Chronic morphine administration enhances nociceptive sensitivity and local cytokine production after incision

DeYong Liang, Xiaoyou Shi, Yanli Qiao, Martin S Angst, David C Yeomans, J David Clark, DeYong Liang, Xiaoyou Shi, Yanli Qiao, Martin S Angst, David C Yeomans, J David Clark

Abstract

Background: The chronic use of opioids prior to surgery leads to lowered pain thresholds and exaggerated pain levels after these procedures. Several mechanisms have been proposed to explain this heightened sensitivity commonly termed opioid-induced hyperalgesia (OIH). Most of these proposed mechanisms involve plastic events in the central or peripheral nervous systems. Alterations in the abundance of peripheral mediators of nociception have not previously been explored.

Results: In these experiments mice were treated with saline (control) or ascending daily doses of morphine to generate a state of OIH followed by hind paw incision. In other experiments morphine treatment was initiated at the time of incision. Both mechanical allodynia and peri-incisional skin cytokine levels were measured. Myeloperoxidase (MPO) assays were used to determine neutrophil activity near the wounds. The cytokine production inhibitor pentoxifylline was used to determine the functional significance of the excess cytokines in previously morphine treated animals. Mice treated chronically treated with morphine prior to incision were found to have enhanced skin levels of IL-1beta, IL-6, G-CSF, KC and TNFalpha after incision at one or more time points compared to saline pretreated controls. The time courses of individual cytokines followed different patterns. There was no discernable effect of chronic morphine treatment on wound area neutrophil infiltration. Pentoxifylline reduced cytokine levels and reversed the excess mechanical sensitization caused by chronic morphine administration prior to incision. Morphine treatment initiated at the time of incision did not lead to a generalized enhancement of cytokine production or nociceptive sensitization in excess of the levels observed after incision alone.

Conclusion: The enhanced level of nociceptive sensitization seen after incision in animals chronically exposed to morphine is associated with elevated levels of several cytokines previously reported to be relevant to this incisional pain model. The cytokines may be functional in supporting nociceptive sensitization because pentoxifylline reverses both peri-incisional skin cytokine levels and OIH. Opioid administration beginning at the time of incision does not seem to have the same cytokine enhancing effect. Approaches to postoperative pain control involving a reduction of cytokines may be an effective way to control excessive pain in patients chronically using opioids prior to surgical procedures.

Figures

Figure 1
Figure 1
Mechanical nociceptive thresholds after drug pretreatment and hindpaw incision. Panel A displays the mechanical thresholds of mice in four different treatment groups: saline pretreatment/no incision, saline pretreatment/incision, morphine pretreatment/no incision, morphine pretreatment/incision. The values labeled pre-incision represent the nociceptive thresholds measured after 4 days of saline or morphine treatment but prior to hind paw incision. In panel B data are presented representing mechanical nociceptive thresholds in mice undergoing saline or morphine treatment beginning at the time of incision. Nociceptive thresholds were measured immediately before that day's dose of morphine. The statistical analysis presented reflects the results of two-way ANOVA comparing saline/incision values to morphine/incision ones. *p < 0.05, **p < 0.01, N = 6/group.
Figure 2
Figure 2
Peri-incisional skin cytokine levels after saline or morphine pretreatment. The figure displays the mechanical thresholds of mice in four different treatment groups: saline pretreatment/no incision, saline pretreatment/incision, morphine pretreatment/no incision, morphine pretreatment/incision. The cytokine levels were assessed at time points out to 72 hours after incision based on the results of the experiments presented in Figure 1. The statistical analysis presented reflects the results of two-way ANOVA comparing saline/incision values to morphine/incision ones over the 2 to 72 hour time period. *p < 0.05, **p < 0.01, N = 8/group.
Figure 3
Figure 3
Peri-incisional skin cytokine levels for mice treated with saline or morphine beginning at the time of incision. The figure displays cytokine measurements from mice in four different treatment groups: saline, incision/saline, no incision/morphine, incision/morphine. The cytokine levels were assessed at 120 hours after the incisions were made (18 hours after the last dose of morphine). Statistical analysis was performed to detect differences between each set of conditions. N = 8/group.
Figure 4
Figure 4
Myeloperoxidase (MPO) activity after hind paw incisions. For these experiments MPO activity was measured in peri-incisional skin as an index of infiltrating neutrophil activity. Skin was harvested from separate groups of mice at the indicated time points and processed for MPO assays as described in Methods. The time course for analysis was the same as that used for the behavioral and cytokine assays. Statistical analysis failed to detect between group differences at any of these time points. N = 8/group.
Figure 5
Figure 5
Mechanical allodynia after incision as affected by morphine and pentoxifylline. In these experiments mice were subjected to nociceptive testing of the hind paw to establish the baseline threshold. Mice were then pre-treated with either saline or morphine for 4 days prior to hind paw incisions being made. After incision, groups of mice were treated daily with daily intraperitoneal saline or pentoxifylline as described in Methods. Nociceptive testing 72 hours after hind paw incision (2 hours after the last dose of pentoxifylline). **p < 0.01, N = 8/group.

References

    1. Clark JD. Chronic pain prevalence and analgesic prescribing in a general medical population. J Pain Symptom Manage. 2002;23:131–137. doi: 10.1016/S0885-3924(01)00396-7.
    1. Angst MS, Clark JD. Opioid-induced hyperalgesia: a qualitative systematic review. Anesthesiology. 2006;104:570–587. doi: 10.1097/00000542-200603000-00025.
    1. Chang G, Chen L, Mao J. Opioid tolerance and hyperalgesia. Med Clin North Am. 2007;91:199–211. doi: 10.1016/j.mcna.2006.10.003.
    1. Koppert W, Schmelz M. The impact of opioid-induced hyperalgesia for postoperative pain. Best Pract Res Clin Anaesthesiol. 2007;21:65–83. doi: 10.1016/j.bpa.2006.12.004.
    1. Mao J. Opioid-induced abnormal pain sensitivity. Curr Pain Headache Rep. 2006;10:67–70. doi: 10.1007/s11916-006-0011-5.
    1. Ruan X. Drug-related side effects of long-term intrathecal morphine therapy. Pain Physician. 2007;10:357–366.
    1. Bulka A, Plesan A, Xu XJ, Wiesenfeld-Hallin Z. Reduced tolerance to the anti-hyperalgesic effect of methadone in comparison to morphine in a rat model of mononeuropathy. Pain. 2002;95:103–109. doi: 10.1016/S0304-3959(01)00382-7.
    1. Compton P, Charuvastra VC, Ling W. Pain intolerance in opioid-maintained former opiate addicts: effect of long-acting maintenance agent. Drug Alcohol Depend. 2001;63:139–146. doi: 10.1016/S0376-8716(00)00200-3.
    1. Doverty M, Somogyi AA, White JM, Bochner F, Beare CH, Menelaou A, Ling W. Methadone maintenance patients are cross-tolerant to the antinociceptive effects of morphine. Pain. 2001;93:155–163. doi: 10.1016/S0304-3959(01)00306-2.
    1. Doverty M, White JM, Somogyi AA, Bochner F, Ali R, Ling W. Hyperalgesic responses in methadone maintenance patients. Pain. 2001;90:91–96. doi: 10.1016/S0304-3959(00)00391-2.
    1. Pud D, Cohen D, Lawental E, Eisenberg E. Opioids and abnormal pain perception: New evidence from a study of chronic opioid addicts and healthy subjects. Drug Alcohol Depend. 2006;82:218–223. doi: 10.1016/j.drugalcdep.2005.09.007.
    1. Chu LF, Clark DJ, Angst MS. Opioid tolerance and hyperalgesia in chronic pain patients after one month of oral morphine therapy: a preliminary prospective study. J Pain. 2006;7:43–48. doi: 10.1016/j.jpain.2005.08.001.
    1. Angst MS, Koppert W, Pahl I, Clark DJ, Schmelz M. Short-term infusion of the mu-opioid agonist remifentanil in humans causes hyperalgesia during withdrawal. Pain. 2003;106:49–57. doi: 10.1016/S0304-3959(03)00276-8.
    1. Hood DD, Curry R, Eisenach JC. Intravenous remifentanil produces withdrawal hyperalgesia in volunteers with capsaicin-induced hyperalgesia. Anesth Analg. 2003;97:810–815. doi: 10.1213/01.ANE.0000078811.80093.88.
    1. Joly V, Richebe P, Guignard B, Fletcher D, Maurette P, Sessler DI, Chauvin M. Remifentanil-induced postoperative hyperalgesia and its prevention with small-dose ketamine. Anesthesiology. 2005;103:147–155. doi: 10.1097/00000542-200507000-00022.
    1. Koppert W, Angst M, Alsheimer M, Sittl R, Albrecht S, Schuttler J, Schmelz M. Naloxone provokes similar pain facilitation as observed after short-term infusion of remifentanil in humans. Pain. 2003;106:91–99. doi: 10.1016/S0304-3959(03)00294-X.
    1. Luginbuhl M, Gerber A, Schnider TW, Petersen-Felix S, Arendt-Nielsen L, Curatolo M. Modulation of remifentanil-induced analgesia, hyperalgesia, and tolerance by small-dose ketamine in humans. Anesth Analg. 2003;96:726–32, table of contents. doi: 10.1213/01.ANE.0000048086.58161.18.
