The effect of metformin on morphine analgesic tolerance and dependence in rats

Iman Fatemi , Morteza Amirteimoury, Ali Shamsizadeh, Ayat Kaeidi

Abstract


Opiate tolerance and dependence is a worldwide public health problem and gives a significant burden to society. The aim of this study was to evaluate the effects of metformin (MET) on development and expression of morphine tolerance and dependence in rats. For induction of tolerance, morphine sulfate was injected (10 mg/kg, twice a day, s.c.) for 7 days. Animals received metformin (5 and 50 mg/kg, orally, daily) during the examination period for assessing the development of morphine tolerance and dependence. In order to evaluate the expression of morphine tolerance and dependence, single doses of MET (5 and 50 mg/kg, orally) were administered on day 7. Tail flick test was performed to assess the induction of morphine tolerance. For evaluation of morphine dependence, naloxone-induced jumping (5 mg/kg, s.c.) was monitored. Our results showed that 7 days coadministration of 50 mg/kg of MET significantly reduced the development of morphine analgesic tolerance versus morphine + saline treated rats (P < 0.001). Treatment with 50 mg/kg MET reduced the incidence and frequency of jumping in naloxone injected animals                  (P < 0.01). It is notable that single dose administration of MET, did not prevent the expression of analgesic tolerance and physical dependence to morphine. Based on these results, it can be concluded that MET attenuates the development of morphine analgesic tolerance and dependence in rats.


Keywords


Metformin; Morphine; Physical dependence; Tolerance

Full Text:

PDF

References


Somogyi AA, Barratt DT, Coller JK. Pharmacogenetics of opioids. Clin Pharmacol Ther. 2007;81(3):429-444.

Hajhashemi V, Dehdashti Kh. Antinociceptive effect of clavulanic acid and its preventive activity against development of morphine tolerance and dependence in animal models. Res Pharm Sci. 2014;9(5):315-321.

Ghannadi A, Hajhashemi V, Abrishami R. Effects of the Persian Carum copticum fruit extracts on morphine withdrawal syndrome in mice. Res Pharm Sci. 2012;7(3):127-131.

Rezazadeh H, Hosseini Kahnouei M, Hassanshahi G, Allahtavakoli M, Shamsizadeh A, Roohbakhsh A, et al. Regulatory effects of chronic low-dose morphine on nitric oxide level along with baroreflex sensitivity in two-kidney one-clip hypertensive rats. Iran J Kidney Dis. 2014;8(3):194-200.

DuPen A, Shen D, Ersek M. Mechanisms of opioid-induced tolerance and hyperalgesia. Pain Manag Nurs. 2007;8(3):113-121.

Morgan MM, Christie MJ. Analysis of opioid efficacy, tolerance, addiction and dependence from cell culture to human. Br J Pharmacol. 2011;164(4):1322-1334.

Xin W, Chun W, Ling L, Wei W. Role of melatonin in the prevention of morphine-induced hyperalgesia and spinal glial activation in rats: protein kinase C pathway involved. Int J Neurosci. 2012;122(3): 154-163.

Koob GF, Sanna PP, and Bloom FE. Neuroscience of addiction. Neuron. 1998;21(3):467-476.

Joseph H, Stancliff S, Langrod J. Methadone maintenance treatment (MMT): a review of historical and clinical issues. Mt Sinai J Med. 2000;67(5-6):347-364.

Song L, Wu C, Zuo Y. Melatonin prevents morphine-induced hyperalgesia and tolerance in rats: role of protein kinase C and N-methyl-D-aspartate receptors. BMC Anesthesiol. 2015;15:12-20.

Maldonado R, Stinus L, Gold LH, Koob GF. Role of different brain structures in the expression of the physical morphine withdrawal syndrome. J Pharmacol Exp Ther. 1992;261(2):669-677.

Mao J, Price DD, Mayer DJ. Mechanisms of hyperalgesia and morphine tolerance: a current view of their possible interactions. Pain. 1995;62(3):259-274.

