<<

Review Acta Neurobiol Exp 2011, 71: 129–138

Opioid in peripheral control

Anna Lesniak*, Andrzej W. Lipkowski

Mossakowski Medical Research Centre Polish Academy of Sciences, Warsaw; *Email: [email protected]

Opioids have a long history of therapeutic use as a remedy for various pain states ranging from mild acute nociceptive pain to unbearable chronic advanced or end-stage disease pain. Analgesia produced by classical is mediated extensively by binding to receptors located in the brain or the spinal cord. Nevertheless, opioid receptors are also expressed outside the CNS in the periphery and may become valuable assets in eliciting analgesia devoid of shortcomings typical for the activation of their central counterparts. The discovery of endogenous opioid peptides that participate in the formation, transmission, modulation and perception of pain signals offers numerous opportunities for the development of new . Novel peptidic analogs, which show limited access through the blood brain barrier may support pain therapy requiring prolonged use of opioid drugs. Key words: immune cells, opioid peptides, pain, peripheral analgesia

Abbreviations: β-FNA - β-funaltrexamine, BBB - blood-brain-barrier, CGRP - -related , CFA - complete Freund adjuvant, CNS - central nervous system, CRF - corticotropin releasing factor, CYP - , DAGO - [Tyr-D-Ala- Gly-Me-Phe-Gly-ol]-, DAMGO - [D-Ala2, N-MePhe4, Gly-ol]-enkephalin, DOR - delta opioid receptor, DPDPE - [D-Pen2,5]-enkephalin, DRG - dorsal root ganglion, EM-1 - 1, EM-2 - endomorphin 2, KOR - kappa opioid receptor, MOR - mu opioid receptor, NLZ – , NTI - , NLXM - methiodide; nor-BNI – nor-, PDYN - , PENK - , PNS - peripheral nervous system, POMC -

Introduction ic pain. This effect of opioids is a consequence of their expression patterns in the areas of the CNS responsible The opioid system is one of the main system for sensorimotor integration and cognitive function- engaged in strongly conserved evolutionary mecha- ing. Nevertheless, in some pain states including nerve nisms like pain perception and modulation, reward, damage, painful inflammation, tissue destruction by and fear behaviors (Herz 1998, Inturrisi cancer expansion opioid receptors located in the 2002, Petrovic et al. 2008, Lehner et al. 2010). Opioid periphery play a significant role in the development of receptors and their corresponding are key analgesia. Besides their profound expression in the players in the inhibition and modulation of pain. It has CNS, opioid receptors are also present in the PNS on long been postulated that sufficient clinically-relevant peripheral sensory nerve terminals as well as on other analgesia is obtained exclusively via activation of cen- nonneural tissues such as the vascular epithelium or tral opioid receptors (Lipkowski and Carr 2002). In keratinocytes (see Table I). addition to inducing an effect, typical cen- The expression of opioid receptors and their trally-active opioids are the source of undesirable side enhanced transport to sensory nerve terminals becomes effects which may limit their therapeutic use in chron- prominent especially in the presence of inflammation. Immune cells are then recruited to the damaged tissue Received 20 December, accepted 19 January 2011 and secrete opioid peptides which bind to peripheral Correspondence should be addressed to: A. Lesniak opioid receptors reducing pain. This endogenous pain- Email: [email protected] relief mechanism became an inspiration for the exog-

© 2011 by Polish Neuroscience Society - PTBUN, Nencki Institute of Experimental Biology 130 A. Lesniak and A.W. Lipkowski

Table I

Localization of opioid receptor expression

Expression Opioid receptor type CNS PNS Non-neural tissues

MOR neocortex, caudate – putamen, peripheral sensory neuron vascular endothelium, cardiac , thalamus, DRG, stomach, duodenum, epithelium, keratinocytes, vas hippocampus, amygdala, jejunum, oleum, proximal and deferens, Sertoli cells nucleus tractus solitarius distal colon

DOR olfactory-related areas, peripheral sensory neuron neocortex, caudate – putamen, DRG nucleus accumbens, amygdala