    1. Singler B, Troster A, Manering N, Schuttler J, Koppert W. Modulation of remifentanil-induced postinfusion hyperalgesia by propofol. Anesth Analg. 2007;104:1397–403, table of contents. doi: 10.1213/01.ane.0000261305.22324.f3.
    1. Aley KO, Levine JD. Different mechanisms mediate development and expression of tolerance and dependence for peripheral mu-opioid antinociception in rat. J Neurosci. 1997;17:8018–8023.
    1. Khasar SG, McCarter G, Levine JD. Epinephrine produces a beta-adrenergic receptor-mediated mechanical hyperalgesia and in vitro sensitization of rat nociceptors. J Neurophysiol. 1999;81:1104–1112.
    1. Liang DY, Liao G, Wang J, Usuka J, Guo Y, Peltz G, Clark JD. A genetic analysis of opioid-induced hyperalgesia in mice. Anesthesiology. 2006;104:1054–1062. doi: 10.1097/00000542-200605000-00023.
    1. Belanger S, Ma W, Chabot JG, Quirion R. Expression of calcitonin gene-related peptide, substance P and protein kinase C in cultured dorsal root ganglion neurons following chronic exposure to mu, delta and kappa opiates. Neuroscience. 2002;115:441–453. doi: 10.1016/S0306-4522(02)00452-9.
    1. Ibuki T, Marsala M, Masuyama T, Yaksh TL. Spinal amino acid release and repeated withdrawal in spinal morphine tolerant rats. Br J Pharmacol. 2003;138:689–697. doi: 10.1038/sj.bjp.0705102.
    1. Mao J, Sung B, Ji RR, Lim G. Chronic morphine induces downregulation of spinal glutamate transporters: implications in morphine tolerance and abnormal pain sensitivity. J Neurosci. 2002;22:8312–8323.
    1. Vanderah TW, Suenaga NM, Ossipov MH, Malan TP, Jr., Lai J, Porreca F. Tonic descending facilitation from the rostral ventromedial medulla mediates opioid-induced abnormal pain and antinociceptive tolerance. J Neurosci. 2001;21:279–286.
    1. Rapp SE, Ready LB, Nessly ML. Acute pain management in patients with prior opioid consumption: a case-controlled retrospective review. Pain. 1995;61:195–201. doi: 10.1016/0304-3959(94)00168-E.
    1. de Leon-Casasola OA, Lema MJ. Epidural sufentanil for acute pain control in a patient with extreme opioid dependency. Anesthesiology. 1992;76:853–856. doi: 10.1097/00000542-199205000-00025.
    1. Carroll IR, Angst MS, Clark JD. Management of perioperative pain in patients chronically consuming opioids. Reg Anesth Pain Med. 2004;29:576–591. doi: 10.1016/j.rapm.2004.06.009.
    1. Li X, Angst MS, Clark JD. Opioid-induced hyperalgesia and incisional pain. Anesth Analg. 2001;93:204–209. doi: 10.1097/00000539-200107000-00040.
    1. Celerier E, Gonzalez JR, Maldonado R, Cabanero D, Puig MM. Opioid-induced hyperalgesia in a murine model of postoperative pain: role of nitric oxide generated from the inducible nitric oxide synthase. Anesthesiology. 2006;104:546–555. doi: 10.1097/00000542-200603000-00023.
    1. Clark JD, Shi X, Li X, Qiao Y, Liang D, Angst MS, Yeomans DC. Morphine reduces local cytokine expression and neutrophil infiltration after incision. Mol Pain. 2007;3:28. doi: 10.1186/1744-8069-3-28.
    1. Ahn DK, Chae JM, Choi HS, Kyung HM, Kwon OW, Park HS, Youn DH, Bae YC. Central cyclooxygenase inhibitors reduced IL-1beta-induced hyperalgesia in temporomandibular joint of freely moving rats. Pain. 2005;117:204–213. doi: 10.1016/j.pain.2005.06.009.
    1. Inglis JJ, Nissim A, Lees DM, Hunt SP, Chernajovsky Y, Kidd BL. The differential contribution of tumour necrosis factor to thermal and mechanical hyperalgesia during chronic inflammation. Arthritis Res Ther. 2005;7:R807–16. doi: 10.1186/ar1743.
