp. 40-50
    • Vacca, V.1    Marinelli, S.2    Luvisetto, S.3    Pavone, F.4
  • 237
    • 0035141560 scopus 로고    scopus 로고
    • Tonic descending facilitation from the rostral ventromedial medulla mediates opioid-induced abnormal pain and antinociceptive tolerance
    • Vanderah, T.W., Suenaga, N.M., Ossipov, M.H., Malan, T.P. Jr., Lai, J., Porreca, F., Tonic descending facilitation from the rostral ventromedial medulla mediates opioid-induced abnormal pain and antinociceptive tolerance. J Neurosci 21 (2001), 279–286.
    • (2001) J Neurosci , vol.21 , pp. 279-286
    • Vanderah, T.W.1    Suenaga, N.M.2    Ossipov, M.H.3    Malan, T.P.4    Lai, J.5    Porreca, F.6
  • 239
    • 34047271295 scopus 로고    scopus 로고
    • Spinal NK-1 receptor expressing neurons mediate opioid-induced hyperalgesia and antinociceptive tolerance via activation of descending pathways
    • Vera-Portocarrero, L.P., Zhang, E.T., King, T., Ossipov, M.H., Vanderah, T.W., Lai, J., Porreca, F., Spinal NK-1 receptor expressing neurons mediate opioid-induced hyperalgesia and antinociceptive tolerance via activation of descending pathways. Pain 129 (2007), 35–45.
    • (2007) Pain , vol.129 , pp. 35-45
    • Vera-Portocarrero, L.P.1    Zhang, E.T.2    King, T.3    Ossipov, M.H.4    Vanderah, T.W.5    Lai, J.6    Porreca, F.7
  • 240
    • 84857287844 scopus 로고    scopus 로고
    • Deconstructing the neuropathic pain phenotype to reveal neural mechanisms
    • von Hehn, C.A., Baron, R., Woolf, C.J., Deconstructing the neuropathic pain phenotype to reveal neural mechanisms. Neuron 73 (2012), 638–652.
    • (2012) Neuron , vol.73 , pp. 638-652
    • von Hehn, C.A.1    Baron, R.2    Woolf, C.J.3
  • 241
    • 78650677080 scopus 로고    scopus 로고
    • Buprenorphine-induced hyperalgesia in the rat
    • Wala, E.P., Holtman, J.R. Jr., Buprenorphine-induced hyperalgesia in the rat. Eur J Pharmacol 651 (2011), 89–95.
    • (2011) Eur J Pharmacol , vol.651 , pp. 89-95
    • Wala, E.P.1    Holtman, J.R.2
  • 242
    • 81855180393 scopus 로고    scopus 로고
    • Effect of prior treatment with ultra-low-dose morphine on opioid- and nerve injury-induced hyperalgesia in rats
    • Wala, E.P., Sloan, P.A., Holtman, J.R. Jr., Effect of prior treatment with ultra-low-dose morphine on opioid- and nerve injury-induced hyperalgesia in rats. J Opioid Manag 7 (2011), 377–389.
    • (2011) J Opioid Manag , vol.7 , pp. 377-389
    • Wala, E.P.1    Sloan, P.A.2    Holtman, J.R.3
  • 244
    • 85027936718 scopus 로고    scopus 로고
    • In vivo veritas: (+)-Naltrexone's actions define translational importance: a letter in response to Skolnick et al. ‘Translational potential of naloxone and naltrexone as TLR4 antagonists’
    • Watkins, L.R., Wang, X., Mustafa, S., Hutchinson, M.R., In vivo veritas: (+)-Naltrexone's actions define translational importance: a letter in response to Skolnick et al. ‘Translational potential of naloxone and naltrexone as TLR4 antagonists’. Trends Pharmacol Sci 35 (2014), 432–433.
    • (2014) Trends Pharmacol Sci , vol.35 , pp. 432-433
    • Watkins, L.R.1    Wang, X.2    Mustafa, S.3    Hutchinson, M.R.4
  • 248
    • 84908054442 scopus 로고    scopus 로고
    • Spinal ephrinB/EphB signalling contributed to remifentanil-induced hyperalgesia via NMDA receptor
    • Xia, W.S., Peng, Y.N., Tang, L.H., Jiang, L.S., Yu, L.N., Zhou, X.L., Zhang, F.J., Yan, M., Spinal ephrinB/EphB signalling contributed to remifentanil-induced hyperalgesia via NMDA receptor. Eur J Pain 18 (2014), 1231–1239.
