The Neuroprotective Effect of α-Lipoic Acid and/or Metformin against the Behavioral and Neurochemical Changes Induced by Hypothyroidism in Rat

Khadrawy Y.A.a· Khoder N.M.a· Sawie H.G.a· Sharada H.M.b· Hosny E.N.a· Abdulla M.S.b

Author affiliations

aMedical Physiology Department, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
bChemistry Department, Faculty of Science, Helwan University, Helwan, Egypt

Log in to MyKarger to check if you already have access to this content.

Buy FullText & PDF Unlimited re-access via MyKarger Unrestricted printing, no saving restrictions for personal use
read more

CHF 38.00 *
EUR 35.00 *
USD 39.00 *

Select

KAB

Buy a Karger Article Bundle (KAB) and profit from a discount!

If you would like to redeem your KAB credit, please log in.

Save over 20% compared to the individual article price.

Learn more

Rent/Cloud Rent for 48h to view Buy Cloud Access for unlimited viewing via different devices Synchronizing in the ReadCube Cloud Printing and saving restrictions apply Rental: USD 8.50
Cloud: USD 20.00

Select

Subscribe Access to all articles of the subscribed year(s) guaranteed for 5 years Unlimited re-access via Subscriber Login or MyKarger Unrestricted printing, no saving restrictions for personal use read more

Subcription rates

Select

* The final prices may differ from the prices shown due to specifics of VAT rules.

Article / Publication Details

First-Page Preview

Abstract of Research Article

Received: December 22, 2021
Accepted: March 11, 2022
Published online: March 30, 2022

Number of Print Pages: 14
Number of Figures: 5
Number of Tables: 2

ISSN: 0028-3835 (Print)
eISSN: 1423-0194 (Online)

For additional information: https://www.karger.com/NEN

Abstract

Objective: The present study evaluates the neuroprotective effect of α-lipoic acid (ALA) and/or metformin (MET) on the behavioral and neurochemical changes induced by hypothyroidism. Methods: Rats were divided into control, rat model of hypothyroidism induced by propylthiouracil, and rat model of hypothyroidism treated with ALA, MET, or their combination. Results: Behaviorally, hypothyroid rats revealed impaired memory and reduced motor activity as indicated from the novel object recognition test and open-field test, respectively. Hypothyroidism induced a significant increase in lipid peroxidation (malondialdehyde [MDA]) and a significant decrease in reduced glutathione (GSH) and nitric oxide (NO) in the cortex and hippocampus. These were associated with a significant increase in tumor necrosis factor-α (TNF-α) and a significant decrease in brain-derived neurotrophic factor (BDNF). Hypothyroidism decreased significantly the levels of serotonin (5-HT), norepinephrine (NE), and dopamine (DA) and reduced the activities of acetylcholinesterase (AchE) and Na+, K+-ATPase in the cortex and hippocampus. Treatment of hypothyroid rats with ALA and/or MET showed an improvement in memory function and motor activity. Moreover, ALA and/or MET prevented the increase in MDA and TNF-α, and the decline in GSH, NO, BDNF, 5-HT, NE, and DA. It also restored AchE and Na+, K+-ATPase activities in the studied brain regions. Conclusion: ALA and/or MET has a potential neuroprotective effect against the adverse behavioral and neurochemical changes induced by hypothyroidism in rats.

© 2022 S. Karger AG, Basel

References Escribano-Serrano J, Mancera-Romero J, Santos-Sánchez V, Payá-Giner C, Méndez-Esteban MI, García-Bonilla A, et al. Prevalence of hypothyroidism in Andalusia, Spain, determined by thyroid hormone comsumption. Rev Esp Salud Publica. 2016;90:e1–e12. Segni M. Disorders of the thyroid gland in infancy, childhood and adolescence. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, editors. South Dartmouth (MA): MDText.com, Inc.; 2000. Blum MR, Wijsman LW, Virgini VS, Bauer DC, den Elzen WP, Jukema JW, et al. Subclinical thyroid dysfunction and depressive symptoms among the elderly: a prospective cohort study. Neuroendocrinology. 2016;103:291–9. Zimmermann MB, Boelaert K. Iodine deficiency and thyroid disorders. Lancet Diabetes Endocrinol. 2015;3(4):286–95. Ahmed OM, El-Gareib AW, El-Bakry AM, Abd El-Tawab SM, Ahmed RG. Thyroid hormones states and brain development interactions. Int J Dev Neurosci. 2008;26(2):147–209. Salazar P, Cisternas P, Martinez M, Inestrosa NC. Hypothyroidism and cognitive disorders during development and adulthood: implications in the central nervous system. Mol Neurobiol. 2019;56:2952–63. Koromilas C, Liapi C, Schulpis KH, Kalafatakis K, Zarros A, Tsakiris S. Structural and functional alterations in the hippocampus due to hypothyroidism. Metab Brain Dis. 2010;25:339–54. Pilhatsch M, Marxen M, Winter C, Smolka MN, Bauer M. Hypothyroidism and mood disorders: integrating novel insights from brain imaging techniques. Thyroid Res. 2011;4 Suppl 1(Suppl 1):S3. Mendes-de-Aguiar CB, Alchini R, Decker H, Alvarez-Silva M, Tasca CI, Trentin AG. Thyroid hormone increases astrocytic glutamate uptake and protects astrocytes and neurons against glutamate toxicity. J Neurosci Res. 2008;86(14):3117–25. Strawn JR, Ekhator NN, D’Souza BB, Geracioti TD. Pituitary-thyroid state correlates with central dopaminergic and serotonergic activity in healthy humans. Neuropsychobiology. 2004;49:84–7. Wiens SC, Trudeau VL. Thyroid hormone and γ-aminobutyric acid (GABA) interactions in neuroendocrine systems. Comp Biochem Physiol. 2006;144:332–44. Smith JW, Evans AT, Costall B, Smythe JW. Thyroid hormones, brain function and cognition: a brief review. Neurosci Biobehav Rev. 2002;26(1):45–60. Mullur R, Liu YY, Brent GA. Thyroid hormone regulation of metabolism. Physiol Rev. 2014;94(2):355–82. Yavuz S, Salgado Nunez Del Prado S, Celi FS. Thyroid hormone action and energy expenditure. J Endocr Soc. 2019;3(7):1345–56. Villanueva I, Alva-Sánchez C, PAcheco-Rosado J. The role of thyroid hormones as inductors of oxidative stress and neurodegeneration. Oxid Med Cell Longev. 2013;2013:218145. Baghcheghi Y, Salmani H, Beheshti F, Hosseini M. Contribution of brain tissue oxidative damage in hypothyroidism-associated learning and memory impairments. Adv Biomed Res. 2017 May 22;6:59. Packer L, Cadenas E. Lipoic acid: energy metabolism and redox regulation of transcription and cell signaling. J Clin Biochem Nutr. 2011;48(1):26–32. Hiller S, DeKroon R, Hamlett ED, Xu L, Osorio C, Robinette J, et al. Alpha-lipoic acid supplementation protects enzymes from damage by nitrosative and oxidative stress. Biochim Biophys Acta. 2016 Jan;1860(1 Pt A):36–45. Bjørklund G, Aaseth J, Crisponi G, Rahman MM, Chirumbolo S. Insights on alpha lipoic and dihydrolipoic acids as promising scavengers of oxidative stress and possible chelators in mercury toxicology. J Inorg Biochem. 