1
|
Schwarting RKW, Wöhr M, Engler H, Sungur AÖ, Schedlowski M. Behaviorally conditioned effects of psychoactive drugs in experimental animals: What we have learned from nearly a century of research and what remains to be learned. Neurosci Biobehav Rev 2024; 162:105721. [PMID: 38754716 DOI: 10.1016/j.neubiorev.2024.105721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 04/26/2024] [Accepted: 05/10/2024] [Indexed: 05/18/2024]
Abstract
Continuous treatment with drugs is a crucial requirement for managing various clinical conditions, including chronic pain and neuropsychiatric disorders such as depression or schizophrenia. Associative learning processes, i.e. Pavlovian conditioning, can play an important role for the effects of drugs and could open new avenues for optimizing patient treatment. In this narrative literature review, we summarize available data in experimental animals regarding the behaviorally conditioned effects of psychostimulants such as d-amphetamine and cocaine, the dopamine receptor agonist apomorphine, the dopamine receptor antagonist haloperidol, morphine and antidepressant drugs. In each section, the drug under discussion is briefly introduced, followed by a detailed examination of conditioning features, including doses and dosing regimens, characteristics of the conditioning process such as test environments or specific conditioned stimuli, testing and conditioned response characteristics, possible extinction or reconditioning or reversal training, neural mechanisms, and finally, the potential clinical relevance of the research area related to the drug. We focus on key outcomes, delve into methodical issues, identify gaps in current knowledge, and suggest future research directions.
Collapse
Affiliation(s)
- Rainer K W Schwarting
- Philipps-University of Marburg, Faculty of Psychology, Experimental and Biological Psychology, Behavioral Neuroscience, Marburg D-35032, Germany; Center for Mind, Brain and Behavior, Marburg D-35032, Germany
| | - Markus Wöhr
- Philipps-University of Marburg, Faculty of Psychology, Experimental and Biological Psychology, Behavioral Neuroscience, Marburg D-35032, Germany; Center for Mind, Brain and Behavior, Marburg D-35032, Germany; KU Leuven, Faculty of Psychology and Educational Sciences, Research Unit Brain and Cognition, Laboratory of Biological Psychology, Social and Affective Neuroscience Research Group, Leuven B-3000, Belgium; KU Leuven, Leuven Brain Institute, Leuven B-3000, Belgium
| | - Harald Engler
- Institute of Medical Psychology and Behavioral Immunobiology, Center for Translational Neuro-, and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Essen D-45147, Germany
| | - A Özge Sungur
- Philipps-University of Marburg, Faculty of Psychology, Experimental and Biological Psychology, Behavioral Neuroscience, Marburg D-35032, Germany; Center for Mind, Brain and Behavior, Marburg D-35032, Germany; KU Leuven, Faculty of Psychology and Educational Sciences, Research Unit Brain and Cognition, Laboratory of Biological Psychology, Social and Affective Neuroscience Research Group, Leuven B-3000, Belgium; KU Leuven, Leuven Brain Institute, Leuven B-3000, Belgium
| | - Manfred Schedlowski
- Institute of Medical Psychology and Behavioral Immunobiology, Center for Translational Neuro-, and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Essen D-45147, Germany; Department of Clinical Neuroscience, Osher Center for Integrative Medicine, Karolinska Institutet, Stockholm 171 77, Sweden
| |
Collapse
|
2
|
Brandão AAC, Deus DLS, Duarte-Filho LAMS, Menezes PMN, Massaranduba ABR, Silva FS, Ribeiro LAA. Nebulized and intraperitoneal ketamine have equivalent antidepressant-like effect in the forced swim and tail suspension tests in mice. Pharmacol Biochem Behav 2023; 233:173674. [PMID: 37949377 DOI: 10.1016/j.pbb.2023.173674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
Major depressive disorder (MDD) is a debilitating illness that affects millions of people worldwide. Currently available antidepressants often take weeks to months to reach their full effect, which leads to an increased risk of suicidal behavior in patients with MMD. Intranasally, esketamine has emerged as an alternative to current antidepressants because of its rapid onset and long-lasting effects in patients with MDD. Animal models are useful for the initial pharmacological screening and for a better understanding of the mechanisms underlying the effects of new drugs with potential against MDD. There is a lack of data on alternative routes of drug administration, either oral or injectable, that can be used in preclinical studies. This study aimed to test whether ketamine has antidepressant-like effects in mice when administered via nebulization using a low-cost apparatus. When mice whose depressive-like behavior was induced by corticosterone were treated with nebulized ketamine at concentrations of 1.3, 2.6, and 5.2 mg/mL, immobility was reduced by 38.6 %, 62.0 %, and 61.1 %, respectively, in the forced swimming test (FST) and 43.6 %, 42.1 %, and 57.9 %, respectively, in the tail suspension test (TST). When depression-like behavior was induced by dexamethasone, nebulization with ketamine reduced immobility by 79.7 %, 49.2 %, and 44.4 % in the FST and 80.9 %, 71.4 %, and 80.4 %, respectively, in the TST. When depression-like behavior was induced by the association between dexamethasone and unpredictable chronic mild stress (UCMS) exposure, immobility was reduced by 26.1 %, 55.3 %, and 19.1 % in FST. Mice treated with nebulized ketamine did not show significant changes in the distance covered or in the time spent moving in the open field test. The efficacy of intraperitoneal and nebulized ketamine is equivalent, which shows that nebulization can be an alternative inexpensive route of drug administration for behavioral studies in rodents.
Collapse
Affiliation(s)
- Aida A C Brandão
- Curso de mestrado em biociências, Universidade Federal do Vale do São Francisco (UNIVASF), Petrolina, PE 56304-917, Brazil
| | - Deborah L S Deus
- Curso de graduação em farmácia, Universidade Federal do Vale do São Francisco (UNIVASF), Petrolina, PE 56304-917, Brazil
| | - Luiz A M S Duarte-Filho
- UMR CNRS 7266 LIENSs, Département de Biotechnologie, La Rochelle Université, La Rochelle, France
| | - Pedro M N Menezes
- Faculdade Maurício de Nassau (UNINASSAU), Av. Cardoso de Sá, 950, Cidade Universitária, Petrolina, PE CEP: 56328-020, Brazil; Faculdade de Petrolina (FACAPE), Campus Universitário, s/n, Vila Eduardo, Petrolina, PE CEP: 56328-903, Brazil
| | - Ana B R Massaranduba
- Universidade Federal do Vale do São Francisco (UNIVASF), Petrolina, PE 56304-917, Brazil
| | - Fabrício S Silva
- Colegiado de Ciências Farmacêuticas (CFARM), Programa de Pós-graduação em Biociências (PGB), Universidade Federal do Vale do São Francisco (UNIVASF), Petrolina, PE 56304-917, Brazil
| | - Luciano A A Ribeiro
- Colegiado de Ciências Farmacêuticas (CFARM), Programa de Pós-graduação em Biociências (PGB), Universidade Federal do Vale do São Francisco (UNIVASF), Petrolina, PE 56304-917, Brazil.
| |
Collapse
|
3
|
Dunham KE, Khaled KH, Weizman L, Venton BJ. Microdosing ketamine in Drosophila does not inhibit SERT like SSRIs, but causes behavioral changes mediated by glutamate and serotonin receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.07.566121. [PMID: 37986873 PMCID: PMC10659355 DOI: 10.1101/2023.11.07.566121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Recently, the FDA approved microdosing ketamine for treatment resistant depression. Traditional antidepressants, like serotonin selective reuptake inhibitors (SSRIs), block serotonin reuptake, but it is not clear if ketamine blocks serotonin reuptake. Here, we tested the effects of feeding ketamine and SSRIs to Drosophila melanogaster larvae, which has a similar serotonin system to mammals, and is a good model to track depression behaviors, such as locomotion and feeding. Fast-scan cyclic voltammetry (FSCV) was used to measure optogenetically-stimulated serotonin changes, and locomotion tracking software and blue dye feeding to monitor behavior. We fed larvae various doses (1-100 mM) of antidepressants for 24 hours and found that 1 mM ketamine did not affect serotonin, but increased locomotion and feeding. Low doses (≤ 10 mM) of escitalopram and fluoxetine inhibited dSERT and also increased feeding and locomotion behaviors. At 100 mM, ketamine inhibited dSERT and increased serotonin concentrations, but decreased locomotion and feeding due to its anesthetic properties. Since microdosing ketamine causes behavioral effects, we also investigated behavior changes with low doses of other NMDA receptor antagonists and 5-HT1A and 2 agonists, which are other possible sites for ketamine action. NMDA receptor antagonism increased feeding, while serotonin receptor agonism increased locomotion, which could explain these effects with ketamine. Ultimately, this work shows that Drosophila is a good model to discern antidepressant mechanisms, and that ketamine does not work on dSERT like SSRIs at microdoses, but affects behavior with other mechanisms.
Collapse
Affiliation(s)
- Kelly E Dunham
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, 22904, USA
| | - Kani H Khaled
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, 22904, USA
| | - Leah Weizman
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, 22904, USA
| | - B Jill Venton
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, 22904, USA
| |
Collapse
|
4
|
Godoy R, Macedo AB, Gervazio KY, Ribeiro LR, Lima JLF, Salvadori MGSS. Effects of ortho-eugenol on anxiety, working memory and oxidative stress in mice. BRAZ J BIOL 2023; 83:e271785. [PMID: 37610945 DOI: 10.1590/1519-6984.271785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 06/21/2023] [Indexed: 08/25/2023] Open
Abstract
Ortho-eugenol is a synthetic derivative from eugenol, the major compound of clove essential oil, which has demonstrated antidepressant and antinociceptive effects in pioneering studies. Additionally, its effects appear to be dependent on the noradrenergic and dopaminergic systems. Depression and anxiety disorders are known to share a great overlap in their pathophysiology, and many drugs are effective in the treatment of both diseases. Furthermore, high levels of anxiety are related to working memory deficits and increased oxidative stress. Thus, in this study we investigated the effects of acute treatment of ortho-eugenol, at 50, 75 and 100 mg/kg, on anxiety, working memory and oxidative stress in male Swiss mice. Our results show that the 100 mg/kg dose increased the number of head-dips and reduced the latency in the hole-board test. The 50 mg/kg dose reduced malondialdehyde levels in the prefrontal cortex and the number of Y-maze entries compared to the MK-801-induced hyperlocomotion group. All doses reduced nitrite levels in the hippocampus. It was also possible to assess a statistical correlation between the reduction of oxidative stress and hyperlocomotion after the administration of ortho-eugenol. However, acute treatment was not able to prevent working memory deficits. Therefore, the present study shows that ortho-eugenol has an anxiolytic and antioxidant effect, and was able to prevent substance-induced hyperlocomotion. Our results contribute to the elucidation of the pharmacological profile of ortho-eugenol, as well as to direct further studies that seek to investigate its possible clinical applications.
Collapse
Affiliation(s)
- R Godoy
- Universidade Federal da Paraíba, Instituto de Pesquisa em Fármacos e Medicamentos, Laboratório de Psicofarmacologia, João Pessoa, PB, Brasil
| | - A B Macedo
- Universidade Federal da Paraíba, Instituto de Pesquisa em Fármacos e Medicamentos, Laboratório de Psicofarmacologia, João Pessoa, PB, Brasil
| | - K Y Gervazio
- Universidade Federal da Paraíba, Instituto de Pesquisa em Fármacos e Medicamentos, Laboratório de Psicofarmacologia, João Pessoa, PB, Brasil
- Universidade Federal da Paraíba, Centro de Ciências da Saúde, Programa de Pós-graduação em Produtos Bioativos Naturais e Sintéticos - PgPNSB, João Pessoa, PB, Brasil
| | - L R Ribeiro
- Universidade Federal da Paraíba, Instituto de Pesquisa em Fármacos e Medicamentos, Laboratório de Psicofarmacologia, João Pessoa, PB, Brasil
| | - J L F Lima
- Universidade Federal da Paraíba, Instituto de Pesquisa em Fármacos e Medicamentos, Laboratório de Psicofarmacologia, João Pessoa, PB, Brasil
- Universidade Federal da Paraíba, Centro de Ciências da Saúde, Programa de Pós-graduação em Produtos Bioativos Naturais e Sintéticos - PgPNSB, João Pessoa, PB, Brasil
| | - M G S S Salvadori
- Universidade Federal da Paraíba, Instituto de Pesquisa em Fármacos e Medicamentos, Laboratório de Psicofarmacologia, João Pessoa, PB, Brasil
- Universidade Federal da Paraíba, Centro de Ciências da Saúde, Programa de Pós-graduação em Produtos Bioativos Naturais e Sintéticos - PgPNSB, João Pessoa, PB, Brasil
| |
Collapse
|
5
|
Zaytseva A, Bouckova E, Wiles MJ, Wustrau MH, Schmidt IG, Mendez-Vazquez H, Khatri L, Kim S. Ketamine's rapid antidepressant effects are mediated by Ca 2+-permeable AMPA receptors. eLife 2023; 12:e86022. [PMID: 37358072 PMCID: PMC10319435 DOI: 10.7554/elife.86022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 06/23/2023] [Indexed: 06/27/2023] Open
Abstract
Ketamine is shown to enhance excitatory synaptic drive in multiple brain areas, which is presumed to underlie its rapid antidepressant effects. Moreover, ketamine's therapeutic actions are likely mediated by enhancing neuronal Ca2+ signaling. However, ketamine is a noncompetitive NMDA receptor (NMDAR) antagonist that reduces excitatory synaptic transmission and postsynaptic Ca2+ signaling. Thus, it is a puzzling question how ketamine enhances glutamatergic and Ca2+ activity in neurons to induce rapid antidepressant effects while blocking NMDARs in the hippocampus. Here, we find that ketamine treatment in cultured mouse hippocampal neurons significantly reduces Ca2+ and calcineurin activity to elevate AMPA receptor (AMPAR) subunit GluA1 phosphorylation. This phosphorylation ultimately leads to the expression of Ca2+-Permeable, GluA2-lacking, and GluA1-containing AMPARs (CP-AMPARs). The ketamine-induced expression of CP-AMPARs enhances glutamatergic activity and glutamate receptor plasticity in cultured hippocampal neurons. Moreover, when a sub-anesthetic dose of ketamine is given to mice, it increases synaptic GluA1 levels, but not GluA2, and GluA1 phosphorylation in the hippocampus within 1 hr after treatment. These changes are likely mediated by ketamine-induced reduction of calcineurin activity in the hippocampus. Using the open field and tail suspension tests, we demonstrate that a low dose of ketamine rapidly reduces anxiety-like and depression-like behaviors in both male and female mice. However, when in vivo treatment of a CP-AMPAR antagonist abolishes the ketamine's effects on animals' behaviors. We thus discover that ketamine at the low dose promotes the expression of CP-AMPARs via reduction of calcineurin activity, which in turn enhances synaptic strength to induce rapid antidepressant actions.
