1
|
Bespalov A, Lütjens R, Doller D. Dusting off old blueprints: Is it time to reconsider metabotropic glutamate receptor 2 for therapeutic drug development? Pharmacol Biochem Behav 2025; 247:173908. [PMID: 39571688 DOI: 10.1016/j.pbb.2024.173908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 01/28/2025]
Abstract
The metabotropic glutamate receptor 2 (mGlu2) is a heavily studied therapeutic target in neuropsychiatry for which we anticipate a renewed interest in the near future. We review the rationale and the outcome of clinical trials with mGlu2/3 receptor agonists in schizophrenia, a field of intense research since a seminal publication by Patel and colleagues (2007). We summarize evidence about selective, potent and safe agents with quantifiable CNS penetration that can be used to test hypotheses of mGlu2 receptors involvement in neuropsychiatric diseases. We summarize lessons learned from previous programs that should be considered to maximize the probability of success when targeting orthosteric and allosteric enhancement of mGlu2 receptor function in schizophrenia and beyond. First, we propose expanding our focus beyond presynaptic mGlu2 receptor stimulation in schizophrenia to novel hypotheses and that the choice of a therapeutic indication no longer be dictated by commercial opportunity but following science as a driver. Second, evidence on internal validity of preclinical studies supporting efficacy claims in the mGlu2 field is very limited. This gap will need to be closed when reviewing the rationale to re-initiate efforts in this field. Third, the pomaglumetad program was halted due to insufficient clinical efficacy, partly because of the inability to identify a treatment responder population. In preclinical studies, effects of mGlu2/3 receptor stimulation also seemed to vary significantly between laboratories. Definition of the responsive subject population and development of response-predicting biomarkers is therefore one of the main avenues of further research in the mGlu2 field.
Collapse
|
2
|
Okubo R, Okada M, Motomura E. Dysfunction of the NMDA Receptor in the Pathophysiology of Schizophrenia and/or the Pathomechanisms of Treatment-Resistant Schizophrenia. Biomolecules 2024; 14:1128. [PMID: 39334894 PMCID: PMC11430065 DOI: 10.3390/biom14091128] [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: 08/15/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
For several decades, the dopamine hypothesis contributed to the discovery of numerous typical and atypical antipsychotics and was the sole hypothesis for the pathophysiology of schizophrenia. However, neither typical nor atypical antipsychotics, other than clozapine, have been effective in addressing negative symptoms and cognitive impairments, which are indices for the prognostic and disability outcomes of schizophrenia. Following the development of atypical antipsychotics, the therapeutic targets for antipsychotics expanded beyond the blockade of dopamine D2 and serotonin 5-HT2A receptors to explore the partial agonism of the D2 receptor and the modulation of new targets, such as D3, 5-HT1A, 5-HT7, and metabotropic glutamate receptors. Despite these efforts, to date, psychiatry has not successfully developed antipsychotics with antipsychotic properties proven to be superior to those of clozapine. The glutamate hypothesis, another hypothesis regarding the pathophysiology/pathomechanism of schizophrenia, was proposed based on clinical findings that N-methyl-D-aspartate glutamate receptor (NMDAR) antagonists, such as phencyclidine and ketamine, induce schizophrenia-like psychotic episodes. Large-scale genome-wide association studies (GWASs) revealed that approximately 30% of the risk genes for schizophrenia (the total number was over one hundred) encode proteins associated with glutamatergic transmission. These findings supported the validation of the glutamate hypothesis, which was inspired by the clinical findings regarding NMDAR antagonists. Additionally, these clinical and genetic findings suggest that schizophrenia is possibly a syndrome with complicated pathomechanisms that are affected by multiple biological and genetic vulnerabilities. The glutamate hypothesis has been the most extensively investigated pathophysiology/pathomechanism hypothesis, other than the dopamine hypothesis. Studies have revealed the possibility that functional abnormalities of the NMDAR play important roles in the pathophysiology/pathomechanism of schizophrenia. However, no antipsychotics derived from the glutamatergic hypothesis have yet been approved for the treatment of schizophrenia or treatment-resistant schizophrenia. Considering the increasing evidence supporting the potential pro-cognitive effects of glutamatergic agents and the lack of sufficient medications to treat the cognitive impairments associated with schizophrenia, these previous setbacks cannot preclude research into potential novel glutamate modulators. Given this background, to emphasize the importance of the dysfunction of the NMDAR in the pathomechanism and/or pathophysiology of schizophrenia, this review introduces the increasing findings on the functional abnormalities in glutamatergic transmission associated with the NMDAR.
Collapse
Affiliation(s)
| | - Motohiro Okada
- Department of Neuropsychiatry, Division of Neuroscience, Graduate School of Medicine, Mie University, Tsu 514-8507, Japan; (R.O.); (E.M.)
| | | |
Collapse
|
3
|
Fujikawa R, Yamada J, Maeda S, Iinuma KM, Moriyama G, Jinno S. Inhibition of reactive oxygen species production accompanying alternatively activated microglia by risperidone in a mouse ketamine model of schizophrenia. J Neurochem 2024; 168:2690-2709. [PMID: 38770640 DOI: 10.1111/jnc.16133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 04/17/2024] [Accepted: 05/06/2024] [Indexed: 05/22/2024]
Abstract
Recent studies have highlighted the potential involvement of reactive oxygen species (ROS) and microglia, a major source of ROS, in the pathophysiology of schizophrenia. In our study, we explored how the second-generation antipsychotic risperidone (RIS) affects ROS regulation and microglial activation in the hippocampus using a mouse ketamine (KET) model of schizophrenia. KET administration resulted in schizophrenia-like behaviors in male C57BL/6J mice, such as impaired prepulse inhibition (PPI) of the acoustic startle response and hyper-locomotion. These behaviors were mitigated by RIS. We found that the gene expression level of an enzyme responsible for ROS production (Nox2), which is primarily associated with activated microglia, was lower in KET/RIS-treated mice than in KET-treated mice. Conversely, the levels of antioxidant enzymes (Ho-1 and Gclc) were higher in KET/RIS-treated mice. The microglial density in the hippocampus was increased in KET-treated mice, which was counteracted by RIS. Hierarchical cluster analysis revealed three morphological subtypes of microglia. In control mice, most microglia were resting-ramified (type I, 89.7%). KET administration shifted the microglial composition to moderately ramified (type II, 44.4%) and hyper-ramified (type III, 25.0%). In KET/RIS-treated mice, type II decreased to 32.0%, while type III increased to 34.0%. An in vitro ROS assay showed that KET increased ROS production in dissociated hippocampal microglia, and this effect was mitigated by RIS. Furthermore, we discovered that a NOX2 inhibitor could counteract KET-induced behavioral deficits. These findings suggest that pharmacological inhibition of ROS production by RIS may play a crucial role in ameliorating schizophrenia-related symptoms. Moreover, modulating microglial activation to regulate ROS production has emerged as a novel avenue for developing innovative treatments for schizophrenia.
Collapse
Affiliation(s)
- Risako Fujikawa
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jun Yamada
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shoichiro Maeda
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kyoko M Iinuma
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | - Shozo Jinno
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
4
|
Gärtner M, Weigand A, Meiering MS, Weigner D, Carstens L, Keicher C, Hertrampf R, Beckmann C, Mennes M, Wunder A, Grimm S. Region- and time- specific effects of ketamine on cerebral blood flow: a randomized controlled trial. Neuropsychopharmacology 2023; 48:1735-1741. [PMID: 37231079 PMCID: PMC10579356 DOI: 10.1038/s41386-023-01605-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 05/04/2023] [Accepted: 05/07/2023] [Indexed: 05/27/2023]
Abstract
There is intriguing evidence suggesting that ketamine might have distinct acute and delayed neurofunctional effects, as its acute administration transiently induces schizophrenia-like symptoms, while antidepressant effects slowly emerge and are most pronounced 24 h after administration. Studies attempting to characterize ketamine's mechanism of action by using blood oxygen level dependent (BOLD) imaging have yielded inconsistent results regarding implicated brain regions and direction of effects. This may be due to intrinsic properties of the BOLD contrast, while cerebral blood flow (CBF), as measured with arterial spin labeling, is a single physiological marker more directly related to neural activity. As effects of acute ketamine challenge are sensitive to modulation by pretreatment with lamotrigine, which inhibits glutamate release, a combination of these approaches should be particularly suited to offer novel insights. In total, 75 healthy participants were investigated in a double blind, placebo-controlled, randomized, parallel-group study and underwent two scanning sessions (acute/post 24 h.). Acute ketamine administration was associated with higher perfusion in interior frontal gyrus (IFG) and dorsolateral prefrontal cortex (DLPFC), but no other investigated brain region. Inhibition of glutamate release by pretreatment with lamotrigine abolished ketamine's effect on perfusion. At the delayed time point, pretreatment with lamotrigine was associated with lower perfusion in IFG. These findings underscore the idea that regionally selective patterns of CBF changes reflect proximate effects of modulated glutamate release on neuronal activity. Furthermore, region- specific sustained effects indicate both a swift restoration of disturbed homeostasis in DLPFC as well changes occurring beyond the immediate effects on glutamate signaling in IFG.
Collapse
Affiliation(s)
- Matti Gärtner
- Medical School Berlin, Berlin, Germany.
- Department of Psychiatry and Psychotherapy, Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany.
| | | | | | | | | | | | | | | | | | - Andreas Wunder
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Simone Grimm
- Medical School Berlin, Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry, University of Zurich, Zurich, Switzerland
| |
Collapse
|
5
|
Ju J, Liu L, Yang X, Men S, Hou ST. Distinctive effects of NMDA receptor modulators on cerebral microcirculation in a schizophrenia mouse model. Biochem Biophys Res Commun 2023; 653:62-68. [PMID: 36857901 DOI: 10.1016/j.bbrc.2023.02.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/16/2023] [Accepted: 02/15/2023] [Indexed: 02/18/2023]
Abstract
Substantial evidence demonstrates that schizophrenia patients have altered cerebral microcirculation. However, little is known regarding how cerebral microcirculatory blood flow (microCBF) changes in schizophrenia. Here, using time-lapse two-photon imaging of individual capillaries, we demonstrated a substantial decrease in cerebral microcirculation in a mouse model of schizophrenia. The involvement of NMDA receptor (NMDAR) functions was investigated to understand further the mechanism of microcirculation reduction in this animal model. Administration of D-serine, a selective full agonist at the glycine site of NMDAR, significantly increased the microCBF in the schizophrenia mouse. Interestingly, administration of GNE-8324, a GluN2A-selective positive allosteric modulator that selectively enhances NMDAR-mediated synaptic responses in inhibitory but not excitatory neurons, had no effect on the microCBF of the schizophrenia mice. Together, these data indicated that NMDAR participated in the regulation of microcirculation in schizophrenia using a mechanism dependent on the tonic NMDAR signaling and the selective modulation of inhibitory neuron activity. Further studies are warranted to establish NMDAR's role in modulating microcirculation in schizophrenia.
Collapse
Affiliation(s)
- Jun Ju
- Brain Research Centre and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Luping Liu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong Special Administrative Region of China
| | - Xinyi Yang
- Brain Research Centre and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Siqi Men
- Brain Research Centre and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Sheng-Tao Hou
- Brain Research Centre and Department of Biology, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
6
|
Gonzalez-Burgos I, Bainier M, Gross S, Schoenenberger P, Ochoa JA, Valencia M, Redondo RL. Glutamatergic and GABAergic Receptor Modulation Present Unique Electrophysiological Fingerprints in a Concentration-Dependent and Region-Specific Manner. eNeuro 2023; 10:ENEURO.0406-22.2023. [PMID: 36931729 PMCID: PMC10124153 DOI: 10.1523/eneuro.0406-22.2023] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 02/13/2023] [Accepted: 02/17/2023] [Indexed: 03/19/2023] Open
Abstract
Brain function depends on complex circuit interactions between excitatory and inhibitory neurons embedded in local and long-range networks. Systemic GABAA-receptor (GABAAR) or NMDA-receptor (NMDAR) modulation alters the excitatory-inhibitory balance (EIB), measurable with electroencephalography (EEG). However, EEG signatures are complex in localization and spectral composition. We developed and applied analytical tools to investigate the effects of two EIB modulators, MK801 (NMDAR antagonist) and diazepam (GABAAR modulator), on periodic and aperiodic EEG features in freely-moving male Sprague Dawley rats. We investigated how, across three brain regions, EEG features are correlated with EIB modulation. We found that the periodic component was composed of seven frequency bands that presented region-dependent and compound-dependent changes. The aperiodic component was also different between compounds and brain regions. Importantly, the parametrization into periodic and aperiodic components unveiled correlations between quantitative EEG and plasma concentrations of pharmacological compounds. MK-801 exposures were positively correlated with the slope of the aperiodic component. Concerning the periodic component, MK-801 exposures correlated negatively with the peak frequency of low-γ oscillations but positively with those of high-γ and high-frequency oscillations (HFOs). As for the power, θ and low-γ oscillations correlated negatively with MK-801, whereas mid-γ correlated positively. Diazepam correlated negatively with the knee of the aperiodic component, positively to β and negatively to low-γ oscillatory power, and positively to the modal frequency of θ, low-γ, mid-γ, and high-γ. In conclusion, correlations between exposures and pharmacodynamic effects can be better-understood thanks to the parametrization of EEG into periodic and aperiodic components. Such parametrization could be key in functional biomarker discovery.
