Browse > Article
http://dx.doi.org/10.4196/kjpp.2019.23.1.81

Chronic administration of ketamine ameliorates the anxiety- and aggressive-like behavior in adolescent mice induced by neonatal maternal separation  

Shin, Sang Yep (Department of Physiology and Biophysics, School of Medicine, Eulji University)
Baek, Nam Jun (Department of Physiology and Biophysics, School of Medicine, Eulji University)
Han, Seung Ho (Department of Physiology and Biophysics, School of Medicine, Eulji University)
Min, Sun Seek (Department of Physiology and Biophysics, School of Medicine, Eulji University)
Publication Information
The Korean Journal of Physiology and Pharmacology / v.23, no.1, 2019 , pp. 81-87 More about this Journal
Abstract
Ketamine has long been used as an anesthetic agent. However, ketamine use is associated with numerous side effects, including flashbacks, amnesia, delirium, and aggressive or violent behavior. Ketamine has also been abused as a cocktail with ecstasy, cocaine, and methamphetamine. Several studies have investigated therapeutic applications of ketamine, demonstrating its antidepressant and anxiolytic effects in both humans and rodents. We recently reported that neonatal maternal separation causes enhanced anxiety- and aggressive-like behaviors in adolescent. In the present study, we evaluated how acute and chronic ketamine administration affected the behavioral consequences of neonatal maternal separation in adolescent mice. Litters were separated from dams for 4 hours per day for 19 days beginning after weaning. Upon reaching adolescence (post-natal day 35-49), mice were acutely (single injection) or chronically (7 daily injections) treated with a sub-anesthetic dose (15 mg/kg) of ketamine. At least 1 h after administration of ketamine, mice were subjected to open-field, elevated-plus maze, and resident-intruder tests. We found that acute ketamine treatment reduced locomotor activity. In contrast, chronic ketamine treatment decreased anxiety, as evidenced by increased time spent on open arms in the elevated-plus maze, and remarkably reduced the number and duration of attacks. In conclusion, the present study suggests that ketamine has potential for the treatment of anxiety and aggressive or violent behaviors.
Keywords
Adolescence; Aggression; Anxiety; Ketamine; Maternal separation;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Liu D, Caldji C, Sharma S, Plotsky PM, Meaney MJ. Influence of neonatal rearing conditions on stress-induced adrenocorticotropin responses and norepinepherine release in the hypothalamic paraventricular nucleus. J Neuroendocrinol. 2000;12:5-12.   DOI
2 Andersen SL, Teicher MH. Delayed effects of early stress on hippocampal development. Neuropsychopharmacology. 2004;29:1988-1993.   DOI
3 Bath KG, Chuang J, Spencer-Segal JL, Amso D, Altemus M, McEwen BS, Lee FS. Variant brain-derived neurotrophic factor (Valine66Methionine) polymorphism contributes to developmental and estrous stage-specific expression of anxiety-like behavior in female mice. Biol Psychiatry. 2012;72:499-504.   DOI
4 Chen YW, Sherpa AD, Aoki C. Single injection of ketamine during mid-adolescence promotes long-lasting resilience to activity-based anorexia of female mice by increasing food intake and attenuating hyperactivity as well as anxiety-like behavior. Int J Eat Disord. 2018;51:1020-1025.   DOI
5 Nagy LR, Featherstone RE, Hahn CG, Siegel SJ. Delayed emergence of behavioral and electrophysiological effects following juvenile ketamine exposure in mice. Transl Psychiatry. 2015;5:e635.   DOI
6 Li M, Xue X, Shao S, Shao F, Wang W. Cognitive, emotional and neurochemical effects of repeated maternal separation in adolescent rats. Brain Res. 2013;1518:82-90.   DOI
7 Harris T, Brown GW, Bifulco A. Loss of parent in childhood and adult psychiatric disorder: the role of lack of adequate parental care. Psychol Med . 1986;16:641-659.   DOI
8 Anisman H, Zaharia MD, Meaney MJ, Merali Z. Do early-life events permanently alter behavioral and hormonal responses to stressors? Int J Dev Neurosci . 1998;16:149-164.   DOI
9 White PF, Way WL, Trevor AJ. Ketamine--its pharmacology and therapeutic uses. Anesthesiology. 1982;56:119-136.   DOI
10 Takahashi RN, Morato GS, Monteiro-de-Lima TC. Effects of ketamine on experimental animal models of aggression. Braz J Med Biol Res. 1984;17:171-178.