2019;195:111–9. Yürük AA, Ayaz A. The health effects of alpha lipoic acid. Hacettepe Üniversitesi Sağlık Bilimleri Fakültesi Dergisi. 2014;1:11–23. Perera J, Tan JH, Jeevathayaparan S, Chakravarthi S, Haleagrahara N. Neuroprotective effects of alpha lipoic acid on haloperidol-induced oxidative stress in the rat brain. Cell Biosci. 2011;1(1):12. Rena G, Hardie DG, Pearson ER. The mechanisms of action of metformin. Diabetologia. 2017;60(9):1577–85. Ahmed S, Mahmood Z, Javed A, Hashmi SN, Zerr I, Zafar S, et al. Effect of metformin on adult hippocampal neurogenesis: comparison with donepezil and links to cognition. J Mol Neurosci. 2017;62(1):88–98. Diaz A, Muñoz-Arenas G, Venegas B, Vázquez-Roque R, Flores G, Guevara J, et al. Metforminium decavanadate (MetfDeca) treatment ameliorates hippocampal neurodegeneration and recognition memory in a metabolic syndrome model. Neurochem Res. 2021 May;46(5):1151–65. Alzoubi KH, Khabour OF, Al-azzam SI, Tashtoush MH, Mhaidat NM. Metformin eased cognitive impairment induced by chronic L-methionine administration: potential role of oxidative stress. Curr Neuropharmacol. 2014;12:186–92. Dehkordi AH, Abbaszadeh A, Mir S, Hasanvand A. Metformin and its anti-inflammatory and anti-oxidative effects; new concepts. J Renal Inj Prev. 2019;8(1):54–61. Ma J, Sarah J, Alzerjawi MH. Effect of propylthiouracil-inducedhypothyroidism on reproductive efficiency of adult male rats. Bas J Vet Res. 2013;12:2. Paseban M, Mohebbati R, Niazmand S, Sathyapalan T, Sahebkar A. Comparison of the neuroprotective effects of aspirin, atorvastatin, captopril and metformin in diabetes mellitus. Biomolecules. 2019;9(4):118. Nalivaeva NN, Belyaev ND, Lewis DI, Pickles AR, Makova NZ, Bagrova DI, et al. Effect of sodium valproate administration on brain neprilysin expression and memory in rats. J Mol Neurosci. 2012;46(3):569–77. Ennaceur A. One-trial object recognition in rats and mice: methodological and theoretical issues. Behav Brain Res. 2010;215(2):244–54. Castillo-Carranza DL, Guerrero-Muñoz MJ, Sengupta U, Hernandez C, Barrett AD, Dineley K, et al. Tau immunotherapy modulates both pathological tau and upstream amyloid pathology in an Alzheimer’s disease mouse model. J Neurosci. 2015;35(12):4857–68. Brown RE, Corey SC, Moore AK. Differences in measures of exploration and fear in MHC-congenic C57BL/6J and B6-H-2K mice. Behav Genet. 1999;29(4):263–71. Ciarlone AE. Further modification of a fluorometric method for analyzing brain amines. Microchem J. 1978;23(1):9–12. Ruiz-Larrea MB, Leal AM, Liza M, Lacort M, de Groot H. Antioxidant effects of estradiol and 2-hydroxyestradiol on iron-induced lipid peroxidation of rat liver microsomes. Steroids. 1994;59:383–8. Moron MS, Depierre JW, Mannervik B. Levels of glutathione, glutathione reductase and glutathione S-transferase activities in rat lung and liver. Biochim Biophys Acta. 1979;582:67–78. Montgomery HAC, Dymock JF. The determination of nitrite in water. Analyst. 1961;86:414–6. Ellman GL, Courtney KD, Andres V Jr, Feather-Stone RM. A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem Pharmacol. 1961;7:88–95. Tsakiris S, Angelogianni P, Schulpis KH, Behrakis P. Protective effect of L-cysteine and glutathione on rat brain Na+, K+-ATPase inhibition induced by free radicals. Z Naturforsch C J Biosci. 2000;55:271–7. Boron WF, Boulpaep EL. Medical physiology. Updated ed. Philadelphia: Elsevier Saunders; 2005. De Las Heras-Gómez I, Medina S, Casas-Pina T, Marín-Soler L, Tomás A, Martínez-Hernández P, et al. Potential applications of lipid peroxidation products – F4-neuroprostanes, F3-neuroprostanesn-6 DPA, F2-dihomo-isoprostanes and F2-isoprostanes – in the evaluation of the allograft function in renal transplantation. Free Radic Biol Med. 2017;104:178–84. Kurutas EB. The importance of antioxidants which play the role in cellular response against oxidative/nitrosative stress: current state. Nutr J. 2016;15(1):71. Cobley JN, Fiorello ML, Bailey DM. 13 reasons why the brain is susceptible to oxidative stress. Redox Biol. 2018 May;15:490–503. Cooke GE, Mullally S, Correia N, O’Mara SM, Gibney J. Hippocampal volume is decreased in adults with hypothyroidism. Thyroid. 2014;24:433–40. Jourd’heuil D, Jourd’heuil FL, Kutchukian PS, Musah RA, Wink DA, Grisham MB. Reaction of superoxide and nitric oxide with peroxynitrite. Implications for peroxynitrite-mediated oxidation reactions in vivo. J Biol Chem. 2001;276(31):28799–805. Blanco S, Hernández R, Franchelli G, Ramos-Álvarez MM, Peinado MÁ. Melatonin influences NO/NOS pathway and reduces oxidative and nitrosative stress in a model of hypoxic-ischemic brain damage. Nitric Oxide. 2017;62:32–43. Serfozo Z, Kiss PB, Kukor Z, Lontay B, Palatka K, Varga V, et al. Thyroid hormones affect the level and activity of nitric oxide synthase in rat cerebral cortex during postnatal development. Neurochem Res. 2008;33(3):569–78. Estrada Sándor P, Komjáti K, Reivich M, Nyáry I. Major role of nitric oxide in the mediation of regional CO2 responsiveness of the cerebral and spinal cord vessels of the cat. J Cereb Blood Flow Metab. 1994;14:49–58. Sinha RA, Pathak A, Mohan V, Bandyopadhyay S, Rastogi L, Godbole MM. Maternal thyroid hormone: a strong repressor of neuronal nitric oxide synthase in rat embryonic neocortex. Endocrinology. 2008;149:4396–401. Baghcheghi Y, Salmani H, Beheshti F, Shafei MN, Sadeghnia HR, Soukhtanloo M, et al. Effects of PPAR-γ agonist, pioglitazone on brain tissues oxidative damage and learning and memory impairment in juvenile hypothyroid rats. Int J Neurosci. 2019;129(10):1024–38. Alderton WK, Cooper CE, Knowles RG. Nitric oxide synthases: structure, function and inhibition. Biochem J. 2001;357(Pt 3):593–615. Saha RN, Pahan K. Signals for the induction of nitric oxide synthase in astrocytes. Neurochem Int. 2006 Jul;49(2):154–63. Estévez AG, Spear N, Thompson JA, Cornwell TL, Radi R, Barbeito L, et al. Nitric oxide-dependent production of cGMP supports the survival of rat embryonic motor neurons cultured with brain-derived neurotrophic factor. J Neurosci. 1998;18:3708–14. Dey P, Panga V, Raghunathan S. A cytokine signalling network for the regulation of inducible nitric oxide synthase expression in rheumatoid arthritis. PLoS One. 2016;11(9):e0161306. Pacher P, Beckman JS, Liaudet L. Nitric oxide and peroxynitrite in health and disease. Physiol Rev. 2007 Jan;87(1):315–424. Siti HN, Kamisah Y, Kamsiah J. The role of oxidative stress, antioxidants and vascular inflammation in cardiovascular disease (a review). Vascul Pharmacol. 2015;71:40–56. Faas MM, de Vos P. Mitochondrial function in immune cells in health and disease. Biochim Biophys Acta Mol Basis Dis. 2020;1866(10):165845. Mishra J, Vishwakarma J, Malik R, Gupta K, Pandey R, Maurya SK, et al. Hypothyroidism induces interleukin-1-dependent autophagy mechanism as a key mediator of hippocampal neuronal apoptosis and cognitive decline in postnatal rats. Mol Neurobiol. 2021;58(3):1196–211. Binder DK, Scharfman HE. Brain-derived neurotrophic factor. Growth Factors. 2004;22:123–31. Peng S, Wuu J, Mufson EJ, Fahnestock M. Precursor form of brain-derived neurotrophic factor and mature brain-derived neurotrophic factor are decreased in the pre-clinical stages of Alzheimer’s disease. J Neurochem. 2005;93:1412–21. Herholz K. Acetylcholine esterase activity in mild cognitive impairment and Alzheimer’s disease. Eur J Nucl Med Mol Imaging. 2008;35(1):S25–9. Minoura A, Kokaze A, Ono K. The relationship between thyroid function and cerebral blood flow in mild cognitive impairment and Alzheimer’s disease. PLoS One. 2019;314(4):e0214676. Wang F, Zeng X, Zhu Y, Ning D, Liu J, Liu C, et al. Effects of thyroxine and donepezil on hippocampal acetylcholine content, acetylcholinesterase activity, synaptotagmin-1 and SNAP-25 expression in hypothyroid adult rats. Mol Med Rep. 2015;11(2):775–82. Erecinska M, Silver IA. Ions and energy in mammalian brain. Prog Neurobiol. 1994;43:37–71. Goldstein I, Levy T, Galili D, Ovadia H, Yirmiya R, Rosen H, et al. Involvement of Na(+), K(+)-ATPase and endogenous digitalis-like compounds in depressive disorders. Biol Psychiatry. 2006;60:491–9. Kurup AR, Kurup PA. Membrane Na(+)-K+ ATPase mediated cascade in bipolar mood disorder, major depressive disorder, and schizophrenia – relationship to hemispheric dominance. Int J Neurosci. 2002;112:965–82. Xie Z, Askari A. Na(+)/K(+)-ATPase as a signal transducer. Eur J Biochem. 2002;269:2434–9. Hattori N, Kitagawa K, Higashida T, Yagyu K, Shimohama S, Wataya T, et al. CI-ATPase and Na+/K(+)-ATPase activities in Alzheimer’s disease brains. Neurosci Lett. 1998;254:141–4. Gamaro GD, Streck EL, Matté C, Prediger ME, Wyse AT, Dalmaz C. Reduction of hippocampal Na+, K+-ATPase activity in rats subjected to an experimental model of depression. Neurochem Res. 2003;28:1339–44. Attwell D, Laughlin SB. An energy budget for signaling in the grey matter of the brain. J Cereb Blood Flow Metab. 2001 Oct;21(10):1133–45. Savard P, Mérand Y, Di Paolo T, Dupont A. Effect of neonatal hypothyroidism on the serotonin system of the rat brain. Brain Res. 1984;292(1):99–108. Hassan WA, Aly MS, Rahman TA, Shahat AS. Impact of experimental hypothyroidism on monoamines level in discrete brain regions and other peripheral tissues of young and adult male rats. Int J Dev Neurosci. 2013;31(4):225–33. Loh HH, Lim LL, Yee A, Loh HS. Association between subclinical hypothyroidism and depression: an updated systematic review and meta-analysis. BMC Psychiatry. 2019;19(1):12. Youdim MBH, Bakhle YS. Monoamine oxidase: isoforms and inhibitors in Parkinson’s disease and depressive illness. Br J Pharmacol. 2006;147(Suppl 1):S287–96. Blier P, de Montigny C. Current advances and trends in the treatment of depression. Trends Pharmacol Sci. 1994;15(7):220–6. He HY, Tian JL, Deng YQ, Xiong X, Xu Y, Liao YM, et al. Association of brain-derived neurotrophic factor levels and depressive symptoms in young adults with acne vulgaris. BMC Psychiatry. 