KOR caudate – putamen, nucleus sensory neuron DRG, accumbens,amygdala, neural stomach, duodenum, jejunum, lobe of the pituitary gland oleum, proximal and distal colon enous administration of endogenous, synthetic opioid share high homology but can also exist in several peptides and novel peptidomimetics in the hope to splice variants due to a differential mRNA processing conquer pain. (Knapp et al. 1995, Wei et al. 2004, Wei and Loh 2011). Different pharmacological profiles of the existing opi- Opioid receptor types oid receptors may be a result of posttranslational modifications and dimerization (Bouvier 2001, Levac Opioid receptors belong to the rhodopsin-like sub- 2002, Rios et al. 2001, Gupta et al. 2006). family of GPCR seven-transmembrane domain metabothropic receptors. Ligand binding facilitates Peripheral opioid receptors coupling to the inhibitory Gi/o protein decreasing cAMP levels by inhibiting Ca2+/Na+ influx. This results Peripheral opioid receptors are synthetized in cell in a decrease of releasing of proalgesic mediators such bodies of primary afferent neurons and intra-axonally as , CGRP and nociceptor excitability. The transported to peripheral sensory nerve endings where existence of opioid receptors was first reported in 1973 they can interact with both endogenous and exogenous in a series of radioligand binding experiments where opioid agonists (Rau et al. 2005, Wang et al. 2010). radiolabeled opioid ligands bind to a receptor in the rat Many studies have shown that the role of peripheral brain membrane homogenate (Pert and Snyder 1973, opioid receptors is more pronounced during peripheral Simon et al. 1973, Terenius 1973). The first attempt to inflammation due to a number of inflammation-driven classify “ receptors” into three types was pro- processes. Under normal conditions nerve fibers are posed by Gilbert and Martin (1976) and Lord and encapsulated by a perineurial barrier preventing the coworkers (1977) based on the efficacy of opiate bind- diffusion of high molecular weight particles and ing in chronic spinal dogs and mouse vas deferens. hydrophilic opioid receptors ligands. In pathological Later in the early nineties the DOR (Evans et al. 1992), conditions such as inflammation easier access of opi- MOR (Chen et al. 1993, Thompson et al. 1993) and oid agonists through the perineurium of sprouting KOR (Li et al. 1993, Yasuda et al. 1993, Meng et al. peripheral nerve fibers is observed (Olsson 1990, 1993) were cloned. Although all three types of Antonijevic et al. 1995). Moreover, the expression of opioid receptors are encoded by a different gene they peripheral opioid receptors and their axonal transport Opioid peptides in peripheral pain control 131 to the sensory nerve terminals is enhanced in the is characterized by highest affinity to DOR, moderate course of inflammation (Hassan et al. 1993, Ji et al. to MOR and very low to KOR. show pref- 1995, Schäfer et al. 1995, Pol and Puig 2004). The most erable binding to KOR but possess some MOR and extensively studied upregulation of MOR expression DOR specificity. Two endogenous opioid peptides was observed in animal inflammatory pain models EM-1 and EM-2 also activate opioid receptors but their (Zhang et al. 1998, Ballet et al. 2003, Puehler et al. precursors are not yet known (Terskiy et al. 2007, 2004, Taguchi et al. 2010) and in human inflammatory Perlikowska et al. 2009). Some postulate a de novo bowel disease (Philippe et al. 2004) but downregulated synthesis of those peptides (Rónai et al. 2009). Several in bone cancer pain in mice (Yamamoto et al. 2008). other non-mammalian opioid peptides which show In parallel, data supporting DOR and KOR overex- affinity to opioid receptors have been discovered to pression in inflammation has also been published but date. This includes amphibian and deltor- some results remain contradictory (Ji et al. 1995, phin (Broccardo et al. 1981, Glaser et al. 1981, Barra et Maekawa et al. 1996, Zhang et al. 1998, Shen et al. al. 1994, Negri et al. 2000, Auvznet et al. 2006, Sinha 2005, Puehler et al. 2006). In addition to opioid recep- et al. 2009), opioid peptides formed in the process tor upregulation, inflammation also changes the envi- of milk like (Rüthrich et al. ronment in local tissue via pH decrease rendering 1992) or digestion such as gluteomorphin (Sun opioid receptors more active due to an increased and Cade 2003) and (Takahashi et al. 2000). G-protein coupling and cAMP level (Rasenik and Endogenous opioid peptides are released from Childers 1989, Reddy and Bhargava 1996, Zöllner et immune cells, which in the presence of inflammatory al. 2003, Shaqura et al. 2004). mediators migrate to inflamed tissue in a process Neuropathic pain resulting from mechanical nerve referred to as “homing“ a centrally-mediated mecha- damage is another condition which may involve opioid nism (Mousa et al. 2001, Shmitt et al. 2003, Heurich et receptor expression changes in peripheral sensory neu- al. 2007). Leukocytes roll along the blood vessel wall rons. In many different studies employing different in a process mediated predominantely by L-selectins neuropathic pain models involving entrapment of the and E- and P-selectins present on endothelial cells peripheral nerve MOR, DOR either KOR expression (Machelska et al. 1998). Adhesion of the leukocytes to was observed to be unregulated (Sung et al. 2000, endothelial cells is mediated by integrins, for example Truong et al. 2003, Kabli and Cahill 2007, Walczak et ICAM-1. After a firm adhesion immune cells squeeze al. 2005, Obara et al. 2009). Some studies however through the endothelium and migrate to the inflamma- found especially MOR expression to be downregulated tion site (Butcher and Picker 1996), release opioid in neuropathic pain (Rashid et al. 2004, Pol et al. 2006, peptides which in turn bind to peripheral opioid recep- Obara et al. 2010). tors. Despite some discrepancies there is overall more and are postulated to play a evidence of opioid receptor overexpression or increased leading role in endogenous antinociception. In binding affinity which makes them more susceptible β-endorphin deficient mice a short stressful swim and more accessable for both engogenous and exoge- challenge failed to produce analgesia (Parikh et al. nous opioid agonists. 