Generic placeholder image

Current Nutrition & Food Science

Editor-in-Chief

ISSN (Print): 1573-4013
ISSN (Online): 2212-3881

Research Article

Tualang Honey and its Methanolic Fraction Improve LPS-induced Learning and Memory Impairment in Male Rats: Comparison with Memantine

Author(s): Wan M.H.W. Yaacob, Idris Long, Rahimah Zakaria* and Zahiruddin Othman

Volume 16, Issue 3, 2020

Page: [333 - 342] Pages: 10

DOI: 10.2174/1573401315666181130103456

Abstract

Background: Tualang honey (TH) has been shown to exert beneficial effects on learning and memory function in various animal models. However, its learning and memory effects in lipopolysaccharide (LPS) rat model have not been elucidated.

Objective: The present study aimed to investigate the cognitive-enhancing effects of TH and its methanolic fraction in comparison to the clinically approved N-methyl-D-aspartate (NMDA) receptor antagonist (memantine) using LPS rat model.

Methods: A total of ninety male Sprague Dawley rats were divided into 5 groups: (i) control, (ii) untreated LPS (iii) LPS treated with 200 mg/kg TH, (iv) LPS treated with 150 mg/kg methanol fraction of TH (MTH) and (v) LPS treated with 10 mg/kg memantine. All treatments were administered intraperitoneally once daily for 14 days. Morris water maze (MWM) and novel object recognition (NOR) tests were performed to assess spatial and recognition memory function.

Results: The present study confirmed that LPS significantly impairs spatial and recognition memory and alone treatment with TH or MTH improved spatial and recognition memory comparable to memantine.

Conclusion: Both TH and its methanolic fraction improved spatial and recognition memory of LPS rat model comparable to memantine. Thus, TH and its methanolic fraction have potential preventivetherapeutic effects for neurodegenerative diseases involving neuroinflammation.

Keywords: Learning and memory, lipopolysaccharide, methanolic function, morris water maze, novel object recognition, tualang honey.