Collapse
Affiliation(s)
- Anastasiya Zaytseva
- Molecular, Cellular and Integrative Neurosciences Program, Colorado State UniversityFort CollinsUnited States
| | - Evelina Bouckova
- Molecular, Cellular and Integrative Neurosciences Program, Colorado State UniversityFort CollinsUnited States
| | - McKennon J Wiles
- Molecular, Cellular and Integrative Neurosciences Program, Colorado State UniversityFort CollinsUnited States
| | - Madison H Wustrau
- Department of Biomedical Sciences, Colorado State University,Fort CollinsUnited States
| | - Isabella G Schmidt
- Molecular, Cellular and Integrative Neurosciences Program, Colorado State UniversityFort CollinsUnited States
| | | | - Latika Khatri
- Department of Cell Biology, New York University Grossman School of MedicineNew YorkUnited States
| | - Seonil Kim
- Molecular, Cellular and Integrative Neurosciences Program, Colorado State UniversityFort CollinsUnited States
- Department of Biomedical Sciences, Colorado State University,Fort CollinsUnited States
| |
Collapse
|
6
|
Chen WC, Wang TS, Chang FY, Chen PA, Chen YC. Age, Dose, and Locomotion: Decoding Vulnerability to Ketamine in C57BL/6J and BALB/c Mice. Biomedicines 2023; 11:1821. [PMID: 37509459 PMCID: PMC10376483 DOI: 10.3390/biomedicines11071821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/15/2023] [Accepted: 06/22/2023] [Indexed: 07/30/2023] Open
Abstract
Ketamine has been abused as a psychedelic agent and causes diverse neurobehavioral changes. Adolescence is a critical developmental stage but vulnerable to substances and environmental stimuli. Growing evidence shows that ketamine affects glutamatergic neurotransmission, which is important for memory storage, addiction, and psychosis. To explore diverse biological responses, this study was designed to assess ketamine sensitivity in mice of different ages and strains. Male C57BL/6J and BALB/c mice were studied in adolescence and adulthood separately. An open field test assessed motor behavioral changes. After a 30-min baseline habituation, mice were injected with ketamine (0, 25, and 50 mg/kg), and their locomotion was measured for 60 min. Following ketamine injection, the travelled distance and speed significantly increased in C57BL/6J mice between both age groups (p < 0.01), but not in BALB/c mice. The pattern of hyperlocomotion showed that mice were delayed at the higher dose (50 mg/kg) compared to the lower dose (25 mg/kg) of ketamine treatment. Ketamine accentuated locomotor activation in adolescent C57BL/6J mice compared to adults, but not in the BALB/c strain. Here, we show that ketamine-induced locomotor behavior is modulated by dose and age. The discrepancy of neurobehaviors in the two strains of mice indicates that sensitivity to ketamine is biologically determined. This study suggests that individual vulnerability to ketamine's pharmacological responses varies biologically.
Collapse
Affiliation(s)
- Wen-Chien Chen
- Department of Psychiatry, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
| | - Tzong-Shi Wang
- Department of Psychiatry, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
| | - Fang-Yu Chang
- Department of Psychiatry, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
| | - Po-An Chen
- Department of Psychiatry, China Medical University Hsinchu Hospital, China Medical University, Hsinchu 302, Taiwan
| | - Yi-Chyan Chen
- Department of Psychiatry, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
- Department of Psychiatry, School of Medicine, Tzu Chi University, Hualien 970, Taiwan
| |
Collapse
|
7
|
Bates MLS, Trujillo KA. Repeated dextromethorphan administration in adolescent rats produces long-lasting behavioral alterations. Pharmacol Biochem Behav 2023; 227-228:173581. [PMID: 37290599 DOI: 10.1016/j.pbb.2023.173581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/30/2023] [Accepted: 06/05/2023] [Indexed: 06/10/2023]
Abstract
Initiation of non-medical dextromethorphan (DXM) use often occurs in adolescence, yet little is known about the consequences when use begins during this developmental period. The current experiments examined the acute response and the effects of repeated exposure to DXM in adolescence on behavior in adulthood. We examined locomotor activity, locomotor sensitization, and cognitive function, in rats that received repeated administration of DXM. Groups of adolescent (PND 30) and adult (PND 60) male rats were treated with DXM (60 mg/kg) once daily for 10 days. Locomotor activity in response to DXM was assessed following the first injection, on the 10th day of injection (adolescent - PND 39; adult - PND 69), and following 20 days of abstinence (adolescent - PND 59; adult - PND 89). Acute locomotor effects and locomotor sensitization were compared in adolescents and adults; cross-sensitization to ketamine, another dissociative with abuse potential, was also examined. In a separate group of rodents cognitive deficits were assessed following a 20 day abstinence period (adolescent - PND 59; adult - PND 89) in spatial learning and novel object recognition tasks. The locomotor stimulant effect of DXM was much greater in adolescents than adults. Also, only adolescent rats that were repeatedly administered DXM demonstrated locomotor sensitization at the end of 10 days of injection. However, sensitization occurred after the abstinence period in all rats regardless of age. Nonetheless, cross-sensitization to ketamine was only evident in adolescent-treated rats. DXM also led to an increase in perseverative errors in reversal learning only in the adolescent-treated group. We conclude that repeated use of DXM produces long-lasting neuroadaptations that may contribute to addiction. Deficits in cognitive flexibility occur in adolescents, although further work is necessary to confirm these findings. The results extend the understanding of potential long-term consequences of DXM use in adolescents and adults.
Collapse
Affiliation(s)
- M L Shawn Bates
- Department of Psychology, California State University San Marcos, 333 S. Twin Oaks Valley Rd, San Marcos, CA 92096, USA.
| | - Keith A Trujillo
- Department of Psychology, California State University San Marcos, 333 S. Twin Oaks Valley Rd, San Marcos, CA 92096, USA.
| |
Collapse
|
8
|
Ionescu TM, Grohs-Metz G, Hengerer B. Functional ultrasound detects frequency-specific acute and delayed S-ketamine effects in the healthy mouse brain. Front Neurosci 2023; 17:1177428. [PMID: 37266546 PMCID: PMC10229773 DOI: 10.3389/fnins.2023.1177428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/21/2023] [Indexed: 06/03/2023] Open
Abstract
Introduction S-ketamine has received great interest due to both its antidepressant effects and its potential to induce psychosis when administered subchronically. However, no studies have investigated both its acute and delayed effects using in vivo small-animal imaging. Recently, functional ultrasound (fUS) has emerged as a powerful alternative to functional magnetic resonance imaging (fMRI), outperforming it in sensitivity and in spatiotemporal resolution. In this study, we employed fUS to thoroughly characterize acute and delayed S-ketamine effects on functional connectivity (FC) within the same cohort at slow frequency bands ranging from 0.01 to 1.25 Hz, previously reported to exhibit FC. Methods We acquired fUS in a total of 16 healthy C57/Bl6 mice split in two cohorts (n = 8 received saline, n = 8 S-ketamine). One day after the first scans, performed at rest, the mice received the first dose of S-ketamine during the second measurement, followed by four further doses administered every 2 days. First, we assessed FC reproducibility and reliability at baseline in six frequency bands. Then, we investigated the acute and delayed effects at day 1 after the first dose and at day 9, 1 day after the last dose, for all bands, resulting in a total of four fUS measurements for every mouse. Results We found reproducible (r > 0.9) and reliable (r > 0.9) group-average readouts in all frequency bands, only the 0.01-0.27 Hz band performing slightly worse. Acutely, S-ketamine induced strong FC increases in five of the six bands, peaking in the 0.073-0.2 Hz band. These increases comprised both cortical and subcortical brain areas, yet were of a transient nature, FC almost returning to baseline levels towards the end of the scan. Intriguingly, we observed robust corticostriatal FC decreases in the fastest band acquired (0.75 Hz-1.25 Hz). These changes persisted to a weaker extent after 1 day and at this timepoint they were accompanied by decreases in the other five bands as well. After 9 days, the decreases in the 0.75-1.25 Hz band were maintained, however no changes between cohorts could be detected in any other bands. Discussion In summary, the study reports that acute and delayed ketamine effects in mice are not only dissimilar but have different directionalities in most frequency bands. The complementary readouts of the employed frequency bands recommend the use of fUS for frequency-specific investigation of pharmacological effects on FC.
Collapse
|
9
|
Kim A, Gu SM, Lee H, Kim DE, Hong JT, Yun J, Cha HJ. Prenatal ketamine exposure impairs prepulse inhibition via arginine vasopressin receptor 1A-mediated GABAergic neuronal dysfunction in the striatum. Biomed Pharmacother 2023; 160:114318. [PMID: 36738499 DOI: 10.1016/j.biopha.2023.114318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 01/17/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023] Open
Abstract
Ketamine is a widely used anesthetic with N-methyl-D-aspartate (NMDA) receptor antagonism. Exposure to ketamine and NMDA receptor antagonists may induce psychosis. However, the mechanism underlying the effects of ketamine on the immature brain remains unclear. In this study, NMDA receptor antagonists, ketamine and methoxetamine, were administered to pregnant F344 rats (E17). These regimens induce psychosis-like behaviors in the offspring, such as hyperlocomotion induced by MK-801, a non-competitive NMDA receptor antagonist. We also observed that prepulse inhibition (PPI) was significantly reduced. Interestingly, ketamine administration increased the arginine vasopressin receptor 1A (Avpr1a) expression levels in the striatum of offspring with abnormal behaviors. Methoxetamine, another NMDA receptor antagonist, also showed similar results. In addition, we demonstrated a viral vector-induced Avpr1a overexpression in the striatum-inhibited PPI. In the striatum of offspring, ketamine or methoxetamine treatment increased glutamate decarboxylase 67 (GAD67) and δ-aminobutyric acid (GABA) levels. These results show that prenatal NMDA receptor antagonist treatment induces GABAergic neuronal dysfunction and abnormalities in sensorimotor gating via regulating Avpr1a expression in the striatum.
Collapse
Affiliation(s)
- Aeseul Kim
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, the Republic of Korea
| | - Sun Mi Gu
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, the Republic of Korea
| | - Haemiru Lee
- Pharmacological Research Division, National Institute of Food and Drug Safety Evaluation (NIFDS), Ministry of Food and Drug Safety (MFDS), OHTAC 187, Osongsaengmyong 2-ro, Cheongju-si, Chungcheongbuk-do 28159, the Republic of Korea
| | - Dong Eun Kim
- Pharmacological Research Division, National Institute of Food and Drug Safety Evaluation (NIFDS), Ministry of Food and Drug Safety (MFDS), OHTAC 187, Osongsaengmyong 2-ro, Cheongju-si, Chungcheongbuk-do 28159, the Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, the Republic of Korea
| | - Jaesuk Yun
- College of Pharmacy, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, the Republic of Korea.
| | - Hye Jin Cha
- College of Veterinary Medicine, Gyeongsang National University, 501, Jinju-daero, Jinju-si, Gyeongsangnam-do 52828, the Republic of Korea.
| |
Collapse
|
10
|
Ma X, Peng J, Chen Y, Wang Z, Zhou Q, Yan J, Jiang H. Esketamine Anesthetizes Mice With a Similar Potency to Racemic Ketamine. Dose Response 2023; 21:15593258231157563. [PMID: 36798635 PMCID: PMC9926386 DOI: 10.1177/15593258231157563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/27/2023] [Indexed: 02/13/2023] Open
Abstract
Esketamine, the right-handed optical isomer of racemic ketamine, has recently become widely used for anesthesia and analgesia as a replacement for racemic ketamine. However, there are limited studies comparing the anesthetic and analgesic effects of esketamine and racemic ketamine in mice. This research was conducted to analyze the dose-dependent anesthetic and analgesic efficacy of esketamine in mice and to compare its potency with that of the racemate. We tested the anesthetic effects of different doses of esketamine and compared its potency with that of the racemate using righting reflex tests. Then, the acetic acid-induced pain model and formalin-induced pain model were used to investigate the analgesic effect. Compared with racemic ketamine, an equivalent dose of esketamine at 100 mg/kg was required to induce stable anesthesia. In contrast, 5 mg/kg esketamine was sufficient to provide analgesic effects similar to those of 10 mg/kg ketamine. Together, esketamine had a similar potency to racemic ketamine for anesthesia and a stronger potency for analgesia in mice.
Collapse
Affiliation(s)
- Xiaofan Ma
- Department of Anesthesiology, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People’s Hospital, Shanghai, China
| | - Jiali Peng
- Department of Anesthesiology, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People’s Hospital, Shanghai, China
| | - Yelin Chen
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Zeyi Wang
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Qiang Zhou
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Jia Yan
- Department of Anesthesiology, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People’s Hospital, Shanghai, China,Jia Yan, Department of Anesthesiology, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People’s Hospital, No. 639, Zhizaoju Road, Shanghai 200011, China.
| | - Hong Jiang
- Department of Anesthesiology, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People’s Hospital, Shanghai, China,Hong Jiang, Department of Anesthesiology, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People's Hospital, No. 639, Zhizaoju Road, Shanghai, China.
| |
Collapse
|
11
|
Ain QU, Saleem U, Ahmad B, Khalid I. Pharmacological screening of silibinin for antischizophrenic activity along with its acute toxicity evaluation in experimental animals. Front Pharmacol 2023; 14:1111915. [PMID: 36817163 PMCID: PMC9936411 DOI: 10.3389/fphar.2023.1111915] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/11/2023] [Indexed: 02/05/2023] Open
Abstract
Silibinin (SIL), a flavolignan extracted from the medicinal plant "silybum marianum (milk thistle)", has traditionally been used to treat liver disease. This phytochemical has displayed neuroprotective properties, its activity against schizophrenia is not elucidated. The present study was designed to evaluate the antipsychotic potential of silibinin and probe its toxic potential. The acute oral toxicity study was assessed as per OECD 425 guidelines. Animals were divided into two groups of female rats (n = 6): one group served as the normal control and the other group received a 2,000 mg/kg dose of SIL. We also evaluated the antipsychotic potential of SIL. To this end, animals were divided into six groups (n = 10) of mice for both the preventive and curative protocols. Group I (CMC 1 mL/kg) served as the normal control and received CMC 1 mL/kg; group II was the diseased group treated with ketamine (10 mg/kg) i.p; group III was the standard group treated with clozapine 1 mg/kg; groups IV, V, and VI served as the treatment groups, receiving SIL 50, 100, and 200 mg/kg, respectively, orally for both protocols. Improvement in positive symptoms of the disease was evaluated by stereotypy and hyperlocomotion, while negative symptoms (behavioral despair) were determined by a forced swim test and a tail suspension test in the mice models. The results suggested that the LD50 of SIL was greater than 2,000 mg/kg. Moreover, SIL prevented and reversed ketamine-induced increase in stereotypy (p < 0.001) and behavioral despair in the forced swim and tail suspension tests (p < 0.001). Taken together, the findings suggest that silibinin is a safe drug with low toxicity which demonstrates significant antipsychotic activity against the positive and negative symptoms of schizophrenia.