2011) whereas antibodies against these opioid peptides abolished analgesia upon CRF stimulation of immune Endogenous peripheral opioid cells secreting β-endorphin and enkephalin (Cabot peptide analgesia 2001). The importance of endogenous peripheral analgesia Endogenous opioid peptides are natural ligands has been examined in various animal pain models which bind to opioid receptors during painful inflam- including chronic inflammatory and cancer pain giv- mation, neuropathy or cancer invasion. In mammals ing hope for a potential therapeutic application of this three types of opioid peptides endorphins, enkephalins natural pain-relief mechanism. In rats with an unilat- and dynorphins are synthetized via cleavage of the eral CFA-induced hindpaw inflammation, stress precursor proteins. Endorphins derived from POMC induced by a cold water swim produced analgesia in exhibit a high affinity for MOR and DOR and low the inflamed but not in the contralateral non-inflamed affinity for KOR. PENK is a enkephalin precursor and paw. This analgesic effect was abolished by BBB- 132 A. Lesniak and A.W. Lipkowski permeable naloxone and naloxone methiodide a non- cally inactive doses of opioid peptides and their ana- selective peripherally restricted opioid receptor antag- logs locally into pathologically changed tissue. In the onist. Nevertheless in chronic CFA inflammation (four visceral pain mouse model an intraperitoneal or subcu- days after the injection of CFA) analgesia was exclu- taneous injection of a cyclic EM-1 analog reduced the sively naloxone methiodide reversible. These results number of acetic acid induced writhes. The analgesic support a notion that acute inflammation is both cen- effect was reversed by MOR specific antagonists trally and peripherally mediated whereas during β-FNA and NLZ and peripherally active NLXM but chronic inflammatory pain analgesia is a result of opi- not by nor-BNI and NTI suggesting a predominant oid peptide binding to peripheral opioid receptors involvement of peripheral MOR. Interestingly, EM-1 (Machelska et al. 2003). failed to abolish pain behavior presumably due to a low At the early stages of the development of mouse stability of the parent compound (Bedini et al. 2010). osteosarcoma β-endorphin containing immune cells [Tyr-Pro-Phe-Pro-NH2] is yet another were detected inside and surrounding the tumoral example of an showing high opioid mass. The local administration of non-selective NLXM, receptor affinity and peripheral activity in visceral MOR-selective CYP and DOR-selective NTI produced pain. It is an amide fragment of bovine β- and in the ipsilateral paw. Further confirma- highly specific for MOR but not for DOR (Chang et al. tion of the involvement of immune cell-derived opioid 1981, 1983, Vogel et al. 1996). Morphiceptin has been peptides in tumor pain attenuation was the injection of shown to elicit strong analgesia after intracerebroven- a CRF . Blockade of the CRF tricular injection but not when injected peripherally receptor inhibits opioid peptide release from immune due to rapid degradation. However, a more stable mor- cells and promoted the development of pain (Baamonde phiceptin analog [Tyr-Pro-NMePhe-Pro-NH2] acted et al. 2006). peripherally inhibiting diarrhea in mice (Shook et al. The study conducted by Stein and coworkers (1993) 1989). Chemical modifications of the morphiceptin aimed to target intra-articular opioid peptides present structure led to a synthesis of analogs resistant to pep- in synovia of human patients who underwent tidase degradation displaying a potent supraspinal and arthroscopic knee surgery. Synovia samples were peripheral MOR-mediated analgesia (Hau et al. 2002). found to contain immune cells abundant in β-endorphin Along with their peripheral analgesic effect new mor- and Met-enkephalin as shown by immunohistochemis- phiceptin analogs inhibited gastrointestinal transit in try staining. The blockage of synovial opioid receptors vivo making them interesting novel therapeutics in the by intra-articular naloxone resulted in higher pain treatment of gastrointestinal mobility disorders e.g., scores in numerical rating scale and increased the the irritable bowel syndrome (Gach et al. 2010). demand for supplementary analgesics. These findings [(Tyr-D-Ala-Gly-PheNH-)2], dimeric pep- further confirm the future application of opioid pep- tide analog of enkephalin, is another example of an tides in pain control not only in animal models but also opioid peptide that has limited permeability of an in human patients. intact BBB (Silbert et al. 1991). Biphalin expresses synergic activities with current drugs used in AIDS Exogenous peripheral opioid therapy. Therefore, biphalin has been proposed to be peptide analgesia applied as a component of antiviral HIV multidrug therapies in combination with chronic pain treatment One of the first clinical attempts to target the in AIDS patients as a primary therapeutic target (Tang peripheral opioid receptor system for pain control was et al. 2008). a study which aimed to examine the local analgesic Allodynia is a frequent condition in multiple clinical