1
|
Olivares-Berjaga D, Martínez-Pinteño A, Rodríguez N, Madero S, Prohens L, Martínez-Serrano I, Mas S, Morén C, Parellada E, Gassó P. Effects of the PAM of mGluR2, JNJ-46356479, on brain apoptotic protein levels in a mouse model of schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2024; 131:110955. [PMID: 38296154 DOI: 10.1016/j.pnpbp.2024.110955] [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: 10/09/2023] [Revised: 01/25/2024] [Accepted: 01/28/2024] [Indexed: 02/12/2024]
Abstract
Current treatment for schizophrenia (SZ) ameliorates the positive symptoms, but is inefficient in treating the negative and cognitive symptoms. The SZ glutamatergic dysfunction hypothesis has opened new avenues in the development of novel drugs targeting the glutamate storm, an inducer of progressive neuropathological changes. Positive allosteric modulators of metabotropic glutamate receptor 2 (mGluR2), such as JNJ-46356479 (JNJ), reduce the presynaptic release of glutamate, which has previously been demonstrated to attenuate glutamate- and dopamine-induced apoptosis in human neuroblastoma cell cultures. We hypothesised that JNJ treatment would modify the brain levels of apoptotic proteins in a mouse model of ketamine (KET)-induced schizophrenia. We analysed the levels of proapoptotic (caspase-3 and Bax) and antiapoptotic (Bcl-2) proteins by western blot in the prefrontal cortex and hippocampus of JNJ-treated mice. JNJ attenuated apoptosis in the brain by partially restoring the levels of the antiapoptotic Bcl-2 protein, which is significantly reduced in animals exposed to KET. Additionally, a significant inverse correlation was observed between proapoptotic protein levels and behavioural deficits in the mice. Our findings suggest that JNJ may attenuate brain apoptosis in vivo, as previously described in cell cultures, providing a link between neuropathological deficits and SZ symptomatology.
Collapse
Affiliation(s)
| | - Albert Martínez-Pinteño
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Natalia Rodríguez
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Santiago Madero
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Barcelona Clínic Schizophrenia Unit (BCSU), Dpt. of Psychiatry, Institute of Neuroscience, Hospital Clínic of Barcelona, University of Barcelona, Spain.
| | - Llucía Prohens
- Dept. of Basic Clinical Practice, University of Barcelona, Spain.
| | | | - Sergi Mas
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain.
| | - Constanza Morén
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Barcelona Clínic Schizophrenia Unit (BCSU), Dpt. of Psychiatry, Institute of Neuroscience, Hospital Clínic of Barcelona, University of Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Spain.
| | - Eduard Parellada
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Barcelona Clínic Schizophrenia Unit (BCSU), Dpt. of Psychiatry, Institute of Neuroscience, Hospital Clínic of Barcelona, University of Barcelona, Spain.
| | - Patricia Gassó
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain.
| |
Collapse
|
2
|
Lan X, Wang C, Zhang F, Liu H, Li W, Ye Y, Hu Z, Mai S, Ning Y, Zhou Y. Short-term cognitive effects of repeated-dose esketamine in adolescents with major depressive disorder and suicidal ideation: a randomized controlled trial. Child Adolesc Psychiatry Ment Health 2023; 17:108. [PMID: 37710297 PMCID: PMC10503003 DOI: 10.1186/s13034-023-00647-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/12/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND Ketamine and its enantiomer have rapid and robust effects on depressive symptom and suicidal ideation. Little is known about their cognitive effects in adolescents. We aimed to evaluate the short-term effect of esketamine on cognition in adolescents with major depressive disorder (MDD) and suicidal ideation. METHOD In this randomized-controlled trial, 51 participants aged 13-18 with MDD and suicidal ideation received three intravenous infusions of either esketamine (0.25 mg/kg) or midazolam (0.02 mg/kg). Four dimensions of the MATRICS Consensus Cognitive Battery (MCCB), including processing speed, working memory, verbal learning and visual learning, were assessed at Days 0, 6 and 12. RESULTS In the linear mixed model, a significant time main effect (F = 12.803, P < 0.001), drug main effect (F = 6.607, P = 0.013), and interaction effect (F = 3.315, P = 0.041) was found in processing speed. Other dimensions including working memory and verbal learning showed significant time main effect (all P < 0.05), but no significant drug or interaction effect (all P > 0.05). Esketamine group showed improvement in processing speed from baseline to Days 6 and 12, and working memory from baseline to Day 12 (all P < 0.05). The generalized estimation equation showed no significant association between baseline cognition and antidepressant or antisuicidal effect (both P > 0.05). CONCLUSIONS The present study suggested that three-dose subanesthetic esketamine infusions did not harm cognition among adolescents with MDD and suicidal ideation. Instead, esketamine may be associated with improvement in processing speed. TRIAL REGISTRATION This trial was registered in the Chinese Clinical Trials Registry ( http://www.chictr.org.cn , ChiCTR2000041232).
Collapse
Affiliation(s)
- Xiaofeng Lan
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Mingxin Road #36, Liwan District, Guangzhou, 510370, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, 510370, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China
| | - Chengyu Wang
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Mingxin Road #36, Liwan District, Guangzhou, 510370, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, 510370, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China
| | - Fan Zhang
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Mingxin Road #36, Liwan District, Guangzhou, 510370, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, 510370, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China
| | - Haiyan Liu
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Mingxin Road #36, Liwan District, Guangzhou, 510370, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, 510370, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China
| | - Weicheng Li
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Mingxin Road #36, Liwan District, Guangzhou, 510370, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, 510370, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China
| | - Yanxiang Ye
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Mingxin Road #36, Liwan District, Guangzhou, 510370, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, 510370, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China
| | - Zhibo Hu
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Mingxin Road #36, Liwan District, Guangzhou, 510370, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, 510370, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China
| | - Siming Mai
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Mingxin Road #36, Liwan District, Guangzhou, 510370, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, 510370, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China
| | - Yuping Ning
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Mingxin Road #36, Liwan District, Guangzhou, 510370, China.
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, 510370, China.
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China.
- Department of Psychology, The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
| | - Yanling Zhou
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Mingxin Road #36, Liwan District, Guangzhou, 510370, China.
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, 510370, China.
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, 510370, China.
| |
Collapse
|
3
|
Oh HA, Yoo JH, Kim YJ, Han KS, Woo DH. 4-EA-NBOMe, an amphetamine derivative, alters glutamatergic synaptic transmission through 5-HT 1A receptors on cortical neurons from SpragueDawley rat and pyramidal neurons from C57BL/6 mouse. Neurotoxicology 2023; 95:144-154. [PMID: 36738894 DOI: 10.1016/j.neuro.2023.02.001] [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: 09/07/2022] [Revised: 01/20/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023]
Abstract
New psychoactive substances (NPSs) are compounds designed to mimic illegal recreational drugs. In particular, there are difficulties in legal restrictions because there is no fast NPS detection method to suppress the initial spread of NPS with criminal records; thus, they expose the public to serious health threats, including toxicity and dependence. However, the effects of NPSs on the brain and the related cellular mechanisms are well unknown. One of the recently emerging drugs is 4-ethylamphetamine-NBOMe (4-EA-NBOMe), a member of the 2 C phenylalanine family with a similar structure to methamphetamine (methA). In this study, we tested the effect of methA analogs on the glutamatergic synaptic transmission on primary cultured cortical neurons of SpragueDawley (SD) rats and C57BL/6 mice, and also layer 2/3 pyramidal neurons of the medial prefrontal cortex (mPFC) of C57BL/6 mice. We found that acute treatment with 4-EA-NBOMe inhibits spontaneous excitatory postsynaptic currents (EPSCs) and that withdrawal after chronic inhibition by 4-EA-NBOMe augments glutamatergic synaptic transmission. These modifications of synaptic responses are mediated by 5-HT1A receptors. These findings suggest that 4-EA-NBOMe directly affects the central nervous system by changing the efficacy of glutamatergic synaptic transmission.
Collapse
Affiliation(s)
- Hyun-A Oh
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, South Korea
| | - Jae Hong Yoo
- Department of Biological Sciences, Chungnam National University, Daejeon 34134 South Korea
| | - Ye-Ji Kim
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, South Korea; Human and Environmental Toxicology, University of Science and Technology, Daejeon 34114, South Korea
| | - Kyung-Seok Han
- Department of Biological Sciences, Chungnam National University, Daejeon 34134 South Korea.
| | - Dong Ho Woo
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, South Korea; Human and Environmental Toxicology, University of Science and Technology, Daejeon 34114, South Korea.
| |
Collapse
|
4
|
Davis MT, DellaGiogia N, Maruff P, Pietrzak RH, Esterlis I. Acute cognitive effects of single-dose intravenous ketamine in major depressive and posttraumatic stress disorder. Transl Psychiatry 2021; 11:205. [PMID: 33833217 PMCID: PMC8032778 DOI: 10.1038/s41398-021-01327-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 01/19/2021] [Accepted: 03/17/2021] [Indexed: 02/02/2023] Open
Abstract
Intravenous (IV) subanesthetic doses of ketamine have been shown to reduce psychiatric distress in both major depressive (MDD) and posttraumatic stress disorder (PTSD). However, the effect of ketamine on cognitive function in these disorders is not well understood. To address this gap, we examined the effect of a single dose of IV ketamine on cognition in individuals with MDD and/or PTSD relative to healthy controls (HC). Psychiatric (n = 29; 15 PTSD, 14 MDD) and sex- age- and IQ matched HC (n = 29) groups were recruited from the community. A single subanesthetic dose of IV ketamine was administered. Mood and cognitive measures were collected prior to, 2 h and 1 day post-ketamine administration. MDD/PTSD individuals evidenced a large-magnitude improvement in severity of depressive symptoms at both 2-hours and 1 day post-ketamine administration (p's < .001, Cohen d's = 0.80-1.02). Controlling for baseline performance and years of education, IV ketamine induced declines in attention (ATTN), executive function (EF), and verbal memory (VM) 2 h post-administration, all of which had resolved by 1 day post-ketamine across groups. The magnitude of cognitive decline was significantly larger in MDD/PTSD relative to HC on attention only (p = .012, d = 0.56). Ketamine did not affect working memory (WM) performance. Cognitive function (baseline, change from baseline to post-ketamine) was not associated with antidepressant response to ketamine. Results suggest that while ketamine may have an acute deleterious effect on some cognitive domains in both MDD/PTSD and HC individuals, most notably attention, this reduction is transient and there is no evidence of ketamine-related cognitive dysfunction at 1 day post-administration.
Collapse
Affiliation(s)
- Margaret T Davis
- Yale University School of Medicine, Department of Psychiatry, New Haven, USA
- Yale University, Department of Psychology, New Haven, USA
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Washington, DC, USA
| | - Nicole DellaGiogia
- Yale University School of Medicine, Department of Psychiatry, New Haven, USA
| | | | - Robert H Pietrzak
- Yale University School of Medicine, Department of Psychiatry, New Haven, USA
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Washington, DC, USA
| | - Irina Esterlis
- Yale University School of Medicine, Department of Psychiatry, New Haven, USA.
- Yale University, Department of Psychology, New Haven, USA.
- U.S. Department of Veterans Affairs, National Center for Posttraumatic Stress Disorder, Washington, DC, USA.
| |
Collapse
|
5
|
Rinaldi R, Bersani G, Marinelli E, Zaami S. The rise of new psychoactive substances and psychiatric implications: A wide-ranging, multifaceted challenge that needs far-reaching common legislative strategies. Hum Psychopharmacol 2020; 35:e2727. [PMID: 32144953 DOI: 10.1002/hup.2727] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/13/2020] [Accepted: 02/17/2020] [Indexed: 12/21/2022]
Abstract
The ever-growing number of novel psychoactive substances (NPS) that have been surfacing globally, as well as related changes in drug abuse trends, undoubtedly constitute a difficult and multifaceted challenge for psychiatry. The intake and abuse of such substances has been linked to a risk of psychopathological disturbances, which stem from imbalances of a range of neurotransmitter pathways and receptors. Through an analysis of relevant research articles and reviews (particularly those outlining NPS neurological and cerebral mechanisms of action and psychopathological consequences arising from NPS abuse; research papers more closely focused on chemical/pharmacological aspects have been ruled out), through a systematic analysis of Pubmed, Medline, PsycLIT and EMBASE literature, as well as data released by health care institutions and drug enforcement agencies (among which the World Health Organization, the United Nations Office on Drugs and Crime, the European Monitoring Centre for Drugs and Drug Addiction, Eurojust, the Novel Psychoactive Treatment UK Network, the Court of Justice of the European Union), the authors aimed to elaborate on the most relevant data relative to NPS-related psychiatric effects, focusing on the conceptual and definition-related complexities inherent to NPS, clinical management and motivations for NPS use; moreover, an effort has been made to highlight the possible measures in order to tackle the unremitting rise of such elusive and potentially harmful substances.
Collapse
Affiliation(s)
- Raffaella Rinaldi
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Departmental Section of Legal Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Giuseppe Bersani
- Department of Medico-Surgical Sciences and Biotechnologies, "Sapienza" University of Rome, Rome, Italy
| | - Enrico Marinelli
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Departmental Section of Legal Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Simona Zaami
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Departmental Section of Legal Medicine, "Sapienza" University of Rome, Rome, Italy
| |
Collapse
|
6
|
Costa G, De Luca MA, Piras G, Marongiu J, Fattore L, Simola N. Neuronal and peripheral damages induced by synthetic psychoactive substances: an update of recent findings from human and animal studies. Neural Regen Res 2020; 15:802-816. [PMID: 31719240 PMCID: PMC6990793 DOI: 10.4103/1673-5374.268895] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Preclinical and clinical studies indicate that synthetic psychoactive substances, in addition to having abuse potential, may elicit toxic effects of varying severity at the peripheral and central levels. Nowadays, toxicity induced by synthetic psychoactive substances poses a serious harm for health, since recreational use of these substances is on the rise among young and adult people. The present review summarizes recent findings on the peripheral and central toxicity elicited by “old” and “new” synthetic psychoactive substances in humans and experimental animals, focusing on amphetamine derivatives, hallucinogen and dissociative drugs and synthetic cannabinoids.
Collapse
Affiliation(s)
- Giulia Costa
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Maria Antonietta De Luca
- Department of Biomedical Sciences; National Institute of Neuroscience (INN), University of Cagliari, Cagliari, Italy
| | - Gessica Piras
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Jacopo Marongiu
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Liana Fattore
- National Research Council of Italy, Institute of Neuroscience, Cagliari, Italy
| | - Nicola Simola
- Department of Biomedical Sciences; National Institute of Neuroscience (INN), University of Cagliari, Cagliari, Italy
| |
Collapse
|
7
|
Strong C, Kabbaj M. On the safety of repeated ketamine infusions for the treatment of depression: Effects of sex and developmental periods. Neurobiol Stress 2018; 9:166-175. [PMID: 30450382 PMCID: PMC6236511 DOI: 10.1016/j.ynstr.2018.09.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 08/23/2018] [Accepted: 09/04/2018] [Indexed: 12/16/2022] Open
Abstract
In this review, we will discuss the safety of repeated treatments with ketamine for patients with treatment-resistant depression (TRD), a condition in which patients with major depression do not show any clinical improvements following treatments with at least two antidepressant drugs. We will discuss the effects of these treatments in both sexes at different developmental periods. Numerous small clinical studies have shown that a single, low-dose ketamine infusion can rapidly alleviate depressive symptoms and thoughts of suicidality in patients with TRD, and these effects can last for about one week. Interestingly, the antidepressant effects of ketamine can be prolonged with intermittent, repeated infusion regimens and produce more robust therapeutic effects when compared to a single infusion. The safety of such repeated treatments with ketamine has not been thoroughly investigated. Although more studies are needed, some clinical and preclinical reports indicated that repeated infusions of low doses of ketamine may have addictive properties, and suggested that adolescent and adult female subjects may be more sensitive to ketamine's addictive effects. Additionally, during ketamine infusions, many TRD patients report hallucinations and feelings of dissociation and depersonalization, and therefore the effects of repeated treatments of ketamine on cognition must be further examined. Some clinical reports indicated that, compared to women, men are more sensitive to the psychomimetic effects of ketamine. Preclinical studies extended these findings to both adolescent and adult male rodents and showed that male rodents at both developmental periods are more sensitive to ketamine's cognitive-altering effects. Accordingly, in this review we shall focus our discussion on the potential addictive and cognitive-impairing effects of repeated ketamine infusions in both sexes at two important developmental periods: adolescence and adulthood. Although more work about the safety of ketamine is warranted, we hope this review will bring some answers about the safety of treating TRD with repeated ketamine infusions.
Collapse
Affiliation(s)
| | - Mohamed Kabbaj
- Corresponding author. Florida State University, 3300-H, 1115 W. Call St, Tallahassee, FL, 32306, USA.
| |
Collapse
|
8
|
Zuo D, Liu Y, Liu Z, Cui J, Zhou X, Liu Y, Li Z, Wu Y. Alcohol aggravates ketamine-induced behavioral, morphological and neurochemical alterations in adolescent rats: The involvement of CREB-related pathways. Behav Brain Res 2018; 349:80-90. [PMID: 29738804 DOI: 10.1016/j.bbr.2018.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/17/2018] [Accepted: 05/03/2018] [Indexed: 01/03/2023]
Abstract
Currently, an increasing proportion of adolescent ketamine users simultaneously consume alcohol. However, the potential behavioural and neurological alterations induced by such a drug combination and the underlying mechanisms have not been systematically examined. Therefore, in the present study, the behavioural and morphological changes and the underlying mechanisms were studied in adolescent rats after repeated alcohol and/or ketamine treatment. This study provided the first evidence that co-administration of alcohol (2 and 4 g/kg, i.g.) in adolescent rats significantly potentiated the neurotoxic properties of repeated ketamine (30 mg/kg, i.p.) treatments over 14 days, manifesting as increased locomotor activity, stereotypic behaviour, ataxia and morphological changes. This potentiation was associated with the enhancement by alcohol of ketamine-induced glutamate (Glu) and dopamine (DA) release in the cortex and hippocampus. Further mechanistic study demonstrated that alcohol potentiated ketamine-induced neurotoxicity through down-regulation of Akt (a serine/threonine kinase or protein kinase, PKB), protein kinase A (PKA), calmodulin-dependent kinase IV (CaMK-IV)-mediated cyclic AMP-responsive element binding protein (CREB) pathways and induction of neuronal apoptosis in the cortex and hippocampus of the adolescent rats. As this study provides strong evidence that repeated alcohol and ketamine co-exposure may cause serious neurotoxicity, attention needs to be drawn to the potential risk of this consumption behaviour, especially for adolescents.
Collapse
Affiliation(s)
- Daiying Zuo
- Department of pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China.
| | - Yumiao Liu
- Department of pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Zi Liu
- Department of pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Jiahui Cui
- Department of pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Xuejiao Zhou
- Department of pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Yang Liu
- Department of pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Zengqiang Li
- Department of pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Yingliang Wu
- Department of pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China.
| |
Collapse
|
9
|
Papazoglou A, Lundt A, Wormuth C, Ehninger D, Henseler C, Soós J, Broich K, Weiergräber M. Non-restraining EEG Radiotelemetry: Epidural and Deep Intracerebral Stereotaxic EEG Electrode Placement. J Vis Exp 2016. [PMID: 27404845 DOI: 10.3791/54216] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Implantable EEG radiotelemetry is of central relevance in the neurological characterization of transgenic mouse models of neuropsychiatric and neurodegenerative diseases as well as epilepsies. This powerful technique does not only provide valuable insights into the underlying pathophysiological mechanisms, i.e., the etiopathogenesis of CNS related diseases, it also facilitates the development of new translational, i.e., therapeutic approaches. Whereas competing techniques that make use of recorder systems used in jackets or tethered systems suffer from their unphysiological restraining to semi-restraining character, radiotelemetric EEG recordings overcome these disadvantages. Technically, implantable EEG radiotelemetry allows for precise and highly sensitive measurement of epidural and deep, intracerebral EEGs under various physiological and pathophysiological conditions. First, we present a detailed protocol of a straight forward, successful, quick and efficient technique for epidural (surface) EEG recordings resulting in high-quality electrocorticograms. Second, we demonstrate how to implant deep, intracerebral EEG electrodes, e.g., in the hippocampus (electrohippocampogram). For both approaches, a computerized 3D stereotaxic electrode implantation system is used. The radiofrequency transmitter itself is implanted into a subcutaneous pouch in both mice and rats. Special attention also has to be paid to pre-, peri- and postoperative treatment of the experimental animals. Preoperative preparation of mice and rats, suitable anesthesia as well as postoperative treatment and pain management are described in detail.
Collapse
Affiliation(s)
- Anna Papazoglou
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM)
| | - Andreas Lundt
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM)
| | - Carola Wormuth
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM)
| | - Dan Ehninger
- Molecular and Cellular Cognition Lab, German Center for Neurodegenerative Diseases (Deutsches Zentrum für Neurodegenerative Erkrankungen, DZNE)
| | - Christina Henseler
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM)
| | - Julien Soós
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM)
| | - Karl Broich
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM)
| | - Marco Weiergräber
- Department of Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM);
| |
Collapse
|
10
|
Dodelet-Devillers A, Zullian C, Beaudry F, Gourdon J, Chevrette J, Hélie P, Vachon P. Physiological and pharmacokinetic effects of multilevel caging on Sprague Dawley rats under ketamine-xylazine anesthesia. Exp Anim 2016; 65:383-392. [PMID: 27263962 PMCID: PMC5111841 DOI: 10.1538/expanim.16-0026] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
While the cage refinement is a necessary step towards improving the welfare of research rats, increasing the complexity and surface area of the living space of an animal may have physiological impacts that need to be taken into consideration. In this study, ketamine (80 mg/kg) and xylazine (10 mg/kg) caused a short duration anesthesia that was significantly decreased in Sprague-Dawley rats housed in multilevel cages (MLC), compared to rats housed in standard cages (SDC). The withdrawal reflex, the palpebral reflexes and the time-to-sternal all occurred earlier in MLC housed rats, suggesting an effect of housing on the physiology of the rats. In addition, during anesthesia, cardiac frequencies were increased in animals housed in the smaller SDC. Respiratory frequencies, the blood oxygen saturation and rectal temperatures during anesthesia did not vary between conditions during the anesthesia. While xylazine pharmacokinetics were unchanged with caging conditions, the clearance and half-lives of ketamine and its metabolite, norketamine, were altered in the rats housed in MLC. Finally, while no difference was ultimately seen in rat body weights, isolated liver and adrenal gland weights were significantly lighter in rats housed in the MLC. Increasing cage sizes, while having a positive impact on wellbeing in rats, can alter anesthetic drug metabolism and thus modify anesthesia parameters and associated physiological processes.
Collapse
Affiliation(s)
- Aurore Dodelet-Devillers
- Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
11
|
Malhi GS, Byrow Y, Cassidy F, Cipriani A, Demyttenaere K, Frye MA, Gitlin M, Kennedy SH, Ketter TA, Lam RW, McShane R, Mitchell AJ, Ostacher MJ, Rizvi SJ, Thase ME, Tohen M. Ketamine: stimulating antidepressant treatment? BJPsych Open 2016; 2:e5-e9. [PMID: 27703782 PMCID: PMC4995167 DOI: 10.1192/bjpo.bp.116.002923] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 03/29/2016] [Accepted: 03/31/2016] [Indexed: 12/16/2022] Open
Abstract
SUMMARY The appeal of ketamine - in promptly ameliorating depressive symptoms even in those with non-response - has led to a dramatic increase in its off-label use. Initial promising results await robust corroboration and key questions remain, particularly concerning its long-term administration. It is, therefore, timely to review the opinions of mood disorder experts worldwide pertaining to ketamine's potential as an option for treating depression and provide a synthesis of perspectives - derived from evidence and clinical experience - and to consider strategies for future investigations. DECLARATION OF INTERESTS G.S.M. Grant/research support: National Health Medical Research Council, NSW Health, Ramsay Health, American Foundation for Suicide Prevention, AstraZeneca, Eli Lilly & Co, Organon, Pfizer, Servier, and Wyeth; has been a speaker for Abbott, AstraZeneca, Eli Lilly & Co, Janssen Cilag, Lundbeck, Pfizer, Ranbaxy, Servier, and Wyeth; consultant: AstraZeneca, Eli Lilly & Co, Janssen Cilag, Lundbeck, and Servier. M.A.F. Grant support: AssureRx, Janssen Research & Development, Mayo Foundation, Myriad, National Institute of Alcohol Abuse and Alcoholism (NIAAA), National Institute of Mental Health (NIMH), Pfizer. Consultant (Mayo): Janssen Research & Development, LLC, Mitsubishi Tanabe Pharma Corporation, Myriad Genetics, Neuralstem Inc., Sunovion, Supernus Pharmaceuticals, Teva Pharmaceuticals. CME/travel support: American Physician Institute, CME Outfitters. Financial interest/Mayo Clinic 2016: AssureRx. S.H.K. Grant/research support: Brain Canada, Bristol Meyer Squibb, CIHR, Janssen, Johnson & Johnson, Lundbeck, Ontario Brain Institute, Pfizer, Servier, St. Jude Medical, Sunovion. T.A.K. Grant/research support (through Stanford University): Sunovion Pharmaceuticals and Merck & Co., Inc.; consultant/advisory board bember: Allergan, Inc., Janssen Pharmaceuticals, Myriad Genetic Laboratories, Inc., and Sunovion Pharmaceuticals; lecture honoraria (not Speaker's Bureau payments): GlaxoSmithKline, and Sunovion Pharmaceuticals; royalties from American Psychiatric Publishing, Inc. Also, AstraZeneca Pharmaceuticals LP provided publication support to Parexel for preparation of a manuscript. Spouse employee and stockholder of Janssen Pharmaceuticals. R.W.L. Honoraria for speaking/advising/consulting, and/or received research funds: AstraZeneca, Brain Canada, Bristol Myers Squibb, Canadian Institutes of Health Research, Canadian Depression Research and Intervention Network, Canadian Network for Mood and Anxiety Treatments, Canadian Psychiatric Association, Coast Capital Savings, Johnson and Johnson, Lundbeck, Lundbeck Institute, Pfizer, Servier, St. Jude Medical, Takeda University, Health Network Foundation, and Vancouver Coastal Health Research Institute. R.M. Investigator Janssen trials of esketamine; 'paid-for' ketamine clinic operated by Oxford Health NHS Foundation Trust - fees used to support the Trust. M.J.O. Consultant: Sunovion and Acadia Pharmaceuticals. Full-time employee of U.S. Department of Veterans Affairs. M.E.T. Advisory/Consultant: Alkermes, Allergan, AstraZeneca, Bristol-Myers Squibb Company, Cerecor inc., Eli Lilly & Co., Forest Laboratories, Gerson Lehrman Group, Fabre-Kramer Pharmaceuticals, Inc., GlaxoSmithKline, Guidepoint Global, H. Lundbeck A/S, MedAvante Inc., Merck and Co. Inc. (formerly Schering Plough and Organon), Moksha8, Naurex Inc., Neuronetics Inc., Novartis, Ortho-McNeil Pharmaceuticals (Johnson & Johnson; Janssen), Otsuka, Pamlab, L.L.C. (Nestle), Pfizer (formerly Wyeth Ayerst Pharmaceuticals), Shire US Inc., Sunovion Pharmaceuticals Inc., Trius Therapeutical Inc. and Takeda. Grant support: Agency for Healthcare Research and Quality, Alkermes, AssureRx, Avanir, Forest Pharmaceuticals, Janssen, National Institute of Mental Health, and Otsuka Pharmaceuticals. Speakers Bureau: None since June, 2010. Equity Holdings: MedAvante, Inc. Royalties: American Psychiatric Foundation, Guilford Publications, Herald House, W.W. Norton & Company, Inc. Spouse's employment: Peloton Advantage, which does business with Pfizer. M.T. Full-time employee at Lilly 1997 to 2008. Honoraria/consulted: Abbott, AstraZeneca, Bristol Myers Squibb, GlaxoSmithKline, Lilly, Johnson & Johnson, Allergan, Otsuka, Merck, Sunovion, Forest, Geodon Richter Plc, Roche, Elan, Alkermes, Lundbeck, Teva, Pamlab, Minerva, Wyeth and Wiley Publishing. Spouse was full time-employee at Lilly 1998-2013. COPYRIGHT AND USAGE © The Royal College of Psychiatrists 2016. This is an open access article distributed under the terms of the Creative Commons Non-Commercial, No Derivatives (CC BY-NC-ND) licence.
Collapse
Affiliation(s)
- Gin S Malhi
- , MD, Department of Psychiatry, Kolling Institute, Sydney Medical School, University of Sydney, Sydney, NSW, Australia; CADE Clinic, Department of Psychiatry, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Yulisha Byrow
- , PhD, CADE Clinic, Department of Psychiatry, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Frederick Cassidy
- , MD, Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| | - Andrea Cipriani
- , PhD, Department of Psychiatry, University of Oxford, Oxford, UK; Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Koen Demyttenaere
- , PhD, KU Leuven, Department of Neurosciences, Research Group of Psychiatry, University Psychiatric Center, Leuven, Belgium
| | - Mark A Frye
- , MD, Psychiatry, Mayo Clinic Depression Center, Mayo Clinic, Rochester, MN, USA
| | - Michael Gitlin
- , MD, Department of Psychiatry, Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Sidney H Kennedy
- , MD, Arthur Sommer Rotenberg Chair in Suicide & Depression Studies, St. Michael's Hospital; Professor of Psychiatry, University Health Network and Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Terence A Ketter
- , MD, Psychiatry and Behavioral Sciences, Chief, Bipolar Disorders Clinic, Stanford University School of Medicine, Stanford, CA, USA
| | - Raymond W Lam
- , MD, Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | | | - Alex J Mitchell
- , MD, Liaison Psychiatry and Psycho-Oncology, University of Leicester, Leicester, UK
| | - Michael J Ostacher
- , MD, Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Sakina J Rizvi
- , PhD, Li Ka Shing Knowledge Institute, ASR Suicide and Depression Studies Program, St. Michael's Hospital; Department of Psychiatry and Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Michael E Thase
- , MD, Department of Psychiatry, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA, USA; Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
| | - Mauricio Tohen
- , MD, Professor and Chairman of the Department of Psychiatry & Behavioural Sciences at the University of New Mexico, Albuquerque, NM, USA
| |
Collapse
|
12
|
EEG Radiotelemetry in Small Laboratory Rodents: A Powerful State-of-the Art Approach in Neuropsychiatric, Neurodegenerative, and Epilepsy Research. Neural Plast 2015; 2016:8213878. [PMID: 26819775 PMCID: PMC4706962 DOI: 10.1155/2016/8213878] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 08/27/2015] [Indexed: 02/04/2023] Open
Abstract
EEG radiotelemetry plays an important role in the neurological characterization of transgenic mouse models of neuropsychiatric and neurodegenerative diseases as well as epilepsies providing valuable insights into underlying pathophysiological mechanisms and thereby facilitating the development of new translational approaches. We elaborate on the major advantages of nonrestraining EEG radiotelemetry in contrast to restraining procedures such as tethered systems or jacket systems containing recorders. Whereas a main disadvantage of the latter is their unphysiological, restraining character, telemetric EEG recording overcomes these disadvantages. It allows precise and highly sensitive measurement under various physiological and pathophysiological conditions. Here we present a detailed description of a straightforward successful, quick, and efficient technique for intraperitoneal as well as subcutaneous pouch implantation of a standard radiofrequency transmitter in mice and rats. We further present computerized 3D-stereotaxic placement of both epidural and deep intracerebral electrodes. Preoperative preparation of mice and rats, suitable anaesthesia, and postoperative treatment and pain management are described in detail. A special focus is on fields of application, technical and experimental pitfalls, and technical connections of commercially available radiotelemetry systems with other electrophysiological setups.
Collapse
|
13
|
A single intraperitoneal injection of ketamine does not affect spatial working, reference memory or neurodegeneration in adult mice: An animal study. Eur J Anaesthesiol 2015; 30:618-26. [PMID: 23736087 DOI: 10.1097/eja.0b013e3283610321] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Ketamine is an anaesthetic and analgesic drug used in research and clinical practice. Little is known about the effects of different doses of this drug on memory and brain cellular death. OBJECTIVE To study the effects of different doses of ketamine on working and reference memory, and neurodegeneration in adult mice. DESIGN A randomised study. SETTINGS The study was carried out in a basic science laboratory, between March 2011 and August 2012. ANIMALS Forty-eight 7-month-old, male C57BL/6 mice were used. INTERVENTION Animals received a single intraperitoneal injection of physiological saline solution or one of three doses of ketamine (25, 75 or 150 mg kg(-1)). Each group consisted of 12 animals (seven animals for behavioural tests and five animals for histopathological and immunohistochemical studies). The animals used for histopathology studies were sacrificed 3 h after anaesthesia. MAIN OUTCOME MEASURES Working and reference memories were assessed using the radial-maze test over 12 consecutive days. The equilibrium was tested using the vertical pole (4 and 24 h after injection), whereas locomotion was assessed using the open field (24, 48 and 72 h after injection). Histopathological (haematoxylin-eosin staining) and immunohistochemical analyses (procaspase-3 and activated caspase-3 detections) were performed 3 h after injection to assess neurodegeneration in the retrosplenial and visual cortices, pyramidal cell layer of the cornu Ammonis 1 and cornu Ammonis 3 areas of the hippocampus, in the granular layer of the dentate gyrus, in the laterodorsal thalamic nucleus, striatum and accumbens nucleus. RESULTS No significant differences were observed between the groups regarding the number of dead cells and cells showing positive immune-reactivity in the different regions of the brain studied. The performance in the vertical pole test and the number of reference and working memory errors in the radial-maze were similar in all groups. Nevertheless, the animals treated with ketamine 75 mg kg(-1) were transiently more active, walking a greater total distance at a greater speed in the open field than other groups (power of 0.96). CONCLUSION These data indicate that a single intraperitoneal injection of ketamine at subanaesthetic and anaesthetic doses does not impair working memory, reference memory or neurodegeneration in adult mice, but an intermediate dose of ketamine produces transitory hyperlocomotion.
Collapse
|
14
|
Giroux MC, Hélie P, Burns P, Vachon P. Anesthetic and pathological changes following high doses of ketamine and xylazine in Sprague Dawley rats. Exp Anim 2015; 64:253-60. [PMID: 25818316 PMCID: PMC4547998 DOI: 10.1538/expanim.14-0088] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The main objective of this study was to compare the effects of ketamine and xylazine in aging rats when coadministered intraperitoneally at high anesthetic doses. Three groups (n=6 rats/group) consisting of rats at 3, 6 and 12 months of age were used. During anesthesia, animals were monitored for heart rate, respiratory frequency, blood oxygen saturation, and rectal temperature. The corneal and paw withdrawal reflex were also examined during anesthesia. During anesthesia, withdrawal and corneal reflexes were absent for progressively longer durations with increasing age. Significant decreases in cardiac and respiratory frequency and, blood oxygen saturation occurred for the 6- and 12-month-old animals. Respiratory frequency and blood oxygen saturation returned to normal at the end of the anesthesia; however, the significant decrease in cardiac frequency persisted in the 6- and 12-month-old animals. Rectal temperature was decreased significantly only in the 3-month-old animals. Pulmonary edema and effusion occurred in 50% of the 12-month-old animals. In conclusion, if ketamine-xylazine are used for anesthesia, the doses should be optimized for the age of the subjects prior to initiation of the research project.
Collapse
Affiliation(s)
- Marie-Chantal Giroux
- Faculty of Veterinary Medicine, Departments of Veterinary Biomedicine, University of Montreal, 3200 rue Sicotte, Saint-Hyacinthe, Quebec, J2S 2M2, Canada
| | | | | | | |
Collapse
|
15
|
Niciu MJ, Henter ID, Luckenbaugh DA, Zarate CA, Charney DS. Glutamate receptor antagonists as fast-acting therapeutic alternatives for the treatment of depression: ketamine and other compounds. Annu Rev Pharmacol Toxicol 2014; 54:119-39. [PMID: 24392693 DOI: 10.1146/annurev-pharmtox-011613-135950] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The N-methyl-D-aspartate (NMDA) receptor antagonist ketamine has rapid and potent antidepressant effects in treatment-resistant major depressive disorder and bipolar depression. These effects are in direct contrast to the more modest effects seen after weeks of treatment with classic monoaminergic antidepressants. Numerous open-label and case studies similarly validate ketamine's antidepressant properties. These clinical findings have been reverse-translated into preclinical models in an effort to elucidate ketamine's antidepressant mechanism of action, and three important targets have been identified: mammalian target of rapamycin (mTOR), eukaryotic elongation factor 2 (eEF2), and glycogen synthase kinase-3 (GSK-3). Current clinical and preclinical research is focused on (a) prolonging/maintaining ketamine's antidepressant effects, (b) developing more selective NMDA receptor antagonists free of ketamine's adverse effects, and (c) identifying predictor, mediator/moderator, and treatment response biomarkers of ketamine's antidepressant effects.
Collapse
Affiliation(s)
- Mark J Niciu
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institutes of Health/National Institute of Mental Health, Bethesda, Maryland 20814-9692;
| | | | | | | | | |
Collapse
|
16
|
Featherstone R, Nagy L, Hahn C, Siegel S. Juvenile exposure to ketamine causes delayed emergence of EEG abnormalities during adulthood in mice. Drug Alcohol Depend 2014; 134:123-127. [PMID: 24210161 PMCID: PMC4009692 DOI: 10.1016/j.drugalcdep.2013.09.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 09/12/2013] [Accepted: 09/17/2013] [Indexed: 01/15/2023]
Abstract
BACKGROUND Increased susceptibility to cognitive impairment or psychosis in adulthood is associated with adolescent drug abuse. Studies in adults have identified impairments in attention and memory, and changes in EEG, as common consequences of ketamine abuse. In contrast, the effects of ketamine on the juvenile brain have not been extensively tested. This is a significant omission, since abuse of ketamine is often observed within this age group. OBJECTIVES Juvenile mice (4-6 weeks of age) were administered ketamine (20mg/kg) for 14 days. EEG was assessed in response to auditory stimulation both at one week following ketamine exposure at 7 weeks of age (juvenile) and again at 12 weeks of age (adult). EEG was analyzed for baseline activity, event-related power and event-related potentials (ERPs). RESULTS While no effects of ketamine exposure were observed during the juvenile period, significant reductions in amplitude of the P20 ERP component and event-related gamma power were seen following ketamine when re-tested as adults. In contrast, reductions in event-related theta were seen in ketamine-exposed mice at both time points. CONCLUSIONS Age related deficits in electrophysiological components such as P20 or event-related gamma may be due to an interruption of normal neural maturation. Reduction of NMDAR signaling during adolescence leads to delayed-onset disruption of gamma oscillations and the P20 component of the ERP. Further, delayed onset of impairment following adolescent ketamine abuse suggests that methods could be developed to detect and treat the early effects of drug exposure prior to the onset of disability.
Collapse
Affiliation(s)
- R.E. Featherstone
- Translational Neuroscience Program, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA
| | - L.R. Nagy
- Translational Neuroscience Program, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA
| | - C.G. Hahn
- Molecular Signaling Program, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA
| | - S.J. Siegel
- Translational Neuroscience Program, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA,Corresponding Author: S.J. Siegel, Professor, University of Pennsylvania, Psychiatry, 125 S 31st street, rm 2202, Philadelphia, UNITED STATES, 2155730278,
| |
Collapse
|
17
|
Veilleux-Lemieux D, Castel A, Carrier D, Beaudry F, Vachon P. Pharmacokinetics of ketamine and xylazine in young and old Sprague-Dawley rats. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2013; 52:567-570. [PMID: 24041212 PMCID: PMC3784662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 01/28/2013] [Accepted: 03/12/2013] [Indexed: 06/02/2023]
Abstract
To compare the pharmacokinetics of coadministered intraperitoneal ketamine and xylazine in young (8 to 10 wk; n = 6) and old rats (2 to 2.4 y; n = 6), blood samples obtained at 15 and 30 min and 1, 2, and 4 h after drug administration were analyzed by HPLC-tandem mass spectrometry. In both groups, the withdrawal reflex was absent during anesthesia and was present at 1.1 (± 0.2) and 2.6 (± 0.7) h after drug administration in young and old rats, respectively, with the first voluntary movement at 1.5 ± 0.2 and 4.9 ± 1.0 h. Drug availability of ketamine and xylazine was 6.0 and 6.7 times greater, respectively, in old than young rats. The rate constant of elimination of both drugs was greatly decreased and the elimination half-life was significantly greater in old compared with young rats. In conclusion, age and associated factors affect the availability of ketamine and xylazine when coadministered to attain clinical anesthesia, changing the pharmacokinetics of these drugs and prolonging anesthesia duration and recovery times with aging. Compared with their young counterparts, aged rats required much higher doses to attain a similar level of anesthesia. Finally, the long half-life of both ketamine and xylazine, when coadministered to old rats, may be a factor in research protocols because residual plasma concentrations could still be present for as long as 3 and 5 d, respectively, after administration.
Collapse
Affiliation(s)
| | - Aude Castel
- Faculty of Veterinary Medicine, Department of Veterinary Biomedicine, Saint-Hyacinthe, Quebec, Canada
| | | | - Francis Beaudry
- Faculty of Veterinary Medicine, Department of Veterinary Biomedicine, Saint-Hyacinthe, Quebec, Canada
| | - Pascal Vachon
- Faculty of Veterinary Medicine, Department of Veterinary Biomedicine, Saint-Hyacinthe, Quebec, Canada
| |
Collapse
|
18
|
Featherstone RE, Liang Y, Saunders JA, Tatard-Leitman VM, Ehrlichman RS, Siegel SJ. Subchronic ketamine treatment leads to permanent changes in EEG, cognition and the astrocytic glutamate transporter EAAT2 in mice. Neurobiol Dis 2012; 47:338-46. [DOI: 10.1016/j.nbd.2012.05.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2011] [Revised: 04/09/2012] [Accepted: 05/11/2012] [Indexed: 01/11/2023] Open
|
19
|
Ribeiro PO, Valentim AM, Rodrigues P, Olsson IA, Antunes LM. Apoptotic neurodegeneration and spatial memory are not affected by sedative and anaesthetics doses of ketamine/medetomidine combinations in adult mice. Br J Anaesth 2012; 108:807-14. [PMID: 22362669 DOI: 10.1093/bja/aes003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Ketamine is increasingly popular in clinical practice and its combination with α(2)-agonists can provide good anaesthetic stability. Little is known about the effects of this combination in the brain. Therefore, we investigated the effects of different concentrations of ketamine combined with medetomidine on cognition and its potential apoptotic neurodegenerative effect in adult mice. METHODS Seventy-eight C57BL/6 adult mice were divided into six different groups (saline solution, 1 mg kg(-1) medetomidine, 25 mg kg(-1) ketamine+1 mg kg(-1) medetomidine, 75 mg kg(-1) ketamine+1 mg kg(-1) medetomidine, 25 mg kg(-1) ketamine, and 75 mg kg(-1) ketamine). Eight animals per group were tested in the T-maze, vertical pole, and open-field test. Five animals per group were used for histopathological [haematoxylin and eosin (HE) staining] and immunohistochemical analyses [caspase-3 activation and expression of neurotrophin brain-derived neurotrophic factor (BDNF)]. Cells showing clear HE staining and positive immunoreactions for caspase-3 and BDNF in the retrosplenial cortex, visual cortex, pyramidal cell layer of the cornu Ammonis 1 and cornu Ammonis 3 areas of the hippocampus, and in the granular layer of the dentate gyrus were counted. RESULTS There were no differences between groups regarding the number of dead cells and cells showing positive immunoreactions in the different areas of the brain studied. Similarly, no differences were detected in the number of trials to complete the T-maze task. Nevertheless, α(2)-agonist decreased hyperlocomotion caused by ketamine in the open field. CONCLUSIONS Neither apoptotic neurodegeneration nor alterations in spatial memory were observed with different concentrations of ketamine combined with medetomidine in adult mice.
Collapse
Affiliation(s)
- P O Ribeiro
- Laboratory Animal Science, Instituto de Biologia Molecular e Celular, Rua do Campo Alegre 823, 4150-180 Porto, Portugal.
| | | | | | | | | |
Collapse
|
20
|
Bechtholt-Gompf AJ, Smith KL, John CS, Kang HH, Carlezon WA, Cohen BM, Ongür D. CD-1 and Balb/cJ mice do not show enduring antidepressant-like effects of ketamine in tests of acute antidepressant efficacy. Psychopharmacology (Berl) 2011; 215:689-95. [PMID: 21274700 DOI: 10.1007/s00213-011-2169-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Accepted: 01/03/2011] [Indexed: 11/30/2022]
Abstract
RATIONALE In patients, ketamine is a fast-acting antidepressant that can induce long-lasting symptom relief. Similar rapid effects have been reported in rodents, but reports of lasting effects are limited. OBJECTIVES We sought to extend past findings by examining dose-response curves that overlap with the individual doses previously reported to induce lasting effects in rodents and determining whether effects generalize to the tail suspension test (TST) and Balb/cJ mice. METHODS Using common tests of antidepressant efficacy we first confirmed our ability to detect the effects of desipramine, a well-characterized antidepressant drug. Next, we sought to determine whether two non-competitive NMDA antagonists, ketamine and MK-801, had long-lasting antidepressant-like effects in CD-1 mice, a strain that has often been used to demonstrate the short-term antidepressant-like effects of ketamine. Finally, we examined the short- and long-term effects of ketamine in a mouse strain that is more sensitive to antidepressant-like effects, Balb/cJ mice. RESULTS In CD-1 mice, desipramine treatment yielded significant short-term antidepressant-like effects in the TST and the forced swimming test (FST). However, no significant enduring effects of ketamine or MK-801 were observed 1 week later. Short-term effects of ketamine in the TST were observed in Balb/cJ mice, but lasting effects were absent 1 week later. CONCLUSIONS Although the TST and FST have been widely used to detect antidepressant-like effects in mice, they do not appear to be sensitive to long-lasting antidepressant-like effects of ketamine in mice and, therefore, do not model the therapeutic effects of ketamine that have been reported in humans with major depression.
Collapse
Affiliation(s)
- Anita J Bechtholt-Gompf
- Department of Psychiatry, Harvard Medical School-McLean Hospital, 115 Mill Street, MRC 215, Belmont, MA 02478, USA.