    1. Vissers KC, De Jongh RF, Hoffmann VL, Meert TF. Exogenous interleukin-6 increases cold allodynia in rats with a mononeuropathy. Cytokine. 2005;30:154–159. doi: 10.1016/j.cyto.2005.01.008.
    1. Li X, Angst MS, Clark JD. A murine model of opioid-induced hyperalgesia. Brain Res Mol Brain Res. 2001;86:56–62. doi: 10.1016/S0169-328X(00)00260-6.
    1. Clark JD, Qiao Y, Li X, Shi X, Angst MS, Yeomans DC. Blockade of the complement C5a receptor reduces incisional allodynia, edema, and cytokine expression. Anesthesiology. 2006;104:1274–1282. doi: 10.1097/00000542-200606000-00024.
    1. Liu J, Feng X, Yu M, Xie W, Zhao X, Li W, Guan R, Xu J. Pentoxifylline attenuates the development of hyperalgesia in a rat model of neuropathic pain. Neurosci Lett. 2007;412:268–272. doi: 10.1016/j.neulet.2006.11.022.
    1. Mika J, Osikowicz M, Makuch W, Przewlocka B. Minocycline and pentoxifylline attenuate allodynia and hyperalgesia and potentiate the effects of morphine in rat and mouse models of neuropathic pain. Eur J Pharmacol. 2007;560:142–149. doi: 10.1016/j.ejphar.2007.01.013.
    1. Redondo P, Garcia-Foncillas J, Espana A, Cuevillas F, Quintanilla E. Differential modulation of IL-8 and TNF-alpha expression in human keratinocytes by buflomedil chlorhydrate and pentoxifylline. Exp Dermatol. 1997;6:186–194. doi: 10.1111/j.1600-0625.1997.tb00204.x.
    1. Bruynzeel I, Stoof TJ, Willemze R. Pentoxifylline and skin inflammation. Clin Exp Dermatol. 1998;23:168–172. doi: 10.1046/j.1365-2230.1998.00316.x.
    1. Oberyszyn TM, Tober KL, Ross MS, Robertson FM. Inhibitory effects of pentoxifylline on ultraviolet B light-induced cutaneous inflammation. Mol Carcinog. 1998;22:16–25. doi: 10.1002/(SICI)1098-2744(199805)22:1<16::AID-MC3>;2-J.
    1. King T, Vardanyan A, Majuta L, Melemedjian O, Nagle R, Cress AE, Vanderah TW, Lai J, Porreca F. Morphine treatment accelerates sarcoma-induced bone pain, bone loss, and spontaneous fracture in a murine model of bone cancer. Pain. 2007;132:154–168. doi: 10.1016/j.pain.2007.06.026. Epub 2007 Aug 15.
    1. Liang DY, Shi X, Li X, Li J, Clark JD. The beta2 adrenergic receptor regulates morphine tolerance and physical dependence. Behav Brain Res. 2007;181:118–126. doi: 10.1016/j.bbr.2007.03.037.
    1. Johnston IN, Milligan ED, Wieseler-Frank J, Frank MG, Zapata V, Campisi J, Langer S, Martin D, Green P, Fleshner M, Leinwand L, Maier SF, Watkins LR. A role for proinflammatory cytokines and fractalkine in analgesia, tolerance, and subsequent pain facilitation induced by chronic intrathecal morphine. J Neurosci. 2004;24:7353–7365. doi: 10.1523/JNEUROSCI.1850-04.2004.
    1. Raghavendra V, Rutkowski MD, DeLeo JA. The role of spinal neuroimmune activation in morphine tolerance/hyperalgesia in neuropathic and sham-operated rats. J Neurosci. 2002;22:9980–9989.
    1. Sacerdote P. Opioids and the immune system. Palliat Med. 2006;20 Suppl 1:s9–15.
    1. Dallos A, Kiss M, Polyanka H, Dobozy A, Kemeny L, Husz S. Effects of the neuropeptides substance P, calcitonin gene-related peptide, vasoactive intestinal polypeptide and galanin on the production of nerve growth factor and inflammatory cytokines in cultured human keratinocytes. Neuropeptides. 2006;40:251–263. doi: 10.1016/j.npep.2006.06.002.
    1. Bigliardi-Qi M, Bigliardi PL, Buchner S, Rufli T. Characterization of mu-opiate receptor in human epidermis and keratinocytes. Ann N Y Acad Sci. 1999;885:368–371.
    1. Bigliardi-Qi M, Sumanovski LT, Buchner S, Rufli T, Bigliardi PL. Mu-opiate receptor and Beta-endorphin expression in nerve endings and keratinocytes in human skin. Dermatology. 2004;209:183–189. doi: 10.1159/000079887.
    1. Schmelz M, Paus R. Opioids and the skin: "itchy" perspectives beyond analgesia and abuse. J Invest Dermatol. 2007;127:1287–1289. doi: 10.1038/sj.jid.5700634.
    1. Ahn DK, Kim KH, Jung CY, Choi HS, Lim EJ, Youn DH, Bae YC. Role of peripheral group I and II metabotropic glutamate receptors in IL-1beta-induced mechanical allodynia in the orofacial area of conscious rats. Pain. 2005;118:53–60. doi: 10.1016/j.pain.2005.07.017.
    1. Jung CY, Choi HS, Ju JS, Park HS, Kwon TG, Bae YC, Ahn DK. Central metabotropic glutamate receptors differentially participate in interleukin-1beta-induced mechanical allodynia in the orofacial area of conscious rats. J Pain. 2006;7:747–756. doi: 10.1016/j.jpain.2006.03.007.
    1. Brack A, Rittner HL, Machelska H, Leder K, Mousa SA, Schafer M, Stein C. Control of inflammatory pain by chemokine-mediated recruitment of opioid-containing polymorphonuclear cells. Pain. 2004;112:229–238. doi: 10.1016/j.pain.2004.08.029.
    1. Mossner R, Beckmann I, Hallermann C, Neumann C, Reich K. Granulocyte colony-stimulating-factor-induced psoriasiform dermatitis resembles psoriasis with regard to abnormal cytokine expression and epidermal activation. Exp Dermatol. 2004;13:340–346. doi: 10.1111/j.0906-6705.2004.00190.x.
    1. de Leon-Casasola OA, Lema MJ. Epidural bupivacaine/sufentanil therapy for postoperative pain control in patients tolerant to opioid and unresponsive to epidural bupivacaine/morphine. Anesthesiology. 1994;80:303–309. doi: 10.1097/00000542-199402000-00010.
    1. de Leon-Casasola OA, Myers DP, Donaparthi S, Bacon DR, Peppriell J, Rempel J, Lema MJ. A comparison of postoperative epidural analgesia between patients with chronic cancer taking high doses of oral opioids versus opioid-naive patients. Anesth Analg. 1993;76:302–307.
    1. Peng PW, Tumber PS, Gourlay D. Review article: perioperative pain management of patients on methadone therapy. Can J Anaesth. 2005;52:513–523.
    1. Pogatzki EM, Raja SN. A mouse model of incisional pain. Anesthesiology. 2003;99:1023–1027. doi: 10.1097/00000542-200310000-00041.
    1. Liang DY, Guo T, Liao G, Kingery WS, Peltz G, Clark JD. Chronic pain and genetic background interact and influence opioid analgesia, tolerance, and physical dependence. Pain. 2006;121:232–240. doi: 10.1016/j.pain.2005.12.026.
    1. Liang DY, Liao G, Lighthall GK, Peltz G, Clark DJ. Genetic variants of the P-glycoprotein gene Abcb1b modulate opioid-induced hyperalgesia, tolerance and dependence. Pharmacogenet Genomics. 2006;16:825–835.
    1. Chaplan SR, Bach FW, Pogrel JW, Chung JM, Yaksh TL. Quantitative assessment of tactile allodynia in the rat paw. J Neurosci Methods. 1994;53:55–63. doi: 10.1016/0165-0270(94)90144-9.
    1. Liang D, Li X, Lighthall G, Clark JD. Heme oxygenase type 2 modulates behavioral and molecular changes during chronic exposure to morphine. Neuroscience. 2003;121:999–1005. doi: 10.1016/S0306-4522(03)00483-4.
    1. Liang DY, Li X, Clark JD. Formalin-induced spinal cord calcium/calmodulin-dependent protein kinase II alpha expression is modulated by heme oxygenase in mice. Neurosci Lett. 2004;360:61–64. doi: 10.1016/j.neulet.2004.02.050.
    1. Poree LR, Guo TZ, Kingery WS, Maze M. The analgesic potency of dexmedetomidine is enhanced after nerve injury: a possible role for peripheral alpha2-adrenoceptors. Anesth Analg. 1998;87:941–948. doi: 10.1097/00000539-199810000-00037.

Source: PubMed

3
Předplatit