effect of intra-articular (Stein et al. 1991). conditions (Peyron et al. 1998, 2004, Becerra et al. Low-dose intra-articular morphine produced more 2006, Moller et al. 2006, Witting et al. 2006) when pronounced postoperative analgesia than in patients patients perceive normally innocuous tactile or ther- injected with intravenous morphine. mal stimuli as painful. Over the years numerous rodent The findings confirming the clinically-relevant and models of neuropathy have been developed to help to selective analgesic effect of opioids in inflamed periph- understand the mechanisms governing persistent pain eral tissue triggered ideas to administer low, systemi- resulting from peripheral nerve injury. Animal models Opioid peptides in peripheral pain control 133 of neuropathic pain include: partial ligation of the sci- and completely abolished by NLXM strongly support atic nerve (Seltzer et al. 1990), chronic constriction the hypothesis that this agonists action is peripherally injury (Bennett and Xie 1988), ligation of the L5/L6 and not centrally mediated (Menéndez et al. 2003). spinal nerves (Kim and Chung 1992) or the spared Very similar results were obtained by Baamonde nerve injury model (Decosterd and Wolf 2000). (2006) where the study confirmed peripheral analgesia Although these models differ in the type of injury or elicited additionally by a DOR - DPDPE and the magnitude or duration of pain symptoms they all KOR agonist - U50,488H. The greatest analgesic effect mimic pain conditions seen in humans (Martin et al. was achieved by stimulation of MOR although analge- 2003). sia produced by DAGO as well as DPDE was shorter In an experimental rat model of neuropathy devel- than that after U50,488H administration. oped by loosely tying four ligatures around the sciatic The potential use of peripheral KOR agonists as nerve, locally injected opioid peptides alleviated novel analgesics was encouraged by the observation mechanical allodynia in the von Fray test (Obara et al. that, unlike MOR agonists, KOR agonists do not cause 2004). In the study it was demonstrated that MOR ago- typical morphine-like aversive side effects. The first nists: morphine, DAMGO, EM-1 and EM-2, were suc- generation KOR agonists however were burdened with cessful in relieving neuropathic pain after intraplantar dose limiting neuropsychiatric side effect like dyspho- injection as opposed to a subcutaneous injection. ria as a result of dopamine release inhibition (Donzanti DAMGO had an antiallodynic effect on the injured et al. 1992; Pande et al. 1996). The second generation paw at a relatively low dose compared with endomor- KOR agonists were targeted towards peripheral KORs phins which required much higher doses to induce a to decrease brain penetration (Barber et al. 1994). similar magnitude of analgesia. EM-2 produced a simi- a prototypic peripherally active KOR lar effect as EM-1 at a similar dose range but its effect agonist has been researched for possible treatment for started to diminish 10 min after injection. Interestingly, human irritable bowel syndrome and was shown effec- opioid peptides were more effective in pain relief than tive in clinical trials (Delvaux et al. 2004, Szarka et al. a prototipical – morphine, which analgesic 2007). Unfortunately, apart from peripheral activity, effect was delayed in time. The reason for these differ- oral asimadoline was not devoid of central psychoto- ences in effectiveness between opioid peptides and mimetic effects (Machelska et al. 1999). In order to morphine are still not cleat-cut. Several possible expla- overcome these therapy-limiting difficulties, a third nations have been raised like the involvement of differ- generation of peptidic KOR agonists has been estab- ent subtypes of MOR (Labuz et al. 2002), unequal lished. A hydrophilic structure of these peptides pre- receptor binding and a change in receptor binding vented from passive transport through biological mem- parameters (Patel et al. 2002). The latter arguments branes acting primarily on the periphery. The novel may be supported by the role of activated macrophages D- tetrapeptide CR665 which is now under which gather around the site of injury and take part in clinical development by Cara Therapeutics when the regeneration process in this model (Stoll et al. injected intravenously in nanomolar range elicited 1992). The change in the specific milieu may in turn potent analgesia in the mouse writhing test without affect opioid receptor binding properties and their producing motor impairment (Vanderah et al. 2008). availability for various opioid peptides. Another plau- sible explanation is the fact that morphine readily Conclusions crosses the BBB exerting profound central analgesia but rudimentary analgesia in the periphery due to a In conclusion, growing knowledge of peripheral rapid uptake by the CNS and escape from local tissue. endogenous pain pathways opens a new chapter in Cancer pain can also be in the area of interest when pain control which may bring substantial benefit for discussing antihyperalgesic effect of opioid peptides. pain sufferers. Many animal and human studies have The peritumoral injection of a selective MOR agonist shown a justification for such claims. The development - DAGO inhibits thermal hyperalgesia in a mouse of novel peptidomimetics characterized by increased model of cancer pain produced by intratibial implanta- plasma stability, lower toxicity, high affinity for opioid tion of NCTC 2472 cells. The analgesic effect of receptors is a milestone for researchers aiming for sat- DAGO was only observed in tumor-bearing animals isfactory long-lasting pain relief in patients. 134 A. Lesniak and A.W. Lipkowski

