Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

General Research Article

A New Look at an Old Drug: Cumulative Effects of Low Ribavirin Doses in Amphetamine-Sensitized Rats

Author(s): Branka Petković*, Srđan Kesić, Slavica Ristić, Željko Pavković, Jelena Podgorac, Gordana Stojadinović and Vesna Pešić

Volume 26, Issue 31, 2020

Page: [3884 - 3894] Pages: 11

DOI: 10.2174/1381612826666200326125821

open access plus

Abstract

Background: Psychotic states related to psychostimulant misuse in patients with hepatitis C virus infection may complicate acceptance and reaction to antiviral treatment. This observation equally applies to the widely used ribavirin therapy.

Objective: We examined psychomotor and body weight gain responses to low ribavirin doses after cessation of intermittent amphetamine treatment in adult rats to assess its role in neurobehavioral outcome during psychostimulant withdrawal.

Method: The model of amphetamine-induced (1.5 mg/kg/day, i.p., 7 consecutive days) motor sensitization and affected body weight gain was established in adult male Wistar rats. Then, additional cohort of amphetaminesensitized rats was subjected to saline (0.9% NaCl; 1 mL/kg/day; i.p.) or ribavirin (10, 20 and 30 mg/kg/day, i.p.) treatment for 7 consecutive days. Animals’ motor activity in a novel environment was monitored after the 1st and the 7th saline/ribavirin injection. Body weight gain was calculated as appropriate. Determination and quantification of ribavirin in the brain tissue were performed also.

Results: The 1st application of ribavirin to amphetamine-sensitized rats affected/decreased their novelty-induced motor activity only at a dose of 30 mg/kg. After the 7th application, ribavirin 30 mg/kg/day still decreased, while 10 and 20 mg/kg/day increased novelty-induced motor activity. These behavioral effects coincided with the time required to reach maximum ribavirin concentration in the brain. Body weight gain during withdrawal was not influenced by any of the doses tested.

Conclusion: Ribavirin displays central effects that in repeated treatment, depending on the applied dose, could significantly influence psychomotor response but not body weight gain during psychostimulant/amphetamine withdrawal.

Keywords: ribavirin, amphetamine, behavioral sensitization, motor activity, environmental novelty, body weight, rats.

