1
|
Diep D, de la Salle S, Thibault Lévesque J, Lifshitz M, Garel N, Greenway KT. The ketamine chameleon: history, pharmacology, and the contested value of experience. Expert Rev Clin Pharmacol 2025:1-21. [PMID: 39868914 DOI: 10.1080/17512433.2025.2459377] [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: 11/24/2024] [Revised: 01/13/2025] [Accepted: 01/23/2025] [Indexed: 01/28/2025]
Abstract
INTRODUCTION Since its synthesis in 1962, ketamine has been widely used in diverse medical contexts, from anesthesia to treatment-resistant depression. However, interpretations of ketamine's subjective effects remain polarized. Biomedical frameworks typically construe the drug's experiential effects as dissociative or psychotomimetic, while psychedelic paradigms emphasize the potential therapeutic merits of these non-ordinary states. AREAS COVERED Ketamine's psychoactive effects have inspired diverse interpretations. In this review, we trace the historical evolution of these perspectives - which we broadly categorize as 'dissociative,' 'dream-like,' and 'psychedelic' - and show how they emerged out of these clinical contexts. We highlight the influence of factors such as language, dose, and environmental context on ketamine's effects and therapeutic outcomes. We discuss potential mechanisms underlying these context-dependent effects and explore the broader clinical and research-related ramifications. EXPERT OPINION Ketamine's subjective effects are undeniably powerful, yet their therapeutic significance remains debated. A nuanced, interdisciplinary approach is essential for maximizing ketamine's potential. Future research should focus on how explanatory models, treatment environments, and patient preparation can optimize ketamine's benefits while minimizing distress. We suggest that, rather than being a tiger to be tamed as its creator once described, ketamine may best be understood as a chameleon whose color shifts depending on its context.
Collapse
Affiliation(s)
- Danny Diep
- Department of Psychiatry, McGill University, Montréal, Québec, Canada
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Sara de la Salle
- Department of Psychiatry, McGill University, Montréal, Québec, Canada
- Lady Davis Institute at the Jewish General Hospital, Montréal, Québec, Canada
| | | | - Michael Lifshitz
- Department of Psychiatry, McGill University, Montréal, Québec, Canada
- Lady Davis Institute at the Jewish General Hospital, Montréal, Québec, Canada
| | - Nicolas Garel
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Department of Psychiatry and Addictology, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Kyle T Greenway
- Department of Psychiatry, McGill University, Montréal, Québec, Canada
- Lady Davis Institute at the Jewish General Hospital, Montréal, Québec, Canada
| |
Collapse
|
2
|
Santander O, Arredondo SB, García-Rojas F, Estay SF, Belforte JE, Chávez AE, Varela-Nallar L, Fuenzalida M. Ketamine administration during adolescence impairs synaptic integration and inhibitory synaptic transmission in the adult dentate gyrus. Prog Neurobiol 2025; 246:102718. [PMID: 39855537 DOI: 10.1016/j.pneurobio.2025.102718] [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: 06/04/2024] [Revised: 12/10/2024] [Accepted: 01/11/2025] [Indexed: 01/27/2025]
Abstract
Ketamine administration during adolescence affects cognitive performance; however, its long-term impact on synaptic function and neuronal integration in the hippocampus a brain region critical for cognition remains unclear. Using functional and molecular analyses, we found that chronic ketamine administration during adolescence exerts long-term effects on synaptic integration, expanding the temporal window in an input-specific manner affecting the inner molecular layer but not the medial perforant path inputs in the adult mouse dorsal hippocampal dentate gyrus. Ketamine also alters the excitatory/inhibitory balance by reducing the efficacy of inhibitory inputs likely due to a reduction in parvalbumin-positive interneurons number and function. These findings indicate that during adolescence, ketamine exerts a strong effect on inhibitory synaptic function mediated by parvalbumin-positive neurons that ultimately impact synaptic integration in the dorsal adult dentate gyrus, which could help to understand the neurobiological and functional bases that confer greater vulnerability to the adolescent brain.
Collapse
Affiliation(s)
- Odra Santander
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile; Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Sebastián B Arredondo
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Science, Universidad Andres Bello, Santiago, Chile
| | - Francisca García-Rojas
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Sebastián F Estay
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2340000, Chile; Instituto de Neurociencias y Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Juan E Belforte
- Instituto de Fisiología y Biofísica "Bernardo Houssay" (IFIBIO-Houssay), Grupo de Neurociencia de Sistemas, Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires 1121, Argentina
| | - Andrés E Chávez
- Instituto de Neurociencias y Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Lorena Varela-Nallar
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Science, Universidad Andres Bello, Santiago, Chile; Millennium Nucleus of Neuroepigenetics and Plasticity (EpiNeuro), Santiago, Chile
| | - Marco Fuenzalida
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Universidad de Valparaíso, Valparaíso 2340000, Chile; Millennium Nucleus of Neuroepigenetics and Plasticity (EpiNeuro), Santiago, Chile.
| |
Collapse
|
3
|
Mares-Barbosa TB, Cuellar-Santoyo AO, Ruiz-Rodríguez VM, Hernández-Balderas K, González-Hernández O, Portales-Pérez DP, Estrada-Sánchez AM. Repeated administration of a subanesthetic dose of ketamine results in impaired motor and cognitive behavior and differential expression of hippocampal P2X1 and P2X7 receptors in adult mice. Behav Brain Res 2025; 482:115441. [PMID: 39842642 DOI: 10.1016/j.bbr.2025.115441] [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: 06/08/2024] [Revised: 12/31/2024] [Accepted: 01/17/2025] [Indexed: 01/24/2025]
Abstract
Ketamine hydrochloride serves multiple purposes, including its use as a general anesthetic, treatment for depression, and recreational drug. In studies involving rodents, ketamine is utilized as a model for schizophrenia. However, it is unclear whether age affects the behavioral response induced by repeated ketamine administration and if it modifies the expression levels of N-methyl-D-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and purinergic receptors (P2X1, P2X4, P2X7). In the present study, we evaluated the effect of intraperitoneal administration of subanesthetic ketamine dose (30 mg/Kg) for fourteen days on young (35 days of age) and adult (76 days of age) mice on different behavioral tests. Nest-building behavior was evaluated during the fourteen-day treatment; short-term memory and social interaction tests were assessed twenty-four hours after the last administration of ketamine. Interestingly, only adult mice treated with ketamine showed impaired nest-building and novel object recognition. In the hippocampus, an area related to memory and cognition, ketamine administration showed no changes in the relative expression of GluN1, P2X4, and P2X7 while increasing GluA2 and P2X1 only in young mice. In contrast, when assessing the protein levels of P2X1 and P2X7 in the hippocampus following ketamine treatment, young mice exhibited a decrease in P2X1 levels while P2X7 levels increased. In contrast, adult mice showed the opposite pattern; P2X1 levels were higher, and P2X7 levels decreased. These results suggest that adult mice are more vulnerable to repeated ketamine administration than young mice and that a differential response of P2X1 and P2X7 might contribute to ketamine-induced behavioral changes.
Collapse
Affiliation(s)
- Teresa Belem Mares-Barbosa
- Laboratorio de Neurobiología, División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico; Laboratorio de Medicina Molecular y Traslacional, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Ares Orlando Cuellar-Santoyo
- Laboratorio de Neurobiología, División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| | - Victor Manuel Ruiz-Rodríguez
- Laboratorio de Neurobiología, División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| | - Karen Hernández-Balderas
- Laboratorio de Neurobiología, División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico
| | - Osiel González-Hernández
- Laboratorio de Medicina Molecular y Traslacional, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Diana Patricia Portales-Pérez
- Laboratorio de Medicina Molecular y Traslacional, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Ana María Estrada-Sánchez
- Laboratorio de Neurobiología, División de Biología Molecular, Instituto Potosino de Investigación Científica y Tecnológica (IPICYT), San Luis Potosí, Mexico.
| |
Collapse
|
4
|
Castro-Zaballa S, González J, Cavelli M, Mateos D, Pascovich C, Tort A, Hunt MJ, Torterolo P. Cortical high-frequency oscillations (≈ 110 Hz) in cats are state-dependent and enhanced by a subanesthetic dose of ketamine. Behav Brain Res 2025; 476:115231. [PMID: 39218075 DOI: 10.1016/j.bbr.2024.115231] [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: 06/11/2024] [Revised: 08/01/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Ketamine is an NMDA receptor antagonist that has antidepressant and anesthetic properties. At subanesthetic doses, ketamine induces transient psychosis in humans, and is used to model psychosis in experimental animals. In rodents, subanesthetic doses of ketamine increase the power of high-frequency oscillations (HFO, > 100 Hz) in the electroencephalogram (EEG), a frequency band linked to cognitive functions. However, to date, the effects of ketamine in carnivores and primates have been poorly investigated. Here, we examined in the cat, cortical HFO during wakefulness, sleep, and after administering a sub-anesthetic dose of ketamine. Four cats were prepared with cortical electrodes for chronic polysomnographic recordings in head-restrained conditions. The cortical HFO power, connectivity, direction of the information flow using Granger Causality (GC) analysis, their relationships with respiratory activity, and the effect of auditory stimulation were analyzed. During wakefulness, but not during sleep, we found that HFO were coupled with the inspiratory phase of the respiration. After ketamine administration, HFO power was enhanced and remained associated with the inspiratory phase. GC analysis suggests that ketamine-enhanced HFO originate from the olfactory bulb (OB) and stream towards the prefrontal cortex (Pf). Accordingly, occluding the nostrils significantly reduced the power of the ketamine-enhanced HFO in both the OB and Pf. Finally, auditory stimulation did not affect HFO. In conclusion, the HFO are associated with respiration during wakefulness, but not during sleep. The enhancement of this rhythm by ketamine may disrupt cortical information processing, which could contribute to some of the neuropsychiatric effects associated with ketamine.
Collapse
Affiliation(s)
- Santiago Castro-Zaballa
- Laboratorio de Neurobiología del Sueño, Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| | - Joaquín González
- Laboratorio de Neurobiología del Sueño, Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay; Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Matías Cavelli
- Laboratorio de Neurobiología del Sueño, Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay; Department of Psychiatry, University of Wisconsin, Madison, United States
| | - Diego Mateos
- Consejo Nacional Investigaciones Científicas y Técnicas (CONICET), Argentina; Universidad Autónoma de Entre Ríos (FCyT-UADER), Entre Ríos, Argentina; Instituto de Matemática Aplicada del Litoral (IMAL-CONICET-UNL), Santa Fe, Argentina; Achucarro Basque Centre for Neuroscience, Spain
| | - Claudia Pascovich
- Laboratorio de Neurobiología del Sueño, Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay; Consciousness and Cognition Laboratory, Department of Psychology, University of Cambridge, United Kingdom
| | - Adriano Tort
- Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil
| | | | - Pablo Torterolo
- Laboratorio de Neurobiología del Sueño, Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
5
|
Shangase KB, Luvuno M, Mabandla M. Effects of combined postweaning social isolation and ketamine administration on schizophrenia-like behaviour in male Sprague Dawley rats. Behav Brain Res 2025; 476:115214. [PMID: 39182622 DOI: 10.1016/j.bbr.2024.115214] [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: 04/10/2024] [Revised: 08/08/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
The pathophysiology behind negative and cognitive symptoms of schizophrenia is not well understood, thus limiting the effectiveness of treatment on these symptoms. Developing reliable animal model of schizophrenia is vital to advance our understanding on the neurobiological basis of the disorder. Double hit is used to refer to the use of two schizophrenia inducing interventions viz ketamine exposure and social isolation. In this study we aim to investigate the robustness of double hit model of schizophrenia in inducing negative and cognitive symptoms of schizophrenia. On postnatal day (PND) 23, thirty-two male Sprague Dawley rats were randomly grouped into four equal groups as follows: group housed + saline (GH), group housed + ketamine (GHK), isolated + saline (SI), and isolated + ketamine (SIK). A single ketamine dose (16 mg/kg) was administered 3 times a week for four weeks. Isolated animals were housed singly throughout the study. The following behavioural tests were carried out: elevated plus maze, three chamber social interaction, resident intruder tests, and novel object recognition (NOR). The SIK group exhibited high anxiety levels, with increased ACTH, corticosterone and norepinephrine concentration when compared to the other groups. The SIK animals also presented with reduced social interaction and decreased oxytocin concentration. SIK rats were more aggressive towards a juvenile intruder but had low testosterone concentration. The SIK group or double hit model showed impaired visual learning and memory and increased expression of proinflammatory cytokines. This suggest that the double hit model is more robust in inducing negative and cognitive symptoms of schizophrenia than each treatment alone.
Collapse
Affiliation(s)
- Khanyiso Bright Shangase
- Department of Human Physiology, School of Laboratory Medicine and Medical Science, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa.
| | - Mluleki Luvuno
- Department of Human Physiology, School of Laboratory Medicine and Medical Science, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| | - Musa Mabandla
- Department of Human Physiology, School of Laboratory Medicine and Medical Science, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| |
Collapse
|
6
|
Lucafò M, Bidoli C, Franzin M, Eitan E, Rau S, Amaddeo A, Fachin A, d'Adamo AP, Decorti G, Stocco G, Barbi E, Cozzi G. Neuron-Derived Extracellular Vesicles miRNA Profiles Identify Children Who Experience Adverse Events after Ketamine Administration for Procedural Sedation. Clin Pharmacol Ther 2025; 117:174-183. [PMID: 39164873 PMCID: PMC11652811 DOI: 10.1002/cpt.3420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 07/30/2024] [Indexed: 08/22/2024]
Abstract
Ketamine provides the highest safety profile among sedatives for procedural sedation and analgesia in the pediatric emergency setting. However, it can cause vomiting and recovery agitation. No studies have examined epigenetic factors, such as microRNAs, for predicting the occurrence of these adverse events. Neuronal-derived extracellular vesicle microRNA profiles were studied to predict the occurrence of ketamine-induced vomiting and recovery agitation in children. For this aim, a single-center prospective pharmacoepigenetic study was performed and 50 children who underwent procedural sedation with intravenous ketamine as the only sedative drug were enrolled between October 2019 and November 2022. MiRNA profiling in plasma neural-derived extracellular vesicles was analyzed through next-generation sequencing and measured before treatment with ketamine. Twenty-two patients experienced vomiting or recovery agitation. Among the 16 differentially expressed microRNAs, the upregulated miR-15a-5p and miR-484 targeted genes related to N-methyl-D-aspartate (NMDA) receptor activity, including glutamate ionotropic receptor NMDA type subunit 2A (GRIN2A). Preliminary data confirmed lower GRIN2A levels in patients who developed these events. Downregulated miR-126-3p and miR-24-3p targeted AMPA receptor-associated genes. Functional analyses of gene targets revealed the enrichment of glutamatergic and neurotrophins signaling. Recovery agitation was associated with this network. Vomiting was related to dopaminergic and cholinergic systems. Three miRNAs (miR-18a-3p, miR-484, and miR-548az-5p) were identified as predictive biomarkers (AUC 0.814; 95% CI: 0.632-0.956) for ketamine-induced vomiting and recovery agitation. MicroRNA profiles can predict the development of ketamine-induced vomiting or recovery agitation in children. This study contributes to the understanding of the mechanisms underlying ketamine-induced adverse events.