Graphical Abstract
[1]
Quintanilla RA, Orellana JA, von Bernhardi R. Understanding risk factors for Alzheimer’s disease: interplay of neuroinflammation, connexin-based communication and oxidative stress. Arch Med Res 2012; 43(8): 632-44.
[http://dx.doi.org/10.1016/j.arcmed.2012.10.016] [PMID: 23142264]
[2]
Song JH, Lee JW, Shim B, et al. Glycyrrhizin alleviates neuroinflammation and memory deficit induced by systemic lipopolysaccharide treatment in mice. Molecules 2013; 18(12): 15788-803.
[http://dx.doi.org/10.3390/molecules181215788] [PMID: 24352029]
[3]
Lyman M, Lloyd DG, Ji X, Vizcaychipi MP, Ma D. Neuroinflammation: the role and consequences. Neurosci Res 2014; 79: 1-12.
[http://dx.doi.org/10.1016/j.neures.2013.10.004] [PMID: 24144733]
[4]
Glass CK, Saijo K, Winner B, Marchetto MC, Gage FH. Mechanisms underlying inflammation in neurodegeneration. Cell 2010; 140(6): 918-34.
[http://dx.doi.org/10.1016/j.cell.2010.02.016] [PMID: 20303880]
[5]
Park SE, Sapkota K, Kim S, Kim H, Kim SJ. Kaempferol acts through mitogen-activated protein kinases and protein kinase B/AKT to elicit protection in a model of neuroinflammation in BV2 microglial cells. Br J Pharmacol 2011; 164(3): 1008-25.
[http://dx.doi.org/10.1111/j.1476-5381.2011.01389.x] [PMID: 21449918]
[6]
Shabab T, Khanabdali R, Moghadamtousi SZ, Kadir HA, Mohan G. Neuroinflammation pathways: a general review. Int J Neurosci 2017; 127(7): 624-33.
[http://dx.doi.org/10.1080/00207454.2016.1212854] [PMID: 27412492]
[7]
Yiannopoulou KG, Papageorgiou SG. Current and future treatments for Alzheimer’s disease. Ther Adv Neurol Disorder 2013; 6(1): 19-33.
[8]
Herrmann N, Chau SA, Kircanski I, Lanctôt KL. Current and emerging drug treatment options for Alzheimer’s disease: a systematic review. Drugs 2011; 71(15): 2031-65.
[http://dx.doi.org/10.2165/11595870-000000000-00000] [PMID: 21985169]
[9]
Summers WK. Current and future treatments of memory complaints and Alzheimer’s disease. Therapy 2011; 8(5): 491-504.
[http://dx.doi.org/10.2217/thy.11.60]
[10]
Ahmad I, Jimenez H, Yaacob NS, Yusuf N. Tualang honey protects keratinocytes from ultraviolet radiation-induced inflammation and DNA damage. Photochem Photobiol 2012; 88(5): 1198-204.
[http://dx.doi.org/10.1111/j.1751-1097.2012.01100.x] [PMID: 22276569]
[11]
Nurul Syazana MS, Halim AS, Gan SH, Shamsuddin S. Antiproliferative effect of methanolic extraction of tualang honey on human keloid fibroblasts. BMC Complement Altern Med 2011; 11: 82.
[http://dx.doi.org/10.1186/1472-6882-11-82] [PMID: 21943200]
[12]
Khalil MI, Alam N, Moniruzzaman M, Sulaiman SA, Gan SH. Phenolic acid composition and antioxidant properties of Malaysian honeys. J Food Sci 2011; 76(6): C921-8.
[http://dx.doi.org/10.1111/j.1750-3841.2011.02282.x] [PMID: 22417491]
[13]
Erejuwa OO, Gurtu S, Sulaiman SA, Ab Wahab MS, Sirajudeen KN, Salleh MS. Hypoglycemic and antioxidant effects of honey supplementation in streptozotocin-induced diabetic rats. Int J Vitam Nutr Res 2010; 80(1): 74-82.
[http://dx.doi.org/10.1024/0300-9831/a000008] [PMID: 20533247]
[14]
Al-Rahbi B, Zakaria R, Othman Z, Hassan A, Mohd Ismail ZI, Muthuraju S. Tualang honey supplement improves memory performance and hippocampal morphology in stressed ovariectomized rats. Acta Histochem 2014; 116(1): 79-88.
[http://dx.doi.org/10.1016/j.acthis.2013.05.004] [PMID: 23810156]
[15]
Azman KF, Zakaria R, AbdAziz C, Othman Z, Al-Rahbi B. Tualang honey improves memory performance and decreases depressive-like behavior in rats exposed to loud noise stress. Noise Health 2015; 17(75): 83-9.
[http://dx.doi.org/10.4103/1463-1741.153388] [PMID: 25774610]
[16]
Kamarulzaidi M, Mohd Yusoff M, Mohamed A, Hasan Adli D. Tualang honey consumption enhanced hippocampal pyramidal count and spatial memory performance of adult male rats. Sains Malays 2016; 45(2): 215-20.
[17]
Saxena A, Phyu HIMA-A, Oothuman P. Improved spatial learning and memory performance following Tualang honey treatment during cerebral hypoperfusion-induced neurodegeneration. J Transl Sci 2016; 2(5): 264-71.
[http://dx.doi.org/10.15761/JTS.1000150]
[18]
Al-Rahbi B, Zakaria R, Othman Z, Hassan A, Ahmad AH. Enhancement of BDNF concentration and restoration of the hypothalamic-pituitary-adrenal axis accompany reduced depressive-like behaviour in stressed ovariectomised rats treated with either Tualang honey or estrogen. Sci World J 2014. 2014; 2014: 310821
[http://dx.doi.org/10.1155/2014/310821]
[19]
Al-Rahbi B, Zakaria R, Othman Z, Hassan A, Ahmad AH. The effects of Tualang honey supplement on medial prefrontal cortex morphology and cholinergic system in stressed ovariectomised rats. Int J Appl Res Nat Prod 2014; 7(3): 28-36.
[20]
Al-Rahbi B, Zakaria R, Othman Z, Hassan A, Ahmad AH. Protective effects of Tualang honey against oxidative stress and anxiety-like behaviour in stressed ovariectomized rats. Int Sch Res Notices 2014; 2014; 2014: 10.
[http://dx.doi.org/10.1155/2014/521065] [http://dx.doi.org/10.1155/20 14/521065]
[21]
Kaškonienė V, Maruška A, Kornyšova O. Quantitative and qualitative determination of phenolic compounds in honey. Cheminė Technologija 2009; 3(52): 74-80.
[22]
Wan Yaacob WMH, Long I, Zakaria R, Othman Z. Protective effects of Tualang honey and its methanolic fraction against lipopolysaccharide-induced oxidative damage in the rats’ hippocampus. Health Environ J 2018; 9(Suppl. 1): 36.
[23]
Lagrèze WA, Knörle R, Bach M, Feuerstein TJ. Memantine is neuroprotective in a rat model of pressure-induced retinal ischemia. Invest Ophthalmol Vis Sci 1998; 39(6): 1063-6.
[PMID: 9579489]
[24]
Rose R, Banerjee A, Ramaiah SK. Characterization of a lipopolysaccharide mediated neutrophilic hepatitis model in Sprague Dawley rats. J Appl Toxicol 2007; 27(6): 602-11.
[http://dx.doi.org/10.1002/jat.1243] [PMID: 17370240]
[25]
Vorhees CV, Williams MT. Morris water maze: procedures for assessing spatial and related forms of learning and memory. Nat Protoc 2006; 1(2): 848-58.
[http://dx.doi.org/10.1038/nprot.2006.116] [PMID: 17406317]
[26]
Bevins RA, Besheer J. Object recognition in rats and mice: a one-trial non-matching-to-sample learning task to study ‘recognition memory’. Nat Protoc 2006; 1(3): 1306-11.
[http://dx.doi.org/10.1038/nprot.2006.205] [PMID: 17406415]
[27]
Kim JH, Wang Q, Choi JM, Lee S, Cho EJ. Protective role of caffeic acid in an Aβ25-35-induced Alzheimer’s disease model. Nutr Res Pract 2015; 9(5): 480-8.
[http://dx.doi.org/10.4162/nrp.2015.9.5.480] [PMID: 26425277]
[28]
Carlini VP, Martini AC, Schiöth HB, Ruiz RD, Fiol de Cuneo M, de Barioglio SR. Decreased memory for novel object recognition in chronically food-restricted mice is reversed by acute ghrelin administration. Neuroscience 2008; 153(4): 929-34.
[http://dx.doi.org/10.1016/j.neuroscience.2008.03.015] [PMID: 18434026]
[29]
Bogdanov S, Jurendic T, Sieber R, Gallmann P. Honey for nutrition and health: a review. J Am Coll Nutr 2008; 27(6): 677-89.
[http://dx.doi.org/10.1080/07315724.2008.10719745] [PMID: 19155427]
[30]
Honey-health and therapeutic qualities. The National Honey Board: Logmont, CO, USA 2003. Available from: http://www.nhb.org/
[31]
Al-Himyari FA. The use of honey as a natural preventive therapy of cognitive decline and dementia in the middle east. Alzheimers Dement 2009; 5(4): 247.
[http://dx.doi.org/10.1016/j.jalz.2009.04.248]
[32]
Lee B, Sur B, Park J, et al. Ginsenoside rg3 alleviates lipopolysaccharide-induced learning and memory impairments by anti-inflammatory activity in rats. Biomol Ther (Seoul) 2013; 21(5): 381-90.
[http://dx.doi.org/10.4062/biomolther.2013.053] [PMID: 24244826]
[33]
Zhang X-Y, Cao J-B, Zhang L-M, Li Y-F, Mi W-D. Deferoxamine attenuates lipopolysaccharide-induced neuroinflammation and memory impairment in mice. J Neuroinflammation 2015; 12: 20.
[http://dx.doi.org/10.1186/s12974-015-0238-3] [PMID: 25644393]
[34]
Klein SL, Nelson RJ. Activation of the immune-endocrine system with lipopolysaccharide reduces affiliative behaviors in voles. Behav Neurosci 1999; 113(5): 1042-8.
[http://dx.doi.org/10.1037/0735-7044.113.5.1042] [PMID: 10571486]
[35]
Hart BL. Biological basis of the behavior of sick animals. Neurosci Biobehav Rev 1988; 12(2): 123-37.
[http://dx.doi.org/10.1016/S0149-7634(88)80004-6] [PMID: 3050629]
[36]
Pugh CR, Kumagawa K, Fleshner M, Watkins LR, Maier SF, Rudy JW. Selective effects of peripheral lipopolysaccharide administration on contextual and auditory-cue fear conditioning. Brain Behav Immun 1998; 12(3): 212-29.
[http://dx.doi.org/10.1006/brbi.1998.0524] [PMID: 9769157]
[37]
Castanon N, Bluthé RM, Dantzer R. Chronic treatment with the atypical antidepressant tianeptine attenuates sickness behavior induced by peripheral but not central lipopolysaccharide and interleukin-1beta in the rat. Psychopharmacology (Berl) 2001; 154(1): 50-60.
[http://dx.doi.org/10.1007/s002130000595] [PMID: 11292006]
[38]
Frank-Cannon TC, Alto LT, McAlpine FE, Tansey MG. Does neuroinflammation fan the flame in neurodegenerative diseases? Mol Neurodegener 2009; 4: 47-7.
[http://dx.doi.org/10.1186/1750-1326-4-47] [PMID: 19917131]
[39]
Zhao W-X, Zhang J-H, Cao J-B, et al. Acetaminophen attenuates lipopolysaccharide-induced cognitive impairment through antioxidant activity. J Neuroinflammation 2017; 14(1): 17.
[http://dx.doi.org/10.1186/s12974-016-0781-6] [PMID: 28109286]
[40]
Rosi S, Vazdarjanova A, Ramirez-Amaya V, Worley PF, Barnes CA, Wenk GL. Memantine protects against LPS-induced neuroinflammation, restores behaviorally-induced gene expression and spatial learning in the rat. Neuroscience 2006; 142(4): 1303-15.
[http://dx.doi.org/10.1016/j.neuroscience.2006.08.017] [PMID: 16989956]
[41]
Devasvaran K, Yong Y-K. Anti-inflammatory and wound healing properties of Malaysia Tualang honey. Curr Sci 2016; 110(1): 47-51.
[http://dx.doi.org/10.18520/cs/v110/i1/48-52]
[42]
Zarifkar A, Choopani S, Ghasemi R, et al. Agmatine prevents LPS-induced spatial memory impairment and hippocampal apoptosis. Eur J Pharmacol 2010; 634(1-3): 84-8.
[http://dx.doi.org/10.1016/j.ejphar.2010.02.029] [PMID: 20184876]
[43]
Lalonde R. The neurobiological basis of spontaneous alternation. Neurosci Biobehav Rev 2002; 26(1): 91-104.
[http://dx.doi.org/10.1016/S0149-7634(01)00041-0] [PMID: 11835987]
[44]
Vogel-Ciernia A, Wood MA. Examining object location and object recognition memory in mice. Curr Protoc Neurosci 2014; 69(8.31): 1-17.
[http://dx.doi.org/10.1002/0471142301.ns0831s69]
[45]
Zakaria R, Wan Yaacob WMH, Othman Z, Long I, Ahmad AH, Al-Rahbi B. Lipopolysaccharide-induced memory impairment in rats: a model of Alzheimer’s disease. Physiol Res 2017; 66(4): 553-65.
[http://dx.doi.org/10.33549/physiolres.933480] [PMID: 28406691]
[46]
Tanaka S, Ide M, Shibutani T, et al. Lipopolysaccharide-induced microglial activation induces learning and memory deficits without neuronal cell death in rats. J Neurosci Res 2006; 83(4): 557-66.
[http://dx.doi.org/10.1002/jnr.20752] [PMID: 16429444]
[47]
Ghosh S, Lertwattanarak R, Garduño JdeJ, et al. Elevated muscle TLR4 expression and metabolic endotoxemia in human aging. J Gerontol A Biol Sci Med Sci 2015; 70(2): 232-46.
[http://dx.doi.org/10.1093/gerona/glu067] [PMID: 24846769]
[48]
Tansey MG, McCoy MK, Frank-Cannon TC. Neuroinflammatory mechanisms in Parkinson’s disease: potential environmental triggers, pathways, and targets for early therapeutic intervention. Exp Neurol 2007; 208(1): 1-25.
[http://dx.doi.org/10.1016/j.expneurol.2007.07.004] [PMID: 17720159]
[49]
Schmid CD, Melchior B, Masek K, et al. Differential gene expression in LPS/IFNgamma activated microglia and macrophages: in vitro versus in vivo. J Neurochem 2009; 109(Suppl. 1): 117-25.
[http://dx.doi.org/10.1111/j.1471-4159.2009.05984.x] [PMID: 19393017]
[50]
Zhu B, Wang Z-G, Ding J, et al. Chronic lipopolysaccharide exposure induces cognitive dysfunction without affecting BDNF expression in the rat hippocampus. Exp Ther Med 2014; 7(3): 750-4.
[http://dx.doi.org/10.3892/etm.2014.1479] [PMID: 24520281]
[51]
Hritcu L, Ciobica A, Stefan M, Mihasan M, Palamiuc L, Nabeshima T. Spatial memory deficits and oxidative stress damage following exposure to lipopolysaccharide in a rodent model of Parkinson’s disease. Neurosci Res 2011; 71(1): 35-43.
[http://dx.doi.org/10.1016/j.neures.2011.05.016] [PMID: 21663772]
[52]
Joshi R, Garabadu D, Teja GR, Krishnamurthy S. Silibinin ameliorates LPS-induced memory deficits in experimental animals. Neurobiol Learn Mem 2014; 116(0): 117-31.
[http://dx.doi.org/10.1016/j.nlm.2014.09.006] [PMID: 25444719]
[53]
Vallianou V, Gounari P, Skourtis A, Panagos J, Kazazis C. Honey and its anti-inflammatory, anti-bacterial and anti-oxidant properties. Gen Med (Los Angel) 2014; 2: 132.
[http://dx.doi.org/10.4172/2327-5146.1000132]
[54]
Mijanur Rahman M, Gan SH, Khalil MI. Neurological effects of honey: current and future prospects. Evid Based Complement Alternat Med 2014; 2014958721
[http://dx.doi.org/10.1155/2014/958721] [PMID: 24876885]

© 2024 Bentham Science Publishers | Privacy Policy