Generic placeholder image

当代阿耳茨海默病研究

Editor-in-Chief

ISSN (Print): 1567-2050
ISSN (Online): 1875-5828

Research Article

复发和不同剂量氯胺酮暴露对幼鼠焦虑样行为和运动活动的影响

卷 19, 期 14, 2022

发表于: 15 March, 2023

页: [933 - 942] 页: 10

弟呕挨: 10.2174/1567205020666230308123718

价格: $65

摘要

背景:氯胺酮是一种广泛使用的麻醉剂。尽管在青少年时期使用氯胺酮的潜在不良影响尚不确定,但某些研究报告称,暴露于反复麻醉的儿童可能面临运动功能和行为风险方面神经发育缺陷的风险增加。我们的目的是研究反复接触不同剂量的氯胺酮对幼年大鼠焦虑行为和运动活动的长期影响。 目的:我们旨在研究反复接触不同剂量的氯胺酮对幼年大鼠焦虑行为和运动活动的长期影响。 方法:将 32 只 Wistar Albino 幼年雄性大鼠随机分为 5 mg/kg、20 mg/kg 和 50 mg/kg 氯胺酮(KET)和生理盐水(C 组)组,连续 3 天在 3-小时间隔 3 剂。最后一次 KET 剂量后十天,通过开放场测试 (OFT)、高架十字迷宫 (EPM) 和明暗盒 (LDB) 分析行为参数。使用 Kruskall-Wallis 检验进行统计分析,然后进行 Dunn 的多重比较检验。 结果:与 C 组相比,50 mg/kg KET 组的无支撑饲养行为减少。与 C 组相比,50 mg/kg KET 组的不正确过渡时间、总梳理时间和转移潜伏期显着增加。 结论:这些结果表明,50 mg/kg KET 会导致类似焦虑的行为并破坏记忆和空间导航。氯胺酮剂量与氯胺酮对幼年大鼠焦虑样行为的后期影响有关。需要进一步的研究来确定在氯胺酮剂量对焦虑和记忆的不同影响中发挥作用的机制。

关键词: 焦虑,氯胺酮,运动活动,记忆,大鼠,空间导航。

[1]
Maddox VH, Godefroi EF, Parcell RF. The synthesis of phencyclidine and other 1-arylcyclohexylamines. J Med Chem 1965; 8(2): 230-5.
[http://dx.doi.org/10.1021/jm00326a019] [PMID: 14332667]
[2]
Li L, Vlisides PE. Ketamine: 50 years of modulating the mind. Front Hum Neurosci 2016; 10: 612.
[http://dx.doi.org/10.3389/fnhum.2016.00612] [PMID: 27965560]
[3]
Gao M, Rejaei D, Liu H. Ketamine use in current clinical practice. Acta Pharmacol Sin 2016; 37(7): 865-72.
[http://dx.doi.org/10.1038/aps.2016.5] [PMID: 27018176]
[4]
Isik B, Bora H, Yılmaz Z, et al. Recurrent anaesthetic management at ten children under radiotherapy: Medical education. Turkiye Klinikleri J Med Sci 2007; 27(6): 883-8.
[5]
Pampal HK. Anestezi Pratiğinde Sedasyon. In: Isık B, Ed. Sedasyon amacıyla kullanılan ilaçlar antidotları ve farmakolojik özellikleri. Ankara: Akademisyen 2019; pp. 21-58.
[6]
Choudhury D, Autry AE, Tolias KF, Krishnan V. Ketamine: Neuroprotective or neurotoxic? Front Neurosci 2021; 15: 672526.
[http://dx.doi.org/10.3389/fnins.2021.672526] [PMID: 34566558]
[7]
Banov MD, Young JR, Dunn T, Szabo ST. Efficacy and safety of ketamine in the management of anxiety and anxiety spectrum disorders: a review of the literature. CNS Spectr 2020; 25(3): 331-42.
[http://dx.doi.org/10.1017/S1092852919001238] [PMID: 31339086 ]
[8]
Raper J, Simon HK, Kamat PP. Long-term evidence of neonatal anaesthesia neurotoxicity linked to behavioural phenotypes in monkeys: where do we go from here? Br J Anaesth 2021; 127(3): 343-5.
[http://dx.doi.org/10.1016/j.bja.2021.06.005] [PMID: 34272059]
[9]
Yan J, Jiang H. Dual effects of ketamine: neurotoxicity versus neuroprotection in anesthesia for the developing brain. J Neurosurg Anesthesiol 2014; 26(2): 155-60.
[http://dx.doi.org/10.1097/ANA.0000000000000027] [PMID: 24275940]
[10]
Lüscher C, Malenka RC. NMDA receptor-dependent long-term potentiation and long-term depression (LTP/LTD). Cold Spring Harb Perspect Biol 2012; 4(6): a005710.
[http://dx.doi.org/10.1101/cshperspect.a005710] [PMID: 22510460]
[11]
Meyer K. The role of dendritic signaling in the anesthetic suppression of consciousness. Anesthesiology 2015; 122(6): 1415-31.
[http://dx.doi.org/10.1097/ALN.0000000000000673] [PMID: 25901843]
[12]
Riedel G, Platt B, Micheau J. Glutamate receptor function in learning and memory. Behav Brain Res 2003; 140(1-2): 1-47.
[http://dx.doi.org/10.1016/S0166-4328(02)00272-3] [PMID: 12644276]
[13]
Sleigh J, Harvey M, Voss L, Denny B. Ketamine – More mechanisms of action than just NMDA blockade. Trends in Anaesthesia and Critical Care 2014; 4(2-3): 76-81.
[http://dx.doi.org/10.1016/j.tacc.2014.03.002]
[14]
Zanos P, Moaddel R, Morris PJ, et al. NMDAR inhibition-independent antidepressant actions of ketamine metabolites. Nature 2016; 533(7604): 481-6.
[http://dx.doi.org/10.1038/nature17998]
[15]
Lazarevic V, Yang Y, Flais I, Svenningsson P. Ketamine decreases neuronally released glutamate via retrograde stimulation of presynaptic adenosine A1 receptors. Mol Psychiatry 2021; 26(12): 7425-35.
[http://dx.doi.org/10.1038/s41380-021-01246-3] [PMID: 34376822]
[16]
Shi M, Ding J, Li L, et al. Effects of ketamine on learning and memory in the hippocampus of rats through ERK, CREB, and Arc. Brain Sci 2020; 11(1): 27.
[http://dx.doi.org/10.3390/brainsci11010027] [PMID: 33383707]
[17]
Ing CH, DiMaggio CJ, Whitehouse AJO, et al. Neurodevelopmental outcomes after initial childhood anesthetic exposure between ages 3 and 10 years. J Neurosurg Anesthesiol 2014; 26(4): 377-86.
[http://dx.doi.org/10.1097/ANA.0000000000000121] [PMID: 25144506]
[18]
Imre G, Fokkema DS, Boer JAD, Ter Horst GJ. Dose–response characteristics of ketamine effect on locomotion, cognitive function and central neuronal activity. Brain Res Bull 2006; 69(3): 338-45.