Collapse
Affiliation(s)
- Irene Gonzalez-Burgos
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel 4070, Switzerland
- Program of Neuroscience, Centro de Investigación Médica Aplicada, Universidad de Navarra, Pamplona 31080, Spain
- Instituto de Investigación Sanitaria de Navarra (Navarra Institute for Health Research), Pamplona 31080, Spain
| | - Marie Bainier
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Simon Gross
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Philipp Schoenenberger
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel 4070, Switzerland
| | - José A Ochoa
- Program of Neuroscience, Centro de Investigación Médica Aplicada, Universidad de Navarra, Pamplona 31080, Spain
- Instituto de Investigación Sanitaria de Navarra (Navarra Institute for Health Research), Pamplona 31080, Spain
| | - Miguel Valencia
- Program of Neuroscience, Centro de Investigación Médica Aplicada, Universidad de Navarra, Pamplona 31080, Spain
- Instituto de Investigación Sanitaria de Navarra (Navarra Institute for Health Research), Pamplona 31080, Spain
- Institute of Data Science and Artificial Intelligence, Universidad de Navarra, Pamplona, Spain
| | - Roger L Redondo
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel 4070, Switzerland
| |
Collapse
|
7
|
Zhang HC, Du Y, Chen L, Yuan ZQ, Cheng Y. MicroRNA schizophrenia: Etiology, biomarkers and therapeutic targets. Neurosci Biobehav Rev 2023; 146:105064. [PMID: 36707012 DOI: 10.1016/j.neubiorev.2023.105064] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/11/2023] [Accepted: 01/22/2023] [Indexed: 01/26/2023]
Abstract
The three sets of symptoms associated with schizophrenia-positive, negative, and cognitive-are burdensome and have serious effects on public health, which affects up to 1% of the population. It is now commonly believed that in addition to the traditional dopaminergic mesolimbic pathway, the etiology of schizophrenia also includes neuronal networks, such as glutamate, GABA, serotonin, BDNF, oxidative stress, inflammation and the immune system. Small noncoding RNA molecules called microRNAs (miRNAs) have come to light as possible participants in the pathophysiology of schizophrenia in recent years by having an impact on these systems. These small RNAs regulate the stability and translation of hundreds of target transcripts, which has an impact on the entire gene network. There may be improved approaches to treat and diagnose schizophrenia if it is understood how these changes in miRNAs alter the critical related signaling pathways that drive the development and progression of the illness.
Collapse
Affiliation(s)
- Heng-Chang Zhang
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Yang Du
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, China
| | - Lei Chen
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, China
| | - Zeng-Qiang Yuan
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China; Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100850, China
| | - Yong Cheng
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China; Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, China; Institute of National Security, Minzu University of China, Beijing, China.
| |
Collapse
|
8
|
Driesen NR, Herman P, Rowland MA, Thompson G, Qiu M, He G, Fineberg S, Barron DS, Helgeson L, Lacadie C, Chow R, Gueorguieva R, Straun TC, Krystal JH, Hyder F. Ketamine Effects on Energy Metabolism, Functional Connectivity and Working Memory in Healthy Humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.21.529425. [PMID: 36865249 PMCID: PMC9980048 DOI: 10.1101/2023.02.21.529425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Working memory (WM) is a crucial resource for temporary memory storage and the guiding of ongoing behavior. N-methyl-D-aspartate glutamate receptors (NMDARs) are thought to support the neural underpinnings of WM. Ketamine is an NMDAR antagonist that has cognitive and behavioral effects at subanesthetic doses. To shed light on subanesthetic ketamine effects on brain function, we employed a multimodal imaging design, combining gas-free calibrated functional magnetic resonance imaging (fMRI) measurement of oxidative metabolism (CMRO 2 ), resting-state cortical functional connectivity assessed with fMRI, and WM-related fMRI. Healthy subjects participated in two scan sessions in a randomized, double-blind, placebo-controlled design. Ketamine increased CMRO 2 and cerebral blood flow (CBF) in prefrontal cortex (PFC) and other cortical regions. However, resting-state cortical functional connectivity was not affected. Ketamine did not alter CBF-CMRO 2 coupling brain-wide. Higher levels of basal CMRO 2 were associated with lower task-related PFC activation and WM accuracy impairment under both saline and ketamine conditions. These observations suggest that CMRO 2 and resting-state functional connectivity index distinct dimensions of neural activity. Ketamine’s impairment of WM-related neural activity and performance appears to be related to its ability to produce cortical metabolic activation. This work illustrates the utility of direct measurement of CMRO 2 via calibrated fMRI in studies of drugs that potentially affect neurovascular and neurometabolic coupling.
Collapse
|
9
|
Gärtner M, de Rover M, Václavů L, Scheidegger M, van Osch MJP, Grimm S. Increase in thalamic cerebral blood flow is associated with antidepressant effects of ketamine in major depressive disorder. World J Biol Psychiatry 2022; 23:643-652. [PMID: 34985394 DOI: 10.1080/15622975.2021.2020900] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Ketamine is a promising treatment option for patients with Major Depressive Disorder (MDD) and has become an important research tool to investigate antidepressant mechanisms of action. However, imaging studies attempting to characterise ketamine's mechanism of action using blood oxygen level-dependent signal (BOLD) imaging have yielded inconsistent results- at least partly due to intrinsic properties of the BOLD contrast, which measures a complex signal related to neural activity. To circumvent the limitations associated with the BOLD signal, we used arterial spin labelling (ASL) as an unambiguous marker of neuronal activity-related changes in cerebral blood flow (CBF). We measured CBF in 21 MDD patients at baseline and 24 h after receiving a single intravenous infusion of subanesthetic ketamine and examined relationships with clinical outcomes. Our findings demonstrate that increase in thalamus perfusion 24 h after ketamine administration is associated with greater improvement of depressive symptoms. Furthermore, lower thalamus perfusion at baseline is associated both with larger increases in perfusion 24 h after ketamine administration and with stronger reduction of depressive symptoms. These findings indicate that ASL is not only a useful tool to broaden our understanding of ketamine's mechanism of action but might also have the potential to inform treatment decisions based on CBF-defined regional disruptions.
Collapse
Affiliation(s)
- Matti Gärtner
- MSB-Medical School Berlin, Berlin, Germany.,Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Mischa de Rover
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands.,Department of Clinical Psychology, Institute of Psychology, Leiden University, Leiden, Netherlands
| | - Lena Václavů
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, Netherlands
| | - Milan Scheidegger
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zurich, Switzerland
| | - Matthias J P van Osch
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, Netherlands
| | - Simone Grimm
- MSB-Medical School Berlin, Berlin, Germany.,Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zurich, Switzerland
| |
Collapse
|
10
|
Suda Y, Uka T. The NMDA receptor antagonist ketamine impairs and delays context-dependent decision making in the parietal cortex. Commun Biol 2022; 5:690. [PMID: 35858997 PMCID: PMC9300646 DOI: 10.1038/s42003-022-03626-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/23/2022] [Indexed: 11/09/2022] Open
Abstract
Flexible decision making is an indispensable ability for humans. A subanesthetic dose of ketamine, an N-methyl-D-aspartate receptor antagonist, impairs this flexibility in a manner that is similar to patients with schizophrenia; however how it affects neural processes related to decision making remains unclear. Here, we report that ketamine administration impairs neural processing related to context-dependent decision making, and delays the onset of decision making. We recorded single unit activity in the lateral intraparietal area (LIP) while monkeys switched between a direction-discrimination task and a depth-discrimination task. Ketamine impaired choice accuracy for incongruent stimuli that required different decisions depending on the task, for the direction-discrimination task. Neural sensitivity to irrelevant depth information increased with ketamine during direction discrimination in LIP, indicating impaired processing of irrelevant information. Furthermore, the onset of decision-related neural activity was delayed in conjunction with an increased reaction time irrespective of task and stimulus congruency. Neural sensitivity and response onset of the middle temporal area (MT) were not modulated by ketamine, indicating that ketamine worked on neural decision processes downstream of MT. These results suggest that ketamine administration may impair what information to process and when to process it for the purpose of achieving flexible decision making.
Collapse
Affiliation(s)
- Yuki Suda
- Department of Integrative Physiology, Graduate School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan.,Brain Science Institute, Tamagawa University, 6-1-1 Tamagawagakuen, Machida, Tokyo, 194-8610, Japan.,Department of Neurophysiology, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan
| | - Takanori Uka
- Department of Integrative Physiology, Graduate School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan. .,Brain Science Institute, Tamagawa University, 6-1-1 Tamagawagakuen, Machida, Tokyo, 194-8610, Japan. .,Department of Neurophysiology, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan.
| |
Collapse
|
11
|
Gärtner M, Weigand A, Scheidegger M, Lehmann M, Wyss PO, Wunder A, Henning A, Grimm S. Acute effects of ketamine on the pregenual anterior cingulate: linking spontaneous activation, functional connectivity, and glutamate metabolism. Eur Arch Psychiatry Clin Neurosci 2022; 272:703-714. [PMID: 35020021 PMCID: PMC9095553 DOI: 10.1007/s00406-021-01377-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/16/2021] [Indexed: 11/29/2022]
Abstract
Ketamine exerts its rapid antidepressant effects via modulation of the glutamatergic system. While numerous imaging studies have investigated the effects of ketamine on a functional macroscopic brain level, it remains unclear how altered glutamate metabolism and changes in brain function are linked. To shed light on this topic we here conducted a multimodal imaging study in healthy volunteers (N = 23) using resting state fMRI and proton (1H) magnetic resonance spectroscopy (MRS) to investigate linkage between metabolic and functional brain changes induced by ketamine. Subjects were investigated before and during an intravenous ketamine infusion. The MRS voxel was placed in the pregenual anterior cingulate cortex (pgACC), as this region has been repeatedly shown to be involved in ketamine's effects. Our results showed functional connectivity changes from the pgACC to the right frontal pole and anterior mid cingulate cortex (aMCC). Absolute glutamate and glutamine concentrations in the pgACC did not differ significantly from baseline. However, we found that stronger pgACC activation during ketamine was linked to lower glutamine concentration in this region. Furthermore, reduced functional connectivity between pgACC and aMCC was related to increased pgACC activation and reduced glutamine. Our results thereby demonstrate how multimodal investigations in a single brain region could help to advance our understanding of the association between metabolic and functional changes.
Collapse
Affiliation(s)
- Matti Gärtner
- MSB Medical School Berlin, Rüdesheimer Straße 50, 14197, Berlin, Germany. .,Department of Psychiatry and Psychotherapy, Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Hindenburgdamm 30, 12203, Berlin, Germany.
| | - Anne Weigand
- grid.466457.20000 0004 1794 7698MSB Medical School Berlin, Rüdesheimer Straße 50, 14197 Berlin, Germany
| | - Milan Scheidegger
- grid.7400.30000 0004 1937 0650Department of Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry, University of Zurich, Zurich, Switzerland
| | - Mick Lehmann
- grid.7400.30000 0004 1937 0650Department of Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry, University of Zurich, Zurich, Switzerland
| | - Patrik O. Wyss
- grid.419769.40000 0004 0627 6016Department of Radiology, Swiss Paraplegic Centre, Nottwil, Switzerland
| | - Andreas Wunder
- grid.420061.10000 0001 2171 7500Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riss, Germany
| | - Anke Henning
- grid.267313.20000 0000 9482 7121Advanced Imaging Research Center, UT Southwestern Medical Center, Dallas, TX USA
| | - Simone Grimm
- grid.466457.20000 0004 1794 7698MSB Medical School Berlin, Rüdesheimer Straße 50, 14197 Berlin, Germany ,grid.6363.00000 0001 2218 4662Department of Psychiatry and Psychotherapy, Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany ,grid.7400.30000 0004 1937 0650Department of Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry, University of Zurich, Zurich, Switzerland
| |
Collapse
|
12
|
Zoccatelli G, Alessandrini F, Rimondo C, Beltramello A, Serpelloni G, M Ciceri EF. Magnetic Resonance Spectroscopy in Adolescent Cannabis Users: Metabolites in the Anterior Cingulate Cortex Reflects Individual Differences in Personality Traits and can Affect Rehabilitation Compliance. Neurol India 2021; 68:640-647. [PMID: 32643678 DOI: 10.4103/0028-3886.288984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Introduction The anterior cingulate cortex (ACC) has shown to play a role in impulsivity, fear, and anxiety. Considering, its high glutamate receptor density, it was chosen as a region of interest to investigate the role of glutamate transmission in drug dependance. We investigated the correlations between personality trait scores and glutamate-to-glutamine (Glx) ratio concentrations in the ACC in order to evaluate if (1) personality traits may increase the probability of drug use and (2) drug use can modify cerebral metabolic pattern contributing to addictive behaviors. Materials and Methods Glx ratio concentrations in the ACC region were measured with high-resolution multivoxel proton magnetic resonance spectroscopy (1H-MRS). Personality traits were evaluated utilizing Cloninger's TCI-revised test. Bivariate correlations between personality scores of 28 teens cannabis users (males, mean age = 18.54 ± 2.80) were evaluated. Results In the ACC, we observed negative correlation between GG concentrations (r = -0.44, P = 0.05) and co-operativeness values (CO), choline (cho), and novelty seeking (NS) values (r = -0,45, P = 0.05). Low levels of glutamate and high levels of cho in the ACC were closely related to the CO and NS personality traits. Conclusions Metabolic and personality patterns seems to be related to the risk of substance predisposition in adolescents. Our data contribute a possible support to the "top-down" control of the ACC on brain metabolism, due to the particular cerebral metabolic pattern found in "drug-using" adolescents.
Collapse
Affiliation(s)
- Giada Zoccatelli
- Department of Diagnostics and Pathology, University Hospital of Verona, Verona, Italy
| | - Franco Alessandrini
- Department of Diagnostics and Pathology, University Hospital of Verona, Verona, Italy
| | - Claudia Rimondo
- Italian Early Warning System on Drugs, Presidency of the Council of Ministers, Rome, Italy
| | | | - Giovanni Serpelloni
- Department of Psychiatry, College of Medicine, University of Florida- Drug Policy Institute, Gainesville, FL, United States
| | - Elisa F M Ciceri
- Department of Diagnostics and Pathology, University Hospital of Verona, Verona; IRCCS Fondazione Istituto Neurologico "C.Besta", Milan, Italy
| |
Collapse
|
13
|
Kraguljac NV, Lahti AC. Neuroimaging as a Window Into the Pathophysiological Mechanisms of Schizophrenia. Front Psychiatry 2021; 12:613764. [PMID: 33776813 PMCID: PMC7991588 DOI: 10.3389/fpsyt.2021.613764] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 02/15/2021] [Indexed: 12/16/2022] Open
Abstract
Schizophrenia is a complex neuropsychiatric disorder with a diverse clinical phenotype that has a substantial personal and public health burden. To advance the mechanistic understanding of the illness, neuroimaging can be utilized to capture different aspects of brain pathology in vivo, including brain structural integrity deficits, functional dysconnectivity, and altered neurotransmitter systems. In this review, we consider a number of key scientific questions relevant in the context of neuroimaging studies aimed at unraveling the pathophysiology of schizophrenia and take the opportunity to reflect on our progress toward advancing the mechanistic understanding of the illness. Our data is congruent with the idea that the brain is fundamentally affected in the illness, where widespread structural gray and white matter involvement, functionally abnormal cortical and subcortical information processing, and neurometabolic dysregulation are present in patients. Importantly, certain brain circuits appear preferentially affected and subtle abnormalities are already evident in first episode psychosis patients. We also demonstrated that brain circuitry alterations are clinically relevant by showing that these pathological signatures can be leveraged for predicting subsequent response to antipsychotic treatment. Interestingly, dopamine D2 receptor blockers alleviate neural abnormalities to some extent. Taken together, it is highly unlikely that the pathogenesis of schizophrenia is uniform, it is more plausible that there may be multiple different etiologies that converge to the behavioral phenotype of schizophrenia. Our data underscore that mechanistically oriented neuroimaging studies must take non-specific factors such as antipsychotic drug exposure or illness chronicity into consideration when interpreting disease signatures, as a clear characterization of primary pathophysiological processes is an imperative prerequisite for rational drug development and for alleviating disease burden in our patients.
Collapse
Affiliation(s)
- Nina Vanessa Kraguljac
- Neuroimaging and Translational Research Laboratory, Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Adrienne Carol Lahti
- Neuroimaging and Translational Research Laboratory, Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
14
|
Hori Y, Schaeffer DJ, Gilbert KM, Hayrynen LK, Cléry JC, Gati JS, Menon RS, Everling S. Altered Resting-State Functional Connectivity Between Awake and Isoflurane Anesthetized Marmosets. Cereb Cortex 2020; 30:5943-5959. [PMID: 32556184 PMCID: PMC7899065 DOI: 10.1093/cercor/bhaa168] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/28/2020] [Accepted: 05/28/2020] [Indexed: 01/02/2023] Open
Abstract
The common marmoset (Callithrix jacchus) is a New World primate that is becoming increasingly popular as a preclinical model. To assess functional connectivity (FC) across the marmoset brain, resting-state functional MRI (RS-fMRI) is often performed under isoflurane anesthesia to avoid the effects of motion, physiological stress, and training requirements. In marmosets, however, it remains unclear how isoflurane anesthesia affects patterns of FC. Here, we investigated the effects of isoflurane on FC when delivered with either medical air or 100% pure oxygen, two canonical methods of inhalant isoflurane anesthesia delivery. The results demonstrated that when delivered with either medical air or 100% oxygen, isoflurane globally decreased FC across resting-state networks that were identified in awake marmosets. Generally, although isoflurane globally decreased FC in resting-state networks, the spatial structure of the networks was preserved. Outside of the context of RS networks, we indexed pair-wise functional connectivity between regions across the brain and found that isoflurane substantially altered interhemispheric and thalamic FC. Taken together, these findings indicate that RS-fMRI under isoflurane anesthesia is useful to evaluate the global structure of functional networks, but may obfuscate important nodes of some network components when compared to data acquired in fully awake marmosets.
Collapse
Affiliation(s)
- Yuki Hori
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - David J Schaeffer
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Kyle M Gilbert
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Lauren K Hayrynen
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Justine C Cléry
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Joseph S Gati
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Ravi S Menon
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5B7, Canada
| | - Stefan Everling
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5B7, Canada
- Department of Physiology and Pharmacology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| |
Collapse
|
15
|
Pharmacodynamic elucidation of glutamate & dopamine in ketamine-induced anaesthesia. Chem Biol Interact 2020; 327:109164. [PMID: 32524992 DOI: 10.1016/j.cbi.2020.109164] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/26/2020] [Accepted: 06/05/2020] [Indexed: 12/30/2022]
Abstract
General anaesthetics are some of the most widely used and essential therapeutic agents. However, despite over a century of research, the molecular mechanisms of general anaesthesia in the central nervous system remain elusive. Ketamine (ketamine hydrochloride) has been approved for use in general anaesthesia either alone or in combination with other medications. It is a superb drug for use in short-term medical procedures that do not require skeletal muscle relaxation, and it has approval for the induction of general anaesthesia as a pre-anaesthetic to other general anaesthetic agents. However, Several questions remain unsolved, including the exact identification of the neural substrate of consciousness and its components, the pharmacodynamic interactions between anaesthetic agents, the mechanisms of cognitive alterations that follow an anaesthetic procedure, the identification of an eventual unitary mechanism of anaesthesia-induced alteration of consciousness, the relationship between network effects and the biochemical targets of anaesthetic agents, leading to difficulties in between-studies comparisons. Thus, the glutamate and dopamine systems play distinct roles in terms of neuronal signalling, yet both have proposed to contribute significantly to the pathophysiology of neuropsychiatric diseases. Imaging of the glutamate system and other aspects of research on the dopamine system have produced less consistent findings, potentially due to methodological limitations and the heterogeneity of the disorder. In this review, we discuss the neural circuits through which the two systems interact and how their disruption may cause psychotic symptoms. We also summarize from a molecular perspective of mechanisms of action of ketamine as general anaesthetics on ligand-gated ion channels mediated modulation of dopamine in the brain region.
Collapse
|
16
|
Beck K, Hindley G, Borgan F, Ginestet C, McCutcheon R, Brugger S, Driesen N, Ranganathan M, D’Souza DC, Taylor M, Krystal JH, Howes OD. Association of Ketamine With Psychiatric Symptoms and Implications for Its Therapeutic Use and for Understanding Schizophrenia: A Systematic Review and Meta-analysis. JAMA Netw Open 2020; 3:e204693. [PMID: 32437573 PMCID: PMC7243091 DOI: 10.1001/jamanetworkopen.2020.4693] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/29/2020] [Indexed: 12/16/2022] Open
Abstract
Importance Ketamine hydrochloride is increasingly used to treat depression and other psychiatric disorders but can induce schizophrenia-like or psychotomimetic symptoms. Despite this risk, the consistency and magnitude of symptoms induced by ketamine or what factors are associated with these symptoms remain unknown. Objective To conduct a meta-analysis of the psychopathological outcomes associated with ketamine in healthy volunteers and patients with schizophrenia and the experimental factors associated with these outcomes. Data Sources MEDLINE, Embase, and PsychINFO databases were searched for within-participant, placebo-controlled studies reporting symptoms using the Brief Psychiatric Rating Scale (BPRS) or the Positive and Negative Syndrome Scale (PANSS) in response to an acute ketamine challenge in healthy participants or patients with schizophrenia. Study Selection Of 8464 citations retrieved, 36 studies involving healthy participants were included. Inclusion criteria were studies (1) including healthy participants; (2) reporting symptoms occurring in response to acute administration of subanesthetic doses of ketamine (racemic ketamine, s-ketamine, r-ketamine) intravenously; (3) containing a placebo condition with a within-subject, crossover design; (4) measuring total positive or negative symptoms using BPRS or PANSS; and (5) providing data allowing the estimation of the mean difference and deviation between the ketamine and placebo condition. Data Extraction and Synthesis Two independent investigators extracted study-level data for a random-effects meta-analysis. Total, positive, and negative BPRS and PANSS scores were extracted. Subgroup analyses were conducted examining the effects of blinding status, ketamine preparation, infusion method, and time between ketamine and placebo conditions. The Meta-analysis of Observational Studies in Epidemiology (MOOSE) and Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines were followed. Main Outcomes and Measures Standardized mean differences (SMDs) were used as effect sizes for individual studies. Standardized mean differences between ketamine and placebo conditions were calculated for total, positive, and negative BPRS and PANSS scores. Results The overall sample included 725 healthy volunteers (mean [SD] age, 28.3 [3.6] years; 533 [73.6%] male) exposed to the ketamine and placebo conditions. Racemic ketamine or S-ketamine was associated with a statistically significant increase in transient psychopathology in healthy participants for total (SMD = 1.50 [95% CI, 1.23-1.77]; P < .001), positive (SMD = 1.55 [95% CI, 1.29-1.81]; P < .001), and negative (SMD = 1.16 [95% CI, 0.96-1.35]; P < .001) symptom ratings relative to the placebo condition. The effect size for this association was significantly greater for positive than negative symptoms of psychosis (estimate, 0.36 [95% CI, 0.12-0.61]; P = .004). There was significant inconsistency in outcomes between studies (I2 range, 77%-83%). Bolus followed by constant infusion increased ketamine's association with positive symptoms relative to infusion alone (effect size, 1.63 [95% CI, 1.36-1.90] vs 0.84 [95% CI, 0.35-1.33]; P = .006). Single-day study design increased ketamine's ability to generate total symptoms (effect size, 2.29 [95% CI, 1.69-2.89] vs 1.39 [95% CI, 1.12-1.66]; P = .007), but age and sex did not moderate outcomes. Insufficient studies were available for meta-analysis of studies in schizophrenia. Of these studies, 2 found a statistically significant increase in symptoms with ketamine administration in total and positive symptoms. Only 1 study found an increase in negative symptom severity with ketamine. Conclusions and Relevance This study found that acute ketamine administration was associated with schizophrenia-like or psychotomimetic symptoms with large effect sizes, but there was a greater increase in positive than negative symptoms and when a bolus was used. These findings suggest that bolus doses should be avoided in the therapeutic use of ketamine to minimize the risk of inducing transient positive (psychotic) symptoms.
Collapse
Affiliation(s)
- Katherine Beck
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Psychiatric Imaging Group, MRC (Medical Research Council) London Institute of Medical Sciences, Hammersmith Hospital, London, United Kingdom
- South London and Maudsley NHS (National Health Service) Foundation Trust, London, United Kingdom
| | - Guy Hindley
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Faith Borgan
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Cedric Ginestet
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Robert McCutcheon
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Psychiatric Imaging Group, MRC (Medical Research Council) London Institute of Medical Sciences, Hammersmith Hospital, London, United Kingdom
- South London and Maudsley NHS (National Health Service) Foundation Trust, London, United Kingdom
| | - Stefan Brugger
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Division of Psychiatry, University College London, London, United Kingdom
- Cardiff University Brain Research Imaging Centre, Cardiff, United Kingdom
| | - Naomi Driesen
- Yale University Medical School, Veterans Affairs Connecticut Health Care System, West Haven
| | - Mohini Ranganathan
- Yale University Medical School, Veterans Affairs Connecticut Health Care System, West Haven
- Department of Psychiatry and National Center for Posttraumatic Stress Disorder (PTSD), Veterans Affairs Connecticut Healthcare System, West Haven
| | - Deepak Cyril D’Souza
- Yale University Medical School, Veterans Affairs Connecticut Health Care System, West Haven
- Department of Psychiatry and National Center for Posttraumatic Stress Disorder (PTSD), Veterans Affairs Connecticut Healthcare System, West Haven
| | - Matthew Taylor
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- University Department of Psychiatry, Warneford Hospital, Oxford, United Kingdom
| | - John H. Krystal
- Yale University Medical School, Veterans Affairs Connecticut Health Care System, West Haven
- Department of Veteran Affairs National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, Veterans Affairs Connecticut Healthcare System, West Haven
| | - Oliver D. Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Psychiatric Imaging Group, MRC (Medical Research Council) London Institute of Medical Sciences, Hammersmith Hospital, London, United Kingdom
- South London and Maudsley NHS (National Health Service) Foundation Trust, London, United Kingdom
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
17
|
Vlerick L, Devreese M, Peremans K, Dockx R, Croubels S, Duchateau L, Polis I. Pharmacokinetics, absolute bioavailability and tolerability of ketamine after intranasal administration to dexmedetomidine sedated dogs. PLoS One 2020; 15:e0227762. [PMID: 31929589 PMCID: PMC6957157 DOI: 10.1371/journal.pone.0227762] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 12/28/2019] [Indexed: 01/13/2023] Open
Abstract
Intranasal ketamine has recently gained interest in human medicine, not only for its sedative, anaesthetic or analgesic properties, but also in the management of treatment resistant depression, where it has been shown to be an effective, fast acting alternative treatment. Since several similarities are reported between human psychiatric disorders and canine anxiety disorders, intranasal ketamine could serve as an alternative treatment for anxiety disordered dogs. However, to the authors knowledge, intranasal administration of ketamine and its pharmacokinetics have never been described in dogs. Therefore, this study aimed to examine the pharmacokinetics, absolute bioavailability and tolerability of intranasal ketamine administration compared with intravenous administration. Seven healthy, adult laboratory Beagle dogs were included in this randomized crossover study. The dogs received 2 mg/kg body weight ketamine intravenously (IV) or intranasally (IN), with a two-week wash-out period. Prior to ketamine administration, dogs were sedated intramuscularly with dexmedetomidine. Venous blood samples were collected at fixed times until 480 min post-administration and ketamine plasma concentrations were determined by liquid chromatography-tandem mass spectrometry. Cardiovascular parameters and sedation scores were recorded at the same time points. Non-compartmental pharmacokinetic analysis revealed a rapid (Tmax = 0.25 ± 0.14 h) and complete IN bioavailability (F = 147.65 ± 49.97%). Elimination half-life was similar between both administration routes (T1/2el IV = 1.47 ± 0.24 h, T1/2el IN = 1.50 ± 0.97 h). Heart rate and sedation scores were significantly higher at 5 and 10 min following IV administration compared to IN administration, but not at the later time-points.
Collapse
Affiliation(s)
- Lise Vlerick
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- * E-mail:
| | - Mathias Devreese
- Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Kathelijne Peremans
- Department of Veterinary Medical Imaging and Small Animal Orthopaedics, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Robrecht Dockx
- Department of Veterinary Medical Imaging and Small Animal Orthopaedics, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- Department of Psychiatry and Medical Psychology, Ghent Experimental Psychiatry (GHEP) lab, Ghent University, Ghent, Belgium
| | - Siska Croubels
- Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Luc Duchateau
- Biometrics Research Centre, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Ingeborgh Polis
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
18
|
Bryant JE, Frölich M, Tran S, Reid MA, Lahti AC, Kraguljac NV. Ketamine induced changes in regional cerebral blood flow, interregional connectivity patterns, and glutamate metabolism. J Psychiatr Res 2019; 117:108-115. [PMID: 31376621 PMCID: PMC7291620 DOI: 10.1016/j.jpsychires.2019.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/19/2019] [Accepted: 07/26/2019] [Indexed: 12/31/2022]
Abstract
Several imaging studies have attempted to characterize the contribution of glutamatergic dysfunction to functional dysconnectivity of large-scale brain networks using ketamine models. However, findings from BOLD imaging studies are conflicting, in part because the signal stems from a complex interaction between blood flow, blood volume, and oxygen consumption. We used arterial spin labelling imaging to measure regional cerebral blood flow (rCBF) in a group of healthy volunteers during a saline and during a ketamine infusion. We examined changes in rCBF and interregional connectivity patterns, as well as their associations with clinical symptom severity and Glx (glutamate + glutamine) assessed with magnetic resonance spectroscopy. We report a regionally selective pattern of rCBF changes following ketamine administration and complex changes in interregional connectivity patterns. We also found that the increase in rCBF in the bilateral putamen and left hippocampus was positively correlated with ketamine induced clinical symptom severity while anterior cingulate rCBF during the ketamine challenge was negatively correlated with change in hippocampal Glx. Our study adds to the efforts to empirically confirm putative links between an NMDA receptor blockage and dysconnectivity of large-scale brain networks, specifically the salience, executive control and default mode networks, suggesting that a glutamatergic imbalance may contribute to dysconnectivity. Development of glutamatergic compounds that alleviate disease burden, possibly through normalizing glutamate excess related increased rCBF, is direly needed.
Collapse
Affiliation(s)
- James Edward Bryant
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, United States
| | - Michael Frölich
- Department of Anesthesiology, University of Alabama at Birmingham, United States
| | - Steve Tran
- Department of Anesthesiology, University of Alabama at Birmingham, United States
| | - Meredith Amanda Reid
- MRI Research Center, Department of Electrical and Computer Engineering, Auburn University, United States
| | - Adrienne Carol Lahti
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, United States
| | - Nina Vanessa Kraguljac
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, United States.
| |
Collapse
|
19
|
The effects of sub-anesthetic ketamine plus ethanol on behaviors and apoptosis in the prefrontal cortex and hippocampus of adolescent rats. Pharmacol Biochem Behav 2019; 184:172742. [PMID: 31348944 DOI: 10.1016/j.pbb.2019.172742] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 07/10/2019] [Accepted: 07/22/2019] [Indexed: 12/14/2022]
Abstract
Ketamine has become increasingly popular in adolescent drug abusers worldwide. Meanwhile, alcohol is usually used by ketamine users. However, little work has been conducted to examine the chronic combined effects of ketamine and ethanol on adolescent brain. Here we probed into the effects of chronic administration of ketamine at recreational doses alone or combined with ethanol on behaviors and neuron damage in an adolescent rat model. 28-day old rats were treated with either 20 or 30 mg/kg ketamine plus or not plus 10% ethanol daily for 21 days. Depressive like behaviors, anxiety like behavior and memory impairment were tested using open field test, forced swimming test, elevated plus maze and Morris water maze. Apoptosis in prefrontal cortex (PFC) and hippocampus (HIP) were determined by the TdT-mediated dUTP Nick-End Labeling (TUNEL) and protein and mRNA levels of caspase-3, Bax and Bcl-2. Results show that co-application of ketamine and ethanol significantly increased immobility time in the forced swimming test, up-regulated TUNEL positive cells and both protein and mRNA expressions of caspase-3 and Bax, compared with the control group and ketamine and ethanol use alone groups in the PFC, but not in the HIP. Our study suggests that chronic co-administration of ketamine and ethanol results in depressive-like behavior and the caspase-dependent apoptosis in the PFC of adolescent rats' brains.
Collapse
|
20
|
Uno Y, Coyle JT. Glutamate hypothesis in schizophrenia. Psychiatry Clin Neurosci 2019; 73:204-215. [PMID: 30666759 DOI: 10.1111/pcn.12823] [Citation(s) in RCA: 225] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/08/2019] [Accepted: 01/16/2019] [Indexed: 12/13/2022]
Abstract
Schizophrenia is a chronic and severe psychiatric disorder that has profound impact on an individual's life and on society. Thus, developing more effective therapeutic interventions is essential. Over the past quarter-century, an abundance of evidence from pharmacologic challenges, post-mortem studies, brain imaging, and genetic studies supports the role of glutamatergic dysregulation in the pathophysiology of schizophrenia, and the results of recent randomized clinical trials based on this evidence have yielded promising results. In this article, we review the evidence that alterations in glutamatergic neurotransmission, especially focusing on the N-methyl-d-aspartate receptor (NMDAR) function, may be a critical causative feature of schizophrenia, how this contributes to pathologic circuit function in the brain, and how these insights are revealing whole new avenues for treatment development that could reduce treatment-resistant symptoms, which account for persistent disability.
Collapse
Affiliation(s)
- Yota Uno
- Department of Psychiatry, Harvard Medical School, Boston, USA.,Laboratory for Psychiatric and Molecular Neuroscience, McLean Hospital, Belmont, USA.,Department of Psychology, University of Bath, Bath, UK
| | - Joseph T Coyle
- Department of Psychiatry, Harvard Medical School, Boston, USA.,Laboratory for Psychiatric and Molecular Neuroscience, McLean Hospital, Belmont, USA
| |
Collapse
|
21
|
Vlerick L, Peremans K, Dockx R, Audenaert K, Baeken C, Saunders JH, Polis I. The long-term effects of single and repeated subanaesthetic ketamine administration on regional cerebral blood flow in healthy dogs measured with 99mTc-HMPAO SPECT. Psychiatry Res Neuroimaging 2019; 285:18-24. [PMID: 30716686 DOI: 10.1016/j.pscychresns.2019.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 01/15/2019] [Accepted: 01/15/2019] [Indexed: 12/13/2022]
Abstract
Subanaesthetic ketamine has recently been established as an effective and rapid treatment for major depressive disorder showing antidepressant effects for up to 1 week on average. The use of repeated ketamine infusions has been put forward to augment and to prolong the antidepressant response and increase the remission rates. The underlying neurobiological mechanisms responsible for ketamine's antidepressant effects remain unclear. Nevertheless, it has been shown, both in dogs and humans, that ketamine can alter neuronal perfusion and therefore neuronal function in brain regions involved in psychiatric and behavioural disorders. Consequently, the aim of the current placebo controlled study was to assess the long-term effects on cerebral perfusion of single and repeated subanaesthetic ketamine infusions in dogs. Twelve healthy, laboratory dogs were scanned at six different time points following single and repeated ketamine administration, using Single Photon Emission Computed Tomography with the radiotracer 99mTc-hexamethylpropylene amine oxime. We hypothesised that repeated infusions could lead to more prolonged perfusion alterations in brain regions critical for behaviour regulation. We found that repeated subanaesthetic ketamine administration did not result in more prolonged cerebral perfusion alterations compared to a single ketamine administration.
Collapse
Affiliation(s)
- Lise Vlerick
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, East Flanders, Belgium.
| | - Kathelijne Peremans
- Department of Veterinary Medical Imaging and Small Animal Orthopaedics, Faculty of Veterinary Medicine, Ghent University, Merelbeke, East Flanders, Belgium
| | - Robrecht Dockx
- Ghent Experimental Psychiatry (GHEP) lab, Department of Psychiatry and Medical Psychology, Ghent University, Ghent, East Flanders, Belgium; Department of Psychiatry, University Hospital (UZBrussel), Brussels, Belgium
| | - Kurt Audenaert
- Ghent Experimental Psychiatry (GHEP) lab, Department of Psychiatry and Medical Psychology, Ghent University, Ghent, East Flanders, Belgium; Department of Psychiatry, University Hospital (UZBrussel), Brussels, Belgium
| | - Chris Baeken
- Ghent Experimental Psychiatry (GHEP) lab, Department of Psychiatry and Medical Psychology, Ghent University, Ghent, East Flanders, Belgium; Department of Psychiatry, University Hospital (UZBrussel), Brussels, Belgium
| | - Jimmy H Saunders
- Department of Veterinary Medical Imaging and Small Animal Orthopaedics, Faculty of Veterinary Medicine, Ghent University, Merelbeke, East Flanders, Belgium
| | - Ingeborgh Polis
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, East Flanders, Belgium
| |
Collapse
|
22
|
Vlerick L, Peremans K, Dockx R, Audenaert K, Baeken C, De Spiegeleer B, Saunders J, Polis I. The influence of subanaesthetic ketamine on regional cerebral blood flow in healthy dogs measured with 99mTc-HMPAO SPECT. PLoS One 2018; 13:e0209316. [PMID: 30562399 PMCID: PMC6298672 DOI: 10.1371/journal.pone.0209316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 12/04/2018] [Indexed: 02/06/2023] Open
Abstract
Subanaesthetic ketamine has recently been proven to be a highly effective and fast acting alternative treatment for several psychiatric disorders. The mechanisms responsible for ketamine's antidepressant effects remain unclear, but a possible explanation could be that ketamine interacts with regional cerebral blood flow (rCBF). Therefore, the effects of two subanaesthetic ketamine doses on rCBF were evaluated. Twelve dogs were randomly assigned to one of the three treatment conditions (condition saline, condition 0.5 mg/kg ketamine or condition 2 mg/kg ketamine) and received in total five saline or ketamine infusions, with one week interval. Single Photon Emission Computed Tomography (SPECT) scans with the radiotracer 99mTc-hexamethylpropylene amine oxime were performed before the start of the infusions (baseline) and 24 hours after the first (single) and last (multiple) infusion. After a wash out period of 3 months, the animals were again assigned to one of the three treatment conditions described above and the infusion/scan protocol was repeated. During the infusions, cardiovascular parameters were evaluated every ten minutes. A one-way repeated measure ANOVA was set up to assess perfusion index for each ketamine dose for the left frontal cortex (alpha = 0.05). The remaining 11 brain regions were post hoc assessed. Perfusion index was significantly increased in the left frontal cortex and in the thalamus 24 hours after single and multiple ketamine infusions compared to baseline in the 2 mg/kg condition. No clinically relevant cardiovascular effects were observed during the ketamine infusions. This study shows that subanaesthetic ketamine can increase neuronal perfusion and therefore alter neuronal function in brain regions involved in depression and anxiety disorders. These perfusion increases may possibly contribute to ketamine's beneficial effects in these psychiatric disorders.
Collapse
Affiliation(s)
- Lise Vlerick
- Department of Small Animal, Faculty of Veterinary Medicine, Ghent University, Merelbeke, East Flanders, Belgium
| | - Kathelijne Peremans
- Department of Veterinary Medical Imaging and Small Animal Orthopaedics, Faculty of Veterinary Medicine, Ghent University, Merelbeke, East Flanders, Belgium
| | - Robrecht Dockx
- Ghent Experimental Psychiatry (GHEP) lab, Department of Psychiatry and Medical Psychology, Ghent University, Ghent, East Flanders, Belgium
| | - Kurt Audenaert
- Ghent Experimental Psychiatry (GHEP) lab, Department of Psychiatry and Medical Psychology, Ghent University, Ghent, East Flanders, Belgium
| | - Chris Baeken
- Ghent Experimental Psychiatry (GHEP) lab, Department of Psychiatry and Medical Psychology, Ghent University, Ghent, East Flanders, Belgium
| | - Bart De Spiegeleer
- Drug Quality and Registration (DruQuaR) group, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, East Flanders, Belgium
| | - Jimmy Saunders
- Department of Veterinary Medical Imaging and Small Animal Orthopaedics, Faculty of Veterinary Medicine, Ghent University, Merelbeke, East Flanders, Belgium
| | - Ingeborgh Polis
- Department of Small Animal, Faculty of Veterinary Medicine, Ghent University, Merelbeke, East Flanders, Belgium
| |
Collapse
|
23
|
Wang X, Pinto-Duarte A, Behrens MM, Zhou X, Sejnowski TJ. Ketamine independently modulated power and phase-coupling of theta oscillations in Sp4 hypomorphic mice. PLoS One 2018. [PMID: 29513708 PMCID: PMC5841791 DOI: 10.1371/journal.pone.0193446] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Reduced expression of Sp4, the murine homolog of human SP4, a risk gene of multiple psychiatric disorders, led to N-methyl-D-aspartate (NMDA) hypofunction in mice, producing behavioral phenotypes reminiscent of schizophrenia, including hypersensitivity to ketamine. As accumulating evidence on molecular mechanisms and behavioral phenotypes established Sp4 hypomorphism as a promising animal model, systems-level neural circuit mechanisms of Sp4 hypomorphism, especially network dynamics underlying cognitive functions, remain poorly understood. We attempted to close this gap in knowledge in the present study by recording multi-channel epidural electroencephalogram (EEG) from awake behaving wildtype and Sp4 hypomorphic mice. We characterized cortical theta-band power and phase-coupling phenotypes, a known neural circuit substrate underlying cognitive functions, and further studied the effects of a subanesthetic dosage of ketamine on theta abnormalities unique to Sp4 hypomorphism. Sp4 hypomorphic mice had markedly elevated theta power localized frontally and parietally, a more pronounced theta phase progression along the neuraxis, and a stronger frontal-parietal theta coupling. Acute subanesthetic ketamine did not affect theta power in wildtype animals but significantly reduced it in Sp4 hypomorphic mice, nearly completely neutralizing their excessive frontal/parietal theta power. Ketamine did not significantly alter cortical theta phase progression in either wildtype or Sp4 hypomorphic animals, but significantly strengthened cortical theta phase-coupling in wildtype, but not in Sp4 hypomorphic animals. Our results suggested that the resting-state phenotypes of cortical theta oscillations unique to Sp4 hypomorphic mice closely mimicked a schizophrenic endophenotype. Further, ketamine independently modulated Sp4 hypomorphic anomalies in theta power and phase-coupling, suggesting separate underlying neural circuit mechanisms.
Collapse
Affiliation(s)
- Xin Wang
- Howard Hughes Medical Institute, the Salk Institute for Biological Studies, La Jolla, California, United States of America
- * E-mail:
| | - António Pinto-Duarte
- Howard Hughes Medical Institute, the Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - M. Margarita Behrens
- Howard Hughes Medical Institute, the Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Xianjin Zhou
- Department of Psychiatry, University of California at San Diego, La Jolla, California, United States of America
| | - Terrence J. Sejnowski
- Howard Hughes Medical Institute, the Salk Institute for Biological Studies, La Jolla, California, United States of America
- Division of Biology, University of California at San Diego, La Jolla, California, United States of America
| |
Collapse
|
24
|
Vutskits L. General Anesthetics to Treat Major Depressive Disorder: Clinical Relevance and Underlying Mechanisms. Anesth Analg 2018; 126:208-216. [PMID: 29135596 DOI: 10.1213/ane.0000000000002594] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Major depressive disorder is a frequent and devastating psychological condition with tremendous public health impact. The underlying pathophysiological mechanisms involve abnormal neurotransmission and a relatedly impaired synaptic plasticity. Since general anesthetics are potent modulators of neuronal activity and, thereby, can exert long-term context-dependent impact on neural networks, an intriguing hypothesis is that these drugs could enhance impaired neural plasticity associated with certain psychiatric diseases. Clinical observations over the past few decades appear to confirm this possibility. Indeed, equipotency of general anesthesia alone in comparison with electroconvulsive therapy under general anesthesia has been demonstrated in several clinical trials. Importantly, in the past 15 years, intravenous administration of subanesthetic doses of ketamine have also been demonstrated to have rapid antidepressant effects. The molecular, cellular, and network mechanisms underlying these therapeutic effects have been partially identified. Although several important questions remain to be addressed, the ensemble of these experimental and clinical observations opens new therapeutic possibilities in the treatment of depressive disorders. Importantly, they also suggest a new therapeutic role for anesthetics that goes beyond their principal use in the perioperative period to facilitate surgery.
Collapse
Affiliation(s)
- Laszlo Vutskits
- From the Department of Anesthesiology, Pharmacology and Intensive Care, University Hospitals of Geneva, Geneva, Switzerland
| |
Collapse
|
25
|
Bojesen KB, Andersen KA, Rasmussen SN, Baandrup L, Madsen LM, Glenthøj BY, Rostrup E, Broberg BV. Glutamate Levels and Resting Cerebral Blood Flow in Anterior Cingulate Cortex Are Associated at Rest and Immediately Following Infusion of S-Ketamine in Healthy Volunteers. Front Psychiatry 2018; 9:22. [PMID: 29467681 PMCID: PMC5808203 DOI: 10.3389/fpsyt.2018.00022] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 01/19/2018] [Indexed: 11/15/2022] Open
Abstract
Progressive loss of brain tissue is seen in some patients with schizophrenia and might be caused by increased levels of glutamate and resting cerebral blood flow (rCBF) alterations. Animal studies suggest that the normalisation of glutamate levels decreases rCBF and prevents structural changes in hippocampus. However, the relationship between glutamate and rCBF in anterior cingulate cortex (ACC) of humans has not been studied in the absence of antipsychotics and illness chronicity. Ketamine is a noncompetitive N-methyl-D-aspartate receptor antagonist that transiently induces schizophrenia-like symptoms and neurobiological disturbances in healthy volunteers (HVs). Here, we used S-ketamine challenge to assess if glutamate levels were associated with rCBF in ACC in 25 male HVs. Second, we explored if S-ketamine changed the neural activity as reflected by rCBF alterations in thalamus (Thal) and accumbens that are connected with ACC. Glutamatergic metabolites were measured in ACC with magnetic resonance (MR) spectroscopy and whole-brain rCBF with pseudo-continuous arterial spin labelling on a 3-T MR scanner before, during, and after infusion of S-ketamine (total dose 0.375 mg/kg). In ACC, glutamate levels were associated with rCBF before (p < 0.05) and immediately following S-ketamine infusion (p = 0.03), but not during and after. S-Ketamine increased rCBF in ACC (p < 0.001) but not the levels of glutamate (p = 0.96). In subcortical regions, S-ketamine altered rCBF in left Thal (p = 0.03). Our results suggest that glutamate levels in ACC are associated with rCBF at rest and in the initial phase of an increase. Furthermore, S-ketamine challenge transiently induces abnormal activation of ACC and left Thal that both are implicated in the pathophysiology of schizophrenia. Future longitudinal studies should investigate if increased glutamate and rCBF are related to the progressive loss of brain tissue in initially first-episode patients.
Collapse
Affiliation(s)
- Kirsten Borup Bojesen
- Centre for Neuropsychiatric Schizophrenia Research (CNSR), Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kasper Aagaard Andersen
- Centre for Neuropsychiatric Schizophrenia Research (CNSR), Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet Glostrup, University of Copenhagen, Copenhagen, Denmark
| | - Sophie Nordahl Rasmussen
- Centre for Neuropsychiatric Schizophrenia Research (CNSR), Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet Glostrup, University of Copenhagen, Copenhagen, Denmark
| | - Lone Baandrup
- Centre for Neuropsychiatric Schizophrenia Research (CNSR), Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark
| | - Line Malmer Madsen
- Department of Anaesthesia, Glostrup Hospital, University of Copenhagen, Glostrup, Denmark
| | - Birte Yding Glenthøj
- Centre for Neuropsychiatric Schizophrenia Research (CNSR), Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Egill Rostrup
- Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet Glostrup, University of Copenhagen, Copenhagen, Denmark
| | - Brian Villumsen Broberg
- Centre for Neuropsychiatric Schizophrenia Research (CNSR), Centre for Clinical Intervention and Neuropsychiatric Schizophrenia Research (CINS), Mental Health Centre Glostrup, University of Copenhagen, Glostrup, Denmark
| |
Collapse
|
26
|
Phencyclidine Discoordinates Hippocampal Network Activity But Not Place Fields. J Neurosci 2017; 37:12031-12049. [PMID: 29118102 DOI: 10.1523/jneurosci.0630-17.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 09/22/2017] [Accepted: 10/17/2017] [Indexed: 11/21/2022] Open
Abstract
We used the psychotomimetic phencyclidine (PCP) to investigate the relationships among cognitive behavior, coordinated neural network function, and information processing within the hippocampus place cell system. We report in rats that PCP (5 mg/kg, i.p.) impairs a well learned, hippocampus-dependent place avoidance behavior in rats that requires cognitive control even when PCP is injected directly into dorsal hippocampus. PCP increases 60-100 Hz medium-freguency gamma oscillations in hippocampus CA1 and these increases correlate with the cognitive impairment caused by systemic PCP administration. PCP discoordinates theta-modulated medium-frequency and slow gamma oscillations in CA1 LFPs such that medium-frequency gamma oscillations become more theta-organized than slow gamma oscillations. CA1 place cell firing fields are preserved under PCP, but the drug discoordinates the subsecond temporal organization of discharge among place cells. This discoordination causes place cell ensemble representations of a familiar space to cease resembling pre-PCP representations despite preserved place fields. These findings point to the cognitive impairments caused by PCP arising from neural discoordination. PCP disrupts the timing of discharge with respect to the subsecond timescales of theta and gamma oscillations in the LFP. Because these oscillations arise from local inhibitory synaptic activity, these findings point to excitation-inhibition discoordination as the root of PCP-induced cognitive impairment.SIGNIFICANCE STATEMENT Hippocampal neural discharge is temporally coordinated on timescales of theta and gamma oscillations in the LFP and the discharge of a subset of pyramidal neurons called "place cells" is spatially organized such that discharge is restricted to locations called a cell's "place field." Because this temporal coordination and spatial discharge organization is thought to represent spatial knowledge, we used the psychotomimetic phencyclidine (PCP) to disrupt cognitive behavior and assess the importance of neural coordination and place fields for spatial cognition. PCP impaired the judicious use of spatial information and discoordinated hippocampal discharge without disrupting firing fields. These findings dissociate place fields from spatial cognitive behavior and suggest that hippocampus discharge coordination is crucial to spatial cognition.
Collapse
|
27
|
Petrilli MA, Kranz TM, Kleinhaus K, Joe P, Getz M, Johnson P, Chao MV, Malaspina D. The Emerging Role for Zinc in Depression and Psychosis. Front Pharmacol 2017; 8:414. [PMID: 28713269 PMCID: PMC5492454 DOI: 10.3389/fphar.2017.00414] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/13/2017] [Indexed: 12/20/2022] Open
Abstract
Zinc participation is essential for all physiological systems, including neural functioning, where it participates in a myriad of cellular processes. Converging clinical, molecular, and genetic discoveries illuminate key roles for zinc homeostasis in association with clinical depression and psychosis which are not yet well appreciated at the clinical interface. Intracellular deficiency may arise from low circulating zinc levels due to dietary insufficiency, or impaired absorption from aging or medical conditions, including alcoholism. A host of medications commonly administered to psychiatric patients, including anticonvulsants, oral medications for diabetes, hormones, antacids, anti-inflammatories and others also impact zinc absorption. Furthermore, inefficient genetic variants in zinc transporter molecules that transport the ion across cellular membranes impede its action even when circulating zinc concentrations is in the normal range. Well powered clinical studies have shown beneficial effects of supplemental zinc in depression and it important to pursue research using zinc as a potential therapeutic option for psychosis as well. Meta-analyses support the adjunctive use of zinc in major depression and a single study now supports zinc for psychotic symptoms. This manuscript reviews the biochemistry and bench top evidence on putative molecular mechanisms of zinc as a psychiatric treatment.
Collapse
Affiliation(s)
| | - Thorsten M Kranz
- Departments of Cell Biology, Physiology and Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York UniversityNew York, NY, United States
| | - Karine Kleinhaus
- Department of Psychiatry, New York University School of MedicineNew York, NY, United States
| | - Peter Joe
- Department of Psychiatry, New York University School of MedicineNew York, NY, United States
| | - Mara Getz
- Department of Psychiatry, New York University School of MedicineNew York, NY, United States
| | - Porsha Johnson
- Department of Psychiatry, New York University School of MedicineNew York, NY, United States
| | - Moses V Chao
- Departments of Cell Biology, Physiology and Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York UniversityNew York, NY, United States
| | - Dolores Malaspina
- Department of Psychiatry, New York University School of MedicineNew York, NY, United States
| |
Collapse
|
28
|
Mao CV, Araujo MFP, Nishimaru H, Matsumoto J, Tran AH, Hori E, Ono T, Nishijo H. Pregenual Anterior Cingulate Gyrus Involvement in Spontaneous Social Interactions in Primates-Evidence from Behavioral, Pharmacological, Neuropsychiatric, and Neurophysiological Findings. Front Neurosci 2017; 11:34. [PMID: 28203143 PMCID: PMC5285368 DOI: 10.3389/fnins.2017.00034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 01/17/2017] [Indexed: 11/23/2022] Open
Abstract
The anterior cingulate cortex (ACC) has been implicated in different aspects of cognition and decision making, including social cognition. Several studies suggest that this region is actually formed by sub-regions concerned with distinct cognitive functions. The ACC is usually divided in its rostro-caudal axis, with the caudal ACC playing a major role in processing own actions, and the rostral ACC being related to social cognition. Recently, it has been suggested that the ACC can also be functionally divided in its dorso-ventral axis into ACC gyrus (ACCg) and ACC sulcus (ACCs), with the ACCg having a central role in processing social information. In this context, we propose that the pregenual ACCg might be especially important for engaging in social interactions. We discuss previous findings that support this hypothesis and present evidence suggesting that the activity of pregenual ACCg neurons is modulated during spontaneous social interactions.
Collapse
Affiliation(s)
- Can Van Mao
- System Emotional Science, Graduate School of Medicine, University of Toyama Toyama, Japan
| | - Mariana F P Araujo
- System Emotional Science, Graduate School of Medicine, University of ToyamaToyama, Japan; Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont InstituteMacaiba, Brazil
| | - Hiroshi Nishimaru
- System Emotional Science, Graduate School of Medicine, University of Toyama Toyama, Japan
| | - Jumpei Matsumoto
- System Emotional Science, Graduate School of Medicine, University of Toyama Toyama, Japan
| | - Ahn Hai Tran
- System Emotional Science, Graduate School of Medicine, University of Toyama Toyama, Japan
| | - Etsuro Hori
- System Emotional Science, Graduate School of Medicine, University of Toyama Toyama, Japan
| | - Taketoshi Ono
- System Emotional Science, Graduate School of Medicine, University of Toyama Toyama, Japan
| | - Hisao Nishijo
- System Emotional Science, Graduate School of Medicine, University of Toyama Toyama, Japan
| |
Collapse
|
29
|
|
30
|
Brandt AS, Unschuld PG, Pradhan S, Lim IAL, Churchill G, Harris AD, Hua J, Barker PB, Ross CA, van Zijl PCM, Edden RAE, Margolis RL. Age-related changes in anterior cingulate cortex glutamate in schizophrenia: A (1)H MRS Study at 7 Tesla. Schizophr Res 2016; 172:101-5. [PMID: 26925800 PMCID: PMC4821673 DOI: 10.1016/j.schres.2016.02.017] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Revised: 02/08/2016] [Accepted: 02/10/2016] [Indexed: 12/27/2022]
Abstract
The extent of age-related changes in glutamate and other neurometabolites in the anterior cingulate cortex (ACC) in individuals with schizophrenia remain unclear. Magnetic resonance spectroscopy (MRS) at 7 T, which yields precise measurements of various metabolites and can distinguish glutamate from glutamine, was used to determine levels of ACC glutamate and other metabolites in 24 individuals with schizophrenia and 24 matched controls. Multiple regression analysis revealed that ACC glutamate decreased with age in patients but not controls. No changes were detected in levels of glutamine, N-acetylaspartate, N-acetylaspartylglutamic acid, myo-inositol, GABA, glutathione, total creatine, and total choline. These results suggest that age may be an important modifier of ACC glutamate in schizophrenia.
Collapse
Affiliation(s)
- Allison S Brandt
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Paul G Unschuld
- Laboratory for Aging Neuroscience and Neuroimaging, Division of Psychiatry Research and Psychogeriatric Medicine, University of Zürich, Zurich, Switzerland
| | - Subechhya Pradhan
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Issel Anne L Lim
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Gregory Churchill
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ashley D Harris
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Jun Hua
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Peter B Barker
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Christopher A Ross
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology and Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Departments of Pharmacology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter C M van Zijl
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Richard A E Edden
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Russell L Margolis
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology and Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
31
|
Yamamoto H, Hagino Y, Kasai S, Ikeda K. Specific Roles of NMDA Receptor Subunits in Mental Disorders. Curr Mol Med 2016; 15:193-205. [PMID: 25817860 PMCID: PMC5384360 DOI: 10.2174/1566524015666150330142807] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 02/28/2015] [Accepted: 03/24/2015] [Indexed: 12/30/2022]
Abstract
N-methyl-D-aspartate (NMDA) receptor plays important roles in learning and memory. NMDA receptors are a tetramer that consists of two glycine-binding subunits GluN1, two glutamate-binding subunits (i.e., GluN2A, GluN2B, GluN2C, and GluN2D), a combination of a GluN2 subunit and glycine-binding GluN3 subunit (i.e., GluN3A or GluN3B), or two GluN3 subunits. Recent studies revealed that the specific expression and distribution of each subunit are deeply involved in neural excitability, plasticity, and synaptic deficits. The present article summarizes reports on the dysfunction of NMDA receptors and responsible subunits in various neurological and psychiatric disorders, including schizophrenia, autoimmune-induced glutamatergic receptor dysfunction, mood disorders, and autism. A key role for the GluN2D subunit in NMDA receptor antagonist-induced psychosis has been recently revealed.
Collapse
Affiliation(s)
| | | | | | - K Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.
| |
Collapse
|
32
|
Pollak TA, De Simoni S, Barimani B, Zelaya FO, Stone JM, Mehta MA. Phenomenologically distinct psychotomimetic effects of ketamine are associated with cerebral blood flow changes in functionally relevant cerebral foci: a continuous arterial spin labelling study. Psychopharmacology (Berl) 2015; 232:4515-24. [PMID: 26438425 DOI: 10.1007/s00213-015-4078-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 09/07/2015] [Indexed: 12/30/2022]
Abstract
RATIONALE The N-methyl-D-aspartate (NMDA) receptor antagonist ketamine provides a pragmatic approach to address the link between glutamate-mediated changes in brain function and psychosis-like experiences. Most studies using PET or BOLD fMRI have assessed these symptoms broadly, which may limit inference about specific mechanisms. OBJECTIVES The objective of this study is to identify the cerebral blood flow (CBF) correlates of ketamine-induced psychopathology, focusing on individual psychotomimetic symptom dimensions, which may have separable neurobiological substrates. METHODS We measured validated psychotomimetic symptom factors following intravenous ketamine administration in 23 healthy male volunteers (10 given a lower dose and 13 a higher dose) and correlated ketamine-induced changes in symptoms with regional changes in CBF, measured non-invasively using arterial spin labelling (ASL). RESULTS The main effect of ketamine paralleled previous studies, with increases in CBF in anterior and subgenual cingulate cortex and decreases in superior and medial temporal cortex. Subjective effects were greater in the high-dose group. For this group, ketamine-induced anhedonia inversely related to orbitofrontal cortex CBF changes and cognitive disorganisation was positively correlated with CBF changes in posterior thalamus and the left inferior and middle temporal gyrus. Perceptual distortion was correlated with different regional CBF changes in the low- and high-dose groups. CONCLUSIONS Here, we provide evidence for the sensitivity of ASL to the effects of ketamine and the strength of subjective experience, suggesting plausible neural mechanisms for ketamine-induced anhedonia and cognitive disorganisation.
Collapse
Affiliation(s)
- T A Pollak
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's Health Partners, King's College London, De Crespigny Park, Denmark Hill, London, SE5 8AF, UK.
| | - S De Simoni
- Computational, Cognitive and Clinical Neuroimaging Laboratory, Department of Medicine, Imperial College London, London, UK
| | - B Barimani
- Faculty of Medicine, Imperial College London, London, UK
| | - F O Zelaya
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - J M Stone
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - M A Mehta
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
33
|
GABA abnormalities in schizophrenia: a methodological review of in vivo studies. Schizophr Res 2015; 167:84-90. [PMID: 25458856 PMCID: PMC4409914 DOI: 10.1016/j.schres.2014.10.011] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Revised: 10/04/2014] [Accepted: 10/07/2014] [Indexed: 12/31/2022]
Abstract
Abnormalities of GABAergic interneurons are some of the most consistent findings from post-mortem studies of schizophrenia. However, linking these molecular deficits with in vivo observations in patients - a critical goal in order to evaluate interventions that would target GABAergic deficits - presents a challenge. Explanatory models have been developed based on animal work and the emerging experimental literature in schizophrenia patients. This literature includes: neuroimaging ligands to GABA receptors, magnetic resonance spectroscopy (MRS) of GABA concentration, transcranial magnetic stimulation of cortical inhibitory circuits and pharmacologic probes of GABA receptors to dynamically challenge the GABA system, usually in combination with neuroimaging studies. Pharmacologic challenges have elicited behavioral changes, and preliminary studies of therapeutic GABAergic interventions have been conducted. This article critically reviews the evidence for GABAergic dysfunction from each of these areas. These methods remain indirect measures of GABAergic function, and a broad array of dysfunction is linked with the putative GABAergic measures, including positive symptoms, cognition, emotion, motor processing and sensory processing, covering diverse brain areas. Measures of receptor binding have not shown replicable group differences in binding, and MRS assays of GABA concentration have yielded equivocal evidence of large-scale alteration in GABA concentration. Overall, the experimental base remains sparse, and much remains to be learned about the role of GABAergic interneurons in healthy brains. Challenges with pharmacologic and functional probes show promise, and may yet enable a better characterization of GABAergic deficits in schizophrenia.
Collapse
|
34
|
Fenton AA. Excitation-inhibition discoordination in rodent models of mental disorders. Biol Psychiatry 2015; 77:1079-88. [PMID: 25895430 PMCID: PMC4444398 DOI: 10.1016/j.biopsych.2015.03.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Revised: 03/11/2015] [Accepted: 03/11/2015] [Indexed: 12/21/2022]
Abstract
Animal models of mental illness provide a foundation for evaluating hypotheses for the mechanistic causes of mental illness. Neurophysiological investigations of neural network activity in rodent models of mental dysfunction are reviewed from the conceptual framework of the discoordination hypothesis, which asserts that failures of neural coordination cause cognitive deficits in the judicious processing and use of information. Abnormal dynamic coordination of excitatory and inhibitory neural discharge in pharmacologic and genetic rodent models supports the discoordination hypothesis. These observations suggest excitation-inhibition discoordination and aberrant neural circuit dynamics as causes of cognitive impairment, as well as therapeutic targets for cognition-promoting treatments.
Collapse
|
35
|
Optogenetic stimulation of infralimbic PFC reproduces ketamine's rapid and sustained antidepressant actions. Proc Natl Acad Sci U S A 2015; 112:8106-11. [PMID: 26056286 DOI: 10.1073/pnas.1414728112] [Citation(s) in RCA: 208] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Ketamine produces rapid and sustained antidepressant actions in depressed patients, but the precise cellular mechanisms underlying these effects have not been identified. Here we determined if modulation of neuronal activity in the infralimbic prefrontal cortex (IL-PFC) underlies the antidepressant and anxiolytic actions of ketamine. We found that neuronal inactivation of the IL-PFC completely blocked the antidepressant and anxiolytic effects of systemic ketamine in rodent models and that ketamine microinfusion into IL-PFC reproduced these behavioral actions of systemic ketamine. We also found that optogenetic stimulation of the IL-PFC produced rapid and long-lasting antidepressant and anxiolytic effects and that these effects are associated with increased number and function of spine synapses of layer V pyramidal neurons. The results demonstrate that ketamine infusions or optogenetic stimulation of IL-PFC are sufficient to produce long-lasting antidepressant behavioral and synaptic responses similar to the effects of systemic ketamine administration.
Collapse
|
36
|
Higa KK, Ji B, Buell MR, Risbrough VB, Powell SB, Young JW, Geyer MA, Zhou X. Restoration of Sp4 in Forebrain GABAergic Neurons Rescues Hypersensitivity to Ketamine in Sp4 Hypomorphic Mice. Int J Neuropsychopharmacol 2015; 18:pyv063. [PMID: 26037489 PMCID: PMC4756721 DOI: 10.1093/ijnp/pyv063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/29/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Ketamine produces schizophrenia-like behavioral phenotypes in healthy people. Prolonged ketamine effects and exacerbation of symptoms after the administration of ketamine have been observed in patients with schizophrenia. More recently, ketamine has been used as a potent antidepressant to treat patients with major depression. The genes and neurons that regulate behavioral responses to ketamine, however, remain poorly understood. Sp4 is a transcription factor for which gene expression is restricted to neuronal cells in the brain. Our previous studies demonstrated that Sp4 hypomorphic mice display several behavioral phenotypes relevant to psychiatric disorders, consistent with human SP4 gene associations with schizophrenia, bipolar disorder, and major depression. Among those behavioral phenotypes, hypersensitivity to ketamine-induced hyperlocomotion has been observed in Sp4 hypomorphic mice. METHODS In the present study, we used the Cre-LoxP system to restore Sp4 gene expression, specifically in either forebrain excitatory or GABAergic inhibitory neurons in Sp4 hypomorphic mice. Mouse behavioral phenotypes related to psychiatric disorders were examined in these distinct rescue mice. RESULTS Restoration of Sp4 in forebrain excitatory neurons did not rescue deficient sensorimotor gating nor ketamine-induced hyperlocomotion. Restoration of Sp4 in forebrain GABAergic neurons, however, rescued ketamine-induced hyperlocomotion, but did not rescue deficient sensorimotor gating. CONCLUSIONS Our studies suggest that the Sp4 gene in forebrain GABAergic neurons regulates ketamine-induced hyperlocomotion.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xianjin Zhou
- Department of Psychiatry, University of California San Diego, La Jolla, CA (Ms Higa, Drs Ji, Risbrough, Powell, Young, Geyer, and Zhou, and Ms Buell); Research Service, VA San Diego Healthcare System, La Jolla, CA (Drs Risbrough, Powell, Young, Geyer, and Zhou, and Ms Buell); Neurosciences Graduate Program, University of California San Diego, La Jolla, CA (Ms Higa).
| |
Collapse
|
37
|
Wang D, Zhou Y, Zhuo C, Qin W, Zhu J, Liu H, Xu L, Yu C. Altered functional connectivity of the cingulate subregions in schizophrenia. Transl Psychiatry 2015; 5:e575. [PMID: 26035059 PMCID: PMC4490280 DOI: 10.1038/tp.2015.69] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 03/06/2015] [Accepted: 04/23/2015] [Indexed: 01/21/2023] Open
Abstract
Schizophrenia patients have shown altered resting-state functional connectivity (rsFC) of the cingulate cortex; however, it is unknown whether rsFCs of the cingulate subregions are differentially affected in this disorder. We aimed to clarify the issue by comparing rsFCs of each cingulate subregion between healthy controls and schizophrenia patients. A total of 102 healthy controls and 94 schizophrenia patients underwent resting-state functional magnetic resonance imaging with a sensitivity-encoded spiral-in imaging sequence to reduce susceptibility-induced signal loss and distortion. The cingulate cortex was divided into nine subregions, including the subgenual anterior cingulate cortex (ACC), areas 24 and 32 of the pregenual ACC, areas 24 and 32 of the anterior mid-cingulate cortex (aMCC), posterior MCC (pMCC), dorsal (dPCC) and ventral (vPCC) posterior cingulate cortex (PCC) and retrosplenial cortex (RSC). The rsFCs of each cingulate subregion were compared between the two groups and the atrophy effect was considered. Results with and without global signal regression were reported. Most cingulate subregions exhibited decreased rsFCs in schizophrenia after global signal regression (GSR). Without GSR, only increased rsFC was found in schizophrenia, which primarily restricted to the aMCC, PCC and RSC. Some of these increased rsFCs were also significant after GSR. These findings suggest that GSR can greatly affect between-group differences in rsFCs and the consistently increased rsFCs may challenge the functional disconnection hypothesis of schizophrenia.
Collapse
Affiliation(s)
- D Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Y Zhou
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - C Zhuo
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Anding Hospital (Tianjin Mental Health Center), Tianjin, China
- Tianjin Anning Hospital, Tianjin, China
| | - W Qin
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - J Zhu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - H Liu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - L Xu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - C Yu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China. E-mail:
| |
Collapse
|
38
|
Khalili-Mahani N, Niesters M, van Osch MJ, Oitzl M, Veer I, de Rooij M, van Gerven J, van Buchem MA, Beckmann CF, Rombouts SARB, Dahan A. Ketamine interactions with biomarkers of stress: a randomized placebo-controlled repeated measures resting-state fMRI and PCASL pilot study in healthy men. Neuroimage 2014; 108:396-409. [PMID: 25554429 DOI: 10.1016/j.neuroimage.2014.12.050] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 11/24/2014] [Accepted: 12/17/2014] [Indexed: 12/17/2022] Open
Abstract
Ketamine, an NMDA receptor antagonist, is increasingly used to study the link between glutamatergic signaling dysregulation and mood and chronic pain disorders. Glutamatergic neurotransmission and stress corticosteroids (cortisol in human) are critical for Ca(2+) mediated neuroplasticity and behavioral adaptation. The mechanisms of action of glutamatergic neurotransmission and stress corticosteroids on the NMDA-receptors of the hippocampus have been long investigated in animals, but given little attention in human studies. In this randomized single-blinded placebo-controlled crossover study (12 healthy young men), five sets of resting-state fMRI (RSFMRI), pseudocontinuous arterial spin labeling (PCASL), and corresponding salivary cortisol samples were acquired over 4h, at given intervals under pharmacokinetically-controlled infusion of subanesthetic ketamine (20 & 40mg/70kg/h). An identical procedure was repeated under a sham placebo condition. Differences in the profile of ketamine versus placebo effect over time were examined. Compared to placebo, ketamine mimicked a stress-like response (increased cortisol, reduced calmness and alertness, and impaired working memory). Ketamine effects on the brain included a transient prefrontal hyperperfusion and a dose-related reduction of relative hippocampal perfusion, plus emerging hyperconnectivity between the hippocampus and the occipital, cingulate, precuneal, cerebellar and basal ganglia regions. The spatiotemporal profiles of ketamine effects on different hippocampal subnetworks suggest a topographically dissociable change in corticohippocampal functional connectivity. We discuss our findings in the context of the negative feedback inhibition theory of the hippocampal stress-control. This pilot study provides a methodological framework for multimodal functional neuroimaging under resting-state conditions, which may be generalized for translational studies of glutamatergic- or stress-related etiology of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Najmeh Khalili-Mahani
- Department of Anesthesiology, Leiden University Medical Center, Leiden, The Netherlands; Leiden Institute for Brain and Cognition (LIBC), Leiden University, Leiden, The Netherlands; Department of Radiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Montreal Neurological Institute, McGill University, Montreal, Canada.
| | - Marieke Niesters
- Department of Anesthesiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Matthias J van Osch
- Department of Radiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Institute of Psychology, Leiden University, Leiden, The Netherlands
| | - Melly Oitzl
- SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Ilya Veer
- Leiden Institute for Brain and Cognition (LIBC), Leiden University, Leiden, The Netherlands; Department of Radiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Charité Universitätsmedizin Berlin, Division of Mind and Brain Research, Department of Psychiatry and Psychotherapy, Berlin, Germany
| | - Mark de Rooij
- Leiden Institute for Brain and Cognition (LIBC), Leiden University, Leiden, The Netherlands
| | - Joop van Gerven
- Department of Neurology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Mark A van Buchem
- Department of Radiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Christian F Beckmann
- Donders Institute for Brain, Cognition and Behaviour; Centre for Cognitive Neuroimaging, Radboud University, Nijmegen, The Netherlands
| | - Serge A R B Rombouts
- Leiden Institute for Brain and Cognition (LIBC), Leiden University, Leiden, The Netherlands; Department of Radiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Institute of Psychology, Leiden University, Leiden, The Netherlands
| | - Albert Dahan
- Department of Anesthesiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
39
|
Troyano-Rodriguez E, Lladó-Pelfort L, Santana N, Teruel-Martí V, Celada P, Artigas F. Phencyclidine inhibits the activity of thalamic reticular gamma-aminobutyric acidergic neurons in rat brain. Biol Psychiatry 2014; 76:937-45. [PMID: 25038984 DOI: 10.1016/j.biopsych.2014.05.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 04/24/2014] [Accepted: 05/21/2014] [Indexed: 01/04/2023]
Abstract
BACKGROUND The neurobiological basis of action of noncompetitive N-methyl-D-aspartate acid receptor (NMDA-R) antagonists is poorly understood. Electrophysiological studies indicate that phencyclidine (PCP) markedly disrupts neuronal activity with an overall excitatory effect and reduces the power of low-frequency oscillations (LFO; <4 Hz) in thalamocortical networks. Because the reticular nucleus of the thalamus (RtN) provides tonic feed-forward inhibition to the rest of the thalamic nuclei, we examined the effect of PCP on RtN activity, under the working hypothesis that NMDA-R blockade in RtN would disinhibit thalamocortical networks. METHODS Drug effects (PCP followed by clozapine) on the activity of RtN (single unit and local field potential recordings) and prefrontal cortex (PFC; electrocorticogram) in anesthetized rats were assessed. RESULTS PCP (.25-.5 mg/kg, intravenous) reduced the discharge rate of 19 of 21 RtN neurons to 37% of baseline (p < .000001) and the power of LFO in RtN and PFC to ~20% of baseline (p < .001). PCP also reduced the coherence between PFC and RtN in the LFO range. A low clozapine dose (1 mg/kg intravenous) significantly countered the effect of PCP on LFO in PFC but not in RtN and further reduced the discharge rate of RtN neurons. However, clozapine administration partly antagonized the fall in coherence and phase-locking values produced by PCP. CONCLUSIONS PCP activates thalamocortical circuits in a bottom-up manner by reducing the activity of RtN neurons, which tonically inhibit thalamic relay neurons. However, clozapine reversal of PCP effects is not driven by restoring RtN activity and may involve a cortical action.
Collapse
Affiliation(s)
- Eva Troyano-Rodriguez
- Department of Neurochemistry and Neuropharmacology, Institut d'Investigacions Biomèdiques de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental, Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Laia Lladó-Pelfort
- Department of Neurochemistry and Neuropharmacology, Institut d'Investigacions Biomèdiques de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental, Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Noemi Santana
- Department of Neurochemistry and Neuropharmacology, Institut d'Investigacions Biomèdiques de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental, Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Vicent Teruel-Martí
- Deptartamento de Anatomia i Embriologia Humana, Facultat de Medicina, Universitat de València, Valencia, Spain
| | - Pau Celada
- Department of Neurochemistry and Neuropharmacology, Institut d'Investigacions Biomèdiques de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental, Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain.
| | - Francesc Artigas
- Department of Neurochemistry and Neuropharmacology, Institut d'Investigacions Biomèdiques de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental, Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| |
Collapse
|
40
|
Ji B, Higa KK, Kim M, Zhou L, Young JW, Geyer MA, Zhou X. Inhibition of protein translation by the DISC1-Boymaw fusion gene from a Scottish family with major psychiatric disorders. Hum Mol Genet 2014; 23:5683-705. [PMID: 24908665 DOI: 10.1093/hmg/ddu285] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The t(1; 11) translocation appears to be the causal genetic lesion with 70% penetrance for schizophrenia, major depression and other psychiatric disorders in a Scottish family. Molecular studies identified the disruption of the disrupted-in-schizophrenia 1 (DISC1) gene by chromosome translocation at chromosome 1q42. Our previous studies, however, revealed that the translocation also disrupted another gene, Boymaw (also termed DISC1FP1), on chromosome 11. After translocation, two fusion genes [the DISC1-Boymaw (DB7) and the Boymaw-DISC1 (BD13)] are generated between the DISC1 and Boymaw genes. In the present study, we report that expression of the DB7 fusion gene inhibits both intracellular NADH oxidoreductase activities and protein translation. We generated humanized DISC1-Boymaw mice with gene targeting to examine the in vivo functions of the fusion genes. Consistent with the in vitro studies on the DB7 fusion gene, protein translation activity is decreased in the hippocampus and in cultured primary neurons from the brains of the humanized mice. Expression of Gad67, Nmdar1 and Psd95 proteins are also reduced. The humanized mice display prolonged and increased responses to the NMDA receptor antagonist, ketamine, on various mouse genetic backgrounds. Abnormal information processing of acoustic startle and depressive-like behaviors are also observed. In addition, the humanized mice display abnormal erythropoiesis, which was reported to associate with depression in humans. Expression of the DB7 fusion gene may reduce protein translation to impair brain functions and thereby contribute to the pathogenesis of major psychiatric disorders.
Collapse
Affiliation(s)
- Baohu Ji
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Kerin K Higa
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Minjung Kim
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Lynn Zhou
- La Jolla High School, 750 Nautilus St., San Diego, CA 92037, USA and
| | - Jared W Young
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA, Research Service, VA San Diego Healthcare System, 3350 La Jolla Village Drive, San Diego, CA 92037, USA
| | - Mark A Geyer
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA, Research Service, VA San Diego Healthcare System, 3350 La Jolla Village Drive, San Diego, CA 92037, USA
| | - Xianjin Zhou
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA, Research Service, VA San Diego Healthcare System, 3350 La Jolla Village Drive, San Diego, CA 92037, USA
| |
Collapse
|
41
|
Daya R, Bhandari J, Hui P, Tian Y, Farncombe T, Mishra R. Effects of MK-801 treatment across several pre-clinical analyses including a novel assessment of brain metabolic function utilizing PET and CT fused imaging in live rats. Neuropharmacology 2014; 77:325-33. [DOI: 10.1016/j.neuropharm.2013.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 09/09/2013] [Accepted: 10/02/2013] [Indexed: 12/16/2022]
|
42
|
Merritt K, McGuire P, Egerton A. Relationship between Glutamate Dysfunction and Symptoms and Cognitive Function in Psychosis. Front Psychiatry 2013; 4:151. [PMID: 24324444 PMCID: PMC3840324 DOI: 10.3389/fpsyt.2013.00151] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Accepted: 11/07/2013] [Indexed: 11/13/2022] Open
Abstract
The glutamate hypothesis of schizophrenia, proposed over two decades ago, originated following the observation that administration of drugs that block NMDA glutamate receptors, such as ketamine, could induce schizophrenia-like symptoms. Since then, this hypothesis has been extended to describe how glutamate abnormalities may disturb brain function and underpin psychotic symptoms and cognitive impairments. The glutamatergic system is now a major focus for the development of new compounds in schizophrenia. Relationships between regional brain glutamate function and symptom severity can be investigated using proton magnetic resonance spectroscopy (1H-MRS) to estimate levels of glutamatergic metabolites in vivo. Here we briefly review the 1H-MRS studies that have explored relationships between glutamatergic metabolites, symptoms, and cognitive function in clinical samples. While some of these studies suggest that more severe symptoms may be associated with elevated glutamatergic function in the anterior cingulate, studies in larger patient samples selected on the basis of symptom severity are required.
Collapse
Affiliation(s)
- Kate Merritt
- Department of Psychosis Studies, Institute of Psychiatry, King's College London , London , UK
| | | | | |
Collapse
|
43
|
Driesen NR, McCarthy G, Bhagwagar Z, Bloch M, Calhoun V, D’Souza DC, Gueorguieva R, He G, Ramachandran R, Suckow RF, Anticevic A, Morgan PT, Krystal JH. Relationship of resting brain hyperconnectivity and schizophrenia-like symptoms produced by the NMDA receptor antagonist ketamine in humans. Mol Psychiatry 2013; 18:1199-204. [PMID: 23337947 PMCID: PMC3646075 DOI: 10.1038/mp.2012.194] [Citation(s) in RCA: 161] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 11/07/2012] [Accepted: 11/12/2012] [Indexed: 12/28/2022]
Abstract
N-methyl-D-aspartate glutamate receptor (NMDA-R) antagonists produce schizophrenia-like positive and negative symptoms in healthy human subjects. Preclinical research suggests that NMDA-R antagonists interfere with the function of gamma-aminobutyric acid (GABA) neurons and alter the brain oscillations. These changes have been hypothesized to contribute to psychosis. In this investigation, we evaluated the hypothesis that the NMDA-R antagonist ketamine produces alterations in cortical functional connectivity during rest that are related to symptoms. We administered ketamine to a primary sample of 22 subjects and to an additional, partially overlapping, sample of 12 subjects. Symptoms before and after the experimental session were rated with the Positive and Negative Syndrome Scale (PANSS). In the primary sample, functional connectivity was measured via functional magnetic resonance imaging almost immediately after infusion began. In the additional sample, this assessment was repeated after 45 min of continuous ketamine infusion. Global, enhanced functional connectivity was observed at both timepoints, and this hyperconnectivity was related to symptoms in a region-specific manner. This study supports the hypothesis that pathological increases in resting brain functional connectivity contribute to the emergence of positive and negative symptoms associated with schizophrenia.
Collapse
|
44
|
Prolonged Ketamine Effects in Sp4 Hypomorphic Mice: Mimicking Phenotypes of Schizophrenia. PLoS One 2013; 8:e66327. [PMID: 23823008 PMCID: PMC3688895 DOI: 10.1371/journal.pone.0066327] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 05/04/2013] [Indexed: 12/11/2022] Open
Abstract
It has been well established that schizophrenia patients display impaired NMDA receptor (NMDAR) functions as well as exacerbation of symptoms in response to NMDAR antagonists. Abnormal NMDAR signaling presumably contributes to cognitive deficits which substantially contribute to functional disability in schizophrenia. Establishing a mouse genetic model will help investigate molecular mechanisms of hypoglutmatergic neurotransmission in schizophrenia. Here, we examined the responses of Sp4 hypomorphic mice to NMDAR antagonists in electroencephalography and various behavioral paradigms. Sp4 hypomorphic mice, previously reported to have reduced NMDAR1 expression and LTP deficit in hippocampal CA1, displayed increased sensitivity and prolonged responses to NMDAR antagonists. Molecular studies demonstrated reduced expression of glutamic acid decarboxylase 67 (GAD67) in both cortex and hippocampus, consistent with abnormal gamma oscillations in Sp4 hypomorphic mice. On the other hand, human SP4 gene was reported to be deleted in schizophrenia. Several human genetic studies suggested the association of SP4 gene with schizophrenia and other psychiatric disorders. Therefore, elucidation of the Sp4 molecular pathway in Sp4 hypomorphic mice may provide novel insights to our understanding of abnormal NMDAR signaling in schizophrenia.
Collapse
|
45
|
Carlson PJ, Diazgranados N, Nugent AC, Ibrahim L, Luckenbaugh DA, Brutsche N, Herscovitch P, Manji HK, Zarate CA, Drevets WC. Neural correlates of rapid antidepressant response to ketamine in treatment-resistant unipolar depression: a preliminary positron emission tomography study. Biol Psychiatry 2013; 73:1213-21. [PMID: 23540908 PMCID: PMC3672258 DOI: 10.1016/j.biopsych.2013.02.008] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 01/16/2013] [Accepted: 02/01/2013] [Indexed: 12/14/2022]
Abstract
BACKGROUND Multiple lines of evidence support a role for the glutamatergic system in the pathophysiology of major depressive disorder (MDD). Ketamine, an N-methyl-D-aspartate antagonist, rapidly improves depressive symptoms in individuals with treatment-resistant depression. The neural mechanisms underlying this effect remain unknown. METHODS In this preliminary study, 20 unmedicated participants with treatment-resistant MDD underwent positron emission tomography to measure regional cerebral glucose metabolism at baseline and following ketamine infusion (single dose of .5mg/kg intravenous over 40minutes). Metabolic data were compared between conditions using a combination of region-of-interest and voxelwise analyses, and differences were correlated with the associated antidepressant response. RESULTS Whole-brain metabolism did not change significantly following ketamine. Regional metabolism decreased significantly under ketamine in the habenula, insula, and ventrolateral and dorsolateral prefrontal cortices of the right hemisphere. Metabolism increased postketamine in bilateral occipital, right sensorimotor, left parahippocampal, and left inferior parietal cortices. Improvement in depression ratings correlated directly with change in metabolism in right superior and middle temporal gyri. Conversely, clinical improvement correlated inversely with metabolic changes in right parahippocampal gyrus and temporoparietal cortex. CONCLUSIONS Although preliminary, these results indicate that treatment-resistant MDD subjects showed decreased metabolism in the right habenula and the extended medial and orbital prefrontal networks in association with rapid antidepressant response to ketamine. Conversely, metabolism increased in sensory association cortices, conceivably related to the illusory phenomena sometimes experienced with ketamine. Further studies are needed to elucidate how these functional anatomical changes relate to the molecular mechanisms underlying ketamine's rapid antidepressant effects.
Collapse
Affiliation(s)
- Paul J. Carlson
- Salt Lake City Veterans Affairs Medical Center and Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT
| | - Nancy Diazgranados
- Department of Psychiatry, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Allison C. Nugent
- Experimental Therapeutics and Pathophysiology Branch, NIMH, NIH, Bethesda, MD
| | - Lobna Ibrahim
- Experimental Therapeutics and Pathophysiology Branch, NIMH, NIH, Bethesda, MD
| | | | - Nancy Brutsche
- Experimental Therapeutics and Pathophysiology Branch, NIMH, NIH, Bethesda, MD
| | - Peter Herscovitch
- National Institutes of Health, Clinical Center, PET Department, Bethesda, MD
| | - Husseini K. Manji
- Johnson & Johnson Pharmaceutical Research & Development, Titusville, NJ
| | - Carlos A. Zarate
- Experimental Therapeutics and Pathophysiology Branch, NIMH, NIH, Bethesda, MD
| | - Wayne C. Drevets
- Johnson & Johnson Pharmaceutical Research & Development, Titusville, NJ,Laureate Institute for Brain Research and University of Oklahoma College of Medicine, Department of Psychiatry, Tulsa, OK
| |
Collapse
|
46
|
Thomases DR, Cass DK, Tseng KY. Periadolescent exposure to the NMDA receptor antagonist MK-801 impairs the functional maturation of local GABAergic circuits in the adult prefrontal cortex. J Neurosci 2013; 33:26-34. [PMID: 23283319 PMCID: PMC3544161 DOI: 10.1523/jneurosci.4147-12.2013] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 10/24/2012] [Accepted: 10/28/2012] [Indexed: 01/28/2023] Open
Abstract
A developmental disruption of prefrontal cortical inhibitory circuits is thought to contribute to the adolescent onset of cognitive deficits observed in schizophrenia. However, the developmental mechanisms underlying such a disruption remain elusive. The goal of this study is to examine how repeated exposure to the NMDA receptor antagonist dizocilpine maleate (MK-801) during periadolescence [from postnatal day 35 (P35) to P40] impacts the normative development of local prefrontal network response in rats. In vivo electrophysiological analyses revealed that MK-801 administration during periadolescence elicits an enduring disinhibited prefrontal local field potential (LFP) response to ventral hippocampal stimulation at 20 Hz (beta) and 40 Hz (gamma) in adulthood (P65-P85). Such a disinhibition was not observed when MK-801 was given during adulthood, indicating that the periadolescent transition is indeed a sensitive period for the functional maturation of prefrontal inhibitory control. Accordingly, the pattern of prefrontal LFP disinhibition induced by periadolescent MK-801 treatment resembles that observed in the normal P30-P40 prefrontal cortex (PFC). Additional pharmacological manipulations revealed that these developmentally immature prefrontal responses can be mimicked by single microinfusion of the GABA(A) receptor antagonist picrotoxin into the normal adult PFC. Importantly, acute administration of the GABA(A)-positive allosteric modulator Indiplon into the PFC reversed the prefrontal disinhibitory state induced by periadolescent MK-801 to normal levels. Together, these results indicate a critical role of NMDA receptors in regulating the periadolescent maturation of GABAergic networks in the PFC and that pharmacologically induced augmentation of local GABA(A)-receptor-mediated transmission is sufficient to overcome the disinhibitory prefrontal state associated with the periadolescent MK-801 exposure.
Collapse
Affiliation(s)
- Daniel R. Thomases
- Department of Cellular and Molecular Pharmacology, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064
| | - Daryn K. Cass
- Department of Cellular and Molecular Pharmacology, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064
| | - Kuei Y. Tseng
- Department of Cellular and Molecular Pharmacology, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064
| |
Collapse
|
47
|
NMDA antagonist ketamine reduces task selectivity in macaque dorsolateral prefrontal neurons and impairs performance of randomly interleaved prosaccades and antisaccades. J Neurosci 2012; 32:12018-27. [PMID: 22933786 DOI: 10.1523/jneurosci.1510-12.2012] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Ketamine, an NMDA receptor antagonist, has been shown to induce behavioral abnormalities in humans that mimic the positive, negative, and most importantly cognitive deficits observed in schizophrenia. Similar cognitive deficits have been observed in nonhuman primates after a subanesthetic dose of ketamine, including an impairment in their ability to perform the antisaccade task, which requires the suppression of a prosaccade toward a flashed stimulus and the generation of a saccade in the opposite direction. The neural basis underlying these cognitive impairments remains unknown. Here, we recorded single-neuron activity in the lateral prefrontal cortex of macaque monkeys before and after the administration of subanesthetic doses of ketamine during the performance of randomly interleaved prosaccade and antisaccade trials. Ketamine impeded the monkeys' ability to maintain and apply the correct task rule and increased reaction times of prosaccades and antisaccades. These behavioral changes were associated with an overall increase in activity of PFC neurons and a reduction in their task selectivity. Our results suggest that the mechanism underlying ketamine-induced cognitive abnormalities may be the nonspecific increase in PFC activity and the associated reduction of task selectivity.
Collapse
|
48
|
Featherstone RE, Liang Y, Saunders JA, Tatard-Leitman VM, Ehrlichman RS, Siegel SJ. Subchronic ketamine treatment leads to permanent changes in EEG, cognition and the astrocytic glutamate transporter EAAT2 in mice. Neurobiol Dis 2012; 47:338-46. [DOI: 10.1016/j.nbd.2012.05.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2011] [Revised: 04/09/2012] [Accepted: 05/11/2012] [Indexed: 01/11/2023] Open
|
49
|
Nagels A, Kirner-Veselinovic A, Wiese R, Paulus FM, Kircher T, Krach S. Effects of ketamine-induced psychopathological symptoms on continuous overt rhyme fluency. Eur Arch Psychiatry Clin Neurosci 2012; 262:403-14. [PMID: 22189657 DOI: 10.1007/s00406-011-0281-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 12/03/2011] [Indexed: 11/29/2022]
Abstract
The N-methyl-D-aspartate receptor (NMDAR) has been implicated in the pathophysiology of schizophrenia. Administered to healthy individuals, a subanesthetic dose of the noncompetitive NMDAR antagonist ketamine reproduces several psychopathological symptoms commonly observed in patients with schizophrenia. In a counterbalanced, placebo-controlled, double-blind, within-participants study, fifteen healthy subjects were administered a continuous subanesthetic S-ketamine infusion while cortical activation was measured using functional magnetic resonance imaging. While being scanned, subjects performed an overt word generation task. Ketamine-induced psychopathological symptoms were assessed with the Positive and Negative Syndrome Scale (PANSS). Ketamine administration elicited effects on psychopathology, including difficulties in abstract thinking, lack of spontaneity and flow of conversation as well as formal thought disorder. On a behavioral level, verbal fluency performance was unaffected. The PANSS score for formal thought disorder positively correlated with activation measures encompassing the left superior temporal gyrus, the right middle and inferior frontal gyrus and the precuneus. Difficulty in abstract thinking was correlated with pronounced activations in prefrontal as well as in anterior cingulate regions, whereas hyperactivations in the left superior temporal gyrus were found in association with a lack of spontaneity and flow of conversation. In the absence of behavioral impairments during verbal fluency, NMDAR blocking evoked psychopathological symptoms and cortical activations in regions previously reported in schizophrenia patients. The results provide further support for the hypothesis of an NMDAR dysfunction in the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Arne Nagels
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Germany.
| | | | | | | | | | | |
Collapse
|
50
|
Fell MJ, McKinzie DL, Monn JA, Svensson KA. Group II metabotropic glutamate receptor agonists and positive allosteric modulators as novel treatments for schizophrenia. Neuropharmacology 2012; 62:1473-83. [DOI: 10.1016/j.neuropharm.2011.06.007] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Revised: 06/07/2011] [Accepted: 06/08/2011] [Indexed: 10/18/2022]
|