    • (2014) Eur J Pain , vol.18 , pp. 1231-1239
    • Xia, W.S.1    Peng, Y.N.2    Tang, L.H.3    Jiang, L.S.4    Yu, L.N.5    Zhou, X.L.6    Zhang, F.J.7    Yan, M.8
  • 249
    • 12144265144 scopus 로고    scopus 로고
    • Cholecystokinin in the rostral ventromedial medulla mediates opioid-induced hyperalgesia and antinociceptive tolerance
    • Xie, J.Y., Herman, D.S., Stiller, C.O., Gardell, L.R., Ossipov, M.H., Lai, J., Porreca, F., Vanderah, T.W., Cholecystokinin in the rostral ventromedial medulla mediates opioid-induced hyperalgesia and antinociceptive tolerance. J Neurosci 25 (2005), 409–416.
    • (2005) J Neurosci , vol.25 , pp. 409-416
    • Xie, J.Y.1    Herman, D.S.2    Stiller, C.O.3    Gardell, L.R.4    Ossipov, M.H.5    Lai, J.6    Porreca, F.7    Vanderah, T.W.8
  • 250
    • 84950263489 scopus 로고    scopus 로고
    • Intrathecal rapamycin attenuates morphine-induced analgesic tolerance and hyperalgesia in rats with neuropathic pain
    • Xu, J.T., Sun, L., Lutz, B.M., Bekker, A., Tao, Y.X., Intrathecal rapamycin attenuates morphine-induced analgesic tolerance and hyperalgesia in rats with neuropathic pain. Transl Perioper Pain Med 2 (2015), 27–34.
    • (2015) Transl Perioper Pain Med , vol.2 , pp. 27-34
    • Xu, J.T.1    Sun, L.2    Lutz, B.M.3    Bekker, A.4    Tao, Y.X.5
  • 252
    • 77953125292 scopus 로고    scopus 로고
    • Expression and distribution of mTOR, p70S6K, 4E-BP1, and their phosphorylated counterparts in rat dorsal root ganglion and spinal cord dorsal horn
    • Xu, J.T., Zhao, X., Yaster, M., Tao, Y.X., Expression and distribution of mTOR, p70S6K, 4E-BP1, and their phosphorylated counterparts in rat dorsal root ganglion and spinal cord dorsal horn. Brain Res 1336 (2010), 46–57.
    • (2010) Brain Res , vol.1336 , pp. 46-57
    • Xu, J.T.1    Zhao, X.2    Yaster, M.3    Tao, Y.X.4
  • 254
    • 1342280526 scopus 로고    scopus 로고
    • Roles of CaMKII, PKA, and PKC in the induction and maintenance of LTP of C-fiber-evoked field potentials in rat spinal dorsal horn
    • Yang, H.W., Hu, X.D., Zhang, H.M., Xin, W.J., Li, M.T., Zhang, T., Zhou, L.J., Liu, X.G., Roles of CaMKII, PKA, and PKC in the induction and maintenance of LTP of C-fiber-evoked field potentials in rat spinal dorsal horn. J Neurophysiol 91 (2004), 1122–1133.
    • (2004) J Neurophysiol , vol.91 , pp. 1122-1133
    • Yang, H.W.1    Hu, X.D.2    Zhang, H.M.3    Xin, W.J.4    Li, M.T.5    Zhang, T.6    Zhou, L.J.7    Liu, X.G.8
  • 255
    • 84955446711 scopus 로고    scopus 로고
    • Endomorphin analog analgesics with reduced abuse liability, respiratory depression, motor impairment, tolerance, and glial activation relative to morphine
    • Zadina, J.E., Nilges, M.R., Morgenweck, J., Zhang, X., Hackler, L., Fasold, M.B., Endomorphin analog analgesics with reduced abuse liability, respiratory depression, motor impairment, tolerance, and glial activation relative to morphine. Neuropharmacology 105 (2015), 215–227.
    • (2015) Neuropharmacology , vol.105 , pp. 215-227
    • Zadina, J.E.1    Nilges, M.R.2    Morgenweck, J.3    Zhang, X.4    Hackler, L.5    Fasold, M.B.6
  • 256
    • 84855880181 scopus 로고    scopus 로고
    • Chronic pain states: pharmacological strategies to restore diminished inhibitory spinal pain control
    • Zeilhofer, H.U., Benke, D., Yevenes, G.E., Chronic pain states: pharmacological strategies to restore diminished inhibitory spinal pain control. Annu Rev Pharmacol Toxicol 52 (2012), 111–133.