Collapse
Affiliation(s)
- Qurat Ul Ain
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan,*Correspondence: Qurat Ul Ain, ; Uzma Saleem,
| | - Uzma Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan,*Correspondence: Qurat Ul Ain, ; Uzma Saleem,
| | - Bashir Ahmad
- Hamza College of Pharmaceutical and Allied Health Sciences, Lahore, Pakistan
| | - Iqra Khalid
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| |
Collapse
|
12
|
The α 2C-adrenoceptor antagonist JP-1302 controls behavioral parameters, tyrosine hydroxylase activity and receptor expression in a rat model of ketamine-induced schizophrenia-like deficits. Pharmacol Biochem Behav 2022; 221:173490. [PMID: 36379444 DOI: 10.1016/j.pbb.2022.173490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/08/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
Schizophrenia is a chronic disabling disease affecting 1 % of the population. Current antipsychotics have limited efficacy in mitigating the severity of the symptoms of the disease. Therefore, searching for new therapeutic targets is essential. Previous studies have shown that α2C-adrenoceptor antagonists may have antipsychotic and pro-cognitive effects. Therefore, the current study evaluates the behavioral and neurochemical effects of JP-1302, a selective α2C-adrenoceptor antagonist, in a model of schizophrenia-like deficits induced by sub-chronic ketamine (KET) administration. Here, we administered ketamine (25 mg/kg, i.p.) to male and female Wistar rats for eight consecutive days. On the last two days of ketamine administration, rats were pretreated with either JP-1302 (1-3-10 μmol/kg, i.p.), chlorpromazine (0.1 mg/kg, i.p.), or saline, and the behavioral tests were performed. Behaviors related to positive (locomotor activity), negative (social interaction), and cognitive (novel object recognition) symptoms of schizophrenia were assessed. Glutamate, glutamine, GABA levels, and α2C-adrenoceptor expression were measured in the frontal cortex and the hippocampus. Tyrosine hydroxylase immunocytochemical reactivity was also shown in the midbrain regions. Sub-chronic ketamine administration increased locomotor activity and produced robust social interaction and object recognition deficits, and JP-1302 significantly ameliorated ketamine-induced cognitive deficits. Ketamine induced a hyperdopaminergic activity in the striatum, which was reversed by the treatment with JP-1302. Also, the α2C-adrenoceptor expression was higher in the frontal cortex and hippocampus in the ketamine-treated rats. Our findings confirm that α2C-adrenoceptor antagonism may be a potential drug target for treating cognitive disorders related to schizophrenia.
Collapse
|
13
|
Porras-Dávila SL, Jiménez-Ferrer E, Román Ramos R, González-Cortazar M, Almanza-Pérez JC, Herrera-Ruiz M. Herniarin, Dimethylfraxetin and Extracts from Tagetes lucida, in Psychosis Secondary to Ketamine and Its Interaction with Haloperidol. PLANTS (BASEL, SWITZERLAND) 2022; 11:2789. [PMID: 36297813 PMCID: PMC9610474 DOI: 10.3390/plants11202789] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 06/16/2023]
Abstract
Tagetes lucida Cav., is a medicinal plant used in Mexico to alleviate different disorders related to alterations of the central nervous system, such as behaviors associated with psychosis. The present work evaluated the effect of different extracts separated from this plant, TlHex, TlEA, TlMet, and TlAq, and of two isolated coumarins, herniarin (HN) and dimethylfraxetin (DF), on haloperidol-induced catalepsy (HAL), and psychotic behaviors provoked with a glutamatergic antagonist, ketamine (KET) on ICR mice. The extracts TlEA, TlAq, and the isolated compounds HN and DF, induced an increment of the cataleptic effect of HAL. Schizophrenia-like symptoms caused by KET were analyzed through the behavior of the animals in the open field (OFT), forced swimming (FST), passive avoidance test (PAT), and social interaction test (SIT). Treatments derived from T. lucida could interact with this substance in all tests except for FST, in which only TlMet blocks its activity. Mainly, TlEA, TlAq, HN, and DF, blocked the effects of KET on stereotyped behavior, hyperlocomotion, cognitive impairment, and detriment in the social interaction of rodents. T. lucida interacted with dopaminergic and glutamatergic systems.
Collapse
Affiliation(s)
- Sandra Liliana Porras-Dávila
- Centro de Investigación Biomédica del Sur, Instituto Mexicano Del Seguro Social, Argentina No. 1, Col. Centro, Xochitepec 62790, Morelos, Mexico
- Doctorado en Ciencias Biológicas y de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana (UAM), Ciudad de México 04960, Mexico
| | - Enrique Jiménez-Ferrer
- Centro de Investigación Biomédica del Sur, Instituto Mexicano Del Seguro Social, Argentina No. 1, Col. Centro, Xochitepec 62790, Morelos, Mexico
| | - Rubén Román Ramos
- Doctorado en Ciencias Biológicas y de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana (UAM), Ciudad de México 04960, Mexico
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Av. Ferrocarril San Rafael Atlixco 186, Leyes de Reforma 1era Secc., Ciudad de México 09310, Mexico
| | - Manasés González-Cortazar
- Centro de Investigación Biomédica del Sur, Instituto Mexicano Del Seguro Social, Argentina No. 1, Col. Centro, Xochitepec 62790, Morelos, Mexico
| | - Julio César Almanza-Pérez
- Doctorado en Ciencias Biológicas y de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana (UAM), Ciudad de México 04960, Mexico
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Av. Ferrocarril San Rafael Atlixco 186, Leyes de Reforma 1era Secc., Ciudad de México 09310, Mexico
| | - Maribel Herrera-Ruiz
- Centro de Investigación Biomédica del Sur, Instituto Mexicano Del Seguro Social, Argentina No. 1, Col. Centro, Xochitepec 62790, Morelos, Mexico
| |
Collapse
|
14
|
Monoamine Neurotransmitters Control Basic Emotions and Affect Major Depressive Disorders. Pharmaceuticals (Basel) 2022; 15:ph15101203. [PMID: 36297314 PMCID: PMC9611768 DOI: 10.3390/ph15101203] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/16/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022] Open
Abstract
Major depressive disorder (MDD) is a common and complex mental disorder, that adversely impacts an individual’s quality of life, but its diagnosis and treatment are not accurately executed and a symptom-based approach is utilized in most cases, due to the lack of precise knowledge regarding the pathophysiology. So far, the first-line treatments are still based on monoamine neurotransmitters. Even though there is a lot of progress in this field, the mechanisms seem to get more and more confusing, and the treatment is also getting more and more controversial. In this study, we try to review the broad advances of monoamine neurotransmitters in the field of MDD, and update its effects in many advanced neuroscience studies. We still propose the monoamine hypothesis but paid special attention to their effects on the new pathways for MDD, such as inflammation, oxidative stress, neurotrophins, and neurogenesis, especially in the glial cells, which have recently been found to play an important role in many neurodegenerative disorders, including MDD. In addition, we will extend the monoamine hypothesis to basic emotions; as suggested in our previous reports, the three monoamine neurotransmitters play different roles in emotions: dopamine—joy, norepinephrine—fear (anger), serotonins—disgust (sadness). Above all, this paper tries to give a full picture of the relationship between the MDD and the monoamine neurotransmitters such as DA, NE, and 5-HT, as well as their contributions to the Three Primary Color Model of Basic Emotions (joy, fear, and disgust). This is done by explaining the contribution of the monoamine from many sides for MDD, such the digestive tract, astrocytes, microglial, and others, and very briefly addressing the potential of monoamine neurotransmitters as a therapeutic approach for MDD patients and also the reasons for its limited clinical efficacy, side effects, and delayed onset of action. We hope this review might offer new pharmacological management of MDD.
Collapse
|
15
|
Viktorov M, Wilkinson MP, Elston VCE, Stone M, Robinson ESJ. A systematic review of studies investigating the acute effects of N-methyl- D-aspartate receptor antagonists on behavioural despair in normal animals suggests poor predictive validity. Brain Neurosci Adv 2022; 6:23982128221081645. [PMID: 35299619 PMCID: PMC8922211 DOI: 10.1177/23982128221081645] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 02/02/2022] [Indexed: 12/16/2022] Open
Abstract
The ability of the N-methyl-D-aspartate receptor antagonist ketamine to induce a rapid and sustained antidepressant effect has led to a surge in pre-clinical studies investigating underlying mechanisms and seeking novel treatments. Animal models are key to this research as they can provide a behavioural readout linking underlying mechanisms to clinical benefits. However, quantifying depression-related behaviours in rodents represents a major challenge with the validity of traditional methods such as models of behavioural despair (forced swim test and tail suspension test) a topic of debate. While there is good evidence to support the value of using these behavioural readouts to study the effects of stress, these approaches have largely failed to detect reliable phenotypic effects in other disease models. In this systematic review, we identified publications which had tested N-methyl-D-aspartate receptor antagonists in normal animals using either the forced swim test or tail suspension test. We compared findings for different doses and time points and also drugs with different clinical profiles to investigate how well the outcomes in the rodent model predicted their effects in the clinic. Despite clear evidence that N-methyl-D-aspartate receptor antagonists reduce immobility time and hence exhibit an antidepressant profile in these tasks, we found similar effects with both clinically effective drugs as well as those which have failed to show efficacy in clinical trials. These findings suggest that behavioural despair tests in normal animals do not provide a good method to predict clinical efficacy of N-methyl-D-aspartate receptor antagonists.
Collapse
Affiliation(s)
- Martin Viktorov
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| | - Matthew P. Wilkinson
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| | - Victoria C. E. Elston
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| | - Medi Stone
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| | - Emma S. J. Robinson
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| |
Collapse
|
16
|
Neurobehavioral Differences of Valproate and Risperidone on MK-801 Inducing Acute Hyperlocomotion in Mice. Behav Neurol 2022; 2022:1048463. [PMID: 35251367 PMCID: PMC8890888 DOI: 10.1155/2022/1048463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/23/2022] [Accepted: 02/07/2022] [Indexed: 11/17/2022] Open
Abstract
Objective The glutamate system plays a major role in the development of neuropsychiatric disorders such as addiction, epilepsy, dementia, and psychosis. MK-801 (dizocilpine), an uncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist, could increase locomotor activity and stereotyped neurobehaviors mimicking schizophrenic-like features in the mouse model. The study would explore the neuropharmacological differences of risperidone and valproic acid on the MK-801-induced neurobehavioral changes. Methods The subjects were male C57BL/6J mice obtained from the National Laboratory Animal Center. Drug effects were assessed using the open field with a video-tracking system and gaiting tests. After habitation, risperidone (0, 0.1 mg/kg) or valproic acid (0, 200 mg/kg) was injected and ran locomotion for 30 mins. Sequentially, mice were followed by intraperitoneal injection (i.p.) with MK-801 (0, 0.2 mg/kg) and ran locomotion for 60 mins. Gaiting behaviors such as step angles, stride lengths, and stance widths were measured following the study drugs. Results The results showed that risperidone and valproic acid alone could not alter the locomotor activities. Following the MK-801 injection, the travelled distance and speed in the entire open field dramatically increased. The dose 0.1 mg/kg of risperidone could totally inhibit the MK-801-induced hyperlocomotion compared with that of the saline-injected group (p < 0.001). The valproic acid (200 mg/kg) partially suppressed the hyperlocomotion which is induced by MK801. Conclusion The more dominant effect of risperidone to rescue MK-801 induced hyperlocomotion compared with that of valproic acid. The partial suppression of valproic acid may imply the psychopharmacological evidence as adjuvant effect to treat psychotic patients through tuning glutamatergic neurotransmission.
Collapse
|
17
|
Han DH, Hong I, Choi JE, Park P, Baek JY, Park H, Ide S, Mishina M, Ikeda K, Kaang BK. Abolished ketamine effects on the spontaneous excitatory postsynaptic current of medial prefrontal cortex neurons in GluN2D knockout mice. Mol Brain 2021; 14:174. [PMID: 34876180 PMCID: PMC8650376 DOI: 10.1186/s13041-021-00883-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/19/2021] [Indexed: 12/02/2022] Open
Abstract
Ketamine, a non-competitive antagonist of the N-methyl-d-aspartate receptor (NMDAR), generates a rapidly-acting antidepressant effect. It exerts psychomimetic effects, yet demands a further investigation of its mechanism. Previous research showed that ketamine did no longer promote hyperlocomotion in GluN2D knockout (KO) mice, which is a subunit of NMDAR. In the present study, we tested whether GluN2D-containing NMDARs participate in the physiological changes in the medial prefrontal cortex (mPFC) triggered by ketamine. Sub-anesthetic dose of ketamine (25 mg/kg) elevated the frequency of spontaneous excitatory postsynaptic currents (sEPSC) in wild-type (WT) mice, but not in GluN2D KO mice, 1 h after the injection. The amplitude of sEPSC and paired-pulse ratio (PPR) were unaltered by ketamine in both WT and GluN2D KO mice. These findings suggest that GluN2D-containing NMDARs might play a role in the ketamine-mediated changes in glutamatergic neurons in mPFC and, presumably, in ketamine-induced hyperlocomotion.
Collapse
Affiliation(s)
- Dae Hee Han
- School of Biological Sciences, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Ilgang Hong
- School of Biological Sciences, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Ja Eun Choi
- School of Biological Sciences, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Pojeong Park
- School of Biological Sciences, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Jun-Yeong Baek
- School of Biological Sciences, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - HyoJin Park
- School of Biological Sciences, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Soichiro Ide
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2- 1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
| | - Masayoshi Mishina
- Brain Science Laboratory, The Research Organization of Science and Technology, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
| | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2- 1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan.
| | - Bong-Kiun Kaang
- School of Biological Sciences, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea.
| |
Collapse
|
18
|
de Araújo FYR, Chaves Filho AJM, Nunes AM, de Oliveira GV, Gomes PXL, Vasconcelos GS, Carletti J, de Moraes MO, de Moraes ME, Vasconcelos SMM, de Sousa FCF, de Lucena DF, Macedo DS. Involvement of anti-inflammatory, antioxidant, and BDNF up-regulating properties in the antipsychotic-like effect of the essential oil of Alpinia zerumbet in mice: a comparative study with olanzapine. Metab Brain Dis 2021; 36:2283-2297. [PMID: 34491479 DOI: 10.1007/s11011-021-00821-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 08/14/2021] [Indexed: 12/30/2022]
Abstract
The current drug therapy for schizophrenia effectively treats acute psychosis and its recurrence; however, this mental disorder's cognitive and negative symptoms are still poorly controlled. Antipsychotics present important side effects, such as weight gain and extrapyramidal effects. The essential oil of Alpinia zerumbet (EOAZ) leaves presents potential antipsychotic properties that need further preclinical investigation. Here, we determined EAOZ effects in preventing and reversing schizophrenia-like symptoms (positive, negative, and cognitive) induced by ketamine (KET) repeated administration in mice and putative neurobiological mechanisms related to this effect. We conducted the behavioral evaluations of prepulse inhibition of the startle reflex (PPI), social interaction, and working memory (Y-maze task), and verified antioxidant (GSH, nitrite levels), anti-inflammatory [interleukin (IL)-6], and neurotrophic [brain-derived neurotrophic factor (BDNF)] effects of this oil in hippocampal tissue. The atypical antipsychotic olanzapine (OLZ) was used as standard drug therapy. EOAZ, similarly to OLZ, prevented and reversed most KET-induced schizophrenia-like behavioral alterations, i.e., sensorimotor gating deficits and social impairment. EOAZ had a modest effect on the prevention of KET-associated working memory deficit. Compared to OLZ, EOAZ showed a more favorable side effects profile, inducing less cataleptic and weight gain changes. EOAZ efficiently protected the hippocampus against KET-induced oxidative imbalance, IL-6 increments, and BDNF impairment. In conclusion, our data add more mechanistic evidence for the anti-schizophrenia effects of EOAZ, based on its antioxidant, anti-inflammatory, and BDNF up-regulating actions. The absence of significant side effects observed in current antipsychotic drug therapy seems to be an essential benefit of the oil.