Collapse
Affiliation(s)
- Marianna Lucafò
- Department of Life SciencesUniversity of TriesteTriesteItaly
| | - Carlotta Bidoli
- Department of Life SciencesUniversity of TriesteTriesteItaly
| | - Martina Franzin
- Department of Advanced Translational DiagnosticsInstitute for Maternal and Child Health IRCCS “Burlo Garofolo”TriesteItaly
| | | | - Sara Rau
- NeuroDex Inc.NatickMassachusettsUSA
| | - Alessandro Amaddeo
- Emergency DepartmentInstitute for Maternal and Child Health‐IRCCS Burlo GarofoloTriesteItaly
| | - Alice Fachin
- Department of Medicine Surgery and Health SciencesUniversity of TriesteTriesteItaly
| | - Adamo Pio d'Adamo
- Department of Medicine Surgery and Health SciencesUniversity of TriesteTriesteItaly
- Laboratory of Medical GeneticsInstitute for Maternal and Child Health ‐ IRCCS “Burlo Garofolo”TriesteItaly
| | - Giuliana Decorti
- Department of Medicine Surgery and Health SciencesUniversity of TriesteTriesteItaly
| | - Gabriele Stocco
- Department of Advanced Translational DiagnosticsInstitute for Maternal and Child Health IRCCS “Burlo Garofolo”TriesteItaly
- Department of Medicine Surgery and Health SciencesUniversity of TriesteTriesteItaly
| | - Egidio Barbi
- Department of Medicine Surgery and Health SciencesUniversity of TriesteTriesteItaly
- Department of PediatricsInstitute for Maternal and Child Health IRCCS “Burlo Garofolo”TriesteItaly
| | - Giorgio Cozzi
- Emergency DepartmentInstitute for Maternal and Child Health‐IRCCS Burlo GarofoloTriesteItaly
| |
Collapse
|
7
|
Mouffok I, Lahogue C, Cailly T, Freret T, Bouet V, Boulouard M. A New Three-Hit Mouse Model of Neurodevelopmental Disorder with Cognitive Impairments and Persistent Sociability Deficits. Brain Sci 2024; 14:1281. [PMID: 39766480 PMCID: PMC11674404 DOI: 10.3390/brainsci14121281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Cognitive deficits and negative symptoms associated with schizophrenia are poorly managed by current antipsychotics. In order to develop effective treatments, refining animal models of neurodevelopmental disorders is essential. METHODS To address their multifactorial etiology, we developed a new three-hit mouse model based on the hypoglutamatergic hypothesis of the pathology combined with early stress, offering strong construct validity. Thus, a genetic susceptibility (serine racemase deletion) was associated with an early environmental stress (24 h maternal separation at 9 days of age) and a further pharmacological treatment with phencyclidine (PCP, a glutamate receptor antagonist treatment, 10 mg/kg/day, from 8 to 10 weeks of age). The face validity of this model was assessed in female mice 1 and 6 weeks after the end of PCP treatment by a set of behavioral experiments investigating positive- and negative-like symptoms and cognitive deficits. RESULTS Our results showed that the three-hit mice displayed persistent hyperlocomotion (positive-like symptoms) and social behavior impairment deficits (negative-like symptoms) but non-persistent spatial working memory deficits (cognitive symptoms). CONCLUSIONS Our work confirms the usefulness of a three-hit combination to model, particularly for negative-like symptoms associated with schizophrenia and other psychiatric disorders. The model therefore gathers powerful construct and face validities and supports an involvement of glutamate dysfunction in behavioral symptoms.
Collapse
Affiliation(s)
- Imane Mouffok
- Department of Health, Normandie Université, UNICAEN (Université de Caen Normandie), INSERM (Institut National de la Santé et de la Recherche Médicale), UMR (Unité Mixte de Recherche) 1075 COMETE, Campus 5, CYCERON, FHU (Fédération Hospitalo-Universitaire) A2M2P, CHU (Centre Hospitalo-Universitaire) Caen, 14000 Caen, France; (I.M.); (C.L.); (T.F.); (M.B.)
| | - Caroline Lahogue
- Department of Health, Normandie Université, UNICAEN (Université de Caen Normandie), INSERM (Institut National de la Santé et de la Recherche Médicale), UMR (Unité Mixte de Recherche) 1075 COMETE, Campus 5, CYCERON, FHU (Fédération Hospitalo-Universitaire) A2M2P, CHU (Centre Hospitalo-Universitaire) Caen, 14000 Caen, France; (I.M.); (C.L.); (T.F.); (M.B.)
| | - Thomas Cailly
- CERMN UR (Unité de Recherche) 4258, Campus 5, Université de Caen Normandie, 14000 Caen, France;
- CYCERON UAR (Unité d’Appui à la Recherche) 3408-US50, IMOGERE, Campus 1, Université de Caen Normandie, 14000 Caen, France
- Department of Nuclear Medicine, CHU Côte de Nacre, 14000 Caen, France
| | - Thomas Freret
- Department of Health, Normandie Université, UNICAEN (Université de Caen Normandie), INSERM (Institut National de la Santé et de la Recherche Médicale), UMR (Unité Mixte de Recherche) 1075 COMETE, Campus 5, CYCERON, FHU (Fédération Hospitalo-Universitaire) A2M2P, CHU (Centre Hospitalo-Universitaire) Caen, 14000 Caen, France; (I.M.); (C.L.); (T.F.); (M.B.)
| | - Valentine Bouet
- Department of Health, Normandie Université, UNICAEN (Université de Caen Normandie), INSERM (Institut National de la Santé et de la Recherche Médicale), UMR (Unité Mixte de Recherche) 1075 COMETE, Campus 5, CYCERON, FHU (Fédération Hospitalo-Universitaire) A2M2P, CHU (Centre Hospitalo-Universitaire) Caen, 14000 Caen, France; (I.M.); (C.L.); (T.F.); (M.B.)
| | - Michel Boulouard
- Department of Health, Normandie Université, UNICAEN (Université de Caen Normandie), INSERM (Institut National de la Santé et de la Recherche Médicale), UMR (Unité Mixte de Recherche) 1075 COMETE, Campus 5, CYCERON, FHU (Fédération Hospitalo-Universitaire) A2M2P, CHU (Centre Hospitalo-Universitaire) Caen, 14000 Caen, France; (I.M.); (C.L.); (T.F.); (M.B.)
| |
Collapse
|
8
|
Xue L, Zhao J, Liu X, Zhao T, Zhang Y, Ye H. MK-801-exposure induces increased translation efficiency and mRNA hyperacetylation of Grin2a in the mouse prefrontal cortex. Epigenetics 2024; 19:2417158. [PMID: 39460980 PMCID: PMC11520555 DOI: 10.1080/15592294.2024.2417158] [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: 03/20/2024] [Revised: 09/19/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Acute exposure to MK-801, the non-competitive N-methyl-D-aspartate (NMDA) receptor antagonist, induces schizophrenia-like behavioural changes in juvenile male mice. However, the effects of acute MK-801 exposure on brain gene expression at the translation level remain unclear. Here, we conducted ribosome profiling analysis on the prefrontal cortex (PFC) of acute MK-801-exposed juvenile male mice. We found 357 differentially translated genes, with the N4-acetylcytidine (ac4C) consensus motif enriched in the transcripts with increased translation efficiency. Acetylated RNA immunoprecipitation sequencing revealed 148 differentially acetylated peaks, of which 121 were hyperacetylated, and 27 were hypoacetylated. Genes harbouring these peaks were enriched in pathways related to axon guidance, Hedgehog signalling pathway, neuron differentiation, and memory. Grin2a encodes an NMDA receptor subunit NMDAR2A, and its human orthologue is a strong susceptibility gene for schizophrenia. Grin2a mRNA was hyperacetylated and exhibited significantly increased translation efficiency. NMDAR2A protein level was increased in MK-801-exposed PFC. Pretreatment of Remodelin, an inhibitor of N-acetyltransferase 10, returned the NMDAR2A protein levels to normal and partially reversed schizophrenia-like behaviours of MK-801-exposed mice, shedding light on the possible role of mRNA acetylation in the aetiology of schizophrenia.
Collapse
Affiliation(s)
- Liting Xue
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jialu Zhao
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xu Liu
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Tian Zhao
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ying Zhang
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Haihong Ye
- Department of Medical Genetics and Developmental Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
9
|
Acero-Castillo MC, Correia MBM, Caixeta FV, Motta V, Barros M, Maior RS. Is the antidepressant effect of ketamine separate from its psychotomimetic effect? A review of rodent models. Neuropharmacology 2024; 258:110088. [PMID: 39032814 DOI: 10.1016/j.neuropharm.2024.110088] [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: 04/16/2024] [Revised: 07/09/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Ketamine is an NMDA (N-methyl-d-aspartate) glutamate receptor antagonist, which has a myriad of dose-dependent pharmacological and behavioral effects, including anesthetic, sedative, amnestic, analgesic, and anti-inflammatory properties. Intriguingly, ketamine at subanesthetic doses displays a relevant profile both in mimicking symptoms of schizophrenia and also as the first fast-acting treatment for depression. Here, we present an overview of the state-of-the-art knowledge about ketamine as an antidepressant as well as a pharmacological model of schizophrenia in animal models and human participants. Ketamine's dual effect appears to arise from its mechanism of action involving NMDA receptors, with both immediate and downstream consequences being triggered as a result. Finally, we discuss the feasibility of a unified approach linking the glutamatergic hypothesis of schizophrenia to the promising preclinical and clinical success of ketamine in the treatment of refractory depression.
Collapse
Affiliation(s)
- M C Acero-Castillo
- Laboratory of Neuroscience, Metabolism, and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil
| | - M B M Correia
- Laboratory of Neuroscience, Metabolism, and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil; Department of Anthropology, Emory University, Atlanta GA, ZIP 30322, USA
| | - F V Caixeta
- Laboratory of Neuroscience, Metabolism, and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil
| | - V Motta
- Department of Basic Psychological Processes, Institute of Psychology, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil
| | - M Barros
- Department of Pharmacy, School of Health Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil
| | - R S Maior
- Laboratory of Neuroscience, Metabolism, and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil.
| |
Collapse
|
10
|
Remali J, Aizat WM. Medicinal plants and plant-based traditional medicine: Alternative treatments for depression and their potential mechanisms of action. Heliyon 2024; 10:e38986. [PMID: 39640650 PMCID: PMC11620067 DOI: 10.1016/j.heliyon.2024.e38986] [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: 09/29/2024] [Accepted: 10/03/2024] [Indexed: 12/07/2024] Open
Abstract
Background Clinical depression is a serious public health issue that affects 4.7 % of the world's population and can lead to suicide tendencies. Although drug medications are available, only 60 % of the depressed patients respond positively to the treatments, while the rest experience side effects that resulted in the discontinuation of their medication. Thus, there is an urgent need for developing a new anti-depressant with a distinct mode of action and manageable side effects. One of the options is using medicinal plants or plant-based traditional medicine as alternative therapies for psychiatric disorders. Objectives Therefore, the objective of this review was twofold; to identify and critically evaluate anti-depressant properties of medicinal plants or those incorporated in traditional medicine; and to discuss their possible mechanism of action as well as challenges and way forward for this alternative treatment approach. Methods Relevant research articles were retrieved from various databases, including Scopus, PubMed, and Web of Science, for the period from 2018 to 2020, and the search was updated in September 2024. The inclusion criterion was relevance to antidepressants, while the exclusion criteria included duplicates, lack of full-text availability, and non-English publications. Results Through an extensive literature review, more than 40 medicinal plant species with antidepressant effects were identified, some of which are part of traditional medicine. The list of the said plant species included Albizia zygia (DC.) J.F.Macbr., Calculus bovis Sativus, Celastrus paniculatus Willd., Cinnamomum sp., Erythrina velutina Willd., Ficus platyphylla Delile, Garcinia mangostana Linn., Hyptis martiusii Benth, and Polygonum multiflorum Thunb. Anti-depressant mechanisms associated with those plants were further characterised based on their modes of action such as anti-oxidation system, anti-inflammation action, modulation of various neurotransmitters, neuroprotective effect, the regulation of hypothalamic-pituitary-adrenal (HPA) axis and anti-depressant mechanism. The challenges and future outlook of this alternative and complementary medicine are also explored and discussed. Conclusion This pool of identified plant species is hoped to offer health care professionals the best possible alternatives of anti-depressants from natural phytocompounds that are efficacious, safe and affordable for applications in future clinical settings.
Collapse
Affiliation(s)
- Juwairiah Remali
- Department of Pathology, Hospital Pulau Pinang, Jalan Residensi, 10450, George Town, Pulau Pinang, Malaysia
| | - Wan Mohd Aizat
- Institute of Systems Biology (INBIOSIS), Universiti Kebangsaan Malaysia (UKM), 43600, Bangi, Selangor, Malaysia
| |
Collapse
|
11
|
Piazza MK, Kavalali ET, Monteggia LM. Ketamine induced synaptic plasticity operates independently of long-term potentiation. Neuropsychopharmacology 2024; 49:1758-1766. [PMID: 38898206 PMCID: PMC11399243 DOI: 10.1038/s41386-024-01895-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/11/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024]
Abstract
Synaptic plasticity occurs via multiple mechanisms to regulate synaptic efficacy. Homeostatic and Hebbian plasticity are two such mechanisms by which neuronal synapses can be altered. Although these two processes are mechanistically distinct, they converge on downstream regulation of AMPA receptor activity to modify glutamatergic neurotransmission. However, much remains to be explored regarding how these two prominent forms of plasticity interact. Ketamine, a rapidly acting antidepressant, increases glutamatergic transmission via pharmacologically-induced homeostatic plasticity. Here, we demonstrate that Hebbian plasticity mechanisms are still intact in synapses that have undergone homeostatic scaling by ketamine after either systemic injection or perfusion onto hippocampal brain slices. We also investigated this relationship in the context of stress induced by chronic exposure to corticosterone (CORT) to better model the circumstances under which ketamine may be used as an antidepressant. We found that CORT induced an anhedonia-like behavioral phenotype in mice but did not impair long-term potentiation (LTP) induction. Furthermore, corticosterone exposure does not impact the intersection of homeostatic and Hebbian plasticity mechanisms, as synapses from CORT-exposed mice also demonstrated intact ketamine-induced plasticity and LTP in succession. These results provide a mechanistic explanation for how ketamine used for the treatment of depression does not impair the integrity of learning and memory processes encoded by mechanisms such as LTP.
Collapse
Affiliation(s)
- Michelle K Piazza
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37240-7933, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37240-7933, USA
| | - Ege T Kavalali
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37240-7933, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37240-7933, USA
| | - Lisa M Monteggia
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37240-7933, USA.
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37240-7933, USA.
| |
Collapse
|
12
|
Zhong J, Wu F, Wu H, He H, Zhang Z, Fan N. Abnormal resting-state functional connectivity of the right anterior cingulate cortex in chronic ketamine users and its correlation with cognitive impairments. Asian J Psychiatr 2024; 102:104199. [PMID: 39298913 DOI: 10.1016/j.ajp.2024.104199] [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: 11/24/2023] [Revised: 08/16/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Chronic ketamine use leads to cognitive impairments, however, the neural mechanisms underpinning these impairments are still unclear. AIMS Many studies showed Anterior cingulate cortex (ACC)is strongly involved in cognition and drug addiction, as supported by our previous studies. The objective of this study was to assess the variations in resting-state functional connectivity (FC) changes in the right anterior cingulate cortex (ACC) of chronic ketamine users (CKUs) and their relationship with cognitive performance. METHODS The study enrolled 28 chronic ketamine users (CKUs) and 30 healthy controls (HCs). Resting-state functional magnetic resonance imaging (fMRI) data were gathered from both groups. Cognitive functions were evaluated using the MATRICS Consensus Cognitive Battery (MCCB). RESULTS CKUs demonstrated significantly poorer cognitive performance than HCs in various cognitive domains, including Visual Learning, Speed of Processing, Working Memory, and the composite score of MCCB. Group-level comparisons revealed that CKUs exhibited enhanced functional connectivity between the right ACC and the right postcentral gyrus (PCG) compared to HCs. There was a positive relationship between the connectivity of right ACC-PCG and reasoning and problem-solving score, but there was no significant association with the characteristics of ketamine use. CONCLUSION CKUs showed enhanced connectivity between the right ACC and the right PCG. This enhanced functional connectivity may indicate functional compensation for cognitive deficits in CKUs, especially for reasoning and problem-solving impairments in CKUs.