[http://dx.doi.org/10.1016/j.brainresbull.2006.01.010] [PMID: 16564431]
[19]
Melo A, Leite-Almeida H, Ferreira C, Sousa N, Pêgo JM. Exposure to ketamine anesthesia affects rat impulsive behavior. Front Behav Neurosci 2016; 10: 226.
[http://dx.doi.org/10.3389/fnbeh.2016.00226]
[20]
Festing MFW. On determining sample size in experiments involving laboratory animals. Lab Anim 2018; 52(4): 341-50.
[http://dx.doi.org/10.1177/0023677217738268] [PMID: 29310487]
[21]
Hall CS. Emotional behavior in the rat. I. Defecation and urination as measures of individual differences in emotionality. J Comp Psychol 1934; 18(3): 385-403.
[http://dx.doi.org/10.1037/h0071444]
[22]
Prut L, Belzung C. The open field as a paradigm to measure the effects of drugs on anxiety-like behaviors: A review. Eur J Pharmacol 2003; 463(1-3): 3-33.
[http://dx.doi.org/10.1016/S0014-2999(03)01272-X] [PMID: 12600700]
[23]
Caliskan H, Akat F, Tatar Y, et al. Effects of exercise training on anxiety in diabetic rats. Behav Brain Res 2019; 30(376): 112084.
[http://dx.doi.org/10.1016/j.bbr.2019.112084]
[24]
Bilgic Y, Demir EA, Bilgic N, Dogan H, Tutuk O, Tumer C. Detrimental effects of chia (Salvia hispanica L.) seeds on learning and memory in aluminum chloride-induced experimental Alzheimer’s disease. Acta Neurobiol Exp 2018; 78(4): 322-31.
[http://dx.doi.org/10.21307/ane-2018-031] [PMID: 30624431]
[25]
Simon P, Dupuis R, Costentin J. Thigmotaxis as an index of anxiety in mice. Influence of dopaminergic transmissions. Behav Brain Res 1994; 61(1): 59-64.
[http://dx.doi.org/10.1016/0166-4328(94)90008-6] [PMID: 7913324]
[26]
Caliskan H, Akat F, Omercioglu G, Bastug G, Ficicilar H, Bastug M. Aerobic exercise has an anxiolytic effect on streptozotocin-induced diabetic rats. Acta Neurobiol Exp 2020; 80(3): 245-55.
[http://dx.doi.org/10.21307/ane-2020-022] [PMID: 32990283]
[27]
Sturman O, Germain PL, Bohacek J. Exploratory rearing: A context- and stress-sensitive behavior recorded in the open-field test. Stress 2018; 21(5): 443-52.
[http://dx.doi.org/10.1080/10253890.2018.1438405] [PMID: 29451062]
[28]
Kalueff AV, Stewart AM, Song C, Berridge KC, Graybiel AM, Fentress JC. Neurobiology of rodent self-grooming and its value for translational neuroscience. Nat Rev Neurosci 2016; 17(1): 45-59.
[http://dx.doi.org/10.1038/nrn.2015.8] [PMID: 26675822]
[29]
Handley SL, Mithani S. Effects of alpha-adrenoceptor agonists and antagonists in a maze-exploration model of? fear?-motivated behaviour. Naunyn Schmiedebergs Arch Pharmacol 1984; 327(1): 1-5.
[http://dx.doi.org/10.1007/BF00504983] [PMID: 6149466]
[30]
Pellow S, Chopin P, File SE, Briley M. Validation of open: Closed arm entries in an elevated plus-maze as a measure of anxiety in the rat. J Neurosci Methods 1985; 14(3): 149-67.
[http://dx.doi.org/10.1016/0165-0270(85)90031-7] [PMID: 2864480]
[31]
Calişkan H, Cihan KH, Güneş E, et al. Duloxetine alleviates high light-induced anxiety-related behaviors in Wistar rats. Trop J Pharm Res 2019; 18(11): 2319-23.
[http://dx.doi.org/10.4314/tjpr.v18i11.13]
[32]
Holly KS, Orndorff CO, Murray TA. MATSAP: An automated analysis of stretch-attend posture in rodent behavioral experiments. Sci Rep 2016; 6(1): 31286.
[http://dx.doi.org/10.1038/srep31286] [PMID: 27503239]
[33]
Morales-Delgado N. Popović N, De la Cruz-Sánchez E, Caballero Bleda M, Popović M. Time-of-day and age impact on memory in elevated plus-maze test in rats. Front Behav Neurosci 2018; 12: 304.
[http://dx.doi.org/10.3389/fnbeh.2018.00304] [PMID: 30574075]
[34]
Crawley J, Goodwin FK. Preliminary report of a simple animal behavior model for the anxiolytic effects of benzodiazepines. Pharmacol Biochem Behav 1980; 13(2): 167-70.