Collapse
Affiliation(s)
- Fernanda Yvelize Ramos de Araújo
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Rua Cel. Nunes de Melo, 1000, 60431-270, Fortaleza, CE, Brazil
| | - Adriano José Maia Chaves Filho
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Rua Cel. Nunes de Melo, 1000, 60431-270, Fortaleza, CE, Brazil
| | - Adriana Mary Nunes
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Rua Cel. Nunes de Melo, 1000, 60431-270, Fortaleza, CE, Brazil
| | - Gersilene Valente de Oliveira
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Rua Cel. Nunes de Melo, 1000, 60431-270, Fortaleza, CE, Brazil
| | - Patrícia Xavier Lima Gomes
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Rua Cel. Nunes de Melo, 1000, 60431-270, Fortaleza, CE, Brazil
| | - Germana Silva Vasconcelos
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Rua Cel. Nunes de Melo, 1000, 60431-270, Fortaleza, CE, Brazil
| | - Jaqueline Carletti
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Rua Cel. Nunes de Melo, 1000, 60431-270, Fortaleza, CE, Brazil
| | - Manoel Odorico de Moraes
- Laboratory of Experimental Oncology, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, Ceará, Brazil
| | - Maria Elisabete de Moraes
- Clinical Pharmacology Unit, Drug Research and Development Center, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Silvânia Maria Mendes Vasconcelos
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Rua Cel. Nunes de Melo, 1000, 60431-270, Fortaleza, CE, Brazil
| | - Francisca Cléa Florenço de Sousa
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Rua Cel. Nunes de Melo, 1000, 60431-270, Fortaleza, CE, Brazil
| | - David Freitas de Lucena
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Rua Cel. Nunes de Melo, 1000, 60431-270, Fortaleza, CE, Brazil
| | - Danielle S Macedo
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Rua Cel. Nunes de Melo, 1000, 60431-270, Fortaleza, CE, Brazil.
- National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, SP, Brazil.
| |
Collapse
|
19
|
Ketamine for psychotic depression: An overview of the glutamatergic system and ketamine's mechanisms associated with antidepressant and psychotomimetic effects. Psychiatry Res 2021; 306:114231. [PMID: 34798487 DOI: 10.1016/j.psychres.2021.114231] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/07/2021] [Accepted: 09/14/2021] [Indexed: 02/08/2023]
Abstract
Approximately 0.35-1% of the general population is afflicted with psychotic depression at some time in their life. Psychotic depression is a subtype of major depressive disorder characterized by mood congruent hallucinations and/or delusions. Patients with psychotic depression often represent the most severe cases, with high relapse and mortality rate. Although treatment guidelines recommend a combination of antidepressants and antipsychotics or electroconvulsive therapy, most patients subsequently relapse due to treatment resistance. Furthermore, with the concern of antipsychotic drug's side effects (e.g., tardive dyskinesia), there is a need for an alternative pharmacotherapy for psychotic depression. Recently, several case studies demonstrated that treatment with ketamine not only ameliorated mood, but also improved psychotic symptoms in patients with treatment-resistant depression and psychotic features. However, the safety of ketamine in these patients is controversial since ketamine is known to induce psychotomimetic and dissociative effects. Additionally, the efficacy and safety of ketamine in patients with psychotic depression has not been established as most clinical trials have excluded these persons due to the theorized risk of aggravating psychotic symptoms. Notwithstanding, it is not established empirically that ketamine treatment in psychotic depression would predictably amplify psychotic symptoms and/or overall illness presentation. Future trials evaluating ketamine in depression should include patients with psychotic features to inform whether ketamine is safe and effective in this subpopulation.
Collapse
|
20
|
Ketamine increases activity of a fronto-striatal projection that regulates compulsive behavior in SAPAP3 knockout mice. Nat Commun 2021; 12:6040. [PMID: 34654803 PMCID: PMC8519915 DOI: 10.1038/s41467-021-26247-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/17/2021] [Indexed: 12/03/2022] Open
Abstract
Obsessive-Compulsive Disorder (OCD), characterized by intrusive thoughts (obsessions) and repetitive behaviors (compulsions), is associated with dysfunction in fronto-striatal circuits. There are currently no fast-acting pharmacological treatments for OCD. However, recent clinical studies demonstrated that an intravenous infusion of ketamine rapidly reduces OCD symptoms. To probe mechanisms underlying ketamine’s therapeutic effect on OCD-like behaviors, we used the SAPAP3 knockout (KO) mouse model of compulsive grooming. Here we recapitulate the fast-acting therapeutic effect of ketamine on compulsive behavior, and show that ketamine increases activity of dorsomedial prefrontal neurons projecting to the dorsomedial striatum in KO mice. Optogenetically mimicking this increase in fronto-striatal activity reduced compulsive grooming behavior in KO mice. Conversely, inhibiting this circuit in wild-type mice increased grooming. Finally, we demonstrate that ketamine blocks the exacerbation of grooming in KO mice caused by optogenetically inhibiting fronto-striatal activity. These studies demonstrate that ketamine increases activity in a fronto-striatal circuit that causally controls compulsive grooming behavior, suggesting this circuit may be important for ketamine’s therapeutic effects in OCD. Intravenous infusion of ketamine rapidly reduces obsessive-compulsive disorder symptoms. Here, the authors show in mice that ketamine acts by increasing activity in a fronto-striatal circuit that causally controls compulsive grooming behaviour.
Collapse
|
21
|
Štefková-Mazochová K, Danda H, Dehaen W, Jurásek B, Šíchová K, Pinterová-Leca N, Mazoch V, Krausová BH, Kysilov B, Smejkalová T, Vyklický L, Kohout M, Hájková K, Svozil D, Horsley RR, Kuchař M, Páleníček T. Pharmacokinetic, pharmacodynamic, and behavioural studies of deschloroketamine in Wistar rats. Br J Pharmacol 2021; 179:65-83. [PMID: 34519023 DOI: 10.1111/bph.15680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Deschloroketamine (DCK), a structural analogue of ketamine, has recently emerged on the illicit drug market as a recreational drug with a modestly long duration of action. Despite it being widely used by recreational users, no systematic research on its effects has been performed to date. EXPERIMENTAL APPROACH Pharmacokinetics, acute effects, and addictive potential in a series of behavioural tests in Wistar rats were performed following subcutaneous (s.c.) administration of DCK (5, 10, and 30 mg·kg-1 ) and its enantiomers S-DCK (10 mg·kg-1 ) and R-DCK (10 mg·kg-1 ). Additionally, activity at human N-methyl-d-aspartate (NMDA) receptors was also evaluated. KEY RESULTS DCK rapidly crossed the blood brain barrier, with maximum brain levels achieved at 30 min and remaining high at 2 h after administration. Its antagonist activity at NMDA receptors is comparable to that of ketamine with S-DCK being more potent. DCK had stimulatory effects on locomotion, induced place preference, and robustly disrupted PPI. Locomotor stimulant effects tended to disappear more quickly than disruptive effects on PPI. S-DCK had more pronounced stimulatory properties than its R-enantiomer. However, the potency in disrupting PPI was comparable in both enantiomers. CONCLUSION AND IMPLICATIONS DCK showed similar behavioural and addictive profiles and pharmacodynamics to ketamine, with S-DCK being in general more active. It has a slightly slower pharmacokinetic profile than ketamine, which is consistent with its reported longer duration of action. These findings have implications and significance for understanding the risks associated with illicit use of DCK.
Collapse
Affiliation(s)
| | - Hynek Danda
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,3rd Faculty of Medicine, Charles University, Prague 10, Czech Republic
| | - Wim Dehaen
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,CZ-OPENSCREEN: National Infrastructure for Chemical Biology, Department of Informatics and Chemistry, Faculty of Chemical Technology, University of Chemistry and Technology, Prague 6, Czech Republic
| | - Bronislav Jurásek
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Forensic Laboratory of Biologically Active Compounds, Department of Chemistry of Natural Compounds, University of Chemistry and Technology, Prague 6, Czech Republic
| | - Klára Šíchová
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic
| | - Nikola Pinterová-Leca
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,3rd Faculty of Medicine, Charles University, Prague 10, Czech Republic
| | - Vladimír Mazoch
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic
| | - Barbora Hrčka Krausová
- Department of Cellular Neurophysiology, Institute of Physiology, CAS, Prague 4, Czech Republic
| | - Bohdan Kysilov
- Department of Cellular Neurophysiology, Institute of Physiology, CAS, Prague 4, Czech Republic
| | - Tereza Smejkalová
- Department of Cellular Neurophysiology, Institute of Physiology, CAS, Prague 4, Czech Republic
| | - Ladislav Vyklický
- Department of Cellular Neurophysiology, Institute of Physiology, CAS, Prague 4, Czech Republic
| | - Michal Kohout
- Department of Organic Chemistry, University of Chemistry and Technology, Prague 6, Czech Republic
| | - Kateřina Hájková
- Forensic Laboratory of Biologically Active Compounds, Department of Chemistry of Natural Compounds, University of Chemistry and Technology, Prague 6, Czech Republic.,Department of Analytical Chemistry, University of Chemistry and Technology, Prague 6, Czech Republic
| | - Daniel Svozil
- CZ-OPENSCREEN: National Infrastructure for Chemical Biology, Department of Informatics and Chemistry, Faculty of Chemical Technology, University of Chemistry and Technology, Prague 6, Czech Republic.,CZ-OPENSCREEN: National Infrastructure for Chemical Biology, Institute of Molecular Genetics, CAS, Prague 4, Czech Republic
| | - Rachel R Horsley
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic
| | - Martin Kuchař
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Forensic Laboratory of Biologically Active Compounds, Department of Chemistry of Natural Compounds, University of Chemistry and Technology, Prague 6, Czech Republic
| | - Tomáš Páleníček
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,3rd Faculty of Medicine, Charles University, Prague 10, Czech Republic
| |
Collapse
|
22
|
Zhang F, Hillhouse TM, Anderson PM, Koppenhaver PO, Kegen TN, Manicka SG, Lane JT, Pottanat E, Van Fossen M, Rice R, Porter JH. Opioid receptor system contributes to the acute and sustained antidepressant-like effects, but not the hyperactivity motor effects of ketamine in mice. Pharmacol Biochem Behav 2021; 208:173228. [PMID: 34224734 DOI: 10.1016/j.pbb.2021.173228] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 11/24/2022]
Abstract
In 2000, a subanesthetic dose (0.5 mg/kg i.v.) of the dissociative anesthetic ketamine was reported to have both rapid and robust antidepressant effects in patients diagnosed with major depressive disorder and later, ketamine also was shown to be effective in treatment-resistant depressed patients. However, the mechanisms responsible for ketamine's antidepressant effects remain unclear. In 2018, a clinical study reported that pretreatment with the nonselective opioid antagonist naltrexone attenuated the rapid antidepressant effect of ketamine in depressed patients. The current study investigated the potential role of the opioid receptor system in the acute and sustained antidepressant-like and hyperactive effects of ketamine. Mice were tested in the tail suspension test (TST) and differential-reinforcement-of-low-rate responding (DRL) 72 s task which are behavioral screens for antidepressant-like properties. Additionally, open field locomotor activity also was measured. In all behavioral assays, mice were pretreated with the nonselective opioid receptor antagonist naltrexone or saline prior to ketamine administration. The current study found that ketamine (10 mg/kg) produced acute (30 min) and sustained (24 h) antidepressant-like effects in TST, which were attenuated by pretreatment of 2 mg/kg naltrexone. Ketamine (32 mg/kg) also produced an acute antidepressant-like effect in the DRL 72 s task that was attenuated by pretreatment of 2 mg/kg naltrexone. Finally, ketamine (10 and 32 mg/kg) produced hyperactivity in the open field; however, pretreatment with 2 mg/kg naltrexone failed to block the hyperactivity effects ketamine. These results, along with recent clinical findings, suggest that ketamine's antidepressant effects, but not its hyperactive effects, involve activation of the opioid system.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
| | - Todd M Hillhouse
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, WI, USA
| | - Paige M Anderson
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, WI, USA
| | | | - Taylor N Kegen
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, WI, USA
| | - Sofia G Manicka
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
| | - Jackson T Lane
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
| | - Elizabeth Pottanat
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
| | - Madeline Van Fossen
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
| | - Remington Rice
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
| | - Joseph H Porter
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
23
|
Silva-Cardoso GK, Nobre MJ. Context-Specific Tolerance and Pharmacological Changes in the Infralimbic Cortex-Nucleus Accumbens Shell Pathway Evoked by Ketamine. Neurochem Res 2021; 46:1686-1700. [PMID: 33786719 DOI: 10.1007/s11064-021-03300-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 02/15/2021] [Accepted: 03/17/2021] [Indexed: 11/30/2022]
Abstract
Like other drugs, ketamine is abused due to its ability to act as a positive reinforcer in the control of behavior, just as natural reinforcers do. Besides, through Pavlovian conditioning, tolerance to drug effects can become conditioned to specific contextual cues showing that environmental stimuli can act as powerful mediators of craving and relapse. In the present study, we shall investigate the effects of long-term ketamine administration and withdrawal on behavioral measures and emotionality, the drug-context-specific influence on the tolerance to the sedative effects of an anesthetic dose of ketamine, and the neuropharmacological events underlying this phenomenon, in rats conditioned with 10 mg/kg of ketamine and later challenged with a dose of ketamine of 80 mg/kg in a familiar and non-familiar environment. Variations in dopamine and serotonin efflux in the infralimbic cortex-nucleus accumbens shell circuitry (IL-NAcSh) was further recorded in the same conditions. Our results highlight that besides its well-known reinforcing properties, ketamine also shares the ability to induce behavioral and pharmacological conditioned tolerance, associated with increases in cortical (IL), and decreases in striatal (NAcSh) dopamine release. To our knowledge, we are presenting the first set of behavioral and neurochemical data showing that, like other drugs of abuse, ketamine can induce learned context-specific tolerance.
Collapse
Affiliation(s)
- Gleice Kelli Silva-Cardoso
- Laboratório de Psicobiologia, Departamento de Psicologia, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo (USP), Av. Bandeirantes, 3900, Ribeirão Preto, SP, 14040-901, Brasil
| | - Manoel Jorge Nobre
- Departamento de Psicologia, Uni-FACEF, Franca, SP, 14401-135, Brasil.
- Laboratório de Psicobiologia, Departamento de Psicologia, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo (USP), Av. Bandeirantes, 3900, Ribeirão Preto, SP, 14040-901, Brasil.
| |
Collapse
|
24
|
Sartim AG, Marques J, Silveira KM, Gobira PH, Guimarães FS, Wegener G, Joca SR. Co-administration of cannabidiol and ketamine induces antidepressant-like effects devoid of hyperlocomotor side-effects. Neuropharmacology 2021; 195:108679. [PMID: 34157363 DOI: 10.1016/j.neuropharm.2021.108679] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/04/2021] [Accepted: 06/17/2021] [Indexed: 01/21/2023]
Abstract
BACKGROUND AND PURPOSE Although useful as a rapid-acting antidepressant drug, ketamine is known to induce psychotomimetic effects, which may interfere with its therapeutic use. Cannabidiol (CBD) is a non-psychostimulant compound from Cannabis sativa, which has shown promising antidepressant effects without inducing hyperlocomotion. AMPA receptor activation is involved in the antidepressant effect induced by ketamine, but its relevance for the effects of CBD is not known. Moreover, given that CBD has antipsychotic and antidepressant properties, it is unknown whether adding CBD to ketamine could potentiate the antidepressant properties of ketamine while also attenuating its psychostimulant effects. EXPERIMENTAL APPROACH S-Ketamine (2.5, 3, 5, 10, 30 mg/kg) and cannabidiol (3, 10, 30 mg/kg) were administered alone or in combination to male Swiss mice. Independent groups received NBQX (AMPA receptor antagonist) 5 min before administration of CBD or S-ketamine. The antidepressant-like effect was assessed in the forced swimming test (FST), and the open field test (OFT) evaluated the psychostimulant effect. KEY RESULTS CBD induced significant dose-dependent antidepressant effects without causing hyperlocomotion in the OFT. S-ketamine produced an antidepressant effect associated with hyperlocomotion in the higher dose. NBQX inhibited the antidepressant effect of both ketamine and CBD. Pretreatment with CBD (10 mg/kg) attenuated the ketamine-induced hyperlocomotion while preserving its antidepressant effect. CONCLUSION AND IMPLICATIONS: Similar to ketamine, the antidepressant-like effect elicited by CBD involves AMPA receptor activation. Additionally, CBD prevents the hyperlocomotion induced by S-ketamine without affecting its antidepressant-like effect. Our findings suggest that CBD and ketamine's combined administration can be a promising therapeutic strategy for achieving an appropriate antidepressant effect without unwanted side-effects. This article is part of the special issue on 'Cannabinoids'.
Collapse
Affiliation(s)
- A G Sartim
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - J Marques
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - K M Silveira
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil; Translational Neuropsychiatry Unit (TNU), Department of Clinical Medicine, Aarhus University, Denmark
| | - P H Gobira
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - F S Guimarães
- Department of Pharmacology, School of Medicine of Ribeirão Preto (FMRP), University of São Paulo, Ribeirão Preto, SP, Brazil; Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of São Paulo, Brazil
| | - G Wegener
- Translational Neuropsychiatry Unit (TNU), Department of Clinical Medicine, Aarhus University, Denmark
| | - S R Joca
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil; Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of São Paulo, Brazil; Translational Neuropsychiatry Unit (TNU), Department of Clinical Medicine, Aarhus University, Denmark; Department of Biomedicine, Aarhus University, Denmark.
| |
Collapse
|
25
|
Ior LD, Otimenyin SO, Okwuasaba FK. Antipsychotic-like effect of ethyl acetate fraction of Terminalia macroptera leaf in mice. IBRO Neurosci Rep 2021; 10:83-89. [PMID: 33842914 PMCID: PMC8019976 DOI: 10.1016/j.ibneur.2021.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 01/19/2021] [Indexed: 10/25/2022] Open
Abstract
Psychosis is a chronic neuropsychiatric disorder that affects millions of individuals worldwide and impairs the quality of life and productivity of the patients. Terminalia macroptera Guill & Perr. (Combrataceae) is a plant that is used in the management of anxiety related disorders. The present study investigates the antipsychotic effects of ethyl acetate fraction of T. macroptera (EFTM) leaf in ketamine-induced psychosis in mice. Acute toxicity of EFTM was determine using Lorke's method. Ketamine (25 mg/kg) was injected once daily for 7 consecutive days in Swiss albino mice to induce psychosis. The effect of the extracts (100, 200 and 400 mg/kg) was evaluated against psychotic-like behaviors induced by ketamine including locomotor activity and stereotypy in the open field test, immobility duration in the forced swim test, and memory impairment using the Y- maze test. The acute antipsychotic effect of EFTM was evaluated on apomorphine climbing test, while woodblock test was performed to assess its extrapyramidal side effects. The LD50 was found to be 3807 mg/kg p.o. which is considered safe. EFTM (100, 200 and 400 mg/kg) exhibited significant antipsychotic effect by reducing ketamine-induced hyperactivity, immobility, and memory deficit in mice, EFTM also suppressed stereotypic climbing behavior due to apomorphine. Accordingly, the antipsychotic activity of EFTM was not associated with extrapyramidal side effects as evidenced by lack of catalepsy. The study revealed that EFTM ameliorated psychotic-like symptoms and is devoid of extrapyramidal side effects in mice, underscoring its antipsychotic-like effect.
Collapse
Affiliation(s)
- Lydia Doosuur Ior
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, University of Jos, Jos, Nigeria
| | | | | |
Collapse
|
26
|
Fujikawa R, Yamada J, Jinno S. Subclass imbalance of parvalbumin-expressing GABAergic neurons in the hippocampus of a mouse ketamine model for schizophrenia, with reference to perineuronal nets. Schizophr Res 2021; 229:80-93. [PMID: 33229224 DOI: 10.1016/j.schres.2020.11.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/24/2020] [Accepted: 11/12/2020] [Indexed: 11/29/2022]
Abstract
Impairments of parvalbumin-expressing GABAergic neurons (PV+ neurons) and specialized extracellular structures called perineuronal nets (PNNs) have been found in schizophrenic patients. In this study, we examined potential alterations in four subclasses of PV+ neurons colocalized with PNNs in the hippocampus of a mouse ketamine model for schizophrenia. Because biosynthesis of human natural killer-1 (HNK-1) is shown to be associated with the risk of schizophrenia, here we used mouse monoclonal Cat-315 antibody, which recognizes HNK-1 glycans on PNNs. Once-daily intraperitoneal injections of ketamine for seven consecutive days induced hyper-locomotor activity in the open field tests. The prepulse inhibition (PPI) test showed that PPI scores declined in ketamine-treated mice compared to vehicle-treated mice. The densities of PV+ neurons and Cat-315+ PNNs declined in the CA1 region of ketamine-treated mice. Interestingly, the density of Cat-315+/PV+ neurons was lower in ketamine-treated mice than in vehicle-treated mice, whereas the density of Cat-315-/PV+ neurons was not affected by ketamine. Among the four subclasses of PV+ neurons, the densities of Cat-315+/PV+ basket cells and Cat-315-/PV+ axo-axonic cells were lower in ketamine-treated mice than in vehicle-treated mice, while the densities of Cat-315-/PV+ basket cells and Cat-315+/PV+ axo-axonic cells were not affected by ketamine. Taken together, PNNs may not play a simple neuroprotective role against ketamine. Because different subclasses of PV+ neurons are considered to play distinct roles in the hippocampal neuronal network, the ketamine-induced subclass imbalance of PV+ neurons may result in abnormal network activity, which underlies the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Risako Fujikawa
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Jun Yamada
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Shozo Jinno
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan.
| |
Collapse
|
27
|
Okubo Eneni AE, Ben-Azu B, Mayowa Ajayi A, Oladele Aderibigbe A. Diosmin attenuates schizophrenia-like behavior, oxidative stress, and acetylcholinesterase activity in mice. Drug Metab Pers Ther 2020; 0:/j/dmdi.ahead-of-print/dmdi-2020-0119/dmdi-2020-0119.xml. [PMID: 33055311 DOI: 10.1515/dmdi-2020-0119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/22/2020] [Indexed: 11/15/2022]
Abstract
Objectives Diosmin (DSM), commonly isolated from various plants, is a citrus nutrient that has been shown to increase intracellular antioxidant capacity and assuage symptoms associated with neurological disorders. Deficiency in the antioxidant system has been implicated in the pathogenesis of schizophrenia. The use of antioxidants as neuroprotectants to suppress schizophrenia pathology is increasingly being sought. Hence, this study investigated the effects of DSM on schizophrenia-like behavior and the underlying changes in biomarkers of oxidative stress and acetylcholinesterase (AChE) activity in mice. Methods The acute antipsychotic effect of DSM (25, 50, and 100 mg/kg, intraperitoneally [i.p.]), haloperidol (1 mg/kg, i.p.), and risperidone (RIS) (0.5 mg/kg, i.p.) was investigated on stereotyped behaviors induced by apomorphine (2 mg/kg, i.p.) and ketamine (10 mg/kg, i.p.). The effect of DSM on ketamine-induced hyperlocomotion, immobility enhancement, and its woodblock cataleptogenic potential was evaluated. Also, the subacute antipsychotic potential of DSM was assessed following intraperitoneal injection of DSM (25-100 mg/kg, i.p.) alone and in combination with ketamine (20 mg/kg, i.p.) for 10 days. The behaviors of the animals were assessed in the open-field, Y-maze, and forced swim tests. Brains of the animals were afterward processed for spectrophotometric assay of oxidative stress and AChE contents. Results DSM (25, 50, and 100 mg/kg) attenuated apormorphine-induced stereotypy and devoid of cataleptogenic effect. DSM and RIS reversed acute and subacute ketamine-induced schizophrenia-like behaviors. Disomin alone increased cognitive function and reduced despair-like phenotype. Furthermore, DSM increased superoxide dismutase and glutathione and decreased malondialdehyde and AChE levels in naïve and ketamine schizophrenic mice. Conclusions DSM prevents schizophrenia-like behavior, attenuates oxidative stress, and AChE activity in naïve and ketamine schizophrenic mice.
Collapse
Affiliation(s)
- Aya-Ebi Okubo Eneni
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Benneth Ben-Azu
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, PAMO University of Medical Sciences, Port Harcourt, Nigeria
| | - Abayomi Mayowa Ajayi
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Adegbuyi Oladele Aderibigbe
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| |
Collapse
|
28
|
Okubo Eneni AE, Ben-Azu B, Mayowa Ajayi A, Oladele Aderibigbe A. Diosmin attenuates schizophrenia-like behavior, oxidative stress, and acetylcholinesterase activity in mice. Drug Metab Pers Ther 2020; 35:dmpt-2020-0119. [PMID: 34704698 DOI: 10.1515/dmpt-2020-0119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/22/2020] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Diosmin (DSM), commonly isolated from various plants, is a citrus nutrient that has been shown to increase intracellular antioxidant capacity and assuage symptoms associated with neurological disorders. Deficiency in the antioxidant system has been implicated in the pathogenesis of schizophrenia. The use of antioxidants as neuroprotectants to suppress schizophrenia pathology is increasingly being sought. Hence, this study investigated the effects of DSM on schizophrenia-like behavior and the underlying changes in biomarkers of oxidative stress and acetylcholinesterase (AChE) activity in mice. METHODS The acute antipsychotic effect of DSM (25, 50, and 100 mg/kg, intraperitoneally [i.p.]), haloperidol (1 mg/kg, i.p.), and risperidone (RIS) (0.5 mg/kg, i.p.) was investigated on stereotyped behaviors induced by apomorphine (2 mg/kg, i.p.) and ketamine (10 mg/kg, i.p.). The effect of DSM on ketamine-induced hyperlocomotion, immobility enhancement, and its woodblock cataleptogenic potential was evaluated. Also, the subacute antipsychotic potential of DSM was assessed following intraperitoneal injection of DSM (25-100 mg/kg, i.p.) alone and in combination with ketamine (20 mg/kg, i.p.) for 10 days. The behaviors of the animals were assessed in the open-field, Y-maze, and forced swim tests. Brains of the animals were afterward processed for spectrophotometric assay of oxidative stress and AChE contents. RESULTS DSM (25, 50, and 100 mg/kg) attenuated apormorphine-induced stereotypy and devoid of cataleptogenic effect. DSM and RIS reversed acute and subacute ketamine-induced schizophrenia-like behaviors. Disomin alone increased cognitive function and reduced despair-like phenotype. Furthermore, DSM increased superoxide dismutase and glutathione and decreased malondialdehyde and AChE levels in naïve and ketamine schizophrenic mice. CONCLUSIONS DSM prevents schizophrenia-like behavior, attenuates oxidative stress, and AChE activity in naïve and ketamine schizophrenic mice.
Collapse
Affiliation(s)
- Aya-Ebi Okubo Eneni
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Benneth Ben-Azu
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria.,Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, PAMO University of Medical Sciences, Port Harcourt, Nigeria
| | - Abayomi Mayowa Ajayi
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Adegbuyi Oladele Aderibigbe
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| |
Collapse
|
29
|
Wang Z, Xu Z, Wu Y, Zhang Z, Li X. Impact of ketamine on the behavior and immune system of adult medaka (Oryzias latipes) at environmentally relevant concentrations and eco-risk assessment in surface water. JOURNAL OF HAZARDOUS MATERIALS 2020; 393:121577. [PMID: 32126430 DOI: 10.1016/j.jhazmat.2019.121577] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/23/2019] [Accepted: 10/30/2019] [Indexed: 06/10/2023]
Abstract
This work for the first time investigated the bioconcentration factor (BCF), toxicity, and eco-risk of KET using adult medaka fish (Oryzias latipes) as model organism after exposure at environmental concentrations (0.05-0.5 μg L-1) and higher levels (5-100 μg L-1) for 90 days. The BCF of KET was approximately 1.07- to 10.94- folds. The behavioral functions, including swimming properties, feeding rate, and food preference, were significantly impacted by KET (≥0.05 μg L-1). After 90-days exposure, KET induced histological abnormalities in liver and kidney tissue at 0.1 and 0.2 μg L-1, respectively. Additionally, the condition factor, hepatic-somatic index (HSI), and nephric-somatic index (NSI) of medaka were markedly impacted by KET treatment at 0.5, 0.5, and 0.1 μg L-1, respectively. Morphological inflammation (i.e., haemorrhage and erosion) in the fish body was observed exposed to KET, and the EC10 value was 0.407 μg L-1. Alterations in the expressions of genes (i.e., cacna1c, oxtr, erk1, and c-fos) and proteins (i.e., OXT and PKA), involved in in calcium ion channels induced by KET, could partly elucidate the underlying mechanism of the toxicity. The inflammatory risk to fish posed by KET in some rivers in southern China was at high level, suggesting the long-term concentration monitoring was required.
Collapse
Affiliation(s)
- Zhenglu Wang
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, PR China; Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210098, PR China
| | - Zeqiong Xu
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, PR China
| | - Yuexia Wu
- Key Laboratory of Watershed Geographic Sciences, Nanjing Institute of Geography & Limnology, Chinese Academy of Sciences, 73 East Beijing Road, 210008, Nanjing, PR China
| | - Zhaobin Zhang
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, PR China
| | - Xiqing Li
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, 100871, PR China.
| |
Collapse
|
30
|
B Hughes R, Whittingham-Dowd J, Simmons RE, Clapcote SJ, Broughton SJ, Dawson N. Ketamine Restores Thalamic-Prefrontal Cortex Functional Connectivity in a Mouse Model of Neurodevelopmental Disorder-Associated 2p16.3 Deletion. Cereb Cortex 2020; 30:2358-2371. [PMID: 31812984 PMCID: PMC7175007 DOI: 10.1093/cercor/bhz244] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 05/01/2019] [Accepted: 06/24/2019] [Indexed: 12/20/2022] Open
Abstract
2p16.3 deletions, involving heterozygous NEUREXIN1 (NRXN1) deletion, dramatically increase the risk of developing neurodevelopmental disorders, including autism and schizophrenia. We have little understanding of how NRXN1 heterozygosity increases the risk of developing these disorders, particularly in terms of the impact on brain and neurotransmitter system function and brain network connectivity. Thus, here we characterize cerebral metabolism and functional brain network connectivity in Nrxn1α heterozygous mice (Nrxn1α+/- mice), and assess the impact of ketamine and dextro-amphetamine on cerebral metabolism in these animals. We show that heterozygous Nrxn1α deletion alters cerebral metabolism in neural systems implicated in autism and schizophrenia including the thalamus, mesolimbic system, and select cortical regions. Nrxn1α heterozygosity also reduces the efficiency of functional brain networks, through lost thalamic "rich club" and prefrontal cortex (PFC) hub connectivity and through reduced thalamic-PFC and thalamic "rich club" regional interconnectivity. Subanesthetic ketamine administration normalizes the thalamic hypermetabolism and partially normalizes thalamic disconnectivity present in Nrxn1α+/- mice, while cerebral metabolic responses to dextro-amphetamine are unaltered. The data provide new insight into the systems-level impact of heterozygous Nrxn1α deletion and how this increases the risk of developing neurodevelopmental disorders. The data also suggest that the thalamic dysfunction induced by heterozygous Nrxn1α deletion may be NMDA receptor-dependent.
