The gut-brain interaction in opioid tolerance

Hamid I Akbarali, William L Dewey, Hamid I Akbarali, William L Dewey

Abstract

The prevailing opioid crisis has necessitated the need to understand mechanisms leading to addiction and tolerance, the major contributors to overdose and death and to develop strategies for developing drugs for pain treatment that lack abuse liability and side-effects. Opioids are commonly used for treatment of pain and symptoms of inflammatory bowel disease. The significant effect of opioids in the gut, both acute and chronic, includes persistent constipation and paradoxically may also worsen pain symptoms. Recent work has suggested a significant role of the gastrointestinal microbiome in behavioral responses to opioids, including the development of tolerance to its pain-relieving effects. In this review, we present current concepts of gut-brain interaction in analgesic tolerance to opioids and suggest that peripheral mechanisms emanating from the gut can profoundly affect central control of opioid function.

Copyright © 2017 Elsevier Ltd. All rights reserved.

Figures

Figure 1
Figure 1
Schema of gut-brain interaction through the dorsal root ganglia neurons. The gut microbiome is altered in the presence of chronic morphine. Bacterial translocation due to disruption of the epithelial barrier results in activation of TLR’s on enteric glia increasing connexin43 and P2X expression. Cytokines (Inflammatory soup) released from glia affect extrinsic sensory afferents whose cell bodies lie within the dorsal root ganglia (DRG) and induce tolerance to opioids.

References

    1. Pappagallo M. Incidence, prevalence, and management of opioid bowel dysfunction. American journal of surgery. 2001;182:11S–18S.
    1. Holzer P. Treatment of opioid-induced gut dysfunction. Expert Opin Investig Drugs. 2007;16:181–194.
    1. Galligan JJ, Akbarali HI. Molecular Physiology of Enteric Opioid Receptors. Am J Gastroenterol. 2014;2:17–21.
    1. Akbarali HI, Inkisar A, Dewey WL. Site and mechanism of morphine tolerance in the gastrointestinal tract. Neurogastroenterol Motil. 2014;26:1361–1367.
    1. Camilleri M, Lembo A, Katzka DA. Opioids in Gastroenterology: Treating Adverse Effects and Creating Therapeutic Benefits. Clin Gastroenterol Hepatol. 2017;15:1338–1349.
    1. North RA, Tonini M. The mechanism of action of narcotic analgesics in the guinea-pig ileum. Br J Pharmacol. 1977;61:541–549.
    1. Smith TH, Grider JR, Dewey WL, Akbarali HI. Morphine Decreases Enteric Neuron Excitability via Inhibition of Sodium Channels. PloS one. 2012;7:e45251.
    1. Smith TH, Ngwainmbi J, Grider JR, Dewey WL, Akbarali HI. An in-vitro preparation of isolated enteric neurons and glia from the myenteric plexus of the adult mouse. Journal of visualized experiments: JoVE. 2013;(78) doi: 10.3791/50688.
    1. Al-Hasani R, Bruchas MR. Molecular mechanisms of opioid receptor-dependent signaling and behavior. Anesthesiology. 2011;115:1363–1381.
    1. Williams JT, Ingram SL, Henderson G, Chavkin C, von Zastrow M, Schulz S, Koch T, Evans CJ, Christie MJ. Regulation of mu-opioid receptors: desensitization, phosphorylation, internalization, and tolerance. Pharmacol Rev. 2013;65:223–254.
    1. White JM, Irvine RJ. Mechanisms of fatal opioid overdose. Addiction. 1999;94:961–972.
    1. Kang M, Maguma HT, Smith TH, Ross GR, Dewey WL, Akbarali HI. The role of beta-arrestin2 in the mechanism of morphine tolerance in the mouse and guinea pig gastrointestinal tract. J Pharmacol Exp Ther. 2012;340:567–576.
    1. Violin JD, Crombie AL, Soergel DG, Lark MW. Biased ligands at G-protein-coupled receptors: promise and progress. Trends Pharmacol Sci. 2014;35:308–316.
    1. DeWire SM, Yamashita DS, Rominger DH, Liu G, Cowan CL, Graczyk TM, Chen XT, Pitis PM, Gotchev D, Yuan C, Koblish M, Lark MW, Violin JD. A G protein-biased ligand at the mu-opioid receptor is potently analgesic with reduced gastrointestinal and respiratory dysfunction compared with morphine. J Pharmacol Exp Ther. 2013;344:708–717.
    1. Altarifi AA, David B, Muchhala KH, Blough BE, Akbarali H, Negus SS. Effects of acute and repeated treatment with the biased mu opioid receptor agonist TRV130 (oliceridine) on measures of antinociception, gastrointestinal function, and abuse liability in rodents. J Psychopharmacol. 2017 269881116689257.
    1. Viscusi ER, Webster L, Kuss M, Daniels S, Bolognese JA, Zuckerman S, Soergel DG, Subach RA, Cook E, Skobieranda F. A randomized, phase 2 study investigating TRV130, a biased ligand of the mu-opioid receptor, for the intravenous treatment of acute pain. Pain. 2016;157:264–272.
    1. Manglik A, Lin H, Aryal DK, McCorvy JD, Dengler D, Corder G, Levit A, Kling RC, Bernat V, Hubner H, Huang XP, Sassano MF, Giguere PM, Lober S, Da D, Scherrer G, Kobilka BK, Gmeiner P, Roth BL, Shoichet BK. Structure-based discovery of opioid analgesics with reduced side effects. Nature. 2016;537:185–190.
    1. Jones JL, Loftus EV., Jr Avoiding the vicious cycle of prolonged opioid use in Crohn’s disease. Am J Gastroenterol. 2005;100:2230–2232.
    1. Cross RK, Wilson KT, Binion DG. Narcotic use in patients with Crohn’s disease. Am J Gastroenterol. 2005;100:2225–2229.
    1. Lichtenstein GR, Feagan BG, Cohen RD, Salzberg BA, Diamond RH, Price S, Langholff W, Londhe A, Sandborn WJ. Serious infection and mortality in patients with Crohn’s disease: more than 5 years of follow-up in the TREAT registry. Am J Gastroenterol. 2012;107:1409–1422.
    1. Hilburger ME, Adler MW, Truant AL, Meissler JJ, Jr, Satishchandran V, Rogers TJ, Eisenstein TK. Morphine induces sepsis in mice. J Infect Dis. 1997;176:183–188.
    1. Roy S, Barke RA, Loh HH. MU-opioid receptor-knockout mice: role of mu-opioid receptor in morphine mediated immune functions. Brain Res Mol Brain Res. 1998;61:190–194.
    1. Acharya C, Betrapally NS, Gillevet PM, Sterling RK, Akbarali H, White MB, Ganapathy D, Fagan A, Sikaroodi M, Bajaj JS. Chronic opioid use is associated with altered gut microbiota and predicts readmissions in patients with cirrhosis. Alimentary pharmacology & therapeutics. 2017;45:319–331. This study examines the effect of opioids on readmissions and on gut microbiota composition in a cohort of cirrhotic patients. There is significant dysbiosis with chronic opioids and is predictive of readmission.
    1. Xu Y, Xie Z, Wang H, Shen Z, Guo Y, Gao Y, Chen X, Wu Q, Li X, Wang K. Bacterial Diversity of Intestinal Microbiota in Patients with Substance Use Disorders Revealed by 16S rRNA Gene Deep Sequencing. Sci Rep. 2017;7:3628. This study examines the composition and changes in the gut microbiota of 45 patients with substance abuse disorders in China. Abundance analysis showed that both age-matched and long-term substance abuse results in significant changes, particularly in the abundance of Bacteioides.
    1. Meng J, Sindberg GM, Roy S. Disruption of gut homeostasis by opioids accelerates HIV disease progression. Frontiers in microbiology. 2015;6:643.
    1. Meng J, Yu H, Ma J, Wang J, Banerjee S, Charboneau R, Barke RA, Roy S. Morphine induces bacterial translocation in mice by compromising intestinal barrier function in a TLR-dependent manner. PLoS One. 2013;8:e54040.
    1. Kang M, Mischel RA, Bhave S, Komla E, Cho A, Huang C, Dewey WL, Akbarali HI. The effect of gut microbiome on tolerance to morphine mediated antinociception in mice. Sci Rep. 2017;7:42658. Depletion of gut bacteria with antibiotics prevents morphine tolerance development to antinociception. Lack of tolerance also occurs in isolated DRG neurons.
    1. Chow AK, Gulbransen BD. Potential roles of enteric glia in bridging neuroimmune communication in the gut. Am J Physiol Gastrointest Liver Physiol. 2017;312:G145–G152.
    1. Bhave S, Gade A, Kang M, Hauser KF, Dewey WL, Akbarali HI. Connexin-purinergic signaling in enteric glia mediates the prolonged effect of morphine on constipation. FASEB J. 2017;31:2649–2660. This paper shows increased P2X mediated currrents in isolated enteric glia from chronic morphine treated mice. P2X and Connexin 43 mRNA were upregulated with lipopolysaccharide treatment of enteric glia. Inhibition of connexin43 with carbenoxolone reduced inflammation in the colon and reduced opioid-induced constipation.
    1. Linan-Rico A, Turco F, Ochoa-Cortes F, Harzman A, Needleman BJ, Arsenescu R, Abdel-Rasoul M, Fadda P, Grants I, Whitaker E, Cuomo R, Christofi FL. Molecular Signaling and Dysfunction of the Human Reactive Enteric Glial Cell Phenotype: Implications for GI Infection, IBD, POI, Neurological, Motility, and GI Disorders. Inflamm Bowel Dis. 2016;22:1812–1834.
    1. Fitting S, Ngwainmbi J, Kang M, Khan FA, Stevens DL, Dewey WL, Knapp PE, Hauser KF, Akbarali HI. Sensitization of enteric neurons to morphine by HIV-1 Tat protein. Neurogastroenterol Motil. 2015;27:468–480.
    1. Galligan JJ. HIV, opiates, and enteric neuron dysfunction. Neurogastroenterol Motil. 2015;27:449–454.
    1. Rao S, Kupfer Y, Pagala M, Chapnick E, Tessler S. Systemic absorption of oral vancomycin in patients with Clostridium difficile infection. Scandinavian journal of infectious diseases. 2011;43:386–388.
    1. Corder G, Tawfik VL, Wang D, Sypek EI, Low SA, Dickinson JR, Sotoudeh C, Clark JD, Barres BA, Bohlen CJ, Scherrer G. Loss of mu opioid receptor signaling in nociceptors, but not microglia, abrogates morphine tolerance without disrupting analgesia. Nature medicine. 2017;23:164–173. This study utilized conditional mu-opioid receptor knock out in DRG neurons and shows that loss of signaling in DRG prevents onset of tolerance and opioid-induced hyperalgesia. Similar effects were obtained with the peripheral opioid antagonist, methylnaltrexone.
    1. Targownik LE, Nugent Z, Singh H, Bugden S, Bernstein CN. The prevalence and predictors of opioid use in inflammatory bowel disease: a population-based analysis. Am J Gastroenterol. 2014;109:1613–1620.
    1. Piekarek K, Israelsson LA. Perforated colonic diverticular disease: the importance of NSAIDs, opioids, corticosteroids, and calcium channel blockers. Int J Colorectal Dis. 2008;23:1193–1197.
    1. Vuong HE, Yano JM, Fung TC, Hsiao EY. The Microbiome and Host Behavior. Annu Rev Neurosci. 2017;40:21–49.
    1. Kiraly DD, Walker DM, Calipari ES, Labonte B, Issler O, Pena CJ, Ribeiro EA, Russo SJ, Nestler EJ. Alterations of the Host Microbiome Affect Behavioral Responses to Cocaine. Sci Rep. 2016;6:35455.

Source: PubMed

3
Prenumerera