Skip to main content
Top
Published in: Journal of Gastroenterology 1/2014

Open Access 01-01-2014 | Review

Physiology, signaling, and pharmacology of opioid receptors and their ligands in the gastrointestinal tract: current concepts and future perspectives

Authors: Marta Sobczak, Maciej Sałaga, Martin A. Storr, Jakub Fichna

Published in: Journal of Gastroenterology | Issue 1/2014

Login to get access

Abstract

Opioid receptors are widely distributed in the human body and are crucially involved in numerous physiological processes. These include pain signaling in the central and the peripheral nervous system, reproduction, growth, respiration, and immunological response. Opioid receptors additionally play a major role in the gastrointestinal (GI) tract in physiological and pathophysiological conditions. This review discusses the physiology and pharmacology of the opioid system in the GI tract. We additionally focus on GI disorders and malfunctions, where pathophysiology involves the endogenous opioid system, such as opioid-induced bowel dysfunction, opioid-induced constipation or abdominal pain. Based on recent reports in the field of pharmacology and medicinal chemistry, we will also discuss the opportunities of targeting the opioid system, suggesting future treatment options for functional disorders and inflammatory states of the GI tract.
Literature
1.
go back to reference Pert CB, Snyder SH. Opiate receptor: demonstration in nervous tissue. Science. 1973;179:1011–4.PubMed Pert CB, Snyder SH. Opiate receptor: demonstration in nervous tissue. Science. 1973;179:1011–4.PubMed
2.
go back to reference Lord JA, Waterfield AA, Hughes J, Kosterlitz HW. Endogenous opioid peptides: multiple agonists and receptors. Nature. 1977;267:495–9.PubMed Lord JA, Waterfield AA, Hughes J, Kosterlitz HW. Endogenous opioid peptides: multiple agonists and receptors. Nature. 1977;267:495–9.PubMed
3.
go back to reference Dupre DJ, Robitaille M, Rebois RV, Hebert TE. The role of Gbetagamma subunits in the organization, assembly, and function of GPCR signaling complexes. Annu Rev Pharmacol Toxicol. 2009;49:31–56.PubMedPubMedCentral Dupre DJ, Robitaille M, Rebois RV, Hebert TE. The role of Gbetagamma subunits in the organization, assembly, and function of GPCR signaling complexes. Annu Rev Pharmacol Toxicol. 2009;49:31–56.PubMedPubMedCentral
4.
go back to reference Piros ET, Hales TG, Evans CJ. Functional analysis of cloned opioid receptors in transfected cell lines. Neurochem Res. 1996;21:1277–85.PubMed Piros ET, Hales TG, Evans CJ. Functional analysis of cloned opioid receptors in transfected cell lines. Neurochem Res. 1996;21:1277–85.PubMed
5.
go back to reference Evans CJ, Keith DE Jr, Morrison H, Magendzo K, Edwards RH. Cloning of a delta opioid receptor by functional expression. Science. 1992;258:1952–5.PubMed Evans CJ, Keith DE Jr, Morrison H, Magendzo K, Edwards RH. Cloning of a delta opioid receptor by functional expression. Science. 1992;258:1952–5.PubMed
6.
go back to reference Li S, Zhu J, Chen C, Chen YW, Deriel JK, Ashby B, et al. Molecular cloning and expression of a rat kappa opioid receptor. Biochem J. 1993;295:629–33.PubMed Li S, Zhu J, Chen C, Chen YW, Deriel JK, Ashby B, et al. Molecular cloning and expression of a rat kappa opioid receptor. Biochem J. 1993;295:629–33.PubMed
7.
go back to reference Chen Y, Mestek A, Liu J, Yu L. Molecular cloning of a rat kappa opioid receptor reveals sequence similarities to the mu and delta opioid receptors. Biochem J. 1993;295:625–8.PubMed Chen Y, Mestek A, Liu J, Yu L. Molecular cloning of a rat kappa opioid receptor reveals sequence similarities to the mu and delta opioid receptors. Biochem J. 1993;295:625–8.PubMed
8.
go back to reference Fukuda K, Kato S, Mori K, Nishi M, Takeshima H. Primary structures and expression from cDNAs of rat opioid receptor delta- and mu-subtypes. FEBS Lett. 1993;327:311–4.PubMed Fukuda K, Kato S, Mori K, Nishi M, Takeshima H. Primary structures and expression from cDNAs of rat opioid receptor delta- and mu-subtypes. FEBS Lett. 1993;327:311–4.PubMed
9.
go back to reference Dietis N, Rowbotham DJ, Lambert DG. Opioid receptor subtypes: fact or artifact? Br J Anaesth. 2011;107:8–18.PubMed Dietis N, Rowbotham DJ, Lambert DG. Opioid receptor subtypes: fact or artifact? Br J Anaesth. 2011;107:8–18.PubMed
10.
go back to reference Zollner C, Stein C. Opioids. Handb Exp Pharmacol. 2007;177:31–63.PubMed Zollner C, Stein C. Opioids. Handb Exp Pharmacol. 2007;177:31–63.PubMed
11.
go back to reference Jordan BA, Cvejic S, Devi LA. Opioids and their complicated receptor complexes. Neuropsychopharmacology. 2000;23:S5–18.PubMed Jordan BA, Cvejic S, Devi LA. Opioids and their complicated receptor complexes. Neuropsychopharmacology. 2000;23:S5–18.PubMed
12.
go back to reference Chaturvedi K, Christoffers KH, Singh K, Howells RD. Structure and regulation of opioid receptors. Biopolymers. 2000;55:334–46.PubMed Chaturvedi K, Christoffers KH, Singh K, Howells RD. Structure and regulation of opioid receptors. Biopolymers. 2000;55:334–46.PubMed
13.
go back to reference Befort K, Tabbara L, Bausch S, Chavkin C, Evans C, Kieffer B. The conserved aspartate residue in the third putative transmembrane domain of the delta-opioid receptor is not the anionic counterpart for cationic opiate binding but is a constituent of the receptor binding site. Mol Pharmacol. 1996;49:216–23.PubMed Befort K, Tabbara L, Bausch S, Chavkin C, Evans C, Kieffer B. The conserved aspartate residue in the third putative transmembrane domain of the delta-opioid receptor is not the anionic counterpart for cationic opiate binding but is a constituent of the receptor binding site. Mol Pharmacol. 1996;49:216–23.PubMed
14.
go back to reference Surratt CK, Johnson PS, Moriwaki A, Seidleck BK, Blaschak CJ, Wang JB, et al. mu opiate receptor. Charged transmembrane domain amino acids are critical for agonist recognition and intrinsic activity. J Biol Chem. 1994;269:20548–53.PubMed Surratt CK, Johnson PS, Moriwaki A, Seidleck BK, Blaschak CJ, Wang JB, et al. mu opiate receptor. Charged transmembrane domain amino acids are critical for agonist recognition and intrinsic activity. J Biol Chem. 1994;269:20548–53.PubMed
15.
go back to reference Chan AS, Law PY, Loh HH, Ho PN, Wu WM, Chan JS, et al. The first and third intracellular loops together with the carboxy terminal tail of the delta-opioid receptor contribute toward functional interaction with Galpha16. J Neurochem. 2003;87:697–708.PubMed Chan AS, Law PY, Loh HH, Ho PN, Wu WM, Chan JS, et al. The first and third intracellular loops together with the carboxy terminal tail of the delta-opioid receptor contribute toward functional interaction with Galpha16. J Neurochem. 2003;87:697–708.PubMed
16.
go back to reference Reisine T, Law SF, Blake A, Tallent M. Molecular mechanisms of opiate receptor coupling to G proteins and effector systems. Ann N Y Acad Sci. 1996;780:168–75.PubMed Reisine T, Law SF, Blake A, Tallent M. Molecular mechanisms of opiate receptor coupling to G proteins and effector systems. Ann N Y Acad Sci. 1996;780:168–75.PubMed
17.
go back to reference Clark MJ, Neubig RR, Traynor JR. Endogenous regulator of G protein signaling proteins suppress Galphao-dependent, mu-opioid agonist-mediated adenylyl cyclase supersensitization. J Pharmacol Exp Ther. 2004;310:215–22.PubMed Clark MJ, Neubig RR, Traynor JR. Endogenous regulator of G protein signaling proteins suppress Galphao-dependent, mu-opioid agonist-mediated adenylyl cyclase supersensitization. J Pharmacol Exp Ther. 2004;310:215–22.PubMed
18.
go back to reference Law PY, Wong YH, Loh HH. Molecular mechanisms and regulation of opioid receptor signaling. Annu Rev Pharmacol Toxicol. 2000;40:389–430.PubMed Law PY, Wong YH, Loh HH. Molecular mechanisms and regulation of opioid receptor signaling. Annu Rev Pharmacol Toxicol. 2000;40:389–430.PubMed
19.
go back to reference Piros ET, Prather PL, Law PY, Evans CJ, Hales TG. Voltage-dependent inhibition of Ca2+ channels in GH3 cells by cloned mu- and delta-opioid receptors. Mol Pharmacol. 1996;50:947–56.PubMed Piros ET, Prather PL, Law PY, Evans CJ, Hales TG. Voltage-dependent inhibition of Ca2+ channels in GH3 cells by cloned mu- and delta-opioid receptors. Mol Pharmacol. 1996;50:947–56.PubMed
20.
go back to reference Georgoussi Z, Georganta EM, Milligan G. The other side of opioid receptor signalling: regulation by protein–protein interaction. Curr Drug Targets. 2012;13:80–102.PubMed Georgoussi Z, Georganta EM, Milligan G. The other side of opioid receptor signalling: regulation by protein–protein interaction. Curr Drug Targets. 2012;13:80–102.PubMed
21.
go back to reference Lefkowitz RJ, Shenoy SK. Transduction of receptor signals by beta-arrestins. Science. 2005;308:512–7.PubMed Lefkowitz RJ, Shenoy SK. Transduction of receptor signals by beta-arrestins. Science. 2005;308:512–7.PubMed
22.
go back to reference Maguma H, Thayne K, Taylor DA. Characteristics of tolerance in the guinea pig ileum produced by chronic in vivo exposure to opioid versus cannabinoid agonists. Biochem Pharmacol. 2010;80:522–32.PubMed Maguma H, Thayne K, Taylor DA. Characteristics of tolerance in the guinea pig ileum produced by chronic in vivo exposure to opioid versus cannabinoid agonists. Biochem Pharmacol. 2010;80:522–32.PubMed
23.
go back to reference Bohn LM, Gainetdinov RR, Lin FT, Lefkowitz RJ, Caron MG. Mu-opioid receptor desensitization by beta-arrestin-2 determines morphine tolerance but not dependence. Nature. 2000;408:720–3.PubMed Bohn LM, Gainetdinov RR, Lin FT, Lefkowitz RJ, Caron MG. Mu-opioid receptor desensitization by beta-arrestin-2 determines morphine tolerance but not dependence. Nature. 2000;408:720–3.PubMed
24.
go back to reference Fernandez Robles CR, Degnan M, Candiotti KA. Pain and genetics. Curr Opin Anaesthesiol. 2012;25:444–9. Fernandez Robles CR, Degnan M, Candiotti KA. Pain and genetics. Curr Opin Anaesthesiol. 2012;25:444–9.
25.
go back to reference Levran O, Yuferov V, Kreek MJ. The genetics of the opioid system and specific drug addictions. Hum Genet. 2012;131:823–42.PubMedPubMedCentral Levran O, Yuferov V, Kreek MJ. The genetics of the opioid system and specific drug addictions. Hum Genet. 2012;131:823–42.PubMedPubMedCentral
26.
go back to reference Raynor K, Kong H, Mestek A, Bye LS, Tian M, Liu J, et al. Characterization of the cloned human mu opioid receptor. J Pharmacol Exp Ther. 1995;272:423–8.PubMed Raynor K, Kong H, Mestek A, Bye LS, Tian M, Liu J, et al. Characterization of the cloned human mu opioid receptor. J Pharmacol Exp Ther. 1995;272:423–8.PubMed
27.
go back to reference Chadzinska M. Opioid system and innate immunity. Comparative studies. I. Opioids and opioid receptors. Adv Cell Biol. 2007;34:251–62. Chadzinska M. Opioid system and innate immunity. Comparative studies. I. Opioids and opioid receptors. Adv Cell Biol. 2007;34:251–62.
28.
go back to reference Cruz-Gordillo P, Fedrigo O, Wray GA, Babbitt CC. Extensive changes in the expression of the opioid genes between humans and chimpanzees. Brain Behav Evol. 2010;76:154–62.PubMed Cruz-Gordillo P, Fedrigo O, Wray GA, Babbitt CC. Extensive changes in the expression of the opioid genes between humans and chimpanzees. Brain Behav Evol. 2010;76:154–62.PubMed
29.
go back to reference Sehgal N, Smith HS, Manchikanti L. Peripherally acting opioids and clinical implications for pain control. Pain Physician. 2011;14:249–58.PubMed Sehgal N, Smith HS, Manchikanti L. Peripherally acting opioids and clinical implications for pain control. Pain Physician. 2011;14:249–58.PubMed
30.
go back to reference Zhang H, Luo X, Kranzler HR, Lappalainen J, Yang BZ, Krupitsky E, et al. Association between two mu-opioid receptor gene (OPRM1) haplotype blocks and drug or alcohol dependence. Hum Mol Genet. 2006;15:807–19.PubMedPubMedCentral Zhang H, Luo X, Kranzler HR, Lappalainen J, Yang BZ, Krupitsky E, et al. Association between two mu-opioid receptor gene (OPRM1) haplotype blocks and drug or alcohol dependence. Hum Mol Genet. 2006;15:807–19.PubMedPubMedCentral
31.
go back to reference Finco G, Pintor M, Sanna D, Orru G, Musu M, De CF, et al. Is target opioid therapy within sight? Minerva Anestesiol. 2012;78:462–72.PubMed Finco G, Pintor M, Sanna D, Orru G, Musu M, De CF, et al. Is target opioid therapy within sight? Minerva Anestesiol. 2012;78:462–72.PubMed
32.
go back to reference Kosarac B, Fox AA, Collard CD. Effect of genetic factors on opioid action. Curr Opin Anaesthesiol. 2009;22:476–82.PubMed Kosarac B, Fox AA, Collard CD. Effect of genetic factors on opioid action. Curr Opin Anaesthesiol. 2009;22:476–82.PubMed
33.
go back to reference Chavkin C, McLaughlin JP, Celver JP. Regulation of opioid receptor function by chronic agonist exposure: constitutive activity and desensitization. Mol Pharmacol. 2001;60:20–5.PubMed Chavkin C, McLaughlin JP, Celver JP. Regulation of opioid receptor function by chronic agonist exposure: constitutive activity and desensitization. Mol Pharmacol. 2001;60:20–5.PubMed
34.
go back to reference Klepstad P, Rakvag TT, Kaasa S, Holthe M, Dale O, Borchgrevink PC, et al. The 118 A>G polymorphism in the human mu-opioid receptor gene may increase morphine requirements in patients with pain caused by malignant disease. Acta Anaesthesiol Scand. 2004;48:1232–9.PubMed Klepstad P, Rakvag TT, Kaasa S, Holthe M, Dale O, Borchgrevink PC, et al. The 118 A>G polymorphism in the human mu-opioid receptor gene may increase morphine requirements in patients with pain caused by malignant disease. Acta Anaesthesiol Scand. 2004;48:1232–9.PubMed
35.
go back to reference Walter C, Lotsch J. Meta-analysis of the relevance of the OPRM1 118A>G genetic variant for pain treatment. Pain. 2009;146:270–5.PubMed Walter C, Lotsch J. Meta-analysis of the relevance of the OPRM1 118A>G genetic variant for pain treatment. Pain. 2009;146:270–5.PubMed
36.
go back to reference Ashenhurst JR, Bujarski S, Ray LA. Delta and kappa opioid receptor polymorphisms influence the effects of naltrexone on subjective responses to alcohol. Pharmacol Biochem Behav. 2012;103:253–9.PubMedPubMedCentral Ashenhurst JR, Bujarski S, Ray LA. Delta and kappa opioid receptor polymorphisms influence the effects of naltrexone on subjective responses to alcohol. Pharmacol Biochem Behav. 2012;103:253–9.PubMedPubMedCentral
37.
go back to reference Crist RC, Ambrose-Lanci LM, Vaswani M, Clarke TK, Zeng A, Yuan C, et al. Case-control association analysis of polymorphisms in the delta-opioid receptor, OPRD1, with cocaine and opioid addicted populations. Drug Alcohol Depend. 2013;127:122–8.PubMedPubMedCentral Crist RC, Ambrose-Lanci LM, Vaswani M, Clarke TK, Zeng A, Yuan C, et al. Case-control association analysis of polymorphisms in the delta-opioid receptor, OPRD1, with cocaine and opioid addicted populations. Drug Alcohol Depend. 2013;127:122–8.PubMedPubMedCentral
38.
go back to reference Arias AJ, Armeli S, Gelernter J, Covault J, Kallio A, Karhuvaara S, et al. Effects of opioid receptor gene variation on targeted nalmefene treatment in heavy drinkers. Alcohol Clin Exp Res. 2008;32:1159–66.PubMedPubMedCentral Arias AJ, Armeli S, Gelernter J, Covault J, Kallio A, Karhuvaara S, et al. Effects of opioid receptor gene variation on targeted nalmefene treatment in heavy drinkers. Alcohol Clin Exp Res. 2008;32:1159–66.PubMedPubMedCentral
39.
go back to reference Zhang H, Gelernter J, Gruen JR, Kranzler HR, Herman AI, Simen AA. Functional impact of a single-nucleotide polymorphism in the OPRD1 promoter region. J Hum Genet. 2010;55:278–84.PubMedPubMedCentral Zhang H, Gelernter J, Gruen JR, Kranzler HR, Herman AI, Simen AA. Functional impact of a single-nucleotide polymorphism in the OPRD1 promoter region. J Hum Genet. 2010;55:278–84.PubMedPubMedCentral
41.
go back to reference Edenberg HJ, Wang J, Tian H, Pochareddy S, Xuei X, Wetherill L, et al. A regulatory variation in OPRK1, the gene encoding the kappa-opioid receptor, is associated with alcohol dependence. Hum Mol Genet. 2008;17:1783–9.PubMed Edenberg HJ, Wang J, Tian H, Pochareddy S, Xuei X, Wetherill L, et al. A regulatory variation in OPRK1, the gene encoding the kappa-opioid receptor, is associated with alcohol dependence. Hum Mol Genet. 2008;17:1783–9.PubMed
42.
go back to reference Peng J, Sarkar S, Chang SL. Opioid receptor expression in human brain and peripheral tissues using absolute quantitative real-time RT-PCR. Drug Alcohol Depend. 2012;124:223–8.PubMedPubMedCentral Peng J, Sarkar S, Chang SL. Opioid receptor expression in human brain and peripheral tissues using absolute quantitative real-time RT-PCR. Drug Alcohol Depend. 2012;124:223–8.PubMedPubMedCentral
43.
go back to reference Holzer P. Opioid antagonists for prevention and treatment of opioid-induced gastrointestinal effects. Curr Opin Anaesthesiol. 2010;23:616–22.PubMed Holzer P. Opioid antagonists for prevention and treatment of opioid-induced gastrointestinal effects. Curr Opin Anaesthesiol. 2010;23:616–22.PubMed
44.
go back to reference Wood JD, Galligan JJ. Function of opioids in the enteric nervous system. Neurogastroenterol Motil. 2004;16(Suppl 2):17–28.PubMed Wood JD, Galligan JJ. Function of opioids in the enteric nervous system. Neurogastroenterol Motil. 2004;16(Suppl 2):17–28.PubMed
45.
go back to reference Sternini C, Patierno S, Selmer IS, Kirchgessner A. The opioid system in the gastrointestinal tract. Neurogastroenterol Motil. 2004;16(Suppl 2):3–16.PubMed Sternini C, Patierno S, Selmer IS, Kirchgessner A. The opioid system in the gastrointestinal tract. Neurogastroenterol Motil. 2004;16(Suppl 2):3–16.PubMed
46.
go back to reference DeHaven-Hudkins DL, DeHaven RN, Little PJ, Techner LM. The involvement of the mu-opioid receptor in gastrointestinal pathophysiology: therapeutic opportunities for antagonism at this receptor. Pharmacol Ther. 2008;117:162–87. DeHaven-Hudkins DL, DeHaven RN, Little PJ, Techner LM. The involvement of the mu-opioid receptor in gastrointestinal pathophysiology: therapeutic opportunities for antagonism at this receptor. Pharmacol Ther. 2008;117:162–87.
47.
go back to reference Poole DP, Pelayo JC, Scherrer G, Evans CJ, Kieffer BL, Bunnett NW. Localization and regulation of fluorescently labeled delta opioid receptor, expressed in enteric neurons of mice. Gastroenterology. 2011;141:982–91.PubMed Poole DP, Pelayo JC, Scherrer G, Evans CJ, Kieffer BL, Bunnett NW. Localization and regulation of fluorescently labeled delta opioid receptor, expressed in enteric neurons of mice. Gastroenterology. 2011;141:982–91.PubMed
48.
go back to reference Phan NQ, Lotts T, Antal A, Bernhard JD, Stander S. Systemic kappa opioid receptor agonists in the treatment of chronic pruritus: a literature review. Acta Derm Venereol. 2012;92:555–60.PubMed Phan NQ, Lotts T, Antal A, Bernhard JD, Stander S. Systemic kappa opioid receptor agonists in the treatment of chronic pruritus: a literature review. Acta Derm Venereol. 2012;92:555–60.PubMed
49.
go back to reference Bagnol D, Mansour A, Akil H, Watson SJ. Cellular localization and distribution of the cloned mu and kappa opioid receptors in rat gastrointestinal tract. Neuroscience. 1997;81:579–91.PubMed Bagnol D, Mansour A, Akil H, Watson SJ. Cellular localization and distribution of the cloned mu and kappa opioid receptors in rat gastrointestinal tract. Neuroscience. 1997;81:579–91.PubMed
50.
go back to reference Kurz A, Sessler DI. Opioid-induced bowel dysfunction: pathophysiology and potential new therapies. Drugs. 2003;63:649–71.PubMed Kurz A, Sessler DI. Opioid-induced bowel dysfunction: pathophysiology and potential new therapies. Drugs. 2003;63:649–71.PubMed
51.
go back to reference Holzer P. Opioids and opioid receptors in the enteric nervous system: from a problem in opioid analgesia to a possible new prokinetic therapy in humans. Neurosci Lett. 2004;361:192–5.PubMed Holzer P. Opioids and opioid receptors in the enteric nervous system: from a problem in opioid analgesia to a possible new prokinetic therapy in humans. Neurosci Lett. 2004;361:192–5.PubMed
52.
go back to reference Donnerer J, Liebmann I. Stimulus-evoked opioid inhibition in guinea-pig longitudinal muscle-myenteric plexus strip is modulated by NMDA receptors. Neurosci Lett. 2007;419:74–7.PubMed Donnerer J, Liebmann I. Stimulus-evoked opioid inhibition in guinea-pig longitudinal muscle-myenteric plexus strip is modulated by NMDA receptors. Neurosci Lett. 2007;419:74–7.PubMed
53.
go back to reference Patierno S, Zellalem W, Ho A, Parsons CG, Lloyd KC, Tonini M, et al. N-methyl-D-aspartate receptors mediate endogenous opioid release in enteric neurons after abdominal surgery. Gastroenterology. 2005;128:2009–19.PubMed Patierno S, Zellalem W, Ho A, Parsons CG, Lloyd KC, Tonini M, et al. N-methyl-D-aspartate receptors mediate endogenous opioid release in enteric neurons after abdominal surgery. Gastroenterology. 2005;128:2009–19.PubMed
54.
go back to reference Donnerer J, Liebmann I. Evidence for opioid-induced release of glutamate in guinea pig longitudinal muscle-myenteric plexus strip. Neurosci Lett. 2009;462:118–20.PubMed Donnerer J, Liebmann I. Evidence for opioid-induced release of glutamate in guinea pig longitudinal muscle-myenteric plexus strip. Neurosci Lett. 2009;462:118–20.PubMed
55.
go back to reference Iwata H, Tsuchiya S, Nakamura T, Yano S. Morphine leads to contraction of the ileal circular muscle via inhibition of the nitrergic pathway in mice. Eur J Pharmacol. 2007;574:66–70.PubMed Iwata H, Tsuchiya S, Nakamura T, Yano S. Morphine leads to contraction of the ileal circular muscle via inhibition of the nitrergic pathway in mice. Eur J Pharmacol. 2007;574:66–70.PubMed
56.
go back to reference De LA, Coupar IM. Insights into opioid action in the intestinal tract. Pharmacol Ther. 1996;69(2):103–15. De LA, Coupar IM. Insights into opioid action in the intestinal tract. Pharmacol Ther. 1996;69(2):103–15.
