The impact of ademetionine and ipidacrine/phenibut on the NCAM distribution and behavior in the rat model of drug-induced liver injury

Authors

DOI:

https://doi.org/10.15584/ejcem.2020.3.1

Keywords:

ademetionine, brain, ipidacrine/phenibut, isoniazid, liver disease, NCAM, rifampicin

Abstract

Introduction. Recently, more attention is being paid to the drug-induced liver injury (DILI) as a consequence of the tuberculos is treatment and the need for new medicine is emphasized. The use of isoniazid and rifampicin has a potentiating effect, which increases the risk of substancial liver damage. In turn, systemic accumulation of toxic metabolites leads to negative changes in various organs, including the brain. It causes an imbalance in biochemical and neurophysiological processes in the brain, ultimately giving the onset to the development of hepatic encephalopathy.

Aim. The effects of rifampicin and isoniazid on the central nervous system have not been studied before and we aimed to evaluate the impact these two substances have on the neuronal cell adhesion molecules (NCAM) distribution and animal behavior in the rat model of DILI.

Material and methods. The 24 male Wistar rats, weighing 180-220 g were used for the experiment and divided to the groups (n=6): 1 – control; 2 – rats with experimental DILI; 3 – rats with DILI plus the intravenous infusion of S-adenosyl-L-methionine at a dose of 35 mg/kg; 4 – rats with DILI plus a fixed combination of ipidacrine hydrochloride at a dose 1 mg/kg body weight and phenibut at a dose 60 mg/kg body weight daily for the last 14 days of the experiment. All experimental procedures were carried out in the accordance with the principles outlined in the current Guide to the Care and Use of Experimental Animals. The locomotor and research activities were studied in the open field test. The activity of aspartate aminotransferase (AST, ЕС 2.6.1.1) and alanine aminotransferase (ALT, ЕС 2.6.1.2) in the serum of rats were tested to confirm the liver damage. The quantitative analyses of soluble and membrane forms of NCAM were performed with ELISA. The ANOVA followed by a Tukey post-hoc test was used to assess statistical differences between groups.

Results. Our investigation in the open field test revealed a significant decrease in the locomotor and research activity of rats after 28 days of rifampicin and isoniazid administration. The recovery of investigated parameters was observed in groups of animals treated with ademetionine (AD group) or combination of ipidacrine and phenibut (IP/PB group). We also observed that changes in rats’ behavior were consistent with alterations of the NCAM levels in the thalamus and hippocampus. Thus, the level of membrane NCAM was significantly decreased under DILI in both investigated brain regions (thalamus and hippocampus), while both AD and IP/PB treatments restored membrane NCAM levels towards those observed in the control group at least in the hippocampus.

Conclusion. Obtained data suggests that both ademetionine and combinated drug containing ipidacrine and phenibut possesses neuroprotective properties and could prevent the decline in synaptic plasticity under antitubercular therapy.

Downloads

Download data is not yet available.

References

Ghabril M, Chalasani N, Björnsson E. Drug-induced liver injury: a clinical update. Curr Opin Gastroenterol. 2010;26(3):222-226.

Björnsson ES. Hepatotoxicity by drugs: the most common implicated agents. Int J Mol Sci. 2016;17(2):224 .

Hakim Z, Waheed A, Bakhtiar S, Hasan N, Hakim B. Potentiating effect of rifampicin on methimazole induced hepatotoxicity in mice. Pak J Pharm Sci. 2018;31(6):2373-2377.

Metushi I, Uetrecht J, Phillips E. Mechanism of isoniazid‐induced hepatotoxicity: then and now. Br J Clin Pharmacol. 2016;81(6):1030-1036.

Ridola L, Nardelli S, Gioia S, Riggio O. Quality of life in patients with minimal hepatic encephalopathy. World J Gastroenterol. 2018;24(48):5446-5453.

Parekh PJ, Balart LA. Ammonia and its role in the pathogenesis of hepatic encephalopathy. Clin Liver Dis. 2015;19(3):529-537.

Lemberg A, Fernández MA. Hepatic encephalopathy, ammonia, glutamate, glutamine and oxidative stress. Ann Hepatol. 2009;8(2):95-102.

Palomero-Gallagher N, Zilles K. Neurotransmitter receptor alterations in hepatic encephalopathy: a review. Arch Biochem Biophys. 2013;536(2):109-121.

Ramachandran A, Duan L, Akakpo JY, Jaeschke H. Mitochondrial dysfunction as a mechanism of drug-induced hepatotoxicity: current understanding and future perspectives. J Clin Transl Res. 2018;4(1):75-100.

Heidari R. Brain mitochondria as potential therapeutic targets for managing hepatic encephalopathy. Life Sci. 2019;218:65-80.

Ho N, Liauw JA, Blaeser F, et al. Impaired Synaptic Plasticity and cAMP Response Element-Binding Protein Activation in Ca2+/Calmodulin-Dependent Protein Kinase Type IV/Gr-Deficient Mice. J Neurosci. 2000;20(17):6459-6472.

Llansola M, Montoliu C, Agusti A, et al. Interplay between glutamatergic and GABAergic neurotransmission alterations in cognitive and motor impairment in minimal hepatic encephalopathy. Neurochem Int. 2015;88:15-19.

Gonzalez-Usano A, Cauli O, Agusti A, Felipo V. Pregnenolone sulfate restores the glutamate-nitric-oxide-cGMP pathway and extracellular GABA in cerebellum and learning and motor coordination in hyperammonemic rats. ACS Chem Neurosci. 2014;5(2):100-105.

Chatterjee M, Schild D, Teunissen CE. Contactins in the central nervous system: role in health and disease. Neural Regen Res. 2019;14(2):206-216.

Mah W, Ko J, Nam J, Han K, Chung WS, Kim E.Selected SALM (Synaptic Adhesion-Like Molecule) Family Proteins Regulate Synapse Formation. J Neurosci. 2010;30(16): 5559-5568.

Lie E, Li Y, Kim R, Kim E. SALM/Lrfn Family Synaptic Adhesion Molecules. Front Mol Neurosci. 2018;11:105.

Bisaz R, Conboy L, Sandi C. Learning under stress: A role for the neural cell adhesion molecule NCAM. Neurobiol Learn Mem. 2009;91(4):333-342.

Yue J, Peng R, Chen J, Liu Y, Dong G. Effects of rifampin on CYP2E1-dependent hepatotoxicity of isoniazid in rats. Pharmacol Res. 2009;59(2):112-119.

Aonurm-Helm A, Jaako K, Jürgenson M, Zharkovsky A. Pharmacological approach for targeting dysfunctional brain plasticity: Focus on neural cell adhesion molecule (NCAM). Pharmacol Res. 2016;113(Pt B):731-738.

Li Q, Cui J, Fang C, Zhang X, Li L.S-adenosylmethionine Administration Attenuates Low Brain-Derived Neurotrophic Factor Expression Induced by Chronic Cerebrovascular Hypoperfusion or Beta Amyloid Treatment. Neurosci Bull. 2016;32(2):153-161.

Li Q, Cui J, Fang C, Liu M, Min G, Li L. S-adenosylmethionine attenuates oxidative stress and neuroinflammation induced by amyloid-β through modulation of glutathione metabolism. J Alzheimers Dis. 2017;58(2):549-558.

Cavallaro RA, Fuso A, Nicolia V, Scarpa S. S-adenosylmethionine prevents oxidative stress and modulates glutathione metabolism in TgCRND8 mice fed a B-vitamin deficient diet. J Alzheimers Dis. 2010;20(4):997-1002.

Lu S, Mato J. S-adenosylmethionine in liver health, injury, and cancer. Physiol Rev. 2012;92(4):1515-1542.

Tyurenkov IN, Borodkina LE, Bagmetova VV, Berestovitskaya VM, Vasil'eva OS. Comparison of nootropic and neuroprotective features of aryl-substituted analogs of gamma-aminobutyric acid. Bull Exp Biol Med. 2016;160(4):465-469.

Dambrova M, Zvejniece L, Liepinsh E, et al. Comparative pharmacological activity of optical isomers of phenibut. Eur J Pharmacol. 2008;583(1):128-134.

Terunuma M. Diversity of structure and function of GABAB receptors: a complexity of GABAB-mediated signaling. Proc Jpn Acad Ser B Phys Biol Sci. 2018;94(10):390-411.

Stodůlka J. Ethanol and physostigmine effects on open field behavior in Wistar rats. Acta Univ Palacki Olomuc Fac Med. 1991;131:39-81.

Karl T, Pabst R, von Hörsten S. Behavioral phenotyping of mice in pharmacological and toxicological research. Exp Toxicol Pathol. 2003;55(1):69-83.

Young D. Effects on clinical laboratory tests: drugs, disease, herbs and natural products», American association for clinical chemistry. 2014, John Wiley & Sons, Inc., Hoboken, New Jersey, USA.

Mand B, Sikaris KA. The De Ritis Ratio: The Test of Time. Clin Biochem Rev. 2013;34(3):117-130.

Almutairi MM, Gong C, Xu YG, Chang Y, Shi H. Factors controlling permeability of the blood-brain barrier. Cell Mol Life Sci. 2016;73(1):57-77.

Awodele O, Akintonwa A, Osunkalu VO, Coker HA. Modulatory activity of antioxidants against the toxicity of Rifampicin in vivo. Rev Inst Med Trop Sao Paulo. 2010;52(1):43-46.

Lee SY, Ko KS. Effects of S-Adenosylmethionine and Its Combinations With Taurine and/or Betaine on Glutathione Homeostasis in Ethanol-induced Acute Hepatotoxicity. J Cancer Prev. 2016;21(3):164-172.

Togashi H, Sakisaka T, Takai Y. Cell adhesion molecules in the central nervous system. Cell Adh Migr. 2009;3(1):29-35.

Mao X, Schwend T, Conrad GW. Expression and localization of neural cell adhesion molecule and polysialic cid during chick corneal development. Invest. Ophthalmol Vis Sci 2012;53(3):1234-1243.

Hinkle CL, Diestel S, Lieberman J, Maness PF. Metalloprotease-induced ectodoma in shedding of neural cell adhesion molecule (NCAM). J Neurobiol. 2006;66:1378-1395.

Abbott NJ. Blood-brain barrier structure and function and the challenges for CNS drug delivery. J Inherit Metab Dis. 2013;36(3):437-449.

Shobo A, Bratkowska D, Baijnath S, et al. Visualization of Time-Dependent Distribution of Rifampicin in Rat Brain Using MALDI MSI and Quantitative LCMS/MS. Assay Drug Dev Technol. 2015;13(5):277-284.

DeMarco VP, Ordonez AA, Klunk M, et al. Determination of [11C]rifampin pharmacokinetics within Mycobacterium tuberculosis-infected mice by using dynamic positron emission tomography bioimaging. Antimicrob Agents Chemother. 2015;59(9):5768-5774.

Aonurm-Helm A, Jaako K, Jürgenson M, Zharkovsky A. Pharmacological approach for targeting dysfunctional brain plasticity: Focus on neural cell adhesion molecule (NCAM). Pharmacol Res. 2016;113(Pt B):731-738.

Pogotova GA. An influence of ademetionine on an energy metabolism, prooxidant-antioxidant system in liver, myocardium, and brain of rats in the presence of the dichloroethane hepatitis. Lik Sprava. 2015;(5-6):120-125.

Washbourne P, Dityatev A, Scheiffele P, Biederer T, Weiner JA, Christopherson KS, El-Husseini A. Cell adhesion molecules in synapse formation. J Neurosci. 2004;24(42):9244-9249.

Hagiyama M, Ichiyanagi N, Kimura KB, Murakami Y, Ito A. Expression of a soluble isoform of cell adhesion molecule 1 in the brain and its involvement in directional neurite outgrowth. Am J Pathol. 2009;174(6):2278-2289.

Vavers E, Zvejniece L, Svalbe B, et al. The neuroprotective effects of R-phenibut after focal cerebral ischemia. Pharmacol Res. 2016;113(Pt B):796-801.

Downloads

Published

2020-09-30

How to Cite

Muraviova, D., Kharchenko, Y., Pierzynowska, K., Pierzynowski, S. G., Wolinski, J., Dyomshyna, O., Zhyliuk, V., & Ushakova, G. (2020). The impact of ademetionine and ipidacrine/phenibut on the NCAM distribution and behavior in the rat model of drug-induced liver injury. European Journal of Clinical and Experimental Medicine, 18(3), 155–164. https://doi.org/10.15584/ejcem.2020.3.1

Issue

Section

ORIGINAL PAPERS