Collapse
Affiliation(s)
- Jun Zhong
- The Affiliated Brain Hospital, Guangzhou Medical University, 36 Mingxin Road, Liwan District, Guangzhou, Guangdong 510370, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, China
| | - Fengchun Wu
- The Affiliated Brain Hospital, Guangzhou Medical University, 36 Mingxin Road, Liwan District, Guangzhou, Guangdong 510370, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, China
| | - Huawang Wu
- The Affiliated Brain Hospital, Guangzhou Medical University, 36 Mingxin Road, Liwan District, Guangzhou, Guangdong 510370, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, China
| | - Hongbo He
- The Affiliated Brain Hospital, Guangzhou Medical University, 36 Mingxin Road, Liwan District, Guangzhou, Guangdong 510370, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, China
| | - Zhaohua Zhang
- The Affiliated Brain Hospital, Guangzhou Medical University, 36 Mingxin Road, Liwan District, Guangzhou, Guangdong 510370, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, China
| | - Ni Fan
- The Affiliated Brain Hospital, Guangzhou Medical University, 36 Mingxin Road, Liwan District, Guangzhou, Guangdong 510370, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, China.
| |
Collapse
|
13
|
Fujikawa R, Yamada J, Maeda S, Iinuma KM, Moriyama G, Jinno S. Inhibition of reactive oxygen species production accompanying alternatively activated microglia by risperidone in a mouse ketamine model of schizophrenia. J Neurochem 2024; 168:2690-2709. [PMID: 38770640 DOI: 10.1111/jnc.16133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 04/17/2024] [Accepted: 05/06/2024] [Indexed: 05/22/2024]
Abstract
Recent studies have highlighted the potential involvement of reactive oxygen species (ROS) and microglia, a major source of ROS, in the pathophysiology of schizophrenia. In our study, we explored how the second-generation antipsychotic risperidone (RIS) affects ROS regulation and microglial activation in the hippocampus using a mouse ketamine (KET) model of schizophrenia. KET administration resulted in schizophrenia-like behaviors in male C57BL/6J mice, such as impaired prepulse inhibition (PPI) of the acoustic startle response and hyper-locomotion. These behaviors were mitigated by RIS. We found that the gene expression level of an enzyme responsible for ROS production (Nox2), which is primarily associated with activated microglia, was lower in KET/RIS-treated mice than in KET-treated mice. Conversely, the levels of antioxidant enzymes (Ho-1 and Gclc) were higher in KET/RIS-treated mice. The microglial density in the hippocampus was increased in KET-treated mice, which was counteracted by RIS. Hierarchical cluster analysis revealed three morphological subtypes of microglia. In control mice, most microglia were resting-ramified (type I, 89.7%). KET administration shifted the microglial composition to moderately ramified (type II, 44.4%) and hyper-ramified (type III, 25.0%). In KET/RIS-treated mice, type II decreased to 32.0%, while type III increased to 34.0%. An in vitro ROS assay showed that KET increased ROS production in dissociated hippocampal microglia, and this effect was mitigated by RIS. Furthermore, we discovered that a NOX2 inhibitor could counteract KET-induced behavioral deficits. These findings suggest that pharmacological inhibition of ROS production by RIS may play a crucial role in ameliorating schizophrenia-related symptoms. Moreover, modulating microglial activation to regulate ROS production has emerged as a novel avenue for developing innovative treatments for schizophrenia.
Collapse
Affiliation(s)
- Risako Fujikawa
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jun Yamada
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shoichiro Maeda
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kyoko M Iinuma
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | - Shozo Jinno
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
14
|
Ponserre M, Ionescu TM, Franz AA, Deiana S, Schuelert N, Lamla T, Williams RH, Wotjak CT, Hobson S, Dine J, Omrani A. Long-term adaptation of prefrontal circuits in a mouse model of NMDAR hypofunction. Neuropharmacology 2024; 254:109970. [PMID: 38685343 DOI: 10.1016/j.neuropharm.2024.109970] [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: 02/15/2024] [Revised: 04/12/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024]
Abstract
Pharmacological approaches to induce N-methyl-d-aspartate receptor (NMDAR) hypofunction have been intensively used to understand the aetiology and pathophysiology of schizophrenia. Yet, the precise cellular and molecular mechanisms that relate to brain network dysfunction remain largely unknown. Here, we used a set of complementary approaches to assess the functional network abnormalities present in male mice that underwent a 7-day subchronic phencyclidine (PCP 10 mg/kg, subcutaneously, once daily) treatment. Our data revealed that pharmacological intervention with PCP affected cognitive performance and auditory evoked gamma oscillations in the prefrontal cortex (PFC) mimicking endophenotypes of some schizophrenia patients. We further assessed PFC cellular function and identified altered neuronal intrinsic membrane properties, reduced parvalbumin (PV) immunostaining and diminished inhibition onto L5 PFC pyramidal cells. A decrease in the strength of optogenetically-evoked glutamatergic current at the ventral hippocampus to PFC synapse was also demonstrated, along with a weaker shunt of excitatory transmission by local PFC interneurons. On a macrocircuit level, functional ultrasound measurements indicated compromised functional connectivity within several brain regions particularly involving PFC and frontostriatal circuits. Herein, we reproduced a panel of schizophrenia endophenotypes induced by subchronic PCP application in mice. We further recapitulated electrophysiological signatures associated with schizophrenia and provided an anatomical reference to critical elements in the brain circuitry. Together, our findings contribute to a better understanding of the physiological underpinnings of deficits induced by subchronic NMDAR antagonist regimes and provide a test system for characterization of pharmacological compounds.
Collapse
Affiliation(s)
- Marion Ponserre
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Tudor M Ionescu
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Alessa A Franz
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Serena Deiana
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Niklas Schuelert
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Thorsten Lamla
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | | | - Carsten T Wotjak
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Scott Hobson
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Julien Dine
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Azar Omrani
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany.
| |
Collapse
|
15
|
Juliani PZ, Rodrigues T, Bressan GN, Camponogara C, Oliveira SM, Brucker N, Fachinetto R. Effects of association between resveratrol and ketamine on behavioral and biochemical analysis in mice. J Neural Transm (Vienna) 2024; 131:971-986. [PMID: 38874765 DOI: 10.1007/s00702-024-02793-z] [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: 11/13/2023] [Accepted: 06/01/2024] [Indexed: 06/15/2024]
Abstract
Resveratrol (3,5,4'-trihydroxy-trans-stilbene), a phenol commonly found in grapes and wine, has been associated as protective in experimental models involving alterations in different neurotransmitter systems. However, studies are reporting that resveratrol could have adverse effects. This study evaluated if the association of a low dose of ketamine and resveratrol could induce behavioral manifestations associated with biochemical alterations. Moreover, the effects of treatment with resveratrol and/or ketamine on monoamine oxidase (MAO) activity, oxidative stress markers, and IL-6 levels in the brain were also investigated. Male Swiss mice received a low dose of ketamine (20 mg/kg) for 14 consecutive days, and resveratrol (10, 30, or 100 mg/kg) from day 8 up to day 14 of the experimental period, intraperitoneally. Locomotor, stereotyped behavior, Y-maze, novel recognition object test (NORT), and social interaction were quantified as well as ex vivo analysis of MAO activity, IL-6 levels, and oxidative stress markers (TBARS and total thiol levels) in brain tissues. Ketamine per se reduced the number of bouts of stereotyped behavior on day 8 of the experimental period. Resveratrol per se reduced the locomotor and exploratory activity in the open field, the time of exploration of new objects in the NORT, MAO-A activity in the striatum and increased the IL-6 levels in the cortex. These effects were attenuated when the mice were co-treated with ketamine and resveratrol. There was a decrease in MAO-A activity in the cortex of mice treated with ketamine + resveratrol 100 mg/kg. No significant alterations were found in oxidative stress markers. Resveratrol does not appear to cause summative effects with ketamine on behavioral alterations. However, the effect of resveratrol per se, mainly on locomotor and exploratory activity, should be better investigated.
Collapse
Affiliation(s)
- Patrícia Zorzi Juliani
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Talita Rodrigues
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Getulio Nicola Bressan
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Camila Camponogara
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Sara Marchesan Oliveira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Natália Brucker
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Roselei Fachinetto
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil.
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil.
- Centro de Ciências da Saúde, Departamento de Fisiologia e Farmacologia, Santa Maria, RS, 97105-900, Brazil.
| |
Collapse
|
16
|
Johnston JN, Zarate CA, Kvarta MD. Esketamine in depression: putative biomarkers from clinical research. Eur Arch Psychiatry Clin Neurosci 2024:10.1007/s00406-024-01865-1. [PMID: 38997425 PMCID: PMC11725628 DOI: 10.1007/s00406-024-01865-1] [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: 06/04/2024] [Accepted: 07/03/2024] [Indexed: 07/14/2024]
Abstract
The discovery of racemic (R, S)-ketamine as a rapid-acting antidepressant and the subsequent FDA approval of its (S)-enantiomer, esketamine, for treatment-resistant depression (TRD) are significant advances in the development of novel neuropsychiatric therapeutics. Esketamine is now recognized as a powerful tool for addressing persistent symptoms of TRD compared to traditional oral antidepressants. However, research on biomarkers associated with antidepressant response to esketamine has remained sparse and, to date, has been largely extrapolated from racemic ketamine studies. Genetic, proteomic, and metabolomic profiles suggest that inflammation and mitochondrial function may play a role in esketamine's antidepressant effects, though these preliminary results require verification. In addition, neuroimaging research has consistently implicated the prefrontal cortex, striatum, and anterior cingulate cortex in esketamine's effects. Esketamine also shows promise in perioperative settings for reducing depression and anxiety, and these effects appear to correlate with increased peripheral biomarkers such as brain-derived neurotrophic factor and serotonin. Further indications are likely to be identified with the continued repurposing of racemic ketamine, providing further opportunity for biomarker study and mechanistic understanding of therapeutic effects. Novel methodologies and well-designed biomarker-focused clinical research trials are needed to more clearly elucidate esketamine's therapeutic actions as well as biologically identify those most likely to benefit from this agent, allowing for the improved personalization of antidepressant treatment.
Collapse
Affiliation(s)
- Jenessa N Johnston
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Mark D Kvarta
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA.
| |
Collapse
|
17
|
Dahan A, Jansen S, van der Schrier R, Sarton E, Dadiomov D, van Velzen M, Olofsen E, Niesters M. Nitric Oxide Donor Sodium Nitroprusside Reduces Racemic Ketamine-But Not Esketamine-Induced Pain Relief. ACS Pharmacol Transl Sci 2024; 7:2044-2053. [PMID: 39022368 PMCID: PMC11249631 DOI: 10.1021/acsptsci.4c00133] [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: 03/12/2024] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 07/20/2024]
Abstract
The anesthetic, analgesic and antidepressant drug ketamine produces dissociation with symptoms of psychosis and anxiety, an effect attributed to neuronal nitric oxide depletion following N-methyl-d-aspartate blockade. There is evidence that dissociation induced by racemic ketamine, containing both ketamine enantiomers (S- and R-ketamine) but not esketamine (the S-isomer) is inhibited by nitric oxide (NO) donor sodium nitroprusside (SNP). We tested whether a similar intervention would reduce racemic and esketamine-induced analgesia in a randomized double-blind placebo-controlled trial. Seventeen healthy volunteers were treated with 0.5 μg.kg-1.min-1 SNP or placebo during a 3-h infusion of escalating doses of racemic ketamine (total dose 140 mg) or esketamine (70 mg). Pain pressure threshold (PPT) and arterial blood samples for measurement of S- and R-ketamine and their metabolites, S- and R-norketamine, were obtained. The data were analyzed with a population pharmacokinetic-pharmacodynamic model that incorporated the measured S- and R- ketamine and S- and R-norketamine isomers as input and PPT as output to the model. The potency of the 2 formulations in increasing PPT from baseline by 100% was 0.47 ± 0.12 (median ± standard error of the estimate) nmol/mL for esketamine and 0.62 ± 0.19 nmol/mL for racemic ketamine, reflecting the 52 ± 27% lower analgesic potency of R-ketamine versus S-ketamine. Modeling showed that SNP had no effect on S-ketamine potency but abolished the R-ketamine analgesic effect. Similar observations were made for S- and R-norketamine. Since SNP had no effect on S-ketamine analgesia, we conclude that SNP interacts on R-ketamine nociceptive pathways, possibly similar to its effects on R-ketamine activated dissociation pathways.
Collapse
Affiliation(s)
- Albert Dahan
- Department
of Anesthesiology, Leiden University Medical
Center, 2333 ZA Leiden, The Netherlands
- PainLess
Foundation, 2333 ZA Leiden, The Netherlands
- Outcomes
Research Consortium, Cleveland, Ohio 90089, United States
| | - Simone Jansen
- Department
of Anesthesiology, Leiden University Medical
Center, 2333 ZA Leiden, The Netherlands
| | - Rutger van der Schrier
- Department
of Anesthesiology, Leiden University Medical
Center, 2333 ZA Leiden, The Netherlands
| | - Elise Sarton
- Department
of Anesthesiology, Leiden University Medical
Center, 2333 ZA Leiden, The Netherlands
| | - David Dadiomov
- USC
Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, Titus
Family Department of Clinical Pharmacy, University of Southern California, Los Angeles, California 90089, United States
| | - Monique van Velzen
- Department
of Anesthesiology, Leiden University Medical
Center, 2333 ZA Leiden, The Netherlands
| | - Erik Olofsen
- Department
of Anesthesiology, Leiden University Medical
Center, 2333 ZA Leiden, The Netherlands
| | - Marieke Niesters
- Department
of Anesthesiology, Leiden University Medical
Center, 2333 ZA Leiden, The Netherlands
- PainLess
Foundation, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
18
|
Hajizadeh Moghaddam A, Malekzadeh Estalkhi F, Khanjani Jelodar S, Ahmed Hasan T, Farhadi-Pahnedari S, Karimian M. Neuroprotective effects of alpha-pinene against behavioral deficits in ketamine-induced mice model of schizophrenia: Focusing on oxidative stress status. IBRO Neurosci Rep 2024; 16:182-189. [PMID: 38318342 PMCID: PMC10839590 DOI: 10.1016/j.ibneur.2023.12.012] [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: 08/02/2023] [Revised: 12/11/2023] [Accepted: 12/30/2023] [Indexed: 02/07/2024] Open
Abstract
Schizophrenia (SCZ) is a profound neurological disorder that affects approximately 1% of the global population. Alpha-pinene (α-pinene) is a natural and active monoterpene found in coniferous tree oil, primarily pine, with diverse pharmacological characteristics, including antioxidative, anxiolytic, and antidepressant properties. This research study delves into the neuroprotective effects of α-pinene on oxidative stress, memory deficits, and depressive and anxiety-like behaviors in a ketamine-induced mice model of SCZ using male mice. The mice were randomly divided into six groups: vehicle, control, positive control, ketamine, α-pinene at 50 mg/kg, and α-pinene at 100 mg/kg. Treatment of the ketamine-induced mice model of SCZ with α-pinene yielded significant improvements in depressive and anxiety-like behaviors and cognitive impairments. Furthermore, it significantly elevated glutathione (GSH) levels, total antioxidant capacity (TAC), dopamine levels, catalase (CAT), and superoxide dismutase (SOD) activities while markedly reducing malondialdehyde (MDA) levels. The current study establishes that α-pinene treatment effectively mitigates oxidative damage, cognitive deficits, and depressive and anxiogenic-like behaviors in the brains of ketamine-treated mice. Therefore, α-pinene treatment is an efficacious approach to forestall the neurobehavioral and neurobiochemical adverse effects of the ketamine-induced SCZ model of mice.
Collapse
Affiliation(s)
| | | | | | - Tabarek Ahmed Hasan
- Department of Animal Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran
| | | | - Mohammad Karimian
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran
| |
Collapse
|
19
|
Busch A, Roussy M, Luna R, Leavitt ML, Mofrad MH, Gulli RA, Corrigan B, Mináč J, Sachs AJ, Palaniyappan L, Muller L, Martinez-Trujillo JC. Neuronal activation sequences in lateral prefrontal cortex encode visuospatial working memory during virtual navigation. Nat Commun 2024; 15:4471. [PMID: 38796480 PMCID: PMC11127969 DOI: 10.1038/s41467-024-48664-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 05/01/2024] [Indexed: 05/28/2024] Open
Abstract
Working memory (WM) is the ability to maintain and manipulate information 'in mind'. The neural codes underlying WM have been a matter of debate. We simultaneously recorded the activity of hundreds of neurons in the lateral prefrontal cortex of male macaque monkeys during a visuospatial WM task that required navigation in a virtual 3D environment. Here, we demonstrate distinct neuronal activation sequences (NASs) that encode remembered target locations in the virtual environment. This NAS code outperformed the persistent firing code for remembered locations during the virtual reality task, but not during a classical WM task using stationary stimuli and constraining eye movements. Finally, blocking NMDA receptors using low doses of ketamine deteriorated the NAS code and behavioral performance selectively during the WM task. These results reveal the versatility and adaptability of neural codes supporting working memory function in the primate lateral prefrontal cortex.
Collapse
Affiliation(s)
- Alexandra Busch
- Robarts Research Institute, University of Western Ontario, London, ON, Canada
- Brain and Mind Institute, University of Western Ontario, London, ON, Canada
- Department of Mathematics, University of Western Ontario, London, ON, Canada
| | - Megan Roussy
- Robarts Research Institute, University of Western Ontario, London, ON, Canada
- Brain and Mind Institute, University of Western Ontario, London, ON, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | - Rogelio Luna
- Robarts Research Institute, University of Western Ontario, London, ON, Canada
- Brain and Mind Institute, University of Western Ontario, London, ON, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | | | - Maryam H Mofrad
- Brain and Mind Institute, University of Western Ontario, London, ON, Canada
- Department of Mathematics, University of Western Ontario, London, ON, Canada
| | - Roberto A Gulli
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Benjamin Corrigan
- Robarts Research Institute, University of Western Ontario, London, ON, Canada
- Brain and Mind Institute, University of Western Ontario, London, ON, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | - Ján Mináč
- Department of Mathematics, University of Western Ontario, London, ON, Canada
| | - Adam J Sachs
- The Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| | - Lena Palaniyappan
- Robarts Research Institute, University of Western Ontario, London, ON, Canada
- Department of Psychiatry, University of Western Ontario, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
| | - Lyle Muller
- Robarts Research Institute, University of Western Ontario, London, ON, Canada.
- Brain and Mind Institute, University of Western Ontario, London, ON, Canada.
- Department of Mathematics, University of Western Ontario, London, ON, Canada.
| | - Julio C Martinez-Trujillo
- Robarts Research Institute, University of Western Ontario, London, ON, Canada.
- Brain and Mind Institute, University of Western Ontario, London, ON, Canada.
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada.
- Lawson Health Research Institute, London, ON, Canada.
| |
Collapse
|
20
|
Sikiric P, Boban Blagaic A, Strbe S, Beketic Oreskovic L, Oreskovic I, Sikiric S, Staresinic M, Sever M, Kokot A, Jurjevic I, Matek D, Coric L, Krezic I, Tvrdeic A, Luetic K, Batelja Vuletic L, Pavic P, Mestrovic T, Sjekavica I, Skrtic A, Seiwerth S. The Stable Gastric Pentadecapeptide BPC 157 Pleiotropic Beneficial Activity and Its Possible Relations with Neurotransmitter Activity. Pharmaceuticals (Basel) 2024; 17:461. [PMID: 38675421 PMCID: PMC11053547 DOI: 10.3390/ph17040461] [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: 02/15/2024] [Revised: 03/24/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
We highlight the particular aspects of the stable gastric pentadecapeptide BPC 157 pleiotropic beneficial activity (not destroyed in human gastric juice, native and stable in human gastric juice, as a cytoprotection mediator holds a response specifically related to preventing or recovering damage as such) and its possible relations with neurotransmitter activity. We attempt to resolve the shortage of the pleiotropic beneficial effects of BPC 157, given the general standard neurotransmitter criteria, in classic terms. We substitute the lack of direct conclusive evidence (i.e., production within the neuron or present in it as a precursor molecule, released eliciting a response on the receptor on the target cells on neurons and being removed from the site of action once its signaling role is complete). This can be a network of interconnected evidence, previously envisaged in the implementation of the cytoprotection effects, consistent beneficial particular evidence that BPC 157 therapy counteracts dopamine, serotonin, glutamate, GABA, adrenalin/noradrenalin, acetylcholine, and NO-system disturbances. This specifically includes counteraction of those disturbances related to their receptors, both blockade and over-activity, destruction, depletion, tolerance, sensitization, and channel disturbances counteraction. Likewise, BPC 157 activates particular receptors (i.e., VGEF and growth hormone). Furthermore, close BPC 157/NO-system relations with the gasotransmitters crossing the cell membrane and acting directly on molecules inside the cell may envisage particular interactions with receptors on the plasma membrane of their target cells. Finally, there is nerve-muscle relation in various muscle disturbance counteractions, and nerve-nerve relation in various encephalopathies counteraction, which is also exemplified specifically by the BPC 157 therapy application.
Collapse
Affiliation(s)
- Predrag Sikiric
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
| | - Alenka Boban Blagaic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
| | - Sanja Strbe
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
| | - Lidija Beketic Oreskovic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
| | - Ivana Oreskovic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
| | - Suncana Sikiric
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Mario Staresinic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
- Department of Surgery, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Marko Sever
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
- Department of Surgery, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Antonio Kokot
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
- Department of Anatomy and Neuroscience, School of Medicine, J.J. Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Ivana Jurjevic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
| | - Danijel Matek
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
| | - Luka Coric
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
| | - Ivan Krezic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
| | - Ante Tvrdeic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
| | - Kresimir Luetic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
| | - Lovorka Batelja Vuletic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Predrag Pavic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
- Department of Surgery, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Tomislav Mestrovic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
- Department of Anatomy and Neuroscience, School of Medicine, J.J. Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Ivica Sjekavica
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
- Department of Diagnostic and Interventional Radiology, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia
| | - Anita Skrtic
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Sven Seiwerth
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.B.B.); (S.S.); (L.B.O.); (I.O.); (S.S.); (M.S.); (M.S.); (A.K.); (I.J.); (D.M.); (L.C.); (I.K.); (A.T.); (K.L.); (L.B.V.); (P.P.); (T.M.); (I.S.); (S.S.)
- Department of Pathology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
21
|
Zhou L, Duan J. The role of NMDARs in the anesthetic and antidepressant effects of ketamine. CNS Neurosci Ther 2024; 30:e14464. [PMID: 37680076 PMCID: PMC11017467 DOI: 10.1111/cns.14464] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/19/2023] [Accepted: 08/26/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND As a phencyclidine (PCP) analog, ketamine can generate rapid-onset and substantial anesthetic effects. Contrary to traditional anesthetics, ketamine is a dissociative anesthetic and can induce loss of consciousness in patients. Recently, the subanaesthetic dose of ketamine was found to produce rapid-onset and lasting antidepressant effects. AIM However, how different concentrations of ketamine can induce diverse actions remains unclear. Furthermore, the molecular mechanisms underlying the NMDAR-mediated anesthetic and antidepressant effects of ketamine are not fully understood. METHOD In this review, we have introduced ketamine and its metabolism, summarized recent advances in the molecular mechanisms underlying NMDAR inhibition in the anesthetic and antidepressant effects of ketamine, explored the possible functions of NMDAR subunits in the effects of ketamine, and discussed the future directions of ketamine-based anesthetic and antidepressant drugs. RESULT Both the anesthetic and antidepressant effects of ketamine were thought to be mediated by N-methyl-D-aspartate receptor (NMDAR) inhibition. CONCLUSION The roles of NMDARs have been extensively studied in the anaesthetic effects of ketamine. However, the roles of NMDARs in antidepressant effects of ketamine are complicated and controversial.
Collapse
Affiliation(s)
- Liang Zhou
- Department of Pharmacology, College of Pharmaceutical SciencesSoochow UniversitySuzhouChina
| | - Jingjing Duan
- Department of Anatomy and Neurobiology, Zhongshan School of MedicineSunYat‐sen UniversityGuangzhouChina
| |
Collapse
|
22
|
Xiao L, Wei Y, Yang H, Fan W, Jiang L, Ye Y, Qin Y, Wang X, Ma C, Liao L. Proteomic Characteristics of the Prefrontal Cortex and Hippocampus in Mice with Chronic Ketamine-Induced Anxiety and Cognitive Impairment. Neuroscience 2024; 541:23-34. [PMID: 38266908 DOI: 10.1016/j.neuroscience.2023.10.008] [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: 04/30/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 01/26/2024]
Abstract
Schizophrenia, a complex psychiatric disorder with diverse symptoms, has been linked to ketamine, known for its N-methyl-D-aspartate (NMDA) receptor antagonistic properties. Understanding the distinct roles and mechanisms of ketamine is crucial, especially regarding its induction of schizophrenia-like symptoms. Recent research highlights the impact of ketamine on key brain regions associated with schizophrenia, specifically the prefrontal cortex (PFC) and hippocampus (Hip). This study focused on these regions to explore proteomic changes related to anxiety and cognitive impairment in a chronic ketamine-induced mouse model of schizophrenia. After twelve consecutive days of ketamine administration, brain tissues from these regions were dissected and analyzed. Using tandem mass tag (TMT) labeling quantitative proteomics techniques, 34,797 and 46,740 peptides were identified in PFC and Hip, corresponding to 5,668 and 6,463 proteins, respectively. In the PFC, a total of 113 proteins showed differential expression, primarily associated with the immuno-inflammatory process, calmodulin, postsynaptic density protein, and mitochondrial function. In the Hip, 129 differentially expressed proteins were screened, mainly related to synaptic plasticity proteins and mitochondrial respiratory chain complex-associated proteins. Additionally, we investigated key proteins within the glutamatergic synapse pathway and observed decreased expression levels of phosphorylated CaMKII and CREB. Overall, the study unveiled a significant proteomic signature in the chronic ketamine-induced schizophrenia mouse model, characterized by anxiety and cognitive impairment in both the PFC and Hip, and this comprehensive proteomic dataset may not only enhance our understanding of the molecular mechanisms underlying ketamine-related mental disorders but also offer valuable insights for future disease treatments.
Collapse
Affiliation(s)
- Li Xiao
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Ying Wei
- College of Pharmacy, North Sichuan Medical College, Nanchong, China
| | - Hong Yang
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Weihao Fan
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Linzhi Jiang
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Yi Ye
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Yongping Qin
- Clinical Pharmacology Laboratory, Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xia Wang
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Chunling Ma
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China.
| | - Linchuan Liao
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China.
| |
Collapse
|
23
|
Luo Y, Yu Y, He H, Fan N. Acute ketamine induces neuronal hyperexcitability and deficits in prepulse inhibition by upregulating IL-6. Prog Neuropsychopharmacol Biol Psychiatry 2024; 130:110913. [PMID: 38103855 DOI: 10.1016/j.pnpbp.2023.110913] [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: 09/11/2023] [Revised: 12/04/2023] [Accepted: 12/10/2023] [Indexed: 12/19/2023]
Abstract
Acute ketamine administration results in psychotic symptoms similar to those observed in schizophrenia and is regarded as a pharmacological model of schizophrenia. Accumulating evidence suggests that patients with schizophrenia show increased IL-6 levels in the blood and cerebrospinal fluid and that IL-6 levels are associated with the severity of psychotic symptoms. In the present study, we found that a single ketamine exposure led to increased expression of IL-6 and IL-6Rα, decreased dendritic spine density, increased expression and currents of T-type calcium channels, and increased neuron excitability in the hippocampal CA1 area 12 h after exposure. Acute ketamine administration also led to impaired prepulse inhibition (PPI) 12 h after administration. Additionally, we found that the expression of signaling molecules IKKα/β, NF-κB, JAK2, and STAT3 was upregulated 12 h after a single ketamine injection. The decreases in dendritic spine density, the increases in calcium currents and neuron excitability, and the impairments in PPI were ameliorated by blocking IL-6 or IL-6Rα. Our findings show that blocking IL-6 or its receptor may protect hippocampal neurons from hyperexcitability, thereby ameliorating ketamine-induced psychotic effects. Our study provides additional evidence that targeting IL-6 and its receptor is a potential strategy for treating psychotic symptoms in acute ketamine-induced psychosis and schizophrenia.
Collapse
Affiliation(s)
- Yayan Luo
- The Affiliated Brain Hospital of Guangzhou Medical University, 36 Mingxin Road, Liwan District, Guangzhou, Guangdong Province 510370, China
| | - Yang Yu
- The Affiliated Brain Hospital of Guangzhou Medical University, 36 Mingxin Road, Liwan District, Guangzhou, Guangdong Province 510370, China
| | - Hongbo He
- The Affiliated Brain Hospital of Guangzhou Medical University, 36 Mingxin Road, Liwan District, Guangzhou, Guangdong Province 510370, China
| | - Ni Fan
- The Affiliated Brain Hospital of Guangzhou Medical University, 36 Mingxin Road, Liwan District, Guangzhou, Guangdong Province 510370, China.
| |
Collapse
|
24
|
Ledesma-Corvi S, Jornet-Plaza J, Gálvez-Melero L, García-Fuster MJ. Novel rapid treatment options for adolescent depression. Pharmacol Res 2024; 201:107085. [PMID: 38309382 DOI: 10.1016/j.phrs.2024.107085] [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: 08/01/2023] [Revised: 12/13/2023] [Accepted: 01/25/2024] [Indexed: 02/05/2024]
Abstract
There is an urgent need for novel fast-acting antidepressants for adolescent treatment-resistant depression and/or suicidal risk, since the selective serotonin reuptake inhibitors that are clinically approved for that age (i.e., fluoxetine or escitalopram) take weeks to work. In this context, one of the main research lines of our group is to characterize at the preclinical level novel approaches for rapid-acting antidepressants for adolescence. The present review summarizes the potential use in adolescence of non-pharmacological options, such as neuromodulators (electroconvulsive therapy and other innovative types of brain stimulation), as well as pharmacological options, including consciousness-altering drugs (mainly ketamine but also classical psychedelics) and cannabinoids (i.e., cannabidiol), with promising fast-acting responses. Following a brief analytical explanation of adolescent depression, we present a general introduction for each therapeutical approach together with the clinical evidence supporting its potential beneficial use in adolescence (mainly extrapolated from prior successful examples for adults), to then report recent and/or ongoing preclinical studies that will aid in improving the inclusion of these therapies in the clinic, by considering potential sex-, age-, and dose-related differences, and/or other factors that might affect efficacy or long-term safety. Finally, we conclude the review by providing future avenues to maximize treatment response, including the need for more clinical studies and the importance of designing and/or testing novel treatment options that are safe and fast-acting for adolescent depression.
Collapse
Affiliation(s)
- Sandra Ledesma-Corvi
- Neuropharmacology Research Group, IUNICS, University of the Balearic Islands, Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Jordi Jornet-Plaza
- Neuropharmacology Research Group, IUNICS, University of the Balearic Islands, Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Laura Gálvez-Melero
- Neuropharmacology Research Group, IUNICS, University of the Balearic Islands, Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - M Julia García-Fuster
- Neuropharmacology Research Group, IUNICS, University of the Balearic Islands, Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain; Department of Medicine, University of the Balearic Islands, Palma, Spain.
| |
Collapse
|
25
|
Howell AM, Anticevic A. Functional Connectivity Biomarkers in Schizophrenia. ADVANCES IN NEUROBIOLOGY 2024; 40:237-283. [PMID: 39562448 DOI: 10.1007/978-3-031-69491-2_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Schizophrenia is a debilitating neuropsychiatric disorder that affects approximately 1% of the population and poses a major public health problem. Despite over 100 years of study, the treatment for schizophrenia remains limited, partially due to the lack of knowledge about the neural mechanisms of the illness and how they relate to symptoms. The US Food and Drug Administration (FDA) and the National Institute of Health (NIH) have provided seven biomarker categories that indicate causes, risks, and treatment responses. However, no FDA-approved biomarkers exist for psychiatric conditions, including schizophrenia, highlighting the need for biomarker development. Over three decades, magnetic resonance imaging (MRI)-based studies have identified patterns of abnormal brain function in schizophrenia. By using functional connectivity (FC) data, which gauges how brain regions interact over time, these studies have differentiated patient subgroups, predicted responses to antipsychotic medication, and correlated neural changes with symptoms. This suggests FC metrics could serve as promising biomarkers. Here, we present a selective review of studies leveraging MRI-derived FC to study neural alterations in schizophrenia, discuss how they align with FDA-NIH biomarkers, and outline the challenges and goals for developing FC biomarkers in schizophrenia.
Collapse
Affiliation(s)
| | - Alan Anticevic
- Yale University, School of Medicine, New Haven, CT, USA.
| |
Collapse
|
26
|
Rodrigues T, Bressan GN, Krum BN, Soares FAA, Fachinetto R. Influence of the dose of ketamine used on schizophrenia-like symptoms in mice: A correlation study with TH, GAD 67, and PPAR-γ. Pharmacol Biochem Behav 2023; 233:173658. [PMID: 37804866 DOI: 10.1016/j.pbb.2023.173658] [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: 07/14/2023] [Revised: 10/03/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Schizophrenia is a chronic, debilitating mental illness that has not yet been completely understood. In this study, we aimed to investigate the effects of different doses of ketamine, a non-competitive NMDA receptor antagonist, on the positive- and negative-like symptoms of schizophrenia. We also explored whether these effects are related to changes in the immunoreactivity of GAD67, TH, and PPAR-γ in brain structures. To conduct the study, male mice received ketamine (20-40 mg/kg) or its vehicle (0.9 % NaCl) intraperitoneally for 14 consecutive days. We quantified stereotyped behavior, the time of immobility in the forced swimming test (FST), and locomotor activity after 7 or 14 days. In addition, we performed ex vivo analysis of the immunoreactivity of GAD, TH, and PPAR-γ, in brain tissues after 14 days. The results showed that ketamine administration for 14 days increased the grooming time in the nose region at all tested doses. It also increased immobility in the FST at 30 mg/kg doses and decreased the number of rearing cycles during stereotyped behavior at 40 mg/kg. These behavioral effects were not associated with changes in locomotor activity. We did not observe any significant alterations regarding the immunoreactivity of brain proteins. However, we found that GAD and TH were positively correlated with the number of rearing during the stereotyped behavior at doses of 20 and 30 mg/kg ketamine, respectively. GAD was positively correlated with the number of rearing in the open field test at a dose of 20 mg/kg. TH was inversely correlated with immobility time in the FST at a dose of 30 mg/kg. PPAR-γ was inversely correlated with the number of bouts of stereotyped behavior at a dose of 40 mg/kg of ketamine. In conclusion, the behavioral alterations induced by ketamine in positive-like symptoms were reproduced with all doses tested and appear to depend on the modulatory effects of TH, GAD, and PPAR-γ. Conversely, negative-like symptoms were associated with a specific dose of ketamine.
Collapse
Affiliation(s)
- Talita Rodrigues
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Getulio Nicola Bressan
- Programa de Pós-Graduação em Ciências Biológicas, Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil
| | - Bárbara Nunes Krum
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Félix Alexandre Antunes Soares
- Programa de Pós-Graduação em Ciências Biológicas, Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil
| | - Roselei Fachinetto
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil.
| |
Collapse
|
27
|
Lehmann M, Ettinger U. Metacognitive monitoring in schizotypy: Systematic literature review and new empirical data. J Behav Ther Exp Psychiatry 2023; 81:101891. [PMID: 37453406 DOI: 10.1016/j.jbtep.2023.101891] [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: 11/14/2022] [Revised: 06/21/2023] [Accepted: 06/25/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND AND OBJECTIVES Deficits in metacognition, the ability to monitor one's own mental states, are key elements of the functional pathology of schizophrenia spectrum disorders. Little is known, however, about the integrity of metacognitive processes in subclinical schizotypy. The purpose of the present investigation was two-fold: First, we conducted a preregistered, systematic literature review to synthesize previous research efforts on the role of metacognition in schizotypy. Second, we investigated the relationship between self-reported dimensions of schizotypy and psychometric as well as behavioral measures of metacognition in a preregistered online study. METHODS A large sample (N = 330) completed a questionnaire battery and an episodic memory experiment; task-based metacognition was tapped via trial-by-trial confidence ratings. RESULTS In keeping with findings from our literature review, higher schizotypy was associated with diminished introspective insight and an overly self-referential and maladaptive metacognitive style in metacognition questionnaires. Importantly, low task-based metacognitive efficiency was predictive of high levels of cognitive disorganization, whereas task-related overconfidence (i.e., increased metacognitive bias) was linked with positive schizotypy. LIMITATIONS Due to the comparatively small number of k = 20 studies meeting our inclusion criteria, the systematic literature review provides only preliminary indications for potential conclusions. Furthermore, control over potential disturbing influences in the experimental study was limited due to its online format. CONCLUSIONS Overall, we provide evidence for specific metacognitive deficits in schizotypy and discuss a potential continuity of preserved and impaired aspects of metacognitive monitoring along the psychosis continuum.
Collapse
Affiliation(s)
- Mirko Lehmann
- Department of Psychology, University of Bonn, Kaiser-Karl-Ring 9, 53111, Bonn, NRW, Germany.
| | - Ulrich Ettinger
- Department of Psychology, University of Bonn, Kaiser-Karl-Ring 9, 53111, Bonn, NRW, Germany.
| |
Collapse
|
28
|
Hilal F, Jeanblanc J, Naassila M. [Interest and mechanisms of action of ketamine in alcohol addiction- A review of clinical and preclinical studies]. Biol Aujourdhui 2023; 217:161-182. [PMID: 38018944 DOI: 10.1051/jbio/2023028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Indexed: 11/30/2023]
Abstract
Alcohol Use Disorder (AUD) is a psychiatric condition characterized by chronic and excessive drinking despite negative consequences on overall health and social or occupational functioning. There are currently limited treatment options available for AUD, and the effects size and the response rates to these treatments are often low to moderate. The World Health Organization has identified the development of medications to treat AUD as one of its 24 priorities. This past decade was marked by a renewed interest in psychedelic use in psychiatry. At the centre of this renaissance, ketamine, an atypical psychedelic already used in the treatment of major depression, is an NMDA receptor antagonist that exists as a racemic compound made of two enantiomers, S-ketamine, and R-ketamine. Each form can be metabolized into different metabolites, some of which having antidepressant properties. In this article, we review both clinical and preclinical studies on ketamine and its metabolites in the treatment of AUD. Preclinical as well as clinical studies have revealed that ketamine is effective in reducing withdrawal symptoms and alcohol craving. Convergent data showed that antidepressant properties of ketamine largely contribute to the decreased likelihood of alcohol relapse, especially in patients undergoing ketamine-assisted psychotherapies. Its effectiveness is believed to be linked with its ability to regulate the glutamatergic pathway, enhance neuroplasticity, rewire brain resting state network functional connectivity and decrease depressive-like states. However, it remains to further investigate (i) why strong differences exist between male and female responses in preclinical studies and (ii) the respective roles of each of the metabolites in the ketamine effects in both genders. Interestingly, current studies are also focusing on ketamine addiction and the comorbidity between alcohol addiction and depression occurring more frequently in females.
Collapse
Affiliation(s)
- Fahd Hilal
- Groupe de recherche sur l'alcool et les pharmacodépendances, INSERM U1247, CURS, Amiens, France
| | - Jérôme Jeanblanc
- Groupe de recherche sur l'alcool et les pharmacodépendances, INSERM U1247, CURS, Amiens, France
| | - Mickaël Naassila
- Groupe de recherche sur l'alcool et les pharmacodépendances, INSERM U1247, CURS, Amiens, France
| |
Collapse
|
29
|
Rajagopal L, Mahjour S, Huang M, Ryan CA, Elzokaky A, Csakai AJ, Orr MJ, Scheidt K, Meltzer HY. NU-1223, a simplified analog of alstonine, with 5-HT 2cR agonist-like activity, rescues memory deficit and positive and negative symptoms in subchronic phencyclidine mouse model of schizophrenia. Behav Brain Res 2023; 454:114614. [PMID: 37572758 DOI: 10.1016/j.bbr.2023.114614] [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: 03/27/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/14/2023]
Abstract
The serotonin (5-HT)2 C receptor(R) is a widely distributed G-protein-coupled receptor, expressed abundantly in the central nervous system. Alstonine is a natural product that has significant properties of atypical antipsychotic drugs (AAPDs), in part attributed to 5-HT2 CR agonism. Based on alstonine, we developed NU-1223, a simplified β carboline analog of alstonine, which shows efficacies comparable to alstonine and to other 5-HT2 CR agonists, Ro-60-0175 and lorcaserin. The 5-HT2 CR antagonism of some APDs, including olanzapine, contributes to weight gain, a major side effect which limits its tolerability, while the 5-HT2 CR agonists and/or modulators, may minimize weight gain. We used the well-established rodent subchronic phencyclidine (PCP) model to test the efficacy of NU-1223 on episodic memory, using novel object recognition (NOR) task, positive (locomotor activity), and negative symptoms (social interaction) of schizophrenia (SCH). We found that NU-1223 produced both transient and prolonged rescue of the subchronic PCP-induced deficits in NOR and SI. Further, NU-1223, but not Ro-60-0175, blocked PCP and amphetamine (AMPH)-induced increase in LMA in subchronic PCP mice. These transient efficacies in LMA were blocked by the 5-HT2 CR antagonist, SB242084. Sub-chronic NU-1223 treatment rescued NOR and SI deficits in subchronic PCP mice for at least 39 days after 3 days injection. Chronic treatment with NU-1223, ip, twice a day for 21 days, did not increase average body weight vs olanzapine. These findings clearly indicate NU-1223 as a class of small molecules with a possible 5-HT2 CR-agonist-like mechanism of action, attributing to its efficacy. Additional in-depth receptor mechanistic studies are warranted, as this small molecule, both transiently and chronically rescued PCP-induced deficits. Furthermore, NU-1223 did not induce weight gain post long-term administrations vs AAPDs such as olanzapine, making NU-1223 a putative therapeutic compound for SCH.
Collapse
Affiliation(s)
- Lakshmi Rajagopal
- Department of Psychiatry and Behavioral Sciences, Northwestern Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Sanaz Mahjour
- Department of Psychiatry and Behavioral Sciences, Northwestern Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Mei Huang
- Department of Psychiatry and Behavioral Sciences, Northwestern Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Chelsea A Ryan
- Department of Psychiatry and Behavioral Sciences, Northwestern Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ahmad Elzokaky
- Department of Psychiatry and Behavioral Sciences, Northwestern Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Adam J Csakai
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA
| | - Meghan J Orr
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA
| | - Karl Scheidt
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA; Department of Pharmacology, Northwestern Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Herbert Y Meltzer
- Department of Psychiatry and Behavioral Sciences, Northwestern Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
30
|
de Oliveira EG, de Lima DA, da Silva Júnior JC, de Souza Barbosa MV, de Andrade Silva SC, de Santana JH, Dos Santos Junior OH, Lira EC, Lagranha CJ, Duarte FS, Gomes DA. (R)-ketamine attenuates neurodevelopmental disease-related phenotypes in a mouse model of maternal immune activation. Eur Arch Psychiatry Clin Neurosci 2023; 273:1501-1512. [PMID: 37249625 DOI: 10.1007/s00406-023-01629-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/22/2023] [Indexed: 05/31/2023]
Abstract
Infections during pregnancy are associated with an increased risk of neuropsychiatric disorders with developmental etiologies, such as schizophrenia and autism spectrum disorders (ASD). Studies have shown that the animal model of maternal immune activation (MIA) reproduces a wide range of phenotypes relevant to the study of neurodevelopmental disorders. Emerging evidence shows that (R)-ketamine attenuates behavioral, cellular, and molecular changes observed in animal models of neuropsychiatric disorders. Here, we investigate whether (R)-ketamine administration during adolescence attenuates some of the phenotypes related to neurodevelopmental disorders in an animal model of MIA. For MIA, pregnant Swiss mice received intraperitoneally (i.p.) lipopolysaccharide (LPS; 100 µg/kg/day) or saline on gestational days 15 and 16. The two MIA-based groups of male offspring received (R)-ketamine (20 mg/kg/day; i.p.) or saline from postnatal day (PND) 36 to 50. At PND 62, the animals were examined for anxiety-like behavior and locomotor activity in the open-field test (OFT), as well as in the social interaction test (SIT). At PND 63, the prefrontal cortex (PFC) was collected for analysis of oxidative balance and gene expression of the cytokines IL-1β, IL-6, and TGF-β1. We show that (R)-ketamine abolishes anxiety-related behavior and social interaction deficits induced by MIA. Additionally, (R)-ketamine attenuated the increase in lipid peroxidation and the cytokines in the PFC of the offspring exposed to MIA. The present work suggests that (R)-ketamine administration may have a long-lasting attenuation in deficits in emotional behavior induced by MIA, and that these effects may be attributed to its antioxidant and anti-inflammatory activity in the PFC.
Collapse
Affiliation(s)
- Elifrances Galdino de Oliveira
- Laboratory of Neuroendocrinology and Metabolism, Department of Physiology and Pharmacology, Bioscience Center, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235 Cidade Universitária, Recife, PE, 50670-901, Brazil.
- Graduate Program of Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, PE, Brazil.
| | - Diógenes Afonso de Lima
- Laboratory of Neuroendocrinology and Metabolism, Department of Physiology and Pharmacology, Bioscience Center, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235 Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - José Carlos da Silva Júnior
- Laboratory of Neuroendocrinology and Metabolism, Department of Physiology and Pharmacology, Bioscience Center, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235 Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Mayara Victória de Souza Barbosa
- Laboratory of Neuroendocrinology and Metabolism, Department of Physiology and Pharmacology, Bioscience Center, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235 Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Severina Cassia de Andrade Silva
- Graduate Program of Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, PE, Brazil
- Laboratory of Biochemistry and Exercise Biochemistry, Department of Physical Education and Sports Science, Federal University of Pernambuco, Vitória de Santo Antão, PE, Brazil
| | - Jonata Henrique de Santana
- Graduate Program of Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, PE, Brazil
- Laboratory of Biochemistry and Exercise Biochemistry, Department of Physical Education and Sports Science, Federal University of Pernambuco, Vitória de Santo Antão, PE, Brazil
| | - Osmar Henrique Dos Santos Junior
- Graduate Program of Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, PE, Brazil
- Laboratory of Biochemistry and Exercise Biochemistry, Department of Physical Education and Sports Science, Federal University of Pernambuco, Vitória de Santo Antão, PE, Brazil
| | - Eduardo Carvalho Lira
- Laboratory of Neuroendocrinology and Metabolism, Department of Physiology and Pharmacology, Bioscience Center, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235 Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Claudia Jacques Lagranha
- Graduate Program of Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, PE, Brazil
- Laboratory of Biochemistry and Exercise Biochemistry, Department of Physical Education and Sports Science, Federal University of Pernambuco, Vitória de Santo Antão, PE, Brazil
| | - Filipe Silveira Duarte
- Laboratory of Neuroendocrinology and Metabolism, Department of Physiology and Pharmacology, Bioscience Center, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235 Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Dayane Aparecida Gomes
- Laboratory of Neuroendocrinology and Metabolism, Department of Physiology and Pharmacology, Bioscience Center, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235 Cidade Universitária, Recife, PE, 50670-901, Brazil
- Graduate Program of Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, PE, Brazil
| |
Collapse
|
31
|
Wang R, Peterson Z, Balasubramanian N, Khan KM, Chimenti MS, Thedens D, Nickl-Jockschat T, Marcinkiewcz CA. Lateral Septal Circuits Govern Schizophrenia-Like Effects of Ketamine on Social Behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.08.552372. [PMID: 37609170 PMCID: PMC10441349 DOI: 10.1101/2023.08.08.552372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Schizophrenia is marked by poor social functioning that can have a severe impact on quality of life and independence, but the underlying neural circuity is not well understood. Here we used a translational model of subanesthetic ketamine in mice to delineate neural pathways in the brain linked to social deficits in schizophrenia. Mice treated with chronic ketamine (30 mg/kg/day for 10 days) exhibit profound social and sensorimotor deficits as previously reported. Using three- dimensional c-Fos immunolabeling and volume imaging (iDISCO), we show that ketamine treatment resulted in hypoactivation of the lateral septum (LS) in response to social stimuli. Chemogenetic activation of the LS rescued social deficits after ketamine treatment, while chemogenetic inhibition of previously active populations in the LS (i.e. social engram neurons) recapitulated social deficits in ketamine-naïve mice. We then examined the translatome of LS social engram neurons and found that ketamine treatment dysregulated genes implicated in neuronal excitability and apoptosis, which may contribute to LS hypoactivation. We also identified 38 differentially expressed genes (DEGs) in common with human schizophrenia, including those involved in mitochondrial function, apoptosis, and neuroinflammatory pathways. Chemogenetic activation of LS social engram neurons induced downstream activity in the ventral part of the basolateral amygdala, subparafascicular nucleus of the thalamus, intercalated amygdalar nucleus, olfactory areas, and dentate gyrus, and it also reduces connectivity of the LS with the piriform cortex and caudate-putamen. In sum, schizophrenia-like social deficits may emerge via changes in the intrinsic excitability of a discrete subpopulation of LS neurons that serve as a central hub to coordinate social behavior via downstream projections to reward, fear extinction, motor and sensory processing regions of the brain.
Collapse
|
32
|
Speers LJ, Sissons DJ, Cleland L, Bilkey DK. Hippocampal phase precession is preserved under ketamine, but the range of precession across a theta cycle is reduced. J Psychopharmacol 2023; 37:809-821. [PMID: 37515458 PMCID: PMC10399102 DOI: 10.1177/02698811231187339] [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] [Indexed: 07/30/2023]
Abstract
BACKGROUND Hippocampal phase precession, which depends on the precise spike timing of place cells relative to local theta oscillations, has been proposed to underlie sequential memory. N-methyl-D-asparate (NMDA) receptor antagonists such as ketamine disrupt memory and also reproduce several schizophrenia-like symptoms, including spatial memory impairments and disorganized cognition. It is possible that these impairments result from disruptions to phase precession. AIMS/METHODS We used an ABA design to test whether an acute, subanesthetic dose (7.5 mg/kg) of ketamine disrupted phase precession in CA1 of male rats as they navigated around a rectangular track for a food reward. RESULTS/OUTCOMES Ketamine did not affect the ability of CA1 place cells to precess despite changes to place cell firing rates, local field potential properties and locomotor speed. However, ketamine reduced the range of phase precession that occurred across a theta cycle. CONCLUSION Phase precession is largely robust to acute NMDA receptor antagonism by ketamine, but the reduced range of precession could have important implications for learning and memory.
Collapse
Affiliation(s)
| | - Daena J Sissons
- Psychology Department, Otago University Dunedin, New Zealand
- Psychology Department, University of Canterbury, Christchurch, New Zealand
| | - Lana Cleland
- Psychology Department, Otago University Dunedin, New Zealand
- Department Psychological Medicine, Otago University, Christchurch, New Zealand
- Department Population Health, Otago University, Christchurch, New Zealand
| | - David K Bilkey
- Psychology Department, Otago University Dunedin, New Zealand
| |
Collapse
|
33
|
Castillo A, Dubois J, Field RM, Fishburn F, Gundran A, Ho WC, Jawhar S, Kates-Harbeck J, M Aghajan Z, Miller N, Perdue KL, Phillips J, Ryan WC, Shafiei M, Scholkmann F, Taylor M. Measuring acute effects of subanesthetic ketamine on cerebrovascular hemodynamics in humans using TD-fNIRS. Sci Rep 2023; 13:11665. [PMID: 37468572 PMCID: PMC10356754 DOI: 10.1038/s41598-023-38258-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 07/05/2023] [Indexed: 07/21/2023] Open
Abstract
Quantifying neural activity in natural conditions (i.e. conditions comparable to the standard clinical patient experience) during the administration of psychedelics may further our scientific understanding of the effects and mechanisms of action. This data may facilitate the discovery of novel biomarkers enabling more personalized treatments and improved patient outcomes. In this single-blind, placebo-controlled study with a non-randomized design, we use time-domain functional near-infrared spectroscopy (TD-fNIRS) to measure acute brain dynamics after intramuscular subanesthetic ketamine (0.75 mg/kg) and placebo (saline) administration in healthy participants (n = 15, 8 females, 7 males, age 32.4 ± 7.5 years) in a clinical setting. We found that the ketamine administration caused an altered state of consciousness and changes in systemic physiology (e.g. increase in pulse rate and electrodermal activity). Furthermore, ketamine led to a brain-wide reduction in the fractional amplitude of low frequency fluctuations, and a decrease in the global brain connectivity of the prefrontal region. Lastly, we provide preliminary evidence that a combination of neural and physiological metrics may serve as predictors of subjective mystical experiences and reductions in depressive symptomatology. Overall, our study demonstrated the successful application of fNIRS neuroimaging to study the physiological effects of the psychoactive substance ketamine in humans, and can be regarded as an important step toward larger scale clinical fNIRS studies that can quantify the impact of psychedelics on the brain in standard clinical settings.
Collapse
Affiliation(s)
| | - Julien Dubois
- Kernel, 5042 Wilshire Blvd, #26878, Los Angeles, CA, 90036, USA
| | - Ryan M Field
- Kernel, 5042 Wilshire Blvd, #26878, Los Angeles, CA, 90036, USA
| | - Frank Fishburn
- Kernel, 5042 Wilshire Blvd, #26878, Los Angeles, CA, 90036, USA
| | - Andrew Gundran
- Kernel, 5042 Wilshire Blvd, #26878, Los Angeles, CA, 90036, USA
| | - Wilson C Ho
- Kernel, 5042 Wilshire Blvd, #26878, Los Angeles, CA, 90036, USA
| | - Sami Jawhar
- Kernel, 5042 Wilshire Blvd, #26878, Los Angeles, CA, 90036, USA
| | | | - Zahra M Aghajan
- Kernel, 5042 Wilshire Blvd, #26878, Los Angeles, CA, 90036, USA
| | - Naomi Miller
- Kernel, 5042 Wilshire Blvd, #26878, Los Angeles, CA, 90036, USA
| | | | - Jake Phillips
- Kernel, 5042 Wilshire Blvd, #26878, Los Angeles, CA, 90036, USA
| | - Wesley C Ryan
- Kernel, 5042 Wilshire Blvd, #26878, Los Angeles, CA, 90036, USA
| | - Mahdi Shafiei
- Kernel, 5042 Wilshire Blvd, #26878, Los Angeles, CA, 90036, USA
| | - Felix Scholkmann
- Scholkmann Data Analysis Services, Scientific Consulting and Physical Engineering, 8057, Zurich, Switzerland
- Neurophotonics and Biosignal Processing Research Group, Biomedical Optics Research Laboratory, Department of Neonatology, University Hospital Zurich, University of Zurich, 8091, Zurich, Switzerland
| | - Moriah Taylor
- Kernel, 5042 Wilshire Blvd, #26878, Los Angeles, CA, 90036, USA
| |
Collapse
|
34
|
Frohlich J, Mediano PAM, Bavato F, Gharabaghi A. Paradoxical pharmacological dissociations result from drugs that enhance delta oscillations but preserve consciousness. Commun Biol 2023; 6:654. [PMID: 37340024 DOI: 10.1038/s42003-023-04988-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/26/2023] [Indexed: 06/22/2023] Open
Abstract
Low-frequency (<4 Hz) neural activity, particularly in the delta band, is generally indicative of loss of consciousness and cortical down states, particularly when it is diffuse and high amplitude. Remarkably, however, drug challenge studies of several diverse classes of pharmacological agents-including drugs which treat epilepsy, activate GABAB receptors, block acetylcholine receptors, or produce psychedelic effects-demonstrate neural activity resembling cortical down states even as the participants remain conscious. Of those substances that are safe to use in healthy volunteers, some may be highly valuable research tools for investigating which neural activity patterns are sufficient for consciousness or its absence.
Collapse
Affiliation(s)
- Joel Frohlich
- Institute for Neuromodulation and Neurotechnology, University Hospital and University of Tuebingen, Tuebingen, Germany.
| | - Pedro A M Mediano
- Department of Computing, Imperial College London, London, UK
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - Francesco Bavato
- Experimental and Clinical Pharmacopsychology, Department of Psychiatry, Psychotherapy, and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Alireza Gharabaghi
- Institute for Neuromodulation and Neurotechnology, University Hospital and University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
35
|
Ebrahimi M, Ahangar N, Zamani E, Shaki F. L-Carnitine Prevents Behavioural Alterations in Ketamine-Induced Schizophrenia in Mice: Possible Involvement of Oxidative Stress and Inflammation Pathways. J Toxicol 2023; 2023:9093231. [PMID: 37363159 PMCID: PMC10289879 DOI: 10.1155/2023/9093231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 01/10/2023] [Accepted: 06/07/2023] [Indexed: 06/28/2023] Open
Abstract
Schizophrenia is a chronic mental complaint known as cognitive impairment. There has been evidence that inflammation and oxidative stress play a main role in schizophrenia pathophysiology. This study aimed to investigate the effects of l-carnitine, as a potent antioxidant, on the treatment of behavioural and biochemical disturbances in mice with ketamine-induced schizophrenia. In this study, schizophrenia was induced in mice by ketamine (25 mg/kg/day, i.p). Before induction of schizophrenia, mice were treated with l-carnitine (100, 200, and 400 mg/kg/day, i.p). Then, behavioural impairments were evaluated by open field (OF) assessment and social interaction test (SIT). After brain tissue isolation, reactive oxygen species (ROS), glutathione concentration (GSH), lipid peroxidation (LPO), protein carbonyl oxidation, superoxide dismutase activity (SOD), and glutathione peroxidase activity (GPx) were assessed as oxidative stress markers. Furthermore, inflammatory biomarkers such as tumour necrosis factor alpha (TNF-α) and nitric oxide (NO) were evaluated in brain tissue. Our results showed ketamine increased inflammation and oxidative damage in brain tissue that was similar to behaviour disorders in mice. Interestingly, l-carnitine significantly decreased oxidative stress and inflammatory markers compared with ketamine-treated mice. In addition, l-carnitine prevented and reversed ketamine-induced alterations in the activities of SOD and GPx enzymes in mice's brains. Also, improved performance in OFT (locomotor activity test) and SIT was observed in l-carnitine-treated mice. These data provided evidence that, due to the antioxidant and anti-inflammatory effects of l-carnitine, it has a neuroprotective effect on mice model of schizophrenia.
Collapse
Affiliation(s)
- Mehrasa Ebrahimi
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
- Students Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nematollah Ahangar
- Department of Pharmacology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ehsan Zamani
- Department of Pharmacology and Toxicology, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Shaki
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
36
|
Shangase KB, Luvuno M, Mabandla MV. Investigating the Robustness of a Rodent "Double Hit" (Post-Weaning Social Isolation and NMDA Receptor Antagonist) Model as an Animal Model for Schizophrenia: A Systematic Review. Brain Sci 2023; 13:848. [PMID: 37371328 DOI: 10.3390/brainsci13060848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/16/2023] [Accepted: 05/21/2023] [Indexed: 06/29/2023] Open
Abstract
Schizophrenia is a debilitating psychiatric disorder comprising positive, negative, and cognitive impairments. Most of the animal models developed to understand the neurobiology and mechanism of schizophrenia do not produce all the symptoms of the disease. Therefore, researchers need to develop new animal models with greater translational reliability, and the ability to produce most if not all symptoms of schizophrenia. This review aimed to evaluate the effectiveness of the rodent "double hit" (post-weaning social isolation and N-methyl-D-aspartate (NMDA) receptor antagonist) model to produce symptoms of schizophrenia. This systematic review was developed according to the 2020 PRISMA guidelines and checklist. The MEDLINE (PubMed) and Ebscohost databases were used to search for studies. The systematic review is based on quantitative animal studies. Studies in languages other than English that could be translated sufficiently using Google translate were also included. Data extraction was performed individually by two independent reviewers and discrepancies between them were resolved by a third reviewer. SYRCLE's risk-of-bias tool was used to test the quality and biases of included studies. Our primary search yielded a total of 47 articles, through different study selection processes. Seventeen articles met the inclusion criteria for this systematic review. Ten of the seventeen studies found that the "double hit" model was more effective in developing various symptoms of schizophrenia. Most studies showed that the "double hit" model is robust and capable of inducing cognitive impairments and positive symptoms of schizophrenia.
Collapse
Affiliation(s)
- Khanyiso Bright Shangase
- Department of Human Physiology, School of Laboratory Medicine and Medical Science, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| | - Mluleki Luvuno
- Department of Human Physiology, School of Laboratory Medicine and Medical Science, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| | - Musa V Mabandla
- Department of Human Physiology, School of Laboratory Medicine and Medical Science, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| |
Collapse
|
37
|
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
|
38
|
Gonzalez-Burgos I, Bainier M, Gross S, Schoenenberger P, Ochoa JA, Valencia M, Redondo RL. Glutamatergic and GABAergic Receptor Modulation Present Unique Electrophysiological Fingerprints in a Concentration-Dependent and Region-Specific Manner. eNeuro 2023; 10:ENEURO.0406-22.2023. [PMID: 36931729 PMCID: PMC10124153 DOI: 10.1523/eneuro.0406-22.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 02/13/2023] [Accepted: 02/17/2023] [Indexed: 03/19/2023] Open
Abstract
Brain function depends on complex circuit interactions between excitatory and inhibitory neurons embedded in local and long-range networks. Systemic GABAA-receptor (GABAAR) or NMDA-receptor (NMDAR) modulation alters the excitatory-inhibitory balance (EIB), measurable with electroencephalography (EEG). However, EEG signatures are complex in localization and spectral composition. We developed and applied analytical tools to investigate the effects of two EIB modulators, MK801 (NMDAR antagonist) and diazepam (GABAAR modulator), on periodic and aperiodic EEG features in freely-moving male Sprague Dawley rats. We investigated how, across three brain regions, EEG features are correlated with EIB modulation. We found that the periodic component was composed of seven frequency bands that presented region-dependent and compound-dependent changes. The aperiodic component was also different between compounds and brain regions. Importantly, the parametrization into periodic and aperiodic components unveiled correlations between quantitative EEG and plasma concentrations of pharmacological compounds. MK-801 exposures were positively correlated with the slope of the aperiodic component. Concerning the periodic component, MK-801 exposures correlated negatively with the peak frequency of low-γ oscillations but positively with those of high-γ and high-frequency oscillations (HFOs). As for the power, θ and low-γ oscillations correlated negatively with MK-801, whereas mid-γ correlated positively. Diazepam correlated negatively with the knee of the aperiodic component, positively to β and negatively to low-γ oscillatory power, and positively to the modal frequency of θ, low-γ, mid-γ, and high-γ. In conclusion, correlations between exposures and pharmacodynamic effects can be better-understood thanks to the parametrization of EEG into periodic and aperiodic components. Such parametrization could be key in functional biomarker discovery.
Collapse
Affiliation(s)
- Irene Gonzalez-Burgos
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel 4070, Switzerland
- Program of Neuroscience, Centro de Investigación Médica Aplicada, Universidad de Navarra, Pamplona 31080, Spain
- Instituto de Investigación Sanitaria de Navarra (Navarra Institute for Health Research), Pamplona 31080, Spain
| | - Marie Bainier
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Simon Gross
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Philipp Schoenenberger
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel 4070, Switzerland
| | - José A Ochoa
- Program of Neuroscience, Centro de Investigación Médica Aplicada, Universidad de Navarra, Pamplona 31080, Spain
- Instituto de Investigación Sanitaria de Navarra (Navarra Institute for Health Research), Pamplona 31080, Spain
| | - Miguel Valencia
- Program of Neuroscience, Centro de Investigación Médica Aplicada, Universidad de Navarra, Pamplona 31080, Spain
- Instituto de Investigación Sanitaria de Navarra (Navarra Institute for Health Research), Pamplona 31080, Spain
- Institute of Data Science and Artificial Intelligence, Universidad de Navarra, Pamplona, Spain
| | - Roger L Redondo
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel 4070, Switzerland
| |
Collapse
|
39
|
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
|
40
|
Functional connectivity signatures of NMDAR dysfunction in schizophrenia-integrating findings from imaging genetics and pharmaco-fMRI. Transl Psychiatry 2023; 13:59. [PMID: 36797233 PMCID: PMC9935542 DOI: 10.1038/s41398-023-02344-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 01/26/2023] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Both, pharmacological and genome-wide association studies suggest N-methyl-D-aspartate receptor (NMDAR) dysfunction and excitatory/inhibitory (E/I)-imbalance as a major pathophysiological mechanism of schizophrenia. The identification of shared fMRI brain signatures of genetically and pharmacologically induced NMDAR dysfunction may help to define biomarkers for patient stratification. NMDAR-related genetic and pharmacological effects on functional connectivity were investigated by integrating three different datasets: (A) resting state fMRI data from 146 patients with schizophrenia genotyped for the disease-associated genetic variant rs7191183 of GRIN2A (encoding the NMDAR 2 A subunit) as well as 142 healthy controls. (B) Pharmacological effects of the NMDAR antagonist ketamine and the GABA-A receptor agonist midazolam were obtained from a double-blind, crossover pharmaco-fMRI study in 28 healthy participants. (C) Regional gene expression profiles were estimated using a postmortem whole-brain microarray dataset from six healthy donors. A strong resemblance was observed between the effect of the genetic variant in schizophrenia and the ketamine versus midazolam contrast of connectivity suggestive for an associated E/I-imbalance. This similarity became more pronounced for regions with high density of NMDARs, glutamatergic neurons, and parvalbumin-positive interneurons. From a functional perspective, increased connectivity emerged between striato-pallido-thalamic regions and cortical regions of the auditory-sensory-motor network, while decreased connectivity was observed between auditory (superior temporal gyrus) and visual processing regions (lateral occipital cortex, fusiform gyrus, cuneus). Importantly, these imaging phenotypes were associated with the genetic variant, the differential effect of ketamine versus midazolam and schizophrenia (as compared to healthy controls). Moreover, the genetic variant was associated with language-related negative symptomatology which correlated with disturbed connectivity between the left posterior superior temporal gyrus and the superior lateral occipital cortex. Shared genetic and pharmacological functional connectivity profiles were suggestive of E/I-imbalance and associated with schizophrenia. The identified brain signatures may help to stratify patients with a common molecular disease pathway providing a basis for personalized psychiatry.
Collapse
|
41
|
Nakamura T, Dinh TH, Asai M, Matsumoto J, Nishimaru H, Setogawa T, Honda S, Yamada H, Mihara T, Nishijo H. Suppressive effects of ketamine on auditory steady-state responses in intact, awake macaques: A non-human primate model of schizophrenia. Brain Res Bull 2023; 193:84-94. [PMID: 36539101 DOI: 10.1016/j.brainresbull.2022.12.006] [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: 11/01/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Auditory steady-state responses (ASSRs) are recurrent neural activities entrained to regular cyclic auditory stimulation. ASSRs are altered in individuals with schizophrenia, and may be related to hypofunction of the N-methyl-D-aspartate (NMDA) glutamate receptor. Noncompetitive NMDA receptor antagonists, including ketamine, have been used in ASSR studies of rodent models of schizophrenia. Although animal studies using non-human primates are required to complement rodent studies, the effects of ketamine on ASSRs are unknown in intact awake non-human primates. In this study, after administration of vehicle or ketamine, click trains at 20-83.3 Hz were presented to elicit ASSRs during recording of electroencephalograms in intact, awake macaque monkeys. The results indicated that ASSRs quantified by event-related spectral perturbation and inter-trial coherence were maximal at 83.3 Hz after vehicle administration, and that ketamine reduced ASSRs at 58.8 and 83.3 Hz, but not at 20 and 40 Hz. The present results demonstrated a reduction of ASSRs by the NMDA receptor antagonist at optimal frequencies with maximal responses in intact, awake macaques, comparable to ASSR reduction in patients with schizophrenia. These findings suggest that ASSR can be used as a neurophysiological biomarker of the disturbance of gamma-oscillatory neural circuits in this ketamine model of schizophrenia using intact, awake macaques. Thus, this model with ASSRs would be useful in the investigation of human brain pathophysiology as well as in preclinical translational research.
Collapse
Affiliation(s)
- Tomoya Nakamura
- System Emotional Science, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan; Department of Anatomy, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Trong Ha Dinh
- System Emotional Science, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan; Department of Physiology, Vietnam Military Medical University, Hanoi 100000, Viet Nam
| | - Makoto Asai
- Candidate Discovery Science Labs, Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| | - Jumpei Matsumoto
- System Emotional Science, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan; Research Center for Idling Brain Science (RCIBS), University of Toyama, Toyama 930-0194, Japan
| | - Hiroshi Nishimaru
- System Emotional Science, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan; Research Center for Idling Brain Science (RCIBS), University of Toyama, Toyama 930-0194, Japan
| | - Tsuyoshi Setogawa
- System Emotional Science, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan; Research Center for Idling Brain Science (RCIBS), University of Toyama, Toyama 930-0194, Japan
| | - Sokichi Honda
- Candidate Discovery Science Labs, Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| | - Hiroshi Yamada
- Candidate Discovery Science Labs, Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| | - Takuma Mihara
- Candidate Discovery Science Labs, Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| | - Hisao Nishijo
- System Emotional Science, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan; Research Center for Idling Brain Science (RCIBS), University of Toyama, Toyama 930-0194, Japan.
| |
Collapse
|
42
|
Koller WN, Cannon TD. Aberrant memory and delusional ideation: A pernicious partnership? Clin Psychol Rev 2023; 99:102231. [PMID: 36469975 DOI: 10.1016/j.cpr.2022.102231] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/02/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022]
Abstract
Delusions can be conceptualized as beliefs that are both at odds with consensus reality and espoused with high conviction. While delusions represent a cardinal symptom of schizophrenia, delusion-like beliefs can be found in the general population. Do similar cognitive mechanisms support delusionality across this spectrum? If so, what are they? Here, we examine evidence for a mechanistic role of the (associative) memory system in the formation and maintenance of delusions and delusion-like beliefs. While general neurocognitive metrics do not tend to associate with delusionality, our scoping review of the clinical and subclinical literature reveals several subdomains of memory function that do. These include a propensity to commit errors of commission (i.e., false alarms and intrusions), source memory biases, and metamemory impairment. We discuss how several of these effects may stem from aberrant associative memory function and offer recommendations for future research. Further, we propose a state/trait interaction model in which underlying traits (i.e., impaired associative and metamemory function) may become coupled with delusionality during states of acute psychosis, when memory function is particularly challenged by aberrant salience attribution and noisy perceptual input. According to this model, delusions may arise as explanations to high-salience (but low-source) mnemonic content that is endorsed with high confidence.
Collapse
Affiliation(s)
- William N Koller
- Department of Psychology, Yale University, Hillhouse Avenue, New Haven, CT 06520-8205, United States of America.
| | - Tyrone D Cannon
- Department of Psychology, Yale University, Hillhouse Avenue, New Haven, CT 06520-8205, United States of America
| |
Collapse
|
43
|
Haaf M, Curic S, Rauh J, Steinmann S, Mulert C, Leicht G. Opposite Modulation of the NMDA Receptor by Glycine and S-Ketamine and the Effects on Resting State EEG Gamma Activity: New Insights into the Glutamate Hypothesis of Schizophrenia. Int J Mol Sci 2023; 24:ijms24031913. [PMID: 36768234 PMCID: PMC9916476 DOI: 10.3390/ijms24031913] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/21/2023] Open
Abstract
NMDA-receptor hypofunction is increasingly considered to be an important pathomechanism in schizophrenia. However, to date, it has not been possible to identify patients with relevant NMDA-receptor hypofunction who would respond to glutamatergic treatments. Preclinical models, such as the ketamine model, could help identify biomarkers related to NMDA-receptor function that respond to glutamatergic modulation, for example, via activation of the glycine-binding site. We, therefore, aimed to investigate the effects of opposing modulation of the NMDA receptor on gamma activity (30-100 Hz) at rest, the genesis of which appears to be highly dependent on NMDA receptors. The effects of subanesthetic doses of S-ketamine and pretreatment with glycine on gamma activity at rest were examined in twenty-five healthy male participants using 64-channel electroencephalography. Psychometric scores were assessed using the PANSS and the 5D-ASC. While S-ketamine significantly increased psychometric scores and gamma activity at the scalp and in the source space, pretreatment with glycine did not significantly attenuate any of these effects when controlled for multiple comparisons. Our results question whether increased gamma activity at rest constitutes a suitable biomarker for the target engagement of glutamatergic drugs in the preclinical ketamine model. They might further point to a differential role of NMDA receptors in gamma activity generation.
Collapse
Affiliation(s)
- Moritz Haaf
- Department of Psychiatry and Psychotherapy, Psychiatry Neuroimaging Branch (PNB), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Correspondence: ; Tel.: +49-(0)40-741059514
| | - Stjepan Curic
- Department of Psychiatry and Psychotherapy, Psychiatry Neuroimaging Branch (PNB), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Jonas Rauh
- Department of Psychiatry and Psychotherapy, Psychiatry Neuroimaging Branch (PNB), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Saskia Steinmann
- Department of Psychiatry and Psychotherapy, Psychiatry Neuroimaging Branch (PNB), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Christoph Mulert
- Department of Psychiatry and Psychotherapy, Psychiatry Neuroimaging Branch (PNB), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Center of Psychiatry, Justus-Liebig University, 35392 Giessen, Germany
| | - Gregor Leicht
- Department of Psychiatry and Psychotherapy, Psychiatry Neuroimaging Branch (PNB), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
44
|
Harda Z, Misiołek K, Klimczak M, Chrószcz M, Rodriguez Parkitna J. C57BL/6N mice show a sub-strain specific resistance to the psychotomimetic effects of ketamine. Front Behav Neurosci 2022; 16:1057319. [PMID: 36505728 PMCID: PMC9731130 DOI: 10.3389/fnbeh.2022.1057319] [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: 09/29/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022] Open
Abstract
Repeated administration of subanesthetic doses of ketamine is a model of psychosis-like state in rodents. In mice, this treatment produces a range of behavioral deficits, including impairment in social interactions and locomotion. To date, these phenotypes were described primarily in the Swiss and C3H/HeHsd mouse strains. A few studies investigated ketamine-induced behaviors in the C57BL/6J strain, but to our knowledge the C57BL/6N strain was not investigated thus far. This is surprising, as both C57BL/6 sub-strains are widely used in behavioral and neuropsychopharmacological research, and are de facto standards for characterization of drug effects. The goal of this study was to determine if C57BL/6N mice are vulnerable to develop social deficits after 5 days withdrawal from sub-chronic ketamine treatment (5 days, 30 mg/kg, i.p.), an experimental schedule shown before to cause deficits in social interactions in C57BL/6J mice. Our results show that sub-chronic administration of ketamine that was reported to cause psychotic-like behavior in C57BL/6J mice does not induce appreciable behavioral alterations in C57BL/6N mice. Thus, we show that the effects of sub-chronic ketamine treatment in mice are sub-strain specific.