Collapse
Affiliation(s)
- Rebecca B Hughes
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK
| | - Jayde Whittingham-Dowd
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK
| | - Rachel E Simmons
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK
| | - Steven J Clapcote
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Susan J Broughton
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK
| | - Neil Dawson
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK
| |
Collapse
|
31
|
Raith H, Schuelert N, Duveau V, Roucard C, Plano A, Dorner-Ciossek C, Ferger B. Differential effects of traxoprodil and S-ketamine on quantitative EEG and auditory event-related potentials as translational biomarkers in preclinical trials in rats and mice. Neuropharmacology 2020; 171:108072. [PMID: 32243874 DOI: 10.1016/j.neuropharm.2020.108072] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 02/14/2020] [Accepted: 03/25/2020] [Indexed: 12/16/2022]
Abstract
Quantitative Electroencephalography (qEEG) and event-related potential (ERP) assessment have emerged as powerful tools to unravel translational biomarkers in preclinical and clinical psychiatric drug discovery trials. The aim of the present study was to compare the GluN2B negative allosteric modulator (NAM) traxoprodil (CP-101,606) with the unselective NMDA receptor channel blocker S-ketamine to give insight into central target engagement and differentiation on multiple EEG readouts. For qEEG recordings telemetric transmitters were implanted in male Wistar rats. Recorded EEG data were analyzed using fast Fourier transformation to determine power spectra and vigilance states. Additionally, body temperature and locomotor activity were assessed via telemetry. For recordings of auditory event-related potentials (AERP) male C57Bl/6J mice were chronically implanted with deep electrodes using a tethered system. Power spectral analysis revealed a significant increase in gamma power following ketamine treatment, whereas traxoprodil (6&18 mg/kg) induced an overall decrease primarily within alpha and beta bands. Additionally, ketamine disrupted sleep and enhanced time spent in wake vigilance states, whereas traxoprodil did not alter sleep-wake architecture. AERP and mismatch negativity (MMN) revealed that ketamine (10 mg/kg) selectively disrupts auditory deviance detection, whereas traxoprodil (6 mg/kg) did not alter MMN at clinically relevant doses. In contrast to ketamine treatment, traxoprodil did not produce hyperactivity and hypothermia. In conclusion, ketamine and traxoprodil showed very different effects on diverse EEG readouts differentiating selective GluN2B antagonism from non-selective pan-NMDA-R antagonists like ketamine. These readouts are thus perfectly suited to support drug discovery efforts on NMDA-R and understanding the different functions of NMDA-R subtypes.
Collapse
Affiliation(s)
- Henrike Raith
- Boehringer Ingelheim Pharma GmbH & Co. KG, CNS Diseases Research Germany, Birkendorferstr. 65, 88397, Biberach an der Riß, Germany.
| | - Niklas Schuelert
- Boehringer Ingelheim Pharma GmbH & Co. KG, CNS Diseases Research Germany, Birkendorferstr. 65, 88397, Biberach an der Riß, Germany.
| | - Venceslas Duveau
- SynapCell SAS, Biopolis and Institut Jean Roget, Université Joseph Fourier-Grenoble 1, Domaine de la merci, 38700, La Tronche, France.
| | - Corinne Roucard
- SynapCell SAS, Biopolis and Institut Jean Roget, Université Joseph Fourier-Grenoble 1, Domaine de la merci, 38700, La Tronche, France.
| | - Andrea Plano
- Plano Consulting, Georg-Schinbain-Str. 70, 88400, Biberach an der Riß, Germany.
| | - Cornelia Dorner-Ciossek
- Boehringer Ingelheim Pharma GmbH & Co. KG, CNS Diseases Research Germany, Birkendorferstr. 65, 88397, Biberach an der Riß, Germany.
| | - Boris Ferger
- Boehringer Ingelheim Pharma GmbH & Co. KG, CNS Diseases Research Germany, Birkendorferstr. 65, 88397, Biberach an der Riß, Germany.
| |
Collapse
|
32
|
Galvanho JP, Manhães AC, Carvalho-Nogueira ACC, Silva JDM, Filgueiras CC, Abreu-Villaça Y. Profiling of behavioral effects evoked by ketamine and the role of 5HT 2 and D 2 receptors in ketamine-induced locomotor sensitization in mice. Prog Neuropsychopharmacol Biol Psychiatry 2020; 97:109775. [PMID: 31676464 DOI: 10.1016/j.pnpbp.2019.109775] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/27/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022]
Abstract
Ketamine has addictive potential, a troublesome fact due to its promising use as a therapeutic drug. An important phenomenon associated with drug addiction is behavioral sensitization, usually characterized as augmented locomotion. However, other behaviors may also be susceptible to sensitization, and/or interfere with locomotor activity. Thus, this study drew a comprehensive behavioral 'profiling' in an animal model of repeated administration of ketamine. Adult Swiss mice received single daily ketamine injections (30 or 50 mg/Kg, i.p.), which were followed by open field testing for 7 days (acquisition period, ACQ). A ketamine challenge (sensitization test, ST) was carried out after a 5-day withdrawal. Locomotion, rearing, grooming, rotation and falling were assessed during ACQ and ST. All behaviors were affected from the first ACQ day onwards, with no indication of competition between locomotion and the other behaviors. Only locomotion in response to 30 mg/Kg of ketamine both escalated during ACQ and expressed increased levels at ST, evidencing development and expression of locomotor sensitization. Considering the involvement of serotonin 5HT(2) and dopamine D(2) receptors on addiction mechanisms, we further tested the involvement of these receptors in ketamine-induced sensitization. Ketanserin (5HT2 antagonist, 3 mg/Kg, s.c.) prevented ketamine-evoked development of locomotor sensitization. However, ketanserin pretreatment during ACQ failed to inhibit its expression during ST. Raclopride (D2 antagonist, 0.5 mg/Kg, s.c.) evoked less robust reductions in locomotion but prevented the development of ketamine-evoked sensitization. Pretreatment during ACQ further inhibited the expression of sensitization during ST. These results indicate that a partial overlap in serotonergic and dopaminergic mechanisms underlies ketamine-induced locomotor sensitization.
Collapse
Affiliation(s)
- Jefferson P Galvanho
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar - Vila Isabel, Rio de Janeiro, RJ 20550-170, Brazil
| | - Alex C Manhães
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar - Vila Isabel, Rio de Janeiro, RJ 20550-170, Brazil.
| | - Ana Cristina C Carvalho-Nogueira
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar - Vila Isabel, Rio de Janeiro, RJ 20550-170, Brazil
| | - Joyce de M Silva
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar - Vila Isabel, Rio de Janeiro, RJ 20550-170, Brazil
| | - Claudio C Filgueiras
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar - Vila Isabel, Rio de Janeiro, RJ 20550-170, Brazil
| | - Yael Abreu-Villaça
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro (UERJ), Av. Prof. Manuel de Abreu 444, 5 andar - Vila Isabel, Rio de Janeiro, RJ 20550-170, Brazil.
| |
Collapse
|
33
|
Wilson C, Li S, Hannan AJ, Renoir T. Antidepressant-like effects of ketamine in a mouse model of serotonergic dysfunction. Neuropharmacology 2020; 168:107998. [PMID: 32061666 DOI: 10.1016/j.neuropharm.2020.107998] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 02/05/2020] [Accepted: 02/09/2020] [Indexed: 12/28/2022]
Abstract
Traditional monoaminergic treatments of depression frequently exhibit suboptimal tolerability and effectiveness. The 'short' (s) allele variant of 5-HTTLPR is known to compromise transcriptional efficacy of the serotonin transporter (5-HTT) and can reduce treatment response to traditional antidepressants (e.g. selective serotonin reuptake inhibitors or SSRIs). This study sought to establish the 5-HTT knock-out (KO) line as a mouse model of SSRI-resistant depression and assess its response to a novel glutamatergic antidepressant, ketamine, a non-competitive N-methyl-d-aspartate receptor (NMDAR) antagonist. Following acute antidepressant treatment, 5-HTT KO mice and wild-type (WT) controls were subjected to the forced-swim test (FST), one of the most widely used techniques to detect acute antidepressant response. As hypothesised, when assessed 30 min after administration in the FST, the SSRI sertraline (20 mg/kg, i.p.) produced antidepressant-like effects in WT control but not in 5-HTT KO mice. In contrast, ketamine (20 mg/kg, i.p.) induced antidepressant-like effects in both genotypes. 5-HTT KO mice also exhibited a reduced locomotor response to both MK-801 (another NMDAR antagonist) and ketamine, and reduced GluN2A protein levels in the hippocampus, suggesting glutamatergic dysfunction in this model. These results highlight the utility of 5-HTT KO mice as a relevant model of SSRI-resistant depression and demonstrate that ketamine can produce acute antidepressant-like effects in conditions of 5-HTT deficiency. These findings extend existing literature that indicates ketamine is effective in ameliorating symptoms of treatment-resistant depression and may have implications for understanding the cellular and molecular mechanisms underlying the antidepressant effects of ketamine. This article is part of the special issue entitled 'Serotonin Research: Crossing Scales and Boundaries'.
Collapse
Affiliation(s)
- Carey Wilson
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Australia; Melbourne School of Psychological Science, University of Melbourne, Parkville, Australia
| | - Shanshan Li
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Australia; Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Australia
| | - Thibault Renoir
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Australia; Facssulty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia.
| |
Collapse
|
34
|
Openshaw RL, Thomson DM, Thompson R, Penninger JM, Pratt JA, Morris BJ, Dawson N. Map2k7 Haploinsufficiency Induces Brain Imaging Endophenotypes and Behavioral Phenotypes Relevant to Schizophrenia. Schizophr Bull 2020; 46:211-223. [PMID: 31219577 PMCID: PMC6942167 DOI: 10.1093/schbul/sbz044] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
c-Jun N-terminal kinase (JNK) signaling contributes to functional plasticity in the brain and cognition. Accumulating evidence implicates a role for MAP kinase kinase 7 (MAP2K7), a JNK activator encoded by the Map2k7 gene, and other JNK pathway components in schizophrenia (ScZ). Mice haploinsufficient for Map2k7 (Map2k7+/- mice) display ScZ-relevant cognitive deficits, although the mechanisms are unclear. Here we show that Map2k7+/- mice display translationally relevant alterations in brain function, including hippocampal and mesolimbic system hypermetabolism with a contrasting prefrontal cortex (PFC) hypometabolism, reminiscent of patients with ScZ. In addition Map2k7+/- mice show alterations in functional brain network connectivity paralleling those reported in early ScZ, including PFC and hippocampal hyperconnectivity and compromised mesolimbic system functional connectivity. We also show that although the cerebral metabolic response to ketamine is preserved, the response to dextroamphetamine (d-amphetamine) is significantly attenuated in Map2k7+/- mice, supporting monoamine neurotransmitter system dysfunction but not glutamate/NMDA receptor (NMDA-R) dysfunction as a consequence of Map2k7 haploinsufficiency. These effects are mirrored behaviorally with an attenuated impact of d-amphetamine on sensorimotor gating and locomotion, whereas similar deficits produced by ketamine are preserved, in Map2k7+/- mice. In addition, Map2k7+/- mice show a basal hyperactivity and sensorimotor gating deficit. Overall, these data suggest that Map2k7 modifies brain and monoamine neurotransmitter system function in a manner relevant to the positive and cognitive symptoms of ScZ.
Collapse
Affiliation(s)
- Rebecca L Openshaw
- Institute of Neuroscience and Psychology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, UK
| | - David M Thomson
- Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow, UK
| | - Rhiannon Thompson
- Institute of Neuroscience and Psychology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, UK
| | - Josef M Penninger
- Institute for Molecular Biotechnology of Austrian Academy of Sciences (IMBA), Vienna, Austria
| | - Judith A Pratt
- Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow, UK
| | - Brian J Morris
- Institute of Neuroscience and Psychology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, UK
| | - Neil Dawson
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, UK,To whom correspondence should be addressed; tel: +44 (0)1524 594 896, e-mail:
| |
Collapse
|
35
|
Kim JW, Monteggia LM. Increasing doses of ketamine curtail antidepressant responses and suppress associated synaptic signaling pathways. Behav Brain Res 2019; 380:112378. [PMID: 31760154 DOI: 10.1016/j.bbr.2019.112378] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 12/13/2022]
Abstract
Clinical findings show that a single subanesthetic dose of ketamine elicits rapid antidepressant effects. Accumulating data suggests that ketamine blocks the N-methyl-D-aspartate receptor and results in specific effects on intracellular signaling including increased brain-derived neurotrophic factor (BDNF) protein expression, which augments synaptic responses required for rapid antidepressant effects. To further investigate this potential mechanism for ketamine's antidepressant action, we examined the effect of increasing ketamine doses on intracellular signaling, synaptic plasticity, and rapid antidepressant effects. Given that ketamine is often used at 2.5-10 mg/kg to examine antidepressant effects and 20-50 mg/kg to model schizophrenia, we compared effects at 5, 20 and 50 mg/kg. We found that intraperitoneal (i.p.) injection of low dose (5 mg/kg) ketamine produces rapid antidepressant effects, which were not observed at 20 or 50 mg/kg. At 5 mg/kg ketamine significantly increased the level of BDNF, a protein necessary for the rapid antidepressant effects, while 20 and 50 mg/kg ketamine did not alter BDNF levels in the hippocampus. Low concentration ketamine also evoked the highest synaptic potentiation in the hippocampal CA1, while higher concentrations significantly decreased the synaptic effects. Our results suggest low dose ketamine produces antidepressant effects and has independent behavioral and synaptic effects compared to higher doses of ketamine that are used to model schizophrenia. These findings strengthen our knowledge on specific signaling associated with ketamine's rapid antidepressant effects.
Collapse
Affiliation(s)
- Ji-Woon Kim
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37240-7933, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37240-7933, USA
| | - Lisa M Monteggia
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37240-7933, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37240-7933, USA.
| |
Collapse
|
36
|
McDougall SA, Rios JW, Apodaca MG, Park GI, Montejano NR, Taylor JA, Moran AE, Robinson JAM, Baum TJ, Teran A, Crawford CA. Effects of dopamine and serotonin synthesis inhibitors on the ketamine-, d-amphetamine-, and cocaine-induced locomotor activity of preweanling and adolescent rats: sex differences. Behav Brain Res 2019; 379:112302. [PMID: 31655095 DOI: 10.1016/j.bbr.2019.112302] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 10/01/2019] [Accepted: 10/12/2019] [Indexed: 12/29/2022]
Abstract
The pattern of ketamine-induced locomotor activity varies substantially across ontogeny and according to sex. Although ketamine is classified as an NMDA channel blocker, it appears to stimulate the locomotor activity of both male and female rats via a monoaminergic mechanism. To more precisely determine the neural mechanisms underlying ketamine's actions, male and female preweanling and adolescent rats were pretreated with vehicle, the dopamine (DA) synthesis inhibitor ∝-methyl-DL-p-tyrosine (AMPT), or the serotonin (5-HT) synthesis inhibitor 4-chloro-DL-phenylalanine methyl ester hydrochloride (PCPA). After completion of the pretreatment regimen, the locomotor activating effects of saline, ketamine, d-amphetamine, and cocaine were assessed during a 2 h test session. In addition, the ability of AMPT and PCPA to reduce dorsal striatal DA and 5-HT content was measured in male and female preweanling, adolescent, and adult rats. Results showed that AMPT and PCPA reduced, but did not fully attenuate, the ketamine-induced locomotor activity of preweanling rats and female adolescent rats. Ketamine (20 and 40 mg/kg) caused a minimal amount of locomotor activity in male adolescent rats, and this effect was not significantly modified by AMPT or PCPA pretreatment. When compared to ketamine, d-amphetamine and cocaine produced different patterns of locomotor activity across ontogeny; moreover, AMPT and PCPA pretreatment affected psychostimulant- and ketamine-induced locomotion differently. When these results are considered together, it appears that both dopaminergic and serotonergic mechanisms mediate the ketamine-induced locomotor activity of preweanling and female adolescent rats. The dichotomous actions of ketamine relative to the psychostimulants in vehicle-, AMPT-, and PCPA-treated rats, suggests that ketamine modulates DA and 5-HT neurotransmission through an indirect mechanism.