References Cabot PJ (2001) Immune-derived opioids and peripheral anti- nociception. Clin Exp Pharmacol Physiol 28: 230–232. Antonijevic I, Mousa SA, Schäfer M, Stein C (1995) Chang KJ, Lillian A, Hazum E , Cuatrecasas P, Chang JK Perineurial defect and peripheral opioid analgesia in (1981) Morphiceptin (NH4-tyr-Pro-Phe-Pro-COHN2): a inflammation. J Neurosci 15: 165–172. potent and specific agonist for morphine (mu) receptors. Auvynet C, Seddiki N, Dunia I, Nicolas P, Amiche M, Science 212: 75–77. Lacombe C (2006) Post-translational amino acid racem- Chang KJ, Wei ET, Killian A, Chang JK (1983) Potent mor- ization in the frog skin peptide I in the secre- phiceptin analogs: structure activity relationships and mor- tion granules of cutaneous serous glands. Eur J Cell Biol. phine-like activities. J Pharmacol Exp Ther 227: 403–408. 85: 25–34. Chen Y, Mestek A, Liu J, Hurley JA, Yu L (1993) Molecular Baamonde A, Lastra A, Juárez L, García-Suárez O, Meana cloning and functional expression of a µ-opioid receptor A, Hidalgo A, Menéndez L (2006) Endogenous beta-en- from rat brain. Mol Pharmacol 44: 8–12. dorphin induces thermal analgesia at the initial stages of Decosterd I, Woolf CJ (2000) Spared nerve injury: an animal a murine osteosarcoma. Peptides 27: 2778–2785. model of persistent peripheral neuropathic pain. Pain 87: Barber A, Bartoszyk GD, Bender HM, Gottschlich R, 149–158. Greiner HE, Harting J, Mauler F, Minck KO, Murray RD, Delvaux M, Beck A, Jacob J, Bouzamondo H, Weber FT, Simon M (1994) A pharmacological profile of the novel, Frexinos J (2004) Effect of asimadoline, a kappa opioid peripherally-selective kappa-opioid receptor agonist, agonist, on pain induced by colonic distension in patients EMD 61753. Br J Pharmacol 113: 1317–1327. with irritable bowel syndrome. Aliment Pharmacol Therap Barra D, Mignogna G, Simmaco M, Pucci P, Severini C, 20: 237–246. Falconieri-Erspamer G, Negri L, Erspamer V (1994) Donzanti BA, Althaus JS, Payson MM, Von Voigtlander PF [D-Leu2]deltorphin, a 17 amino acid opioid peptide from (1992) Kappa agonist-induced reduction in dopamine the skin of the Brazilian hylid frog, Phyllomedusa bur- release: site of action and tolerance. Res Commun Chem meisteri. Peptides 15: 199–202. Pathol Pharmacol 78: 193–210. Ballet S, Conrath M, Fischer J, Kaneko T, Hamon M, Evans CJ, Keith DE, Morrison H, Magendzo K, Edwards Cesselin F (2003) Expression and G-protein coupling of RH (1992) cloning of a delta opioid receptor by func- mu-opioid receptors in the spinal cord and dorsal root tional expression. Science 258: 1952–1955. ganglia of polyarthritic rats. 37: 211–219. Gach K, do-Rego JC, Fichna J, Storr M, Delbro D, Toth G, Becerra L, Morris S, Bazes S, Gostic R, Sherman S, Gostic J, Janecka A (2010) Synthesis and biological evaluation of Pendse G, Moulton E, Scrivani S, Keith D, Chizh B, novel peripherally active morphiceptin analogs. Peptides Borsook D (2006) Trigeminal neuropathic pain alters 31: 1617–1624. responses in CNS circuits to mechanical (brush) and ther- Gilbert PE, Martin WR (1976) The effects of morphine- and mal (cold and heat) stimuli. J Neurosci 26: 10646–10657. naltorphine-like drugs in the nondependent morphine- Bedini A,, Baiula M, Gentilucci L, Tolomelli A, De Marco dependent and -dependent chronic spinal dog. R, Spampinato S (2010) Peripheral antinociceptive effects J Pharmacol Exp Ther 198: 66–82. of the cyclic endomorphin-1 analog c[YpwFG] in a Glaser T, Hübner K, de Castiglione R, Hamprecht B (1981) mouse visceral pain model. Peptides 31: 2135–2140. , opioid peptides from amphibian skin, act Bennett GJ, Xie YK (1988) A peripheral mononeuropathy in on opioid receptors of mouse neuroblastoma x rat glioma rat that produces disorders of pain sensation like those hybrid cells. J Neurochem 37: 1613–1617. seen in man. Pain 33: 87–107. Gupta A, Décaillot FM, Devi LA (2006) Targeting opioid Bouvier M (2001) Oligomerization of G-protein-coupled trans- receptor heterodimers: strategies for screening and drug mitter μ, σ, κ, receptors. Nat Rev Neurosci 2: 274–286. development. AAPS J 8: E153–E159. Broccardo M, Erspamer V, Falconieri Erspamer G, Improta Hassan AH, Ableitner A, Stein C, Herz A (1993) Inflammation G, Linari G, Melchiorri P, Montecucchi PC (1981) of the rat paw enhances axonal transport of opioid recep- Pharmacological data on dermorphins, a new class of tors in the sciatic nerve and increases their density in the potent opioid peptides from amphibian skin. Br J inflamed tissue. Neurosci 55: 185–195. Pharmacol 73: 625–631. Hau VS, Huber JD, Campos CR, Lipkowski AW, Misicka A, Butcher EC, Picker LJ (1996) Lymphocyte homing and Davis TP (2002) Effect of guanidino modification and homeostasis. Science 272: 60–66. proline substitution on the in vitro stability and blood- Opioid peptides in peripheral pain control 135