    • (2012) Annu Rev Pharmacol Toxicol , vol.52 , pp. 111-133
    • Zeilhofer, H.U.1    Benke, D.2    Yevenes, G.E.3
  • 257
    • 78651498932 scopus 로고    scopus 로고
    • TNF-alpha contributes to spinal cord synaptic plasticity and inflammatory pain: distinct role of TNF receptor subtypes 1 and 2
    • Zhang, L., Berta, T., Xu, Z.Z., Liu, T., Park, J.Y., Ji, R.R., TNF-alpha contributes to spinal cord synaptic plasticity and inflammatory pain: distinct role of TNF receptor subtypes 1 and 2. Pain 152 (2011), 419–427.
    • (2011) Pain , vol.152 , pp. 419-427
    • Zhang, L.1    Berta, T.2    Xu, Z.Z.3    Liu, T.4    Park, J.Y.5    Ji, R.R.6
  • 258
    • 2942731605 scopus 로고    scopus 로고
    • Proinflammatory chemokines, such as C-C chemokine ligand 3, desensitize mu-opioid receptors on dorsal root ganglia neurons
    • Zhang, N., Rogers, T.J., Caterina, M., Oppenheim, J.J., Proinflammatory chemokines, such as C-C chemokine ligand 3, desensitize mu-opioid receptors on dorsal root ganglia neurons. J Immunol 173 (2004), 594–599.
    • (2004) J Immunol , vol.173 , pp. 594-599
    • Zhang, N.1    Rogers, T.J.2    Caterina, M.3    Oppenheim, J.J.4
  • 259
    • 84870901086 scopus 로고    scopus 로고
    • Spinal MCP-1 contributes to the development of morphine antinociceptive tolerance in rats
    • Zhao, C.M., Guo, R.X., Hu, F., Meng, J.L., Mo, L.Q., Chen, P.X., Liao, X.X., Cui, Y., Feng, J.Q., Spinal MCP-1 contributes to the development of morphine antinociceptive tolerance in rats. Am J Med Sci 344 (2012), 473–479.
    • (2012) Am J Med Sci , vol.344 , pp. 473-479
    • Zhao, C.M.1    Guo, R.X.2    Hu, F.3    Meng, J.L.4    Mo, L.Q.5    Chen, P.X.6    Liao, X.X.7    Cui, Y.8    Feng, J.Q.9
  • 260
    • 84864108171 scopus 로고    scopus 로고
    • Chronic opioid potentiates presynaptic but impairs postsynaptic N-methyl-D-aspartic acid receptor activity in spinal cords: implications for opioid hyperalgesia and tolerance
    • Zhao, Y.L., Chen, S.R., Chen, H., Pan, H.L., Chronic opioid potentiates presynaptic but impairs postsynaptic N-methyl-D-aspartic acid receptor activity in spinal cords: implications for opioid hyperalgesia and tolerance. J Biol Chem 287 (2012), 25073–25085.
    • (2012) J Biol Chem , vol.287 , pp. 25073-25085
    • Zhao, Y.L.1    Chen, S.R.2    Chen, H.3    Pan, H.L.4
  • 261
    • 77954217806 scopus 로고    scopus 로고
    • The direction of synaptic plasticity mediated by C-fibers in spinal dorsal horn is decided by Src-family kinases in microglia: the role of tumor necrosis factor-alpha
    • Zhong, Y., Zhou, L.J., Ren, W.J., Xin, W.J., Li, Y.Y., Zhang, T., Liu, X.G., The direction of synaptic plasticity mediated by C-fibers in spinal dorsal horn is decided by Src-family kinases in microglia: the role of tumor necrosis factor-alpha. Brain Behav Immun 24 (2010), 874–880.
    • (2010) Brain Behav Immun , vol.24 , pp. 874-880
    • Zhong, Y.1    Zhou, L.J.2    Ren, W.J.3    Xin, W.J.4    Li, Y.Y.5    Zhang, T.6    Liu, X.G.7
  • 262
    • 77949839792 scopus 로고    scopus 로고
    • Opioid-induced long-term potentiation in the spinal cord is a presynaptic event
    • Zhou, H.Y., Chen, S.R., Chen, H., Pan, H.L., Opioid-induced long-term potentiation in the spinal cord is a presynaptic event. J Neurosci 30 (2010), 4460–4466.
    • (2010) J Neurosci , vol.30 , pp. 4460-4466
    • Zhou, H.Y.1    Chen, S.R.2    Chen, H.3    Pan, H.L.4
  • 263


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.