UK Prospective Diabetes Study (UKPDS) Group. Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). Lancet. 1998;352(9131):854-865.

Malin SK, Kashyap SR. Effects of metformin on weight loss: potential mechanisms. Curr Opin Endocrinol Diabetes Obes. 2014;21(5):323-329.

Gundelach T, Rodewald M, Bekes I, Janni W, Hancke K. Metformin for the treatment of polycystic ovary syndrome. Med Monatsschr Pharm. 2016;39(2):75-78.

Liu SN, Liu Q, Sun SJ, Hou SC, Wang Y, Shen ZF. Metformin ameliorates β-cell dysfunction by regulating inflammation production, ion and hormone homeostasis of pancreas in diabetic KKAy mice. Yao Xue Xue Bao. 2014;49(11):1554-1562.

Zhou C, Sun R, Zhuang S, Sun C, Jiang Y, Cui Y, et al. Metformin prevents cerebellar granule neurons against glutamate-induced neurotoxicity. Brain Res Bull. 2016;121:241-245.

Fatemi I, Khaluoi A, Kaeidi A, Shamsizadeh A, Heydari S, Allahtavakoli M. Protective effect of metformin on D-galactose-induced aging model in mice. Iran J Basic Med Sci. 2017;21(1):19-25.

Bonnefont-Rousselot D, Raji B, Walrand S, Gardes-Albert M, Jore D, Legrand A, et al. An intracellular modulation of free radical production could contribute to the beneficial effects of metformin towards oxidative stress. Metabolism. 2003;52(5):586-589.

Bułdak Ł, Machnik G, Bułdak RJ, Łabuzek K, Bołdys A, Okopień B. Exenatide and metformin express their anti-inflammatory effects on human monocytes/macrophages by the attenuation of MAPKs and NFκB signaling. Naunyn Schmiedebergs Arch Pharmacol. 2016;389(10):1103-1115.

Deng T, Zheng YR, Hou WW, Yuan Y, Shen Z, Wu XL, et al. Pre-stroke metformin treatment is neuroprotective involving AMPK reduction. Neurochem Res. 2016;41(10):2719-2727.

Wang C, Liu C, Gao K, Zhao H, Zhou Z, Shen Z, et al. Metformin preconditioning provide neuroprotection through enhancement of autophagy and suppression of inflammation and apoptosis after spinal cord injury. Biochem Biophys Res Commun. 2016;477(4):534-540.

Pan Y, Sun X, Jiang L, Hu L, Kong H, Han Y, et al. Metformin reduces morphine tolerance by inhibiting microglial-mediated neuroinflammation. J Neuroinflammation. 2016;13(1):294-301.

Hajializadeh Z, Nasri S, Kaeidi A, Sheibani V, Rasoulian B, Esmaeili-Mahani S. Inhibitory effect of Thymus caramanicus Jalas on hyperglycemia-induced apoptosis in in vitro and in vivo models of diabetic neuropathic pain. J Ethnopharmacol. 2014;153(3):596-603.

Nunez C, Foldes A, Laorden ML, Milanes MV, Kovacs KJ. Activation of stress-related hypothalamic neuropeptide gene expression during morphine withdrawal. J Neurochem. 2007;101(4): 1060-1071.

Robinson TE, Kolb B. Structural plasticity associated with exposure to drugs of abuse. Neuropharmacology. 2004;47(1):33-46.

Atici S, Cinel L, Cinel I, Doruk N, Aktekin M, Akca A, et al. Opioid neurotoxicity: comparison of morphine and tramadol in an experimental rat model. Int J Neurosci. 2004;114(8):1001-1011.

DeLeo JA, Tanga FY, Tawfik VL. Neuroimmune activation and neuroinflammation in chronic pain and opioid tolerance/hyperalgesia. Neuroscientist. 2004;10(1):40-52.

Muscoli C, Cuzzocrea S, Ndengele MM, Mollace V, Porreca F, Fabrizi F, et al. Therapeutic manipulation of peroxynitrite attenuates the development of opiate-induced antinociceptive tolerance in mice. J Clin Invest. 2007;117(11):3530-3539.

Zhao J, Xin X, Xie GX, Palmer PP, Huang YG. Molecular and cellular mechanisms of the age-dependency of opioid analgesia and tolerance. Mol Pain. 2012;8:38-43.

Bujalska-Zadrozny M, Duda K. Additive effect of combined application of magnesium and MK-801 on analgesic action of morphine. Pharmacology. 2014;93(3-4):113-119.

Lee J, Chan SL, Lu C, Lane MA, Mattson MP. Phenformin suppresses calcium responses to glutamate and protects hippocampal neurons against excitotoxicity. Exp Neurol. 2002;175(1):161-167.

Dambisya YM, Lee TL. Role of nitric oxide in the induction and expression of morphine tolerance and dependence in mice. Br J Pharmacol. 1996;117(5):914-918.

Cao JL, Ding HL, He JH, Zhang LC, Duan SM, Zeng YM. The spinal nitric oxide involved in the inhibitory effect of midazolam on morphine-induced analgesia tolerance. Pharmacol Biochem Behav. 2005;80(3):493-503.

Machelska H, Ziólkowska B, Mika J, Przewlocka B, Przewlocki R. Chronic morphine increases biosynthesis of nitric oxide synthase in the rat spinal cord. Neuroreport. 1997;8(12):2743-2747.

Chung MM, Nicol CJ, Cheng YC, Lin KH, Chen YL, Pei D, et al. Metformin activation of AMPK suppresses AGE-induced inflammatory response in hNSCs. Exp Cell Res. 2017;352(1):75-83.

Garg G, Singh S, Singh AK, and Rizvi SI. Antiaging effect of metformin on brain in naturally aged and accelerated senescence model of rat. Rejuvenation Res. 2017;20(3):173-182.

Ndengele MM, Cuzzocrea S, Masini E, Vinci MC, Esposito E, Muscoli C, et al. Spinal ceramide modulates the development of morphine antinociceptive tolerance via peroxynitrite-mediated nitroxidative stress and neuroimmune activation. J Pharmacol Exp Ther. 2009;329(1):64-75.

Salvemini D, Little JW, Doyle T, Neumann WL. Roles of reactive oxygen and nitrogen species in pain. Free Radic Biol Med. 2011;51(5):951-966.

Mori T, Ito S, Matsubayashi K, Sawaguchi T. Comparison of nitric oxide synthase inhibitors, phospholipase A2 inhibitor and free radical scavengers as attenuators of opioid withdrawal syndrome. Behav Pharmacol. 2007;18(8):725-729.

Ozmen I, Naziroglu M, Alici HA, Sahin F, Cengiz M, Eren I. Spinal morphine administration reduces the fatty acid contents in spinal cord and brain by increasing oxidative stress. Neurochem Res. 2007;32(1):19-25.

Muscoli C, Cuzzocrea S, Ndengele MM, Mollace V, Porreca F, Fabrizi F, et al. Therapeutic manipulation of peroxynitrite attenuates the development of opiate-induced antinociceptive tolerance in mice. J Clin Invest. 2007;117(11):3530-3539.

Hou X, Song J, Li XN, Zhang L, Wang X, Chen L, et al. Metformin reduces intracellular reactive oxygen species levels by upregulating expression of the antioxidant thioredoxin via the AMPK-FOXO3 pathway. Biochem Biophys Res Commun. 2010;396(2):199-205.

Faure P, Rossini E, Wiernsperger N, Richard MJ, Favier A, Halimi S. An insulin sensitizer improves the free radical defense system potential and insulin sensitivity in high fructose-fed rats. Diabetes. 1999;48(2):353-357.


Refbacks

  • There are currently no refbacks.


Creative Commons LicenseThis work is licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International License which allows users to read, copy, distribute and make derivative works for non-commercial purposes from the material, as long as the author of the original work is cited properly.