57.
go back to reference Brock C, Olesen SS, Olesen AE, Frokjaer JB, Andresen T, Drewes AM. Opioid-induced bowel dysfunction: pathophysiology and management. Drugs. 2012;72:1847–65.PubMed Brock C, Olesen SS, Olesen AE, Frokjaer JB, Andresen T, Drewes AM. Opioid-induced bowel dysfunction: pathophysiology and management. Drugs. 2012;72:1847–65.PubMed
58.
go back to reference Barrett KE. New insights into the pathogenesis of intestinal dysfunction: secretory diarrhea and cystic fibrosis. World J Gastroenterol. 2000;6:470–4.PubMed Barrett KE. New insights into the pathogenesis of intestinal dysfunction: secretory diarrhea and cystic fibrosis. World J Gastroenterol. 2000;6:470–4.PubMed
59.
go back to reference Barrett KE, Keely SJ. Chloride secretion by the intestinal epithelium: molecular basis and regulatory aspects. Annu Rev Physiol. 2000;62:535–72.PubMed Barrett KE, Keely SJ. Chloride secretion by the intestinal epithelium: molecular basis and regulatory aspects. Annu Rev Physiol. 2000;62:535–72.PubMed
60.
go back to reference Herman MA, Gillis RA, Vicini S, Dretchen KL, Sahibzada N. Tonic GABAA receptor conductance in medial subnucleus of the tractus solitarius neurons is inhibited by activation of mu-opioid receptors. J Neurophysiol. 2012;107:1022–31.PubMed Herman MA, Gillis RA, Vicini S, Dretchen KL, Sahibzada N. Tonic GABAA receptor conductance in medial subnucleus of the tractus solitarius neurons is inhibited by activation of mu-opioid receptors. J Neurophysiol. 2012;107:1022–31.PubMed
61.
go back to reference Herman MA, Alayan A, Sahibzada N, Bayer B, Verbalis J, Dretchen KL, et al. micro-Opioid receptor stimulation in the medial subnucleus of the tractus solitarius inhibits gastric tone and motility by reducing local GABA activity. Am J Physiol Gastrointest Liver Physiol. 2010;299:G494–506.PubMed Herman MA, Alayan A, Sahibzada N, Bayer B, Verbalis J, Dretchen KL, et al. micro-Opioid receptor stimulation in the medial subnucleus of the tractus solitarius inhibits gastric tone and motility by reducing local GABA activity. Am J Physiol Gastrointest Liver Physiol. 2010;299:G494–506.PubMed
62.
go back to reference Vilardaga JP, Bunemann M, Feinstein TN, Lambert N, Nikolaev VO, Engelhardt S, et al. GPCR and G proteins: drug efficacy and activation in live cells. Mol Endocrinol. 2009;23:590–9.PubMed Vilardaga JP, Bunemann M, Feinstein TN, Lambert N, Nikolaev VO, Engelhardt S, et al. GPCR and G proteins: drug efficacy and activation in live cells. Mol Endocrinol. 2009;23:590–9.PubMed
63.
go back to reference Gomez MP, Nasi E. Light transduction in invertebrate hyperpolarizing photoreceptors: possible involvement of a Go-regulated guanylate cyclase. J Neurosci. 2000;20:5254–63.PubMed Gomez MP, Nasi E. Light transduction in invertebrate hyperpolarizing photoreceptors: possible involvement of a Go-regulated guanylate cyclase. J Neurosci. 2000;20:5254–63.PubMed
64.
go back to reference Law PY, Erickson-Herbrandson LJ, Zha QQ, Solberg J, Chu J, Sarre A, et al. Heterodimerization of mu- and delta-opioid receptors occurs at the cell surface only and requires receptor-G protein interactions. J Biol Chem. 2000;280:11152–64. Law PY, Erickson-Herbrandson LJ, Zha QQ, Solberg J, Chu J, Sarre A, et al. Heterodimerization of mu- and delta-opioid receptors occurs at the cell surface only and requires receptor-G protein interactions. J Biol Chem. 2000;280:11152–64.
65.
go back to reference Berger AC, Whistler JL. Morphine-induced mu opioid receptor trafficking enhances reward yet prevents compulsive drug use. EMBO Mol Med. 2011;3:385–97.PubMedPubMedCentral Berger AC, Whistler JL. Morphine-induced mu opioid receptor trafficking enhances reward yet prevents compulsive drug use. EMBO Mol Med. 2011;3:385–97.PubMedPubMedCentral
66.
go back to reference Milligan G. G protein-coupled receptor dimerisation: molecular basis and relevance to function. Biochim Biophys Acta. 2007;1768:825–35.PubMed Milligan G. G protein-coupled receptor dimerisation: molecular basis and relevance to function. Biochim Biophys Acta. 2007;1768:825–35.PubMed
67.
go back to reference Milligan G, Parenty G, Stoddart LA, Lane JR. Novel pharmacological applications of G-protein-coupled receptor-G protein fusions. Curr Opin Pharmacol. 2007;7:521–6.PubMed Milligan G, Parenty G, Stoddart LA, Lane JR. Novel pharmacological applications of G-protein-coupled receptor-G protein fusions. Curr Opin Pharmacol. 2007;7:521–6.PubMed
68.
go back to reference Al-Hasani R, Bruchas MR. Molecular mechanisms of opioid receptor-dependent signaling and behavior. Anesthesiology. 2011;115:1363–81.PubMedPubMedCentral Al-Hasani R, Bruchas MR. Molecular mechanisms of opioid receptor-dependent signaling and behavior. Anesthesiology. 2011;115:1363–81.PubMedPubMedCentral
69.
go back to reference Rios C, Gomes I, Devi LA. mu opioid and CB1 cannabinoid receptor interactions: reciprocal inhibition of receptor signaling and neuritogenesis. Br J Pharmacol. 2006;148:387–95.PubMed Rios C, Gomes I, Devi LA. mu opioid and CB1 cannabinoid receptor interactions: reciprocal inhibition of receptor signaling and neuritogenesis. Br J Pharmacol. 2006;148:387–95.PubMed
71.
go back to reference Bunchorntavakul C, Reddy KR. Pruritus in chronic cholestatic liver disease. Clin Liver Dis. 2012;16:331–46.PubMed Bunchorntavakul C, Reddy KR. Pruritus in chronic cholestatic liver disease. Clin Liver Dis. 2012;16:331–46.PubMed
72.
go back to reference Headrick JP, Pepe S, Peart JN. Non-analgesic effects of opioids: cardiovascular effects of opioids and their receptor systems. Curr Pharm Des. 2012;18:6090–100.PubMed Headrick JP, Pepe S, Peart JN. Non-analgesic effects of opioids: cardiovascular effects of opioids and their receptor systems. Curr Pharm Des. 2012;18:6090–100.PubMed
73.
go back to reference Husain S, Abdul Y, Potter DE. Non-Analgesic Effects of Opioids: neuroprotection in the Retina. Curr Pharm Des. 2012;18:6101–8.PubMed Husain S, Abdul Y, Potter DE. Non-Analgesic Effects of Opioids: neuroprotection in the Retina. Curr Pharm Des. 2012;18:6101–8.PubMed
74.
go back to reference Kromer W. Endogenous and exogenous opioids in the control of gastrointestinal motility and secretion. Pharmacol Rev. 1988;40:121–62.PubMed Kromer W. Endogenous and exogenous opioids in the control of gastrointestinal motility and secretion. Pharmacol Rev. 1988;40:121–62.PubMed
75.
go back to reference Panchal SJ, Muller-Schwefe P, Wurzelmann JI. Opioid-induced bowel dysfunction: prevalence, pathophysiology and burden. Int J Clin Pract. 2007;61:1181–7.PubMedPubMedCentral Panchal SJ, Muller-Schwefe P, Wurzelmann JI. Opioid-induced bowel dysfunction: prevalence, pathophysiology and burden. Int J Clin Pract. 2007;61:1181–7.PubMedPubMedCentral
76.
go back to reference Collins S, Verma-Gandhu M. The putative role of endogenous and exogenous opiates in inflammatory bowel disease. Gut. 2006;55:756–7.PubMed Collins S, Verma-Gandhu M. The putative role of endogenous and exogenous opiates in inflammatory bowel disease. Gut. 2006;55:756–7.PubMed
77.
go back to reference Owczarek D, Cibor D, Mach T, Ciesla A, Pierzchala-Koziec K, Salapa K, et al. Met-enkephalins in patients with inflammatory bowel diseases. Adv Med Sci. 2011;56:158–64.PubMed Owczarek D, Cibor D, Mach T, Ciesla A, Pierzchala-Koziec K, Salapa K, et al. Met-enkephalins in patients with inflammatory bowel diseases. Adv Med Sci. 2011;56:158–64.PubMed
78.
go back to reference Al-Khrasani M, Lacko E, Riba P, Kiraly K, Sobor M, Timar J, et al. The central versus peripheral antinociceptive effects of mu-opioid receptor agonists in the new model of rat visceral pain. Brain Res Bull. 2012;87:238–43.PubMed Al-Khrasani M, Lacko E, Riba P, Kiraly K, Sobor M, Timar J, et al. The central versus peripheral antinociceptive effects of mu-opioid receptor agonists in the new model of rat visceral pain. Brain Res Bull. 2012;87:238–43.PubMed
79.
go back to reference Xu GY, Winston JH, Chen JD. Electroacupuncture attenuates visceral hyperalgesia and inhibits the enhanced excitability of colon specific sensory neurons in a rat model of irritable bowel syndrome. Neurogastroenterol Motil. 2009;21:1302–e125. Xu GY, Winston JH, Chen JD. Electroacupuncture attenuates visceral hyperalgesia and inhibits the enhanced excitability of colon specific sensory neurons in a rat model of irritable bowel syndrome. Neurogastroenterol Motil. 2009;21:1302–e125.