11 Magarinos AM, Verdugo JM, McEwen BS. Chronic stress alters synaptic terminal structure in hippocampus. Proc Natl Acad Sci U S A. 1997;94:14002-14008.   DOI
12 McEwen BS, Albeck D, Cameron H, Chao HM, Gould E, Hastings N, Kuroda Y, Luine V, Magarinos AM, McKittrick CR, Orchinik M, Pavlides C, Vaher P, Watanabe Y, Weiland N. Stress and the brain: a paradoxical role for adrenal steroids. Vitam Horm. 1995;51:371-402.   DOI
13 Clarke M, Razmjou S, Prowse N, Dwyer Z, Litteljohn D, Pentz R, Anisman H, Hayley S. Ketamine modulates hippocampal neurogenesis and pro-inflammatory cytokines but not stressor induced neurochemical changes. Neuropharmacology. 2017;112:210-220.   DOI
14 Neumann ID, Veenema AH, Beiderbeck DI. Aggression and anxiety: social context and neurobiological links. Front Behav Neurosci. 2010;4:12. doi: 10.3389/fnbeh.2010.00012.   DOI
15 Baudry M, Oliver M, Creager R, Wieraszko A, Lynch G. Increase in glutamate receptors following repetitive electrical stimulation in hippocampal slices. Life Sci. 1980;27:325-330.   DOI
16 Covington HE 3rd, Newman EL, Tran S, Walton L, Hayek W, Leonard MZ, DeBold JF, Miczek KA. The urge to fight: persistent escalation by alcohol and role of NMDA receptors in mice. Front Behav Neurosci. 2018;12:206. doi: 10.3389/fnbeh.2018.00206.   DOI
17 Crawford TN, Cohen PR, Chen H, Anglin DM, Ehrensaft M. Early maternal separation and the trajectory of borderline personality disorder symptoms. Dev Psychopathol. 2009;21:1013-1030.   DOI
18 Haller J, Harold G, Sandi C, Neumann ID. Effects of adverse earlylife events on aggression and anti-social behaviours in animals and humans. J Neuroendocrinol. 2014;26:724-738.   DOI
19 David-Ferdon C, Simon TR, Spivak H, Gorman-Smith D, Savannah SB, Listenbee RL, Iskander J. Centers for Disease C, Prevention. CDC grand rounds: preventing youth violence. MMWR Morb Mortal Wkly Rep. 2015;64:171-174.
20 Kendall T, Pilling S, Tyrer P, Duggan C, Burbeck R, Meader N, Taylor C; Guideline Development Groups. Borderline and antisocial personality disorders: summary of NICE guidance. BMJ. 2009;338:b93. doi: 10.1136/bmj.693.   DOI
21 Bolshakov KV, Gmiro VE, Tikhonov DB, Magazanik LG. Determinants of trapping block of N-methyl-d-aspartate receptor channels. J Neurochem. 2003;87:56-65.   DOI
22 Maeng S, Zarate CA Jr. The role of glutamate in mood disorders: results from the ketamine in major depression study and the presumed cellular mechanism underlying its antidepressant effects. Curr Psychiatry Rep. 2007;9:467-474.   DOI
23 Michelotti P, Quadros VA, Pereira ME, Rosemberg DB. Ketamine modulates aggressive behavior in adult zebrafish. Neurosci Lett. 2018;684:164-168.   DOI
24 Hirota K, Lambert DG. Ketamine: its mechanism(s) of action and unusual clinical uses. Br J Anaesth. 1996;77:441-444.   DOI
25 Brown JA, Ramikie TS, Schmidt MJ, Baldi R, Garbett K, Everheart MG, Warren LE, Gellert L, Horvath S, Patel S, Mirnics K. Inhibition of parvalbumin-expressing interneurons results in complex behavioral changes. Mol Psychiatry. 2015;20:1499-1507.   DOI
26 Andersen SL, Lyss PJ, Dumont NL, Teicher MH. Enduring neurochemical effects of early maternal separation on limbic structures. Ann N Y Acad Sci . 1999;877:756-759.   DOI
27 Behrens MM, Ali SS, Dao DN, Lucero J, Shekhtman G, Quick KL, Dugan LL. Ketamine-induced loss of phenotype of fast-spiking interneurons is mediated by NADPH-oxidase. Science. 2007;318:1645-1647.   DOI
28 MacDonald JF, Miljkovic Z, Pennefather P. Use-dependent block of excitatory amino acid currents in cultured neurons by ketamine. J Neurophysiol. 1987;58:251-266.   DOI
29 Shin SY, Han SH, Woo RS, Jang SH, Min SS. Adolescent mice show anxiety- and aggressive-like behavior and the reduction of longterm potentiation in mossy fiber-CA3 synapses after neonatal maternal separation. Neuroscience. 2016;316:221-231.   DOI