[1]
Grassi A, Ballardini G. Hepatitis C in injection drug users: It is time to treat. World J Gastroenterol 2017; 23(20): 3569-71.
[http://dx.doi.org/10.3748/wjg.v23.i20.3569] [PMID: 28611509]
[2]
Zając M, Muszalska I, Sobczak A, et al. Hepatitis C - New drugs and treatment prospects. Eur J Med Chem 2019; 165: 225-49.
[http://dx.doi.org/10.1016/j.ejmech.2019.01.025] [PMID: 30685524]
[3]
Malaguarnera M, Laurino A, Di Fazio I, et al. Neuropsychiatric effects and type of IFN-alpha in chronic hepatitis C. J Interferon Cytokine Res 2001; 21(5): 273-8.
[http://dx.doi.org/10.1089/107999001300177457] [PMID: 11429157]
[4]
de Knegt RJ, Bezemer G, Van Gool AR, et al. Randomised clinical trial: escitalopram for the prevention of psychiatric adverse events during treatment with peginterferon-alfa-2a and ribavirin for chronic hepatitis C. Aliment Pharmacol Ther 2011; 34(11-12): 1306-17.
[http://dx.doi.org/10.1111/j.1365-2036.2011.04867.x] [PMID: 21999489]
[5]
Fioravante M, Alegre SM, Marin DM, Lorena SL, Pereira TS, Soares EC. Weight loss and resting energy expenditure in patients with chronic hepatitis C before and during standard treatment. Nutrition 2012; 28(6): 630-4.
[http://dx.doi.org/10.1016/j.nut.2011.08.010] [PMID: 22196981]
[6]
Cattie JE, Letendre SL, Woods SP, et al. Persistent neurocognitive decline in a clinic sample of hepatitis C virus-infected persons receiving interferon and ribavirin treatment. J Neurovirol 2014; 20(6): 561-70.
[http://dx.doi.org/10.1007/s13365-014-0265-3] [PMID: 25326107]
[7]
Mahajan S, Avasthi A, Grover S, Chawla YK. Role of baseline depressive symptoms in the development of depressive episode in patients receiving antiviral therapy for hepatitis C infection. J Psychosom Res 2014; 77(2): 109-15.
[http://dx.doi.org/10.1016/j.jpsychores.2014.05.008] [PMID: 25077851]
[8]
Robinson TE, Becker JB. Enduring changes in brain and behavior produced by chronic amphetamine administration: a review and evaluation of animal models of amphetamine psychosis. Brain Res 1986; 396(2): 157-98.
[http://dx.doi.org/10.1016/0165-0173(86)90002-0] [PMID: 3527341]
[9]
Steketee JD, Kalivas PW. Drug wanting: behavioral sensitization and relapse to drug-seeking behavior. Pharmacol Rev 2011; 63(2): 348-65.
[http://dx.doi.org/10.1124/pr.109.001933] [PMID: 21490129]
[10]
Wolgin DL. Amphetamine stereotypy, the basal ganglia, and the “selection problem”. Behav Brain Res 2012; 231(2): 297-308.
[http://dx.doi.org/10.1016/j.bbr.2011.11.003] [PMID: 22101067]
[11]
Kelley AE, Berridge KC. The neuroscience of natural rewards: relevance to addictive drugs. J Neurosci 2002; 22(9): 3306-11.
[http://dx.doi.org/10.1523/JNEUROSCI.22-09-03306.2002] [PMID: 11978804]
[12]
Robinson TE, Kolb B. Structural plasticity associated with exposure to drugs of abuse. Neuropharmacology 2004; 47(Suppl. 1): 33-46.
[http://dx.doi.org/10.1016/j.neuropharm.2004.06.025] [PMID: 15464124]
[13]
Cabib S. The neurobiology of stereotypy II: The role of stressStereotypic animal behaviour: Fundamentals and applications to welfare. CAB International Oxford 2006; pp. 227-52.
[http://dx.doi.org/10.1079/9780851990040.0227]
[14]
Tanimura Y, Ogoegbunam FC, Lewis MH. Amphetamine-induced sensitization and spontaneous stereotypy in deer mice. Pharmacol Biochem Behav 2009; 92(4): 670-5.
[http://dx.doi.org/10.1016/j.pbb.2009.03.006] [PMID: 19324069]
[15]
Janać B, Pešić V, Peković S, Rakić L, Stojiljković M. Different effects of adenosine A1 agonist ribavirin on amphetamine-induced total locomotor and stereotypic activities in rats. Ann N Y Acad Sci 2005; 1048: 396-9.
[http://dx.doi.org/10.1196/annals.1342.048] [PMID: 16154961]
[16]
Janać B, Pešić V, Peković S, Rakić L, Stojiljković M. The time-course of ribavirin-provoked changes of basal and AMPH-induced motor activities in rats. Exp Brain Res 2005; 165(3): 402-6.
[http://dx.doi.org/10.1007/s00221-005-2311-0] [PMID: 15883801]
[17]
Peković S, Filipović R, Subasić S, et al. Downregulation of glial scarring after brain injury: the effect of purine nucleoside analogue ribavirin. Ann N Y Acad Sci 2005; 1048: 296-310.
[http://dx.doi.org/10.1196/annals.1342.027] [PMID: 16154942]
[18]
Stojkov D, Lavrnja I, Pekovic S, et al. Therapeutic effects of combined treatment with ribavirin and tiazofurin on experimental autoimmune encephalomyelitis development: clinical and histopathological evaluation. J Neurol Sci 2008; 267(1-2): 76-85.
[http://dx.doi.org/10.1016/j.jns.2007.10.010] [PMID: 17996253]
[19]
Lavrnja I, Savic D, Bjelobaba I, et al. The effect of ribavirin on reactive astrogliosis in experimental autoimmune encephalomyelitis. J Pharmacol Sci 2012; 119(3): 221-32.
[http://dx.doi.org/10.1254/jphs.12004FP] [PMID: 22785017]
[20]
Petković B, Stojadinović G, Kesić S, et al. Psychomotor activity and body weight gain after exposure to low ribavirin doses in rats: Role of treatment duration. Arch Biol Sci 2019; 71: 357-68.
[http://dx.doi.org/10.2298/ABS190205018P]
[21]
Franchetti P, Cappellacci L, Grifantini M, Senatore G, Martini C, Lucacchini A. Tiazofurin analogues as selective agonists of A1 adenosine receptors. Res Commun Mol Pathol Pharmacol 1995; 87: 103-5.
[22]
Ballarin M, Reiriz J, Ambrosio S, Mahy N. Effect of locally infused 2-chloroadenosine, an A1 receptor agonist, on spontaneous and evoked dopamine release in rat neostriatum. Neurosci Lett 1995; 185(1): 29-32.
[http://dx.doi.org/10.1016/0304-3940(94)11217-7] [PMID: 7731548]
[23]
Turgeon SM, Pollack AE, Schusheim L, Fink JS. Effects of selective adenosine A1 and A2a agonists on amphetamine-induced locomotion and c-Fos in striatum and nucleus accumbens. Brain Res 1996; 707(1): 75-80.
[http://dx.doi.org/10.1016/0006-8993(95)01223-0] [PMID: 8866715]
[24]
Golembiowska K, Zylewska A. Agonists of A1 and A2A adenosine receptors attenuate methamphetamine-induced overflow of dopamine in rat striatum. Brain Res 1998; 806(2): 202-9.
[http://dx.doi.org/10.1016/S0006-8993(98)00743-4] [PMID: 9739141]
[25]
Hobson BD, Merritt KE, Bachtell RK. Stimulation of adenosine receptors in the nucleus accumbens reverses the expression of cocaine sensitization and cross-sensitization to dopamine D2 receptors in rats. Neuropharmacology 2012; 63(6): 1172-81.
[http://dx.doi.org/10.1016/j.neuropharm.2012.06.038] [PMID: 22749927]
[26]
Song WJ, Tkatch T, Surmeier DJ. Adenosine receptor expression and modulation of Ca(2+) channels in rat striatal cholinergic interneurons. J Neurophysiol 2000; 83(1): 322-32.
[http://dx.doi.org/10.1152/jn.2000.83.1.322] [PMID: 10634875]
[27]
Yoon KW, Rothman SM. Adenosine inhibits excitatory but not inhibitory synaptic transmission in the hippocampus. J Neurosci 1991; 11(5): 1375-80.
[http://dx.doi.org/10.1523/JNEUROSCI.11-05-01375.1991] [PMID: 1851219]
[28]
Qi G, van Aerde K, Abel T, Feldmeyer D. Adenosine differentially modulates synaptic transmission of excitatory and inhibitory microcircuits in layer 4 of rat barrel cortex. Cereb Cortex 2017; 27(9): 4411-22.
[http://dx.doi.org/10.1093/cercor/bhw243] [PMID: 27522071]
[29]
Fuxe K, Ferré S, Zoli M, Agnati LF. Integrated events in central dopamine transmission as analyzed at multiple levels. Evidence for intramembrane adenosine A2A/dopamine D2 and adenosine A1/dopamine D1 receptor interactions in the basal ganglia. Brain Res Brain Res Rev 1998; 26(2-3): 258-73.
[http://dx.doi.org/10.1016/S0165-0173(97)00049-0] [PMID: 9651540]
[30]
Franco R, Lluis C, Canela EI, et al. Receptor-receptor interactions involving adenosine A1 or dopamine D1 receptors and accessory proteins. J Neural Transm (Vienna) 2007; 114(1): 93-104.
[http://dx.doi.org/10.1007/s00702-006-0566-7] [PMID: 17024327]
[31]
Cechova S, Elsobky AM, Venton BJ. A1 receptors self-regulate adenosine release in the striatum: evidence of autoreceptor characteristics. Neuroscience 2010; 171(4): 1006-15.
[http://dx.doi.org/10.1016/j.neuroscience.2010.09.063] [PMID: 20933584]
[32]
Ciruela F, Gómez-Soler M, Guidolin D, et al. Adenosine receptor containing oligomers: their role in the control of dopamine and glutamate neurotransmission in the brain. Biochim Biophys Acta 2011; 1808(5): 1245-55.
[http://dx.doi.org/10.1016/j.bbamem.2011.02.007] [PMID: 21316336]
[33]
Franco R, Ferré S, Agnati L, et al. Evidence for adenosine/dopamine receptor interactions: indications for heteromerization. Neuropsychopharmacology 2000; 23(4)(Suppl.): S50-9.
[http://dx.doi.org/10.1016/S0893-133X(00)00144-5] [PMID: 11008067]
[34]
Fuxe K, Ferré S, Genedani S, Franco R, Agnati LF. Adenosine receptor-dopamine receptor interactions in the basal ganglia and their relevance for brain function. Physiol Behav 2007; 92(1-2): 210-7.
[http://dx.doi.org/10.1016/j.physbeh.2007.05.034] [PMID: 17572452]
[35]
Vezina P. D1 dopamine receptor activation is necessary for the induction of sensitization by amphetamine in the ventral tegmental area. J Neurosci 1996; 16(7): 2411-20.
[http://dx.doi.org/10.1523/JNEUROSCI.16-07-02411.1996] [PMID: 8601820]
[36]
Magendzo K, Bustos G. Expression of amphetamine-induced behavioral sensitization after short- and long-term withdrawal periods: participation of mu- and delta-opioid receptors. Neuropsychopharmacology 2003; 28(3): 468-77.
[http://dx.doi.org/10.1038/sj.npp.1300063] [PMID: 12629526]
[37]
Nair AB, Jacob S. A simple practice guide for dose conversion between animals and human. J Basic Clin Pharm 2016; 7(2): 27-31.
[http://dx.doi.org/10.4103/0976-0105.177703] [PMID: 27057123]
[38]
Janać B, Pešić V, Veskov R, et al. The effects of tiazofurin on basal and amphetamine-induced motor activity in rats. Pharmacol Biochem Behav 2004; 77(3): 575-82.
[http://dx.doi.org/10.1016/j.pbb.2003.12.025] [PMID: 15006469]
[39]
Berman SM, Kuczenski R, McCracken JT, London ED. Potential adverse effects of amphetamine treatment on brain and behavior: a review. Mol Psychiatry 2009; 14(2): 123-42.
[http://dx.doi.org/10.1038/mp.2008.90] [PMID: 18698321]
[40]
Petković B, Kesić S, Pešić V. Critical view on the usage of ribavirin in already existing psychostimulant-use disorder. Curr Pharm Des 2020.
[http://dx.doi.org/10.2174/1381612826666200115094642] [PMID: 31939725]
[41]
Hooks MS, Jones GH, Neill DB, Justice JB Jr. Individual differences in amphetamine sensitization: dose-dependent effects. Pharmacol Biochem Behav 1992; 41(1): 203-10.
[http://dx.doi.org/10.1016/0091-3057(92)90083-R] [PMID: 1539070]
[42]
Strakowski SM, Sax KW, Rosenberg HL, DelBello MP, Adler CM. Human response to repeated low-dose d-amphetamine: evidence for behavioral enhancement and tolerance. Neuropsychopharmacology 2001; 25(4): 548-54.
[http://dx.doi.org/10.1016/S0893-133X(01)00253-6] [PMID: 11557168]
[43]
Rognli EB, Bramness JG. Understanding the relationship between amphetamines and psychosis. Curr Addict Rep 2015; 2: 285-92.
[http://dx.doi.org/10.1007/s40429-015-0077-4]
[44]
Solanto MV. Neuropsychopharmacological mechanisms of stimulant drug action in attention-deficit hyperactivity disorder: a review and integration. Behav Brain Res 1998; 94(1): 127-52.
[http://dx.doi.org/10.1016/S0166-4328(97)00175-7] [PMID: 9708845]
[45]
Crombag HS, Badiani A, Chan J, Dell’Orco J, Dineen SP, Robinson TE. The ability of environmental context to facilitate psychomotor sensitization to amphetamine can be dissociated from its effect on acute drug responsiveness and on conditioned responding. Neuropsychopharmacology 2001; 24(6): 680-90.
[http://dx.doi.org/10.1016/S0893-133X(00)00238-4] [PMID: 11331148]
[46]
Fraioli S, Cioli I, Nencini P. Amphetamine reinstates polydipsia induced by chronic exposure to quinpirole, a dopaminergic D2 agonist, in rats. Behav Brain Res 1997; 89(1-2): 199-215.
[http://dx.doi.org/10.1016/S0166-4328(97)00063-6] [PMID: 9475627]
[47]
Wolgin DL, Thompson GB, Oslan IA. Tolerance to amphetamine: contingent suppression of stereotypy mediates recovery of feeding. Behav Neurosci 1987; 101(2): 264-71.
[http://dx.doi.org/10.1037/0735-7044.101.2.264] [PMID: 3580129]
[48]
Olsson IAS, Westlund K. More than numbers matter: the effect of social factors on behaviour and welfare of laboratory rodents and non-human primates. Appl Anim Behav Sci 2007; 103: 229-54.
[http://dx.doi.org/10.1016/j.applanim.2006.05.022]
[49]
Rowland N, Antelman SM, Kocan D. Elevated water intake in rats treated chronically with amphetamine: drinking in excess of need? Appetite 1981; 2(1): 51-66.
[http://dx.doi.org/10.1016/S0195-6663(81)80036-0] [PMID: 7337439]
[50]
Camanni S, Nencini P. Physiological and environmental aspects of drinking stimulated by chronic exposure to amphetamine in rats. Gen Pharmacol 1994; 25(1): 7-13.
[http://dx.doi.org/10.1016/0306-3623(94)90003-5] [PMID: 8026715]
[51]
Evans KR, Vaccarino FJ. Amphetamine- and morphine-induced feeding: evidence for involvement of reward mechanisms. Neurosci Biobehav Rev 1990; 14(1): 9-22.
[http://dx.doi.org/10.1016/S0149-7634(05)80156-3] [PMID: 2325945]
[52]
Biederman J, Faraone SV, Monuteaux MC, Plunkett EA, Gifford J, Spencer T. Growth deficits and attention-deficit/hyperactivity disorder revisited: impact of gender, development, and treatment. Pediatrics 2003; 111(5 Pt 1): 1010-6.
[http://dx.doi.org/10.1542/peds.111.5.1010] [PMID: 12728081]
[53]
Rivkees SA, Price SL, Zhou FC. Immunohistochemical detection of A1 adenosine receptors in rat brain with emphasis on localization in the hippocampal formation, cerebral cortex, cerebellum, and basal ganglia. Brain Res 1995; 677(2): 193-203.
[http://dx.doi.org/10.1016/0006-8993(95)00062-U] [PMID: 7552243]
[54]
Seyam MS, Freshwater DA, O’Donnell K, Mutimer DJ. Weight loss during pegylated interferon and ribavirin treatment of chronic hepatitis C*. J Viral Hepat 2005; 12(5): 531-5.
[http://dx.doi.org/10.1111/j.1365-2893.2005.00637.x] [PMID: 16108770]
[55]
Lin Z, Li Y, Gong G, et al. Restriction of H1N1 influenza virus infection by selenium nanoparticles loaded with ribavirin via resisting caspase-3 apoptotic pathway. Int J Nanomedicine 2018; 13: 5787-97.
[http://dx.doi.org/10.2147/IJN.S177658] [PMID: 30310281]
[56]
Festing MF, Altman DG. Guidelines for the design and statistical analysis of experiments using laboratory animals. ILAR J 2002; 43(4): 244-58.
[http://dx.doi.org/10.1093/ilar.43.4.244] [PMID: 12391400]
[57]
Kitchen CM. Nonparametric vs parametric tests of location in biomedical research. Am J Ophthalmol 2009; 147(4): 571-2.
[http://dx.doi.org/10.1016/j.ajo.2008.06.031] [PMID: 19327444]
[58]
Stojanović M, Andjelković-Apostolović M, Milošević Z, Ignjatović A. Parametric versus nonparametric tests in biomedical research. Acta Medica Medianae 2018; 57: 75-80.
[http://dx.doi.org/10.5633/amm.2018.0212]
[59]
Baker M. Statisticians issue warning over misuse of P values. Nature 2016; 531(7593): 151.
[http://dx.doi.org/10.1038/nature.2016.19503] [PMID: 26961635]

© 2024 Bentham Science Publishers | Privacy Policy