Collapse
Affiliation(s)
- Sanders A McDougall
- Department of Psychology, California State University, San Bernardino, CA, USA.
| | - Jasmine W Rios
- Department of Psychology, California State University, San Bernardino, CA, USA
| | - Matthew G Apodaca
- Department of Psychology, California State University, San Bernardino, CA, USA
| | - Ginny I Park
- Department of Psychology, California State University, San Bernardino, CA, USA
| | - Nazaret R Montejano
- Department of Psychology, California State University, San Bernardino, CA, USA
| | - Jordan A Taylor
- Department of Psychology, California State University, San Bernardino, CA, USA
| | - Andrea E Moran
- Department of Psychology, California State University, San Bernardino, CA, USA; Department of Psychology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | | | - Timothy J Baum
- Department of Psychology, California State University, San Bernardino, CA, USA
| | - Angie Teran
- Department of Psychology, California State University, San Bernardino, CA, USA
| | - Cynthia A Crawford
- Department of Psychology, California State University, San Bernardino, CA, USA
| |
Collapse
|
37
|
Trujillo KA, Heller CY. Ketamine sensitization: Influence of dose, environment, social isolation and treatment interval. Behav Brain Res 2019; 378:112271. [PMID: 31593791 DOI: 10.1016/j.bbr.2019.112271] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/12/2019] [Accepted: 09/30/2019] [Indexed: 12/18/2022]
Abstract
Ketamine is a dissociative anesthetic first developed in the 1960s but is increasingly used at subanesthetic doses for both clinical and non-clinical purposes. There is evidence from human recreational users of compulsive use and addiction. Sensitization is an increase in an effect of a drug with repeated use that is thought to be important in the development of addiction. Research on psychomotor stimulants has shown the development of sensitization in laboratory animals to be modified by factors that influence addiction. In the current paper we describe four experiments on the development of sensitization in laboratory rats aimed at determining if ketamine sensitization is also influenced by factors thought to be important in addiction. Adult, male Sprague-Dawley rats received ketamine (5, 10, 20 or 50 mg/kg i.p.) for five or more days and the development of locomotor sensitization was followed. Experiment 1 examined the ability of low doses of ketamine to produce sensitization and found sensitization at 5, 10 and 20 mg/kg. Experiment 2 examined the influence of environmental context and found that ketamine sensitization (20 mg/kg) was greater when administration occurred in a novel environment (the experimental apparatus) than in home cages. Experiment 3 found that ketamine sensitization (20 mg/kg) did not occur when animals were housed in social isolation but occurred readily in pair-housed animals. Finally, Experiment 4 found that ketamine sensitization (20 or 50 mg/kg) was similar whether drug was administered daily or at 3-day intervals. Together, the results demonstrate that ketamine sensitization is robust and reliable, occurring under a variety of circumstances. Moreover, ketamine sensitization is influenced by factors that influence the development of addiction in humans. The current results may lead to a better understanding of ketamine abuse and addiction and may help inform clinical use of the drug.
Collapse
Affiliation(s)
- Keith A Trujillo
- Department of Psychology and Office for Training, Research, and Education in the Sciences, California State University San Marcos, 333 S. Twin Oaks Valley Road, San Marcos, CA 92096-0001, USA.
| | - Colleen Y Heller
- Department of Psychology and Office for Training, Research, and Education in the Sciences, California State University San Marcos, 333 S. Twin Oaks Valley Road, San Marcos, CA 92096-0001, USA
| |
Collapse
|
38
|
Crawford CA, Moran AE, Baum TJ, Apodaca MG, Montejano NR, Park GI, Gomez V, McDougall SA. Effects of monoamine depletion on the ketamine-induced locomotor activity of preweanling, adolescent, and adult rats: Sex and age differences. Behav Brain Res 2019; 379:112267. [PMID: 31593789 DOI: 10.1016/j.bbr.2019.112267] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 09/23/2019] [Accepted: 09/29/2019] [Indexed: 12/19/2022]
Abstract
Ketamine significantly increases the locomotor activity of rodents, however this effect varies according to the sex and age of the animal being tested. To determine the role monoamine systems play in ketamine's locomotor activating effects: (a) male and female preweanling, adolescent, and adult rats were pretreated with vehicle or the monoamine depleting agent reserpine (1 or 5 mg/kg), and (b) the behavioral actions of ketamine (20 or 40 mg/kg) were then compared to d-amphetamine (2 mg/kg) and cocaine (10 or 15 mg/kg). The ability of reserpine to deplete dorsal striatal dopamine (DA) and serotonin (5-HT) in male and female rats was determined using HPLC. Ketamine caused substantial increases in the locomotion of preweanling rats and older female rats (adolescents and adults), but had only small stimulatory effects on adolescent and adult male rats. When compared to cocaine and d-amphetamine, ketamine was especially sensitive to the locomotor-inhibiting effects of monoamine depletion. Ketamine-induced locomotion is at least partially mediated by monoamine systems, since depleting DA and 5-HT levels by 87-96% significantly attenuated the locomotor activating effects of ketamine in male and female rats from all three age groups. When administered to reserpine-pretreated rats, ketamine produced a different pattern of behavioral effects than either psychostimulant, suggesting that ketamine does not stimulate locomotor activity via actions at the presynaptic terminal. Instead, our results are consistent with the hypothesis that ketamine increases locomotor activity through a down-stream mechanism, possibly involving ascending DA and/or 5-HT projection neurons.
Collapse
Affiliation(s)
- Cynthia A Crawford
- Department of Psychology, California State University, San Bernardino, CA, USA.
| | - Andrea E Moran
- Department of Psychology, California State University, San Bernardino, CA, USA; Department of Psychology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Timothy J Baum
- Department of Psychology, California State University, San Bernardino, CA, USA
| | - Matthew G Apodaca
- Department of Psychology, California State University, San Bernardino, CA, USA
| | - Nazaret R Montejano
- Department of Psychology, California State University, San Bernardino, CA, USA
| | - Ginny I Park
- Department of Psychology, California State University, San Bernardino, CA, USA
| | - Vanessa Gomez
- Department of Psychology, California State University, San Bernardino, CA, USA
| | - Sanders A McDougall
- Department of Psychology, California State University, San Bernardino, CA, USA
| |
Collapse
|
39
|
PT-31, a putative α2-adrenoceptor agonist, is effective in schizophrenia cognitive symptoms in mice. Behav Pharmacol 2019; 30:574-587. [DOI: 10.1097/fbp.0000000000000494] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
40
|
Lee G, Zhou Y. NMDAR Hypofunction Animal Models of Schizophrenia. Front Mol Neurosci 2019; 12:185. [PMID: 31417356 PMCID: PMC6685005 DOI: 10.3389/fnmol.2019.00185] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/17/2019] [Indexed: 12/20/2022] Open
Abstract
The N-methyl-d-aspartate receptor (NMDAR) hypofunction hypothesis has been proposed to help understand the etiology and pathophysiology of schizophrenia. This hypothesis was based on early observations that NMDAR antagonists could induce a full range of symptoms of schizophrenia in normal human subjects. Accumulating evidence in humans and animal studies points to NMDAR hypofunctionality as a convergence point for various symptoms of schizophrenia. Here we review animal models of NMDAR hypofunction generated by pharmacological and genetic approaches, and how they relate to the pathophysiology of schizophrenia. In addition, we discuss the limitations of animal models of NMDAR hypofunction and their potential utility for therapeutic applications.
Collapse
Affiliation(s)
| | - Yi Zhou
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| |
Collapse
|
41
|
Lee CW, Chen YJ, Wu HF, Chung YJ, Lee YC, Li CT, Lin HC. Ketamine ameliorates severe traumatic event-induced antidepressant-resistant depression in a rat model through ERK activation. Prog Neuropsychopharmacol Biol Psychiatry 2019; 93:102-113. [PMID: 30940482 DOI: 10.1016/j.pnpbp.2019.03.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 03/12/2019] [Accepted: 03/29/2019] [Indexed: 12/28/2022]
Abstract
Treatment-resistant depression (TRD) is a major public health issue, as it is common for patients with depression to fail to respond to adequate trials of antidepressants. However, a well-established animal model of TRD is still warranted. The present study focused on selective serotonin reuptake inhibitor (SSRI) resistance, and aimed to investigate whether higher levels of traumatic stress caused by greater numbers of foot-shocks may lead to severe depression and to examine the feasibility of this as an animal model of SSRI-resistant depression. To reveal the correlation between traumatic stress and severe depression, rats received 3, 6 and 10 tone (conditioned stimulus, CS)-shock (unconditioned stimulus, US) pairings to mimic mild, moderate, and severe traumatic events, and subsequent depressive-like behaviors and protein immunocontents were analyzed. The antidepressant efficacy was assessed for ketamine and SSRI (i.e., fluoxetine) treatment. We found that only the severe stress group presented depressive-like behaviors. Phosphorylation of extracellular signal-regulated kinases (ERKs) was decreased in the amygdala and prefrontal cortex (PFC). The immunocontents of GluA1 and PSD 95 were increased in the amygdala and decreased in the PFC. Moreover, the glutamate-related abnormalities in the amygdala and PFC were normalized by single-dose (10 mg/kg, i.p.) ketamine treatment. In contrast, the depressive-like behaviors were not reversed by 28 days of fluoxetine treatment (10 mg/kg, i.p.) in the severe stress group. Our data demonstrated that high levels of traumatic stress could lead to SSRI-resistant depressive symptoms through impacts on the glutamatergic system, and that this rat model has the potential to be a feasible animal model of SSRI-resistant depression.
Collapse
Affiliation(s)
- Chi-Wei Lee
- Department and Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan; Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Instiutes, Taiwan
| | - Yi-Ju Chen
- Department and Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Han-Fang Wu
- Department and Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Optometry, Hsin-Sheng College of Medical Care and Management, Taoyuan, Taiwan
| | - Yueh-Jung Chung
- Department and Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Chao Lee
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Instiutes, Taiwan
| | - Cheng-Ta Li
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Brain Research Center, National Yang-Ming University, Taipei, Taiwan; Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan.
| | - Hui-Ching Lin
- Department and Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan; Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Instiutes, Taiwan; Brain Research Center, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
42
|
McDougall SA, Park GI, Ramirez GI, Gomez V, Adame BC, Crawford CA. Sex-dependent changes in ketamine-induced locomotor activity and ketamine pharmacokinetics in preweanling, adolescent, and adult rats. Eur Neuropsychopharmacol 2019; 29:740-755. [PMID: 30981586 PMCID: PMC7059997 DOI: 10.1016/j.euroneuro.2019.03.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/22/2019] [Accepted: 03/27/2019] [Indexed: 01/06/2023]
Abstract
Although ketamine has long been known to increase locomotor activity, only recently was it realized that this behavioral effect varies according to both sex and age. The purpose of the present study was threefold: first, to measure the locomotor activating effects of ketamine in male and female rats across early ontogeny and into adulthood; second, to assess ketamine and norketamine pharmacokinetics in the dorsal striatum and hippocampus of the same age groups; and, third, to use curvilinear regression to determine the relationship between locomotor activity and dorsal striatal concentrations of ketamine and norketamine. A high dose of ketamine (80 mg/kg, i.p.) was administered in order to examine the complete cycle of locomotor responsiveness across a 280-min testing session. In separate groups of rats, the dorsal striata and hippocampi were removed at 10 time points (0-360 min) after ketamine administration and samples were assayed for ketamine, norketamine, and dopamine using HPLC. In female rats, ketamine produced high levels of locomotor activity that varied only slightly among age groups. Male preweanling rats responded like females, but adolescent and adult male rats exhibited lesser amounts of ketamine-induced locomotor activity. Ketamine and norketamine pharmacokinetics, especially peak values and area under the curve, generally mirrored age- and sex-dependent differences in locomotor activity. Among male rats and younger female rats, dorsal striatal ketamine and norketamine levels accounted for a large proportion of the variance in locomotor activity. In adult female rats, however, an additional factor, perhaps involving other ketamine and norketamine metabolites, was influencing locomotor activity.
Collapse
Affiliation(s)
- Sanders A McDougall
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA 92407, USA.
| | - Ginny I Park
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA 92407, USA
| | - Goretti I Ramirez
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA 92407, USA
| | - Vanessa Gomez
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA 92407, USA
| | - Brittnee C Adame
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA 92407, USA
| | - Cynthia A Crawford
- Department of Psychology, California State University, 5500 University Parkway, San Bernardino, CA 92407, USA
| |
Collapse
|
43
|
Bates MLS, Trujillo KA. Long-lasting effects of repeated ketamine administration in adult and adolescent rats. Behav Brain Res 2019; 369:111928. [PMID: 31034850 DOI: 10.1016/j.bbr.2019.111928] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/22/2019] [Accepted: 04/25/2019] [Indexed: 12/19/2022]
Abstract
Initiation of ketamine use often occurs in adolescence, yet little is known about long-term consequences when use begins in this developmental period. The current experiments were designed to examine the effects of repeated exposure to ketamine in adolescence on behavior in adulthood. We examined locomotor activity, as well as cognitive function, in animals that received repeated administration of ketamine. Groups of adolescent and adult male rats were treated with ketamine (25 mg/kg) once daily for 10 days. Locomotor activity was assessed following the first injection, following 10 days of injection, and following 20 days of abstinence. Acute locomotor effects and locomotor sensitization were compared in adolescents and adults; cross-sensitization to dextromethorphan, another dissociative with abusive potential, was also examined. In a separate group of animals cognitive deficits were assessed following the 20 day abstinence period in spatial learning and novel object recognition tasks. The locomotor stimulant effect of ketamine was much greater in adolescents than adults. Animals that were repeatedly administered ketamine demonstrated locomotor sensitization immediately after the final injection. However, sensitization only persisted after the abstinence period in animals treated as adults. No cross-sensitization to dextromethorphan was evident. Ketamine failed to produce statistically significant cognitive deficits in either age group, although drug-treated adults showed a trend towards deficits in spatial learning. Repeated use of ketamine produces long-lasting neuroadaptations that may contribute to addiction. Mild lasting memory deficits may occur in adults, although further work is necessary to confirm these findings. The results extend the understanding of potential long-term consequences of ketamine use in adolescents and adults.