80.
go back to reference Zadina JE, Hackler L, Ge LJ, Kastin AJ. A potent and selective endogenous agonist for the mu-opiate receptor. Nature. 1997;386:499–502.PubMed Zadina JE, Hackler L, Ge LJ, Kastin AJ. A potent and selective endogenous agonist for the mu-opiate receptor. Nature. 1997;386:499–502.PubMed
81.
go back to reference Storr M, Geisler F, Neuhuber WL, Schusdziarra V, Allescher HD. Endomorphin-1 and -2, endogenous ligands for the mu-opioid receptor, inhibit striated and smooth muscle contraction in the rat oesophagus. Neurogastroenterol Motil. 2000;12:441–8.PubMed Storr M, Geisler F, Neuhuber WL, Schusdziarra V, Allescher HD. Endomorphin-1 and -2, endogenous ligands for the mu-opioid receptor, inhibit striated and smooth muscle contraction in the rat oesophagus. Neurogastroenterol Motil. 2000;12:441–8.PubMed
82.
go back to reference Nishiwaki H, Saitoh N, Nishio H, Takeuchi T, Hata F. Relationship between muscarinic autoinhibition and the inhibitory effect of morphine on acetylcholine release from myenteric plexus of guinea pig ileum. Jpn J Pharmacol. 1998;77:271–8.PubMed Nishiwaki H, Saitoh N, Nishio H, Takeuchi T, Hata F. Relationship between muscarinic autoinhibition and the inhibitory effect of morphine on acetylcholine release from myenteric plexus of guinea pig ileum. Jpn J Pharmacol. 1998;77:271–8.PubMed
83.
go back to reference Nishiwaki H, Saitoh N, Nishio H, Takeuchi T, Hata F. Inhibitory effect of endomorphin-1 and -2 on acetylcholine release from myenteric plexus of guinea pig ileum. Jpn J Pharmacol. 1998;78:83–6.PubMed Nishiwaki H, Saitoh N, Nishio H, Takeuchi T, Hata F. Inhibitory effect of endomorphin-1 and -2 on acetylcholine release from myenteric plexus of guinea pig ileum. Jpn J Pharmacol. 1998;78:83–6.PubMed
84.
go back to reference Yu Y, Cui Y, Wang X, Lai LH, Wang CL, Fan YZ, et al. In vitro characterization of the effects of endomorphin 1 and 2, endogenous ligands for mu-opioid receptors, on mouse colonic motility. Biochem Pharmacol. 2007;73:1384–93.PubMed Yu Y, Cui Y, Wang X, Lai LH, Wang CL, Fan YZ, et al. In vitro characterization of the effects of endomorphin 1 and 2, endogenous ligands for mu-opioid receptors, on mouse colonic motility. Biochem Pharmacol. 2007;73:1384–93.PubMed
85.
go back to reference Azuma Y, Ohura K. Endomorphin-2 modulates productions of TNF-alpha, IL-1beta, IL-10, and IL-12, and alters functions related to innate immune of macrophages. Inflammation. 2002;26:223–32.PubMed Azuma Y, Ohura K. Endomorphin-2 modulates productions of TNF-alpha, IL-1beta, IL-10, and IL-12, and alters functions related to innate immune of macrophages. Inflammation. 2002;26:223–32.PubMed
86.
go back to reference Janecka A, Perlikowska R, Gach K, Wyrebska A, Fichna J. Development of opioid peptide analogs for pain relief. Curr Pharm Des. 2010;16:1126–35.PubMed Janecka A, Perlikowska R, Gach K, Wyrebska A, Fichna J. Development of opioid peptide analogs for pain relief. Curr Pharm Des. 2010;16:1126–35.PubMed
87.
go back to reference Varamini P, Mansfeld FM, Blanchfield JT, Wyse BD, Smith MT, Toth I. Synthesis and biological evaluation of an orally active glycosylated endomorphin-1. J Med Chem. 2012;55:5859–67.PubMed Varamini P, Mansfeld FM, Blanchfield JT, Wyse BD, Smith MT, Toth I. Synthesis and biological evaluation of an orally active glycosylated endomorphin-1. J Med Chem. 2012;55:5859–67.PubMed
88.
go back to reference Kaneko K, Iwasaki M, Yoshikawa M, Ohinata K. Orally administered soymorphins, soy-derived opioid peptides, suppress feeding and intestinal transit via gut mu(1)-receptor coupled to 5-HT(1A), D(2), and GABA(B) systems. Am J Physiol Gastrointest Liver Physiol. 2010;299:G799–805.PubMed Kaneko K, Iwasaki M, Yoshikawa M, Ohinata K. Orally administered soymorphins, soy-derived opioid peptides, suppress feeding and intestinal transit via gut mu(1)-receptor coupled to 5-HT(1A), D(2), and GABA(B) systems. Am J Physiol Gastrointest Liver Physiol. 2010;299:G799–805.PubMed
89.
go back to reference Chiba H, Tani F, Yoshikawa M. Opioid antagonist peptides derived from kappa-casein. J Dairy Res. 1989;56:363–6.PubMed Chiba H, Tani F, Yoshikawa M. Opioid antagonist peptides derived from kappa-casein. J Dairy Res. 1989;56:363–6.PubMed
90.
go back to reference Patten GS, Head RJ, Abeywardena MY. Effects of casoxin 4 on morphine inhibition of small animal intestinal contractility and gut transit in the mouse. Clin Exp Gastroenterol. 2011;4:23–31.PubMedPubMedCentral Patten GS, Head RJ, Abeywardena MY. Effects of casoxin 4 on morphine inhibition of small animal intestinal contractility and gut transit in the mouse. Clin Exp Gastroenterol. 2011;4:23–31.PubMedPubMedCentral
91.
go back to reference Stefano GB, Ptacek R, Kuzelova H, Kream RM. Endogenous morphine: up-to-date review 2011. Folia Biol. 2012;58:49–56. Stefano GB, Ptacek R, Kuzelova H, Kream RM. Endogenous morphine: up-to-date review 2011. Folia Biol. 2012;58:49–56.
92.
go back to reference Netzer P, Sendensky A, Wissmeyer MP, Baumeler S, Batista C, Scheurer U, et al. The effect of naloxone-3-glucuronide on colonic transit time in healthy men after acute morphine administration: a placebo-controlled double-blinded crossover preclinical volunteer study. Aliment Pharmacol Ther. 2008;28:1334–41.PubMed Netzer P, Sendensky A, Wissmeyer MP, Baumeler S, Batista C, Scheurer U, et al. The effect of naloxone-3-glucuronide on colonic transit time in healthy men after acute morphine administration: a placebo-controlled double-blinded crossover preclinical volunteer study. Aliment Pharmacol Ther. 2008;28:1334–41.PubMed
93.
go back to reference Reber P, Brenneisen R, Flogerzi B, Batista C, Netzer P, Scheurer U. Effect of naloxone-3-glucuronide and N-methylnaloxone on the motility of the isolated rat colon after morphine. Dig Dis Sci. 2007;52:502–7.PubMed Reber P, Brenneisen R, Flogerzi B, Batista C, Netzer P, Scheurer U. Effect of naloxone-3-glucuronide and N-methylnaloxone on the motility of the isolated rat colon after morphine. Dig Dis Sci. 2007;52:502–7.PubMed
94.
go back to reference Ross GR, Gabra BH, Dewey WL, Akbarali HI. Morphine tolerance in the mouse ileum and colon. J Pharmacol Exp Ther. 2008;327:561–72.PubMedPubMedCentral Ross GR, Gabra BH, Dewey WL, Akbarali HI. Morphine tolerance in the mouse ileum and colon. J Pharmacol Exp Ther. 2008;327:561–72.PubMedPubMedCentral
95.
go back to reference Aquilante CL, Letrent SP, Pollack GM, Brouwer KL. Increased brain P-glycoprotein in morphine tolerant rats. Life Sci. 2000;66:L47–51. Aquilante CL, Letrent SP, Pollack GM, Brouwer KL. Increased brain P-glycoprotein in morphine tolerant rats. Life Sci. 2000;66:L47–51.
96.
go back to reference King M, Su W, Chang A, Zuckerman A, Pasternak GW. Transport of opioids from the brain to the periphery by P-glycoprotein: peripheral actions of central drugs. Nat Neurosci. 2001;4:268–74.PubMed King M, Su W, Chang A, Zuckerman A, Pasternak GW. Transport of opioids from the brain to the periphery by P-glycoprotein: peripheral actions of central drugs. Nat Neurosci. 2001;4:268–74.PubMed
97.
go back to reference Yousif S, Saubamea B, Cisternino S, Marie-Claire C, Dauchy S, Scherrmann JM, et al. Effect of chronic exposure to morphine on the rat blood-brain barrier: focus on the P-glycoprotein. J Neurochem. 2008;107:647–57.PubMed Yousif S, Saubamea B, Cisternino S, Marie-Claire C, Dauchy S, Scherrmann JM, et al. Effect of chronic exposure to morphine on the rat blood-brain barrier: focus on the P-glycoprotein. J Neurochem. 2008;107:647–57.PubMed
98.
go back to reference Anastassopoulos KP, Chow W, Tapia CI, Baik R, Ackerman SJ, Biondi D, et al. Economic study on the impact of side effects in patients taking oxycodone controlled-release for noncancer pain. J Manag Care Pharm. 2012;18:615–26.PubMed Anastassopoulos KP, Chow W, Tapia CI, Baik R, Ackerman SJ, Biondi D, et al. Economic study on the impact of side effects in patients taking oxycodone controlled-release for noncancer pain. J Manag Care Pharm. 2012;18:615–26.PubMed
99.
go back to reference Peng X, Cebra JJ, Adler MW, Meissler JJ Jr, Cowan A, Feng P, et al. Morphine inhibits mucosal antibody responses and TGF-beta mRNA in gut-associated lymphoid tissue following oral cholera toxin in mice. J Immunol. 2001;167:3677–81.PubMed Peng X, Cebra JJ, Adler MW, Meissler JJ Jr, Cowan A, Feng P, et al. Morphine inhibits mucosal antibody responses and TGF-beta mRNA in gut-associated lymphoid tissue following oral cholera toxin in mice. J Immunol. 2001;167:3677–81.PubMed
100.
go back to reference Breslow JM, Feng P, Meissler JJ, Pintar JE, Gaughan J, Adler MW, et al. Potentiating effect of morphine on oral Salmonella enterica serovar Typhimurium infection is mu-opioid receptor-dependent. Microb Pathog. 2010;49:330–5.PubMedPubMedCentral Breslow JM, Feng P, Meissler JJ, Pintar JE, Gaughan J, Adler MW, et al. Potentiating effect of morphine on oral Salmonella enterica serovar Typhimurium infection is mu-opioid receptor-dependent. Microb Pathog. 2010;49:330–5.PubMedPubMedCentral
101.
go back to reference Feng P, Rahim RT, Cowan A, Liu-Chen LY, Peng X, Gaughan J, et al. Effects of mu, kappa or delta opioids administered by pellet or pump on oral Salmonella infection and gastrointestinal transit. Eur J Pharmacol. 2006;534:250–7.PubMed Feng P, Rahim RT, Cowan A, Liu-Chen LY, Peng X, Gaughan J, et al. Effects of mu, kappa or delta opioids administered by pellet or pump on oral Salmonella infection and gastrointestinal transit. Eur J Pharmacol. 2006;534:250–7.PubMed
102.
go back to reference Babrowski T, Holbrook C, Moss J, Gottlieb L, Valuckaite V, Zaborin A, et al. Pseudomonas aeruginosa virulence expression is directly activated by morphine and is capable of causing lethal gut-derived sepsis in mice during chronic morphine administration. Ann Surg. 2012;255:386–93.PubMedPubMedCentral Babrowski T, Holbrook C, Moss J, Gottlieb L, Valuckaite V, Zaborin A, et al. Pseudomonas aeruginosa virulence expression is directly activated by morphine and is capable of causing lethal gut-derived sepsis in mice during chronic morphine administration. Ann Surg. 2012;255:386–93.PubMedPubMedCentral
103.
go back to reference Glattard E, Welters ID, Lavaux T, Muller AH, Laux A, Zhang D, et al. Endogenous morphine levels are increased in sepsis: a partial implication of neutrophils. PLoS ONE. 2010;5:e8791.PubMedPubMedCentral Glattard E, Welters ID, Lavaux T, Muller AH, Laux A, Zhang D, et al. Endogenous morphine levels are increased in sepsis: a partial implication of neutrophils. PLoS ONE. 2010;5:e8791.PubMedPubMedCentral
104.
go back to reference Capasso R, Borrelli F, Capasso F, Siebert DJ, Stewart DJ, Zjawiony JK, et al. The hallucinogenic herb Salvia divinorum and its active ingredient salvinorin A inhibit enteric cholinergic transmission in the guinea-pig ileum. Neurogastroenterol Motil. 2006;18:69–75.PubMed Capasso R, Borrelli F, Capasso F, Siebert DJ, Stewart DJ, Zjawiony JK, et al. The hallucinogenic herb Salvia divinorum and its active ingredient salvinorin A inhibit enteric cholinergic transmission in the guinea-pig ileum. Neurogastroenterol Motil. 2006;18:69–75.PubMed
105.
go back to reference Fichna J, Schicho R, Andrews CN, Bashashati M, Klompus M, McKay DM, et al. Salvinorin A inhibits colonic transit and neurogenic ion transport in mice by activating kappa-opioid and cannabinoid receptors. Neurogastroenterol Motil. 2009;21:1326-e128. Fichna J, Schicho R, Andrews CN, Bashashati M, Klompus M, McKay DM, et al. Salvinorin A inhibits colonic transit and neurogenic ion transport in mice by activating kappa-opioid and cannabinoid receptors. Neurogastroenterol Motil. 2009;21:1326-e128.
106.
go back to reference Capasso R, Borrelli F, Cascio MG, Aviello G, Huben K, Zjawiony JK, et al. Inhibitory effect of salvinorin A, from Salvia divinorum, on ileitis-induced hypermotility: cross-talk between kappa-opioid and cannabinoid CB(1) receptors. Br J Pharmacol. 2008;155:681–9.PubMed Capasso R, Borrelli F, Cascio MG, Aviello G, Huben K, Zjawiony JK, et al. Inhibitory effect of salvinorin A, from Salvia divinorum, on ileitis-induced hypermotility: cross-talk between kappa-opioid and cannabinoid CB(1) receptors. Br J Pharmacol. 2008;155:681–9.PubMed
107.
go back to reference Fichna J, Dicay M, Hirota SA, Traboulsi D, Macdonald JA, Janecka A, et al. Differential effects of salvinorin A on endotoxin-induced hypermotility and neurogenic ion transport in mouse ileum. Neurogastroenterol Motil. 2011;23:583-e212. Fichna J, Dicay M, Hirota SA, Traboulsi D, Macdonald JA, Janecka A, et al. Differential effects of salvinorin A on endotoxin-induced hypermotility and neurogenic ion transport in mouse ileum. Neurogastroenterol Motil. 2011;23:583-e212.
108.
go back to reference Fichna J, Dicay M, Lewellyn K, Janecka A, Zjawiony JK, MacNaughton WK, et al. Salvinorin A has antiinflammatory and antinociceptive effects in experimental models of colitis in mice mediated by KOR and CB1 receptors. Inflamm Bowel Dis. 2012;18:1137–45.PubMed Fichna J, Dicay M, Lewellyn K, Janecka A, Zjawiony JK, MacNaughton WK, et al. Salvinorin A has antiinflammatory and antinociceptive effects in experimental models of colitis in mice mediated by KOR and CB1 receptors. Inflamm Bowel Dis. 2012;18:1137–45.PubMed
109.
go back to reference Fichna J, Schicho R, Janecka A, Zjawiony JK, Storr M. Selective natural kappa opioid and cannabinoid receptor agonists with a potential role in the treatment of gastrointestinal dysfunction. Drug News Perspect. 2009;22:383–92.PubMed Fichna J, Schicho R, Janecka A, Zjawiony JK, Storr M. Selective natural kappa opioid and cannabinoid receptor agonists with a potential role in the treatment of gastrointestinal dysfunction. Drug News Perspect. 2009;22:383–92.PubMed
110.
go back to reference Laugsand EA, Kaasa S, Klepstad P. Management of opioid-induced nausea and vomiting in cancer patients: systematic review and evidence-based recommendations. Palliat Med. 2011;25:442–53.PubMed Laugsand EA, Kaasa S, Klepstad P. Management of opioid-induced nausea and vomiting in cancer patients: systematic review and evidence-based recommendations. Palliat Med. 2011;25:442–53.PubMed
111.
go back to reference Chen W, Chung HH, Cheng JT. Opiate-induced constipation related to activation of small intestine opioid mu2-receptors. World J Gastroenterol. 2012;18:1391–6.PubMed Chen W, Chung HH, Cheng JT. Opiate-induced constipation related to activation of small intestine opioid mu2-receptors. World J Gastroenterol. 2012;18:1391–6.PubMed
112.
go back to reference Kumar R, Reeta KH, Ray SB. Antinociceptive effect of intrathecal loperamide: role of mu-opioid receptor and calcium channels. Eur J Pharmacol. 2012;696:77–82.PubMed Kumar R, Reeta KH, Ray SB. Antinociceptive effect of intrathecal loperamide: role of mu-opioid receptor and calcium channels. Eur J Pharmacol. 2012;696:77–82.PubMed
113.
go back to reference Primi MP, Bueno L, Baumer P, Berard H, Lecomte JM. Racecadotril demonstrates intestinal antisecretory activity in vivo. Aliment Pharmacol Ther. 1999;13:3–7.PubMed Primi MP, Bueno L, Baumer P, Berard H, Lecomte JM. Racecadotril demonstrates intestinal antisecretory activity in vivo. Aliment Pharmacol Ther. 1999;13:3–7.PubMed
114.
go back to reference Duncker SC, Philippe D, Martin-Paschoud C, Moser M, Mercenier A, Nutten S. Nigella sativa (Black Cumin) seed extract alleviates symptoms of allergic diarrhea in mice, involving opioid receptors. PLoS One. 2012;7:e39841.PubMedPubMedCentral Duncker SC, Philippe D, Martin-Paschoud C, Moser M, Mercenier A, Nutten S. Nigella sativa (Black Cumin) seed extract alleviates symptoms of allergic diarrhea in mice, involving opioid receptors. PLoS One. 2012;7:e39841.PubMedPubMedCentral
115.
go back to reference Isik F, Tunali AT, Yarat A, Genc Z, Pisiriciler R, Caliskan-Ak E, et al. Protective effects of black cumin (Nigella sativa) oil on TNBS-induced experimental colitis in rats. Dig Dis Sci. 2011;56:721–30.PubMed Isik F, Tunali AT, Yarat A, Genc Z, Pisiriciler R, Caliskan-Ak E, et al. Protective effects of black cumin (Nigella sativa) oil on TNBS-induced experimental colitis in rats. Dig Dis Sci. 2011;56:721–30.PubMed
116.
go back to reference Boskabady MH, Mohsenpoor N, Takaloo L. Antiasthmatic effect of Nigella sativa in airways of asthmatic patients. Phytomedicine. 2010;17:707–13.PubMed Boskabady MH, Mohsenpoor N, Takaloo L. Antiasthmatic effect of Nigella sativa in airways of asthmatic patients. Phytomedicine. 2010;17:707–13.PubMed
117.
go back to reference Banerjee S, Padhye S, Azmi A, Wang Z, Philip PA, Kucuk O, et al. Review on molecular and therapeutic potential of thymoquinone in cancer. Nutr Cancer. 2010;62:938–46.PubMed Banerjee S, Padhye S, Azmi A, Wang Z, Philip PA, Kucuk O, et al. Review on molecular and therapeutic potential of thymoquinone in cancer. Nutr Cancer. 2010;62:938–46.PubMed
118.
go back to reference Hirata T, Keto Y, Nakata M, Takeuchi A, Funatsu T, Akuzawa S, et al. Effects of serotonin 5-HT3 receptor antagonists on stress-induced colonic hyperalgesia and diarrhoea in rats: a comparative study with opioid receptor agonists, a muscarinic receptor antagonist and a synthetic polymer. Neurogastroenterol Motil. 2008;20:557–65.PubMed Hirata T, Keto Y, Nakata M, Takeuchi A, Funatsu T, Akuzawa S, et al. Effects of serotonin 5-HT3 receptor antagonists on stress-induced colonic hyperalgesia and diarrhoea in rats: a comparative study with opioid receptor agonists, a muscarinic receptor antagonist and a synthetic polymer. Neurogastroenterol Motil. 2008;20:557–65.PubMed
119.
go back to reference McNicol E, Horowicz-Mehler N, Fisk RA, Bennett K, Gialeli-Goudas M, Chew PW, et al. Management of opioid side effects in cancer-related and chronic noncancer pain: a systematic review. J Pain. 2003;4:231–56.PubMed McNicol E, Horowicz-Mehler N, Fisk RA, Bennett K, Gialeli-Goudas M, Chew PW, et al. Management of opioid side effects in cancer-related and chronic noncancer pain: a systematic review. J Pain. 2003;4:231–56.PubMed
120.
go back to reference Gwee KA. Irritable bowel syndrome in developing countries–a disorder of civilization or colonization? Neurogastroenterol Motil. 2005;17:317–24.PubMed Gwee KA. Irritable bowel syndrome in developing countries–a disorder of civilization or colonization? Neurogastroenterol Motil. 2005;17:317–24.PubMed
121.
go back to reference Pappagallo M. Incidence, prevalence, and management of opioid bowel dysfunction. Am J Surg. 2001;182:11S–8S.PubMed Pappagallo M. Incidence, prevalence, and management of opioid bowel dysfunction. Am J Surg. 2001;182:11S–8S.PubMed
122.
go back to reference Deibert P, Xander C, Blum HE, Becker G. Methylnaltrexone: the evidence for its use in the management of opioid-induced constipation. Core Evid. 2010;4:247–58.PubMedPubMedCentral Deibert P, Xander C, Blum HE, Becker G. Methylnaltrexone: the evidence for its use in the management of opioid-induced constipation. Core Evid. 2010;4:247–58.PubMedPubMedCentral
123.
go back to reference Leppert W. The role of opioid receptor antagonists in the treatment of opioid-induced constipation: a review. Adv Ther. 2010;27:714–30.PubMed Leppert W. The role of opioid receptor antagonists in the treatment of opioid-induced constipation: a review. Adv Ther. 2010;27:714–30.PubMed
124.
go back to reference Thomas J, Karver S, Cooney GA, Chamberlain BH, Watt CK, Slatkin NE, et al. Methylnaltrexone for opioid-induced constipation in advanced illness. N Engl J Med. 2008;358:2332–43.PubMed Thomas J, Karver S, Cooney GA, Chamberlain BH, Watt CK, Slatkin NE, et al. Methylnaltrexone for opioid-induced constipation in advanced illness. N Engl J Med. 2008;358:2332–43.PubMed
125.
go back to reference Jakab RL, Collaco AM, Ameen NA. Lubiprostone targets prostanoid signaling and promotes ion transporter trafficking, mucus exocytosis, and contractility. Dig Dis Sci. 2012;57:2826–45.PubMedPubMedCentral Jakab RL, Collaco AM, Ameen NA. Lubiprostone targets prostanoid signaling and promotes ion transporter trafficking, mucus exocytosis, and contractility. Dig Dis Sci. 2012;57:2826–45.PubMedPubMedCentral
126.
go back to reference Norimatsu Y, Moran AR, Macdonald KD. Lubiprostone activates CFTR, but not ClC-2, via the prostaglandin receptor (EP(4)). Biochem Biophys Res Commun. 2012;426:374–9.PubMedPubMedCentral Norimatsu Y, Moran AR, Macdonald KD. Lubiprostone activates CFTR, but not ClC-2, via the prostaglandin receptor (EP(4)). Biochem Biophys Res Commun. 2012;426:374–9.PubMedPubMedCentral
127.
go back to reference Ao M, Venkatasubramanian J, Boonkaewwan C, Ganesan N, Syed A, Benya RV, et al. Lubiprostone activates Cl- secretion via cAMP signaling and increases membrane CFTR in the human colon carcinoma cell line, T84. Dig Dis Sci. 2011;56:339–51.PubMed Ao M, Venkatasubramanian J, Boonkaewwan C, Ganesan N, Syed A, Benya RV, et al. Lubiprostone activates Cl- secretion via cAMP signaling and increases membrane CFTR in the human colon carcinoma cell line, T84. Dig Dis Sci. 2011;56:339–51.PubMed
128.
go back to reference Sun X, Wang X, Wang GD, Xia Y, Liu S, Qu M, et al. Lubiprostone reverses the inhibitory action of morphine on mucosal secretion in human small intestine. Dig Dis Sci. 2011;56:330–8.PubMed Sun X, Wang X, Wang GD, Xia Y, Liu S, Qu M, et al. Lubiprostone reverses the inhibitory action of morphine on mucosal secretion in human small intestine. Dig Dis Sci. 2011;56:330–8.PubMed
129.
go back to reference Fei G, Raehal K, Liu S, Qu MH, Sun X, Wang GD, et al. Lubiprostone reverses the inhibitory action of morphine on intestinal secretion in guinea pig and mouse. J Pharmacol Exp Ther. 2010;334:333–40.PubMed Fei G, Raehal K, Liu S, Qu MH, Sun X, Wang GD, et al. Lubiprostone reverses the inhibitory action of morphine on intestinal secretion in guinea pig and mouse. J Pharmacol Exp Ther. 2010;334:333–40.PubMed
130.
go back to reference Chamberlain SM, Rao SS. Safety evaluation of lubiprostone in the treatment of constipation and irritable bowel syndrome. Expert Opin Drug Saf. 2012;11:841–50.PubMed Chamberlain SM, Rao SS. Safety evaluation of lubiprostone in the treatment of constipation and irritable bowel syndrome. Expert Opin Drug Saf. 2012;11:841–50.PubMed
131.
go back to reference Kalff JC, Schraut WH, Billiar TR, Simmons RL, Bauer AJ. Role of inducible nitric oxide synthase in postoperative intestinal smooth muscle dysfunction in rodents. Gastroenterology. 2000;118:316–27.PubMed Kalff JC, Schraut WH, Billiar TR, Simmons RL, Bauer AJ. Role of inducible nitric oxide synthase in postoperative intestinal smooth muscle dysfunction in rodents. Gastroenterology. 2000;118:316–27.PubMed
132.
go back to reference Bauer AJ, Boeckxstaens GE. Mechanisms of postoperative ileus. Neurogastroenterol Motil. 2004;16:54–60.PubMed Bauer AJ, Boeckxstaens GE. Mechanisms of postoperative ileus. Neurogastroenterol Motil. 2004;16:54–60.PubMed
133.
go back to reference Beard TL, Leslie JB, Nemeth J. The opioid component of delayed gastrointestinal recovery after bowel resection. J Gastrointest Surg. 2011;15:1259–68.PubMed Beard TL, Leslie JB, Nemeth J. The opioid component of delayed gastrointestinal recovery after bowel resection. J Gastrointest Surg. 2011;15:1259–68.PubMed
134.
go back to reference Thompson M, Magnuson B. Management of postoperative ileus. Orthopedics. 2012;35:213–7.PubMed Thompson M, Magnuson B. Management of postoperative ileus. Orthopedics. 2012;35:213–7.PubMed
135.
go back to reference Pfab F, Nowak-Machen M, Napadow V, Fleckenstein J. Alternatives to prokinetics to move the pylorus and colon. Curr Opin Clin Nutr Metab Care. 2012;15:166–73.PubMed Pfab F, Nowak-Machen M, Napadow V, Fleckenstein J. Alternatives to prokinetics to move the pylorus and colon. Curr Opin Clin Nutr Metab Care. 2012;15:166–73.PubMed
136.
go back to reference Schmidt J, Stoffels B, Nazir A. haven-Hudkins DL, Bauer AJ. Alvimopan and COX-2 inhibition reverse opioid and inflammatory components of postoperative ileus. Neurogastroenterol Motil. 2008;20:689–99.PubMed Schmidt J, Stoffels B, Nazir A. haven-Hudkins DL, Bauer AJ. Alvimopan and COX-2 inhibition reverse opioid and inflammatory components of postoperative ileus. Neurogastroenterol Motil. 2008;20:689–99.PubMed
137.
go back to reference Delaney CP, Weese JL, Hyman NH, Bauer J, Techner L, Gabriel K, et al. Phase III trial of alvimopan, a novel, peripherally acting, mu opioid antagonist, for postoperative ileus after major abdominal surgery. Dis Colon Rectum. 2005;48:1114–25.PubMed Delaney CP, Weese JL, Hyman NH, Bauer J, Techner L, Gabriel K, et al. Phase III trial of alvimopan, a novel, peripherally acting, mu opioid antagonist, for postoperative ileus after major abdominal surgery. Dis Colon Rectum. 2005;48:1114–25.PubMed
138.
go back to reference Apfel CC, Jalota L. Can central antiemetic effects of opioids counter-balance opioid-induced nausea and vomiting? Acta Anaesthesiol Scand. 2010;54:129–31.PubMed Apfel CC, Jalota L. Can central antiemetic effects of opioids counter-balance opioid-induced nausea and vomiting? Acta Anaesthesiol Scand. 2010;54:129–31.PubMed
139.
go back to reference Marderstein EL, Delaney CP. Management of postoperative ileus: focus on alvimopan. Ther Clin Risk Manag. 2008;4:965–73.PubMedPubMedCentral Marderstein EL, Delaney CP. Management of postoperative ileus: focus on alvimopan. Ther Clin Risk Manag. 2008;4:965–73.PubMedPubMedCentral
140.
go back to reference Webster L, Jansen JP, Peppin J, Lasko B, Irving G, Morlion B, et al. Alvimopan, a peripherally acting mu-opioid receptor (PAM-OR) antagonist for the treatment of opioid-induced bowel dysfunction: results from a randomized, double-blind, placebo-controlled, dose-finding study in subjects taking opioids for chronic non-cancer pain. Pain. 2008;137:428–40.PubMed Webster L, Jansen JP, Peppin J, Lasko B, Irving G, Morlion B, et al. Alvimopan, a peripherally acting mu-opioid receptor (PAM-OR) antagonist for the treatment of opioid-induced bowel dysfunction: results from a randomized, double-blind, placebo-controlled, dose-finding study in subjects taking opioids for chronic non-cancer pain. Pain. 2008;137:428–40.PubMed
141.
go back to reference Paulson DM, Kennedy DT, Donovick RA, Carpenter RL, Cherubini M, Techner L, et al. Alvimopan: an oral, peripherally acting, mu-opioid receptor antagonist for the treatment of opioid-induced bowel dysfunction—a 21-day treatment-randomized clinical trial. J Pain. 2005;6:184–92.PubMed Paulson DM, Kennedy DT, Donovick RA, Carpenter RL, Cherubini M, Techner L, et al. Alvimopan: an oral, peripherally acting, mu-opioid receptor antagonist for the treatment of opioid-induced bowel dysfunction—a 21-day treatment-randomized clinical trial. J Pain. 2005;6:184–92.PubMed
142.
go back to reference Jansen JP, Lorch D, Langan J, Lasko B, Hermanns K, Kleoudis CS, et al. A randomized, placebo-controlled phase 3 trial (Study SB-767905/012) of alvimopan for opioid-induced bowel dysfunction in patients with non-cancer pain. J Pain. 2011;12:185–93.PubMed Jansen JP, Lorch D, Langan J, Lasko B, Hermanns K, Kleoudis CS, et al. A randomized, placebo-controlled phase 3 trial (Study SB-767905/012) of alvimopan for opioid-induced bowel dysfunction in patients with non-cancer pain. J Pain. 2011;12:185–93.PubMed
143.
go back to reference Irving G, Penzes J, Ramjattan B, Cousins M, Rauck R, Spierings EL, et al. A randomized, placebo-controlled phase 3 trial (Study SB-767905/013) of alvimopan for opioid-induced bowel dysfunction in patients with non-cancer pain. J Pain. 2011;12:175–84.PubMed Irving G, Penzes J, Ramjattan B, Cousins M, Rauck R, Spierings EL, et al. A randomized, placebo-controlled phase 3 trial (Study SB-767905/013) of alvimopan for opioid-induced bowel dysfunction in patients with non-cancer pain. J Pain. 2011;12:175–84.PubMed
144.
go back to reference Wolff BG, Michelassi F, Gerkin TM, Techner L, Gabriel K, Du W, et al. Alvimopan, a novel, peripherally acting mu opioid antagonist: results of a multicenter, randomized, double-blind, placebo-controlled, phase III trial of major abdominal surgery and postoperative ileus. Ann Surg. 2004;240:728–34.PubMed Wolff BG, Michelassi F, Gerkin TM, Techner L, Gabriel K, Du W, et al. Alvimopan, a novel, peripherally acting mu opioid antagonist: results of a multicenter, randomized, double-blind, placebo-controlled, phase III trial of major abdominal surgery and postoperative ileus. Ann Surg. 2004;240:728–34.PubMed
145.
go back to reference Viscusi ER, Goldstein S, Witkowski T, Andonakakis A, Jan R, Gabriel K, et al. Alvimopan, a peripherally acting mu-opioid receptor antagonist, compared with placebo in postoperative ileus after major abdominal surgery: results of a randomized, double-blind, controlled study. Surg Endosc. 2006;20:64–70.PubMed Viscusi ER, Goldstein S, Witkowski T, Andonakakis A, Jan R, Gabriel K, et al. Alvimopan, a peripherally acting mu-opioid receptor antagonist, compared with placebo in postoperative ileus after major abdominal surgery: results of a randomized, double-blind, controlled study. Surg Endosc. 2006;20:64–70.PubMed
146.
go back to reference Buchler MW, Seiler CM, Monson JR, Flamant Y, Thompson-Fawcett MW, Byrne MM, et al. Clinical trial: alvimopan for the management of post-operative ileus after abdominal surgery: results of an international randomized, double-blind, multicentre, placebo-controlled clinical study. Aliment Pharmacol Ther. 2008;28:312–25.PubMed Buchler MW, Seiler CM, Monson JR, Flamant Y, Thompson-Fawcett MW, Byrne MM, et al. Clinical trial: alvimopan for the management of post-operative ileus after abdominal surgery: results of an international randomized, double-blind, multicentre, placebo-controlled clinical study. Aliment Pharmacol Ther. 2008;28:312–25.PubMed
147.
go back to reference Taguchi A, Sharma N, Saleem RM, Sessler DI, Carpenter RL, Seyedsadr M, et al. Selective postoperative inhibition of gastrointestinal opioid receptors. N Engl J Med. 2001;345:935–40.PubMed Taguchi A, Sharma N, Saleem RM, Sessler DI, Carpenter RL, Seyedsadr M, et al. Selective postoperative inhibition of gastrointestinal opioid receptors. N Engl J Med. 2001;345:935–40.PubMed
148.
go back to reference Ludwig K, Enker WE, Delaney CP, Wolff BG, Du W, Fort JG, et al. Gastrointestinal tract recovery in patients undergoing bowel resection: results of a randomized trial of alvimopan and placebo with a standardized accelerated postoperative care pathway. Arch Surg. 2008;143:1098–105.PubMed Ludwig K, Enker WE, Delaney CP, Wolff BG, Du W, Fort JG, et al. Gastrointestinal tract recovery in patients undergoing bowel resection: results of a randomized trial of alvimopan and placebo with a standardized accelerated postoperative care pathway. Arch Surg. 2008;143:1098–105.PubMed
149.
go back to reference Herzog TJ, Coleman RL, Guerrieri JP Jr, Gabriel K, Du W, Techner L, et al. A double-blind, randomized, placebo-controlled phase III study of the safety of alvimopan in patients who undergo simple total abdominal hysterectomy. Am J Obstet Gynecol. 2006;195:445–53.PubMed Herzog TJ, Coleman RL, Guerrieri JP Jr, Gabriel K, Du W, Techner L, et al. A double-blind, randomized, placebo-controlled phase III study of the safety of alvimopan in patients who undergo simple total abdominal hysterectomy. Am J Obstet Gynecol. 2006;195:445–53.PubMed
150.
go back to reference Ludwig K, Viscusi ER, Wolff BG, Delaney CP, Senagore A, Techner L. Alvimopan for the management of postoperative ileus after bowel resection: characterization of clinical benefit by pooled responder analysis. World J Surg. 2010;34:2185–90.PubMedPubMedCentral Ludwig K, Viscusi ER, Wolff BG, Delaney CP, Senagore A, Techner L. Alvimopan for the management of postoperative ileus after bowel resection: characterization of clinical benefit by pooled responder analysis. World J Surg. 2010;34:2185–90.PubMedPubMedCentral
151.
go back to reference Itawi EA, Savoie LM, Hanna AJ, Apostolides GY. Alvimopan addition to a standard perioperative recovery pathway. JSLS. 2011;15:492–8.PubMedPubMedCentral Itawi EA, Savoie LM, Hanna AJ, Apostolides GY. Alvimopan addition to a standard perioperative recovery pathway. JSLS. 2011;15:492–8.PubMedPubMedCentral
152.
go back to reference Poston S, Broder MS, Gibbons MM, Maclaren R, Chang E, Vandepol CJ, et al. Impact of alvimopan (entereg) on hospital costs after bowel resection: results from a large inpatient database. P T. 2011;36:209–20.PubMedPubMedCentral Poston S, Broder MS, Gibbons MM, Maclaren R, Chang E, Vandepol CJ, et al. Impact of alvimopan (entereg) on hospital costs after bowel resection: results from a large inpatient database. P T. 2011;36:209–20.PubMedPubMedCentral
153.
go back to reference Delaney CP, Craver C, Gibbons MM, Rachfal AW, VandePol CJ, Cook SF, et al. Evaluation of clinical outcomes with alvimopan in clinical practice: a national matched-cohort study in patients undergoing bowel resection. Ann Surg. 2012;255:731–8.PubMed Delaney CP, Craver C, Gibbons MM, Rachfal AW, VandePol CJ, Cook SF, et al. Evaluation of clinical outcomes with alvimopan in clinical practice: a national matched-cohort study in patients undergoing bowel resection. Ann Surg. 2012;255:731–8.PubMed
154.
go back to reference Vaughan-Shaw PG, Fecher IC, Harris S, Knight JS. A meta-analysis of the effectiveness of the opioid receptor antagonist alvimopan in reducing hospital length of stay and time to GI recovery in patients enrolled in a standardized accelerated recovery program after abdominal surgery. Dis Colon Rectum. 2012;55:611–20.PubMed Vaughan-Shaw PG, Fecher IC, Harris S, Knight JS. A meta-analysis of the effectiveness of the opioid receptor antagonist alvimopan in reducing hospital length of stay and time to GI recovery in patients enrolled in a standardized accelerated recovery program after abdominal surgery. Dis Colon Rectum. 2012;55:611–20.PubMed
155.
go back to reference Bader S, Jaroslawski K, Blum HE, Becker G. Opioid-induced constipation in advanced illness: safety and efficacy of methylnaltrexone bromide. Clin Med Insights Oncol. 2011;5:201–11.PubMedPubMedCentral Bader S, Jaroslawski K, Blum HE, Becker G. Opioid-induced constipation in advanced illness: safety and efficacy of methylnaltrexone bromide. Clin Med Insights Oncol. 2011;5:201–11.PubMedPubMedCentral
156.
go back to reference Wong BS, Rao AS, Camilleri M, Manabe N, McKinzie S, Busciglio I, et al. The effects of methylnaltrexone alone and in combination with acutely administered codeine on gastrointestinal and colonic transit in health. Aliment Pharmacol Ther. 2010;32:884–93.PubMed Wong BS, Rao AS, Camilleri M, Manabe N, McKinzie S, Busciglio I, et al. The effects of methylnaltrexone alone and in combination with acutely administered codeine on gastrointestinal and colonic transit in health. Aliment Pharmacol Ther. 2010;32:884–93.PubMed
157.
go back to reference Murphy DB, Sutton JA, Prescott LF, Murphy MB. Opioid-induced delay in gastric emptying: a peripheral mechanism in humans. Anesthesiology. 1997;87:765–70.PubMed Murphy DB, Sutton JA, Prescott LF, Murphy MB. Opioid-induced delay in gastric emptying: a peripheral mechanism in humans. Anesthesiology. 1997;87:765–70.PubMed
158.
go back to reference Yuan CS, Foss JF, Osinski J, Toledano A, Roizen MF, Moss J. The safety and efficacy of oral methylnaltrexone in preventing morphine-induced delay in oral-cecal transit time. Clin Pharmacol Ther. 1997;61:467–75.PubMed Yuan CS, Foss JF, Osinski J, Toledano A, Roizen MF, Moss J. The safety and efficacy of oral methylnaltrexone in preventing morphine-induced delay in oral-cecal transit time. Clin Pharmacol Ther. 1997;61:467–75.PubMed
159.
go back to reference Michna E, Weil AJ, Duerden M, Schulman S, Wang W, Tzanis E, et al. Efficacy of subcutaneous methylnaltrexone in the treatment of opioid-induced constipation: a responder post hoc analysis. Pain Med. 2011;12:1223–30.PubMed Michna E, Weil AJ, Duerden M, Schulman S, Wang W, Tzanis E, et al. Efficacy of subcutaneous methylnaltrexone in the treatment of opioid-induced constipation: a responder post hoc analysis. Pain Med. 2011;12:1223–30.PubMed
160.
go back to reference Iyer SS, Randazzo BP, Tzanis EL, Schulman SL, Zhang H, Wang W, et al. Effect of subcutaneous methylnaltrexone on patient-reported constipation symptoms. Value Health. 2011;14:177–83.PubMed Iyer SS, Randazzo BP, Tzanis EL, Schulman SL, Zhang H, Wang W, et al. Effect of subcutaneous methylnaltrexone on patient-reported constipation symptoms. Value Health. 2011;14:177–83.PubMed
161.
go back to reference Michna E, Blonsky ER, Schulman S, Tzanis E, Manley A, Zhang H, et al. Subcutaneous methylnaltrexone for treatment of opioid-induced constipation in patients with chronic, nonmalignant pain: a randomized controlled study. J Pain. 2011;12:554–62.PubMed Michna E, Blonsky ER, Schulman S, Tzanis E, Manley A, Zhang H, et al. Subcutaneous methylnaltrexone for treatment of opioid-induced constipation in patients with chronic, nonmalignant pain: a randomized controlled study. J Pain. 2011;12:554–62.PubMed
162.
go back to reference Yu CS, Chun HK, Stambler N, Carpenito J, Schulman S, Tzanis E, et al. Safety and efficacy of methylnaltrexone in shortening the duration of postoperative ileus following segmental colectomy: results of two randomized, placebo-controlled phase 3 trials. Dis Colon Rectum. 2011;54:570–8.PubMed Yu CS, Chun HK, Stambler N, Carpenito J, Schulman S, Tzanis E, et al. Safety and efficacy of methylnaltrexone in shortening the duration of postoperative ileus following segmental colectomy: results of two randomized, placebo-controlled phase 3 trials. Dis Colon Rectum. 2011;54:570–8.PubMed
163.
go back to reference Anissian L, Schwartz HW, Vincent K, Vincent HK, Carpenito J, Stambler N, et al. Subcutaneous methylnaltrexone for treatment of acute opioid-induced constipation: phase 2 study in rehabilitation after orthopedic surgery. J Hosp Med. 2012;7:67–72.PubMed Anissian L, Schwartz HW, Vincent K, Vincent HK, Carpenito J, Stambler N, et al. Subcutaneous methylnaltrexone for treatment of acute opioid-induced constipation: phase 2 study in rehabilitation after orthopedic surgery. J Hosp Med. 2012;7:67–72.PubMed
164.
go back to reference Chamberlain BH, Cross K, Winston JL, Thomas J, Wang W, Su C, et al. Methylnaltrexone treatment of opioid-induced constipation in patients with advanced illness. J Pain Symptom Manage. 2009;38:683–90.PubMed Chamberlain BH, Cross K, Winston JL, Thomas J, Wang W, Su C, et al. Methylnaltrexone treatment of opioid-induced constipation in patients with advanced illness. J Pain Symptom Manage. 2009;38:683–90.PubMed
165.
go back to reference Slatkin N, Thomas J, Lipman AG, Wilson G, Boatwright ML, Wellman C, et al. Methylnaltrexone for treatment of opioid-induced constipation in advanced illness patients. J Support Oncol. 2009;7:39–46.PubMed Slatkin N, Thomas J, Lipman AG, Wilson G, Boatwright ML, Wellman C, et al. Methylnaltrexone for treatment of opioid-induced constipation in advanced illness patients. J Support Oncol. 2009;7:39–46.PubMed
166.
go back to reference Portenoy RK, Thomas J, Moehl Boatwright ML, Tran D, Galasso FL, Stambler N, et al. Subcutaneous methylnaltrexone for the treatment of opioid-induced constipation in patients with advanced illness: a double-blind, randomized, parallel group, dose-ranging study. J Pain Symptom Manage. 2008;35:458–68. Portenoy RK, Thomas J, Moehl Boatwright ML, Tran D, Galasso FL, Stambler N, et al. Subcutaneous methylnaltrexone for the treatment of opioid-induced constipation in patients with advanced illness: a double-blind, randomized, parallel group, dose-ranging study. J Pain Symptom Manage. 2008;35:458–68.
167.
go back to reference Lipman AG, Karver S, Cooney GA, Stambler N, Israel RJ. Methylnaltrexone for opioid-induced constipation in patients with advanced illness: a 3-month open-label treatment extension study. J Pain Palliat Care Pharmacother. 2011;25:136–45.PubMed Lipman AG, Karver S, Cooney GA, Stambler N, Israel RJ. Methylnaltrexone for opioid-induced constipation in patients with advanced illness: a 3-month open-label treatment extension study. J Pain Palliat Care Pharmacother. 2011;25:136–45.PubMed
168.
go back to reference Yuan CS, Foss JF, O’connor M, Osinski J, Karrison T, Moss J, et al. Methylnaltrexone for reversal of constipation due to chronic methadone use: a randomized controlled trial. JAMA. 2000;283:367–72.PubMed Yuan CS, Foss JF, O’connor M, Osinski J, Karrison T, Moss J, et al. Methylnaltrexone for reversal of constipation due to chronic methadone use: a randomized controlled trial. JAMA. 2000;283:367–72.PubMed
169.
go back to reference Garnock-Jones KP, McKeage K. Methylnaltrexone. Drugs. 2010;70:919–28. Garnock-Jones KP, McKeage K. Methylnaltrexone. Drugs. 2010;70:919–28.
170.
go back to reference Garten L, Degenhardt P, Buhrer C. Resolution of opioid-induced postoperative ileus in a newborn infant after methylnaltrexone. J Pediatr Surg. 2011;46:e13–5.PubMed Garten L, Degenhardt P, Buhrer C. Resolution of opioid-induced postoperative ileus in a newborn infant after methylnaltrexone. J Pediatr Surg. 2011;46:e13–5.PubMed
171.
go back to reference Fichna J, Gach K, Perlikowska R, Cravezic A, Bonnet JJ, do-Rego JC, et al. Novel endomorphin analogues with antagonist activity at the mu-opioid receptor in the gastrointestinal tract. Regul Pept. 2010;162:109–14. Fichna J, Gach K, Perlikowska R, Cravezic A, Bonnet JJ, do-Rego JC, et al. Novel endomorphin analogues with antagonist activity at the mu-opioid receptor in the gastrointestinal tract. Regul Pept. 2010;162:109–14.
172.
go back to reference Fichna J, Storr MA. Brain-gut interactions in IBS. Front Pharmacol. 2012;3:127. Fichna J, Storr MA. Brain-gut interactions in IBS. Front Pharmacol. 2012;3:127.
174.
go back to reference Andrews EB, Eaton SC, Hollis KA, Hopkins JS, Ameen V, Hamm LR, et al. Prevalence and demographics of irritable bowel syndrome: results from a large web-based survey. Aliment Pharmacol Ther. 2005;22:935–42.PubMed Andrews EB, Eaton SC, Hollis KA, Hopkins JS, Ameen V, Hamm LR, et al. Prevalence and demographics of irritable bowel syndrome: results from a large web-based survey. Aliment Pharmacol Ther. 2005;22:935–42.PubMed
175.
go back to reference Wilkins T, Pepitone C, Alex B, Schade RR. Diagnosis and management of IBS in adults. Am Fam Physician. 2012;86:419–26.PubMed Wilkins T, Pepitone C, Alex B, Schade RR. Diagnosis and management of IBS in adults. Am Fam Physician. 2012;86:419–26.PubMed
176.
go back to reference Philpott H, Gibson P, Thien F. Irritable bowel syndrome—an inflammatory disease involving mast cells. Asia Pac Allergy. 2011;1:36–42.PubMedPubMedCentral Philpott H, Gibson P, Thien F. Irritable bowel syndrome—an inflammatory disease involving mast cells. Asia Pac Allergy. 2011;1:36–42.PubMedPubMedCentral
177.
go back to reference Alaradi O, Barkin JS. Irritable bowel syndrome: update on pathogenesis and management. Med Princ Pract. 2002;11:2–17.PubMed Alaradi O, Barkin JS. Irritable bowel syndrome: update on pathogenesis and management. Med Princ Pract. 2002;11:2–17.PubMed
178.
go back to reference Distrutti E, Mencarelli A, Renga B, Caliendo G, Santagada V, Severino B, et al. A nitro-arginine derivative of trimebutine (NO2-Arg-Trim) attenuates pain induced by colorectal distension in conscious rats. Pharmacol Res. 2009;59:319–29.PubMed Distrutti E, Mencarelli A, Renga B, Caliendo G, Santagada V, Severino B, et al. A nitro-arginine derivative of trimebutine (NO2-Arg-Trim) attenuates pain induced by colorectal distension in conscious rats. Pharmacol Res. 2009;59:319–29.PubMed
179.
go back to reference Lee N, Wald A. Linaclotide: evidence for its potential use in irritable bowel syndrome and chronic constipation. Core Evid. 2012;7:39–47.PubMedPubMedCentral Lee N, Wald A. Linaclotide: evidence for its potential use in irritable bowel syndrome and chronic constipation. Core Evid. 2012;7:39–47.PubMedPubMedCentral
180.
go back to reference Olden KW. Targeted therapies for diarrhea-predominant irritable bowel syndrome. Clin Exp Gastroenterol. 2012;5:69–100.PubMedPubMedCentral Olden KW. Targeted therapies for diarrhea-predominant irritable bowel syndrome. Clin Exp Gastroenterol. 2012;5:69–100.PubMedPubMedCentral
181.
go back to reference Gross KJ, Pothoulakis C. Role of neuropeptides in inflammatory bowel disease. Inflamm Bowel Dis. 2007;13:918–32.PubMed Gross KJ, Pothoulakis C. Role of neuropeptides in inflammatory bowel disease. Inflamm Bowel Dis. 2007;13:918–32.PubMed
182.
go back to reference Philippe D, Dubuquoy L, Groux H, Brun V, Chuoi-Mariot MT, Gaveriaux-Ruff C, et al. Anti-inflammatory properties of the mu opioid receptor support its use in the treatment of colon inflammation. J Clin Invest. 2003;111:1329–38.PubMedPubMedCentral Philippe D, Dubuquoy L, Groux H, Brun V, Chuoi-Mariot MT, Gaveriaux-Ruff C, et al. Anti-inflammatory properties of the mu opioid receptor support its use in the treatment of colon inflammation. J Clin Invest. 2003;111:1329–38.PubMedPubMedCentral
183.
go back to reference Srinath AI, Walter C, Newara MC, Szigethy EM. Pain management in patients with inflammatory bowel disease: insights for the clinician. Therap Adv Gastroenterol. 2012;5:339–57.PubMedPubMedCentral Srinath AI, Walter C, Newara MC, Szigethy EM. Pain management in patients with inflammatory bowel disease: insights for the clinician. Therap Adv Gastroenterol. 2012;5:339–57.PubMedPubMedCentral
184.
go back to reference Cabot PJ, Carter L, Gaiddon C, Zhang Q, Schafer M, Loeffler JP, et al. Immune cell-derived beta-endorphin. Production, release, and control of inflammatory pain in rats. J Clin Invest. 1997;100:142–8.PubMedPubMedCentral Cabot PJ, Carter L, Gaiddon C, Zhang Q, Schafer M, Loeffler JP, et al. Immune cell-derived beta-endorphin. Production, release, and control of inflammatory pain in rats. J Clin Invest. 1997;100:142–8.PubMedPubMedCentral
185.
go back to reference Tomassini N, Renaud FL, Roy S, Loh HH. Mu and delta receptors mediate morphine effects on phagocytosis by murine peritoneal macrophages. J Neuroimmunol. 2003;136:9–16.PubMed Tomassini N, Renaud FL, Roy S, Loh HH. Mu and delta receptors mediate morphine effects on phagocytosis by murine peritoneal macrophages. J Neuroimmunol. 2003;136:9–16.PubMed
186.
go back to reference Philippe D, Chakass D, Thuru X, Zerbib P, Tsicopoulos A, Geboes K, et al. Mu opioid receptor expression is increased in inflammatory bowel diseases: implications for homeostatic intestinal inflammation. Gut. 2006;55:815–23.PubMed Philippe D, Chakass D, Thuru X, Zerbib P, Tsicopoulos A, Geboes K, et al. Mu opioid receptor expression is increased in inflammatory bowel diseases: implications for homeostatic intestinal inflammation. Gut. 2006;55:815–23.PubMed
187.
go back to reference Goldsmith JR, Uronis JM, Jobin C. Mu opioid signaling protects against acute murine intestinal injury in a manner involving Stat3 signaling. Am J Pathol. 2011;179:673–83.PubMed Goldsmith JR, Uronis JM, Jobin C. Mu opioid signaling protects against acute murine intestinal injury in a manner involving Stat3 signaling. Am J Pathol. 2011;179:673–83.PubMed
188.
go back to reference Matters GL, Harms JF, McGovern C, Fitzpatrick L, Parikh A, Nilo N, et al. The opioid antagonist naltrexone improves murine inflammatory bowel disease. J Immunotoxicol. 2008;5:179–87.PubMed Matters GL, Harms JF, McGovern C, Fitzpatrick L, Parikh A, Nilo N, et al. The opioid antagonist naltrexone improves murine inflammatory bowel disease. J Immunotoxicol. 2008;5:179–87.PubMed
189.
go back to reference Jan WC, Chen CH, Hsu K, Tsai PS, Huang CJ. L-type calcium channels and mu-opioid receptors are involved in mediating the anti-inflammatory effects of naloxone. J Surg Res. 2011;167:e263–72.PubMed Jan WC, Chen CH, Hsu K, Tsai PS, Huang CJ. L-type calcium channels and mu-opioid receptors are involved in mediating the anti-inflammatory effects of naloxone. J Surg Res. 2011;167:e263–72.PubMed
190.
go back to reference Smith JP, Bingaman SI, Ruggiero F, Mauger DT, Mukherjee A, McGovern CO, et al. Therapy with the opioid antagonist naltrexone promotes mucosal healing in active Crohn’s disease: a randomized placebo-controlled trial. Dig Dis Sci. 2011;56:2088–97.PubMedPubMedCentral Smith JP, Bingaman SI, Ruggiero F, Mauger DT, Mukherjee A, McGovern CO, et al. Therapy with the opioid antagonist naltrexone promotes mucosal healing in active Crohn’s disease: a randomized placebo-controlled trial. Dig Dis Sci. 2011;56:2088–97.PubMedPubMedCentral
191.
go back to reference Hilburger ME, Adler MW, Truant AL, Meissler JJ Jr, Satishchandran V, Rogers TJ, et al. Morphine induces sepsis in mice. J Infect Dis. 1997;176:183–8.PubMed Hilburger ME, Adler MW, Truant AL, Meissler JJ Jr, Satishchandran V, Rogers TJ, et al. Morphine induces sepsis in mice. J Infect Dis. 1997;176:183–8.PubMed
192.
go back to reference Topcu I, Ekici NZ, Isik R, Sakarya M. The effects of tramadol and fentanyl on gastrointestinal motility in septic rats. Anesth Analg. 2006;102:876–81.PubMed Topcu I, Ekici NZ, Isik R, Sakarya M. The effects of tramadol and fentanyl on gastrointestinal motility in septic rats. Anesth Analg. 2006;102:876–81.PubMed
194.
go back to reference Tang CW, Feng WM, Du HM, Bao Y, Zhu M. Delayed administration of D-Ala2-D-Leu5-enkephalin, a delta-opioid receptor agonist, improves survival in a rat model of sepsis. Tohoku J Exp Med. 2011;224:69–76.PubMed Tang CW, Feng WM, Du HM, Bao Y, Zhu M. Delayed administration of D-Ala2-D-Leu5-enkephalin, a delta-opioid receptor agonist, improves survival in a rat model of sepsis. Tohoku J Exp Med. 2011;224:69–76.PubMed
195.
go back to reference Goins WF, Cohen JB, Glorioso JC. Gene therapy for the treatment of chronic peripheral nervous system pain. Neurobiol Dis. 2012;48:255–70.PubMedPubMedCentral Goins WF, Cohen JB, Glorioso JC. Gene therapy for the treatment of chronic peripheral nervous system pain. Neurobiol Dis. 2012;48:255–70.PubMedPubMedCentral
196.
go back to reference Fink DJ, Wechuck J, Mata M, Glorioso JC, Goss J, Krisky D, et al. Gene therapy for pain: results of a phase I clinical trial. Ann Neurol. 2011;70:207–12.PubMedPubMedCentral Fink DJ, Wechuck J, Mata M, Glorioso JC, Goss J, Krisky D, et al. Gene therapy for pain: results of a phase I clinical trial. Ann Neurol. 2011;70:207–12.PubMedPubMedCentral
Metadata
Title
Physiology, signaling, and pharmacology of opioid receptors and their ligands in the gastrointestinal tract: current concepts and future perspectives
Authors
Marta Sobczak
Maciej Sałaga
Martin A. Storr
Jakub Fichna
Publication date
01-01-2014
Publisher
Springer Japan
Published in
Journal of Gastroenterology / Issue 1/2014
Print ISSN: 0944-1174
Electronic ISSN: 1435-5922
DOI
https://doi.org/10.1007/s00535-013-0753-x

Other articles of this Issue 1/2014

Journal of Gastroenterology 1/2014 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.