Korean J Physiol Pharmacol.  2019 Jan;23(1):81-87. 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

Affiliations
  • 1Department of Physiology and Biophysics, School of Medicine, Eulji University, Daejeon 34824, Korea. hans0424@eulji.ac.kr, ssmin@eulji.ac.kr

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.

Keyword

Adolescence; Aggression; Anxiety; Ketamine; Maternal separation

MeSH Terms

Adolescent*
Aggression
Amnesia
Animals
Anti-Anxiety Agents
Anxiety
Arm
Cocaine
Delirium
Humans
Ketamine*
Methamphetamine
Mice*
Motor Activity
Rodentia
Weaning
Anti-Anxiety Agents
Cocaine
Ketamine
Methamphetamine

Figure

  • Fig. 1 Experimental timeline for behavioral tests.Litters were randomly assigned to maternal separation (MS) and handled (HD; control) groups. After the neonatal maternal separation procedure (PND2–20), all litters were weaned at PND22 and then separated according to gender and treatment. Adolescent mice (PND35–49) were administered a single intraperitoneal injection of a sub-anesthetic dose of ketamine (acute treatment) or a daily injection of the same dose for 1 week (chronic treatment). Open-field (OF) and elevated-plus maze (EPM) tests were performed sequentially at each mice (PND42). Resident-intruder test (RIT) was performed at the other mice (PND49). Before RIT, adolescent male mice were isolated in their cages for 2 weeks (PND35–49). At least 1 h after administration of ketamine, mice were subjected to OF, EPM test and RIT.

  • Fig. 2 Effects of maternal separation on open-field test performance in mice acutely or repeatedly treated with ketamine (15 mg/kg).(A) Distanced moved and (B) velocity were recorded in an acrylic cage (30 cm2). Results are expressed as means±SEM (HD group, n=23; MS group, n=24; **p<0.001 vs. HD+Sal, **p<0.001 vs . MS+Sal; one-way ANOVA with Tukey's post hoc test).

  • Fig. 3 Effects of maternal separation on elevated plus-maze performance in mice acutely or repeatedly treated with ketamine (15 mg/kg).Anxiety-like behavior was reflected in the percentage of time spent in open arms of the elevated plus maze. Results are expressed as means±SEM (HD group, n=22; MS group, n=26; *p<0.05, **p<0.001 vs . HD+Sal; *p<0.05, **p<0.001 vs. MS+Sal; one-way ANOVA followed by Tukey's post hoc test).

  • Fig. 4 Effects of maternal separation on performance in the resident-intruder test in mice acutely or repeatedly treated with ketamine (15 mg/kg).Aggressive-like behavior was measured as the latency to first attack (A), number of attacks (B), and cumulative attack duration (C). Results are expressed as means±SEM (HD group, n=21; MS group, n=22; *p<0.05 vs . HD+Sal, *p<0.05 vs. MS+Sal; one-way ANOVA followed by Tukey's post hoc test).


Reference

1. 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. PMID: 25719677.
2. 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. PMID: 18221626.
Article
3. Michelotti P, Quadros VA, Pereira ME, Rosemberg DB. Ketamine modulates aggressive behavior in adult zebrafish. Neurosci Lett. 2018; 684:164–168. PMID: 30102959.
Article
4. Hirota K, Lambert DG. Ketamine: its mechanism(s) of action and unusual clinical uses. Br J Anaesth. 1996; 77:441–444. PMID: 8942324.
Article
5. 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. PMID: 12969252.
Article
6. Brown JA, Ramikie TS, Schmidt MJ, Báldi R, Garbett K, Everheart MG, Warren LE, Gellért L, Horváth S, Patel S, Mirnics K. Inhibition of parvalbumin-expressing interneurons results in complex behavioral changes. Mol Psychiatry. 2015; 20:1499–1507. PMID: 25623945.
Article
7. 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. PMID: 18063801.
Article
8. 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. PMID: 2443623.
Article
9. Shin SY, Han SH, Woo RS, Jang SH, Min SS. Adolescent mice show anxiety- and aggressive-like behavior and the reduction of long-term potentiation in mossy fiber-CA3 synapses after neonatal maternal separation. Neuroscience. 2016; 316:221–231. PMID: 26733385.
Article
10. 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. PMID: 10415699.
Article
11. 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. PMID: 10692138.
Article
12. Andersen SL, Teicher MH. Delayed effects of early stress on hippocampal development. Neuropsychopharmacology. 2004; 29:1988–1993. PMID: 15316569.
Article
13. 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. PMID: 22552045.
Article
14. 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. PMID: 30102796.
Article
15. 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. PMID: 26371763.
Article
16. 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. PMID: 23623774.
Article
17. 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. PMID: 3763778.
Article
18. 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. PMID: 9785112.
Article
19. White PF, Way WL, Trevor AJ. Ketamine--its pharmacology and therapeutic uses. Anesthesiology. 1982; 56:119–136. PMID: 6892475.
Article
20. 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. PMID: 6542807.
21. McEwen BS, Albeck D, Cameron H, Chao HM, Gould E, Hastings N, Kuroda Y, Luine V, Magariños 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. PMID: 7483328.
Article
22. 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. PMID: 27106168.
Article
23. 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. PMID: 20407578.
Article
24. Magariños AM, Verdugo JM, McEwen BS. Chronic stress alters synaptic terminal structure in hippocampus. Proc Natl Acad Sci U S A. 1997; 94:14002–14008. PMID: 9391142.
25. 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. PMID: 7412479.
Article
26. 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. PMID: 30271332.
Article
27. 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. PMID: 19583895.
Article
28. Haller J, Harold G, Sandi C, Neumann ID. Effects of adverse early-life events on aggression and anti-social behaviours in animals and humans. J Neuroendocrinol. 2014; 26:724–738. PMID: 25059307.
Article
29. 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. PMID: 19176682.
Article
Full Text Links
  • KJPP
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr