1
|
Ali I, Jupp B, Hudson MR, Major B, Silva J, Yamakawa GR, Casillas-Espinosa PM, Braine E, Thergarajan P, Haskali MB, Vivash L, Brkljaca R, Shultz SR, Kwan P, Fukushima K, Sachdev P, Cheng JY, Mychasiuk R, Jones NC, Wright DK, OBrien TJ. In vivo biomarkers of GABAergic function in epileptic rats treated with the GAT-1 inhibitor E2730. Epilepsia 2024; 65:3376-3390. [PMID: 39302665 DOI: 10.1111/epi.18119] [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: 05/21/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/22/2024]
Abstract
OBJECTIVE E2730, an uncompetitive γ-aminobutyric acid (GABA) transporter-1 (GAT-1) inhibitor, has potent anti-seizure effects in a rodent model of chronic temporal lobe epilepsy, the kainic acid status epilepticus (KASE) rat model. In this study, we examined purported neuroimaging and physiological surrogate biomarkers of the effect of E2730 on brain GABAergic function. METHODS We conducted a randomized cross-over study, incorporating 1-week treatments with E2730 (100 mg/kg/day subcutaneous infusion) or vehicle in epileptic post-KASE rats. KASE rats underwent serial 9.4 T magnetic resonance spectroscopy (MRS) measuring GABA and other brain metabolites, [18F]Flumazenil positron emission tomography (PET) quantifying GABAA receptor availability, quantitative electroencephalography (qEEG) and transcranial magnetic stimulation (TMS)-mediated motor activity, as well as continuous video-EEG recording to measure spontaneous seizures during each treatment. Age-matched, healthy control animals treated with E2730 or vehicle were also studied. RESULTS E2730 treatment significantly reduced spontaneous seizures, with 8 of 11 animals becoming seizure-free. MRS revealed that E2730-treated animals had significantly reduced taurine levels. [18F]Flumazenil PET imaging revealed no changes in GABA receptor affinity or density during E2730 treatment. The power of gamma frequency oscillations in the EEG was decreased significantly in the auditory cortex and hippocampus of KASE and control rats during E2730 treatment. Auditory evoked gamma frequency power was enhanced by E2730 treatment in the auditory cortex of KASE and healthy controls, but only in the hippocampus of KASE rats. E2730 did not influence motor evoked potentials triggered by TMS. SIGNIFICANCE This study identified clinically relevant changes in multimodality imaging and functional purported biomarkers of GABAergic activity during E2730 treatment in epileptic and healthy control animals. These biomarkers could be utilized in clinical trials of E2730 and potentially other GABAergic drugs to provide surrogate endpoints, thereby reducing the cost of such trials.
Collapse
Affiliation(s)
- Idrish Ali
- The Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Bianca Jupp
- The Department of Neuroscience, Monash University, Melbourne, Australia
| | - Matthew R Hudson
- The Department of Neuroscience, Monash University, Melbourne, Australia
| | - Brendan Major
- The Department of Neuroscience, Monash University, Melbourne, Australia
| | - Juliana Silva
- The Department of Neuroscience, Monash University, Melbourne, Australia
| | - Glenn R Yamakawa
- The Department of Neuroscience, Monash University, Melbourne, Australia
| | - Pablo M Casillas-Espinosa
- The Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Emma Braine
- The Department of Neuroscience, Monash University, Melbourne, Australia
| | | | - Mohammad B Haskali
- Department of Radiopharmaceutical Sciences, Cancer Imaging, The Peter MacCallum Cancer Centre, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia
| | - Lucy Vivash
- The Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | | | - Sandy R Shultz
- The Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
- Centre for Trauma and Mental Health Research, Vancouver Island University, Nanaimo, Canada
| | - Patrick Kwan
- The Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | | | - Pallavi Sachdev
- Clinical Evidence Generation, Translational Sciences, Eisai Inc., Bunkyo, Japan
| | - Jocelyn Y Cheng
- Clinical Evidence Generation, Translational Sciences, Eisai Inc., Bunkyo, Japan
| | | | - Nigel C Jones
- The Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - David K Wright
- The Department of Neuroscience, Monash University, Melbourne, Australia
| | - Terence J OBrien
- The Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| |
Collapse
|
2
|
Vinnakota C, Hudson MR, Jones NC, Sundram S, Hill RA. Potential Roles for the GluN2D NMDA Receptor Subunit in Schizophrenia. Int J Mol Sci 2023; 24:11835. [PMID: 37511595 PMCID: PMC10380280 DOI: 10.3390/ijms241411835] [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: 05/12/2023] [Revised: 07/19/2023] [Accepted: 07/22/2023] [Indexed: 07/30/2023] Open
Abstract
Glutamate N-methyl-D-aspartate receptor (NMDAR) hypofunction has been proposed to underlie schizophrenia symptoms. This theory arose from the observation that administration of NMDAR antagonists, which are compounds that inhibit NMDAR activity, reproduces behavioural and molecular schizophrenia-like phenotypes, including hallucinations, delusions and cognitive impairments in healthy humans and animal models. However, the role of specific NMDAR subunits in these schizophrenia-relevant phenotypes is largely unknown. Mounting evidence implicates the GluN2D subunit of NMDAR in some of these symptoms and pathology. Firstly, genetic and post-mortem studies show changes in the GluN2D subunit in people with schizophrenia. Secondly, the psychosis-inducing effects of NMDAR antagonists are blunted in GluN2D-knockout mice, suggesting that the GluN2D subunit mediates NMDAR-antagonist-induced psychotomimetic effects. Thirdly, in the mature brain, the GluN2D subunit is relatively enriched in parvalbumin (PV)-containing interneurons, a cell type hypothesized to underlie the cognitive symptoms of schizophrenia. Lastly, the GluN2D subunit is widely and abundantly expressed early in development, which could be of importance considering schizophrenia is a disorder that has its origins in early neurodevelopment. The limitations of currently available therapies warrant further research into novel therapeutic targets such as the GluN2D subunit, which may help us better understand underlying disease mechanisms and develop novel and more effective treatment options.
Collapse
Affiliation(s)
- Chitra Vinnakota
- Department of Psychiatry, School of Clinical Sciences, Faculty of Medical, Nursing and Health Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Matthew R Hudson
- Department of Neuroscience, Faculty of Medical, Nursing and Health Sciences, Monash University, Melbourne, VIC 3004, Australia
| | - Nigel C Jones
- Department of Neuroscience, Faculty of Medical, Nursing and Health Sciences, Monash University, Melbourne, VIC 3004, Australia
| | - Suresh Sundram
- Department of Psychiatry, School of Clinical Sciences, Faculty of Medical, Nursing and Health Sciences, Monash University, Clayton, VIC 3168, Australia
- Mental Health Program, Monash Health, Clayton, VIC 3168, Australia
| | - Rachel A Hill
- Department of Psychiatry, School of Clinical Sciences, Faculty of Medical, Nursing and Health Sciences, Monash University, Clayton, VIC 3168, Australia
| |
Collapse
|
3
|
de Bartolomeis A, Vellucci L, Barone A, Manchia M, De Luca V, Iasevoli F, Correll CU. Clozapine's multiple cellular mechanisms: What do we know after more than fifty years? A systematic review and critical assessment of translational mechanisms relevant for innovative strategies in treatment-resistant schizophrenia. Pharmacol Ther 2022; 236:108236. [PMID: 35764175 DOI: 10.1016/j.pharmthera.2022.108236] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 12/21/2022]
Abstract
Almost fifty years after its first introduction into clinical care, clozapine remains the only evidence-based pharmacological option for treatment-resistant schizophrenia (TRS), which affects approximately 30% of patients with schizophrenia. Despite the long-time experience with clozapine, the specific mechanism of action (MOA) responsible for its superior efficacy among antipsychotics is still elusive, both at the receptor and intracellular signaling level. This systematic review is aimed at critically assessing the role and specific relevance of clozapine's multimodal actions, dissecting those mechanisms that under a translational perspective could shed light on molecular targets worth to be considered for further innovative antipsychotic development. In vivo and in vitro preclinical findings, supported by innovative techniques and methods, together with pharmacogenomic and in vivo functional studies, point to multiple and possibly overlapping MOAs. To better explore this crucial issue, the specific affinity for 5-HT2R, D1R, α2c, and muscarinic receptors, the relatively low occupancy at dopamine D2R, the interaction with receptor dimers, as well as the potential confounder effects resulting in biased ligand action, and lastly, the role of the moiety responsible for lipophilic and alkaline features of clozapine are highlighted. Finally, the role of transcription and protein changes at the synaptic level, and the possibility that clozapine can directly impact synaptic architecture are addressed. Although clozapine's exact MOAs that contribute to its unique efficacy and some of its severe adverse effects have not been fully understood, relevant information can be gleaned from recent mechanistic understandings that may help design much needed additional therapeutic strategies for TRS.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment Resistant Psychosis, Department of Neuroscience, Reproductive Science and Dentistry, University Medical School of Naples "Federico II", Naples, Italy.
| | - Licia Vellucci
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment Resistant Psychosis, Department of Neuroscience, Reproductive Science and Dentistry, University Medical School of Naples "Federico II", Naples, Italy
| | - Annarita Barone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment Resistant Psychosis, Department of Neuroscience, Reproductive Science and Dentistry, University Medical School of Naples "Federico II", Naples, Italy
| | - Mirko Manchia
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy; Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | - Felice Iasevoli
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment Resistant Psychosis, Department of Neuroscience, Reproductive Science and Dentistry, University Medical School of Naples "Federico II", Naples, Italy
| | - Christoph U Correll
- The Zucker Hillside Hospital, Department of Psychiatry, Northwell Health, Glen Oaks, NY, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Department of Psychiatry and Molecular Medicine, Hempstead, NY, USA; Charité Universitätsmedizin Berlin, Department of Child and Adolescent Psychiatry, Berlin, Germany
| |
Collapse
|
4
|
Present and future antipsychotic drugs: a systematic review of the putative mechanisms of action for efficacy and a critical appraisal under a translational perspective. Pharmacol Res 2022; 176:106078. [PMID: 35026403 DOI: 10.1016/j.phrs.2022.106078] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/23/2021] [Accepted: 01/07/2022] [Indexed: 01/10/2023]
Abstract
Antipsychotics represent the mainstay of schizophrenia pharmacological therapy, and their role has been expanded in the last years to mood disorders treatment. Although introduced in 1952, many years of research were required before an accurate picture of how antipsychotics work began to emerge. Despite the well-recognized characterization of antipsychotics in typical and atypical based on their liability to induce motor adverse events, their main action at dopamine D2R to elicit the "anti-psychotic" effect, as well as the multimodal action at other classes of receptors, their effects on intracellular mechanisms starting with receptor occupancy is still not completely understood. Significant lines of evidence converge on the impact of these compounds on multiple molecular signaling pathways implicated in the regulation of early genes and growth factors, dendritic spine shape, brain inflammation, and immune response, tuning overall the function and architecture of the synapse. Here we present, based on PRISMA approach, a comprehensive and systematic review of the above mechanisms under a translational perspective to disentangle those intracellular actions and signaling that may underline clinically relevant effects and represent potential targets for further innovative strategies in antipsychotic therapy.
Collapse
|
5
|
Speers LJ, Bilkey DK. Disorganization of Oscillatory Activity in Animal Models of Schizophrenia. Front Neural Circuits 2021; 15:741767. [PMID: 34675780 PMCID: PMC8523827 DOI: 10.3389/fncir.2021.741767] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/16/2021] [Indexed: 01/02/2023] Open
Abstract
Schizophrenia is a chronic, debilitating disorder with diverse symptomatology, including disorganized cognition and behavior. Despite considerable research effort, we have only a limited understanding of the underlying brain dysfunction. In this article, we review the potential role of oscillatory circuits in the disorder with a particular focus on the hippocampus, a region that encodes sequential information across time and space, as well as the frontal cortex. Several mechanistic explanations of schizophrenia propose that a loss of oscillatory synchrony between and within these brain regions may underlie some of the symptoms of the disorder. We describe how these oscillations are affected in several animal models of schizophrenia, including models of genetic risk, maternal immune activation (MIA) models, and models of NMDA receptor hypofunction. We then critically discuss the evidence for disorganized oscillatory activity in these models, with a focus on gamma, sharp wave ripple, and theta activity, including the role of cross-frequency coupling as a synchronizing mechanism. Finally, we focus on phase precession, which is an oscillatory phenomenon whereby individual hippocampal place cells systematically advance their firing phase against the background theta oscillation. Phase precession is important because it allows sequential experience to be compressed into a single 120 ms theta cycle (known as a 'theta sequence'). This time window is appropriate for the induction of synaptic plasticity. We describe how disruption of phase precession could disorganize sequential processing, and thereby disrupt the ordered storage of information. A similar dysfunction in schizophrenia may contribute to cognitive symptoms, including deficits in episodic memory, working memory, and future planning.
Collapse
Affiliation(s)
| | - David K. Bilkey
- Department of Psychology, Otago University, Dunedin, New Zealand
| |
Collapse
|
6
|
Lahogue C, Pinault D. Frontoparietal anodal tDCS reduces ketamine-induced oscillopathies. Transl Neurosci 2021; 12:282-296. [PMID: 34239718 PMCID: PMC8240415 DOI: 10.1515/tnsci-2020-0157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/05/2021] [Accepted: 05/14/2021] [Indexed: 12/26/2022] Open
Abstract
During the prodromal phase of schizophrenia with its complex and insidious clinical picture, electroencephalographic recordings detect widespread oscillation disturbances (or oscillopathies) during the wake-sleep cycle. Neural oscillations are electrobiomarkers of the connectivity state within systems. A single-systemic administration of ketamine, a non-competitive NMDA glutamate receptor antagonist, transiently reproduces the oscillopathies with a clinical picture reminiscent of the psychosis prodrome. This acute pharmacological model may help the research and development of innovative treatments against psychotic transition. Transcranial electrical stimulation is recognized as an appropriate non-invasive therapeutic modality since it can increase cognitive performance and modulate neural oscillations with little or no side effects. Therefore, our objective was to set up, in the sedated adult rat, a stimulation method that is able to normalize ketamine-induced increase in gamma-frequency (30-80 Hz) oscillations and decrease in sigma-frequency (10-17 Hz) oscillations. Unilateral and bipolar frontoparietal (FP), transcranial anodal stimulation by direct current (<+1 mA) was applied in ketamine-treated rats. A concomitant bilateral electroencephalographic recording of the parietal cortex measured the stimulation effects on its spontaneously occurring oscillations. A 5 min FP anodal tDCS immediately and quickly reduced, significantly with an intensity-effect relationship, the ketamine-induced gamma hyperactivity, and sigma hypoactivity at least in the bilateral parietal cortex. A duration effect was also recorded. The tDCS also tended to diminish the ketamine-induced delta hypoactivity. These preliminary neurophysiological findings are promising for developing a therapeutic proof-of-concept against neuropsychiatric disorders.
Collapse
Affiliation(s)
- Caroline Lahogue
- Université de Strasbourg, Strasbourg, France
- INSERM U1114, Neuropsychologie Cognitive et Physiopathologie de la Schizophrénie, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Centre de Recherche en Biomédecine de Strasbourg (CRBS), Faculté de médecine, Strasbourg, France
| | - Didier Pinault
- Université de Strasbourg, Strasbourg, France
- INSERM U1114, Neuropsychologie Cognitive et Physiopathologie de la Schizophrénie, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Centre de Recherche en Biomédecine de Strasbourg (CRBS), Faculté de médecine, Strasbourg, France
| |
Collapse
|
7
|
Thériault RK, St-Denis M, Hewitt T, Khokhar JY, Lalonde J, Perreault ML. Sex-Specific Cannabidiol- and Iloperidone-Induced Neuronal Activity Changes in an In Vitro MAM Model System of Schizophrenia. Int J Mol Sci 2021; 22:ijms22115511. [PMID: 34073710 PMCID: PMC8197248 DOI: 10.3390/ijms22115511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/20/2022] Open
Abstract
Cortical circuit dysfunction is thought to be an underlying mechanism of schizophrenia (SZ) pathophysiology with normalization of aberrant circuit activity proposed as a biomarker for antipsychotic efficacy. Cannabidiol (CBD) shows potential as an adjunctive antipsychotic therapy; however, potential sex effects in these drug interactions remain unknown. In the present study, we sought to elucidate sex effects of CBD coadministration with the atypical antipsychotic iloperidone (ILO) on the activity of primary cortical neuron cultures derived from the rat methylazoxymethanol acetate (MAM) model used for the study of SZ. Spontaneous network activity measurements were obtained using a multielectrode array at baseline and following administration of CBD or ILO alone, or combined. At baseline, MAM male neurons displayed increased bursting activity whereas MAM female neurons exhibited no difference in bursting activity compared to sex-matched controls. CBD administered alone showed a rapid but transient increase in neuronal activity in the MAM networks, an effect more pronounced in females. Furthermore, ILO had an additive effect on CBD-induced elevations in activity in the MAM male neurons. In the MAM female neurons, CBD or ILO administration resulted in time-dependent elevations in neuronal activity, but the short-term CBD-induced increases in activity were lost when CBD and ILO were combined. Our findings indicate that CBD induces rapid increases in cortical neuronal activity, with sex-specific drug interactions upon ILO coadministration. This suggests that sex should be a consideration when implementing adjunct therapy for treatment of SZ.
Collapse
Affiliation(s)
- Rachel-Karson Thériault
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada; (R.-K.T.); (M.S.-D.); (T.H.); (J.L.)
- Collaborative Program in Neuroscience, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - Myles St-Denis
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada; (R.-K.T.); (M.S.-D.); (T.H.); (J.L.)
| | - Tristen Hewitt
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada; (R.-K.T.); (M.S.-D.); (T.H.); (J.L.)
- Collaborative Program in Neuroscience, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - Jibran Y. Khokhar
- Collaborative Program in Neuroscience, University of Guelph, Guelph, ON N1G 2W1, Canada;
- Department of Biomedical Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Jasmin Lalonde
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada; (R.-K.T.); (M.S.-D.); (T.H.); (J.L.)
- Collaborative Program in Neuroscience, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - Melissa L. Perreault
- Collaborative Program in Neuroscience, University of Guelph, Guelph, ON N1G 2W1, Canada;
- Department of Biomedical Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
- Correspondence: ; Tel.: +1-(519)-824-4120 (ext. 52013)
| |
Collapse
|
8
|
Irwin MN, VandenBerg A. Retracing our steps to understand ketamine in depression: A focused review of hypothesized mechanisms of action. Ment Health Clin 2021; 11:200-210. [PMID: 34026396 PMCID: PMC8120982 DOI: 10.9740/mhc.2021.05.200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Introduction MDD represents a significant burden worldwide, and while a number of approved treatments exist, there are high rates of treatment resistance and refractoriness. Ketamine, an N-methyl-d-aspartate receptor (NMDAR) antagonist, is a novel, rapid-acting antidepressant, however the mechanisms underlying the efficacy of ketamine are not well understood and many other mechanisms outside of NMDAR antagonism have been postulated based on preclinical data. This focused review aims to present a summary of the proposed mechanisms of action by which ketamine functions in depressive disorders supported by preclinical data and clinical studies in humans. Methods A literature search was completed using the PubMed and Google Scholar databases. Results were limited to clinical trials and case studies in humans that were published in English. The findings were used to compile this article. Results The antidepressant effects associated with ketamine are mediated via a complex interplay of mechanisms; key steps include NMDAR blockade on γ-aminobutyric acid interneurons, glutamate surge, and subsequent activation and upregulation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor. Discussion Coadministration of ketamine for MDD with other psychotropic agents, for example benzodiazepines, may attenuate antidepressant effects. Limited evidence exists for these effects and should be evaluated on a case-by-case basis.
Collapse
Affiliation(s)
- Madison N Irwin
- Clinical Pharmacist Specialist in Psychology and Neurology, Department of Pharmacy, Michigan Medicine, Ann Arbor, Michigan
| | - Amy VandenBerg
- Clinical Pharmacist Specialist in Psychology and Neurology, Department of Pharmacy, Michigan Medicine, Ann Arbor, Michigan
| |
Collapse
|
9
|
Gregory KJ, Goudet C. International Union of Basic and Clinical Pharmacology. CXI. Pharmacology, Signaling, and Physiology of Metabotropic Glutamate Receptors. Pharmacol Rev 2020; 73:521-569. [PMID: 33361406 DOI: 10.1124/pr.119.019133] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Metabotropic glutamate (mGlu) receptors respond to glutamate, the major excitatory neurotransmitter in the mammalian brain, mediating a modulatory role that is critical for higher-order brain functions such as learning and memory. Since the first mGlu receptor was cloned in 1992, eight subtypes have been identified along with many isoforms and splice variants. The mGlu receptors are transmembrane-spanning proteins belonging to the class C G protein-coupled receptor family and represent attractive targets for a multitude of central nervous system disorders. Concerted drug discovery efforts over the past three decades have yielded a wealth of pharmacological tools including subtype-selective agents that competitively block or mimic the actions of glutamate or act allosterically via distinct sites to enhance or inhibit receptor activity. Herein, we review the physiologic and pathophysiological roles for individual mGlu receptor subtypes including the pleiotropic nature of intracellular signal transduction arising from each. We provide a comprehensive analysis of the in vitro and in vivo pharmacological properties of prototypical and commercially available orthosteric agonists and antagonists as well as allosteric modulators, including ligands that have entered clinical trials. Finally, we highlight emerging areas of research that hold promise to facilitate rational design of highly selective mGlu receptor-targeting therapeutics in the future. SIGNIFICANCE STATEMENT: The metabotropic glutamate receptors are attractive therapeutic targets for a range of psychiatric and neurological disorders. Over the past three decades, intense discovery efforts have yielded diverse pharmacological tools acting either competitively or allosterically, which have enabled dissection of fundamental biological process modulated by metabotropic glutamate receptors and established proof of concept for many therapeutic indications. We review metabotropic glutamate receptor molecular pharmacology and highlight emerging areas that are offering new avenues to selectively modulate neurotransmission.
Collapse
Affiliation(s)
- Karen J Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.) and Institut de Génomique Fonctionnelle (IGF), University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Sante et de la Recherche Medicale (INSERM), Montpellier, France (C.G.)
| | - Cyril Goudet
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.) and Institut de Génomique Fonctionnelle (IGF), University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Sante et de la Recherche Medicale (INSERM), Montpellier, France (C.G.)
| |
Collapse
|
10
|
Nasal respiration is necessary for ketamine-dependent high frequency network oscillations and behavioral hyperactivity in rats. Sci Rep 2020; 10:18981. [PMID: 33149202 PMCID: PMC7642442 DOI: 10.1038/s41598-020-75641-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/05/2020] [Indexed: 12/12/2022] Open
Abstract
Changes in oscillatory activity are widely reported after subanesthetic ketamine, however their mechanisms of generation are unclear. Here, we tested the hypothesis that nasal respiration underlies the emergence of high-frequency oscillations (130–180 Hz, HFO) and behavioral activation after ketamine in freely moving rats. We found ketamine 20 mg/kg provoked “fast” theta sniffing in rodents which correlated with increased locomotor activity and HFO power in the OB. Bursts of ketamine-dependent HFO were coupled to “fast” theta frequency sniffing. Theta coupling of HFO bursts were also found in the prefrontal cortex and ventral striatum which, although of smaller amplitude, were coherent with OB activity. Haloperidol 1 mg/kg pretreatment prevented ketamine-dependent increases in fast sniffing and instead HFO coupling to slower basal respiration. Consistent with ketamine-dependent HFO being driven by nasal respiration, unilateral naris blockade led to an ipsilateral reduction in ketamine-dependent HFO power compared to the control side. Bilateral nares blockade reduced ketamine-induced hyperactivity and HFO power and frequency. These findings suggest that nasal airflow entrains ketamine-dependent HFO in diverse brain regions, and that the OB plays an important role in the broadcast of this rhythm.
Collapse
|
11
|
Friesner ID, Martinez E, Zhou H, Gould JD, Li A, Chen ZS, Zhang Q, Wang J. Ketamine normalizes high-gamma power in the anterior cingulate cortex in a rat chronic pain model. Mol Brain 2020; 13:129. [PMID: 32967695 PMCID: PMC7513294 DOI: 10.1186/s13041-020-00670-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/14/2020] [Indexed: 11/18/2022] Open
Abstract
Chronic pain alters cortical and subcortical plasticity, causing enhanced sensory and affective responses to peripheral nociceptive inputs. Previous studies have shown that ketamine had the potential to inhibit abnormally amplified affective responses of single neurons by suppressing hyperactivity in the anterior cingulate cortex (ACC). However, the mechanism of this enduring effect has yet to be understood at the network level. In this study, we recorded local field potentials from the ACC of freely moving rats. Animals were injected with complete Freund’s adjuvant (CFA) to induce persistent inflammatory pain. Mechanical stimulations were administered to the hind paw before and after CFA administration. We found a significant increase in the high-gamma band (60–100 Hz) power in response to evoked pain after CFA treatment. Ketamine, however, reduced the high-gamma band power in response to evoked pain in CFA-treated rats. In addition, ketamine had a sustained effect on the high-gamma band power lasting up to five days after a single dose administration. These results demonstrate that ketamine has the potential to alter maladaptive neural responses in the ACC induced by chronic pain.
Collapse
Affiliation(s)
- Isabel D Friesner
- Department of Anesthesiology, Perioperative Care and Pain, New York University School of Medicine, New York, NY, 10016, USA
| | - Erik Martinez
- Department of Anesthesiology, Perioperative Care and Pain, New York University School of Medicine, New York, NY, 10016, USA
| | - Haocheng Zhou
- Department of Anesthesiology, Perioperative Care and Pain, New York University School of Medicine, New York, NY, 10016, USA
| | | | - Anna Li
- Department of Anesthesiology, Perioperative Care and Pain, New York University School of Medicine, New York, NY, 10016, USA
| | - Zhe Sage Chen
- Department of Psychiatry, New York University School of Medicine, New York, NY, 10016, USA.,Department of Neuroscience & Physiology, New York University School of Medicine, New York, NY, 10016, USA.,Neuroscience Institute, New York University School of Medicine, New York, NY, 10016, USA
| | - Qiaosheng Zhang
- Department of Anesthesiology, Perioperative Care and Pain, New York University School of Medicine, New York, NY, 10016, USA.
| | - Jing Wang
- Department of Anesthesiology, Perioperative Care and Pain, New York University School of Medicine, New York, NY, 10016, USA. .,Department of Neuroscience & Physiology, New York University School of Medicine, New York, NY, 10016, USA. .,Neuroscience Institute, New York University School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
12
|
Sokolenko E, Nithianantharajah J, Jones NC. MK-801 impairs working memory on the Trial-Unique Nonmatch-to-Location test in mice, but this is not exclusively mediated by NMDA receptors on PV+ interneurons or forebrain pyramidal cells. Neuropharmacology 2020; 171:108103. [DOI: 10.1016/j.neuropharm.2020.108103] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 03/11/2020] [Accepted: 04/06/2020] [Indexed: 01/13/2023]
|
13
|
Farmer CA, Gilbert JR, Moaddel R, George J, Adeojo L, Lovett J, Nugent AC, Kadriu B, Yuan P, Gould TD, Park LT, Zarate CA. Ketamine metabolites, clinical response, and gamma power in a randomized, placebo-controlled, crossover trial for treatment-resistant major depression. Neuropsychopharmacology 2020; 45:1398-1404. [PMID: 32252062 PMCID: PMC7297997 DOI: 10.1038/s41386-020-0663-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/10/2020] [Accepted: 03/19/2020] [Indexed: 12/27/2022]
Abstract
A single, subanesthetic dose of (R,S)-ketamine (ketamine) exerts rapid and robust antidepressant effects. Several groups previously reported that (2S,6S;2R,6R)-hydroxynorketamine (HNK) had antidepressant effects in rodents, and that (2R,6R)-HNK increased cortical electroencephalographic gamma power. This exploratory study examined the relationship between ketamine metabolites, clinical response, psychotomimetic symptoms, and gamma power changes in 34 individuals (ages 18-65) with treatment-resistant depression (TRD) who received a single ketamine infusion (0.5 mg/kg) over 40 min. Plasma concentrations of ketamine, norketamine, and HNKs were measured at 40, 80, 120, and 230 min and at 1, 2, and 3 days post-infusion. Linear mixed models evaluated ketamine metabolites as mediators of antidepressant and psychotomimetic effects and their relationship to resting-state whole-brain magnetoencephalography (MEG) gamma power 6-9 h post-infusion. Three salient findings emerged. First, ketamine concentration positively predicted distal antidepressant response at Day 11 post-infusion, and an inverse relationship was observed between (2S,6S;2R,6R)-HNK concentration and antidepressant response at 3 and 7 days post-infusion. Norketamine concentration was not associated with antidepressant response. Second, ketamine, norketamine, and (2S,6S;2R,6R)-HNK concentrations at 40 min were positively associated with contemporaneous psychotomimetic symptoms; post-hoc analysis revealed that ketamine was the predominant contributor. Third, increased (2S,6S;2R,6R)-HNK maximum observed concentration (Cmax) was associated with increased MEG gamma power. While contrary to preclinical observations and our a priori hypotheses, these exploratory results replicate those of a recently published study documenting a relationship between higher (2S,6S;2R,6R)-HNK concentrations and weaker antidepressant response in humans and provide further rationale for studying gamma power changes as potential biomarkers of antidepressant response.
Collapse
Affiliation(s)
- Cristan A Farmer
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Jessica R Gilbert
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Ruin Moaddel
- National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Jomy George
- Clinical Pharmacokinetics Research Unit, Pharmacy Department, National Institutes of Health, Bethesda, MD, USA
| | - Lilian Adeojo
- Clinical Pharmacokinetics Research Unit, Pharmacy Department, National Institutes of Health, Bethesda, MD, USA
| | - Jacqueline Lovett
- National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Allison C Nugent
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
- Magnetoencephalography Core Facility, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Bashkim Kadriu
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Peixiong Yuan
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Todd D Gould
- Departments of Psychiatry, Pharmacology, and Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
| | - Lawrence T Park
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Carlos A Zarate
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
14
|
Toader O, von Heimendahl M, Schuelert N, Nissen W, Rosenbrock H. Suppression of Parvalbumin Interneuron Activity in the Prefrontal Cortex Recapitulates Features of Impaired Excitatory/Inhibitory Balance and Sensory Processing in Schizophrenia. Schizophr Bull 2020; 46:981-989. [PMID: 31903492 PMCID: PMC7342098 DOI: 10.1093/schbul/sbz123] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Accumulating evidence supports parvalbumin expressing inhibitory interneuron (PV IN) dysfunction in the prefrontal cortex as a cause for cognitive impairment associated with schizophrenia (CIAS). PV IN decreased activity is suggested to be the culprit for many of the EEG deficits measured in patients, which correlate with deficits in working memory (WM), cognitive flexibility and attention. In the last few decades, CIAS has been recognized as a heavy burden on the quality of life of patients with schizophrenia, but little progress has been made in finding new treatment options. An important limiting factor in this process is the lack of adequate preclinical models and an incomplete understanding of the circuits engaged in cognition. In this study, we back-translated an auditory stimulation protocol regularly used in human EEG studies into mice and combined it with optogenetics to investigate the role of prefrontal cortex PV INs in excitatory/inhibitory balance and cortical processing. We also assessed spatial WM and reversal learning (RL) during inhibition of prefrontal cortex PV INs. We found significant impairments in trial-to-trial reliability, increased basal network activity and increased oscillation power at 20-60 Hz, and a decreased signal-to-noise ratio, but no significant impairments in behavior. These changes reflect some but not all neurophysiological deficits seen in patients with schizophrenia, suggesting that other neuronal populations and possibly brain regions are involved as well. Our work supports and expands previous findings and highlights the versatility of an approach that combines innovative technologies with back-translated tools used in humans.
Collapse
Affiliation(s)
- Oana Toader
- Central Nervous System Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach Riß, Germany,To whom correspondence should be addressed; Central Nervous System Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorferstr. 65, 88397 Biberach Riß, Germany; tel: +49 735154188280, e-mail:
| | - Moritz von Heimendahl
- Central Nervous System Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach Riß, Germany
| | - Niklas Schuelert
- Central Nervous System Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach Riß, Germany
| | - Wiebke Nissen
- Central Nervous System Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach Riß, Germany
| | - Holger Rosenbrock
- Central Nervous System Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach Riß, Germany
| |
Collapse
|
15
|
Jazayeri D, Braine E, McDonald S, Dworkin S, Powell KL, Griggs K, Vajda FJE, O'Brien TJ, Jones NC. A rat model of valproate teratogenicity from chronic oral treatment during pregnancy. Epilepsia 2020; 61:1291-1300. [PMID: 32415786 DOI: 10.1111/epi.16536] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 04/23/2020] [Accepted: 04/23/2020] [Indexed: 01/26/2023]
Abstract
OBJECTIVE Sodium valproate (VPA), the most effective antiepileptic drug for patients with genetic generalized epilepsy (GGE), is a potent human teratogen that increases the risk of a range of congenital malformations, including spina bifida. The mechanisms underlying this teratogenicity are not known, but may involve genetic risk factors. This study aimed to develop an animal model of VPA-induced birth defects. METHODS We used three different rat strains: inbred Genetic Absence Epilepsy Rats From Strasbourg (GAERS), a model of GGE with absence seizures; inbred Non-Epileptic Controls (NEC); and outbred nonepileptic Wistars. Female rats were fed standard chow or VPA (20 g/kg food) mixed in standard chow for 2 weeks prior to conception, and then mated with same-strain males. Treatment continued throughout pregnancy. Fetuses were extracted via C-section on gestational day 21 and examined for birth defects, including external assessment and spinal measurements. RESULTS VPA-exposed pups showed significant reductions in weight, length, and whole-body development compared with controls of all three strains (P < .0001). Gestational VPA treatment altered intravertebral distances, and resulted in underdeveloped vertebral arches between thoracic region T11 and caudal region C2 in most pups (GAERS, 100%; NEC, 95%; Wistar, 80%), more frequently than in controls (9%, 13%, 19%). SIGNIFICANCE Gestational VPA treatment results in similar developmental and morphological abnormalities in three rat strains, including one with GGE, indicating that the genetic underpinnings of epilepsy do not contribute markedly to VPA-induced birth defects. This model may be used in future studies to investigate mechanisms involved in the pathogenesis of antiepileptic drug-induced birth defects.
Collapse
Affiliation(s)
- Dana Jazayeri
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia.,La Trobe Centre for Sport and Exercise Medicine Research, School of Allied Health, Human Services, and Sport, La Trobe University, Bundoora, Victoria, Australia
| | - Emma Braine
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
| | - Stuart McDonald
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia.,Department of Physiology, Anatomy, and Microbiology, School of Life Sciences, La Trobe University, Melbourne, Victoria, Australia
| | - Sebastian Dworkin
- Department of Physiology, Anatomy, and Microbiology, School of Life Sciences, La Trobe University, Melbourne, Victoria, Australia
| | - Kim L Powell
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
| | - Karen Griggs
- Department of Physiology, Anatomy, and Microbiology, School of Life Sciences, La Trobe University, Melbourne, Victoria, Australia
| | - Frank J E Vajda
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
| | - Terence J O'Brien
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
| | - Nigel C Jones
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
16
|
NMDA receptors on parvalbumin-positive interneurons and pyramidal neurons both contribute to MK-801 induced gamma oscillatory disturbances: Complex relationships with behaviour. Neurobiol Dis 2020; 134:104625. [DOI: 10.1016/j.nbd.2019.104625] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 08/26/2019] [Accepted: 09/23/2019] [Indexed: 12/23/2022] Open
|
17
|
Sokolenko E, Hudson MR, Nithianantharajah J, Jones NC. The mGluR 2/3 agonist LY379268 reverses NMDA receptor antagonist effects on cortical gamma oscillations and phase coherence, but not working memory impairments, in mice. J Psychopharmacol 2019; 33:1588-1599. [PMID: 31580222 DOI: 10.1177/0269881119875976] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Abnormalities in neural oscillations that occur in the gamma frequency range (30-80 Hz) may underlie cognitive deficits in schizophrenia. Both cognitive impairments and gamma oscillatory disturbances can be induced in healthy people and rodents by administration of N-methyl-D-aspartate receptor (NMDAr) antagonists. AIMS We studied relationships between cognitive impairment and gamma abnormalities following NMDAr antagonism, and attempted to reverse deficits with the metabotropic glutamate receptor type 2/3 (mGluR2/3) agonist LY379268. METHODS C57/Bl6 mice were trained to perform the Trial-Unique Nonmatching to Location (TUNL) touchscreen test for working memory. They were then implanted with local field potential (LFP) recording electrodes in prefrontal cortex and dorsal hippocampus. Mice were administered either LY379268 (3 mg/kg) or vehicle followed by the NMDAr antagonist MK-801 (0.3 or 1 mg/kg) or vehicle prior to testing on the TUNL task, or recording LFPs during the presentation of an auditory stimulus. RESULTS MK-801 impaired working memory and increased perseveration, but these behaviours were not improved by LY379268 treatment. MK-81 increased the power of ongoing gamma and high gamma (130-180 Hz) oscillations in both brain regions and regional coherence between regions, and these signatures were augmented by LY379268. However, auditory-evoked gamma oscillation deficits caused by MK-801 were not affected by LY379268 pretreatment. CONCLUSIONS NMDA receptor antagonism impairs working memory in mice, but this is not reversed by stimulation of mGluR2/3. Since elevations in ongoing gamma power and regional coherence caused by MK-801 were improved by LY379268, it appears unlikely that these specific oscillatory abnormalities underlie the working memory impairment caused by NMDAr antagonism.
Collapse
Affiliation(s)
- Elysia Sokolenko
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne Brain Centre, Parkville, VIC, Australia
| | - Matthew R Hudson
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne Brain Centre, Parkville, VIC, Australia.,Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne, VIC, Australia
| | - Jess Nithianantharajah
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Nigel C Jones
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne Brain Centre, Parkville, VIC, Australia.,Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne, VIC, Australia
| |
Collapse
|
18
|
Nugent AC, Ballard ED, Gould TD, Park LT, Moaddel R, Brutsche NE, Zarate CA. Ketamine has distinct electrophysiological and behavioral effects in depressed and healthy subjects. Mol Psychiatry 2019; 24:1040-1052. [PMID: 29487402 PMCID: PMC6111001 DOI: 10.1038/s41380-018-0028-2] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 07/13/2017] [Accepted: 11/03/2017] [Indexed: 01/19/2023]
Abstract
Ketamine's mechanism of action was assessed using gamma power from magnetoencephalography (MEG) as a proxy measure for homeostatic balance in 35 unmedicated subjects with major depressive disorder (MDD) and 25 healthy controls enrolled in a double-blind, placebo-controlled, randomized cross-over trial of 0.5 mg/kg ketamine. MDD subjects showed significant improvements in depressive symptoms, and healthy control subjects exhibited modest but significant increases in depressive symptoms for up to 1 day after ketamine administration. Both groups showed increased resting gamma power following ketamine. In MDD subjects, gamma power was not associated with the magnitude of the antidepressant effect. However, baseline gamma power was found to moderate the relationship between post-ketamine gamma power and antidepressant response; specifically, higher post-ketamine gamma power was associated with better response in MDD subjects with lower baseline gamma, with an inverted relationship in MDD subjects with higher baseline gamma. This relationship was observed in multiple regions involved in networks hypothesized to be involved in the pathophysiology of MDD. This finding suggests biological subtypes based on the direction of homeostatic dysregulation and has important implications for inferring ketamine's mechanism of action from studies of healthy controls alone.
Collapse
Affiliation(s)
- Allison C Nugent
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Elizabeth D Ballard
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Todd D Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Lawrence T Park
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Ruin Moaddel
- Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
| | - Nancy E Brutsche
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
19
|
Park L, Furey M, Nugent AC, Farmer C, Ellis J, Szczepanik J, Lener MS, Zarate CA. Neurophysiological Changes Associated with Antidepressant Response to Ketamine Not Observed in a Negative Trial of Scopolamine in Major Depressive Disorder. Int J Neuropsychopharmacol 2019; 22:10-18. [PMID: 30184133 PMCID: PMC6313153 DOI: 10.1093/ijnp/pyy051] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 05/09/2018] [Accepted: 07/27/2018] [Indexed: 12/11/2022] Open
Abstract
Background This randomized, placebo-controlled, crossover trial examined the antidepressant efficacy of the muscarinic antagonist scopolamine in major depressive disorder subjects with more severe and refractory forms of major depressive disorder relative to previous reports. Methods Participants included 23 medication-free major depressive disorder subjects (12 F/11 M, 20-55 years) currently experiencing a major depressive episode. Subjects had scored ≥20 on the Montgomery-Asberg Depression Rating Scale. Following a single-blind, placebo lead-in, participants were randomized to receive 2 counterbalanced blocks of 3 i.v. infusions of scopolamine (4 μg/kg) and placebo in a double-blind manner. The primary and secondary outcomes were the Montgomery-Asberg Depression Rating Scale and the Hamilton Anxiety Rating Scale, respectively. Magnetoencephalography and plasma brain-derived neurotrophic factor concentrations were obtained prior to and after each treatment phase. Results As assessed by both the Montgomery-Asberg Depression Rating Scale and Hamilton Anxiety Rating Scale, scopolamine had no significant antidepressant or anxiolytic effects relative to placebo. No significant drug vs placebo effects were seen in magnetoencephalography gamma power or brain-derived neurotrophic factor plasma concentrations, and brain-derived neurotrophic factor changes did not correlate with change in Montgomery-Asberg Depression Rating Scale score in response to scopolamine. Conclusions These results do not support the efficacy of scopolamine for more severe or refractory forms of depression. No pre- to post-infusion changes in plasma brain-derived neurotrophic factor were detected, and magnetoencephalography gamma power changed only in the placebo lead-in, suggesting that these biomarker measures were not affected by scopolamine in this cohort. While difficult to interpret given the lack of antidepressant response, the findings suggest that the neurobiological effects of ketamine and scopolamine are at least partly distinct.
Collapse
Affiliation(s)
- Lawrence Park
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Maura Furey
- Janssen Research and Development, LLC, La Jolla, California
| | - Allison C Nugent
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Cristan Farmer
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Jessica Ellis
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Joanna Szczepanik
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Marc S Lener
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
20
|
Hudson MR, Hannan AJ, O’Brien TJ, Jones NC. High-Frequency Neuronal Oscillatory Abnormalities in the Phospholipase C-β1 Knockout Mouse Model of Schizophrenia. Int J Neuropsychopharmacol 2018; 22:221-231. [PMID: 30517689 PMCID: PMC6403088 DOI: 10.1093/ijnp/pyy097] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 11/27/2018] [Accepted: 11/30/2018] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Schizophrenia is a complex neuropsychiatric disorder characterized by psychoses, socioaffective disturbances, and cognitive deficits. The phosphodiesterase enzyme phospholipase C-β1 has been reported to be reduced in postmortem tissue of schizophrenia patients. Dysregulation of neuronal oscillations, particularly those in the higher frequency range such as beta (12-30 Hz) and gamma (30-80 Hz), are also associated with this disorder. We investigated the influence of phospholipase C-β1 gene deletion on cortical oscillatory activity and sensorimotor gating behavior. METHODS Adult phospholipase C-β1 knockout and wild-type C57Bl/6J control mice (total n = 26) underwent surgical implantation of extradural electrodes to allow electrocorticography recordings. Electrocorticography was recorded during prepulse inhibition behavior sessions to measure ongoing and auditory-evoked electrophysiological responses. Mice were also pretreated with antipsychotic drugs haloperidol (0.25 mg/kg), clozapine (2.5 mg/kg), and olanzapine (5 mg/kg). RESULTS Phospholipase C-β1 knockout mice exhibited reduced prepulse inhibition and diminished power and phase synchrony of beta and gamma oscillatory responses to auditory stimuli as well as elevated ongoing beta oscillations. Reductions in prepulse inhibition were highly correlated with the power and phase synchrony of evoked oscillations. Clozapine and olanzapine ameliorated the prepulse inhibition deficit in phospholipase C-β1 knockout mice, but not the electrophysiology abnormalities. CONCLUSIONS Phospholipase C-β1 reduction leads to disturbances to beta and gamma oscillatory dynamics and prepulse inhibition behavior. The strong relationships between these measures demonstrate the importance of event-related oscillatory activity to sensorimotor gating behavior. However, dissociation of these measures observed in the drug studies suggests that abnormalities in neuronal networks may not necessarily need to be corrected for behavioral improvement.
Collapse
Affiliation(s)
- Matthew R Hudson
- Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Melbourne Brain Centre, Parkville, Victoria, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne Brain Centre, Parkville, Victoria, Australia
| | - Terence J O’Brien
- Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Melbourne Brain Centre, Parkville, Victoria, Australia,Department of Neuroscience, Central Clinical School, Monash University and Department of Neurology, The Alfred Hospital, Melbourne, Victoria, Australia
| | - Nigel C Jones
- Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Melbourne Brain Centre, Parkville, Victoria, Australia,Department of Neuroscience, Central Clinical School, Monash University and Department of Neurology, The Alfred Hospital, Melbourne, Victoria, Australia,Correspondence: Nigel C. Jones, PhD, Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, 3004, Australia ()
| |
Collapse
|
21
|
Popova P, Rockstroh B, Miller GA, Wienbruch C, Carolus AM, Popov T. The impact of cognitive training on spontaneous gamma oscillations in schizophrenia. Psychophysiology 2018; 55:e13083. [PMID: 29624694 DOI: 10.1111/psyp.13083] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 03/08/2018] [Accepted: 03/08/2018] [Indexed: 12/27/2022]
Abstract
Schizophrenia patients exhibit less gamma-frequency EEG/MEG activity (>30 Hz), a finding interpreted as evidence of poor temporal neural organization and functional network communication. Research has shown that neuroplasticity-oriented training can improve task-related oscillatory dynamics, indicating some reorganization capacity in schizophrenia. Demonstrating a generalization of such task training effects to spontaneous oscillations at rest would not only enrich understanding of this neuroplastic potential but inform the interpretation of spontaneous gamma oscillations in the service of normal cognitive function. In the present study, neuromagnetic resting-state oscillatory brain activity and cognitive performance were assessed before and after training in 61 schizophrenia patients, who were randomly assigned to 4 weeks of neuroplasticity-oriented targeted cognitive training or treatment as usual (TAU). Gamma power of 40-90 Hz increased after training, but not after TAU, in a frontoparietal network. Across two types of training, this increase was related to improved cognitive test performance. These results indicate that abnormal oscillatory dynamics in schizophrenia patients manifested in spontaneous gamma activity can be changed with neuroplasticity-oriented training parallel to cognitive performance.
Collapse
Affiliation(s)
- Petia Popova
- Department of Psychology, University of Konstanz, Konstanz, Germany
| | | | - Gregory A Miller
- Department of Psychology and Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California, USA
| | | | - Almut M Carolus
- Department of Psychology, University of Konstanz, Konstanz, Germany
| | - Tzvetan Popov
- Department of Psychology, University of Konstanz, Konstanz, Germany
| |
Collapse
|
22
|
Anderson PM, Jones NC, O'Brien TJ, Pinault D. The N-Methyl d-Aspartate Glutamate Receptor Antagonist Ketamine Disrupts the Functional State of the Corticothalamic Pathway. Cereb Cortex 2018; 27:3172-3185. [PMID: 27261525 DOI: 10.1093/cercor/bhw168] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The non-competitive N-methyl d-aspartate glutamate receptor (NMDAR) antagonist ketamine elicits a brain state resembling high-risk states for developing psychosis and early stages of schizophrenia characterized by sensory and cognitive deficits and aberrant ongoing gamma (30-80 Hz) oscillations in cortical and subcortical structures, including the thalamus. The underlying mechanisms are unknown. The goal of the present study was to determine whether a ketamine-induced psychotic-relevant state disturbs the functional state of the corticothalamic (CT) pathway. Multisite field recordings were performed in the somatosensory CT system of the sedated rat. Baseline activity was challenged by activation of vibrissa-related prethalamic inputs. The sensory-evoked thalamic response was characterized by a short-latency (∼4 ms) prethalamic-mediated negative sharp potential and a longer latency (∼10 ms) CT-mediated negative potential. Following a single subcutaneous injection of ketamine (2.5 mg/kg), spontaneously occurring and sensory-evoked thalamic gamma oscillations increased and decreased in power, respectively. The power of the sensory-related gamma oscillations was positively correlated with both the amplitude and the area under the curve of the corresponding CT potential but not with the prethalamic potential. The present results show that the layer VI CT pathway significantly contributes in thalamic gamma oscillations, and they support the hypothesis that reduced NMDAR activation disturbs the functional state of CT and corticocortical networks.
Collapse
Affiliation(s)
- Paul M Anderson
- Neuropsychologie cognitive et physiopathologie de la schizophrénie, INSERM U1114, Strasbourg, France.,FMTS, Faculté de Médecine, Université de Strasbourg, Strasbourg, France.,Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia.,Current address: Department of Cognitive Neuroscience, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Nigel C Jones
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
| | - Terence J O'Brien
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
| | - Didier Pinault
- Neuropsychologie cognitive et physiopathologie de la schizophrénie, INSERM U1114, Strasbourg, France.,FMTS, Faculté de Médecine, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
23
|
Jones NC, Hudson M, Foreman J, Rind G, Hill R, Manning EE, Buuse M. Brain‐derived neurotrophic factor haploinsufficiency impairs high‐frequency cortical oscillations in mice. Eur J Neurosci 2017; 48:2816-2825. [DOI: 10.1111/ejn.13722] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/30/2017] [Accepted: 09/13/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Nigel C. Jones
- Department of Medicine Melbourne Brain Centre Royal Melbourne Hospital University of Melbourne Parkville Vic. 3052 Australia
- Department of Neuroscience Central Clinical School Monash University Melbourne Vic. Australia
- Department of Neurology The Alfred Hospital Melbourne Vic. Australia
| | - Matthew Hudson
- Department of Medicine Melbourne Brain Centre Royal Melbourne Hospital University of Melbourne Parkville Vic. 3052 Australia
| | - Joshua Foreman
- Department of Medicine Melbourne Brain Centre Royal Melbourne Hospital University of Melbourne Parkville Vic. 3052 Australia
| | - Gil Rind
- Department of Medicine Melbourne Brain Centre Royal Melbourne Hospital University of Melbourne Parkville Vic. 3052 Australia
| | - Rachel Hill
- Department of Psychiatry Monash University Melbourne Vic. Australia
- Melbourne Brain Centre Florey Institutes of Neuroscience and Mental Health University of Melbourne Parkville Vic. Australia
| | - Elizabeth E. Manning
- Melbourne Brain Centre Florey Institutes of Neuroscience and Mental Health University of Melbourne Parkville Vic. Australia
| | - Maarten Buuse
- Melbourne Brain Centre Florey Institutes of Neuroscience and Mental Health University of Melbourne Parkville Vic. Australia
- School of Psychology and Public Health La Trobe University Melbourne Vic. Australia
- Department of Pharmacology University of Melbourne Melbourne Vic. Australia
- The College of Public Health, Medical and Veterinary Sciences James Cook University Townsville QLD Australia
| |
Collapse
|
24
|
Furth KE, McCoy AJ, Dodge C, Walters JR, Buonanno A, Delaville C. Neuronal correlates of ketamine and walking induced gamma oscillations in the medial prefrontal cortex and mediodorsal thalamus. PLoS One 2017; 12:e0186732. [PMID: 29095852 PMCID: PMC5667758 DOI: 10.1371/journal.pone.0186732] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 10/08/2017] [Indexed: 01/19/2023] Open
Abstract
Alterations in the function of the medial prefrontal cortex (mPFC) and its major thalamic source of innervation, the mediodorsal (MD) thalamus, have been hypothesized to contribute to the symptoms of schizophrenia. The NMDAR antagonist ketamine, used to model schizophrenia, elicits a brain state resembling early stage schizophrenia characterized by cognitive deficits and increases in cortical low gamma (40-70 Hz) power. Here we sought to determine how ketamine differentially affects spiking and gamma local field potential (LFP) activity in the rat mPFC and MD thalamus. Additionally, we investigated the ability of drugs targeting the dopamine D4 receptor (D4R) to modify the effects of ketamine on gamma activity as a measure of potential cognitive therapeutic efficacy. Rats were trained to walk on a treadmill to reduce confounds related to hyperactivity after ketamine administration (10 mg/kg s.c.) while recordings were obtained from electrodes chronically implanted in the mPFC and MD thalamus. Ketamine increased gamma LFP power in mPFC and MD thalamus in a similar frequency range, yet did not increase thalamocortical synchronization. Ketamine also increased firing rates and spike synchronization to gamma oscillations in the mPFC but decreased both measures in MD thalamus. Conversely, walking alone increased both firing rates and spike-gamma LFP correlations in both mPFC and MD thalamus. The D4R antagonist alone (L-745,870) had no effect on gamma LFP power during treadmill walking, although it reversed increases induced by the D4R agonist (A-412997) in both mPFC and MD thalamus. Neither drug altered ketamine-induced changes in gamma power or firing rates in the mPFC. However, in MD thalamus, the D4R agonist increased ketamine-induced gamma power and prevented ketamine's inhibitory effect on firing rates. Results provide new evidence that ketamine differentially modulates spiking and gamma power in MD thalamus and mPFC, supporting a potential role for both areas in contributing to ketamine-induced schizophrenia-like symptoms.
Collapse
Affiliation(s)
- Katrina E. Furth
- Neurophysiological Pharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
- Graduate Program for Neuroscience, Boston University, Boston, Massachusetts, United States of America
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alex J. McCoy
- Neurophysiological Pharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Caroline Dodge
- Neurophysiological Pharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Judith R. Walters
- Neurophysiological Pharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Andres Buonanno
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Claire Delaville
- Neurophysiological Pharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
25
|
Lee J, Hudson MR, O'Brien TJ, Nithianantharajah J, Jones NC. Local NMDA receptor hypofunction evokes generalized effects on gamma and high-frequency oscillations and behavior. Neuroscience 2017; 358:124-136. [DOI: 10.1016/j.neuroscience.2017.06.039] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 06/21/2017] [Accepted: 06/22/2017] [Indexed: 10/19/2022]
|
26
|
Cooper MD, Rosenblat JD, Cha DS, Lee Y, Kakar R, McIntyre RS. Strategies to mitigate dissociative and psychotomimetic effects of ketamine in the treatment of major depressive episodes: a narrative review. World J Biol Psychiatry 2017; 18:410-423. [PMID: 26752601 DOI: 10.3109/15622975.2016.1139747] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Objectives Replicated evidence has demonstrated that ketamine exerts rapid-acting and potent antidepressant effects. Notwithstanding, its promise to mitigate depressive symptoms and suicidality in antidepressant-resistant populations, several limitations and safety concerns accompany ketamine including, but not limited to, the potential for abuse and psychotomimetic/dissociative experiences. The focus of the current narrative review is to synthesise available evidence of strategies that may mitigate and fully prevent treatment-emergent psychotomimetic and dissociative effects associated with ketamine administration. Methods PubMed, Google Scholar and ClinicalTrials.gov were searched for relevant articles. Results Potential avenues investigated to minimise psychotomimetic effects associated with ketamine administration include the following: (1) altering dosing and infusion rates; (2) route of administration; (3) enantiomer choice; (4) co-administration with mood stabilisers of antipsychotics; and (5) use of alternative N-methyl-d-aspartate (NMDA)-modulating agents. Emerging evidence indicates that dissociative experiences can be significantly mitigated by using an intranasal route of administration, lower dosages, or use of alternative NMDA-modulating agents, namely lanicemine (AZD6765) and GLYX-13. Conclusions Currently, intranasal administration presents as the most promising strategy to mitigate dissociative and psychotomimetic effects; however, studies of strategies to mitigate the adverse events of ketamine are limited in number and quality and thus further investigation is still needed.
Collapse
Affiliation(s)
- Matthew D Cooper
- a Medical Sciences, Dalhousie University , Halifax , NS , Canada.,b Mood Disorders Psychopharmacology Unit, University Health Network , Toronto , ON , Canada
| | - Joshua D Rosenblat
- b Mood Disorders Psychopharmacology Unit, University Health Network , Toronto , ON , Canada.,c Department of Psychiatry , University of Toronto , Toronto , ON , Canada
| | - Danielle S Cha
- b Mood Disorders Psychopharmacology Unit, University Health Network , Toronto , ON , Canada.,d Institute of Medical Science, University of Toronto , Toronto , ON , Canada
| | - Yena Lee
- b Mood Disorders Psychopharmacology Unit, University Health Network , Toronto , ON , Canada
| | - Ron Kakar
- b Mood Disorders Psychopharmacology Unit, University Health Network , Toronto , ON , Canada.,e Department of Psychiatry , Western University , London and Windsor , ON , Canada
| | - Roger S McIntyre
- b Mood Disorders Psychopharmacology Unit, University Health Network , Toronto , ON , Canada.,c Department of Psychiatry , University of Toronto , Toronto , ON , Canada.,f Department of Pharmacology , University of Toronto , Toronto , ON , Canada
| |
Collapse
|
27
|
Johnson MP, Muhlhauser MA, Nisenbaum ES, Simmons RMA, Forster BM, Knopp KL, Yang L, Morrow D, Li DL, Kennedy JD, Swanson S, Monn JA. Broad spectrum efficacy with LY2969822, an oral prodrug of metabotropic glutamate 2/3 receptor agonist LY2934747, in rodent pain models. Br J Pharmacol 2017; 174:822-835. [PMID: 28177520 DOI: 10.1111/bph.13740] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 01/27/2017] [Accepted: 01/31/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND AND PURPOSE A body of evidence suggests activation of metabotropic glutamate 2/3 (mGlu2/3 ) receptors would be an effective analgesic in chronic pain conditions. Thus, the analgesic properties of a novel mGlu2/3 receptor agonist prodrug were investigated. EXPERIMENTAL APPROACH After oral absorption, the prodrug LY2969822 rapidly converts to the brain penetrant, potent and subtype-selective mGlu2/3 receptor agonist LY2934747. Behavioural assessments of allodynia, hyperalgesia and nocifensive behaviours were determined in preclinical pain models after administration of LY2969822 0.3-10 mg·kg-1 . In addition, the ability of i.v. LY2934747 to modulate dorsal horn spinal cord wide dynamic range (WDR) neurons in spinal nerve ligated (SNL) rats was assessed. KEY RESULTS Following treatment with LY2934747, the spontaneous activity and electrically-evoked wind-up of WDR neurons in rats that had undergone spinal nerve ligation and developed mechanical allodynia were suppressed. In a model of sensitization, orally administered LY2969822 prevented the nociceptive behaviours induced by an intraplantar injection of formalin. The on-target nature of this effect was confirmed by blockade with an mGlu2/3 receptor antagonist. LY2969822 prevented capsaicin-induced tactile hypersensitivity, reversed the SNL-induced tactile hypersensitivity and reversed complete Freund's adjuvant - induced mechanical hyperalgesia. The mGlu2/3 receptor agonist prodrug demonstrated efficacy in visceral pain models, including a colorectal distension model and partially prevented the nocifensive behaviours in the mouse acetic acid writhing model. CONCLUSIONS AND IMPLICATIONS Following oral administration of the prodrug LY2969822, the mGlu2/3 receptor agonist LY2934747 was formed and this attenuated pain behaviours across a broad range of preclinical pain models.
Collapse
Affiliation(s)
- Michael P Johnson
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Mark A Muhlhauser
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Eric S Nisenbaum
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Rosa M A Simmons
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Beth M Forster
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Kelly L Knopp
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Lijuan Yang
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Denise Morrow
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Dominic L Li
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Jeffrey D Kennedy
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Steven Swanson
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - James A Monn
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
28
|
Sampaio LRL, Borges LTN, Barbosa TM, Matos NCB, Lima RDF, Oliveira MND, Gularte VN, Patrocínio MCA, Macêdo D, Vale OCD, Vasconcelos SMMD. Electroencephalographic study of chlorpromazine alone or combined with alpha-lipoic acid in a model of schizophrenia induced by ketamine in rats. J Psychiatr Res 2017; 86:73-82. [PMID: 27951451 DOI: 10.1016/j.jpsychires.2016.12.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 11/20/2016] [Accepted: 12/01/2016] [Indexed: 11/18/2022]
Abstract
Schizophrenia is characterized by behavioral symptoms, brain function impairments and electroencephalographic (EEG) changes. Dysregulation of immune responses and oxidative imbalance underpins this mental disorder. The present study aimed to investigate the effects of the typical antipsychotic chlorpromazine (CP) alone or combined with the natural antioxidant alpha-lipoic acid (ALA) on changes in the hippocampal average spectral power induced by ketamine (KET). Three days after stereotactic implantation of electrodes, male Wistar rats were divided into groups treated for 10 days with saline (control) or KET (10 mg/kg, IP). CP (1 or 5 mg/kg, IP) alone or combined with ALA (100 mg/kg, P.O.) was administered 30 min before KET or saline. Hippocampal EEG recordings were taken on the 1st, 5th and 10th days of treatment immediately after the last drug administration. KET significantly increased average spectral power of delta and gamma-high bands on the 5th and 10th days of treatment when compared to control. Gamma low-band significantly increased on the 1st, 5th and 10th days when compared to control group. This effect of KET was prevented by CP alone or combined with ALA. Indeed, the combination of ALA 100 + CP1 potentiated the inhibitory effects of CP1 on gamma low-band oscillations. In conclusion, our results showed that KET presents excitatory and time-dependent effects on hippocampal EEG bands activity. KET excitatory effects on EEG were prevented by CP alone and in some situations potentiated by its combination with ALA.
Collapse
Affiliation(s)
- Luis Rafael Leite Sampaio
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil; Health Science Center, School of Nursing, University of Fortaleza, Fortaleza, Ceará, Brazil
| | - Lucas Teixeira Nunes Borges
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil; Health Science Center, School of Nursing, University of Fortaleza, Fortaleza, Ceará, Brazil
| | - Talita Matias Barbosa
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Natalia Castelo Branco Matos
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Ricardo de Freitas Lima
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | | | - Viviane Nóbrega Gularte
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | | | - Danielle Macêdo
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Otoni Cardoso do Vale
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Silvânia Maria Mendes de Vasconcelos
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil.
| |
Collapse
|
29
|
Hudson MR, Rind G, O'Brien TJ, Jones NC. Reversal of evoked gamma oscillation deficits is predictive of antipsychotic activity with a unique profile for clozapine. Transl Psychiatry 2016; 6:e784. [PMID: 27093066 PMCID: PMC4872409 DOI: 10.1038/tp.2016.51] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 02/29/2016] [Accepted: 03/05/2016] [Indexed: 11/09/2022] Open
Abstract
Recent heuristic models of schizophrenia propose that abnormalities in the gamma frequency cerebral oscillations may be closely tied to the pathophysiology of the disorder, with hypofunction of N-methyl-d-aspartate receptors (NMDAr) implicated as having a crucial role. Prepulse inhibition (PPI) is a behavioural measure of sensorimotor gating that is disrupted in schizophrenia. We tested the ability for antipsychotic drugs with diverse pharmacological actions to (1) ameliorate NMDAr antagonist-induced disruptions to gamma oscillations and (2) attenuate NMDAr antagonist-induced disruptions to PPI. We hypothesized that antipsychotic-mediated improvement of PPI deficits would be accompanied by a normalization of gamma oscillatory activity. Wistar rats were implanted with extradural electrodes to facilitate recording of electroencephalogram during PPI behavioural testing. In each session, the rats were administered haloperidol (0.25 mg kg(-1)), clozapine (5 mg kg(-1)), olanzapine (5 mg kg(-1)), LY379268 (3 mg kg(-1)), NFPS (sarcosine, 1 mg kg(-1)), d-serine (1800 mg kg(-1)) or vehicle, followed by the NMDAr antagonists MK-801(0.16 mg kg(-1)), ketamine (5 mg kg(-1)) or vehicle. Outcome measures were auditory-evoked, as well as ongoing, gamma oscillations and PPI. Although treatment with all the clinically validated antipsychotic drugs reduced ongoing gamma oscillations, clozapine was the only compound that prevented the sensory-evoked gamma deficit produced by ketamine and MK-801. In addition, clozapine was also the only antipsychotic that attenuated the disruption to PPI produced by the NMDAr antagonists. We conclude that disruptions to evoked, but not ongoing, gamma oscillations caused by NMDAr antagonists are functionally relevant, and suggest that compounds, which restore sensory-evoked gamma oscillations may improve sensory processing in patients with schizophrenia.
Collapse
Affiliation(s)
- M R Hudson
- Department of Medicine, Royal Melbourne Hospital, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia
| | - G Rind
- Department of Medicine, Royal Melbourne Hospital, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia
| | - T J O'Brien
- Department of Medicine, Royal Melbourne Hospital, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia
| | - N C Jones
- Department of Medicine, Royal Melbourne Hospital, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia,Department of Medicine, Royal Melbourne Hospital, Melbourne Brain Centre, University of Melbourne, Parkville, VIC 3052, Australia. E-mail:
| |
Collapse
|
30
|
Ahnaou A, de Boer P, Lavreysen H, Huysmans H, Sinha V, Raeymaekers L, Van De Casteele T, Cid J, Van Nueten L, Macdonald G, Kemp J, Drinkenburg W. Translational neurophysiological markers for activity of the metabotropic glutamate receptor (mGluR2) modulator JNJ-40411813: Sleep EEG correlates in rodents and healthy men. Neuropharmacology 2016; 103:290-305. [DOI: 10.1016/j.neuropharm.2015.11.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/28/2015] [Accepted: 11/28/2015] [Indexed: 12/31/2022]
|
31
|
Robson SE, Brookes MJ, Hall EL, Palaniyappan L, Kumar J, Skelton M, Christodoulou NG, Qureshi A, Jan F, Katshu MZ, Liddle EB, Liddle PF, Morris PG. Abnormal visuomotor processing in schizophrenia. NEUROIMAGE-CLINICAL 2015; 12:869-878. [PMID: 27872809 PMCID: PMC5107643 DOI: 10.1016/j.nicl.2015.08.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 08/11/2015] [Accepted: 08/13/2015] [Indexed: 11/30/2022]
Abstract
Subtle disturbances of visual and motor function are known features of schizophrenia and can greatly impact quality of life; however, few studies investigate these abnormalities using simple visuomotor stimuli. In healthy people, electrophysiological data show that beta band oscillations in sensorimotor cortex decrease during movement execution (event-related beta desynchronisation (ERBD)), then increase above baseline for a short time after the movement (post-movement beta rebound (PMBR)); whilst in visual cortex, gamma oscillations are increased throughout stimulus presentation. In this study, we used a self-paced visuomotor paradigm and magnetoencephalography (MEG) to contrast these responses in patients with schizophrenia and control volunteers. We found significant reductions in the peak-to-peak change in amplitude from ERBD to PMBR in schizophrenia compared with controls. This effect was strongest in patients who made fewer movements, whereas beta was not modulated by movement in controls. There was no significant difference in the amplitude of visual gamma between patients and controls. These data demonstrate that clear abnormalities in basic sensorimotor processing in schizophrenia can be observed using a very simple MEG paradigm. Visual and motor deficits in schizophrenia are rarely investigated. We use MEG to non-invasively assess the neural basis of these deficits. Patients showed abnormalities in neuronal oscillations in motor cortex. Beta band power, reflecting cortical inhibition, was reduced after movements. Increased movement frequency may be a behavioural compensation for this reduction.
Collapse
Affiliation(s)
- Siân E Robson
- Sir Peter Mansfield Imaging Centre, School of Physics and Astronomy, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Matthew J Brookes
- Sir Peter Mansfield Imaging Centre, School of Physics and Astronomy, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Emma L Hall
- Sir Peter Mansfield Imaging Centre, School of Physics and Astronomy, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Lena Palaniyappan
- Centre for Translational Neuroimaging in Mental Health, Institute of Mental Health, School of Medicine, University of Nottingham, Jubilee Campus, Triumph Road, Nottingham NG7 2TU, UK
| | - Jyothika Kumar
- Centre for Translational Neuroimaging in Mental Health, Institute of Mental Health, School of Medicine, University of Nottingham, Jubilee Campus, Triumph Road, Nottingham NG7 2TU, UK
| | - Michael Skelton
- Centre for Translational Neuroimaging in Mental Health, Institute of Mental Health, School of Medicine, University of Nottingham, Jubilee Campus, Triumph Road, Nottingham NG7 2TU, UK
| | - Nikolaos G Christodoulou
- Centre for Translational Neuroimaging in Mental Health, Institute of Mental Health, School of Medicine, University of Nottingham, Jubilee Campus, Triumph Road, Nottingham NG7 2TU, UK
| | - Ayaz Qureshi
- Kevin White Unit, Smithdown Health Park, Smithdown Road, Liverpool L15 2HE, UK
| | - Fiesal Jan
- Herschel Prins Centre, Glenfield Hospital, Leicestershire Partnership NHS Trust, Groby Road, Leicester LE3 9QP, UK
| | - Mohammad Z Katshu
- Centre for Translational Neuroimaging in Mental Health, Institute of Mental Health, School of Medicine, University of Nottingham, Jubilee Campus, Triumph Road, Nottingham NG7 2TU, UK
| | - Elizabeth B Liddle
- Centre for Translational Neuroimaging in Mental Health, Institute of Mental Health, School of Medicine, University of Nottingham, Jubilee Campus, Triumph Road, Nottingham NG7 2TU, UK
| | - Peter F Liddle
- Centre for Translational Neuroimaging in Mental Health, Institute of Mental Health, School of Medicine, University of Nottingham, Jubilee Campus, Triumph Road, Nottingham NG7 2TU, UK
| | - Peter G Morris
- Sir Peter Mansfield Imaging Centre, School of Physics and Astronomy, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| |
Collapse
|
32
|
Leishman E, O’Donnell BF, Millward JB, Vohs JL, Rass O, Krishnan GP, Bolbecker AR, Morzorati SL. Phencyclidine Disrupts the Auditory Steady State Response in Rats. PLoS One 2015; 10:e0134979. [PMID: 26258486 PMCID: PMC4530939 DOI: 10.1371/journal.pone.0134979] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 07/12/2015] [Indexed: 12/20/2022] Open
Abstract
The Auditory Steady-State Response (ASSR) in the electroencephalogram (EEG) is usually reduced in schizophrenia (SZ), particularly to 40 Hz stimulation. The gamma frequency ASSR deficit has been attributed to N-methyl-D-aspartate receptor (NMDAR) hypofunction. We tested whether the NMDAR antagonist, phencyclidine (PCP), produced similar ASSR deficits in rats. EEG was recorded from awake rats via intracranial electrodes overlaying the auditory cortex and at the vertex of the skull. ASSRs to click trains were recorded at 10, 20, 30, 40, 50, and 55 Hz and measured by ASSR Mean Power (MP) and Phase Locking Factor (PLF). In Experiment 1, the effect of different subcutaneous doses of PCP (1.0, 2.5 and 4.0 mg/kg) on the ASSR in 12 rats was assessed. In Experiment 2, ASSRs were compared in PCP treated rats and control rats at baseline, after acute injection (5 mg/kg), following two weeks of subchronic, continuous administration (5 mg/kg/day), and one week after drug cessation. Acute administration of PCP increased PLF and MP at frequencies of stimulation below 50 Hz, and decreased responses at higher frequencies at the auditory cortex site. Acute administration had a less pronounced effect at the vertex site, with a reduction of either PLF or MP observed at frequencies above 20 Hz. Acute effects increased in magnitude with higher doses of PCP. Consistent effects were not observed after subchronic PCP administration. These data indicate that acute administration of PCP, a NMDAR antagonist, produces an increase in ASSR synchrony and power at low frequencies of stimulation and a reduction of high frequency (> 40 Hz) ASSR activity in rats. Subchronic, continuous administration of PCP, on the other hand, has little impact on ASSRs. Thus, while ASSRs are highly sensitive to NMDAR antagonists, their translational utility as a cross-species biomarker for NMDAR hypofunction in SZ and other disorders may be dependent on dose and schedule.
Collapse
Affiliation(s)
- Emma Leishman
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States of America
| | - Brian F. O’Donnell
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States of America
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States of America
- Larue D. Carter Memorial Hospital, Indianapolis, Indiana, United States of America
- * E-mail:
| | - James B. Millward
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Jenifer L. Vohs
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States of America
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States of America
- Larue D. Carter Memorial Hospital, Indianapolis, Indiana, United States of America
| | - Olga Rass
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States of America
| | - Giri P. Krishnan
- University of California Riverside, Riverside, CA, United States of America
| | - Amanda R. Bolbecker
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, United States of America
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States of America
- Larue D. Carter Memorial Hospital, Indianapolis, Indiana, United States of America
| | - Sandra L. Morzorati
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States of America
| |
Collapse
|
33
|
Puig MV, Gener T. Serotonin Modulation of Prefronto-Hippocampal Rhythms in Health and Disease. ACS Chem Neurosci 2015; 6:1017-25. [PMID: 25799292 DOI: 10.1021/cn500350e] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
There is mounting evidence that most cognitive functions depend upon the coordinated activity of neuronal networks often located far from each other in the brain. Ensembles of neurons synchronize their activity, generating oscillations at different frequencies that may encode behavior by allowing an efficient communication between brain areas. The serotonin system, by virtue of the widespread arborisation of serotonergic neurons, is in an excellent position to exert strong modulatory actions on brain rhythms. These include specific oscillatory activities in the prefrontal cortex and the hippocampus, two brain areas essential for many higher-order cognitive functions. Psychiatric patients show abnormal oscillatory activities in these areas, notably patients with schizophrenia who display psychotic symptoms as well as affective and cognitive impairments. Synchronization of neural activity between the prefrontal cortex and the hippocampus seems to be important for cognition and, in fact, reduced prefronto-hippocampal synchrony has been observed in a genetic mouse model of schizophrenia. Here, we review recent advances in the field of neuromodulation of brain rhythms by serotonin, focusing on the actions of serotonin in the prefrontal cortex and the hippocampus. Considering that the serotonergic system plays a crucial role in cognition and mood and is a target of many psychiatric treatments, it is surprising that this field of research is still in its infancy. In that regard, we point to future investigations that are much needed in this field.
Collapse
Affiliation(s)
- M. Victoria Puig
- Neuroscience Programme, Hospital del Mar Medical Research Institute (IMIM), Barcelona Biomedical Research Park (PRBB), Barcelona, Spain
| | - Thomas Gener
- Neuroscience Programme, Hospital del Mar Medical Research Institute (IMIM), Barcelona Biomedical Research Park (PRBB), Barcelona, Spain
- Systems Biology Program, Centre for Genomic Regulation (CRG), Barcelona Biomedical Research Park (PRBB), Barcelona, Spain
| |
Collapse
|
34
|
Skoblenick KJ, Womelsdorf T, Everling S. Ketamine Alters Outcome-Related Local Field Potentials in Monkey Prefrontal Cortex. Cereb Cortex 2015; 26:2743-2752. [PMID: 26045564 DOI: 10.1093/cercor/bhv128] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
A subanesthetic dose of the noncompetitive N-methyl-d-aspartate receptor antagonist ketamine is known to induce a schizophrenia-like phenotype in humans and nonhuman primates alike. The transient behavioral changes mimic the positive, negative, and cognitive symptoms of the disease but the neural mechanisms behind these changes are poorly understood. A growing body of evidence indicates that the cognitive control processes associated with prefrontal cortex (PFC) regions relies on groups of neurons synchronizing at narrow-band frequencies measurable in the local field potential (LFP). Here, we recorded LFPs from the caudo-lateral PFC of 2 macaque monkeys performing an antisaccade task, which requires the suppression of an automatic saccade toward a stimulus and the initiation of a goal-directed saccade in the opposite direction. Preketamine injection activity showed significant differences in a narrow 20-30 Hz beta frequency band between correct and error trials in the postsaccade response epoch. Ketamine significantly impaired the animals' performance and was associated with a loss of the differences in outcome-specific beta-band power. Instead, we observed a large increase in high-gamma-band activity. Our results suggest that the PFC employs beta-band synchronization to prepare for top-down cognitive control of saccades and the monitoring of task outcome.
Collapse
Affiliation(s)
| | | | - Stefan Everling
- Department of Anatomy and Cell Biology.,Department of Physiology and Pharmacology.,Department of Psychology, Western University, London, ON, Canada.,Robarts Research Institute, London, ON, Canada
| |
Collapse
|
35
|
Lovelace JW, Corches A, Vieira PA, Hiroto AS, Mackie K, Korzus E. An animal model of female adolescent cannabinoid exposure elicits a long-lasting deficit in presynaptic long-term plasticity. Neuropharmacology 2015; 99:242-55. [PMID: 25979486 DOI: 10.1016/j.neuropharm.2015.04.034] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 04/16/2015] [Accepted: 04/29/2015] [Indexed: 11/29/2022]
Abstract
Cannabis continues to be the most accessible and popular illicit recreational drug. Whereas current data link adolescence cannabinoid exposure to increased risk for dependence on other drugs, depression, anxiety disorders and psychosis, the mechanism(s) underlying these adverse effects remains controversial. Here we show in a mouse model of female adolescent cannabinoid exposure deficient endocannabinoid (eCB)-mediated signaling and presynaptic forms of long-term depression at adult central glutamatergic synapses in the prefrontal cortex. Increasing endocannabinoid levels by blockade of monoacylglycerol lipase, the primary enzyme responsible for degrading the endocannabinoid 2-arachidonoylglycerol (2-AG), with the specific inhibitor JZL 184 ameliorates eCB-LTD deficits. The observed deficit in cortical presynaptic signaling may represent a neural maladaptation underlying network instability and abnormal cognitive functioning. Our study suggests that adolescent cannabinoid exposure may permanently impair brain functions, including the brain's intrinsic ability to appropriately adapt to external influences.
Collapse
Affiliation(s)
- Jonathan W Lovelace
- Department of Psychology & Neuroscience Program, University of California Riverside, CA 92521, USA
| | - Alex Corches
- Biomedical Sciences Program, University of California Riverside, CA 92521, USA
| | - Philip A Vieira
- Department of Psychology & Neuroscience Program, University of California Riverside, CA 92521, USA
| | - Alex S Hiroto
- Department of Psychology & Neuroscience Program, University of California Riverside, CA 92521, USA
| | - Ken Mackie
- Department of Psychological & Brain Sciences, Gill Center for Biomedical Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Edward Korzus
- Department of Psychology & Neuroscience Program, University of California Riverside, CA 92521, USA; Biomedical Sciences Program, University of California Riverside, CA 92521, USA.
| |
Collapse
|