2019;19(1):193. Chaalal A, Poirier R, Blum D, Gillet B, Le Blanc P, Basquin M, et al. PTU-induced hypothyroidism in rats leads to several early neuropathological signs of Alzheimer’s disease in the hippocampus and spatial memory impairments. Hippocampus. 2014;24:1381–93. Rezk BM, Haenen GR, Van der Vijgh WJ, Bast A. Lipoic acid protects efficiently only against a specific form of peroxynitrite-induced damage. J Biol Chem. 2004;279(11):9693–7. Kim SM, Ha JS, Han AR, Cho SW, Yang SJ. Effects of α-lipoic acid on LPS-induced neuroinflammation and NLRP3 inflammasome activation through the regulation of BV-2 microglial cells activation. BMB Rep. 2019;52(10):613–8. Hadjighassem M, Kamalidehghan B, Shekarriz N, Baseerat A, Molavi N, Mehrpour M, et al. Oral consumption of α-linolenic acid increases serum BDNF levels in healthy adult humans. Nutr J. 2015;14(14):20. Souza GF, Saldanha GB, Freitas RM. Lipoic acid increases glutathione peroxidase, Na+, K+-ATPase and acetylcholinesterase activities in rat hippocampus after pilocarpine-induced seizures? Arq Neuropsiquiatr. 2010;68(4):586–91. Ahmed HH. Modulatory effects of vitamin E, acetyl-L-carnitine and α-lipoic acid on new potential biomarkers for Alzheimer’s disease in rat model. Exp Toxicol Pathol. 2012;64(6):549–56. Muñoz-Arenas G, Pulido G, Treviño S, Vázquez-Roque R, Flores G, Moran C, et al. Effects of metformin on recognition memory and hippocampal neuroplasticity in rats with metabolic syndrome. Synapse. 2020;74(9):e22153. Vial G, Detaille D, Guigas B. Role of Mitochondria in the mechanism(s) of action of metformin. Front Endocrinol. 2019;10:294. Bonnefont-Rousselot D, Raji B, Walrand S, Gardès-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;52:586–9. 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;477:534–40. Markowicz-Piasecka M, Sikora J, Mateusiak Ł, Mikiciuk-Olasik E, Huttunen KM. Metformin and its sulfenamide prodrugs inhibit human cholinesterase activity. Oxid Med Cell Longev. 2017;2017:7303096. Molitoris BA, Geerdes A, McIntosh JR. Dissociation and redistribution of Na+, K(+)-ATPase from its surface membrane actin cytoskeletal complex during cellular ATP depletion. J Clin Invest. 1991;88:462–9. Zhang J, Huang L, Shi X, Yang L, Hua F, Ma J, et al. Metformin protects against myocardial ischemia-reperfusion injury and cell pyroptosis via AMPK/NLRP3 inflammasome pathway. Aging. 2020;12(23):24270–87. Guo M, Mi J, Jiang QM, Xu JM, Tang YY, Tian G, et al. Metformin may produce antidepressant effects through improvement of cognitive function among depressed patients with diabetes mellitus. Clin Exp Pharmacol Physiol. 2014;41(9):650–6. Ahuja S, Uniyal A, Akhtar A, Sah SP. Alpha lipoic acid and metformin alleviates experimentally induced insulin resistance and cognitive deficit by modulation of TLR2 signalling. Pharmacol Rep. 2019;71(4):614–23. Article / Publication Details

First-Page Preview

Abstract of Research Article

Received: December 22, 2021
Accepted: March 11, 2022
Published online: March 30, 2022

Number of Print Pages: 14
Number of Figures: 5
Number of Tables: 2

ISSN: 0028-3835 (Print)
eISSN: 1423-0194 (Online)

For additional information: https://www.karger.com/NEN

Copyright / Drug Dosage / Disclaimer Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.

留言 (0)

沒有登入
gif