Collapse
|
45
|
Grieco SF, Castrén E, Knudsen GM, Kwan AC, Olson DE, Zuo Y, Holmes TC, Xu X. Psychedelics and Neural Plasticity: Therapeutic Implications. J Neurosci 2022; 42:8439-8449. [PMID: 36351821 PMCID: PMC9665925 DOI: 10.1523/jneurosci.1121-22.2022] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
Psychedelic drugs have reemerged as tools to treat several brain disorders. Cultural attitudes toward them are changing, and scientists are once again investigating the neural mechanisms through which these drugs impact brain function. The significance of this research direction is reflected by recent work, including work presented by these authors at the 2022 meeting of the Society for Neuroscience. As of 2022, there were hundreds of clinical trials recruiting participants for testing the therapeutic effects of psychedelics. Emerging evidence suggests that psychedelic drugs may exert some of their long-lasting therapeutic effects by inducing structural and functional neural plasticity. Herein, basic and clinical research attempting to elucidate the mechanisms of these compounds is showcased. Topics covered include psychedelic receptor binding sites, effects of psychedelics on gene expression, and on dendrites, and psychedelic effects on microcircuitry and brain-wide circuits. We describe unmet clinical needs and the current state of translation to the clinic for psychedelics, as well as other unanswered basic neuroscience questions addressable with future studies.
Collapse
Affiliation(s)
- Steven F Grieco
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, California 92697
| | - Eero Castrén
- Neuroscience Center-HiLIFE, University of Helsinki, Helsinki, Finland 00014
| | - Gitte M Knudsen
- Neurobiology Research Unit, Copenhagen University Hospital Rigshospitalet and Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark 2200
| | - Alex C Kwan
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853
| | - David E Olson
- Department of Chemistry, University of California-Davis, Davis, California 95616
- Department of Biochemistry & Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, California 95817
- Center for Neuroscience, University of California-Davis, Davis, California 95618
| | - Yi Zuo
- Department of Molecular, Cell and Developmental Biology, University of California-Santa Cruz, Santa Cruz, California 95064
| | - Todd C Holmes
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California 92697
- Center for Neural Circuit Mapping, University of California-Irvine, Irvine, California 92697
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, California 92697
- Center for Neural Circuit Mapping, University of California-Irvine, Irvine, California 92697
| |
Collapse
|
46
|
Rajpal H, Mediano PAM, Rosas FE, Timmermann CB, Brugger S, Muthukumaraswamy S, Seth AK, Bor D, Carhart-Harris RL, Jensen HJ. Psychedelics and schizophrenia: Distinct alterations to Bayesian inference. Neuroimage 2022; 263:119624. [PMID: 36108798 PMCID: PMC7614773 DOI: 10.1016/j.neuroimage.2022.119624] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/11/2022] [Accepted: 09/10/2022] [Indexed: 11/28/2022] Open
Abstract
Schizophrenia and states induced by certain psychotomimetic drugs may share some physiological and phenomenological properties, but they differ in fundamental ways: one is a crippling chronic mental disease, while the others are temporary, pharmacologically-induced states presently being explored as treatments for mental illnesses. Building towards a deeper understanding of these different alterations of normal consciousness, here we compare the changes in neural dynamics induced by LSD and ketamine (in healthy volunteers) against those associated with schizophrenia, as observed in resting-state M/EEG recordings. While both conditions exhibit increased neural signal diversity, our findings reveal that this is accompanied by an increased transfer entropy from the front to the back of the brain in schizophrenia, versus an overall reduction under the two drugs. Furthermore, we show that these effects can be reproduced via different alterations of standard Bayesian inference applied on a computational model based on the predictive processing framework. In particular, the effects observed under the drugs are modelled as a reduction of the precision of the priors, while the effects of schizophrenia correspond to an increased precision of sensory information. These findings shed new light on the similarities and differences between schizophrenia and two psychotomimetic drug states, and have potential implications for the study of consciousness and future mental health treatments.
Collapse
Affiliation(s)
- Hardik Rajpal
- Centre for Complexity Science, Imperial College London, South Kensington, London, United Kingdom; Department of Mathematics, Imperial College London, South Kensington, London, United Kingdom; Public Policy Program, The Alan Turing Institute, London, United Kingdom.
| | - Pedro A M Mediano
- Department of Computing, Imperial College London, South Kensington, London, United Kingdom; Department of Psychology, University of Cambridge, Cambridge, United Kingdom; Department of Psychology, Queen Mary University of London, London, United Kingdom.
| | - Fernando E Rosas
- Centre for Complexity Science, Imperial College London, South Kensington, London, United Kingdom; Centre for Psychedelic Research, Department of Brain Sciences, Imperial College London, London, United Kingdom; Data Science Institute, Imperial College London, London, United Kingdom; Department of Informatics, University of Sussex, Brighton, United Kingdom
| | - Christopher B Timmermann
- Centre for Psychedelic Research, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Stefan Brugger
- Cardiff University Brain Research Imaging Centre, School of Psychology, Cardiff University, United Kingdom; Centre for Academic Mental Health, Bristol Medical School, University of Bristol, United Kingdom
| | | | - Anil K Seth
- School of Engineering and Informatics, University of Sussex, United Kingdom; CIFAR Program on Brain, Mind, and Consciousness, Toronto, Canada
| | - Daniel Bor
- Department of Psychology, University of Cambridge, Cambridge, United Kingdom; Department of Psychology, Queen Mary University of London, London, United Kingdom
| | - Robin L Carhart-Harris
- Centre for Psychedelic Research, Department of Brain Sciences, Imperial College London, London, United Kingdom; Psychedelics Division, Neuroscape, Department of Neurology, University of California San Francisco, US
| | - Henrik J Jensen
- Centre for Complexity Science, Imperial College London, South Kensington, London, United Kingdom; Department of Mathematics, Imperial College London, South Kensington, London, United Kingdom; Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| |
Collapse
|
47
|
Limoges A, Yarur HE, Tejeda HA. Dynorphin/kappa opioid receptor system regulation on amygdaloid circuitry: Implications for neuropsychiatric disorders. Front Syst Neurosci 2022; 16:963691. [PMID: 36276608 PMCID: PMC9579273 DOI: 10.3389/fnsys.2022.963691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Amygdaloid circuits are involved in a variety of emotional and motivation-related behaviors and are impacted by stress. The amygdala expresses several neuromodulatory systems, including opioid peptides and their receptors. The Dynorphin (Dyn)/kappa opioid receptor (KOR) system has been implicated in the processing of emotional and stress-related information and is expressed in brain areas involved in stress and motivation. Dysregulation of the Dyn/KOR system has also been implicated in various neuropsychiatric disorders. However, there is limited information about the role of the Dyn/KOR system in regulating amygdala circuitry. Here, we review the literature on the (1) basic anatomy of the amygdala, (2) functional regulation of synaptic transmission by the Dyn/KOR system, (3) anatomical architecture and function of the Dyn/KOR system in the amygdala, (4) regulation of amygdala-dependent behaviors by the Dyn/KOR system, and (5) future directions for the field. Future work investigating how the Dyn/KOR system shapes a wide range of amygdala-related behaviors will be required to increase our understanding of underlying circuitry modulation by the Dyn/KOR system. We anticipate that continued focus on the amygdala Dyn/KOR system will also elucidate novel ways to target the Dyn/KOR system to treat neuropsychiatric disorders.
Collapse
Affiliation(s)
- Aaron Limoges
- Unit on Neuromodulation and Synaptic Integration, Bethesda, MD, United States
- NIH-Columbia University Individual Graduate Partnership Program, National Institutes of Health, Bethesda, MD, United States
- Department of Biological Sciences, Columbia University, New York, NY, United States
| | - Hector E. Yarur
- Unit on Neuromodulation and Synaptic Integration, Bethesda, MD, United States
| | - Hugo A. Tejeda
- Unit on Neuromodulation and Synaptic Integration, Bethesda, MD, United States
- *Correspondence: Hugo A. Tejeda,
| |
Collapse
|
48
|
Afrin S, Hossain A, Begum S. Effects of Moringa oleifera on working memory: an experimental study with memory-impaired Wistar rats tested in radial arm maze. BMC Res Notes 2022; 15:314. [PMID: 36192762 PMCID: PMC9528094 DOI: 10.1186/s13104-022-06219-5] [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: 04/27/2022] [Accepted: 09/21/2022] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Memory impairment is a serious problem that has a significant negative impact on survival and quality of life. When used for a long time, drugs used to treat memory loss become less effective and have more side effects, making therapy more difficult. Different medicinal plants are now being highlighted because of their valuable applications and low risk of adverse effects. Moringa oleifera is one of these plants that has gained much attention due to its diverse biological functions. The study aimed to determine the effects of Moringa oleifera on working memory in memory-impaired Wistar rats. RESULTS For this experimental study, 30 male Wistar rats having 150-250 g bodyweight were divided equally into three groups: Group-I/normal memory group (treated with oral normal saline 5 ml/kg body weight), Group-II/memory-impaired group (induced by intraperitoneal ketamine 15 mg/kg body weight), and Group-III/experimental group (treated with oral Moringa oleifera 200 mg/kg bodyweight and intraperitoneal ketamine 15 mg/kg body weight). The experimental group showed significantly fewer working memory errors than the memory-impaired group. The experimental group also provides the lowest variability of WMEs among groups. Thus, the study concludes that M. oleifera can prevent ketamine-induced memory impairment in Wistar rats.
Collapse
Affiliation(s)
- Sadia Afrin
- Department of Physiology, Marks Medical College and Hospital, Dhaka, Bangladesh
| | - Ahmed Hossain
- College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates. .,Department of Public Health, North South University, Dhaka, Bangladesh.
| | - Shelina Begum
- Department of Physiology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| |
Collapse
|
49
|
Cui K, Yu Z, Xu L, Jiang W, Wang L, Wang X, Zou D, Gu J, Gao F, Zhang X, Wang Z. Behavioral features and disorganization of oscillatory activity in C57BL/6J mice after acute low dose MK-801 administration. Front Neurosci 2022; 16:1001869. [PMID: 36188453 PMCID: PMC9515662 DOI: 10.3389/fnins.2022.1001869] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
Low dose acute administration of N-methyl-D-aspartate receptor (NMDAR) antagonist MK-801 is widely used to model cognition impairments associated with schizophrenia (CIAS) in rodents. However, due to no unified standards for animal strain, dose, route of drug delivery, and the duration of administration, how different doses of MK-801 influence behavior and fundamental frequency bands of the local field potential (LFP) in cortical and subcortical brain regions without consistent conclusions. The optimal dose of MK-801 as a valid cognition impairers to model CIAS in C57BL/6J mice remains unclear. The current study characterizes the behavior and neural oscillation alterations induced by different low doses of MK-801 in medial prefrontal cortex (mPFC) and hippocampus CA1 of C57BL/6J mice. The results reveal that mice treated with 0.1 and 0.3 mg/kg MK-801 demonstrate increased locomotion and diminished prepulse inhibition (PPI), while not when treated with 0.05 mg/kg MK-801. We also find that MK-801 dose as low as 0.05 mg/kg can significantly diminishes spontaneous alteration during the Y-maze test. Additionally, the oscillation power in delta, theta, alpha, gamma and HFO bands of the LFP in mPFC and CA1 was potentiated by different dose levels of MK-801 administration. The current findings revealed that the observed sensitivity against spontaneous alteration impairment and neural oscillation at 0.05 mg/kg MK-801 suggest that 0.05 mg/kg will produce changes in CIAS-relevant behavior without overt changes in locomotion and sensorimotor processing in C57BL/6J mice.
Collapse
Affiliation(s)
- Keke Cui
- Department of Pharmacology, Ningbo University School of Medicine, Ningbo, China
- Key Laboratory of Addiction Research of Zhejiang Province, Kang Ning Hospital, Ningbo, China
| | - Zhipeng Yu
- Department of Pharmacology, Ningbo University School of Medicine, Ningbo, China
| | - Le Xu
- Department of Pharmacology, Ningbo University School of Medicine, Ningbo, China
| | - Wangcong Jiang
- Department of Pharmacology, Ningbo University School of Medicine, Ningbo, China
| | - Luwan Wang
- Department of Pharmacology, Ningbo University School of Medicine, Ningbo, China
| | - Xiangqun Wang
- Department of Pharmacology, Ningbo University School of Medicine, Ningbo, China
| | - Dandan Zou
- Department of Pharmacology, Ningbo University School of Medicine, Ningbo, China
| | - Jiajie Gu
- The Affiliated People’s Hospital of Ningbo University, Ningbo, China
| | - Feng Gao
- The Affiliated People’s Hospital of Ningbo University, Ningbo, China
| | - Xiaoqing Zhang
- Department of Pharmacology, Ningbo University School of Medicine, Ningbo, China
- The Affiliated People’s Hospital of Ningbo University, Ningbo, China
- Key Laboratory of Addiction Research of Zhejiang Province, Kang Ning Hospital, Ningbo, China
| | - Zhengchun Wang
- Department of Pharmacology, Ningbo University School of Medicine, Ningbo, China
- The Affiliated People’s Hospital of Ningbo University, Ningbo, China
- Key Laboratory of Addiction Research of Zhejiang Province, Kang Ning Hospital, Ningbo, China
- *Correspondence: Zhengchun Wang,
| |
Collapse
|
50
|
Wu Q, Tang J, Qi C, Xie A, Liu J, O’Neill J, Liu T, Hao W, Liao Y. Higher glutamatergic activity in the medial prefrontal cortex in chronic ketamine users. J Psychiatry Neurosci 2022; 47:E263-E271. [PMID: 35882477 PMCID: PMC9343127 DOI: 10.1503/jpn.210179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 02/13/2022] [Accepted: 04/13/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The medial prefrontal cortex (mPFC) plays an important role in depression and addiction. Previous studies have shown alterations in glutamatergic activity in the mPFC following the administration of ketamine in patients with depression and healthy controls. However, it remains unclear whether chronic, nonmedical use of ketamine affects metabolites in the mPFC. METHODS Using proton magnetic resonance spectroscopy, we measured metabolites (glutamate and glutamine [Glx]; phosphocreatine and creatine [PCr+Cr]; myo-inositol; N-acetyl-aspartate; and glycerophosphocholine and phosphocholine [GPC+PC]) in the mPFC of chronic ketamine users (n = 20) and healthy controls (n = 43). Among ketamine users, 60% consumed ketamine once per day or more, 10% consumed it every 2 days and 30% consumed it every 3 or more days. Using analysis of covariance, we evaluated between-group differences in the ratios of Glx:PCr+Cr, myo-inositol:PCr+Cr, N-acetyl-aspartate:PCr+Cr and GPC+PC:PCr+Cr. RESULTS Chronic ketamine users showed significantly higher Glx:PCr+Cr ratios than healthy controls (median 1.05 v. 0.95, p = 0.008). We found no significant differences in myoinositol:PCr+Cr, N-acetyl-aspartate:PCr+Cr or GPC+PC:PCr+Cr ratios between the 2 groups. We found a positive relationship between N-acetyl-aspartate:PCr+Cr and Glx:PCr+Cr ratios in the healthy control group (R = 0.345, p = 0.023), but the ketamine use group failed to show such an association (ρ = 0.197, p = 0.40). LIMITATIONS The cross-sectional design of this study did not permit causal inferences related to higher Glx:PCr+Cr ratios and chronic ketamine use. CONCLUSION This study provides the first evidence that chronic ketamine users have higher glutamatergic activity in the mPFC than healthy controls; this finding may provide new insights relevant to the treatment of depression with ketamine.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yanhui Liao
- From the Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China (Wu, Tang, Liao); the Department of Psychiatry and National Clinical Research Centre for Mental Disorders, Second Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China (Wu, T. Liu, Hao); the Department of Psychiatry, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, P.R. China (Qi); the Department of Radiology, Hunan Provincial People's Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, P.R. China (Xie, J. Liu); the Division of Child and Adolescent Psychiatry, UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, Calif., USA (O'Neill)
| |
Collapse
|