| | | | | | | | | | | | | |
Collapse
|
21
|
Bender C, de Olmos S, Bueno A, de Olmos J, Lorenzo A. Comparative analyses of the neurodegeneration induced by the non-competitive NMDA-receptor-antagonist drug MK801 in mice and rats. Neurotoxicol Teratol 2010; 32:542-50. [DOI: 10.1016/j.ntt.2010.05.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Revised: 05/03/2010] [Accepted: 05/05/2010] [Indexed: 10/19/2022]
|
22
|
Green SM, Coté CJ. Ketamine and Neurotoxicity: Clinical Perspectives and Implications for Emergency Medicine. Ann Emerg Med 2009; 54:181-90. [DOI: 10.1016/j.annemergmed.2008.10.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2008] [Revised: 09/23/2008] [Accepted: 10/01/2008] [Indexed: 10/21/2022]
|
23
|
Amann L, Halene T, Ehrlichman R, Luminais S, Ma N, Abel T, Siegel S. Chronic ketamine impairs fear conditioning and produces long-lasting reductions in auditory evoked potentials. Neurobiol Dis 2009; 35:311-7. [PMID: 19467327 PMCID: PMC2726963 DOI: 10.1016/j.nbd.2009.05.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Revised: 04/27/2009] [Accepted: 05/17/2009] [Indexed: 11/29/2022] Open
Abstract
Ketamine is an NMDA receptor antagonist with a variety of uses, ranging from recreational drug to pediatric anesthetic and chronic pain reliever. Despite its value in the clinical setting, little is known about the immediate and long-lasting effects of repeated ketamine treatment. We assessed the effects of chronic administration of a subanesthetic dose of ketamine on contextual fear conditioning, detection of pitch deviants and auditory gating. After four, but not two, weeks of daily ketamine injections, mice exhibited decreased freezing in the fear conditioning paradigm. Gating of the P80 component of auditory evoked potentials was also significantly altered by treatment condition, as ketamine caused a significant decrease in S1 amplitude. Additionally, P20 latency was significantly increased as a result of ketamine treatment. Though no interactions were found involving test week, stimulus and treatment condition, these results suggest that repeated ketamine administration impairs fear memory and has lasting effects on encoding of sensory stimuli.
Collapse
Affiliation(s)
- L.C. Amann
- SMRI Laboratory for Experimental Therapeutics in Psychiatry, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - T.B. Halene
- SMRI Laboratory for Experimental Therapeutics in Psychiatry, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- International Research Training Group 1328 Schizophrenia and Autism, Department of Psychiatry and Psychotherapy, RWTH Aachen University, Aachen, Germany
- Transdisciplinary Tobacco Use Research Center, University of Pennsylvania., Philadelphia, PA, USA
| | - R.S. Ehrlichman
- SMRI Laboratory for Experimental Therapeutics in Psychiatry, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - S.N. Luminais
- SMRI Laboratory for Experimental Therapeutics in Psychiatry, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - N. Ma
- Department of Biology, University of Pennsylvania., Philadelphia, PA, USA
| | - T. Abel
- Department of Biology, University of Pennsylvania., Philadelphia, PA, USA
| | - S.J. Siegel
- SMRI Laboratory for Experimental Therapeutics in Psychiatry, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Transdisciplinary Tobacco Use Research Center, University of Pennsylvania., Philadelphia, PA, USA
| |
Collapse
|
24
|
Bodarky CL, Halene TB, Ehrlichman RS, Banerjee A, Ray R, Hahn CG, Jonak G, Siegel SJ. Novel environment and GABA agonists alter event-related potentials in N-methyl-D-aspartate NR1 hypomorphic and wild-type mice. J Pharmacol Exp Ther 2009; 331:308-18. [PMID: 19602553 DOI: 10.1124/jpet.109.150938] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Clinical and experimental data suggest dysregulation of N-methyl-d-aspartate receptor (NMDAR)-mediated glutamatergic pathways in schizophrenia. The interaction between NMDAR-mediated abnormalities and the response to novel environment has not been studied. Mice expressing 5 to 10% of normal N-methyl-d-aspartate receptor subunit 1 (NR1) subunits [NR1(neo)(-/-)] were compared with wild-type littermates for positive deflection at 20 ms (P20) and negative deflection at 40 ms (N40) auditory event-related potentials (ERPs). Groups were tested for habituation within and across five testing sessions, with novel environment tested during a sixth session. Subsequently, we examined the effects of a GABA(A) positive allosteric modulator (chlordiazepoxide) and a GABA(B) receptor agonist (baclofen) as potential interventions to normalize aberrant responses. There was a reduction in P20, but not N40 amplitude within each habituation day. Although there was no amplitude or gating change across habituation days, there was a reduction in P20 and N40 amplitude and gating in the novel environment. There was no difference between genotypes for N40. Only NR1(neo)(-/-) mice had reduced P20 in the novel environment. Chlordiazepoxide increased N40 amplitude in wild-type mice, whereas baclofen increased P20 amplitude in NR1(neo)(-/-) mice. As noted in previous publications, the pattern of ERPs in NR1(neo)(-/-) mice does not recapitulate abnormalities in schizophrenia. In addition, reduced NR1 expression does not influence N40 habituation but does affect P20 in a novel environment. Thus, the pattern of P50 (positive deflection at 50 ms) but not N100 (negative deflection at 100 ms) in human studies may relate to subjects' reactions to unfamiliar environments. In addition, NR1 reduction decreased GABA(A) receptor-mediated effects on ERPs while causing increased GABA(B) receptor-mediated effects. Future studies will examine changes in GABA receptor subunits after reductions in NR1 expression.
Collapse
|
25
|
Halene TB, Ehrlichman RS, Liang Y, Christian EP, Jonak GJ, Gur TL, Blendy JA, Dow HC, Brodkin ES, Schneider F, Gur RC, Siegel SJ. Assessment of NMDA receptor NR1 subunit hypofunction in mice as a model for schizophrenia. GENES BRAIN AND BEHAVIOR 2009; 8:661-75. [PMID: 19563516 DOI: 10.1111/j.1601-183x.2009.00504.x] [Citation(s) in RCA: 131] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
N-methyl-D-aspartate receptors (NMDARs) play a pivotal role in excitatory neurotransmission, synaptic plasticity and brain development. Clinical and experimental evidence suggests a dysregulation of NMDAR function and glutamatergic pathways in the pathophysiology of schizophrenia. We evaluated electrophysiological and behavioral properties of NMDAR deficiency utilizing mice that express only 5-10% of the normal level of NMDAR NR1 subunit. Auditory and visual event related potentials yielded significantly increased amplitudes for the P20 and N40 components in NMDAR deficient (NR1(neo)-/-) mice suggesting decreased inhibitory tone. Compared to wild types, NR1(neo)-/- mice spent less time in social interactions and showed reduced nest building. NR1(neo)-/- mice displayed a preference for open arms of a zero maze and central zone of an open field, possibly reflecting decreased anxiety-related behavioral inhibition. However, locomotor activity did not differ between groups in either home cage environment or during behavioral testing. NR1(neo)-/- mice displayed hyperactivity only when placed in a large unfamiliar environment, suggesting that neither increased anxiety nor non-specific motor activation accounts for differential behavioral patterns. Data suggest that NMDAR NR1 deficiency causes disinhibition in sensory processing as well as reduced behavioral inhibition and impaired social interactions. The behavioral signature in NR1(neo)-/- mice supports the impact of impaired NMDAR function in a mouse model with possible relevance to negative symptoms in schizophrenia.
Collapse
Affiliation(s)
- T B Halene
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Response to the comment on Viberg et al. (2008) “Neonatal ketamine exposure results in changes in biochemical substrates of neuronal growth and synaptogenesis, and alters adult behavior irreversibly” by Ching-Hung Hsu. Toxicology 2008. [DOI: 10.1016/j.tox.2008.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|