brain barrier permeability of endomorphin II. J Pharm Sci and inflammation in rats and humans. J Pharmacol Exp 91: 2140–2149. Ther 290: 354–361. Herz A (1998) Opioid reward mechanisms: a key role in Machelska H, Schopohl JK, Mousa SA, Labuz D, Schäfer drug abuse? Can J Physiol Pharmacol 76: 252–258. M, Stein C (2003) Different mechanisms of intrinsic pain Heurich M, Mousa SA, Lenzner M, Morciniec P, Kopf inhibition in early and late inflammation. J Neuroimmunol A,Welte, M, Stein C (2007) Influence of pain treatmentby 141: 30–39. epidural and bupivacaine on homing of opioid- Maekawa K, Minami M, Masuda T, Satoh M (1996) containing leukocytes to surgical wounds. Brain Behav Expression of mu- and kappa-, but not delta-, opioid Immun 21: 544–552. receptor mRNAs is enhanced in the spinal dorsal horn of Inturrisi CE (2002) Clinical pharmacology of opioids for the arthritic rats. Pain 64: 365–371. pain. Clin J Pain 18: S3–S13. Martin WJ, Stewart LA, Tarpley JW (2003) Animal models Ji RR, Zhang Q, Law PY, Low HH, Elde R, Hökfelt T (1995) of neuropathic pain. Method Mol Med 84: 233–242. Expression of mu-, delta-, and kappa-opioid receptor-like Menéndez L, Lastra A, Fresno MF, Llames S, Meana A, immunoreactivities in rat dorsal root ganglia after carra- Hidalgo A, Baamonde A (2003) Initial thermal heat geenan-induced inflammation. J Neurosci 15: 8156–8166. hypoalgesia and delayed hyperalgesia in a murine model Kabli N, Cahill CM (2007) Anti-allodynic effects of peripheral of bone cancer pain. Brain Res 969: 102–109. delta opioid receptors in neuropathic pain. Pain 127: 84–93. Meng F, Xie GX, Thompson RC, Mansour A, Goldstein A, Kim SH, Chung JM (1992) An experimental model for Watson SJ, Akil H (1993) Cloning and pharmacological peripheral neuropathy produced by segmental spinal characterization of a rat kappa opioid receptor. Proc Natl nerve ligation in the rat. Pain 50: 355–363. Acad Sci USA 90: 9954–9958. Knapp RJ, Malatynska E, Collins N, Fang L, Wang JY, Moller AT, Feldt-Rasmussen U, Rasmussen AK, Sommer C, Hruby VJ, Roeske WR, Yamamura HI (1995) Molecular Hasholt L, Bach FW, Kølvraa S, Jensen TS (2006) Small- biology and pharmacology of cloned opioid receptors. fibre neuropathy in female Fabry patients: reduced allo- Faseb J 9: 516–525. dynia and skin blood flow after topical . J Labuz D, Przewlocki R, Przewlocka B (2002) Cross- Peripher Nerv Syst 11: 119–125. tolerance between the different mu-opioid receptor ago- Mousa SA, Zhang Q, Sitte N, Ji RR, Stein C (2001) nists endomorphin-1, endomorphin-2 and morphine at the β-Endorphin-containing memory-cells and m-opioid spinal level in the rat, Neurosci Lett 334: 127–130. receptors undergo transport to peripheral inflamed tissue. Lehner M, Wislowska-Stanek A, Maciejak P, Szyndler J, J Neuroimmunol 115: 71–78. Sobolewska A, Krzascik P, Plaznik A (2010) The relation- Negri L, Melchiorri P, Lattanzi R (2000) Pharmacology of ship between pain sensitivity and conditioned fear amphibian opiate peptides. Peptides. 21: 1639–1647. response in rats. Acta Neurobiol Exp (Wars) 70: 56–66. Obara I, Przewlocki R, B Przewlocka B (2004) Local Levac B, O’Dowdand BF, George SR (2002) Oligomerization peripheral effects of m-opioid receptor agonists in neuro- of opioid receptors: generation of novel signaling units. pathic pain in rats. Neurosci Lett 360: 85–89. Curr Opin Pharmacol 2: 76–81. Obara I, Parkitna JR, Korostynski M, Makuch W, Kaminska Li S, Zhu J, Chen G, Chen YW, Deriel JK, Ashby B, Liu- D, Przewlocka B, Przewlocki R (2009) Local peripheral Chen LY (1993) Molecular cloning and expression of a opioid effects and expression of opioid genes in the spinal rat κ opioid receptor. Biochem J 295: 629–633. cord and dorsal root ganglia in neuropathic and inflam- Lipkowski AW, Carr DB (2002) Rethinking opioid equiva- matory pain. Pain 141: 283–291. lence. Pain-Clinical Updates 10: 1–7. Obara I, Gunduz Cinar O, Starowicz K, Benyhe S, Borsodi Lord JAH, Waterfield AA, Hughes J, Kosterlitz HW (1997) A, Przewlocka B (2010) Agonist-dependent attenuation Endogenous opioid peptides: multiple agonists and recep- of mu-opioid receptor-mediated G-protein activation in tors. Nature 267: 495–499. the dorsal root ganglia of neuropathic rats. J Neural Machelska H, Cabot PJ, Mousa SA, Zhang Q, Stein C Transm. 117: 421–429. (1998) Pain control in inflammation governed by selec- Olsson Y (1990) Microenvironment of the peripheral ner- tins. Nat Med 4: 1425–1428. vous system under normal and pathological conditions. Machelska H, Pfluger M, Weber W, Piranvisseh-Volk M, Crit Rev Neurobiol 5: 265–311. Daubert JD, Dehaven R, Stein C (1999). Peripheral Pande AC, Pyke RE, Greiner M, Wideman GL, Benjamin R, effects of the kappa-opioid agonist EMD 61753 on pain Pierce MW (1996) Analgesic efficacy of versus 136 A. Lesniak and A.W. Lipkowski

placebo or morphine in postsurgical pain. Clin root ganglia in response to peripheral inflammation. Neuropharmacol 19: 451–456. Neurosci 141: 989–998. Patel MB, Patel CN, Rajashekara V, Yoburn BC (2002) Rasenick MM, Childers SR (1989) Modification of Opioid agonists differentially regulate mu-opioid recep- Gs-stimulated adenylate cyclase in brain membranes by tors and trafficking proteins in vivo. Mol Pharmacol 62: low pH pretreatment: correlation with altered guanine 1464–1470. nucleotide exchange. J Neurochem 53: 219–225. Parikh D, Hamid A, Friedman TC, Nguyen K, Tseng A, Rashid MH, Inoue M, Toda K, Ueda H (2004) Loss of Marquez P, Lutfy K (2011) Stress-induced analgesia and peripheral morphine analgesia contributes to the reduced endogenous opioid peptides: the importance of stress effectiveness of systemic morphine in neuropathic pain. J duration. Eur J Pharmacol 650: 563–567. Pharmacol Exp Ther 309: 380–387. Perlikowska R, Fichna J, Janecka A (2009) - Rau KK, Caudle RM, Cooper BY, Johnson RD (2005) endogenous ligands of the mu-opioid receptor. Postepy Diverse immunocytochemical expression of opioid recep- Biochem 55: 388–394. tors in electrophysiologically defined cells of rat dorsal Pert CB, Snyder SH (1973) Opiate receptor: demonstration root ganglia. J Chem Neuroanat 29: 255–264. in nervous tissue. Science 179: 1011–1014. Reddy PL, Bhargava HN (1996) Effect of low pH treatment Petrovic P, Pleger B, Seymour B, Klöppel S, De Martino B, on opioid peptides binding to their receptors and func- Critchley H, Dolan RJ (2008) Blocking central opiate tional coupling of G-proteins to adenylyl cyclase in the function modulates hedonic impact and anterior cingulate rat spinal cord. Peptides 17: 625–628. response to rewards and losses. J Neurosci 28: 10509– Rónai AZ, Király K, Szebeni A, Szemenyei E, Prohászka Z, 10516. Darula Z, Tóth G, Till I, Szalay B, Kató E, Barna I (2009) Peyron R, García-Larrea L, Grégoire MC, Convers P, Immunoreactive endomorphin 2 is generated extracellu- Lavenne F, Veyre L, Froment JC, Mauguière F, Michel D, larly in rat isolated L4,5 dorsal root ganglia by DPP-IV. Laurent B (1998) Allodynia after lateral-medullary Regul Pept 157: 1–2. (Wallenberg) infarct. A PET study. Brain 121: 345–356. Rüthrich HL, Grecksch G, Schmidt R, Neubert K (1992) Peyron R, Schneider F, Faillenot I, Convers P, Barral FG, Linear and cyclic beta analogues with high Garcia-Larrea L, Laurent MD (2004) An fMRI study of analgesic activity. Peptides 13: 483–485. cortical representation of mechanical allodynia in patients Rios CD, Jordan BA, Gomes I, Devi LA (2001) G-protein with neuropathic pain. Neurology 63: 1838–1846. receptor dimerization: Modulation of receptor function. Philippe D, Chakass D, Thuru X, Zerbib P, Tsicopoulos A, Pharmacol Ther 92: 71–87. Geboes K, Bulois P, Breisse M, Vorng H, Gay J, Colombel Schäfer M, Imai Y, Uhl GR, Stein C (1995) Inflammation JF, Desreumaux P, Chamaillard M (2004) Mu opioid enhances peripheral µ-opioid analgesia, but not µ-opioid receptor expression is increased in inflammatory bowel receptor transcription in dorsal root ganglia. Eur J diseases: implications for homeostatic intestinal inflam- Pharmacol 279: 165–169. mation. Gut 55: 815–23. Schmitt TK, Mousa SA, Brack A, Schmidt DK, Rittner HL, Pol O, Puig MM (2004) Expression of opioid receptors dur- Welte M, Schäfer M, Stein C (2003) Modulation of ing peripheral inflammation. Curr Top Med Chem 4: peripheral endogenous opioid analgesia by central affer- 51–61. ent blockade. Anesthesiology 98: 195–202. Pol O, Murtra P, Caracuel L, Valverde O, Puig MM, Seltzer Z, Dubner R, Shir Y (1990) A novel behavioral Maldonado R (2006) Expression of opioid receptors and model ofneuropathic pain disorders produced in rats by c-fos in CB1 knockout mice exposed to neuropathic pain. partial sciatic nerve injury. Pain 43: 205–218. Neuropharmacol 50: 123–132. Shaqura MA, Zöllner C, Mousa SA, Stein C, and Schäfer M Puehler W, Zöllner C, Brack A, Shaqura MA, Krause H, (2004) Characterization of μ opioid receptor binding and Schäfer M, Stein C (2004) Rapid upregulation of mu G-protein coupling in rat hypothalamus, spinal cord and opioid receptor mRNA in dorsal root ganglia in response primary afferent neurons during inflammatory pain. J to peripheral inflammation depends on neuronal conduc- Pharmacol Exp Ther 308: 712–718. tion. Neurosci 129: 473–479. Shen H, Aeschlimann A, Reisch N, Gay RE, Simmen BR, Puehler W, Rittner HL, Mousa SA, Brack A, Krause H, Stein Michel BA, Gay S, Sprott H (2005) Kappa and delta C, Schafer M (2006) Interleukin-1 beta contributes to the opioid receptors are expressed but down-regulated in upregulation of kappa opioid receptor mRNA in dorsal fibroblast-like synoviocytes of patients with rheuma- Opioid peptides in peripheral pain control 137

toid arthritis and osteoarthritis. Arthritis Rheum 52: with biphalin and AZT for Friend leukemia virus infec- 1402–1410. tion in vitro. Pharmacol Rep 60: 190–198. Shook JE, Lemcke PK, Gehrig CA, Hruby VJ, Burks TF Taguchi R, Taguchi T, Kitakoji H (2010) Involvement of (1989) Antidiarrheal properties of supraspinal mu and peripheral opioid receptors in electroacupuncture analge- delta and peripheral mu, delta and kappa opioid receptors: sia for carrageenan-induced hyperalgesia. Brain Res inhibition of diarrhea without constipation. J Pharmacol 1355: 97–103. Exp Ther 249: 83–90. Terenius L (1973) Characteristics of the “receptor” for nar- Silbert BS, Lipkowski AW, Cepeda MS, Szyfelbein SK, cotic analgesics in synaptic plasma membrane fraction Osgood PF, Carr DB (1991) Analgesic activity of a novel from rat brain. Acta Pharmacol Toxicol 33: 377–384. bivalent opioid peptide compared to morphine via differ- Terskiy A, Wannemacher KM, Yadav PN, Tsai M, Tian B, ent routes of administration. Agent Action 33: 382–387. Howells RD (2007) Search of the human proteome for Simon EJ, Hiller JM, Edelman I (1973) Stereospecific bind- endomorphin-1 and endomorphin-2 precursor proteins. ing of the potent analgesic [H3] to rat Life Sci 81:1593–1601. brain homogenate. Proc Natl Acad Sci USA 70: 1947– Thompson RC, Mansour A, Akil H, Watson SJ (1993) 1949. Cloning and pharmacological characterization of a rat µ Sinha B, Cao Z, Murray TF, Aldrich JV (2009) Discovery of opioid receptor. Neuron 11: 903–913. dermorphin-based affinity labels with subnanomolar Truong W, Cheng C, Xu QG, Li XQ, Zochodne DW (2003) affinity for mu opioid receptors. J Med Chem 52: 7372– Mu opioid receptors and analgesia at the site of a periph- 7375. eral nerve injury. Ann Neurol 53: 366–375. Stein C, Comisel K, Haimerl E, Yassouridis A, Lehrberger Vanderah TW, Largent-Milnes T, Lai J, Porreca F, Houghten K, Herz A, Peter K (1991) Analgesic effect of in­traarticular RA, Menzaghi F, Wisniewski K, Stalewski J, Sueiras-Diaz morphine after arthroscopic knee surgery. N Engl J Med J, Galyean R, Schteingart C, Junien JL, Trojnar J, Rivière 325: 1123–1126. PJM (2008) Novel D-amino acid tetrapeptides produce Stein C, Hassan AH, Lehrberger K, Giefing J, Yassouridis A potent antinociception by selectively acting at peripheral (1993) Local analgesic effect of endogenous opioid pep- κ-opioid receptors. Eur J Pharmacol 583: 62–72. tides. Lancet 342: 321–324. Vogel D, Schmidt R, Hartung K, Demuth HU, Chung NN, Stoll G, Hartung HP (1992) The role of macrophages in Schiller PW (1996) Cyclic morphiceptin analogs: cycl- degeneration and immune mediated demyelination of the ization studies and opioid activities in vitro. Int J Pept peripheral nervous system. Adv Neuroimmunol 2: 163– Protein Res 48: 495–502. 179. Walczak JS, Pichette V, Leblond F, Desbiens K, Beaulieu P Sun Z, Cade R (2003) Findings in normal rats following (2005) Behavioral, pharmacological and molecular char- administration of -7 (GD-7). Peptides 24: acterization of the saphenous nerve partial ligation: a new 321–323. model of neuropathic pain. Neurosci 132: 1093–1102. Sung B, Loh HH, Wei L (2000) Association of kappa opioid Wanga HB, Zhaoa B, Zhonga YQ, Lia KC, Lib ZY, , Wang receptor mRNA upregulation in dorsal root ganglia with Q, , Lua YJ, Zhang ZN, Hea SQ, Zheng HC, Wud SX, mechanical allodynia in mice following nerve injury. Hökfelte GM, Baob L, Zhanga X (2010) Coexpression of Neurosci Lett 291: 163–166. δ- and μ-opioid receptors in nociceptive sensory neurons. Szarka LA, Camilleri M, Burton D, Fox JC, McKinzie S, PNAS 29: 13117–13122. Stanislav T, Simonson J, Sullivan N, Zinsmeister AR Wei LN, Law PY, Loh HH (2004) Post-transcriptional regu- (2007) Efficacy of on-demand asimadoline, a peripheral lation of opioid receptors in the nervous system. Front kappa-opioid agonist, in females with irritable bowel Biosci 9: 1665–1679. syndrome. Clin Gastroenterol Hepatol 5: 1268–1275. Wei LN, Loh HH (2011) Transcriptional and Epigenetic Takahashi M, Fukunaga H, Kaneto H, Fukudome S, Regulation of Opioid Receptor Genes-Present and Future. Yoshikawa M (2000) Behavioral and pharmacological Annu Rev Pharmacol Toxicol 51: 75–97. studies on gluten exorphin A5, a newly isolated bioactive Witting N, Kupers RC, Svensson P, Jensen TS (2006) A PET food protein fragment, in mice. Jpn J Pharmacol 84: activation study of brush-evoked allodynia in patients 259–265. with nerve injury pain. Pain 120: 145–54. Tang JL, Lipkowski AW, Specter S (2008) Molecular assess- Yamamoto J, Kawamata T, Niiyama Y, Omote K, Namiki A ment of the potential combination therapy of cytokines (2008) Down-regulation of mu opioid receptor expres- 138 A. Lesniak and A.W. Lipkowski

sion within distinct subpopulations of dorsal root gangli- Zhang Q, Schäfer M, Elde R, Stein C (1998) Effects of neuro- on neurons in a murine model of bone cancer pain. toxins and hindpaw inflammation on opioid receptor immu- Neurosci 151: 843–53. noreactivities in dorsal root ganglia. Neurosci 85: 281–291. Yasuda K, Raynor K, Kong H, Breder CD, Takeda J, Reisine Zöllner C, Shaqura MA, Bopaiah CP, Mousa S, Stein C, and T, Bell GI (1993). Cloning and functional comparison of Schäfer M (2003) Painful inflammation-induced increase n and y opioid receptors from mouse brain. Proceedings in μ-opioid receptor binding and G-protein coupling in Natl Acad Sci USA 90: 6736– 6740. primary afferent neurons. Mol Pharmacol 64: 202–210.