151
|
Ingram R, Kang H, Lightman S, Jane DE, Bortolotto ZA, Collingridge GL, Lodge D, Volianskis A. Some distorted thoughts about ketamine as a psychedelic and a novel hypothesis based on NMDA receptor-mediated synaptic plasticity. Neuropharmacology 2018; 142:30-40. [PMID: 29885421 DOI: 10.1016/j.neuropharm.2018.06.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 05/27/2018] [Accepted: 06/05/2018] [Indexed: 12/12/2022]
Abstract
Ketamine, a channel blocking NMDA receptor antagonist, is used off-label for its psychedelic effects, which may arise from a combination of several inter-related actions. Firstly, reductions of the contribution of NMDA receptors to afferent information from external and internal sensory inputs may distort sensations and their processing in higher brain centres. Secondly, reductions of NMDA receptor-mediated excitation of GABAergic interneurons can result in glutamatergic overactivity. Thirdly, limbic cortical disinhibition may indirectly enhance dopaminergic and serotonergic activity. Fourthly, inhibition of NMDA receptor mediated synaptic plasticity, such as short-term potentiation (STP) and long-term potentiation (LTP), could lead to distorted memories. Here, for the first time, we compared quantitatively the effects of ketamine on STP and LTP. We report that ketamine inhibits STP in a double sigmoidal fashion with low (40 nM) and high (5.6 μM) IC50 values. In contrast, ketamine inhibits LTP in a single sigmoidal manner (IC50 value ∼ 15 μM). A GluN2D-subunit preferring NMDA receptor antagonist, UBP145, has a similar pharmacological profile. We propose that the psychedelic effects of ketamine may involve the inhibition of STP and, potentially, associated forms of working memory. This article is part of the Special Issue entitled 'Psychedelics: New Doors, Altered Perceptions'.
Collapse
Affiliation(s)
- Rachael Ingram
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Heather Kang
- School of Clinical Sciences, University of Bristol, Bristol, UK; Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK; Dept Physiology, University of Toronto and Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| | | | - David E Jane
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Zuner A Bortolotto
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Graham L Collingridge
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK; Dept Physiology, University of Toronto and Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| | - David Lodge
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Arturas Volianskis
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK; School of Clinical Sciences, University of Bristol, Bristol, UK.
| |
Collapse
|
152
|
Jeong YH, Choi JH, Lee D, Kim S, Kim KT. Vaccinia-related kinase 2 modulates role of dysbindin by regulating protein stability. J Neurochem 2018; 147:609-625. [PMID: 30062698 DOI: 10.1111/jnc.14562] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/13/2018] [Accepted: 07/24/2018] [Indexed: 12/23/2022]
Abstract
Vaccinia-related kinase 2 (VRK2) is a serine/threonine kinase that belongs to the casein kinase 1 family. VRK2 has long been known for its relationship with neurodegenerative disorders such as schizophrenia. However, the role of VRK2 and the substrates associated with it are unknown. Dysbindin is known as one of the strong risk factors for schizophrenia. The expression of dysbindin is indeed significantly reduced in schizophrenia patients. Moreover, dysbindin is involved in neurite outgrowth and regulation of NMDA receptor signaling. Here, we first identified dysbindin as a novel interacting protein of VRK2 through immunoprecipitation. We hypothesized that dysbindin is phosphorylated by VRK2 and further that this phosphorylation plays an important role in the function of dysbindin. We show that VRK2 phosphorylates Ser 297 and Ser 299 of dysbindin using in vitro kinase assay. In addition, we found that VRK2-mediated phosphorylation of dysbindin enhanced ubiquitination of dysbindin and consequently resulted in the decrease in its protein stability through western blotting. Over-expression of VRK2 in human neuroblastoma (SH-SY5Y) cells reduced neurite outgrowth induced by retinoic acid. Furthermore, a phosphomimetic mutant of dysbindin alleviated neurite outgrowth and affected surface expression of N-methyl-d-aspartate 2A, a subunit of NMDA receptor in mouse hippocampal neurons. Together, our work reveals the regulation of dysbindin by VRK2, providing the association of these two proteins, which are commonly implicated in schizophrenia. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Young-Hun Jeong
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Korea
| | - Jung-Hyun Choi
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Korea
| | - Dohyun Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Korea.,R&D Center, NovMetaPharma Co., Ltd., Pohang, 37668, Korea
| | - Sangjune Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Korea.,Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kyong-Tai Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Korea.,Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Korea
| |
Collapse
|
153
|
Gu Y, Servello D, Han Z, Lalchandani RR, Ding JB, Huang K, Gu C. Balanced Activity between Kv3 and Nav Channels Determines Fast-Spiking in Mammalian Central Neurons. iScience 2018; 9:120-137. [PMID: 30390433 PMCID: PMC6218699 DOI: 10.1016/j.isci.2018.10.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 08/11/2018] [Accepted: 10/12/2018] [Indexed: 10/29/2022] Open
Abstract
Fast-spiking (FS) neurons can fire action potentials (APs) up to 1,000 Hz and play key roles in vital functions such as sound location, motor coordination, and cognition. Here we report that the concerted actions of Kv3 voltage-gated K+ (Kv) and Na+ (Nav) channels are sufficient and necessary for inducing and maintaining FS. Voltage-clamp analysis revealed a robust correlation between the Kv3/Nav current ratio and FS. Expressing Kv3 channels alone could convert ∼30%-60% slow-spiking (SS) neurons to FS in culture. In contrast, co-expression of either Nav1.2 or Nav1.6 together with Kv3.1 or Kv3.3, but not alone or with Kv1.2, converted SS to FS with 100% efficiency. Furthermore, RNA-sequencing-based genome-wide analysis revealed that the Kv3/Nav ratio and Kv3 expression levels strongly correlated with the maximal AP frequencies. Therefore, FS is established by the properly balanced activities of Kv3 and Nav channels and could be further fine-tuned by channel biophysical features and localization patterns.
Collapse
Affiliation(s)
- Yuanzheng Gu
- Department of Biological Chemistry and Pharmacology, The Ohio State University, 182 Rightmire Hall, 1060 Carmack Road, Columbus, OH 43210, USA
| | - Dustin Servello
- Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Zhi Han
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; College of Software, Nankai University, Tianjin 300071, China; Regenstrief Institute, Indianapolis, IN 46202, USA
| | - Rupa R Lalchandani
- Department of Neurosurgery, and Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Jun B Ding
- Department of Neurosurgery, and Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Kun Huang
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Regenstrief Institute, Indianapolis, IN 46202, USA; School of Biomedical Engineering, Shenzhen University, Shenzhen 518037, China
| | - Chen Gu
- Department of Biological Chemistry and Pharmacology, The Ohio State University, 182 Rightmire Hall, 1060 Carmack Road, Columbus, OH 43210, USA; Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
154
|
Heermann T, Garrett L, Wurst W, Fuchs H, Gailus-Durner V, Hrabě de Angelis M, Graw J, Hölter SM. Crybb2 Mutations Consistently Affect Schizophrenia Endophenotypes in Mice. Mol Neurobiol 2018; 56:4215-4230. [PMID: 30291584 DOI: 10.1007/s12035-018-1365-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 09/25/2018] [Indexed: 10/28/2022]
Abstract
As part of the βγ-superfamily, βB2-crystallin (CRYBB2) is an ocular structural protein in the lens, and mutation of the corresponding gene can cause cataracts. CRYBB2 also is expressed in non-lens tissue such as the adult mouse brain and is associated with neuropsychiatric disorders such as schizophrenia. Nevertheless, the robustness of this association as well as how CRYBB2 may contribute to disease-relevant phenotypes is unknown. To add further clarity to this issue, we performed a comprehensive analysis of behavioral and neurohistological alterations in mice with an allelic series of mutations in the C-terminal end of the Crybb2 gene. Behavioral phenotyping of these three βB2-mutant lines Crybb2O377, Crybb2Philly, and Crybb2Aey2 included assessment of exploratory activity and anxiety-related behavior in the open field, sensorimotor gating measured by prepulse inhibition (PPI) of the acoustic startle reflex, cognitive performance measured by social discrimination, and spontaneous alternation in the Y-maze. In each mutant line, we also quantified the number of parvalbumin-positive (PV+) GABAergic interneurons in selected brain regions that express CRYBB2. While there were allele-specific differences in individual behaviors and affected brain areas, all three mutant lines exhibited consistent alterations in PPI that paralleled alterations in the PV+ cell number in the thalamic reticular nucleus (TRN). The direction of the PPI change mirrored that of the TRN PV+ cell number thereby suggesting a role for TRN PV+ cell number in modulating PPI. Moreover, as both altered PPI and PV+ cell number are schizophrenia-associated endophenotypes, our result implicates mutated Crybb2 in the development of this neuropsychiatric disorder.
Collapse
Affiliation(s)
- Tamara Heermann
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764, Neuherberg, Germany.,Max Planck Institute of Biochemistry, Munich, Germany
| | - Lillian Garrett
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764, Neuherberg, Germany.,German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764, Neuherberg, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764, Neuherberg, Germany.,Developmental Genetics, Technische Universität München- Weihenstephan, c/o Helmholtz Zentrum München, Munich, Germany.,German Centre of Neurodegenerative Diseases (DZNE), Site Munich, Feodor-Lynen-Str. 17, 81377, Munich, Germany.,Munich Cluster of Systems Neurology (SyNergy), Adolf-Butenandt-Institut, Ludwig-Maximilians-Universität München, Schillerstr.44, 80336, Munich, Germany
| | - Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764, Neuherberg, Germany
| | - Valerie Gailus-Durner
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764, Neuherberg, Germany
| | - Martin Hrabě de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764, Neuherberg, Germany.,Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, 85354, Freising, Germany.,German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Jochen Graw
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764, Neuherberg, Germany
| | - Sabine M Hölter
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764, Neuherberg, Germany. .,German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764, Neuherberg, Germany.
| |
Collapse
|
155
|
Ballesteros A, Sánchez-Torres AM, López-Ilundain JM, Cabrera B, Lobo A, González-Pinto AM, Díaz-Caneja C, Corripio I, Vieta E, de la Serna E, Bobes J, Usall J, Contreras F, Lorente-Omeñaca R, Mezquida G, Bernardo M, Cuesta MJ. Is cognitive impairment associated with antipsychotic dose and anticholinergic equivalent loads in first-episode psychosis? Psychol Med 2018; 48:2247-2256. [PMID: 29331153 DOI: 10.1017/s0033291717003774] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Cognitive deficits are a core feature of early stages in schizophrenia. However, the extent to which antipsychotic (AP) have a deleterious effect on cognitive performance remains under debate. We aim to investigate whether anticholinergic loadings and dose of AP drugs in first episode of psychosis (FEP) in advanced phase of remission are associated with cognitive impairment and the differences between premorbid intellectual quotient (IQ) subgroups. METHODS Two hundred and sixty-six patients participated. The primary outcomes were cognitive dimensions, dopaminergic/anticholinergic load of AP [in chlorpromazine equivalents (Eq-CPZ) and the Anticholinergic Risk Scale (ARS), respectively]. RESULTS Impairments in processing speed, verbal memory and global cognition were significantly associated with high Eq-CPZ and verbal impairment with high ARS score. Moreover, this effect was higher in the low IQ subgroup. CONCLUSIONS Clinicians should be aware of the potential cognitive impairment associated with AP in advanced remission FEP, particularly in lower premorbid IQ patients.
Collapse
Affiliation(s)
| | | | | | - Bibiana Cabrera
- Barcelona Clínic Schizophrenia Unit,Neuroscience Institute,Hospital Clínic de Barcelona,Barcelona,Spain
| | - Antonio Lobo
- Department of Medicine and Psychiatry,University of Zaragoza. Aragon Institute for Health Research (IIS Aragon),Zaragoza,Spain
| | | | | | - Iluminada Corripio
- Network Centre for Biomedical Research in Mental Health (CIBERSAM),Madrid,Spain
| | - Eduard Vieta
- Network Centre for Biomedical Research in Mental Health (CIBERSAM),Madrid,Spain
| | - Elena de la Serna
- Network Centre for Biomedical Research in Mental Health (CIBERSAM),Madrid,Spain
| | - Julio Bobes
- Network Centre for Biomedical Research in Mental Health (CIBERSAM),Madrid,Spain
| | - Judith Usall
- Network Centre for Biomedical Research in Mental Health (CIBERSAM),Madrid,Spain
| | - Fernando Contreras
- Network Centre for Biomedical Research in Mental Health (CIBERSAM),Madrid,Spain
| | | | - Gisela Mezquida
- Barcelona Clínic Schizophrenia Unit,Neuroscience Institute,Hospital Clínic de Barcelona,Barcelona,Spain
| | - Miguel Bernardo
- Barcelona Clínic Schizophrenia Unit,Neuroscience Institute,Hospital Clínic de Barcelona,Barcelona,Spain
| | | |
Collapse
|
156
|
Higher ambient synaptic glutamate at inhibitory versus excitatory neurons differentially impacts NMDA receptor activity. Nat Commun 2018; 9:4000. [PMID: 30275542 PMCID: PMC6167324 DOI: 10.1038/s41467-018-06512-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 08/31/2018] [Indexed: 01/28/2023] Open
Abstract
Selective disruption of synaptic drive to inhibitory neurons could contribute to the pathophysiology of various brain disorders. We have previously identified a GluN2A-selective positive allosteric modulator, GNE-8324, that selectively enhances N-methyl-d-aspartate receptor (NMDAR)-mediated synaptic responses in inhibitory but not excitatory neurons. Here, we demonstrate that differences in NMDAR subunit composition do not underlie this selective potentiation. Rather, a higher ambient glutamate level in the synaptic cleft of excitatory synapses on inhibitory neurons is a key factor. We show that increasing expression of glutamate transporter 1 (GLT-1) eliminates GNE-8324 potentiation in inhibitory neurons, while decreasing GLT-1 activity enables potentiation in excitatory neurons. Our results reveal an unsuspected difference between excitatory synapses onto different neuronal types, and a more prominent activation of synaptic NMDARs by ambient glutamate in inhibitory than excitatory neurons. This difference has implications for tonic NMDAR activity/signaling and the selective modulation of inhibitory neuron activity to treat brain disorders. Inhibitory interneurons play important roles in brain circuits and in several neuropsychiatric disorders. Here, the authors show that excitatory synapses onto interneurons vs. excitatory neurons differ in their ambient synaptic glutamate level, a finding with important implications for selective pharmacological targeting of inhibitory neuron NMDA receptors.
Collapse
|
157
|
Elucidating the developmental trajectories of GABAergic cortical interneuron subtypes. Neurosci Res 2018; 138:26-32. [PMID: 30227162 DOI: 10.1016/j.neures.2018.09.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 08/20/2018] [Accepted: 08/20/2018] [Indexed: 12/21/2022]
Abstract
GABAergic interneurons in the neocortex play pivotal roles in the feedforward and feedback inhibition that control higher order information processing and thus, malfunction in the inhibitory circuits often leads to neurodevelopmental disorders. Very interestingly, a large diversity of morphology, synaptic targeting specificity, electrophysiological properties and molecular expression profiles are found in cortical interneurons, which originate within the distantly located embryonic ganglionic eminences. Here, I will review the still ongoing effort to understand the developmental trajectories of GABAergic cortical interneuron subtypes.
Collapse
|
158
|
Murray JD, Demirtaş M, Anticevic A. Biophysical Modeling of Large-Scale Brain Dynamics and Applications for Computational Psychiatry. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2018; 3:777-787. [PMID: 30093344 PMCID: PMC6537601 DOI: 10.1016/j.bpsc.2018.07.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/11/2018] [Accepted: 07/11/2018] [Indexed: 01/09/2023]
Abstract
Noninvasive neuroimaging has revolutionized the study of the organization of the human brain and how its structure and function are altered in psychiatric disorders. A critical explanatory gap lies in our mechanistic understanding of how systems-level neuroimaging biomarkers emerge from underlying synaptic-level perturbations associated with a disease state. We describe an emerging computational psychiatry approach leveraging biophysically based computational models of large-scale brain dynamics and their potential integration with clinical and pharmacological neuroimaging. In particular, we focus on neural circuit models, which describe how patterns of functional connectivity observed in resting-state functional magnetic resonance imaging emerge from neural dynamics shaped by inter-areal interactions through underlying structural connectivity defining long-range projections. We highlight the importance of local circuit physiological dynamics, in combination with structural connectivity, in shaping the emergent functional connectivity. Furthermore, heterogeneity of local circuit properties across brain areas, which impacts large-scale dynamics, may be critical for modeling whole-brain phenomena and alterations in psychiatric disorders and pharmacological manipulation. Finally, we discuss important directions for future model development and biophysical extensions, which will expand their utility to link clinical neuroimaging to neurobiological mechanisms.
Collapse
Affiliation(s)
- John D Murray
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut.
| | - Murat Demirtaş
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Alan Anticevic
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
159
|
Laminar Distribution of Neurochemically-Identified Interneurons and Cellular Co-expression of Molecular Markers in Epileptic Human Cortex. Neurosci Bull 2018; 34:992-1006. [PMID: 30171525 PMCID: PMC6246828 DOI: 10.1007/s12264-018-0275-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 05/20/2018] [Indexed: 12/20/2022] Open
Abstract
Inhibitory GABAergic interneurons are fundamental elements of cortical circuits and play critical roles in shaping network activity. Dysfunction of interneurons can lead to various brain disorders, including epilepsy, schizophrenia, and anxiety. Based on the electrophysiological properties, cell morphology, and molecular identity, interneurons could be classified into various subgroups. In this study, we investigated the density and laminar distribution of different interneuron types and the co-expression of molecular markers in epileptic human cortex. We found that parvalbumin (PV) and somatostatin (SST) neurons were distributed in all cortical layers except layer I, while tyrosine hydroxylase (TH) and neuropeptide Y (NPY) were abundant in the deep layers and white matter. Cholecystokinin (CCK) neurons showed a high density in layers IV and VI. Neurons with these markers constituted ~7.2% (PV), 2.6% (SST), 0.5% (TH), 0.5% (NPY), and 4.4% (CCK) of the gray-matter neuron population. Double- and triple-labeling revealed that NPY neurons were also SST-immunoreactive (97.7%), and TH neurons were more likely to express SST (34.2%) than PV (14.6%). A subpopulation of CCK neurons (28.0%) also expressed PV, but none contained SST. Together, these results revealed the density and distribution patterns of different interneuron populations and the overlap between molecular markers in epileptic human cortex.
Collapse
|
160
|
Roach BJ, Ford JM, Mathalon DH. Gamma Band Phase Delay in Schizophrenia. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2018; 4:131-139. [PMID: 30314905 DOI: 10.1016/j.bpsc.2018.08.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 08/15/2018] [Accepted: 08/15/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND In 1999, Kwon et al. reported several electroencephalographic gamma band auditory steady-state response (ASSR) abnormalities in schizophrenia, spawning approximately 100 subsequent studies. While many studies replicated the finding of reduced 40-Hz ASSR power in schizophrenia and extended this by showing that 40-Hz phase synchrony (phase-locking factor [PLF]) was also reduced, none attempted to replicate the original phase delay finding of Kwon et al. Accordingly, we measured the 40-Hz ASSR phase-locking angle (PLA) to assess phase delay and examined its differential sensitivity to schizophrenia, relative to power and PLF measures. METHODS To obtain ASSRs, electroencephalography data were recorded from 28 patients with schizophrenia and 25 healthy control subjects listening to repeated 40-Hz 500-ms click trains. Evoked power, total power, PLF, and PLA were calculated after Morlet wavelet time-frequency decomposition of single trial data from electrode Fz. RESULTS In patients with schizophrenia, 40-Hz PLA was significantly reduced (i.e., phase delayed) (p < .0001) and was unrelated to reductions in their 40-Hz power or PLF. PLA discriminated patients from healthy control subjects with 85% accuracy compared with 67% for power and 65% for PLF. CONCLUSIONS Consistent with the original Kwon et al. study, 40-Hz click train-driven gamma oscillations were phase delayed in schizophrenia. Importantly, this phase delay abnormality was substantially larger than the gamma power and phase synchrony abnormalities that have been the focus of prior 40-Hz ASSR studies in schizophrenia. PLA provides a unique neurobiological measure of gamma band abnormalities in schizophrenia, likely reflecting a distinct pathophysiological mechanism from those underlying PLF and power abnormalities.
Collapse
Affiliation(s)
- Brian J Roach
- Mental Health Service, San Francisco Veterans Affairs Health Care System, University of California, San Francisco, San Francisco, California; Northern California Institute for Research and Education, San Francisco Veterans Affairs Medical Center, University of California, San Francisco, San Francisco, California
| | - Judith M Ford
- Mental Health Service, San Francisco Veterans Affairs Health Care System, University of California, San Francisco, San Francisco, California; Department of Psychiatry, University of California, San Francisco, San Francisco, California
| | - Daniel H Mathalon
- Mental Health Service, San Francisco Veterans Affairs Health Care System, University of California, San Francisco, San Francisco, California; Department of Psychiatry, University of California, San Francisco, San Francisco, California.
| |
Collapse
|
161
|
Andersson JD, Matuskey D, Finnema SJ. Positron emission tomography imaging of the γ-aminobutyric acid system. Neurosci Lett 2018; 691:35-43. [PMID: 30102960 DOI: 10.1016/j.neulet.2018.08.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 08/06/2018] [Accepted: 08/09/2018] [Indexed: 01/08/2023]
Abstract
In this review, we summarize the recent development of positron emission tomography (PET) radioligands for γ-aminobutyric acid A (GABAA) receptors and their potential to measure changes in endogenous GABA levels and highlight the clinical and translational applications of GABA-sensitive PET radioligands. We review the basic physiology of the GABA system with a focus on the importance of GABAA receptors in the brain and specifically the benzodiazepine binding site. Challenges for the development of central nervous system radioligands and particularly for radioligands with increased GABA sensitivity are outlined, as well as the status of established benzodiazepine site PET radioligands and agonist GABAA radioligands. We underline the challenge of using allosteric interactions to measure GABA concentrations and review the current state of PET imaging of changes in GABA levels. We conclude that PET tracers with increased GABA sensitivity are required to efficiently measure GABA release and that such a tool could be broadly applied to assess GABA transmission in vivo across several disorders.
Collapse
Affiliation(s)
- Jan D Andersson
- University of Alberta, Medical Isotope and Cyclotron Facility, Edmonton, Canada
| | - David Matuskey
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Sjoerd J Finnema
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA; Center for Psychiatric Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
162
|
Affiliation(s)
- Jeffrey A Lieberman
- From the Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, and the New York State Psychiatric Institute - both in New York
| | - Michael B First
- From the Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, and the New York State Psychiatric Institute - both in New York
| |
Collapse
|
163
|
Gabrb2-knockout mice displayed schizophrenia-like and comorbid phenotypes with interneuron-astrocyte-microglia dysregulation. Transl Psychiatry 2018; 8:128. [PMID: 30013074 PMCID: PMC6048160 DOI: 10.1038/s41398-018-0176-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/30/2018] [Accepted: 06/04/2018] [Indexed: 12/05/2022] Open
Abstract
Intronic polymorphisms of the GABAA receptor β2 subunit gene (GABRB2) under adaptive evolution were associated with schizophrenia and reduced expression, especially of the long isoform which differs in electrophysiological properties from the short isoform. The present study was directed to examining the gene dosage effects of Gabrb2 in knockout mice of both heterozygous (HT) and homozygous (KO) genotypes with respect to possible schizophrenia-like and comorbid phenotypes. The KO mice, and HT mice to a lesser extent, were found to display prepulse inhibition (PPI) deficit, locomotor hyperactivity, stereotypy, sociability impairments, spatial-working and spatial-reference memory deficits, reduced depression and anxiety, and accelerated pentylenetetrazol (PTZ)-induced seizure. In addition, the KO mice were highly susceptible to audiogenic epilepsy. Some of the behavioral phenotypes showed evidence of imprinting, gender effect and amelioration by the antipsychotic risperidone, and the audiogenic epilepsy was inhibited by the antiepileptic diazepam. GABAergic parvalbumin (PV)-positive interneuron dystrophy, astrocyte dystrophy, and extensive microglia activation were observed in the frontotemporal corticolimbic regions, and reduction of newborn neurons was observed in the hippocampus by immunohistochemical staining. The neuroinflammation indicated by microglial activation was accompanied by elevated brain levels of oxidative stress marker malondialdehyde (MDA) and the pro-inflammatory cytokines tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6). These extensive schizophrenia-like and comorbid phenotypes brought about by Gabrb2 knockout, in conjunction with our previous findings on GABRB2 association with schizophrenia, support a pivotal role of GABRB2 in schizophrenia etiology.
Collapse
|
164
|
Kim P, Scott MR, Meador-Woodruff JH. Abnormal expression of ER quality control and ER associated degradation proteins in the dorsolateral prefrontal cortex in schizophrenia. Schizophr Res 2018; 197:484-491. [PMID: 29496332 PMCID: PMC6109614 DOI: 10.1016/j.schres.2018.02.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 11/27/2017] [Accepted: 02/12/2018] [Indexed: 11/19/2022]
Abstract
Abnormalities in posttranslational protein modifications (PTMs) that regulate protein targeting, trafficking, synthesis, and function have been implicated in the pathophysiology of schizophrenia. The endoplasmic reticulum (ER) contains specialized machinery that facilitate protein synthesis, ER entry and exit, quality control, and post-translational processing, steps required for protein maturation. Dysregulation of these systems could represent potential mechanisms for abnormalities of neurotransmitter associated proteins in schizophrenia. We hypothesized that expression of ER processing pathways is dysregulated in schizophrenia. We characterized protein and complex expression of essential components from protein folding, ER quality control (ERQC), and ER associated degradation (ERAD) processes in the dorsolateral prefrontal cortex of 12 matched pairs of elderly schizophrenia and comparison subjects. We found increased expression of proteins associated with recognizing and modifying misfolded proteins, including UDP-glucose/glycoprotein glucosyltransferase 2 (UGGT2), ER degradation enhancing alpha-mannosidase like protein 2 (EDEM2), and synoviolin (SYVN1)/HRD1. As SYVN1/HRD1 is a component of the ubiquitin ligase HRD1-SEL1L complex that facilitates ERAD, we immunoprecipitated SEL1L and measured expression of other proteins in this complex. In schizophrenia, SYVN1/HRD1 and OS-9, ERAD promoters, have increased association with SEL1L, while XTP3-B, which can prevent ERAD of substrates, has decreased association. Abnormal expression of proteins associated with ERQC and ERAD suggests dysregulation in ER localized protein processing pathways in schizophrenia. Interestingly, the deficits we found are not in the protein processing machinery itself, but in proteins that recognize and target incompletely or misfolded proteins. These changes may reflect potential mechanisms of abnormal neurotransmitter associated protein expression previously observed in schizophrenia.
Collapse
Affiliation(s)
- Pitna Kim
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Madeline R Scott
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - James H Meador-Woodruff
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
165
|
Selesnick SA, Piccinini G. Quantum-like behavior without quantum physics II. A quantum-like model of neural network dynamics. J Biol Phys 2018; 44:501-538. [PMID: 29948555 DOI: 10.1007/s10867-018-9504-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/22/2018] [Indexed: 10/14/2022] Open
Abstract
In earlier work, we laid out the foundation for explaining the quantum-like behavior of neural systems in the basic kinematic case of clusters of neuron-like units. Here we extend this approach to networks and begin developing a dynamical theory for them. Our approach provides a novel mathematical foundation for neural dynamics and computation which abstracts away from lower-level biophysical details in favor of information-processing features of neural activity. The theory makes predictions concerning such pathologies as schizophrenia, dementias, and epilepsy, for which some evidence has accrued. It also suggests a model of memory retrieval mechanisms. As further proof of principle, we analyze certain energy-like eigenstates of the 13 three-neuron motif classes according to our theory and argue that their quantum-like superpositional nature has a bearing on their observed structural integrity.
Collapse
Affiliation(s)
- S A Selesnick
- Department of Mathematics and Computer Science, University of Missouri - St. Louis, St. Louis, Missouri, 63121, USA.
| | - Gualtiero Piccinini
- Department of Philosophy, Associate Director, Center for Neurodynamics, University of Missouri - St. Louis, St. Louis, Missouri, 63121, USA
| |
Collapse
|
166
|
Saito H, Kato R, Hashimoto T, Uchida Y, Hase T, Tsuruga K, Takita K, Morimoto Y. Influence of nitrous oxide on granule cell migration in the dentate gyrus of the neonatal rat. Biomed Res 2018; 39:39-45. [PMID: 29467350 DOI: 10.2220/biomedres.39.39] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
For several decades, the neurotoxicities of anesthetics to the developing brain have been reported by many researchers focusing on various phenomena such as apoptosis, neurodegeneration, electrophysiological aberrations, and behavioral abnormalities. According to these reports, signals via N-methyl-D-aspartate receptors (NMDA-r) and/or γ-aminobutyric acid type A receptors (GABAA-r) are implicated in the anesthetic neurotoxicity. On the other hand, during brain development, NMDA-r and GABAA-r are also recognized to play primary roles in neural cell migration. Therefore, anesthetics exposed in this period may influence the neural cell migration of neonates, and increase the number of hilar ectopic granule cells, which are reported to be a cause of continuous neurological deficits. To examine this hypothesis, we investigated immunohistochemically granule cell distribution in the hippocampal dentate gyrus of Wistar/ST rats after nitrous oxide (N2O) exposure. At postnatal day (P) 6, 5-bromo-2'-deoxyuridine (BrdU) was administered to label newly generated cells. Then, rats were divided into groups (n = 6 each group), exposed to 50% N2O at P7, and evaluated at P21. As a result, we found that ectopic ratios (ratio of hilar/total granule cells generated at P6) were decreased in rats at P21 compared with those at P7, and increased in N2O exposed rats for over 120 min compared with the other groups. These results suggest that 50% N2O exposure for over 120 min increases the ratios of ectopic granule cells in the rat dentate gyrus.
Collapse
|
167
|
Katahira T, Miyazaki N, Motoyama J. Immediate effects of maternal separation on the development of interneurons derived from medial ganglionic eminence in the neonatal mouse hippocampus. Dev Growth Differ 2018; 60:278-290. [DOI: 10.1111/dgd.12540] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 04/25/2018] [Accepted: 04/25/2018] [Indexed: 01/21/2023]
Affiliation(s)
- Tatsuya Katahira
- Organization of Advanced Research and Education; Doshisha University; Kyoto Japan
| | | | - Jun Motoyama
- Laboratory of Developmental Neurobiology; Graduate School of Brain Science; Doshisha University; Kyoto Japan
| |
Collapse
|
168
|
Symmonds M, Moran CH, Leite MI, Buckley C, Irani SR, Stephan KE, Friston KJ, Moran RJ. Ion channels in EEG: isolating channel dysfunction in NMDA receptor antibody encephalitis. Brain 2018; 141:1691-1702. [PMID: 29718139 PMCID: PMC6207885 DOI: 10.1093/brain/awy107] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 01/31/2018] [Accepted: 02/22/2018] [Indexed: 12/15/2022] Open
Abstract
See Roberts and Breakspear (doi:10.1093/brain/awy136) for a scientific commentary on this article.Neurological and psychiatric practice frequently lack diagnostic probes that can assess mechanisms of neuronal communication non-invasively in humans. In N-methyl-d-aspartate (NMDA) receptor antibody encephalitis, functional molecular assays are particularly important given the presence of NMDA antibodies in healthy populations, the multifarious symptomology and the lack of radiological signs. Recent advances in biophysical modelling techniques suggest that inferring cellular-level properties of neural circuits from macroscopic measures of brain activity is possible. Here, we estimated receptor function from EEG in patients with NMDA receptor antibody encephalitis (n = 29) as well as from encephalopathic and neurological patient controls (n = 36). We show that the autoimmune patients exhibit distinct fronto-parietal network changes from which ion channel estimates can be obtained using a microcircuit model. Specifically, a dynamic causal model of EEG data applied to spontaneous brain responses identifies a selective deficit in signalling at NMDA receptors in patients with NMDA receptor antibody encephalitis but not at other ionotropic receptors. Moreover, though these changes are observed across brain regions, these effects predominate at the NMDA receptors of excitatory neurons rather than at inhibitory interneurons. Given that EEG is a ubiquitously available clinical method, our findings suggest a unique re-purposing of EEG data as an assay of brain network dysfunction at the molecular level.
Collapse
Affiliation(s)
- Mkael Symmonds
- Division of Clinical Neurology, John Radcliffe Hospital, Oxford, UK
- Department of Clinical Neurophysiology, John Radcliffe Hospital, Oxford, UK
- Epilepsy Research Group, Nuffield Department of Clinical Neurosciences, Oxford University, John Radcliffe Hospital, Oxford, Oxford, UK
| | - Catherine H Moran
- Department of Neurological Surgery, Beaumont Hospital, Dublin, Ireland
| | - M Isabel Leite
- Division of Clinical Neurology, John Radcliffe Hospital, Oxford, UK
- Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, Oxford University, John Radcliffe Hospital, Oxford, Oxford, UK
| | - Camilla Buckley
- Division of Clinical Neurology, John Radcliffe Hospital, Oxford, UK
| | - Sarosh R Irani
- Division of Clinical Neurology, John Radcliffe Hospital, Oxford, UK
- Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, Oxford University, John Radcliffe Hospital, Oxford, Oxford, UK
| | - Klaas Enno Stephan
- Translational Neuromodeling Unit, Institute for Biomedical Engineering, University of Zurich and ETH Zurich, 6 Wilfriedstrasse, Zurich, Switzerland
| | - Karl J Friston
- Wellcome Trust Centre for Neuroimaging, University College London, 12 Queen Square, London, UK
| | - Rosalyn J Moran
- Department of Engineering Mathematics, Merchant Venturers School of Engineering, University of Bristol, 75 Woodland Rd, Bristol, UK
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
169
|
Imai H, Shoji H, Ogata M, Kagawa Y, Owada Y, Miyakawa T, Sakimura K, Terashima T, Katsuyama Y. Dorsal Forebrain-Specific Deficiency of Reelin-Dab1 Signal Causes Behavioral Abnormalities Related to Psychiatric Disorders. Cereb Cortex 2018; 27:3485-3501. [PMID: 26762856 DOI: 10.1093/cercor/bhv334] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Reelin-Dab1 signaling is involved in brain development and neuronal functions. The abnormalities in the signaling through either reduction of Reelin and Dab1 gene expressions or the genomic mutations in the brain have been reported to be associated with psychiatric disorders. However, it has not been clear if the deficiency in Reelin-Dab1 signaling is responsible for symptoms of the disorders. Here, to examine the function of Reelin-Dab1 signaling in the forebrain, we generated dorsal forebrain-specific Dab1 conditional knockout mouse (Dab1 cKO) and performed a behavioral test battery on the Dab1 cKO mice. Although conventional Dab1 null mutant mice exhibit cerebellar atrophy and cerebellar ataxia, the Dab1 cKO mice had normal cerebellum and showed no motor dysfunction. Dab1 cKO mice exhibited behavioral abnormalities, including hyperactivity, decreased anxiety-like behavior, and impairment of working memory, which are reminiscent of symptoms observed in patients with psychiatric disorders such as schizophrenia and bipolar disorder. These results suggest that deficiency of Reelin-Dab1 signal in the dorsal forebrain is involved in the pathogenesis of some symptoms of human psychiatric disorders.
Collapse
Affiliation(s)
- Hideaki Imai
- Division of Developmental Neurobiology, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan
| | - Hirotaka Shoji
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake 470-1192, Japan.,Japan Science and Technology Agency, Core Research for Evolutional Science and Technology, Kawaguchi 332-0012, Japan
| | - Masaki Ogata
- Department of Organ Anatomy, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Yoshiteru Kagawa
- Department of Organ Anatomy, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Yuji Owada
- Department of Organ Anatomy, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake 470-1192, Japan.,Japan Science and Technology Agency, Core Research for Evolutional Science and Technology, Kawaguchi 332-0012, Japan.,Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Toshio Terashima
- Division of Developmental Neurobiology, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan
| | - Yu Katsuyama
- Division of Developmental Neurobiology, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan.,Department of Organ Anatomy, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| |
Collapse
|
170
|
Cardarelli RA, Martin R, Jaaro-Peled H, Sawa A, Powell EM, O'Donnell P. Dominant-Negative DISC1 Alters the Dopaminergic Modulation of Inhibitory Interneurons in the Mouse Prefrontal Cortex. MOLECULAR NEUROPSYCHIATRY 2018; 4:20-29. [PMID: 29998115 DOI: 10.1159/000488030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 02/26/2018] [Indexed: 11/19/2022]
Abstract
A truncated disrupted in schizophrenia 1 (Disc1) gene increases the risk of psychiatric disorders, probably affecting cortical interneurons. Here, we sought to determine whether this cell population is affected in mice carrying a truncated (Disc1) allele (DN-DISC1). We utilized whole cell recordings to assess electrophysiological properties and modulation by dopamine (DA) in two classes of interneurons: fast-spiking (FS) and low threshold-spiking (LTS) interneurons in wild-type and DN-DISC1 mice. In DN-DISC1 mice, FS interneurons, but not LTS interneurons, exhibited altered action potentials. Further, the perineuronal nets that surround FS interneurons exhibited abnormal morphology in DN-DISC1 mice, and the DA modulation of this cell type was altered in DN-DISC1 mice. We conclude that early-life manipulation of a gene associated with risk of psychiatric disease can result in dysfunction, but not loss, of specific GABAergic interneurons. The resulting alteration of excitatory-inhibitory balance is a critical element in DISC1 pathophysiology.
Collapse
Affiliation(s)
- Ross A Cardarelli
- Program in Neuroscience, University of Maryland Medical School, Baltimore, Maryland, USA
| | - Rolicia Martin
- Department of Anatomy and Neurobiology, University of Maryland Medical School, Baltimore, Maryland, USA
| | - Hanna Jaaro-Peled
- Department of Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Akira Sawa
- Department of Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Elizabeth M Powell
- Department of Anatomy and Neurobiology, University of Maryland Medical School, Baltimore, Maryland, USA.,Department of Psychiatry, University of Maryland Medical School, Baltimore, Maryland, USA
| | - Patricio O'Donnell
- Program in Neuroscience, University of Maryland Medical School, Baltimore, Maryland, USA.,Department of Anatomy and Neurobiology, University of Maryland Medical School, Baltimore, Maryland, USA.,Department of Psychiatry, University of Maryland Medical School, Baltimore, Maryland, USA
| |
Collapse
|
171
|
Neuropsychiatric Phenotypes Produced by GABA Reduction in Mouse Cortex and Hippocampus. Neuropsychopharmacology 2018; 43:1445-1456. [PMID: 29362511 PMCID: PMC5916365 DOI: 10.1038/npp.2017.296] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 10/25/2017] [Accepted: 11/24/2017] [Indexed: 01/09/2023]
Abstract
Whereas cortical GAD67 reduction and subsequent GABA level decrease are consistently observed in schizophrenia and depression, it remains unclear how these GABAergic abnormalities contribute to specific symptoms. We modeled cortical GAD67 reduction in mice, in which the Gad1 gene is genetically ablated from ~50% of cortical and hippocampal interneurons. Mutant mice showed a reduction of tissue GABA in the hippocampus and cortex including mPFC, and exhibited a cluster of effort-based behavior deficits including decreased home-cage wheel running and increased immobility in both tail suspension and forced swim tests. Since saccharine preference, progressive ratio responding to food, and learned helplessness task were normal, such avolition-like behavior could not be explained by anhedonia or behavioral despair. In line with the prevailing view that dopamine in anterior cingulate cortex (ACC) plays a role in evaluating effort cost for engaging in actions, we found that tail-suspension triggered dopamine release in ACC of controls, which was severely attenuated in the mutant mice. Conversely, ACC dopamine release by progressive ratio responding to reward, during which animals were allowed to effortlessly perform the nose-poking, was not affected in mutants. These results suggest that cortical GABA reduction preferentially impairs the effort-based behavior which requires much effort with little benefit, through a deficit of ACC dopamine release triggered by high-effort cost behavior, but not by reward-seeking behavior. Collectively, a subset of negative symptoms with a reduced willingness to expend costly effort, often observed in patients with schizophrenia and depression, may be attributed to cortical GABA level reduction.
Collapse
|
172
|
Włodarczyk A. Benzodiazepine use in schizophrenia. Schizophr Res 2018; 195:576. [PMID: 28958478 DOI: 10.1016/j.schres.2017.09.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 09/19/2017] [Accepted: 09/22/2017] [Indexed: 10/18/2022]
Affiliation(s)
- Adam Włodarczyk
- Department of Psychiatry, Medical University of Gdansk, Dębinki 7 St. build. 25, 80-952 Gdansk, Poland.
| |
Collapse
|
173
|
Newman EL, Terunuma M, Wang TL, Hewage N, Bicakci MB, Moss SJ, DeBold JF, Miczek KA. A Role for Prefrontal Cortical NMDA Receptors in Murine Alcohol-Heightened Aggression. Neuropsychopharmacology 2018; 43:1224-1234. [PMID: 29052618 PMCID: PMC5916347 DOI: 10.1038/npp.2017.253] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/03/2017] [Accepted: 10/15/2017] [Indexed: 11/09/2022]
Abstract
Alcohol is associated with nearly half of all violent crimes committed in the United States; yet, a potential neural basis for this type of pathological aggression remains elusive. Alcohol may act on N-methyl-d-aspartate receptors (NMDARs) within cortical circuits to impede processing and to promote aggression. Here, male mice were characterized as alcohol-heightened (AHAs) or alcohol non-heightened aggressors (ANAs) during resident-intruder confrontations after self-administering 1.0 g/kg alcohol (6% w/v) or water. Alcohol produced a pathological-like pattern of aggression in AHAs; these mice shifted their bites to more vulnerable locations on the body of a submissive animal, including the anterior back and ventrum after consuming alcohol. In addition, through immunoblotting, we found that AHAs overexpressed the NMDAR GluN2D subunit in the prefrontal cortex (PFC) as compared to ANAs while the two phenotypes expressed similar levels of GluN1, GluN2A and GluN2B. After identifying several behavioral and molecular characteristics that distinguish AHAs from ANAs, we tested additional mice for their aggression following preferential antagonism of GluN2D-containing NMDARs. In these experiments, groups of AHAs and ANAs self-administered 1.0 g/kg alcohol (6% w/v) or water before receiving intraperitoneal (i.p.) doses of ketamine or memantine, or infusions of memantine directly into the prelimbic (PLmPFC) or infralimbic medial PFC (ILmPFC). Moderate doses of IP ketamine, IP memantine, or intra-PLmPFC memantine increased aggression in AHAs, but only in the absence of alcohol. Prior alcohol intake blocked the pro-aggressive effects of ketamine or memantine. In contrast, only memantine, administered systemically or intra-PLmPFC, interacted with prior alcohol intake to escalate aggression in ANAs. Intra-ILmPFC memantine had no effect on aggression in either AHAs or ANAs. In sum, this work illustrates a potential role of GluN2D-containing NMDARs in the PLmPFC in alcohol-heightened aggression. GluN2D-containing NMDARs are highly expressed on cortical parvalbumin-containing interneurons, suggesting that, in a subset of individuals, alcohol may functionally alter signal integration within cortical microcircuits to dysregulate threat reactivity and promote aggression. This work suggests that targeting GluN2D-NMDARs may be of use in reducing the impact of alcohol-related violence in the human population.
Collapse
Affiliation(s)
- Emily L Newman
- Psychology Department, Tufts University, Medford, MA, USA
| | - Miho Terunuma
- Division of Oral Biochemistry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Tiffany L Wang
- Psychology Department, Tufts University, Medford, MA, USA
| | - Nishani Hewage
- Psychology Department, Tufts University, Medford, MA, USA
| | | | - Stephen J Moss
- Department of Neuroscience, Sackler School of Graduate Biomedical Sciences, Boston, MA, USA
| | | | - Klaus A Miczek
- Psychology Department, Tufts University, Medford, MA, USA
- Department of Neuroscience, Sackler School of Graduate Biomedical Sciences, Boston, MA, USA
| |
Collapse
|
174
|
Xu MY, Wong AHC. GABAergic inhibitory neurons as therapeutic targets for cognitive impairment in schizophrenia. Acta Pharmacol Sin 2018; 39:733-753. [PMID: 29565038 DOI: 10.1038/aps.2017.172] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 12/25/2017] [Indexed: 12/24/2022] Open
Abstract
Schizophrenia is considered primarily as a cognitive disorder. However, functional outcomes in schizophrenia are limited by the lack of effective pharmacological and psychosocial interventions for cognitive impairment. GABA (gamma-aminobutyric acid) interneurons are the main inhibitory neurons in the central nervous system (CNS), and they play a critical role in a variety of pathophysiological processes including modulation of cortical and hippocampal neural circuitry and activity, cognitive function-related neural oscillations (eg, gamma oscillations) and information integration and processing. Dysfunctional GABA interneuron activity can disrupt the excitatory/inhibitory (E/I) balance in the cortex, which could represent a core pathophysiological mechanism underlying cognitive dysfunction in schizophrenia. Recent research suggests that selective modulation of the GABAergic system is a promising intervention for the treatment of schizophrenia-associated cognitive defects. In this review, we summarized evidence from postmortem and animal studies for abnormal GABAergic neurotransmission in schizophrenia, and how altered GABA interneurons could disrupt neuronal oscillations. Next, we systemically reviewed a variety of up-to-date subtype-selective agonists, antagonists, positive and negative allosteric modulators (including dual allosteric modulators) for α5/α3/α2 GABAA and GABAB receptors, and summarized their pro-cognitive effects in animal behavioral tests and clinical trials. Finally, we also discuss various representative histone deacetylases (HDAC) inhibitors that target GABA system through epigenetic modulations, GABA prodrug and presynaptic GABA transporter inhibitors. This review provides important information on current potential GABA-associated therapies and future insights for development of more effective treatments.
Collapse
|
175
|
Riordan AJ, Schaler AW, Fried J, Paine TA, Thornton JE. Estradiol and luteinizing hormone regulate recognition memory following subchronic phencyclidine: Evidence for hippocampal GABA action. Psychoneuroendocrinology 2018. [PMID: 29529524 DOI: 10.1016/j.psyneuen.2018.02.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The cognitive symptoms of schizophrenia are poorly understood and difficult to treat. Estrogens may mitigate these symptoms via unknown mechanisms. To examine these mechanisms, we tested whether increasing estradiol (E) or decreasing luteinizing hormone (LH) could mitigate short-term episodic memory loss in a phencyclidine (PCP) model of schizophrenia. We then assessed whether changes in cortical or hippocampal GABA may underlie these effects. Female rats were ovariectomized and injected subchronically with PCP. To modulate E and LH, animals received estradiol capsules or Antide injections. Short-term episodic memory was assessed using the novel object recognition task (NORT). Brain expression of GAD67 was analyzed via western blot, and parvalbumin-containing cells were counted using immunohistochemistry. Some rats received hippocampal infusions of a GABAA agonist, GABAA antagonist, or GAD inhibitor before behavioral testing. We found that PCP reduced hippocampal GAD67 and abolished recognition memory. Antide restored hippocampal GAD67 and rescued recognition memory in PCP-treated animals. Estradiol prevented PCP's amnesic effect in NORT but failed to restore hippocampal GAD67. PCP did not cause significant differences in number of parvalbumin-expressing cells or cortical expression of GAD67. Hippocampal infusions of a GABAA agonist restored recognition memory in PCP-treated rats. Blocking hippocampal GAD or GABAA receptors in ovx animals reproduced recognition memory loss similar to PCP and inhibited estradiol's protection of recognition memory in PCP-treated animals. In summary, decreasing LH or increasing E can lessen short-term episodic memory loss, as measured by novel object recognition, in a PCP model of schizophrenia. Alterations in hippocampal GABA may contribute to both PCP's effects on recognition memory and the hormones' ability to prevent or reverse them.
Collapse
Affiliation(s)
- Alexander J Riordan
- Oberlin College, Neuroscience Department, 119 Woodland St, Oberlin, OH 44074, USA.
| | - Ari W Schaler
- Oberlin College, Neuroscience Department, 119 Woodland St, Oberlin, OH 44074, USA
| | - Jenny Fried
- Oberlin College, Neuroscience Department, 119 Woodland St, Oberlin, OH 44074, USA
| | - Tracie A Paine
- Oberlin College, Neuroscience Department, 119 Woodland St, Oberlin, OH 44074, USA
| | - Janice E Thornton
- Oberlin College, Neuroscience Department, 119 Woodland St, Oberlin, OH 44074, USA
| |
Collapse
|
176
|
Excitatory and inhibitory synaptic dysfunction in mania: an emerging hypothesis from animal model studies. Exp Mol Med 2018; 50:1-11. [PMID: 29628501 PMCID: PMC5938027 DOI: 10.1038/s12276-018-0028-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 11/29/2017] [Indexed: 12/26/2022] Open
Abstract
Bipolar disorder (BD) is a common psychiatric disorder characterized by recurrent mood swings between depression and mania, and is associated with high treatment costs. The existence of manic episodes is the defining feature of BD, during which period, patients experience extreme elevation in activity, energy, and mood, with changes in sleep patterns that together severely impair their ability to function in daily life. Despite some limitations in recapitulating the complex features of human disease, several rodent models of mania have been generated and characterized, which have provided important insights toward understanding its underlying pathogenic mechanisms. Among the mechanisms, neuronal excitatory and inhibitory (E/I) synaptic dysfunction in some brain regions, including the frontal cortex, hippocampus, and striatum, is an emerging hypothesis explaining mania. In this review, we highlight recent studies of rodent manic models having impairments in the E/I synaptic development and function. We also summarize the molecular and functional changes of E/I synapses by some mood stabilizers that may contribute to the therapeutic efficacy of drugs. Furthermore, we discuss potential future directions in the study of this emerging hypothesis to better connect the outcomes of basic research to the treatment of patients with this devastating mental illness. Studies in rodents offer insights into bipolar disorder that may help understanding and treatment of this common and debilitating condition. Kihoon Han and colleagues at Korea University in Seoul review research using mice and rats to model the episodes of mania in patients with bipolar disorder. The research supports an emerging hypothesis implicating specific problems with nervous transmission in the brain in the onset of mania. The hypothesis suggests that the transmission of signals between particular nerve cells whose normal function is either to excite or to inhibit other nerve cells may be involved. It also indicates regions of the brain most involved in manic episodes. Changes at the affected nerve junctions—called synapses—brought about by mood-stabilizing drugs are examined. The hypothesis suggests new approaches to treatment options for researchers to explore.
Collapse
|
177
|
Ravikrishnan A, Gandhi PJ, Shelkar GP, Liu J, Pavuluri R, Dravid SM. Region-specific Expression of NMDA Receptor GluN2C Subunit in Parvalbumin-Positive Neurons and Astrocytes: Analysis of GluN2C Expression using a Novel Reporter Model. Neuroscience 2018; 380:49-62. [PMID: 29559384 DOI: 10.1016/j.neuroscience.2018.03.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/26/2018] [Accepted: 03/09/2018] [Indexed: 12/12/2022]
Abstract
Hypofunction of NMDA receptors in parvalbumin (PV)-positive interneurons has been proposed as a potential mechanism for cortical abnormalities and symptoms in schizophrenia. GluN2C-containing receptors have been linked to this hypothesis due to the higher affinity of psychotomimetic doses of ketamine for GluN1/2C receptors. However, the precise cell-type expression of GluN2C subunit remains unknown. We describe the expression of the GluN2C subunit using a novel EGFP reporter model. We observed EGFP(GluN2C) localization in PV-positive neurons in the nucleus reticularis of the thalamus, globus pallidus externa and interna, ventral pallidum and substantia nigra. In contrast, EGFP(GluN2C)-expressing cells did not co-localize with PV-positive neurons in the cortex, striatum, hippocampus or amygdala. Instead, EGFP(GluN2C) expression in these regions co-localized with an astrocytic marker. We confirmed functional expression of GluN2C-containing receptors in the PV-neurons in substantia nigra and cortical astrocytes using electrophysiology. GluN2C was found to be enriched in several first-order and higher order thalamic nuclei. Interestingly, we found that a previous GluN2C β-gal reporter model excluded expression from PV-neurons and certain thalamic nuclei but exhibited expression in the retrosplenial cortex. GluN2C's unique distribution in neuronal and non-neuronal cells in a brain region-specific manner raises interesting questions regarding the role of GluN2C-containing receptors in the central nervous system.
Collapse
Affiliation(s)
| | - Pauravi J Gandhi
- Department of Pharmacology, Creighton University, Omaha, NE 68178, USA
| | - Gajanan P Shelkar
- Department of Pharmacology, Creighton University, Omaha, NE 68178, USA
| | - Jinxu Liu
- Department of Pharmacology, Creighton University, Omaha, NE 68178, USA
| | | | - Shashank M Dravid
- Department of Pharmacology, Creighton University, Omaha, NE 68178, USA.
| |
Collapse
|
178
|
Olloquequi J, Cornejo-Córdova E, Verdaguer E, Soriano FX, Binvignat O, Auladell C, Camins A. Excitotoxicity in the pathogenesis of neurological and psychiatric disorders: Therapeutic implications. J Psychopharmacol 2018; 32:265-275. [PMID: 29444621 DOI: 10.1177/0269881118754680] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurological and psychiatric disorders are leading contributors to the global disease burden, having a serious impact on the quality of life of both patients and their relatives. Although the molecular events underlying these heterogeneous diseases remain poorly understood, some studies have raised the idea of common mechanisms involved. In excitotoxicity, there is an excessive activation of glutamate receptors by excitatory amino acids, leading to neuronal damage. Thus, the excessive release of glutamate can lead to a dysregulation of Ca2+ homeostasis, triggering the production of free radicals and oxidative stress, mitochondrial dysfunction and eventually cell death. Although there is a consensus in considering excitotoxicity as a hallmark in most neurodegenerative diseases, increasing evidence points to the relevant role of this pathological mechanism in other illnesses affecting the central nervous system. Consequently, antagonists of glutamate receptors are used in current treatments or in clinical trials in both neurological and psychiatric disorders. However, drugs modulating other aspects of the excitotoxic mechanism could be more beneficial. This review discusses how excitotoxicity is involved in the pathogenesis of different neurological and psychiatric disorders and the promising strategies targeting the excitotoxic insult.
Collapse
Affiliation(s)
- Jordi Olloquequi
- 1 Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | | | - Ester Verdaguer
- 3 Departament de Biologia Cellular, Fisiologia i Immunologia, Universitat de Barcelona, Spain.,4 Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,5 Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Francesc X Soriano
- 3 Departament de Biologia Cellular, Fisiologia i Immunologia, Universitat de Barcelona, Spain.,5 Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Octavio Binvignat
- 6 Laboratorio de Ciencias Morfológicas, Pontificia Universidad Católica de Valparaíso, Chile
| | - Carme Auladell
- 3 Departament de Biologia Cellular, Fisiologia i Immunologia, Universitat de Barcelona, Spain.,4 Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,5 Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Antoni Camins
- 4 Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,5 Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,7 Departament de Farmacologia, Toxicologia i Química Terapèutica, Universitat de Barcelona, Spain
| |
Collapse
|
179
|
Ramyead A, Kometer M, Studerus E, Andreou C, Ward LM, Riecher-Rössler A. Abnormal brain connectivity during error-monitoring in the psychosis high-risk state. Schizophr Res 2018; 193:261-262. [PMID: 28689757 DOI: 10.1016/j.schres.2017.06.055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 06/22/2017] [Accepted: 06/27/2017] [Indexed: 10/19/2022]
Affiliation(s)
- Avinash Ramyead
- Department of Psychiatry, Weill Institute for Neurosciences, University of California, San Francisco, (UCSF), San Francisco, USA; University of Basel Psychiatric Hospital, Center for Gender Research and Early Detection, Basel, Switzerland
| | - Michael Kometer
- Neuropsychopharmacology and Brain Imaging Research Unit, Department of Psychiatry, Psychotherapy and Psychosomatics, Hospital of Psychiatry, University of Zurich, Switzerland
| | - Erich Studerus
- University of Basel Psychiatric Hospital, Center for Gender Research and Early Detection, Basel, Switzerland
| | - Christina Andreou
- University of Basel Psychiatric Hospital, Center for Gender Research and Early Detection, Basel, Switzerland
| | - Lawrence M Ward
- Department of Psychology and Brain Research Centre, University of British Columbia, Vancouver, BC, Canada
| | - Anita Riecher-Rössler
- University of Basel Psychiatric Hospital, Center for Gender Research and Early Detection, Basel, Switzerland.
| |
Collapse
|
180
|
Chiu PW, Lui SSY, Hung KSY, Chan RCK, Chan Q, Sham PC, Cheung EFC, Mak HKF. In vivo gamma-aminobutyric acid and glutamate levels in people with first-episode schizophrenia: A proton magnetic resonance spectroscopy study. Schizophr Res 2018; 193:295-303. [PMID: 28751130 DOI: 10.1016/j.schres.2017.07.021] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 07/07/2017] [Accepted: 07/07/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND Gamma-aminobutyric acid (GABA) dysfunction and its consequent imbalance are implicated in the pathophysiology of schizophrenia. Reduced GABA production would lead to a disinhibition of glutamatergic neurons and subsequently cause a disruption of the modulation between GABAergic interneurons and glutamatergic neurons. In this study, levels of GABA, Glx (summation of glutamate and glutamine), and other metabolites in the anterior cingulate cortex were measured and compared between first-episode schizophrenia subjects and healthy controls (HC). Diagnostic potential of GABA and Glx as upstream biomarkers for schizophrenia was explored. METHODS Nineteen first-episode schizophrenia subjects and fourteen HC participated in this study. Severity of clinical symptoms of patients was measured with Positive and Negative Syndrome Scale (PANSS). Metabolites were measured using proton magnetic resonance spectroscopy, and quantified using internal water as reference. RESULTS First-episode schizophrenia subjects revealed reduced GABA and myo-inositol (mI), and increased Glx and choline (Cho), compared to HC. No significant correlation was found between metabolite levels and PANSS scores. Receiver operator characteristics analyses showed Glx had higher sensitivity and specificity (84.2%, 92.9%) compared to GABA (73.7%, 64.3%) for differentiating schizophrenia patients from HC. Combined model of both GABA and Glx revealed the best sensitivity and specificity (89.5%, 100%). CONCLUSION This study simultaneously showed reduction in GABA and elevation in Glx in first-episode schizophrenia subjects, and this might provide insights on explaining the disruption of modulation between GABAergic interneurons and glutamatergic neurons. Elevated Cho might indicate increased membrane turnover; whereas reduced mI might reflect dysfunction of the signal transduction pathway. In vivo Glx and GABA revealed their diagnostic potential for schizophrenia.
Collapse
Affiliation(s)
- P W Chiu
- Department of Diagnostic Radiology, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
| | - Simon S Y Lui
- Castle Peak Hospital, Hong Kong, China; Neuropsychology and Applied Cognitive Neuroscience Laboratory, CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | | | - Raymond C K Chan
- Neuropsychology and Applied Cognitive Neuroscience Laboratory, CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China; Department of Psychiatry, The University of Hong Kong, Hong Kong, China
| | | | - P C Sham
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China; Department of Psychiatry, The University of Hong Kong, Hong Kong, China
| | | | - Henry K F Mak
- Department of Diagnostic Radiology, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China; Alzheimer's Disease Research Network, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
181
|
Kotzadimitriou D, Nissen W, Paizs M, Newton K, Harrison PJ, Paulsen O, Lamsa K. Neuregulin 1 Type I Overexpression Is Associated with Reduced NMDA Receptor-Mediated Synaptic Signaling in Hippocampal Interneurons Expressing PV or CCK. eNeuro 2018; 5:ENEURO.0418-17.2018. [PMID: 29740596 PMCID: PMC5938717 DOI: 10.1523/eneuro.0418-17.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/25/2018] [Accepted: 02/28/2018] [Indexed: 11/21/2022] Open
Abstract
Hypofunction of N-methyl-d-aspartate receptors (NMDARs) in inhibitory GABAergic interneurons is implicated in the pathophysiology of schizophrenia (SZ), a heritable disorder with many susceptibility genes. However, it is still unclear how SZ risk genes interfere with NMDAR-mediated synaptic transmission in diverse inhibitory interneuron populations. One putative risk gene is neuregulin 1 (NRG1), which signals via the receptor tyrosine kinase ErbB4, itself a schizophrenia risk gene. The type I isoform of NRG1 shows increased expression in the brain of SZ patients, and ErbB4 is enriched in GABAergic interneurons expressing parvalbumin (PV) or cholecystokinin (CCK). Here, we investigated ErbB4 expression and synaptic transmission in interneuronal populations of the hippocampus of transgenic mice overexpressing NRG1 type I (NRG1tg-type-I mice). Immunohistochemical analyses confirmed that ErbB4 was coexpressed with either PV or CCK in hippocampal interneurons, but we observed a reduced number of ErbB4-immunopositive interneurons in the NRG1tg-type-I mice. NMDAR-mediated currents in interneurons expressing PV (including PV+ basket cells) or CCK were reduced in NRG1tg-type-I mice compared to their littermate controls. We found no difference in AMPA receptor-mediated currents. Optogenetic activation (5 pulses at 20 Hz) of local glutamatergic fibers revealed a decreased NMDAR-mediated contribution to disynaptic GABAergic inhibition of pyramidal cells in the NRG1tg-type-I mice. GABAergic synaptic transmission from either PV+ or CCK+ interneurons, and glutamatergic transmission onto pyramidal cells, did not significantly differ between genotypes. The results indicate that synaptic NMDAR-mediated signaling in hippocampal interneurons is sensitive to chronically elevated NGR1 type I levels. This may contribute to the pathophysiological consequences of increased NRG1 expression in SZ.
Collapse
Affiliation(s)
| | - Wiebke Nissen
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Melinda Paizs
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, 6720, Hungary
| | - Kathryn Newton
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Paul J. Harrison
- Department of Psychiatry, University of Oxford, and Oxford Health NHS Foundation Trust, Oxford, UK
| | - Ole Paulsen
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Karri Lamsa
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, 6720, Hungary
| |
Collapse
|
182
|
Réus GZ, Becker IRT, Scaini G, Petronilho F, Oses JP, Kaddurah-Daouk R, Ceretta LB, Zugno AI, Dal-Pizzol F, Quevedo J, Barichello T. The inhibition of the kynurenine pathway prevents behavioral disturbances and oxidative stress in the brain of adult rats subjected to an animal model of schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2018; 81:55-63. [PMID: 29030243 DOI: 10.1016/j.pnpbp.2017.10.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 10/09/2017] [Accepted: 10/09/2017] [Indexed: 12/19/2022]
Abstract
Evidence has shown that the kynurenine pathway (KP) plays a role in the onset of oxidative stress and also in the pathophysiology of schizophrenia. The aim of this study was to use a pharmacological animal model of schizophrenia induced by ketamine to investigate if KP inhibitors could protect the brains of Wistar rats against oxidative stress and behavioral changes. Ketamine, injected at the dose of 25mg/kg, increased spontaneous locomotor activity. However, the inhibitors of tryptophan 2,3-dioxygenase (TDO), indoleamine 2,3-dioxygenase (IDO) and kynurenine-3-monooxygenase (KMO) were able to reverse these changes. In addition, the IDO inhibitor prevented lipid peroxidation, and decreased the levels of protein carbonyl in the prefrontal cortex (PFC), hippocampus and striatum. It also increased the activity of superoxide dismutase (SOD) in the hippocampus, as well as increasing the levels of catalase activity in the PFC and hippocampus. The TDO inhibitor prevented lipid damage in the striatum and reduced the levels of protein carbonyl in the hippocampus and striatum. Also, the TDO inhibitor increased the levels of SOD activity in the striatum and CAT activity in the hippocampus of ketamine-induced pro-oxidant effects. Lipid damage was not reversed by the KMO inhibitor. The KMO inhibitor increased the levels of SOD activity in the hippocampus, and reduced the levels of protein carbonyl while elevating the levels of CAT activity in the striatum of rats that had been injected with ketamine. Our findings revealed that the KP pathway could be a potential mechanism by which a schizophrenia animal model induced by ketamine could cause interference by producing behavioral disturbance and inducing oxidative stress in the brain, suggesting that the inhibition of the KP pathway could be a potential target in treating schizophrenia.
Collapse
Affiliation(s)
- Gislaine Z Réus
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| | - Indianara R T Becker
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Giselli Scaini
- Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Fabricia Petronilho
- Laboratory of Clinical and Experimental Pathophysiology, Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Tubarão, SC, Brazil
| | - Jean P Oses
- Translational Science on Brain Disorders, Department of Health and Behavior, Catholic University of Pelotas, Pelotas, RS, Brazil
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke Institute for Brain Sciences, Duke University, Durham, NC, USA; Programa de Pós-graduação em Saúde Coletiva, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Luciane B Ceretta
- Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| | - Alexandra I Zugno
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - João Quevedo
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil; Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| | - Tatiana Barichello
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Laboratory of Experimental Microbiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| |
Collapse
|
183
|
Hagihara H, Catts VS, Katayama Y, Shoji H, Takagi T, Huang FL, Nakao A, Mori Y, Huang KP, Ishii S, Graef IA, Nakayama KI, Shannon Weickert C, Miyakawa T. Decreased Brain pH as a Shared Endophenotype of Psychiatric Disorders. Neuropsychopharmacology 2018; 43:459-468. [PMID: 28776581 PMCID: PMC5770757 DOI: 10.1038/npp.2017.167] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 07/14/2017] [Accepted: 08/01/2017] [Indexed: 01/25/2023]
Abstract
Although the brains of patients with schizophrenia and bipolar disorder exhibit decreased brain pH relative to those of healthy controls upon postmortem examination, it remains controversial whether this finding reflects a primary feature of the diseases or is a result of confounding factors such as medication and agonal state. To date, systematic investigation of brain pH has not been undertaken using animal models that can be studied without confounds inherent in human studies. In the present study, we first reevaluated the pH of the postmortem brains of patients with schizophrenia and bipolar disorder by conducting a meta-analysis of existing data sets from 10 studies. We then measured pH, lactate levels, and related metabolite levels in brain homogenates from five neurodevelopmental mouse models of psychiatric disorders, including schizophrenia, bipolar disorder, and autism spectrum disorder. All mice were drug naive with the same agonal state, postmortem interval, and age within each strain. Our meta-analysis revealed that brain pH was significantly lower in patients with schizophrenia and bipolar disorder than in control participants, even when a few potential confounding factors (postmortem interval, age, and history of antipsychotic use) were considered. In animal experiments, we observed significantly lower pH and higher lactate levels in the brains of model mice relative to controls, as well as a significant negative correlation between pH and lactate levels. Our findings suggest that lower pH associated with increased lactate levels is not a mere artifact, but rather implicated in the underlying pathophysiology of schizophrenia and bipolar disorder.
Collapse
Affiliation(s)
- Hideo Hagihara
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Vibeke S Catts
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, Australia,School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Yuta Katayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Hirotaka Shoji
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Tsuyoshi Takagi
- Institute for Developmental Research, Aichi Human Service Center, Kasugai, Japan,RIKEN Tsukuba Institute, Tsukuba, Japan
| | - Freesia L Huang
- Program of Developmental Neurobiology, National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, USA
| | - Akito Nakao
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Kuo-Ping Huang
- Program of Developmental Neurobiology, National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, USA
| | | | - Isabella A Graef
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Cynthia Shannon Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, Australia,School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan,Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Kutsukake-cho, Toyoake, Aichi 470-1192, Japan, Tel: +81 562 93 9376, Fax: +81 562 92 5382, E-mail:
| |
Collapse
|
184
|
Fond G, Berna F, Boyer L, Godin O, Brunel L, Andrianarisoa M, Aouizerate B, Capdevielle D, Chereau I, Danion JM, Dubertret C, Dubreucq J, Faget C, Gabayet F, Le Gloahec T, Llorca PM, Mallet J, Misdrahi D, Rey R, Richieri R, Passerieux C, Portalier C, Roux P, Vehier A, Yazbek H, Schürhoff F, Bulzacka E. Benzodiazepine long-term administration is associated with impaired attention/working memory in schizophrenia: results from the national multicentre FACE-SZ data set. Eur Arch Psychiatry Clin Neurosci 2018; 268:17-26. [PMID: 28349247 DOI: 10.1007/s00406-017-0787-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 03/13/2017] [Indexed: 10/19/2022]
Abstract
OBJECTIVE The effect of benzodiazepine long-term administration (BLTA) in cognitive functioning of subjects with schizophrenia (SZ) has been partially explored to date. The objective was to assess BLTA-associated cognitive impairment with a comprehensive cognitive battery in a non-selected multicentric/national community-dwelling sample of stabilized SZ subjects. METHOD 407 community-dwelling stabilized SZ subjects were consecutively included in the FondaMental Academic Centers of Expertise for Schizophrenia Cohort (FACE-SZ). Patients taking daily benzodiazepine were defined as BLTA+ as all patients examined by the Expert Center were clinically stabilized and under stable dose of treatment for at least 3 months. Each patient has been administered a 1-day long comprehensive cognitive battery (including The National Adult Reading Test, the Wechsler Adult Intelligence Scale, the Trail Making Test, the California Verbal Learning Test, the Doors test, and The Continuous Performance Test-Identical Pairs). RESULTS In the multivariate analyses, results showed that BLTA was associated with impaired attention/working memory (OR 0.60, 95% confidence interval 0.42-0.86; p = 0.005) independently of socio-demographic variables and illness characteristics. Verbal and performance current IQ-[respectively, OR 0.98, 95% CI (0.96;0.99), p = 0.016 and 0.98, 95% CI(0.97;0.99), p = 0.034] but not premorbid IQ-(p > 0.05) have been associated with BLTA in a multivariate model including the same confounding variables. CONCLUSION BLTA is associated with impaired attention/working memory in schizophrenia. The BLTA benefit/risk ratio should be regularly reevaluated. Alternative pharmacological and non-pharmacological strategies for comorbid anxiety disorders and sleep disorders should be preferred when possible. It seems reasonable to withdraw BLTA before the start of cognitive remediation therapy, as soon as possible, to improve the effectiveness of this therapy. Limits: the delay between the last benzodiazepine intake and testing, as well as the specific class of benzodiazepines (long half-life vs. short half-life), and the number of benzodiazepine daily intakes have not been recorded in the present study. The precise motive for BLTA prescription and sleep disturbances have not been reported, which is a limit for the interpretation of the present results.
Collapse
Affiliation(s)
- Guillaume Fond
- Fondation FondaMental, Créteil, France. .,INSERM U955, Translational Psychiatry Team, DHU Pe-PSY, Centre Expert Schizophrénie, Pôle de Psychiatrie et d'Addictologie des Hôpitaux Universitaires Henri Mondor,, Paris Est University, 40 rue de Mesly, 94000, Créteil, France. .,Clinique Jeanne d'arc-Hôpital Privé Parisien, 55 rue du commandant Mouchotte, 94160, Saint-Mandé, France. .,CHU Carémeau, 30000, Nîmes, France.
| | - F Berna
- Fondation FondaMental, Créteil, France.,Hôpitaux Universitaires de Strasbourg, INSERM U1114, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - L Boyer
- Fondation FondaMental, Créteil, France.,Pôle Psychiatrie Universitaire, CHU Sainte-Marguerite, 13274, Marseille cedex 09, France
| | - O Godin
- Fondation FondaMental, Créteil, France.,Sorbonne Universités, UPMC Univ Paris 06, INSERM, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Institut Pierre Louis d'Epidémiologie et de Santé Publique, 75013, Paris, France
| | - L Brunel
- Fondation FondaMental, Créteil, France.,INSERM U955, Translational Psychiatry Team, DHU Pe-PSY, Centre Expert Schizophrénie, Pôle de Psychiatrie et d'Addictologie des Hôpitaux Universitaires Henri Mondor,, Paris Est University, 40 rue de Mesly, 94000, Créteil, France
| | - M Andrianarisoa
- Fondation FondaMental, Créteil, France.,INSERM U955, Translational Psychiatry Team, DHU Pe-PSY, Centre Expert Schizophrénie, Pôle de Psychiatrie et d'Addictologie des Hôpitaux Universitaires Henri Mondor,, Paris Est University, 40 rue de Mesly, 94000, Créteil, France
| | - B Aouizerate
- Fondation FondaMental, Créteil, France.,Centre Hospitalier Charles Perrens, Université de Bordeaux, 33076, Bordeaux, France.,Inserm, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U862, 33000, Bordeaux, France
| | - D Capdevielle
- Fondation FondaMental, Créteil, France.,Service Universitaire de Psychiatrie Adulte, Hôpital la Colombière, CHRU Montpellier, Université Montpellier 1, Inserm 1061, Montpellier, France
| | - I Chereau
- Fondation FondaMental, Créteil, France.,CMP B, CHU, EA 7280 Faculté de Médecine, Université d'Auvergne, BP 69 63003, Clermont-Ferrand Cedex 1, France
| | - J M Danion
- Fondation FondaMental, Créteil, France.,Hôpitaux Universitaires de Strasbourg, INSERM U1114, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - C Dubertret
- Fondation FondaMental, Créteil, France.,AP-HP, Department of Psychiatry, Inserm U894, Sorbonne Paris Cité, Faculté de médecine, Louis Mourier Hospital, Université Paris Diderot, Colombes, France
| | - J Dubreucq
- Fondation FondaMental, Créteil, France.,Centre Référent de Réhabilitation Psychosociale, CH Alpes Isère, Grenoble, France
| | - C Faget
- Fondation FondaMental, Créteil, France.,Assistance Publique des Hôpitaux de Marseille (AP-HM), pôle universitaire de psychiatrie, Marseille, France
| | - F Gabayet
- Fondation FondaMental, Créteil, France.,Centre Référent de Réhabilitation Psychosociale, CH Alpes Isère, Grenoble, France
| | - T Le Gloahec
- Fondation FondaMental, Créteil, France.,INSERM U955, Translational Psychiatry Team, DHU Pe-PSY, Centre Expert Schizophrénie, Pôle de Psychiatrie et d'Addictologie des Hôpitaux Universitaires Henri Mondor,, Paris Est University, 40 rue de Mesly, 94000, Créteil, France
| | - P M Llorca
- Fondation FondaMental, Créteil, France.,CMP B, CHU, EA 7280 Faculté de Médecine, Université d'Auvergne, BP 69 63003, Clermont-Ferrand Cedex 1, France
| | - J Mallet
- Fondation FondaMental, Créteil, France.,AP-HP, Department of Psychiatry, Inserm U894, Sorbonne Paris Cité, Faculté de médecine, Louis Mourier Hospital, Université Paris Diderot, Colombes, France
| | - D Misdrahi
- Fondation FondaMental, Créteil, France.,Centre Hospitalier Charles Perrens, Université de Bordeaux, 33076, Bordeaux, France.,CNRS UMR 5287-INCIA, Bordeaux, France
| | - R Rey
- Fondation FondaMental, Créteil, France.,Université Claude Bernard Lyon 1/Centre Hospitalier Le Vinatier Pole Est, 95 bd Pinel, BP 300 39, 69678, BRON Cedex, France
| | - R Richieri
- Fondation FondaMental, Créteil, France.,Assistance Publique des Hôpitaux de Marseille (AP-HM), pôle universitaire de psychiatrie, Marseille, France
| | - C Passerieux
- Fondation FondaMental, Créteil, France.,Service de Psychiatrie d'Adulte, Centre Hospitalier de Versailles, UFR des Sciences de la Santé Simone Veil, Université Versailles Saint-Quentin en Yvelines, Versailles, France
| | - C Portalier
- Fondation FondaMental, Créteil, France.,AP-HP, Department of Psychiatry, Inserm U894, Sorbonne Paris Cité, Faculté de médecine, Louis Mourier Hospital, Université Paris Diderot, Colombes, France
| | - P Roux
- Fondation FondaMental, Créteil, France.,Service de Psychiatrie d'Adulte, Centre Hospitalier de Versailles, UFR des Sciences de la Santé Simone Veil, Université Versailles Saint-Quentin en Yvelines, Versailles, France
| | - A Vehier
- Fondation FondaMental, Créteil, France.,Université Claude Bernard Lyon 1/Centre Hospitalier Le Vinatier Pole Est, 95 bd Pinel, BP 300 39, 69678, BRON Cedex, France
| | - H Yazbek
- Fondation FondaMental, Créteil, France.,Service Universitaire de Psychiatrie Adulte, Hôpital la Colombière, CHRU Montpellier, Université Montpellier 1, Inserm 1061, Montpellier, France
| | - F Schürhoff
- Fondation FondaMental, Créteil, France.,INSERM U955, Translational Psychiatry Team, DHU Pe-PSY, Centre Expert Schizophrénie, Pôle de Psychiatrie et d'Addictologie des Hôpitaux Universitaires Henri Mondor,, Paris Est University, 40 rue de Mesly, 94000, Créteil, France
| | - E Bulzacka
- Fondation FondaMental, Créteil, France.,INSERM U955, Translational Psychiatry Team, DHU Pe-PSY, Centre Expert Schizophrénie, Pôle de Psychiatrie et d'Addictologie des Hôpitaux Universitaires Henri Mondor,, Paris Est University, 40 rue de Mesly, 94000, Créteil, France
| | | |
Collapse
|
185
|
Zhang J, Jing Y, Zhang H, Bilkey DK, Liu P. Effects of maternal immune activation on brain arginine metabolism of postnatal day 2 rat offspring. Schizophr Res 2018; 192:431-441. [PMID: 28526281 DOI: 10.1016/j.schres.2017.05.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 05/10/2017] [Accepted: 05/13/2017] [Indexed: 12/19/2022]
Abstract
l-Arginine is a versatile semi-essential amino acid with a number of bioactive metabolites, and altered arginine metabolism has been implicated in the pathogenesis of schizophrenia. Earlier research has demonstrated that maternal immune activation (MIA; a risk factor for schizophrenia) alters arginine metabolism in the prefrontal cortex and hippocampus of the adult offspring. The present study investigated how MIA affected the levels of l-arginine and its downstream metabolites in the whole forebrain, frontal cortex, hippocampus and cerebellum of male and female rat offspring at the age of postnatal day 2. While no effects were evident in the forebrain, MIA significantly increased l-arginine, glutamate, putrescine, spermidine and spermine levels and the glutamate/GABA ratio, but decreased the glutamine/glutamate ratio, in the frontal cortex, hippocampus and/or cerebellum with no marked sex differences. Cluster analyses revealed that l-arginine and its main metabolites formed distinct groups, which changed as a function of MIA or sex in all four brain regions examined. These results demonstrate, for the first time, that MIA alters brain arginine metabolism in the rat offspring during early neonatal development, and further support the involvement of arginine metabolism in the pathogenesis of schizophrenia.
Collapse
Affiliation(s)
- Jiaxian Zhang
- Department of Anatomy, University of Otago, Dunedin, New Zealand; Brain Health and Research Centre, University of Otago, Dunedin, New Zealand
| | - Yu Jing
- Department of Anatomy, University of Otago, Dunedin, New Zealand; Brain Health and Research Centre, University of Otago, Dunedin, New Zealand
| | - Hu Zhang
- School of Pharmacy, University of Otago, Dunedin, New Zealand; Brain Health and Research Centre, University of Otago, Dunedin, New Zealand
| | - David K Bilkey
- Department of Psychology, University of Otago, Dunedin, New Zealand; Brain Health and Research Centre, University of Otago, Dunedin, New Zealand
| | - Ping Liu
- Department of Anatomy, University of Otago, Dunedin, New Zealand; Brain Health and Research Centre, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
186
|
Ren SQ, Yao W, Yan JZ, Jin C, Yin JJ, Yuan J, Yu S, Cheng Z. Amyloid β causes excitation/inhibition imbalance through dopamine receptor 1-dependent disruption of fast-spiking GABAergic input in anterior cingulate cortex. Sci Rep 2018; 8:302. [PMID: 29321592 PMCID: PMC5762926 DOI: 10.1038/s41598-017-18729-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 12/17/2017] [Indexed: 11/21/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of dementia in the elderly. At the early stages of AD development, the soluble β-amyloid (Aβ) induces synaptic dysfunction, perturbs the excitation/inhibition balance of neural circuitries, and in turn alters the normal neural network activity leading to cognitive decline, but the underlying mechanisms are not well established. Here by using whole-cell recordings in acute mouse brain slices, we found that 50 nM Aβ induces hyperexcitability of excitatory pyramidal cells in the cingulate cortex, one of the most vulnerable areas in AD, via depressing inhibitory synaptic transmission. Furthermore, by simultaneously recording multiple cells, we discovered that the inhibitory innervation of pyramidal cells from fast-spiking (FS) interneurons instead of non-FS interneurons is dramatically disrupted by Aβ, and perturbation of the presynaptic inhibitory neurotransmitter gamma-aminobutyric acid (GABA) release underlies this inhibitory input disruption. Finally, we identified the increased dopamine action on dopamine D1 receptor of FS interneurons as a key pathological factor that contributes to GABAergic input perturbation and excitation/inhibition imbalance caused by Aβ. Thus, we conclude that the dopamine receptor 1-dependent disruption of FS GABAergic inhibitory input plays a critical role in Aβ-induced excitation/inhibition imbalance in anterior cingulate cortex.
Collapse
Affiliation(s)
- Si-Qiang Ren
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China.
| | - Wen Yao
- Department of Pharmacology, Wuxi Higher Health Vocational Technology School, Wuxi, China
| | - Jing-Zhi Yan
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical College, Xuzhou, China
| | - Chunhui Jin
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Jia-Jun Yin
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Jianmin Yuan
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Shui Yu
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China
| | - Zaohuo Cheng
- Wuxi Mental Health Center, Nanjing Medical University, Wuxi, China.
| |
Collapse
|
187
|
Lippert T, Gelineau L, Napoli E, Borlongan CV. Harnessing neural stem cells for treating psychiatric symptoms associated with fetal alcohol spectrum disorder and epilepsy. Prog Neuropsychopharmacol Biol Psychiatry 2018; 80:10-22. [PMID: 28365374 DOI: 10.1016/j.pnpbp.2017.03.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 03/09/2017] [Indexed: 12/20/2022]
Abstract
Brain insults with progressive neurodegeneration are inherent in pathological symptoms that represent many psychiatric illnesses. Neural network disruptions characterized by impaired neurogenesis have been recognized to precede, accompany, and possibly even exacerbate the evolution and progression of symptoms of psychiatric disorders. Here, we focus on the neurodegeneration and the resulting psychiatric symptoms observed in fetal alcohol spectrum disorder and epilepsy, in an effort to show that these two diseases are candidate targets for stem cell therapy. In particular, we provide preclinical evidence in the transplantation of neural stem cells (NSCs) in both conditions, highlighting the potential of this cell-based treatment for correcting the psychiatric symptoms that plague these two disorders. Additionally, we discuss the challenges of NSC transplantation and offer insights into the mechanisms that may mediate the therapeutic benefits and can be exploited to overcome the hurdles of translating this therapy from the laboratory to the clinic. Our ultimate goal is to advance stem cell therapy for the treatment of psychiatric disorders.
Collapse
Affiliation(s)
- Trenton Lippert
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd. MDC 78, Tampa, FL 33612, USA
| | - Lindsey Gelineau
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd. MDC 78, Tampa, FL 33612, USA
| | - Eleonora Napoli
- Department of Molecular Biosciences, 3011 VM3B 1089 Veterinary Medicine Drive, University of California Davis, Davis, CA 95616, USA..
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd. MDC 78, Tampa, FL 33612, USA.
| |
Collapse
|
188
|
Rosas-Arellano A, Estrada-Mondragón A, Mantellero CA, Tejeda-Guzmán C, Castro MA. The adjustment of γ-aminobutyric acid A tonic subunits in Huntington's disease: from transcription to translation to synaptic levels into the neostriatum. Neural Regen Res 2018; 13:584-590. [PMID: 29722299 PMCID: PMC5950657 DOI: 10.4103/1673-5374.230270] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
γ-Aminobutyric acid (GABA), plays a key role in all stages of life, also is considered the main inhibitory neurotransmitter. GABA activates two kind of membrane receptors known as GABAA and GABAB, the first one is responsible to render tonic inhibition by pentameric receptors containing α4−6, β3, δ, or ρ1−3 subunits, they are located at perisynaptic and/or in extrasynaptic regions. The biophysical properties of GABAA tonic inhibition have been related with cellular protection against excitotoxic injury and cell death in presence of excessive excitation. On this basis, GABAA tonic inhibition has been proposed as a potential target for therapeutic intervention of Huntington's disease. Huntington's disease is a neurodegenerative disorder caused by a genetic mutation of the huntingtin protein. For experimental studies of Huntington's disease mouse models have been developed, such as R6/1, R6/2, HdhQ92, HdhQ150, as well as YAC128. In all of them, some key experimental reports are focused on neostriatum. The neostriatum is considered as the most important connection between cerebral cortex and basal ganglia structures, its cytology display two pathways called direct and indirect constituted by medium sized spiny neurons expressing dopamine D1 and D2 receptors respectively, they display strong expression of many types of GABAA receptors, including tonic subunits. The studies about of GABAA tonic subunits and Huntington's disease into the neostriatum are rising in recent years, suggesting interesting changes in their expression and localization which can be used as a strategy to delay the cellular damage caused by the imbalance between excitation and inhibition, a hallmark of Huntington's disease.
Collapse
Affiliation(s)
- Abraham Rosas-Arellano
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile; Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile; Departamento de Fisiología, Biofísica y Neurociencias, Cinvestav del IPN, Ciudad de México, México
| | | | - Carola A Mantellero
- Laboratorio de Neurociencias, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago de Chile, Chile
| | - Carlos Tejeda-Guzmán
- Departamento de Fisiología, Biofísica y Neurociencias, Cinvestav del IPN, Ciudad de México, México
| | - Maite A Castro
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias; Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
189
|
Renard J, Rushlow WJ, Laviolette SR. Effects of Adolescent THC Exposure on the Prefrontal GABAergic System: Implications for Schizophrenia-Related Psychopathology. Front Psychiatry 2018; 9:281. [PMID: 30013490 PMCID: PMC6036125 DOI: 10.3389/fpsyt.2018.00281] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/11/2018] [Indexed: 12/14/2022] Open
Abstract
Marijuana is the most commonly used drug of abuse among adolescents. Considerable clinical evidence supports the hypothesis that adolescent neurodevelopmental exposure to high levels of the principal psychoactive component in marijuana, -delta-9-tetrahydrocanabinol (THC), is associated with a high risk of developing psychiatric diseases, such as schizophrenia later in life. This marijuana-associated risk is believed to be related to increasing levels of THC found within commonly used marijuana strains. Adolescence is a highly vulnerable period for the development of the brain, where the inhibitory GABAergic system plays a pivotal role in the maturation of regulatory control mechanisms in the central nervous system (CNS). Specifically, adolescent neurodevelopment represents a critical period wherein regulatory connectivity between higher-order cortical regions and sub-cortical emotional processing circuits such as the mesolimbic dopamine (DA) system is established. Emerging preclinical evidence demonstrates that adolescent exposure to THC selectively targets schizophrenia-related molecular and neuropharmacological signaling pathways in both cortical and sub-cortical regions, including the prefrontal cortex (PFC) and mesolimbic DA pathway, comprising the ventral tegmental area (VTA) and nucleus accumbens (NAc). Prefrontal cortical GABAergic hypofunction is a key feature of schizophrenia-like neuropsychopathology. This GABAergic hypofunction may lead to the loss of control of the PFC to regulate proper sub-cortical DA neurotransmission, thereby leading to schizophrenia-like symptoms. This review summarizes preclinical evidence demonstrating that reduced prefrontal cortical GABAergic neurotransmission has a critical role in the sub-cortical DAergic dysregulation and schizophrenia-like behaviors observed following adolescent THC exposure.
Collapse
Affiliation(s)
- Justine Renard
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada
| | - Walter J Rushlow
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada.,Department of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Steven R Laviolette
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON, Canada.,Department of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
190
|
Bagasrawala I, Memi F, V. Radonjić N, Zecevic N. N-Methyl d-Aspartate Receptor Expression Patterns in the Human Fetal Cerebral Cortex. Cereb Cortex 2017; 27:5041-5053. [PMID: 27664962 PMCID: PMC6077866 DOI: 10.1093/cercor/bhw289] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 08/01/2016] [Accepted: 08/23/2016] [Indexed: 01/09/2023] Open
Abstract
N-methyl d-aspartate receptors (NMDARs), a subtype of glutamate receptor, have important functional roles in cellular activity and neuronal development. They are well-studied in rodent and adult human brains, but limited information is available about their distribution in the human fetal cerebral cortex. Here we show that 3 NMDAR subunits, NR1, NR2A, and NR2B, are expressed in the human cerebral cortex during the second trimester of gestation, a period of intense neurogenesis and synaptogenesis. With increasing fetal age, expression of the NMDAR-encoding genes Grin1 (NR1) and Grin2a (NR2A) increased while Grin2b (NR2B) expression decreased. The protein levels of all 3 subunits paralleled the changes in gene expression. On cryosections, all 3 subunits were expressed in proliferative ventricular and subventricular zones, in radial glia, and in intermediate progenitor cells, consistent with their role in the proliferation of cortical progenitor cells and in the determination of their respective fates. The detection of NR1, NR2A, and NR2B in both glutamatergic and GABAergic neurons of the cortical plate suggests the involvement of NMDARs in the maturation of human cortical neurons and in early synapse formation. Our results and previous studies in rodents suggest that NMDAR expression in the developing human brain is evolutionarily conserved.
Collapse
Affiliation(s)
- Inseyah Bagasrawala
- Department of Neuroscience, University of Connecticut Health, Farmington, CT 06030, USA
| | - Fani Memi
- Department of Neuroscience, University of Connecticut Health, Farmington, CT 06030, USA
| | - Nevena V. Radonjić
- Psychiatry Department, University of Connecticut Health, Farmington, CT 06030, USA
| | - Nada Zecevic
- Department of Neuroscience, University of Connecticut Health, Farmington, CT 06030, USA
| |
Collapse
|
191
|
Berridge MJ. Vitamin D deficiency: infertility and neurodevelopmental diseases (attention deficit hyperactivity disorder, autism, and schizophrenia). Am J Physiol Cell Physiol 2017; 314:C135-C151. [PMID: 29070492 DOI: 10.1152/ajpcell.00188.2017] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The process of development depends on a number of signaling systems that regulates the progressive sequence of developmental events. Infertility and neurodevelopmental diseases, such as attention deficit hyperactivity disorder, autism spectrum disorders, and schizophrenia, are caused by specific alterations in these signaling processes. Calcium signaling plays a prominent role throughout development beginning at fertilization and continuing through early development, implantation, and organ differentiation such as heart and brain development. Vitamin D plays a major role in regulating these signaling processes that control development. There is an increase in infertility and an onset of neurodevelopmental diseases when vitamin D is deficient. The way in which vitamin D deficiency acts to alter development is a major feature of this review. One of the primary functions of vitamin D is to maintain the phenotypic stability of both the Ca2+ and redox signaling pathways that play such a key role throughout development.
Collapse
Affiliation(s)
- Michael J Berridge
- Laboratory of Molecular Signalling, The Babraham Institute , Cambridge , United Kingdom
| |
Collapse
|
192
|
Pelkey KA, Chittajallu R, Craig MT, Tricoire L, Wester JC, McBain CJ. Hippocampal GABAergic Inhibitory Interneurons. Physiol Rev 2017; 97:1619-1747. [PMID: 28954853 DOI: 10.1152/physrev.00007.2017] [Citation(s) in RCA: 495] [Impact Index Per Article: 70.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 05/16/2017] [Accepted: 05/26/2017] [Indexed: 12/11/2022] Open
Abstract
In the hippocampus GABAergic local circuit inhibitory interneurons represent only ~10-15% of the total neuronal population; however, their remarkable anatomical and physiological diversity allows them to regulate virtually all aspects of cellular and circuit function. Here we provide an overview of the current state of the field of interneuron research, focusing largely on the hippocampus. We discuss recent advances related to the various cell types, including their development and maturation, expression of subtype-specific voltage- and ligand-gated channels, and their roles in network oscillations. We also discuss recent technological advances and approaches that have permitted high-resolution, subtype-specific examination of their roles in numerous neural circuit disorders and the emerging therapeutic strategies to ameliorate such pathophysiological conditions. The ultimate goal of this review is not only to provide a touchstone for the current state of the field, but to help pave the way for future research by highlighting where gaps in our knowledge exist and how a complete appreciation of their roles will aid in future therapeutic strategies.
Collapse
Affiliation(s)
- Kenneth A Pelkey
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Ramesh Chittajallu
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Michael T Craig
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Ludovic Tricoire
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Jason C Wester
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Chris J McBain
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| |
Collapse
|
193
|
Ribolsi M, Lisi G, Ponzo V, Siracusano A, Caltagirone C, Niolu C, Koch G. Left hemispheric breakdown of LTP-like cortico-cortical plasticity in schizophrenic patients. Clin Neurophysiol 2017; 128:2037-2042. [DOI: 10.1016/j.clinph.2017.06.255] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Revised: 05/21/2017] [Accepted: 06/26/2017] [Indexed: 12/23/2022]
|
194
|
Grain R, Lally J, Stubbs B, Malik S, LeMince A, Nicholson TR, Murray RM, Gaughran F. Autoantibodies against voltage-gated potassium channel and glutamic acid decarboxylase in psychosis: A systematic review, meta-analysis, and case series. Psychiatry Clin Neurosci 2017; 71:678-689. [PMID: 28573688 DOI: 10.1111/pcn.12543] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 05/17/2017] [Accepted: 05/19/2017] [Indexed: 01/21/2023]
Abstract
Antibodies to the voltage-gated potassium channel (VGKC) complex and glutamic acid decarboxylase (GAD) have been reported in some cases of psychosis. We conducted the first systematic review and meta-analysis to investigate their prevalence in people with psychosis and report a case series of VGKC-complex antibodies in refractory psychosis. Only five studies presenting prevalence rates of VGKC seropositivity in psychosis were identified, in addition to our case series, with an overall prevalence of 1.5% (25/1720) compared to 0.7% in healthy controls (12/1753). Meta-analysis established that the pooled prevalence of GAD65 autoantibodies was 5.8% (95% confidence interval [CI]: 2.0-15.6%; I2 = 91%; nine studies) in psychotic disorders, with a prevalence of 4.6% (95%CI: 1.2-15.9%; nine studies; I2 = 89%) and 6.2% (95%CI: 1.2-27.0%; two studies; I2 = 69%) in schizophrenia and bipolar disorder, respectively. People with psychosis were more likely to have GAD65 antibodies than controls (odds ratio [OR], 2.24; 95%CI: 1.28-3.92%; P = 0.005; eight studies; I2 = 0%). Among 21 participants with treatment-resistant psychosis, none had VGKC antibodies. The prevalence of VGKC antibodies is low in psychosis. Our preliminary meta-analysis suggests that GAD autoantibodies are more common in people with psychosis than in controls, although few studies accounted for the possibility of co-existing type 1 diabetes mellitus and the clinical significance of reported GAD titers remains unclear. The paucity of studies reporting thresholds for defining GAD abnormality and rates of comorbid type 1 diabetes mellitus precludes interpretations regarding the influence of GAD antibodies on the development of psychotic disorders and may have led to an overestimate of the prevalence of GAD. Our case series fails to support the hypothesis that VGKC antibodies are linked to treatment resistance in psychosis, but the literature to date is remarkably sparse.
Collapse
Affiliation(s)
- Rosemary Grain
- GKT School of Medicine, King's College London, London, UK
| | - John Lally
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,National Psychosis Service, South London and Maudsley NHS Foundation Trust, London, UK.,Department of Psychiatry, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland.,Department of Psychiatry, University College Dublin, Dublin, Ireland
| | - Brendon Stubbs
- Physiotherapy Department, South London and Maudsley NHS Foundation Trust, London, UK.,Health Service and Population Research Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Steffi Malik
- Medical School, University of Bristol, Bristol, UK
| | - Anne LeMince
- National Psychosis Service, South London and Maudsley NHS Foundation Trust, London, UK
| | - Timothy R Nicholson
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Robin M Murray
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,National Psychosis Service, South London and Maudsley NHS Foundation Trust, London, UK
| | - Fiona Gaughran
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,National Psychosis Service, South London and Maudsley NHS Foundation Trust, London, UK.,The Collaboration for Leadership in Applied Health Research and Care (CLAHRC), South London Psychosis Research Team, London, UK
| |
Collapse
|
195
|
Laclef C, Métin C. Conserved rules in embryonic development of cortical interneurons. Semin Cell Dev Biol 2017; 76:86-100. [PMID: 28918121 DOI: 10.1016/j.semcdb.2017.09.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 09/08/2017] [Accepted: 09/11/2017] [Indexed: 12/24/2022]
Abstract
This review will focus on early aspects of cortical interneurons (cIN) development from specification to migration and final positioning in the human cerebral cortex. These mechanisms have been largely studied in the mouse model, which provides unique possibilities of genetic analysis, essential to dissect the molecular and cellular events involved in cortical development. An important goal here is to discuss the conservation and the potential divergence of these mechanisms, with a particular interest for the situation in the human embryo. We will thus cover recent works, but also revisit older studies in the light of recent data to better understand the developmental mechanisms underlying cIN differentiation in human. Because cIN are implicated in severe developmental disorders, understanding the molecular and cellular mechanisms controlling their differentiation might clarify some causes and potential therapeutic approaches to these important clinical conditions.
Collapse
Affiliation(s)
- Christine Laclef
- INSERM, UMR-S839, Paris, France; Sorbonne Universités, UPMC University Paris 6, UMR-S839, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Christine Métin
- INSERM, UMR-S839, Paris, France; Sorbonne Universités, UPMC University Paris 6, UMR-S839, Paris, France; Institut du Fer à Moulin, Paris, France.
| |
Collapse
|
196
|
Renard J, Szkudlarek HJ, Kramar CP, Jobson CEL, Moura K, Rushlow WJ, Laviolette SR. Adolescent THC Exposure Causes Enduring Prefrontal Cortical Disruption of GABAergic Inhibition and Dysregulation of Sub-Cortical Dopamine Function. Sci Rep 2017; 7:11420. [PMID: 28900286 PMCID: PMC5595795 DOI: 10.1038/s41598-017-11645-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 08/29/2017] [Indexed: 02/07/2023] Open
Abstract
Chronic adolescent marijuana use has been linked to the later development of psychiatric diseases such as schizophrenia. GABAergic hypofunction in the prefrontal cortex (PFC) is a cardinal pathological feature of schizophrenia and may be a mechanism by which the PFC loses its ability to regulate sub-cortical dopamine (DA) resulting in schizophrenia-like neuropsychopathology. In the present study, we exposed adolescent rats to Δ-9-tetra-hydrocannabinol (THC), the psychoactive component in marijuana. At adulthood, we characterized the functionality of PFC GABAergic neurotransmission and its regulation of sub-cortical DA function using molecular, behavioral and in-vivo electrophysiological analyses. Our findings revealed a persistent attenuation of PFC GABAergic function combined with a hyperactive neuronal state in PFC neurons and associated disruptions in cortical gamma oscillatory activity. These PFC abnormalities were accompanied by hyperactive DAergic neuronal activity in the ventral tegmental area (VTA) and behavioral and cognitive abnormalities similar to those observed in psychiatric disorders. Remarkably, these neuronal and behavioral effects were reversed by pharmacological activation of GABAA receptors in the PFC. Together, these results identify a mechanistic link between dysregulated frontal cortical GABAergic inhibition and sub-cortical DAergic dysregulation, characteristic of well-established neuropsychiatric endophenotypes.
Collapse
Affiliation(s)
- Justine Renard
- Dept. of Anatomy and Cell Biology & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Hanna J Szkudlarek
- Dept. of Anatomy and Cell Biology & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Cecilia P Kramar
- Dept. of Anatomy and Cell Biology & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Christina E L Jobson
- Dept. of Anatomy and Cell Biology & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Kyra Moura
- Dept. of Anatomy and Cell Biology & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Walter J Rushlow
- Dept. of Anatomy and Cell Biology & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada.,Dept. of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada
| | - Steven R Laviolette
- Dept. of Anatomy and Cell Biology & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada. .,Dept. of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, N6A 5C1, Canada.
| |
Collapse
|
197
|
Ben-Shachar D. Mitochondrial multifaceted dysfunction in schizophrenia; complex I as a possible pathological target. Schizophr Res 2017; 187:3-10. [PMID: 27802911 DOI: 10.1016/j.schres.2016.10.022] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/10/2016] [Accepted: 10/14/2016] [Indexed: 01/09/2023]
Abstract
Mitochondria are key players in various essential cellular processes beyond being the main energy supplier of the cell. Accordingly, they are involved in neuronal synaptic transmission, neuronal growth and sprouting and consequently neuronal plasticity and connectivity. In addition, mitochondria participate in the modulation of gene transcription and inflammation as well in physiological responses in health and disease. Schizophrenia is currently regarded as a neurodevelopmental disorder associated with impaired immune system, aberrant neuronal differentiation and abnormalities in various neurotransmitter systems mainly the dopaminergic, glutaminergic and GABAergic. Ample evidence has been accumulated over the last decade indicating a multifaceted dysfunction of mitochondria in schizophrenia. Indeed, mitochondrial deficit can be of relevance for the majority of the pathologies observed in this disease. In the present article, we overview specific deficits of the mitochondria in schizophrenia, with a focus on the first complex (complex I) of the mitochondrial electron transport chain (ETC). We argue that complex I, being a major factor in the regulation of mitochondrial ETC, is a possible key modulator of various functions of the mitochondria. We review biochemical, molecular, cellular and functional evidence for mitochondrial impairments and their possible convergence to impact in-vitro neuronal differentiation efficiency in schizophrenia. Mitochondrial function in schizophrenia may advance our knowledge of the disease pathophysiology and open the road for new treatment targets for the benefit of the patients.
Collapse
Affiliation(s)
- Dorit Ben-Shachar
- Laboratory of Psychobiology, Department of Psychiatry, Rambam Health Care Campus, B. Rappaport Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Technion-IIT, Haifa, Israel.
| |
Collapse
|
198
|
Jagannath V, Gerstenberg M, Correll CU, Walitza S, Grünblatt E. A systematic meta-analysis of the association of Neuregulin 1 (NRG1), D-amino acid oxidase (DAO), and DAO activator (DAOA)/G72 polymorphisms with schizophrenia. J Neural Transm (Vienna) 2017; 125:89-102. [PMID: 28864885 DOI: 10.1007/s00702-017-1782-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 08/23/2017] [Indexed: 02/07/2023]
Abstract
The glutamate hypothesis of schizophrenia is related to the proposed dysregulation of D-amino acid oxidase (DAO), DAO activator (DAOA)/G72, and Neuregulin 1 (NRG1) genes. Genetic studies have shown significant associations between DAO, DAOA, NRG1 single-nucleotide polymorphisms (SNPs), and schizophrenia. The systematic literature search yielded 6, 5, and 18 new studies for DAO, DAOA, and NRG1 published after 2011 and not included in the previous SchizophreniaGene (SZGene) meta-analysis. We conducted meta-analyses of 20, 23, and 48 case-control studies, respectively, to comprehensively evaluate the association of 8 DAO, 12 DAOA, and 14 NRG1 SNPs with schizophrenia. The updated meta-analyses resulted in the following findings: the C-allele of DAO rs4623951 was associated with schizophrenia across all pooled studies [Odds ratio (OR) = 0.88, 95% confidence interval (CI) = 0.79-0.98, p = 0.02, N = 3143]; however, no new reports could be included. The G-allele of DAOA rs778293 was associated with schizophrenia in Asian patients (OR = 1.17, 95% CI = 1.08-1.27, p = 0.00008, N = 6117), and the T-allele of DAOA rs3916971 was associated with schizophrenia across all studies (OR = 0.84, 95% CI = 0.73-0.96, p = 0.01, N = 1765). Again, for both SNPs, no new eligible studies were available. After adding new reports, the T-allele of NRG1 SNP8NRG241930 (rs62510682) across all studies (OR = 0.95, 95% CI = 0.91-0.997, p = 0.04, N = 22,898) and in Caucasian samples (OR = 0.95, 95% CI = 0.90-0.99, p = 0.03, N = 16,014), and the C-allele of NRG1 rs10503929 across all studies (OR = 0.89, 95% CI = 0.81-0.97, p = 0.01, N = 6844) and in Caucasian samples (OR = 0.89, 95% CI = 0.81-0.98, p = 0.01, N = 6414) were protective against schizophrenia. Our systematic meta-analysis is the most updated one for the association of DAO, DAOA, and NRG1 SNPs with schizophrenia.
Collapse
Affiliation(s)
- Vinita Jagannath
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, Centre for Child and Adolescent Psychiatry Research, University of Zurich, 5th Floor, Room K118, Wagistrasse 12, 8952, Schlieren, Switzerland
| | - Miriam Gerstenberg
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, Centre for Child and Adolescent Psychiatry Research, University of Zurich, 5th Floor, Room K118, Wagistrasse 12, 8952, Schlieren, Switzerland
| | - Christoph U Correll
- The Zucker Hillside Hospital, Psychiatry Research, Northwell Health, Glen Oaks, NY, USA.,Hofstra Northwell School of Medicine, Hempstead, NY, USA.,The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Susanne Walitza
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, Centre for Child and Adolescent Psychiatry Research, University of Zurich, 5th Floor, Room K118, Wagistrasse 12, 8952, Schlieren, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Edna Grünblatt
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, Centre for Child and Adolescent Psychiatry Research, University of Zurich, 5th Floor, Room K118, Wagistrasse 12, 8952, Schlieren, Switzerland. .,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland. .,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
199
|
Comprehensive review: Computational modelling of schizophrenia. Neurosci Biobehav Rev 2017; 83:631-646. [PMID: 28867653 DOI: 10.1016/j.neubiorev.2017.08.022] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 07/08/2017] [Accepted: 08/30/2017] [Indexed: 12/21/2022]
Abstract
Computational modelling has been used to address: (1) the variety of symptoms observed in schizophrenia using abstract models of behavior (e.g. Bayesian models - top-down descriptive models of psychopathology); (2) the causes of these symptoms using biologically realistic models involving abnormal neuromodulation and/or receptor imbalance (e.g. connectionist and neural networks - bottom-up realistic models of neural processes). These different levels of analysis have been used to answer different questions (i.e. understanding behavioral vs. neurobiological anomalies) about the nature of the disorder. As such, these computational studies have mostly supported diverging hypotheses of schizophrenia's pathophysiology, resulting in a literature that is not always expanding coherently. Some of these hypotheses are however ripe for revision using novel empirical evidence. Here we present a review that first synthesizes the literature of computational modelling for schizophrenia and psychotic symptoms into categories supporting the dopamine, glutamate, GABA, dysconnection and Bayesian inference hypotheses respectively. Secondly, we compare model predictions against the accumulated empirical evidence and finally we identify specific hypotheses that have been left relatively under-investigated.
Collapse
|
200
|
Kaneta H, Ukai W, Tsujino H, Furuse K, Kigawa Y, Tayama M, Ishii T, Hashimoto E, Kawanishi C. Antipsychotics promote GABAergic interneuron genesis in the adult rat brain: Role of heat-shock protein production. J Psychiatr Res 2017; 92:108-118. [PMID: 28414930 DOI: 10.1016/j.jpsychires.2017.03.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 03/07/2017] [Accepted: 03/08/2017] [Indexed: 02/07/2023]
Abstract
Current antipsychotics reduce positive symptoms and reverse negative symptoms in conjunction with cognitive behavioral issues with the goal of restoring impaired occupational and social functioning. However, limited information is available on their influence on gliogenesis or their neurogenic properties in adult schizophrenia brains, particularly on GABAergic interneuron production. In the present study, we used young adult subventricular zone (SVZ)-derived progenitor cells expressing proteoglycan NG2 cultures to examine the oligodendrocyte and GABAergic interneuron genesis effects of several kinds of antipsychotics on changes in differentiation function induced by exposure to the NMDA receptor antagonist MK-801. We herein demonstrated that antipsychotics promoted or restored changes in the oligodendrocyte/GABAergic interneuron differentiation functions of NG2(+) cells induced by the exposure to MK-801, which was considered to be one of the drug-induced schizophrenia model. We also demonstrated that antipsychotics restored heat-shock protein (HSP) production in NG2(+) cells with differentiation impairment. The antipsychotics olanzapine, aripiprazole, and blonanserin, but not haloperidol increased HSP90 levels, which were reduced by the exposure to MK-801. Our results showed that antipsychotics, particularly those recently synthesized, exerted similar GABAergic interneuron genesis effects on NG2(+) neuronal/glial progenitor cells in the adult rat brain by increasing cellular HSP production, and also suggest that HSP90 may play a crucial role in the pathophysiology of schizophrenia and is a key target for next drug development.
Collapse
Affiliation(s)
- Hiroo Kaneta
- Department of Neuropsychiatry, Sapporo Medical University, School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| | - Wataru Ukai
- Department of Neuropsychiatry, Sapporo Medical University, School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| | - Hanako Tsujino
- Department of Neuropsychiatry, Sapporo Medical University, School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| | - Kengo Furuse
- Department of Neuropsychiatry, Sapporo Medical University, School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| | - Yoshiyasu Kigawa
- Department of Neuropsychiatry, Sapporo Medical University, School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| | - Masaya Tayama
- Department of Neuropsychiatry, Sapporo Medical University, School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| | - Takao Ishii
- Department of Neuropsychiatry, Sapporo Medical University, School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| | - Eri Hashimoto
- Department of Neuropsychiatry, Sapporo Medical University, School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| | - Chiaki Kawanishi
- Department of Neuropsychiatry, Sapporo Medical University, School of Medicine, S-1, W-16, Chuo-ku, Sapporo, 0608543, Japan.
| |
Collapse
|