Collapse
Affiliation(s)
- M L Shawn Bates
- Department of Psychology and Office for Training, Research and Education in the Sciences (OTRES), California State University, San Marcos, 333 S. Twin Oaks Valley Rd, San Marcos, CA 92096, USA.
| | - Keith A Trujillo
- Department of Psychology and Office for Training, Research and Education in the Sciences (OTRES), California State University, San Marcos, 333 S. Twin Oaks Valley Rd, San Marcos, CA 92096, USA.
| |
Collapse
|
44
|
Ben-Azu B, Nwoke EE, Aderibigbe AO, Omogbiya IA, Ajayi AM, Olonode ET, Umukoro S, Iwalewa EO. Possible neuroprotective mechanisms of action involved in the neurobehavioral property of naringin in mice. Biomed Pharmacother 2018; 109:536-546. [PMID: 30399589 DOI: 10.1016/j.biopha.2018.10.055] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 10/10/2018] [Accepted: 10/10/2018] [Indexed: 01/01/2023] Open
Abstract
Flavonoids are naturally occurring bioactive phytochemical metabolites widely known to prevent and suppress several human diseases, and are important sources of therapeutic compounds from plants. Evidence derived from previous studies suggests that naringin, a neuroactive flavonoid possess functional beneficial neurobehavioral effects including anxiolytic, antidepressant and memory enhancing properties. However, literature search revealed that no studies have been carried out to evaluate the possible biochemical mechanisms involved in the neurobehavioral property of naringin alone following repeated treatment. Hence, this study was designed to evaluate the possible neuro-biochemical mechanisms involved in the neurobehavioral property of naringin following repeated administration in mice. The effects of naringin (2.5, 5 and 10 mg/kg), diazepam (2 mg/kg), imipramine (15 mg/kg) and donepezil (1 mg/kg) or vehicle on neurobehavioral and biochemical effects were evaluated in mice following repeated intraperitoneal injection for 7 consecutive days. Neurobehavioral activities consisting of open-field (locomotor), elevated-plus maze (anxiolytic), forced swim and social interaction (antidepressant and social preference), and Y-maze (memory enhancing) tests were assessed. Thereafter, brains levels of biomarkers of oxidative, nitrosative and cholinergic parameters were determined. Repeated treatment with naringin produced increased locomotor activity, and demonstrated antidepressant-like effects evidenced by decreased immobility time in forced swim test and increased % social preference in the social interaction test relative to controls. Also, naringin induced anxiolytic-like effect and increased cognitive performance in mice. Mechanistically, naringin significantly increased the activities of superoxide dismutase and catalase, and glutathione concentration relative to vehicle-controls. However, naringin significantly decreased malondialdehyde and nitrite contents, and reduced brain acetylcholinesterase activity in mice brains in a significant manner relative to controls. Taken together, these findings suggest that treatment with naringin might be useful to produce functional behavioral effects via mechanisms related to enhancement of cholinergic transmission, antioxidant defense systems, inhibition of lipid peroxidation and nitrosative processes.
Collapse
Affiliation(s)
- Benneth Ben-Azu
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria.
| | - Ekene Enekabokom Nwoke
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| | - Adegbuyi Oladele Aderibigbe
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Itivere Adrian Omogbiya
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria; Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, Delta State University, Abraka, Nigeria
| | - Abayomi Mayowa Ajayi
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Elizabeth Toyin Olonode
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria; Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Afe Babalola University, Ado Ekiti, Ekiti State, Nigeria
| | - Solomon Umukoro
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Ezekiel O Iwalewa
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| |
Collapse
|
45
|
Morin Attenuates Neurochemical Changes and Increased Oxidative/Nitrergic Stress in Brains of Mice Exposed to Ketamine: Prevention and Reversal of Schizophrenia-Like Symptoms. Neurochem Res 2018; 43:1745-1755. [PMID: 29956036 DOI: 10.1007/s11064-018-2590-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 06/20/2018] [Accepted: 06/24/2018] [Indexed: 12/22/2022]
Abstract
Previous studies have revealed that morin (MOR), a neuroactive bioflavonoid, with proven psychotropic and neuroprotective properties reduced schizophrenic-like behaviors in mice. This study further evaluated the ability of MOR to prevent and reverse ketamine-induced schizophrenic-like behaviors and the underlying neurochemical changes and increased oxidative/nitrergic stress in mice. In the preventive protocol, mice received intraperitoneal injection of MOR (100 mg/kg), reference antipsychotic drugs [haloperidol (1 mg/kg), risperidone (0.5 mg/kg)], or saline daily for 14 consecutive days prior to i.p. injection of ketamine (KET) (20 mg/kg/day) from the 8th to the 14th day. In the reversal protocol, the animals received KET or saline for 14 days prior to MOR, haloperidol, risperidone, or saline treatments. Schizophrenic-like behaviors: positive (open-field test), negative (social-interaction test) and cognitive (Y-maze test) symptoms were evaluated. Thereafter, the brain levels of dopamine, glutamate, 5-hydroxytryptamine and acetyl-cholinesterase, as well as biomarkers of oxidative/nitrergic stress were measured in the striatum, prefrontal-cortex (PFC) and hippocampus (HC). Morin prevented and reversed KET-induced hyperlocomotion, social and cognitive deficits. Also, MOR or risperidone attenuated altered dopaminergic, glutamatergic, 5-hydroxytryptaminergic and cholinergic neurotransmissions in brain region-dependent manner. The increased malondialdehyde and nitrite levels accompanied by decreased glutathione concentrations in the striatum, PFC and HC in KET-treated mice were significantly attenuated by MOR or risperidone. Taken together, these findings suggest that the anti-schizophrenic-like activity of MOR may be mediated via mechanisms related to attenuation of neurochemical changes and oxidative/nitrergic alterations in mice.
Collapse
|
46
|
Ben-Azu B, Omogbiya IA, Aderibigbe AO, Umukoro S, Ajayi AM, Iwalewa EO. Doxycycline prevents and reverses schizophrenic-like behaviors induced by ketamine in mice via modulation of oxidative, nitrergic and cholinergic pathways. Brain Res Bull 2018; 139:114-124. [DOI: 10.1016/j.brainresbull.2018.02.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 01/21/2018] [Accepted: 02/02/2018] [Indexed: 11/24/2022]
|
47
|
Advantages of the Alpha-lipoic Acid Association with Chlorpromazine in a Model of Schizophrenia Induced by Ketamine in Rats: Behavioral and Oxidative Stress evidences. Neuroscience 2018; 373:72-81. [PMID: 29337238 DOI: 10.1016/j.neuroscience.2018.01.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/27/2017] [Accepted: 01/03/2018] [Indexed: 11/20/2022]
Abstract
Schizophrenia is a chronic mental disorder reported to compromise about 1% of the world's population. Although its pathophysiological process is not completely elucidated, evidence showing the presence of an oxidative imbalance has been increasingly highlighted in the literature. Thus, the use of antioxidant substances may be of importance for schizophrenia treatment. The objective of this study was to evaluate the behavioral and oxidative alterations by the combination of chlorpromazine (CP) and alpha-lipoic acid (ALA), a potent antioxidant, in the ketamine (KET) model of schizophrenia in rats. Male Wistar rats (200-300 g) were treated for 10 days with saline, CP or ALA alone or in combination with CP previous to KET and the behavioral (open field, Y-maze and PPI tests) and oxidative tests were performed on the last day of treatment. The results showed that KET induced hyperlocomotion, impaired working memory and decreased PPI. CP alone or in combination with ALA prevented KET-induced behavioral effects. In addition, the administration of KET decreased GSH and increased nitrite, lipid peroxidation and myeloperoxidase activity. CP alone or combined with ALA prevented the oxidative alterations induced by KET. In conclusion, the treatment with KET in rats induced behavioral impairments accompanied by hippocampal oxidative alterations, possibly related to NMDA receptors hypofunction. Besides that, CP alone or combined with ALA prevented these effects, showing a beneficial activity as antipsychotic agents.
Collapse
|
48
|
Kokkinou M, Ashok AH, Howes OD. The effects of ketamine on dopaminergic function: meta-analysis and review of the implications for neuropsychiatric disorders. Mol Psychiatry 2018; 23:59-69. [PMID: 28972576 PMCID: PMC5754467 DOI: 10.1038/mp.2017.190] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 05/16/2017] [Accepted: 07/17/2017] [Indexed: 12/23/2022]
Abstract
Ketamine is a non-competitive antagonist at the N-methyl-d-aspartate receptor. It has recently been found to have antidepressant effects and is a drug of abuse, suggesting it may have dopaminergic effects. To examine the effect of ketamine on the dopamine systems, we carried out a systematic review and meta-analysis of dopamine measures in the rodent, human and primate brain following acute and chronic ketamine administration relative to a drug-free baseline or control condition. Systematic search of PubMed and PsychInfo electronic databases yielded 40 original peer-reviewed studies. There were sufficient rodent studies of the acute effects of ketamine at sub-anaesthetic doses for meta-analysis. Acute ketamine administration in rodents is associated with significantly increased dopamine levels in the cortex (Hedge's g= 1.33, P<0.01), striatum (Hedge's g=0.57, P<0.05) and the nucleus accumbens (Hedge's g=1.30, P<0.05) compared to control conditions, and 62-180% increases in dopamine neuron population activity. Sub-analysis indicated elevations were more marked in in vivo (g=1.93) than ex vivo (g=0.50) studies. There were not enough studies for meta-analysis in other brain regions studied (hippocampus, ventral pallidum and cerebellum), or of the effects of chronic ketamine administration, although consistent increases in cortical dopamine levels (from 88 to 180%) were reported in the latter studies. In contrast, no study showed an effect of anaesthetic doses (>100 mg kg-1) of ketamine on dopamine levels ex vivo, although this remains to be tested in vivo. Findings in non-human primates and in human studies using positron emission tomography were not consistent. The studies reviewed here provide evidence that acute ketamine administration leads to dopamine release in the rodent brain. We discuss the inter-species variation in the ketamine induced dopamine release as well as the implications for understanding psychiatric disorders, in particular substance abuse, schizophrenia, and the potential antidepressant properties of ketamine, and comparisons with stimulants and other NMDA antagonists. Finally we identify future research needs.
Collapse
Affiliation(s)
- M Kokkinou
- Robert Steiner MR Unit, Psychiatric Imaging Group, MRC London Institute of Medical Sciences (LMS), Hammersmith Hospital, London, UK,Psychiatric Imaging Group, Faculty of Medicine, MRC London Institute of Medical Sciences (LMS), Imperial College London, London, UK
| | - A H Ashok
- Robert Steiner MR Unit, Psychiatric Imaging Group, MRC London Institute of Medical Sciences (LMS), Hammersmith Hospital, London, UK,Psychiatric Imaging Group, Faculty of Medicine, MRC London Institute of Medical Sciences (LMS), Imperial College London, London, UK,Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, Kings College London, London, UK
| | - O D Howes
- Robert Steiner MR Unit, Psychiatric Imaging Group, MRC London Institute of Medical Sciences (LMS), Hammersmith Hospital, London, UK,Psychiatric Imaging Group, Faculty of Medicine, MRC London Institute of Medical Sciences (LMS), Imperial College London, London, UK,Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, Kings College London, London, UK,Psychiatric Imaging Group, MRC Clinical Sciences Centre, Hammersmith Hospital, Imperial College London, Du Cane Road, London W12 0NN, UK. E-mail:
| |
Collapse
|
49
|
Robinson BL, Dumas M, Ali SF, Paule MG, Gu Q, Kanungo J. Mechanistic studies on ketamine-induced mitochondrial toxicity in zebrafish embryos. Neurotoxicol Teratol 2017; 69:63-72. [PMID: 29225006 DOI: 10.1016/j.ntt.2017.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 12/06/2017] [Accepted: 12/06/2017] [Indexed: 12/26/2022]
Abstract
Ketamine, a phencyclidine derivative, is an antagonist of the Ca2+-permeable N-methyl-d-aspartate (NMDA)-type glutamate receptors. It is a pediatric anesthetic and has been implicated in developmental neurotoxicity. Ketamine has also been shown to deplete ATP in mammalian cells. Our previous studies showed that acetyl l-carnitine (ALCAR) prevented ketamine-induced cardiotoxicity and neurotoxicity in zebrafish embryos. Based on our finding that ALCAR's protective effect was blunted by oligomycin A, an inhibitor of ATP synthase, we further investigated the effects of ketamine and ALCAR on ATP levels, mitochondria and ATP synthase in zebrafish embryos. The results demonstrated that ketamine reduced ATP levels in the embryos but not in the presence of ALCAR. Ketamine reduced total mitochondrial protein levels and mitochondrial potential, which were prevented with ALCAR co-treatment. To determine the cause of ketamine-induced ATP deficiency, we explored the status of ATP synthase. The results showed that a subunit of ATP synthase, atp5α1, was transcriptionally down-regulated by ketamine, but not in the presence of ALCAR, although ketamine caused a significant upregulation in another ATP synthase subunit, atp5β and total ATP synthase protein levels. Most of the ATP generated by heart mitochondria are utilized for its contraction and relaxation. Ketamine-treated embryos showed abnormal heart structure, which was abolished with ALCAR co-treatment. This study offers evidence for a potential mechanism by which ketamine could cause ATP deficiency mediated by mitochondrial dysfunction.
Collapse
Affiliation(s)
- Bonnie L Robinson
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR 72079, USA
| | - Melanie Dumas
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR 72079, USA
| | - Syed F Ali
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR 72079, USA
| | - Merle G Paule
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR 72079, USA
| | - Qiang Gu
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR 72079, USA
| | - Jyotshna Kanungo
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR 72079, USA.
| |
Collapse
|
50
|
Viana GSDB, Xavier CC, do Vale EM, Lopes MJP, Alves VDJ, Costa RDO, Neves KRT. The monoaminergic pathways and inhibition of monoamine transporters interfere with the antidepressive-like behavior of ketamine. IBRO Rep 2017; 4:7-13. [PMID: 30135946 PMCID: PMC6084823 DOI: 10.1016/j.ibror.2017.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 11/28/2017] [Indexed: 01/02/2023] Open
Abstract
Ketamine (KET), a NMDA receptor antagonist, has been studied for its rapid and efficacious antidepressant effect, even for the treatment-resistant depression. Although depression is a major cause of disability worldwide, the treatment can be feasible, affordable and cost-effective, decreasing the population health burden. We evaluated the antidepressive-like effects of KET and its actions on monoamine contents (DA and its metabolites, as well as 5-HT) and on tyrosine hydroxylase (TH). In addition DAT and SERT (DA and 5-HT transporters, respectively) were also assessed. Male Swiss mice were divided into Control and KET-treated groups. The animals were acutely treated with KET (2, 5 or 10 mg/kg, i.p.) and subjected to the forced swimming test, for evaluation of the antidepressive-like behavior. Imipramine and fluoxetine were used as references. The results showed that KET decreased dose-dependently the immobility time and shortly after the test, the animals were euthanized for striatal dissections and monoamine determinations. In addition, the brain (striata, hippocampi and prefrontal cortices) was immunohistochemically processed for TH, DAT and SERT. KET at its higher dose increased DA and its metabolites (DOPAC and HVA) and mainly 5-HT contents, in mice striata, effects associated with increases in TH and decreases in DAT immunoreactivities. Furthermore, reductions in SERT immunoreactivities were observed in the striatum and hippocampus. The results indicate that KET antidepressive-like effect probably involves, among other factors, monoaminergic pathways, as suggested by the increased striatal TH immunoreactivity and reduced brain DA (DAT) and 5-HT (SERT) transporters.
Collapse
Affiliation(s)
- Glauce Socorro de Barros Viana
- Faculty of Medicine Estácio of Juazeiro do Norte, Ceará, Brazil.,Faculty of Medicine of the Federal University of Ceará, Ceará, Brazil
| | | | | | | | | | | | | |
Collapse
|