[http://dx.doi.org/10.1016/0091-3057(80)90067-2] [PMID: 6106204]
[35]
Flecknell P. Laboratory Animal Anaesthesia. Amsterdam: Elsevier 1996.
[36]
Carrier N, Kabbaj M. Sex differences in the antidepressant-like effects of ketamine. Neuropharmacology 2013; 70: 27-34.
[http://dx.doi.org/10.1016/j.neuropharm.2012.12.009] [PMID: 23337256]
[37]
Franceschelli A, Sens J, Herchick S, Thelen C, Pitychoutis PM. Sex differences in the rapid and the sustained antidepressant-like effects of ketamine in stress-naïve and “depressed” mice exposed to chronic mild stress. Neuroscience 2015; 290: 49-60.
[http://dx.doi.org/10.1016/j.neuroscience.2015.01.008] [PMID: 25595985]
[38]
Parise EM, Alcantara LF, Warren BL, et al. Repeated ketamine exposure induces an enduring resilient phenotype in adolescent and adult rats. Biol Psychiatry 2013; 74(10): 750-9.
[http://dx.doi.org/10.1016/j.biopsych.2013.04.027] [PMID: 23790225]
[39]
Saland SK, Schoepfer KJ, Kabbaj M. Hedonic sensitivity to low-dose ketamine is modulated by gonadal hormones in a sex-dependent manner. Sci Rep 2016; 6(1): 21322.
[http://dx.doi.org/10.1038/srep21322] [PMID: 26888470]
[40]
Seibenhener ML, Wooten MC. Use of the Open Field Maze to measure locomotor and anxiety-like behavior in mice. J Vis Exp 2015; 96(96): e52434.
[http://dx.doi.org/10.3791/52434] [PMID: 25742564]
[41]
Sharma S, Rakoczy S, Brown-Borg H. Assessment of spatial memory in mice. Life Sci 2010; 87(17-18): 521-36.
[http://dx.doi.org/10.1016/j.lfs.2010.09.004] [PMID: 20837032]
[42]
Quillfeldt JA. Behavioral methods to study learning and memory in rats Rodent Model as Tools in Ethical Biomedical Research. Cham: Springer 2006; pp. 1-42.
[43]
Weston RG, Fitzgerald PJ, Watson BO. Repeated dosing of ketamine in the forced swim test: Are multiple shots better than one? Front Psychiatry 2021; 12: 659052.
[http://dx.doi.org/10.3389/fpsyt.2021.659052] [PMID: 34045982]
[44]
Hetzler BE, Swain Wautlet B. Ketamine-induced locomotion in rats in an open-field. Pharmacol Biochem Behav 1985; 22(4): 653-5.
[http://dx.doi.org/10.1016/0091-3057(85)90291-6] [PMID: 3991775]
[45]
Fraga DB, Olescowicz G, Moretti M, et al. Anxiolytic effects of ascorbic acid and ketamine in mice. J Psychiatr Res 2018; 100: 16-23.
[http://dx.doi.org/10.1016/j.jpsychires.2018.02.006] [PMID: 29475017]
[46]
Shin SY, Baek NJ, Han SH, Min SS. Chronic administration of ketamine ameliorates the anxiety- and aggressive-like behavior in adolescent mice induced by neonatal maternal separation. Korean J Physiol Pharmacol 2019; 23(1): 81-7.
[http://dx.doi.org/10.4196/kjpp.2019.23.1.81] [PMID: 30627013]
[47]
Pitsikas N, Georgiadou G, Delis F, Antoniou K. Effects of anesthetic ketamine on anxiety-like behaviour in rats. Neurochem Res 2019; 44(4): 829-38.
[http://dx.doi.org/10.1007/s11064-018-02715-y] [PMID: 30656595]
[48]
Akillioglu K, Karadepe M. Effect neonatal ketamine treatment on exploratory and anxiety-like behaviours in adulthood. Clin Psychopharmacol Neurosci 2021; 19(1): 93-103.
[http://dx.doi.org/10.9758/cpn.2021.19.1.93] [PMID: 33508792]
[49]
Amorim M, Bravo J, Silva AI, et al. Repeated exposure to ketamine in adolescent rats results in persistent anxiety in the adulthood. J Drug Alcohol Res 2018; 7(1): 1-10.
[http://dx.doi.org/10.4303/jdar/236060]
[50]
Ghosal S, Duman CH, Liu RJ, et al. Ketamine rapidly reverses stress-induced impairments in GABAergic transmission in the prefrontal cortex in male rodents. Neurobiol Dis 2020; 134: 104669.
[http://dx.doi.org/10.1016/j.nbd.2019.104669] [PMID: 31707118]
[51]
Neves G, Borsoi M, Antonio CB, Pranke MA, Betti AH, Rates SMK. Is forced swimming immobility a good endpoint for modeling negative symptoms of schizophrenia?-study of sub-anesthetic ketamine repeated administration effects. An Acad Bras Cienc 2017; 89(3): 1655-69.
[52]
Pribish A, Wood N, Kalava A. A review of nonanesthetic uses of ketamine. Anesthesiol Res Pract 2020; 2020: 1-15.
[http://dx.doi.org/10.1155/2020/5798285] [PMID: 32308676]
[53]
Grandjean P, Landrigan PJ. Neurobehavioural effects of developmental toxicity. Lancet Neurol 2014; 13(3): 330-8.
[http://dx.doi.org/10.1016/S1474-4422(13)70278-3] [PMID: 24556010]
[54]
Chen Y, Yang Z, Wei L, et al. Yes associated protein protects and rescues SH SY5Y cells from ketamine induced apoptosis. Mol Med Rep 2020; 22(3): 2342-50.
[http://dx.doi.org/10.3892/mmr.2020.11328] [PMID: 32705208]
[55]
Gerb SA, Cook JE, Gochenauer AE, et al. Ketamine tolerance in Sprague-Dawley rats after chronic administration of ketamine, morphine, or cocaine. Comp Med 2019; 69(1): 29-34.
[http://dx.doi.org/10.30802/AALAS-CM-18-000053] [PMID: 30696519]
[56]
Corriger A, Pickering G. Ketamine and depression: A narrative review. Drug Des Devel Ther 2019; 13(13): 3051-67.
[http://dx.doi.org/10.2147/DDDT.S221437] [PMID: 31695324]
[57]
Yankelevitch-Yahav R, Franko M, Huly A, Doron R. The forced swim test as a model of depressive-like behavior. J Vis Exp 2015; 97(97): 52587.
[http://dx.doi.org/10.3791/52587] [PMID: 25867960]
[58]
Autry AE, Adachi M, Nosyreva E, et al. NMDA receptor blockade at rest triggers rapid behavioural antidepressant responses. Nature 2011; 475(7354): 91-5.
[http://dx.doi.org/10.1038/nature10130] [PMID: 21677641]
[59]
Polat Çorumlu E. Aydın OÖ, Aydın EG, Ulupınar E. Effects of single-dose ketamine infusion on behavioral parameters and neuronal activation in the medial prefrontal cortex of juvenile rats exposed to prenatal stress. Anatomy 2015; 9(3): 142-50.
[http://dx.doi.org/10.2399/ana.15.027]
[60]
Li N, Lee B, Liu RJ, et al. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science 2010; 329(5994): 959-64.
[61]
Shiroma PR, Albott CS, Johns B, Thuras P, Wels J, Lim KO. Neurocognitive performance and serial intravenous subanesthetic ketamine in treatment-resistant depression. Int J Neuropsychopharmacol 2014; 17(11): 1805-13.
[http://dx.doi.org/10.1017/S1461145714001011] [PMID: 24963561]
[62]
Zarate CA Jr, Singh JB, Carlson PJ, et al. A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch Gen Psychiatry 2006; 63(8): 856-64.
[http://dx.doi.org/10.1001/archpsyc.63.8.856] [PMID: 16894061]
[63]
Zou X, Patterson TA, Divine RL, et al. Prolonged exposure to ketamine increases neurodegeneration in the developing monkey brain. Int J Dev Neurosci 2009; 27(7): 727-31.
[http://dx.doi.org/10.1016/j.ijdevneu.2009.06.010] [PMID: 19580862]
[64]
Zou X, Patterson TA, Sadovova N, et al. Potential neurotoxicity of ketamine in the developing rat brain. Toxicol Sci 2009; 108(1): 149-58.
[http://dx.doi.org/10.1093/toxsci/kfn270] [PMID: 19126600]
[65]
Ikonomidou C, Bosch F, Miksa M, et al. Blockade of NMDA receptors and apoptotic neurodegeneration in the developing brain. Science 1999; 283(5398): 70-4.
[http://dx.doi.org/10.1126/science.283.5398.70] [PMID: 9872743]
[66]
Johnson SC, Pan A, Li L, Sedensky M, Morgan P. Neurotoxicity of anesthetics: Mechanisms and meaning from mouse intervention studies. Neurotoxicol Teratol 2019; 71: 22-31.
[http://dx.doi.org/10.1016/j.ntt.2018.11.004] [PMID: 30472095]
[67]
Liu F, Paule MG, Ali S, Wang C. Ketamine-induced neurotoxicity and changes in gene expression in the developing rat brain. Curr Neuropharmacol 2011; 9(1): 256-61.
[http://dx.doi.org/10.2174/157015911795017155] [PMID: 21886601]
[68]
Mion G, Villevieille T. Ketamine pharmacology: An update (pharmacodynamics and molecular aspects, recent findings). CNS Neurosci Ther 2013; 19(6): 370-80.
[http://dx.doi.org/10.1111/cns.12099] [PMID: 23575437]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy