1
|
Cong L, Zhang T, Zhang T, Liu Y, Li Y, Pang X, Zhao L, Wu T, Ding S, Liu Y, Wu H, Shen H, Li Y. Gene Deficiency of δ Subunit-Containing GABA A Receptor in mPFC Lead Learning and Memory Impairment in Mice. Neurochem Res 2025; 50:71. [PMID: 39751665 DOI: 10.1007/s11064-024-04320-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025]
Abstract
Maintaining GABAergic inhibition within physiological limits in the medial prefrontal cortex (mPFC) is critical for working memory. While synaptic GABAAR typically mediate the primary component of mPFC inhibition, the role of extrasynaptic δ-GABAAR in working memory remains unclear. To investigate this, we used fiber photometry to examine the effects of δ-GABAAR in freely moving mice. Our results indicate that the loss of δ-GABAAR expression leads to learning and memory impairment. Specifically, activation of δ-GABAAR impaired learning and memory in WT mice but enhanced learning and memory in δ+/- knockout mice. Furthermore, δ-GABAAR activation increased calcium activity in the mPFC pyramidal neurons, an effect not observed in δ-Cas9-sgRNA virus-infected mice. Collectively, these findings suggest that δ-GABAAR deficiency impairs learning and memory by modulating the excitability of pyramidal neurons in the mPFC. These results delineate the functional contribution of δ-GABAAR to learning and memory, suggesting their role extends beyond the mere maintenance of information.
Collapse
Affiliation(s)
- Lin Cong
- Laboratory of Chinese Medicine Brain Science, Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Tianshu Zhang
- Laboratory of Chinese Medicine Brain Science, Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Institute of Brain Science and Brain-inspired Research, Shandong Academy of Medical Sciences, Shandong First Medical University, Jinan, 250117, China
| | - Teng Zhang
- Laboratory of Chinese Medicine Brain Science, Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yifan Liu
- Laboratory of Chinese Medicine Brain Science, Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yunxiao Li
- Laboratory of Chinese Medicine Brain Science, Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xiaogang Pang
- Laboratory of Chinese Medicine Brain Science, Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Lianbin Zhao
- Laboratory of Chinese Medicine Brain Science, Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Tongrui Wu
- Department of Cellular Biology, School of Basic Medicine, Tianjin Medical University, Tianjin, 300070, China
| | - Shengkai Ding
- Laboratory of Chinese Medicine Brain Science, Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yanling Liu
- Laboratory of Chinese Medicine Brain Science, Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Hao Wu
- Laboratory of Chinese Medicine Brain Science, Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Hui Shen
- Laboratory of Chinese Medicine Brain Science, Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Department of Cellular Biology, School of Basic Medicine, Tianjin Medical University, Tianjin, 300070, China.
| | - Yuanyuan Li
- Laboratory of Chinese Medicine Brain Science, Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
2
|
Wei X, Campagna JJ, Jagodzinska B, Wi D, Cohn W, Lee JT, Zhu C, Huang CS, Molnár L, Houser CR, John V, Mody I. A therapeutic small molecule enhances γ-oscillations and improves cognition/memory in Alzheimer's disease model mice. Proc Natl Acad Sci U S A 2024; 121:e2400420121. [PMID: 39106304 PMCID: PMC11331084 DOI: 10.1073/pnas.2400420121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 07/08/2024] [Indexed: 08/09/2024] Open
Abstract
Brain rhythms provide the timing for recruitment of brain activity required for linking together neuronal ensembles engaged in specific tasks. The γ-oscillations (30 to 120 Hz) orchestrate neuronal circuits underlying cognitive processes and working memory. These oscillations are reduced in numerous neurological and psychiatric disorders, including early cognitive decline in Alzheimer's disease (AD). Here, we report on a potent brain-permeable small molecule, DDL-920 that increases γ-oscillations and improves cognition/memory in a mouse model of AD, thus showing promise as a class of therapeutics for AD. We employed anatomical, in vitro and in vivo electrophysiological, and behavioral methods to examine the effects of our lead therapeutic candidate small molecule. As a novel in central nervous system pharmacotherapy, our lead molecule acts as a potent, efficacious, and selective negative allosteric modulator of the γ-aminobutyric acid type A receptors most likely assembled from α1β2δ subunits. These receptors, identified through anatomical and pharmacological means, underlie the tonic inhibition of parvalbumin (PV) expressing interneurons (PV+INs) critically involved in the generation of γ-oscillations. When orally administered twice daily for 2 wk, DDL-920 restored the cognitive/memory impairments of 3- to 4-mo-old AD model mice as measured by their performance in the Barnes maze. Our approach is unique as it is meant to enhance cognitive performance and working memory in a state-dependent manner by engaging and amplifying the brain's endogenous γ-oscillations through enhancing the function of PV+INs.
Collapse
Affiliation(s)
- Xiaofei Wei
- Department of Neurology, The David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA90095
- Department of Neurosurgery, The David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA90095
| | - Jesus J. Campagna
- Department of Neurology, Drug Development Laboratory, Mary S. Easton Center for Alzheimer’s Disease Research and Care, The David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA90095
| | - Barbara Jagodzinska
- Department of Neurology, Drug Development Laboratory, Mary S. Easton Center for Alzheimer’s Disease Research and Care, The David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA90095
| | - Dongwook Wi
- Department of Neurology, Drug Development Laboratory, Mary S. Easton Center for Alzheimer’s Disease Research and Care, The David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA90095
| | - Whitaker Cohn
- Department of Neurology, Drug Development Laboratory, Mary S. Easton Center for Alzheimer’s Disease Research and Care, The David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA90095
| | - Jessica T. Lee
- Department of Neurology, Drug Development Laboratory, Mary S. Easton Center for Alzheimer’s Disease Research and Care, The David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA90095
| | - Chunni Zhu
- Department of Neurology, Drug Development Laboratory, Mary S. Easton Center for Alzheimer’s Disease Research and Care, The David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA90095
| | - Christine S. Huang
- Department of Neurobiology, The David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA90095
| | - László Molnár
- Department of Electrical Engineering, Sapientia Hungarian University of Transylvania, Târgu Mureş540485, Romania
| | - Carolyn R. Houser
- Department of Neurobiology, The David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA90095
| | - Varghese John
- Department of Neurology, Drug Development Laboratory, Mary S. Easton Center for Alzheimer’s Disease Research and Care, The David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA90095
| | - Istvan Mody
- Department of Neurology, The David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA90095
- Department of Physiology, The David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA90095
| |
Collapse
|
3
|
Smith SS, Benanni S, Jones Q, Kenney L, Evrard MR. Manipulation of α4βδ GABA A receptors alters synaptic pruning in layer 3 prelimbic prefrontal cortex and impairs temporal order recognition: Implications for schizophrenia and autism. Brain Res 2024; 1835:148929. [PMID: 38599510 DOI: 10.1016/j.brainres.2024.148929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/04/2024] [Accepted: 04/06/2024] [Indexed: 04/12/2024]
Abstract
Temporal order memory is impaired in autism spectrum disorder (ASD) and schizophrenia (SCZ). These disorders, more prevalent in males, result in abnormal dendritic spine pruning during adolescence in layer 3 (L3) medial prefrontal cortex (mPFC), yielding either too many (ASD) or too few (SCZ) spines. Here we tested whether altering spine density in neural circuits including the mPFC could be associated with impaired temporal order memory in male mice. We have shown that α4βδ GABAA receptors (GABARs) emerge at puberty on spines of L5 prelimbic mPFC (PL) where they trigger pruning. We show here that α4βδ receptors also increase at puberty in L3 PL (P < 0.0001) and used these receptors as a target to manipulate spine density here. Pubertal injection (14 d) of the GABA agonist gaboxadol, at a dose (3 mg/kg) selective for α4βδ, reduced L3 spine density by half (P < 0.0001), while α4 knock-out increased spine density ∼ 40 % (P < 0.0001), mimicking spine densities in SCZ and ASD, respectively. In both cases, performance on the mPFC-dependent temporal order recognition task was impaired, resulting in decreases in the discrimination ratio which assesses preference for the novel object: -0.39 ± 0.15, gaboxadol versus 0.52 ± 0.09, vehicle; P = 0.0002; -0.048 ± 0.10, α4 KO versus 0.49 ± 0.04, wild-type; P < 0.0001. In contrast, the number of approaches was unaltered, reflecting unchanged locomotion. These data suggest that altering α4βδ GABAR expression/activity alters spine density in L3 mPFC and impairs temporal order memory to mimic changes in ASD and SCZ. These findings may provide insight into these disorders.
Collapse
Affiliation(s)
- Sheryl S Smith
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY 11203, USA.
| | - Safae Benanni
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY 11203, USA.
| | - Quiana Jones
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY 11203, USA.
| | - Lindsay Kenney
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY 11203, USA.
| | - Matthew R Evrard
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY 11203, USA.
| |
Collapse
|
4
|
Menzikov SA, Zaichenko DM, Moskovtsev AA, Morozov SG, Kubatiev AA. Phenols and GABA A receptors: from structure and molecular mechanisms action to neuropsychiatric sequelae. Front Pharmacol 2024; 15:1272534. [PMID: 38303988 PMCID: PMC10831359 DOI: 10.3389/fphar.2024.1272534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/03/2024] [Indexed: 02/03/2024] Open
Abstract
γ-Aminobutyric acid type A receptors (GABAARs) are members of the pentameric ligand-gated ion channel (pLGIC) family, which are widespread throughout the invertebrate and vertebrate central nervous system. GABAARs are engaged in short-term changes of the neuronal concentrations of chloride (Cl-) and bicarbonate (HCO3 -) ions by their passive permeability through the ion channel pore. GABAARs are regulated by various structurally diverse phenolic substances ranging from simple phenols to complex polyphenols. The wide chemical and structural variability of phenols suggest similar and different binding sites on GABAARs, allowing them to manifest themselves as activators, inhibitors, or allosteric ligands of GABAAR function. Interest in phenols is associated with their great potential for GABAAR modulation, but also with their subsequent negative or positive role in neurological and psychiatric disorders. This review focuses on the GABAergic deficit hypotheses during neurological and psychiatric disorders induced by various phenols. We summarize the structure-activity relationship of general phenol groups concerning their differential roles in the manifestation of neuropsychiatric symptoms. We describe and analyze the role of GABAAR subunits in manifesting various neuropathologies and the molecular mechanisms underlying their modulation by phenols. Finally, we discuss how phenol drugs can modulate GABAAR activity via desensitization and resensitization. We also demonstrate a novel pharmacological approach to treat neuropsychiatric disorders via regulation of receptor phosphorylation/dephosphorylation.
Collapse
|
5
|
Wei X, Campagna JJ, Jagodzinska B, Wi D, Cohn W, Lee J, Zhu C, Huang CS, Molnár L, Houser CR, John V, Mody I. A therapeutic small molecule lead enhances γ-oscillations and improves cognition/memory in Alzheimer's disease model mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.04.569994. [PMID: 38106006 PMCID: PMC10723366 DOI: 10.1101/2023.12.04.569994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Brain rhythms provide the timing and concurrence of brain activity required for linking together neuronal ensembles engaged in specific tasks. In particular, the γ-oscillations (30-120 Hz) orchestrate neuronal circuits underlying cognitive processes and working memory. These oscillations are reduced in numerous neurological and psychiatric disorders, including early cognitive decline in Alzheimer's disease (AD). Here we report on a potent brain permeable small molecule, DDL-920 that increases γ-oscillations and improves cognition/memory in a mouse model of AD, thus showing promise as a new class of therapeutics for AD. As a first in CNS pharmacotherapy, our lead candidate acts as a potent, efficacious, and selective negative allosteric modulator (NAM) of the γ-aminobutyric acid type A receptors (GABA A Rs) assembled from α1β2δ subunits. We identified these receptors through anatomical and pharmacological means to mediate the tonic inhibition of parvalbumin (PV) expressing interneurons (PV+INs) critically involved in the generation of γ-oscillations. Our approach is unique as it is meant to enhance cognitive performance and working memory in a state-dependent manner by engaging and amplifying the brain's endogenous γ-oscillations through enhancing the function of PV+INs.
Collapse
|
6
|
Adraoui FW, Douw L, Martens GJM, Maas DA. Connecting Neurobiological Features with Interregional Dysconnectivity in Social-Cognitive Impairments of Schizophrenia. Int J Mol Sci 2023; 24:ijms24097680. [PMID: 37175387 PMCID: PMC10177877 DOI: 10.3390/ijms24097680] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Schizophrenia (SZ) is a devastating psychiatric disorder affecting about 1% of the world's population. Social-cognitive impairments in SZ prevent positive social interactions and lead to progressive social withdrawal. The neurobiological underpinnings of social-cognitive symptoms remain poorly understood, which hinders the development of novel treatments. At the whole-brain level, an abnormal activation of social brain regions and interregional dysconnectivity within social-cognitive brain networks have been identified as major contributors to these symptoms. At the cellular and subcellular levels, an interplay between oxidative stress, neuroinflammation and N-methyl-D-aspartate receptor hypofunction is thought to underly SZ pathology. However, it is not clear how these molecular processes are linked with interregional dysconnectivity in the genesis of social-cognitive symptoms. Here, we aim to bridge the gap between macroscale (connectivity analyses) and microscale (molecular and cellular mechanistic) knowledge by proposing impaired myelination and the disinhibition of local microcircuits as possible causative biological pathways leading to dysconnectivity and abnormal activity of the social brain. Furthermore, we recommend electroencephalography as a promising translational technique that can foster pre-clinical drug development and discuss attractive drug targets for the treatment of social-cognitive symptoms in SZ.
Collapse
Affiliation(s)
- Florian W Adraoui
- Biotrial, Preclinical Pharmacology Department, 7-9 rue Jean-Louis Bertrand, 35000 Rennes, France
| | - Linda Douw
- Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan, 1081 HZ Amsterdam, The Netherlands
| | - Gerard J M Martens
- Donders Centre for Neuroscience (DCN), Department of Molecular Animal Physiology, Faculty of Science, Donders Institute for Brain, Cognition and Behavior, Radboud University, 6525 GA Nijmegen, The Netherlands
- NeuroDrug Research Ltd., 6525 ED Nijmegen, The Netherlands
| | - Dorien A Maas
- Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
7
|
El Haj M, Moustafa AA. "Ten dollars today or 50 dollars after one month?" Temporal discounting in Korsakoff syndrome. Cogn Neuropsychiatry 2023; 28:116-129. [PMID: 36724487 DOI: 10.1080/13546805.2023.2173059] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Little research has investigated decision making in patients with Korsakoff syndrome (KS). Specifically, to our knowledge, there is a lack of research investigating whether patients with KS may tend to prefer immediate over future rewards (i.e., temporal discounting). Further, we investigated the relationship between temporal discounting and inhibition. METHODS We, for the first time, invited patients with KS and control participants to perform a temporal discounting task, in which they answered questions probing preferences between an immediate, but smaller amount of money, and a delayed, but larger amount of money (e.g., "would you prefer 10 dollars today or 50 dollars after one month?"). Furthermore, inhibition was measured using the Stroop Colour Word Test. RESULTS Analysis demonstrated higher temporal discounting in patients with KS than in control participants. Temporal discounting in both populations was significantly correlated with inhibition. CONCLUSIONS Patients with KS may have difficulties to suppress the temptation of smaller, but immediate, rewards.
Collapse
Affiliation(s)
- Mohamad El Haj
- Laboratoire de Psychologie des Pays de la Loire (LPPL - EA 4638), Nantes Université, Univ Angers Nantes, France
- Unité de Gériatrie, Centre Hospitalier de Tourcoing, Tourcoing, France
- Institut Universitaire de France, Paris, France
| | - Ahmed A Moustafa
- School of Psychology, Faculty of Society and Design, Bond University, Gold Coast, Australia
- Department of Human Anatomy and Physiology, the Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| |
Collapse
|
8
|
Dorsolateral Prefrontal Cortex Glutamate/Gamma-Aminobutyric Acid (GABA) Alterations in Clinical High Risk and First-Episode Schizophrenia: A Preliminary 7-T Magnetic Resonance Spectroscopy Imaging Study. Int J Mol Sci 2022; 23:ijms232415846. [PMID: 36555487 PMCID: PMC9781166 DOI: 10.3390/ijms232415846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Converging lines of evidence suggest that an imbalance between excitation and inhibition is present in the dorsolateral prefrontal cortex (DLPFC) of schizophrenia (SCZ). Gamma-aminobutyric-acid (GABA) and, to a lesser extent, glutamate (Glu) abnormalities were reported in the DLPFC of SCZ patients, especially on the right hemisphere, by post-mortem studies. However, in vivo evidence of GABA, Glu, and Glu/GABA DLPFC abnormalities, particularly on the right side and the early stages of illness, is limited. In this preliminary study, we utilized 7-Tesla magnetic resonance spectroscopic imaging (MRSI) to investigate bilateral Glu/Creatine (Cre), GABA/Cre, and Glu/GABA in the DLPFC of sixteen first episode schizophrenia (FES), seventeen clinical high risk (CHR), and twenty-six healthy comparison (HC) subjects. FES and CHR had abnormal GABA/Cre and Glu/GABA in the right DLPFC (rDLPFC) compared with HC participants, while no differences were observed in the left DLPFC (lDLPFC) among the three groups. Furthermore, HC had higher Glu/GABA in rDLPFC compared to lDLPFC (R > L), whereas the opposite relationship (R < L) was observed in the DLPFC Glu/GABA of FES patients. Altogether, these findings indicate that GABA/Cre and Glu/GABA DLPFC alterations are present before illness manifestation and worsen in FES patients, thus representing a putative early pathophysiological biomarker for SCZ and related psychotic disorders.
Collapse
|
9
|
Lukow PB, Martins D, Veronese M, Vernon AC, McGuire P, Turkheimer FE, Modinos G. Cellular and molecular signatures of in vivo imaging measures of GABAergic neurotransmission in the human brain. Commun Biol 2022; 5:372. [PMID: 35440709 PMCID: PMC9018713 DOI: 10.1038/s42003-022-03268-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 03/14/2022] [Indexed: 11/21/2022] Open
Abstract
Diverse GABAergic interneuron networks orchestrate information processing in the brain. Understanding the principles underlying the organisation of this system in the human brain, and whether these principles are reflected by available non-invasive in vivo neuroimaging methods, is crucial for the study of GABAergic neurotransmission. Here, we use human gene expression data and state-of-the-art imaging transcriptomics to uncover co-expression patterns between genes encoding GABAA receptor subunits and inhibitory interneuron subtype-specific markers, and their association with binding patterns of the gold-standard GABA PET radiotracers [11C]Ro15-4513 and [11C]flumazenil. We found that the inhibitory interneuron marker somatostatin covaries with GABAA receptor-subunit genes GABRA5 and GABRA2, and that their distribution followed [11C]Ro15-4513 binding. In contrast, the inhibitory interneuron marker parvalbumin covaried with GABAA receptor-subunit genes GABRA1, GABRB2 and GABRG2, and their distribution tracked [11C]flumazenil binding. Our findings indicate that existing PET radiotracers may provide complementary information about key components of the GABAergic system.
Collapse
Affiliation(s)
- Paulina Barbara Lukow
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 16 De Crespigny Park, SE5 8AF, London, UK.
| | - Daniel Martins
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 16 De Crespigny Park, SE5 8AF, London, UK
- NIHR Maudsley Biomedical Research Centre, De Crespigny Park, Denmark Hill, London, SE5 8AF, UK
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 16 De Crespigny Park, SE5 8AF, London, UK
- NIHR Maudsley Biomedical Research Centre, De Crespigny Park, Denmark Hill, London, SE5 8AF, UK
- Department of Information Engineering, University of Padua, Via Giovanni Gradenigo, 6, 35131, Padova, PD, Italy
| | - Anthony Christopher Vernon
- Department of Basic & Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe Road, Brixton, London, SE5 9RT, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, New Hunt's House, Guy's Campus, London, UK
| | - Philip McGuire
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 16 De Crespigny Park, SE5 8AF, London, UK
- NIHR Maudsley Biomedical Research Centre, De Crespigny Park, Denmark Hill, London, SE5 8AF, UK
| | - Federico Edoardo Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 16 De Crespigny Park, SE5 8AF, London, UK
| | - Gemma Modinos
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 16 De Crespigny Park, SE5 8AF, London, UK
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 16 De Crespigny Park, SE5 8AF, London, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, New Hunt's House, Guy's Campus, London, UK
| |
Collapse
|
10
|
Reversing the Psychiatric Effects of Neurodevelopmental Cannabinoid Exposure: Exploring Pharmacotherapeutic Interventions for Symptom Improvement. Int J Mol Sci 2021; 22:ijms22157861. [PMID: 34360626 PMCID: PMC8346164 DOI: 10.3390/ijms22157861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 12/16/2022] Open
Abstract
Neurodevelopmental exposure to psychoactive compounds in cannabis, specifically THC, is associated with a variety of long-term psychopathological outcomes. This increased risk includes a higher prevalence of schizophrenia, mood and anxiety disorders, and cognitive impairments. Clinical and pre-clinical research continues to identify a wide array of underlying neuropathophysiological sequelae and mechanisms that may underlie THC-related psychiatric risk vulnerability, particularly following adolescent cannabis exposure. A common theme among these studies is the ability of developmental THC exposure to induce long-term adaptations in the mesocorticolimbic system which resemble pathological endophenotypes associated with these disorders. This narrative review will summarize recent clinical and pre-clinical evidence that has elucidated these THC-induced developmental risk factors and examine how specific pharmacotherapeutic interventions may serve to reverse or perhaps prevent these cannabis-related risk outcomes.
Collapse
|
11
|
Laviolette SR. Exploring the impact of adolescent exposure to cannabinoids and nicotine on psychiatric risk: insights from translational animal models. Psychol Med 2021; 51:940-947. [PMID: 31801641 DOI: 10.1017/s0033291719003325] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Adolescence represents a highly sensitive period of mammalian neurodevelopment wherein critical synaptic and structural changes are taking place in brain regions involved in cognition, self-regulation and emotional processing. Importantly, neural circuits such as the mesocorticolimbic pathway, comprising the prefrontal cortex, sub-cortical mesolimbic dopamine system and their associated input/output centres, are particularly vulnerable to drug-related insults. Human adolescence represents a life-period wherein many individuals first begin to experiment with recreational drugs such as nicotine and cannabis, both of which are known to profoundly modulate neurochemical signalling within the mesocorticolimbic pathway and to influence both long-term and acute neuropsychiatric symptoms. While a vast body of epidemiological clinical research has highlighted the effects of adolescent exposure to drugs such as nicotine and cannabis on the developing adolescent brain, many of these studies are limited to correlative analyses and rely on retrospective self-reports from subjects, making causal interpretations difficult to discern. The use of pre-clinical animal studies can avoid these issues by allowing for precise temporal and dose-related experimental control over drug exposure during adolescence. In addition, such animal-based research has the added advantage of allowing for in-depth molecular, pharmacological, genetic and neuronal analyses of how recreational drug exposure may set up the brain for neuropsychiatric risk. This review will explore some of the advantages and disadvantages of these models, with a focus on the common, divergent and synergistic effects of adolescent nicotine and cannabis exposure on neuropsychiatric risk.
Collapse
Affiliation(s)
- Steven R Laviolette
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, CanadaN6A3K7
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, CanadaN6A3K7
- Department of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, CanadaN6A3K7
| |
Collapse
|
12
|
Liao VWY, Chebib M, Ahring PK. Efficient expression of concatenated α1β2δ and α1β3δ GABA A receptors, their pharmacology and stoichiometry. Br J Pharmacol 2021; 178:1556-1573. [PMID: 33491192 DOI: 10.1111/bph.15380] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/28/2020] [Accepted: 01/11/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE GABAA receptors containing δ-subunits are notorious for being difficult to study in vitro due to heterogeneity of expressed receptor populations and low GABA-evoked current amplitudes. Thus, there are some published misconceptions and contradictory conclusions made regarding the pharmacology and stoichiometry of δ-containing receptors. The aim of this study was to obtain robust homogenous expression of α1βδ receptors for in-depth investigation. EXPERIMENTAL APPROACH Novel δ-containing pentameric concatenated constructs were designed. The resulting α1β2δ and α1β3δ GABAA receptor concatemers were investigated by two-electrode voltage-clamp electrophysiology using Xenopus laevis oocytes. KEY RESULTS First, while homogenous α1βδ GABAA receptor pools could not be obtained by manipulating the ratio of injected cRNAs of free α1, β2/3, and δ subunits, concatenated pentameric α1β2δ and α1β3δ constructs resulted in robust expression levels of concatemers. Second, by using optimised constructs that give unidirectional assembly of concatemers, we found that the δ subunit cannot directly participate in GABA binding and receptor activation. Hence, functional δ-containing receptors are likely to all have a conventional 2α:2β:1δ stoichiometry arranged as βαβαδ when viewed counterclockwise from the extracellular side. Third, α1β2/3δ receptors were found to express efficiently in X. laevis oocytes but have a low estimated open probability of ~0.5% upon GABA activation. Because of this, these receptors are uniquely susceptible to positive allosteric modulation by, for example, neurosteroids. CONCLUSION AND IMPLICATIONS Our data answer important outstanding questions regarding the pharmacology and stoichiometry of α1δ-containing GABAA receptors and pave the way for future analysis and drug discovery efforts.
Collapse
Affiliation(s)
- Vivian Wan Yu Liao
- Brain and Mind Centre, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Mary Chebib
- Brain and Mind Centre, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Philip Kiaer Ahring
- Brain and Mind Centre, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
13
|
Gault JM, Thompson JA, Maharajh K, Hosokawa P, Stevens KE, Olincy A, Liedtke EI, Ojemann A, Ojemann S, Abosch A. Striatal and Thalamic Auditory Response During Deep Brain Stimulation for Essential Tremor: Implications for Psychosis. Neuromodulation 2020; 23:478-488. [PMID: 32022409 DOI: 10.1111/ner.13101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 12/04/2019] [Accepted: 01/02/2020] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The P50, a positive auditory-evoked potential occurring 50 msec after an auditory click, has been characterized extensively with electroencephalography (EEG) to detect aberrant auditory electrophysiology in disorders like schizophrenia (SZ) where 61-74% have an auditory gating deficit. The P50 response occurs in primary auditory cortex and several thalamocortical regions. In rodents, the gated P50 response has been identified in the reticular thalamic nucleus (RT)-a deep brain structure traversed during deep brain stimulation (DBS) targeting of the ventral intermediate nucleus (VIM) of the thalamus to treat essential tremor (ET) allowing for interspecies comparison. The goal was to utilize the unique opportunity provided by DBS surgery for ET to map the P50 response in multiple deep brain structures in order to determine the utility of intraoperative P50 detection for facilitating DBS targeting of auditory responsive subterritories. MATERIALS AND METHODS We developed a method to assess P50 response intraoperatively with local field potentials (LFP) using microelectrode recording during routine clinical electrophysiologic mapping for awake DBS surgery in seven ET patients. Recording sites were mapped into a common stereotactic space. RESULTS Forty significant P50 responses of 155 recordings mapped to the ventral thalamus, RT and CN head/body interface at similar rates of 22.7-26.7%. P50 response exhibited anatomic specificity based on distinct positions of centroids of positive and negative responses within brain regions and the fact that P50 response was not identified in the recordings from either the internal capsule or the dorsal thalamus. CONCLUSIONS Detection of P50 response intraoperatively may guide DBS targeting RT and subterritories within CN head/body interface-DBS targets with the potential to treat psychosis and shown to modulate schizophrenia-like aberrancies in mouse models.
Collapse
Affiliation(s)
- Judith M Gault
- Department of Neurosurgery, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA.,Department of Psychiatry, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - John A Thompson
- Department of Neurosurgery, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Keeran Maharajh
- Department of Psychiatry, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA.,Department of Neurology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Patrick Hosokawa
- Department of Neurosurgery, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Karen E Stevens
- Department of Psychiatry, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Ann Olincy
- Department of Psychiatry, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Erin I Liedtke
- Department of Neurosurgery, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Alex Ojemann
- Department of Neurosurgery, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Steven Ojemann
- Department of Neurosurgery, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Aviva Abosch
- Department of Neurosurgery, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA.,Department of Neurology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
14
|
Impaired Interneuron Development in a Novel Model of Neonatal Brain Injury. eNeuro 2019; 6:eN-NWR-0300-18. [PMID: 30809588 PMCID: PMC6390196 DOI: 10.1523/eneuro.0300-18.2019] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 12/27/2018] [Accepted: 01/03/2019] [Indexed: 12/20/2022] Open
Abstract
Prematurity is associated with significantly increased risk of neurobehavioral pathologies, including autism and schizophrenia. A common feature of these psychiatric disorders is prefrontal cortex (PFC) inhibitory circuit disruption due to GABAergic interneuron alteration. Cortical interneurons are generated and migrate throughout late gestation and early infancy, making them highly susceptible to perinatal insults such as preterm birth. Term and preterm PFC pathology specimens were assessed using immunohistochemical markers for interneurons. Based on the changes seen, a new preterm encephalopathy mouse model was developed to produce similar PFC interneuron loss. Maternal immune activation (MIA; modeling chorioamnionitis, associated with 85% of extremely preterm births) was combined with chronic sublethal hypoxia (CSH; modeling preterm respiratory failure), with offspring of both sexes assessed anatomically, molecularly and neurobehaviorally. In the PFC examined from the human preterm samples compared to matched term samples at corrected age, a decrease in somatostatin (SST) and calbindin (CLB) interneurons was seen in upper cortical layers. This pattern of interneuron loss in upper cortical layers was mimicked in the mouse PFC following the combination of MIA and CSH, but not after either insult alone. This persistent interneuron loss is associated with postnatal microglial activation that occurs during CSH only after MIA. The combined insults lead to long-term neurobehavioral deficits which parallel human psychopathologies that may be seen after extremely preterm birth. This new preclinical model supports a paradigm in which specific cellular alterations seen in preterm encephalopathy can be linked with a risk of neuropsychiatric sequela. Specific interneuron subtypes may provide therapeutic targets to prevent or ameliorate these neurodevelopmental risks.
Collapse
|
15
|
Chemogenetic Isolation Reveals Synaptic Contribution of δ GABA A Receptors in Mouse Dentate Granule Neurons. J Neurosci 2018; 38:8128-8145. [PMID: 30076210 DOI: 10.1523/jneurosci.0799-18.2018] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 07/18/2018] [Accepted: 07/24/2018] [Indexed: 11/21/2022] Open
Abstract
Two major GABAA receptor classes mediate ionotropic GABA signaling, those containing a δ subunit and those with a γ2 subunit. The classical viewpoint equates γ2-containing receptors with IPSCs and δ-containing receptors with tonic inhibition because of differences in receptor localization, but significant questions remain because the populations cannot be pharmacologically separated. We removed this barrier using gene editing to confer a point mutation on the δ subunit in mice, rendering receptors containing the subunit picrotoxin resistant. By pharmacologically isolating δ-containing receptors, our results demonstrate their contribution to IPSCs in dentate granule neurons and weaker contributions to thalamocortical IPSCs. Despite documented extrasynaptic localization, we found that receptor localization does not preclude participation in isolated IPSCs, including mIPSCs. Further, phasic inhibition from δ subunit-containing receptors strongly inhibited summation of EPSPs, whereas tonic activity had little impact. In addition to any role that δ-containing receptors may play in canonical tonic inhibition, our results highlight a previously underestimated contribution of δ-containing receptors to phasic inhibition.SIGNIFICANCE STATEMENT GABAA receptors play key roles in transient and tonic inhibition. The prevailing view suggests that synaptic γ2-containing GABAA receptors drive phasic inhibition, whereas extrasynaptic δ-containing receptors mediate tonic inhibition. To re-evaluate the impact of δ receptors, we took a chemogenetic approach that offers a sensitive method to probe the synaptic contribution of δ-containing receptors. Our results reveal that localization does not strongly limit the contribution of δ receptors to IPSCs and that δ receptors make an unanticipated robust contribution to phasic inhibition.
Collapse
|
16
|
Yang GJ, Murray JD, Glasser M, Pearlson GD, Krystal JH, Schleifer C, Repovs G, Anticevic A. Altered Global Signal Topography in Schizophrenia. Cereb Cortex 2018; 27:5156-5169. [PMID: 27702810 DOI: 10.1093/cercor/bhw297] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 08/28/2016] [Indexed: 02/04/2023] Open
Abstract
Schizophrenia (SCZ) is a disabling neuropsychiatric disease associated with disruptions across distributed neural systems. Resting-state functional magnetic resonance imaging has identified extensive abnormalities in the blood-oxygen level-dependent signal in SCZ patients, including alterations in the average signal over the brain-i.e. the "global" signal (GS). It remains unknown, however, if these "global" alterations occur pervasively or follow a spatially preferential pattern. This study presents the first network-by-network quantification of GS topography in healthy subjects and SCZ patients. We observed a nonuniform GS contribution in healthy comparison subjects, whereby sensory areas exhibited the largest GS component. In SCZ patients, we identified preferential GS representation increases across association regions, while sensory regions showed preferential reductions. GS representation in sensory versus association cortices was strongly anti-correlated in healthy subjects. This anti-correlated relationship was markedly reduced in SCZ. Such shifts in GS topography may underlie profound alterations in neural information flow in SCZ, informing development of pharmacotherapies.
Collapse
Affiliation(s)
- Genevieve J Yang
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, New Haven, CT 06511, USA.,Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA.,Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT 06519, USA
| | - John D Murray
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, New Haven, CT 06511, USA
| | - Matthew Glasser
- Department of Neurobiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Godfrey D Pearlson
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, New Haven, CT 06511, USA.,Olin Neuropsychiatry Research Center, Institute of Living, Hartford Hospital, 200 Retreat Avenue, Hartford, CT 06106, USA
| | - John H Krystal
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, New Haven, CT 06511, USA.,Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA.,NIAAA Center for the Translational Neuroscience of Alcoholism, New Haven, CT 06519, USA
| | - Charlie Schleifer
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, New Haven, CT 06511, USA
| | - Grega Repovs
- Department of Psychology, University of Ljubljana, Ljubljana, Slovenia
| | - Alan Anticevic
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, New Haven, CT 06511, USA.,Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA.,Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT 06519, USA.,Olin Neuropsychiatry Research Center, Institute of Living, Hartford Hospital, 200 Retreat Avenue, Hartford, CT 06106, USA.,NIAAA Center for the Translational Neuroscience of Alcoholism, New Haven, CT 06519, USA.,Department of Psychology, Yale University, 2 Hillhouse Avenue, New Haven, CT 06520, USA
| |
Collapse
|
17
|
Puvvada KC, Summerfelt A, Du X, Krishna N, Kochunov P, Rowland LM, Simon JZ, Hong LE. Delta Vs Gamma Auditory Steady State Synchrony in Schizophrenia. Schizophr Bull 2018; 44:378-387. [PMID: 29036430 PMCID: PMC5814801 DOI: 10.1093/schbul/sbx078] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Background Delta band (1-4 Hz) neuronal responses support the precision and stability of auditory processing, and a deficit in delta band synchrony may be relevant to auditory domain symptoms in schizophrenia patients. Methods Delta band synchronization elicited by a 2.5 Hz auditory steady state response (ASSR) paradigm, along with those from theta (5 Hz), alpha (10 Hz), beta (20 Hz), gamma (40 Hz), and high gamma (80 Hz) frequency ASSR, were compared in 128 patients with schizophrenia, 108 healthy controls, and 55 first-degree relatives (FDR) of patients. Results Delta band synchronization was significantly impaired in patients compared with controls (F = 18.3, P < .001). There was a significant 2.5 Hz by 40 Hz ASSR interaction (P = .023), arising from a greater reduction of 2.5 Hz ASSR than of 40 Hz ASSR, in patients compared with controls. Greater deficit in delta ASSR was associated with auditory perceptual abnormality (P = .007) and reduced verbal working memory (P < .001). Gamma frequency ASSR impairment was also significant but more modest (F = 8.7, P = .004), and this deficit was also present in FDR (P = .022). Conclusions The ability to sustain delta band oscillation entrainment in the auditory pathway is significantly reduced in schizophrenia patients and appears to be clinically relevant.
Collapse
Affiliation(s)
- Krishna C Puvvada
- Department of Electrical & Computer Engineering, University of Maryland, College Park, MD
| | - Ann Summerfelt
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD
| | - Xiaoming Du
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD
| | - Nithin Krishna
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD
| | - Peter Kochunov
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD
| | - Laura M Rowland
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD
| | - Jonathan Z Simon
- Department of Electrical & Computer Engineering, University of Maryland, College Park, MD
- Department of Biology, University of Maryland, College Park, MD
- Institute for Systems Research, University of Maryland, College Park, MD
| | - L Elliot Hong
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
18
|
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: 4.3] [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
|
19
|
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: 90] [Impact Index Per Article: 11.3] [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
|
20
|
Zhang WH, Zhou J, Pan HQ, Wang XY, Liu WZ, Zhang JY, Yin XP, Pan BX. δ Subunit-containing GABA A receptor prevents overgeneralization of fear in adult mice. ACTA ACUST UNITED AC 2017; 24:381-384. [PMID: 28716958 PMCID: PMC5516689 DOI: 10.1101/lm.045856.117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 05/08/2017] [Indexed: 11/25/2022]
Abstract
The role of δ subunit-containing GABAA receptor (GABAA(δ)R) in fear generalization is uncertain. Here, by using mice with or without genetic deletion of GABAA(δ)R and using protocols in which the conditioned tone stimuli were cross presented with different nonconditioned stimuli, we observed that when the two tone stimuli were largely similar, both genotypes froze similarly to either of them. However, when they differed markedly, the knockout mice froze much more than their wild-type littermates to the nonconditioned but not conditioned stimuli. Thus, GABAA(δ)R may prevent inappropriate fear generalization when the incoming stimuli differ clearly from the learned threat.
Collapse
Affiliation(s)
- Wen-Hua Zhang
- Department of Neurology, the 2nd Affiliated Hospital, Nanchang University, Nanchang, 330031, China.,Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Jin Zhou
- Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, Nanchang, 330031, China.,School of Life Science, Nanchang University, Nanchang, 330031, China
| | - Han-Qing Pan
- Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, Nanchang, 330031, China.,School of Life Science, Nanchang University, Nanchang, 330031, China
| | - Xiao-Yang Wang
- Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, Nanchang, 330031, China.,School of Life Science, Nanchang University, Nanchang, 330031, China
| | - Wei-Zhu Liu
- Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, Nanchang, 330031, China.,School of Life Science, Nanchang University, Nanchang, 330031, China
| | - Jun-Yu Zhang
- Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Xiao-Ping Yin
- Department of Neurology, the 2nd Affiliated Hospital, Nanchang University, Nanchang, 330031, China
| | - Bing-Xing Pan
- Department of Neurology, the 2nd Affiliated Hospital, Nanchang University, Nanchang, 330031, China.,Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, Nanchang, 330031, China.,School of Life Science, Nanchang University, Nanchang, 330031, China
| |
Collapse
|
21
|
Simonsen C, Boddum K, von Schoubye NL, Kloppenburg A, Sønderskov K, Hansen SL, Kristiansen U. Anticonvulsive evaluation of THIP in the murine pentylenetetrazole kindling model: lack of anticonvulsive effect of THIP despite functional δ-subunit-containing GABA A receptors in dentate gyrus granule cells. Pharmacol Res Perspect 2017; 5. [PMID: 28805971 PMCID: PMC5684853 DOI: 10.1002/prp2.322] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 04/20/2017] [Accepted: 04/21/2017] [Indexed: 12/29/2022] Open
Abstract
THIP (4,5,6,7‐tetrahydroisoxazolo[5,4‐c]pyridin‐3‐ol) is a GABAA receptor agonist with varying potencies and efficacies at γ‐subunit‐containing receptors. More importantly, THIP acts as a selective superagonist at δ‐subunit‐containing receptors (δ‐GABAARs) at clinically relevant concentrations. Evaluation of THIP as a potential anticonvulsant has given contradictory results in different animal models and for this reason, we reevaluated the anticonvulsive properties of THIP in the murine pentylenetetrazole (PTZ) kindling model. As loss of δ‐GABAAR in the dentate gyrus has been associated with several animal models of epilepsy, we first investigated the presence of functional δ‐GABAA receptors. Both immunohistochemistry and Western blot data demonstrated that δ‐GABAAR expression is not only present in the dentate gyrus, but also the expression level was enhanced in the early phase after PTZ kindling. Whole‐cell patch‐clamp studies in acute hippocampal brain slices revealed that THIP was indeed able to induce a tonic inhibition in dentate gyrus granule cells. However, THIP induced a tonic current of similar magnitude in the PTZ‐kindled mice compared to saline‐treated animals despite the observed upregulation of δ‐GABAARs. Even in the demonstrated presence of functional δ‐GABAARs, THIP (0.5–4 mg/kg) showed no anticonvulsive effect in the PTZ kindling model using a comprehensive in vivo evaluation of the anticonvulsive properties.
Collapse
Affiliation(s)
- Charlotte Simonsen
- Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Kim Boddum
- Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nadia L von Schoubye
- Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alissa Kloppenburg
- Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Kasper Sønderskov
- Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Suzanne L Hansen
- Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Uffe Kristiansen
- Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Neuroimaging studies of GABA in schizophrenia: a systematic review with meta-analysis. Transl Psychiatry 2017; 7:e1147. [PMID: 28585933 PMCID: PMC5537645 DOI: 10.1038/tp.2017.124] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 04/26/2017] [Accepted: 05/03/2017] [Indexed: 01/17/2023] Open
Abstract
Data from animal models and from postmortem studies suggest that schizophrenia is associated with brain GABAergic dysfunction. The extent to which this is reflected in data from in vivo studies of GABA function in schizophrenia is unclear. The Medline database was searched to identify articles published until 21 October 2016. The search terms included GABA, proton magnetic resonance spectroscopy (1H-MRS), positron emission tomography (PET), single photon emission computed tomography (SPECT), schizophrenia and psychosis. Sixteen GABA 1H-MRS studies (538 controls, 526 patients) and seven PET/SPECT studies of GABAA/benzodiazepine receptor (GABAA/BZR) availability (118 controls, 113 patients) were identified. Meta-analyses of 1H-MRS GABA in the medial prefrontal cortex (mPFC), parietal/occipital cortex (POC) and striatum did not show significant group differences (mFC: g=-0.3, 409 patients, 495 controls, 95% confidence interval (CI): -0.6 to 0.1; POC: g=-0.3, 139 patients, 111 controls, 95% CI: -0.9 to 0.3; striatum: g=-0.004, 123 patients, 95 controls, 95% CI: -0.7 to 0.7). Heterogeneity across studies was high (I2>50%), and this was not explained by subsequent moderator or meta-regression analyses. There were insufficient PET/SPECT receptor availability studies for meta-analyses, but a systematic review did not suggest replicable group differences in regional GABAA/BZR availability. The current literature does not reveal consistent alterations in in vivo GABA neuroimaging measures in schizophrenia, as might be hypothesized from animal models and postmortem data. The analysis highlights the need for further GABA neuroimaging studies with improved methodology and addressing potential sources of heterogeneity.
Collapse
|
23
|
Fatemi SH, Folsom TD, Thuras PD. GABA A and GABA B receptor dysregulation in superior frontal cortex of subjects with schizophrenia and bipolar disorder. Synapse 2017; 71. [PMID: 28316115 DOI: 10.1002/syn.21973] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 02/21/2017] [Accepted: 03/14/2017] [Indexed: 11/12/2022]
Abstract
Schizophrenia and bipolar disorder are complex psychiatric disorders that affect millions of people worldwide. Evidence from gene association and postmortem studies has identified abnormalities of the gamma-aminobutyric acid (GABA) signaling system in both disorders. Abnormal GABAergic signaling and transmission could contribute to the symptomatology of these disorders, potentially through impaired gamma oscillations which normally occur during cognitive processing. In the current study, we examined the protein expression of 14 GABAA and two GABAB receptor subunits in the superior frontal cortex of subjects with schizophrenia, bipolar disorder, and healthy controls. Analyses of Variance (ANOVAs) identified significant group effects for protein levels for the α1, α6, β1, β3, δ, ɛ, and π GABAA receptor subunits and R1 and R2 GABAB receptor subunits. Follow-up t tests confirmed changes for these subunits in subjects with schizophrenia, subjects with bipolar disorder, or both groups. Alterations in stoichiometry of GABA receptor subunits could result in altered ligand binding, transmission, and pharmacology of GABA receptors in superior frontal cortex. Thus, impaired GABAergic transmission may negatively contribute to symptoms such as anxiety or panic as well as impaired learning and information processing, all of which are disrupted in schizophrenia and bipolar disorder. Taken together, these results provide additional evidence of GABAergic receptor abnormalities in these disorders.
Collapse
Affiliation(s)
- S Hossein Fatemi
- Department of Psychiatry, Division of Neuroscience Research, University of Minnesota Medical School, 420 Delaware St. SE, MMC 392, Minneapolis, Minnesota, 55455.,Department of Neuroscience, University of Minnesota Medical School, 321 Church St. SE, Minneapolis, Minnesota, 55455
| | - Timothy D Folsom
- Department of Psychiatry, Division of Neuroscience Research, University of Minnesota Medical School, 420 Delaware St. SE, MMC 392, Minneapolis, Minnesota, 55455
| | - Paul D Thuras
- Department of Psychiatry, VA Medical Center, 1 Veterans Drive Minneapolis, Minnesota, 55417-2399
| |
Collapse
|
24
|
de Jonge JC, Vinkers CH, Hulshoff Pol HE, Marsman A. GABAergic Mechanisms in Schizophrenia: Linking Postmortem and In Vivo Studies. Front Psychiatry 2017; 8:118. [PMID: 28848455 PMCID: PMC5554536 DOI: 10.3389/fpsyt.2017.00118] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/22/2017] [Indexed: 01/11/2023] Open
Abstract
Schizophrenia is a psychiatric disorder characterized by hallucinations, delusions, disorganized thinking, and impairments in cognitive functioning. Evidence from postmortem studies suggests that alterations in cortical γ-aminobutyric acid (GABAergic) neurons contribute to the clinical features of schizophrenia. In vivo measurement of brain GABA levels using magnetic resonance spectroscopy (MRS) offers the possibility to provide more insight into the relationship between problems in GABAergic neurotransmission and clinical symptoms of schizophrenia patients. This study reviews and links alterations in the GABA system in postmortem studies, animal models, and human studies in schizophrenia. Converging evidence implicates alterations in both presynaptic and postsynaptic components of GABAergic neurotransmission in schizophrenia, and GABA may thus play an important role in the pathophysiology of schizophrenia. MRS studies can provide direct insight into the GABAergic mechanisms underlying the development of schizophrenia as well as changes during its course.
Collapse
Affiliation(s)
- Jeroen C de Jonge
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, Netherlands
| | - Christiaan H Vinkers
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, Netherlands
| | - Hilleke E Hulshoff Pol
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, Netherlands
| | - Anouk Marsman
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, Netherlands.,Danish Research Centre for Magnetic Resonance, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| |
Collapse
|
25
|
Modgil A, Parakala ML, Ackley MA, Doherty JJ, Moss SJ, Davies PA. Endogenous and synthetic neuroactive steroids evoke sustained increases in the efficacy of GABAergic inhibition via a protein kinase C-dependent mechanism. Neuropharmacology 2016; 113:314-322. [PMID: 27743930 DOI: 10.1016/j.neuropharm.2016.10.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 10/03/2016] [Accepted: 10/09/2016] [Indexed: 11/16/2022]
Abstract
The neuroactive steroid (NAS) tetrahydrodeoxycorticosterone (THDOC) increases protein kinase C (PKC) mediated phosphorylation of extrasynaptic GABAA receptor (GABAAR) subunits leading to increased surface expression of α4/β3 subunit-containing extrasynaptic GABAARs, leading to a sustained increase in GABAAR tonic current density. Whether other naturally occurring and synthetic NASs share both an allosteric and metabotropic action on GABAARs is unknown. Here, we examine the allosteric and metabotropic properties of allopregnanolone (ALLO), and synthetic NASs SGE-516 and ganaxolone. ALLO, SGE-516, and ganaxolone all allosterically enhanced prototypical synaptic and extrasynaptic recombinant GABAARs. In dentate gyrus granule cells (DGGCs) all three NASs, when applied acutely, allosterically enhanced tonic and phasic GABAergic currents. In separate experiments, slices were exposed to NASs for 15 min, and then transferred to a steroid naïve recording chamber followed by ≥ 30 min wash before tonic currents were measured. A sustained increase in tonic current was observed following exposure to ALLO, or SGE-516 and was prevented by inhibiting PKC with GF 109203X. No increase in tonic current was observed with exposure to ganaxolone. In agreement with the observations of an increased tonic current, the NASs ALLO and SGE-516 increased the phosphorylation and surface expression of the β3 subunit-containing GABAARs. Our studies demonstrate that neuroactive steroids have differential abilities to induce sustained increases in the efficacy of tonic inhibition by promoting GABAAR phosphorylation and membrane trafficking dependent on PKC activity.
Collapse
Affiliation(s)
- Amit Modgil
- Tufts University School of Medicine, Department of Neuroscience, 136 Harrison Ave, Boston, MA 02111, USA
| | - Manasa L Parakala
- Tufts University School of Medicine, Department of Neuroscience, 136 Harrison Ave, Boston, MA 02111, USA
| | | | | | - Stephen J Moss
- Tufts University School of Medicine, Department of Neuroscience, 136 Harrison Ave, Boston, MA 02111, USA; Department of Neuroscience, Physiology and Pharmacology, University College, London WC1E6BT, United Kingdom
| | - Paul A Davies
- Tufts University School of Medicine, Department of Neuroscience, 136 Harrison Ave, Boston, MA 02111, USA.
| |
Collapse
|
26
|
Yuan NY, Poe MM, Witzigmann C, Cook JM, Stafford D, Arnold LA. Characterization of GABA A receptor ligands with automated patch-clamp using human neurons derived from pluripotent stem cells. J Pharmacol Toxicol Methods 2016; 82:109-114. [PMID: 27544543 DOI: 10.1016/j.vascn.2016.08.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 05/31/2016] [Accepted: 08/14/2016] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Automated patch clamp is a recent but widely used technology to assess pre-clinical drug safety. With the availability of human neurons derived from pluripotent stem cells, this technology can be extended to determine CNS effects of drug candidates, especially those acting on the GABAA receptor. METHODS iCell Neurons (Cellular Dynamics International, A Fujifilm Company) were cultured for ten days and analyzed by patch clamp in the presence of agonist GABA or in combination with positive allosteric GABAA receptor modulators. Both efficacy and affinity were determined. In addition, mRNA of GABAA receptor subunits were quantified by qRT-PCR. RESULTS We have shown that iCell Neurons are compatible with the IonFlux microfluidic system of the automated patch clamp instrument. Resistance ranging from 15 to 25MΩ was achieved for each trap channel of patch clamped cells in a 96-well plate format. GABA induced a robust change of current with an EC50 of 0.43μM. Positive GABAA receptor modulators diazepam, HZ-166, and CW-04-020 exhibited EC50 values of 0.42μM, 1.56μM, and 0.23μM, respectively. The α2/α3/α5 selective compound HZ-166-induced the highest potentiation (efficacy) of 810% of the current induced by 100nM GABA. Quantification of GABAA receptor mRNA in iCell Neurons revealed high levels of α5 and β3 subunits and low levels of α1, which is similar to the configuration in human neonatal brain. DISCUSSION iCell Neurons represent a new cellular model to characterize GABAergic compounds using automated patch clamp. These cells have excellent representation of cellular GABAA receptor distribution that enable determination of total small molecule efficacy and affinity as measured by cell membrane current change.
Collapse
Affiliation(s)
- Nina Y Yuan
- Department of Chemistry and Biochemistry and Milwaukee Institute for Drug Discovery, University of Wisconsin - Milwaukee, 3210 N. Cramer Street, Milwaukee, WI 53211, United States.
| | - Michael M Poe
- Department of Chemistry and Biochemistry and Milwaukee Institute for Drug Discovery, University of Wisconsin - Milwaukee, 3210 N. Cramer Street, Milwaukee, WI 53211, United States.
| | - Christopher Witzigmann
- Department of Chemistry and Biochemistry and Milwaukee Institute for Drug Discovery, University of Wisconsin - Milwaukee, 3210 N. Cramer Street, Milwaukee, WI 53211, United States.
| | - James M Cook
- Department of Chemistry and Biochemistry and Milwaukee Institute for Drug Discovery, University of Wisconsin - Milwaukee, 3210 N. Cramer Street, Milwaukee, WI 53211, United States.
| | - Douglas Stafford
- Department of Chemistry and Biochemistry and Milwaukee Institute for Drug Discovery, University of Wisconsin - Milwaukee, 3210 N. Cramer Street, Milwaukee, WI 53211, United States.
| | - Leggy A Arnold
- Department of Chemistry and Biochemistry and Milwaukee Institute for Drug Discovery, University of Wisconsin - Milwaukee, 3210 N. Cramer Street, Milwaukee, WI 53211, United States.
| |
Collapse
|
27
|
Canetta S, Bolkan S, Padilla-Coreano N, Song L, Sahn R, Harrison N, Gordon JA, Brown A, Kellendonk C. Maternal immune activation leads to selective functional deficits in offspring parvalbumin interneurons. Mol Psychiatry 2016; 21:956-68. [PMID: 26830140 PMCID: PMC4914410 DOI: 10.1038/mp.2015.222] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 12/04/2015] [Accepted: 12/15/2015] [Indexed: 12/26/2022]
Abstract
Abnormalities in prefrontal gamma aminobutyric acid (GABA)ergic transmission, particularly in fast-spiking interneurons that express parvalbumin (PV), are hypothesized to contribute to the pathophysiology of multiple psychiatric disorders, including schizophrenia, bipolar disorder, anxiety disorders and depression. While primarily histological abnormalities have been observed in patients and in animal models of psychiatric disease, evidence for abnormalities in functional neurotransmission at the level of specific interneuron populations has been lacking in animal models and is difficult to establish in human patients. Using an animal model of a psychiatric disease risk factor, prenatal maternal immune activation (MIA), we found reduced functional GABAergic transmission in the medial prefrontal cortex (mPFC) of adult MIA offspring. Decreased transmission was selective for interneurons expressing PV, resulted from a decrease in release probability and was not observed in calretinin-expressing neurons. This deficit in PV function in MIA offspring was associated with increased anxiety-like behavior and impairments in attentional set shifting, but did not affect working memory. Furthermore, cell-type specific optogenetic inhibition of mPFC PV interneurons was sufficient to impair attentional set shifting and enhance anxiety levels. Finally, we found that in vivo mPFC gamma oscillations, which are supported by PV interneuron function, were linearly correlated with the degree of anxiety displayed in adult mice, and that this correlation was disrupted in MIA offspring. These results demonstrate a selective functional vulnerability of PV interneurons to MIA, leading to affective and cognitive symptoms that have high relevance for schizophrenia and other psychiatric disorders.
Collapse
Affiliation(s)
- Sarah Canetta
- Department of Psychiatry, Mailman School of Public Health, Columbia University Medical Center, New York, NY 10032, USA
| | - Scott Bolkan
- Department of Psychiatry, Mailman School of Public Health, Columbia University Medical Center, New York, NY 10032, USA
| | - Nancy Padilla-Coreano
- Department of Psychiatry, Mailman School of Public Health, Columbia University Medical Center, New York, NY 10032, USA,Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA
| | - LouJin Song
- Department of Pharmacology, Mailman School of Public Health, Columbia University Medical Center, New York, NY 10032, USA
| | | | - Neil Harrison
- Department of Pharmacology, Mailman School of Public Health, Columbia University Medical Center, New York, NY 10032, USA,Department of Anesthesiology, Mailman School of Public Health, Columbia University Medical Center, New York, NY 10032, USA
| | - Joshua A. Gordon
- Department of Psychiatry, Mailman School of Public Health, Columbia University Medical Center, New York, NY 10032, USA,Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Alan Brown
- Department of Psychiatry, Mailman School of Public Health, Columbia University Medical Center, New York, NY 10032, USA,Department of Epidemiology, Mailman School of Public Health, Columbia University Medical Center, New York, NY 10032, USA,Divison of Epidemiology, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Christoph Kellendonk
- Department of Psychiatry, Mailman School of Public Health, Columbia University Medical Center, New York, NY 10032, USA,Department of Pharmacology, Mailman School of Public Health, Columbia University Medical Center, New York, NY 10032, USA,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| |
Collapse
|
28
|
Iura A, Takahashi A, Hakata S, Mashimo T, Fujino Y. Reductions in tonic GABAergic current in substantia gelatinosa neurons and GABAAreceptor δ subunit expression after chronic constriction injury of the sciatic nerve in mice. Eur J Pain 2016; 20:1678-1688. [DOI: 10.1002/ejp.891] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2016] [Indexed: 11/09/2022]
Affiliation(s)
- A. Iura
- Department of Anesthesiology and Intensive Care Medicine; Graduate School of Medicine; Osaka University; Japan
| | - A. Takahashi
- Department of Anesthesiology and Intensive Care Medicine; Graduate School of Medicine; Osaka University; Japan
| | - S. Hakata
- Department of Anesthesiology and Intensive Care Medicine; Graduate School of Medicine; Osaka University; Japan
| | - T. Mashimo
- Toyonaka Municipal Hospital; Osaka Japan
| | - Y. Fujino
- Department of Anesthesiology and Intensive Care Medicine; Graduate School of Medicine; Osaka University; Japan
| |
Collapse
|
29
|
GABAergic Function as a Limiting Factor for Prefrontal Maturation during Adolescence. Trends Neurosci 2016; 39:441-448. [PMID: 27233681 DOI: 10.1016/j.tins.2016.04.010] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 04/22/2016] [Accepted: 04/25/2016] [Indexed: 11/24/2022]
Abstract
Adolescence is a vulnerable period for the onset of mental illnesses including schizophrenia and affective disorders, yet the neurodevelopmental processes underlying this vulnerability remain poorly understood. The prefrontal cortex (PFC) and its local GABAergic system are thought to contribute to the core of cognitive deficits associated with such disorders. However, clinical and preclinical end-point analyses performed in adults are likely to give limited insight into the cellular mechanisms that are altered during adolescence but are only manifested in adulthood. This perspective summarizes work regarding the developmental trajectories of the GABAergic system in the PFC during adolescence to provide an insight into the increased susceptibility to psychiatric disorders during this critical developmental period.
Collapse
|
30
|
Bolkan SS, Carvalho Poyraz F, Kellendonk C. Using human brain imaging studies as a guide toward animal models of schizophrenia. Neuroscience 2016; 321:77-98. [PMID: 26037801 PMCID: PMC4664583 DOI: 10.1016/j.neuroscience.2015.05.055] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/15/2015] [Accepted: 05/21/2015] [Indexed: 12/17/2022]
Abstract
Schizophrenia is a heterogeneous and poorly understood mental disorder that is presently defined solely by its behavioral symptoms. Advances in genetic, epidemiological and brain imaging techniques in the past half century, however, have significantly advanced our understanding of the underlying biology of the disorder. In spite of these advances clinical research remains limited in its power to establish the causal relationships that link etiology with pathophysiology and symptoms. In this context, animal models provide an important tool for causally testing hypotheses about biological processes postulated to be disrupted in the disorder. While animal models can exploit a variety of entry points toward the study of schizophrenia, here we describe an approach that seeks to closely approximate functional alterations observed with brain imaging techniques in patients. By modeling these intermediate pathophysiological alterations in animals, this approach offers an opportunity to (1) tightly link a single functional brain abnormality with its behavioral consequences, and (2) to determine whether a single pathophysiology can causally produce alterations in other brain areas that have been described in patients. In this review we first summarize a selection of well-replicated biological abnormalities described in the schizophrenia literature. We then provide examples of animal models that were studied in the context of patient imaging findings describing enhanced striatal dopamine D2 receptor function, alterations in thalamo-prefrontal circuit function, and metabolic hyperfunction of the hippocampus. Lastly, we discuss the implications of findings from these animal models for our present understanding of schizophrenia, and consider key unanswered questions for future research in animal models and human patients.
Collapse
Affiliation(s)
- S S Bolkan
- Department of Pharmacology, Columbia University, New York, NY 10032, USA; Department of Psychiatry, Columbia University, New York, NY 10032, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - F Carvalho Poyraz
- Department of Pharmacology, Columbia University, New York, NY 10032, USA; Department of Psychiatry, Columbia University, New York, NY 10032, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - C Kellendonk
- Department of Pharmacology, Columbia University, New York, NY 10032, USA; Department of Psychiatry, Columbia University, New York, NY 10032, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA.
| |
Collapse
|
31
|
Beas BS, McQuail JA, Ban Uelos C, Setlow B, Bizon JL. Prefrontal cortical GABAergic signaling and impaired behavioral flexibility in aged F344 rats. Neuroscience 2016; 345:274-286. [PMID: 26873002 DOI: 10.1016/j.neuroscience.2016.02.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 02/03/2016] [Accepted: 02/05/2016] [Indexed: 02/07/2023]
Abstract
The prefrontal cortex (PFC) is critical for the ability to flexibly adapt established patterns of behavior in response to a change in environmental contingencies. Impaired behavioral flexibility results in maladaptive strategies such as perseveration on response options that no longer produce a desired outcome. Pharmacological manipulations of prefrontal cortical GABAergic signaling modulate behavioral flexibility in animal models, and prefrontal cortical interneuron dysfunction is implicated in impaired behavioral flexibility that accompanies neuropsychiatric disease. As deficits in behavioral flexibility also emerge during the normal aging process, the goal of this study was to determine the role of GABAergic signaling, specifically via prefrontal cortical GABA(B) receptors, in such age-related deficits. Young and aged rats were trained in a set shifting task performed in operant chambers. First, rats learned to discriminate between two response levers to obtain a food reward on the basis of a cue light illuminated above the correct lever. Upon acquisition of this initial discrimination, the contingencies were shifted such that rats had to ignore the cue light and respond on the levers according to their left/right positions. Both young and aged rats acquired the initial discrimination similarly; however, aged rats were impaired relative to young following the set shift. Among aged rats, GABA(B) receptor expression in the medial prefrontal cortex (mPFC) was strongly correlated with set shifting, such that lower expression was associated with worse performance. Subsequent experiments showed that intra-mPFC administration of the GABA(B) receptor agonist baclofen enhanced set shifting performance in aged rats. These data directly link GABAergic signaling via GABA(B) receptors to impaired behavioral flexibility associated with normal aging.
Collapse
Affiliation(s)
- B S Beas
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States.
| | - J A McQuail
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States.
| | - C Ban Uelos
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States.
| | - B Setlow
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States; Department of Psychiatry, University of Florida College of Medicine, Gainesville, FL, United States; Department of Psychology, University of Florida, Gainesville, FL, United States.
| | - J L Bizon
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States; Department of Psychiatry, University of Florida College of Medicine, Gainesville, FL, United States.
| |
Collapse
|
32
|
Bohnsack JP, Carlson SL, Morrow AL. Differential regulation of synaptic and extrasynaptic α4 GABA(A) receptor populations by protein kinase A and protein kinase C in cultured cortical neurons. Neuropharmacology 2016; 105:124-132. [PMID: 26767953 DOI: 10.1016/j.neuropharm.2016.01.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 12/07/2015] [Accepted: 01/04/2016] [Indexed: 11/24/2022]
Abstract
The GABAA α4 subunit exists in two distinct populations of GABAA receptors. Synaptic GABAA α4 receptors are localized at the synapse and mediate phasic inhibitory neurotransmission, while extrasynaptic GABAA receptors are located outside of the synapse and mediate tonic inhibitory transmission. These receptors have distinct pharmacological and biophysical properties that contribute to interest in how these different subtypes are regulated under physiological and pathological states. We utilized subcellular fractionation procedures to separate these populations of receptors in order to investigate their regulation by protein kinases in cortical cultured neurons. Protein kinase A (PKA) activation decreases synaptic α4 expression while protein kinase C (PKC) activation increases α4 subunit expression, and these effects are associated with increased β3 S408/409 or γ2 S327 phosphorylation respectively. In contrast, PKA activation increases extrasynaptic α4 and δ subunit expression, while PKC activation has no effect. Our findings suggest synaptic and extrasynaptic GABAA α4 subunit expression can be modulated by PKA to inform the development of more specific therapeutics for neurological diseases that involve deficits in GABAergic transmission.
Collapse
Affiliation(s)
- John Peyton Bohnsack
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7365, USA; Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill NC, 27599-7178, USA
| | - Stephen L Carlson
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill NC, 27599-7178, USA
| | - A Leslie Morrow
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7365, USA; Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7365, USA; Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill NC, 27599-7178, USA.
| |
Collapse
|
33
|
Functional hierarchy underlies preferential connectivity disturbances in schizophrenia. Proc Natl Acad Sci U S A 2015; 113:E219-28. [PMID: 26699491 DOI: 10.1073/pnas.1508436113] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Schizophrenia may involve an elevated excitation/inhibition (E/I) ratio in cortical microcircuits. It remains unknown how this regulatory disturbance maps onto neuroimaging findings. To address this issue, we implemented E/I perturbations within a neural model of large-scale functional connectivity, which predicted hyperconnectivity following E/I elevation. To test predictions, we examined resting-state functional MRI in 161 schizophrenia patients and 164 healthy subjects. As predicted, patients exhibited elevated functional connectivity that correlated with symptom levels, and was most prominent in association cortices, such as the fronto-parietal control network. This pattern was absent in patients with bipolar disorder (n = 73). To account for the pattern observed in schizophrenia, we integrated neurobiologically plausible, hierarchical differences in association vs. sensory recurrent neuronal dynamics into our model. This in silico architecture revealed preferential vulnerability of association networks to E/I imbalance, which we verified empirically. Reported effects implicate widespread microcircuit E/I imbalance as a parsimonious mechanism for emergent inhomogeneous dysconnectivity in schizophrenia.
Collapse
|
34
|
Distinctive transcriptome alterations of prefrontal pyramidal neurons in schizophrenia and schizoaffective disorder. Mol Psychiatry 2015; 20:1397-405. [PMID: 25560755 PMCID: PMC4492919 DOI: 10.1038/mp.2014.171] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 10/14/2014] [Accepted: 11/12/2014] [Indexed: 12/12/2022]
Abstract
Schizophrenia is associated with alterations in working memory that reflect dysfunction of dorsolateral prefrontal cortex (DLPFC) circuitry. Working memory depends on the activity of excitatory pyramidal cells in DLPFC layer 3 and, to a lesser extent, in layer 5. Although many studies have profiled gene expression in DLPFC gray matter in schizophrenia, little is known about cell-type-specific transcript expression in these two populations of pyramidal cells. We hypothesized that interrogating gene expression, specifically in DLPFC layer 3 or 5 pyramidal cells, would reveal new and/or more robust schizophrenia-associated differences that would provide new insights into the nature of pyramidal cell dysfunction in the illness. We also sought to determine the impact of other variables, such as a diagnosis of schizoaffective disorder or medication use at the time of death, on the patterns of gene expression in pyramidal neurons. Individual pyramidal cells in DLPFC layers 3 or 5 were captured by laser microdissection from 36 subjects with schizophrenia or schizoaffective disorder and matched normal comparison subjects. The mRNA from cell collections was subjected to transcriptome profiling by microarray followed by quantitative PCR validation. Expression of genes involved in mitochondrial (MT) or ubiquitin-proteasome system (UPS) functions were markedly downregulated in the patient group (P-values for MT-related and UPS-related pathways were <10(-7) and <10(-5), respectively). MT-related gene alterations were more prominent in layer 3 pyramidal cells, whereas UPS-related gene alterations were more prominent in layer 5 pyramidal cells. Many of these alterations were not present, or found to a lesser degree, in samples of DLPFC gray matter from the same subjects, suggesting that they are pyramidal cell specific. Furthermore, these findings principally reflected alterations in the schizophrenia subjects were not present or present to a lesser degree in the schizoaffective disorder subjects (diagnosis of schizoaffective disorder was the most significant covariate, P<10(-6)) and were not attributable to factors frequently comorbid with schizophrenia. In summary, our findings reveal expression deficits in MT- and UPS-related genes specific to layer 3 and/or layer 5 pyramidal cells in the DLPFC of schizophrenia subjects. These cell type-specific transcriptome signatures are not characteristic of schizoaffective disorder, providing a potential molecular-cellular basis of differences in clinical phenotypes.
Collapse
|
35
|
Bristow GC, Bostrom JA, Haroutunian V, Sodhi MS. Sex differences in GABAergic gene expression occur in the anterior cingulate cortex in schizophrenia. Schizophr Res 2015; 167:57-63. [PMID: 25660468 PMCID: PMC4524801 DOI: 10.1016/j.schres.2015.01.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 01/15/2015] [Accepted: 01/17/2015] [Indexed: 01/30/2023]
Abstract
GABAergic dysfunction has been strongly implicated in the pathophysiology of schizophrenia. In this study, we analyzed the expression levels of several GABAergic genes in the anterior cingulate cortex (ACC) of postmortem subjects with schizophrenia (n=21) and a comparison group of individuals without a history of psychiatric illness (n=18). Our analyses revealed a significant sex by diagnosis effect, along with significant differences in GABAergic gene expression based on medication status. Analyses revealed that in male groups, the expression of GABAergic genes was generally lower in schizophrenia cases compared to the controls, with significantly lower expression levels of GABA-Aα5, GABA-Aβ1, and GABA-Aε. In females, the expression of GABAergic genes was higher in the schizophrenia cases, with significantly higher expression of the GABA-Aβ1 and GAD67 genes. Analysis of the effect of medication in the schizophrenia subjects revealed significantly higher expression of GABA-Aα1-3, GABA-Aβ2, GABA-Aγ2, and GAD67 in the medicated group compared to the unmedicated group. These data show that sex differences in the expression of GABAergic genes occur in the ACC in schizophrenia. Therefore, our data support previous findings of GABAergic dysfunction in schizophrenia and emphasize the importance of considering sex in analyses of the pathophysiology of schizophrenia. Sex differences in the GABAergic regulation of ACC function may contribute to the differences observed in the symptoms of male and female patients with schizophrenia. In addition, our findings indicate that antipsychotic medications may alter GABAergic signaling in the ACC, supporting the potential of GABAergic targets for the development of novel antipsychotic medication.
Collapse
Affiliation(s)
- Greg C. Bristow
- Department of Pharmacy Practice and Center for Pharmaceutical Biotechnology, College of Pharmacy, University of Illinois at Chicago, Chicago IL 60607, USA.,Corresponding author: Dr. Greg Bristow, Department of Pharmacy Practice, University of Illinois at Chicago, 900 S. Ashland Ave., MC870, Chicago IL 60607-4067, USA. Tel: +1-312-996-1413, Fax: +1-312-413-9303,
| | - John A. Bostrom
- Department of Pharmacy Practice and Center for Pharmaceutical Biotechnology, College of Pharmacy, University of Illinois at Chicago, Chicago IL 60607, USA
| | - Vahram Haroutunian
- Department of Psychiatry, Mount Sinai School of Medicine, New York, NY, USA
| | - Monsheel S. Sodhi
- Department of Pharmacy Practice and Center for Pharmaceutical Biotechnology, College of Pharmacy, University of Illinois at Chicago, Chicago IL 60607, USA.,Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
36
|
Fatemi SH, Folsom TD. GABA receptor subunit distribution and FMRP-mGluR5 signaling abnormalities in the cerebellum of subjects with schizophrenia, mood disorders, and autism. Schizophr Res 2015; 167:42-56. [PMID: 25432637 PMCID: PMC5301472 DOI: 10.1016/j.schres.2014.10.010] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 10/06/2014] [Accepted: 10/08/2014] [Indexed: 12/24/2022]
Abstract
Gamma-aminobutyric acid (GABA) is the main inhibitory neurotransmitter in the brain. GABAergic receptor abnormalities have been documented in several major psychiatric disorders including schizophrenia, mood disorders, and autism. Abnormal expression of mRNA and protein for multiple GABA receptors has also been observed in multiple brain regions leading to alterations in the balance between excitatory/inhibitory signaling in the brain with potential profound consequences for normal cognition and maintenance of mood and perception. Altered expression of GABAA receptor subunits has been documented in fragile X mental retardation 1 (FMR1) knockout mice, suggesting that loss of its protein product, fragile X mental retardation protein (FMRP), impacts GABAA subunit expression. Recent postmortem studies from our laboratory have shown reduced expression of FMRP in the brains of subjects with schizophrenia, bipolar disorder, major depression, and autism. FMRP acts as a translational repressor and, under normal conditions, inhibits metabotropic glutamate receptor 5 (mGluR5)-mediated signaling. In fragile X syndrome (FXS), the absence of FMRP is hypothesized to lead to unregulated mGluR5 signaling, ultimately resulting in the behavioral and intellectual impairments associated with this disorder. Our laboratory has identified changes in mGluR5 expression in autism, schizophrenia, and mood disorders. In the current review article, we discuss our postmortem data on GABA receptors, FMRP, and mGluR5 levels and compare our results with other laboratories. Finally, we discuss the interactions between these molecules and the potential for new therapeutic interventions that target these interconnected signaling systems.
Collapse
Affiliation(s)
- S Hossein Fatemi
- Department of Psychiatry, Division of Neuroscience Research, University of Minnesota Medical School, 420 Delaware St SE, MMC 392, Minneapolis, MN 55455, USA; Department of Neuroscience, University of Minnesota Medical School, 321 Church St. SE, Minneapolis, MN 55455, USA.
| | - Timothy D Folsom
- Department of Psychiatry, Division of Neuroscience Research, University of Minnesota Medical School, 420 Delaware St SE, MMC 392, Minneapolis, MN 55455, USA.
| |
Collapse
|
37
|
Martin BS, Martinez-Botella G, Loya CM, Salituro FG, Robichaud AJ, Huntsman MM, Ackley MA, Doherty JJ, Corbin JG. Rescue of deficient amygdala tonic γ-aminobutyric acidergic currents in the Fmr-/y mouse model of fragile X syndrome by a novel γ-aminobutyric acid type A receptor-positive allosteric modulator. J Neurosci Res 2015; 94:568-78. [PMID: 26308557 DOI: 10.1002/jnr.23632] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 08/01/2015] [Accepted: 08/04/2015] [Indexed: 11/07/2022]
Abstract
Alterations in the ratio of excitatory to inhibitory transmission are emerging as a common component of many nervous system disorders, including autism spectrum disorders (ASDs). Tonic γ-aminobutyric acidergic (GABAergic) transmission provided by peri- and extrasynaptic GABA type A (GABAA ) receptors powerfully controls neuronal excitability and plasticity and, therefore, provides a rational therapeutic target for normalizing hyperexcitable networks across a variety of disorders, including ASDs. Our previous studies revealed tonic GABAergic deficits in principal excitatory neurons in the basolateral amygdala (BLA) in the Fmr1(-/y) knockout (KO) mouse model fragile X syndrome. To correct amygdala deficits in tonic GABAergic neurotransmission in Fmr1(-/y) KO mice, we developed a novel positive allosteric modulator of GABAA receptors, SGE-872, based on endogenously active neurosteroids. This study shows that SGE-872 is nearly as potent and twice as efficacious for positively modulating GABAA receptors as its parent molecule, allopregnanolone. Furthermore, at submicromolar concentrations (≤1 μM), SGE-872 is selective for tonic, extrasynaptic α4β3δ-containing GABAA receptors over typical synaptic α1β2γ2 receptors. We further find that SGE-872 strikingly rescues the tonic GABAergic transmission deficit in principal excitatory neurons in the Fmr1(-/y) KO BLA, a structure heavily implicated in the neuropathology of ASDs. Therefore, the potent and selective action of SGE-872 on tonic GABAA receptors containing α4 subunits may represent a novel and highly useful therapeutic avenue for ASDs and related disorders involving hyperexcitability of neuronal networks.
Collapse
Affiliation(s)
- Brandon S Martin
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC
| | | | - Carlos M Loya
- SAGE Therapeutics, 215 First Street, Cambridge, MA 0214243
| | | | | | - Molly M Huntsman
- Pharmaceutical Sciences and Department of Pediatrics, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Mike A Ackley
- SAGE Therapeutics, 215 First Street, Cambridge, MA 0214243
| | | | - Joshua G Corbin
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC
| |
Collapse
|
38
|
Kumar M, González LA, Dillon GH. Assessment of subunit-dependent direct gating and allosteric modulatory effects of carisoprodol at GABA(A) receptors. Neuropharmacology 2015; 97:414-25. [PMID: 25896767 DOI: 10.1016/j.neuropharm.2015.04.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 04/02/2015] [Accepted: 04/03/2015] [Indexed: 10/23/2022]
Abstract
Carisoprodol is a widely prescribed muscle relaxant, abuse of which has grown considerably in recent years. It directly activates and allosterically modulates α1β2γ2 GABAARs, although the site(s) of action are unknown. To gain insight into the actions of carisoprodol, subunit-dependent effects of this drug were assessed. Whole-cell patch clamp recordings were obtained from HEK293 cells expressing α1β2, α1β3 or αxβzγ2 (where x = 1-6 and z = 1-3) GABAARs, and in receptors incorporating the δ subunit (modeling extrasynaptic receptors). The ability to directly gate and allosterically potentiate GABA-gated currents was observed for all configurations. Presence or absence of the γ2 subunit did not affect the ability of carisoprodol to directly gate or allosterically modulate the receptor. Presence of the β1 subunit conferred highest efficacy for direct activation relative to maximum GABA currents, while presence of the β2 subunit conferred highest efficacy for allosteric modulation of the GABA response. With regard to α subunits, carisoprodol was most efficacious at enhancing the actions of GABA in receptors incorporating the α1 subunit. The ability to directly gate the receptor was generally comparable regardless of the α subunit isoform, although receptors incorporating the α3 subunit showed significantly reduced direct gating efficacy and affinity. In extrasynaptic (α1β3δ and α4β3δ) receptors, carisoprodol had greater efficacy than GABA as a direct gating agonist. In addition, carisoprodol allosterically potentiated both EC20 and saturating GABA concentrations in these receptors. In assessing voltage-dependence, we found direct gating and inhibitory effects were insensitive to membrane voltage, whereas allosteric modulatory effects were affected by membrane voltage. Our findings demonstrate direct and allosteric effects of carisoprodol at synaptic and extrasynpatic GABAARs and that subunit isoform influences these effects.
Collapse
Affiliation(s)
- Manoj Kumar
- Department of Physiology and Pharmacology and Center for Neuroscience, West Virginia University School of Medicine, Robert C. Byrd Health Sciences Center, Morgantown, WV 26506, USA
| | - Lorie A González
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Glenn H Dillon
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; Department of Physiology and Pharmacology and Center for Neuroscience, West Virginia University School of Medicine, Robert C. Byrd Health Sciences Center, Morgantown, WV 26506, USA.
| |
Collapse
|
39
|
Schmidt MJ, Mirnics K. Neurodevelopment, GABA system dysfunction, and schizophrenia. Neuropsychopharmacology 2015; 40:190-206. [PMID: 24759129 PMCID: PMC4262918 DOI: 10.1038/npp.2014.95] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/03/2014] [Accepted: 04/11/2014] [Indexed: 02/07/2023]
Abstract
The origins of schizophrenia have eluded clinicians and researchers since Kraepelin and Bleuler began documenting their findings. However, large clinical research efforts in recent decades have identified numerous genetic and environmental risk factors for schizophrenia. The combined data strongly support the neurodevelopmental hypothesis of schizophrenia and underscore the importance of the common converging effects of diverse insults. In this review, we discuss the evidence that genetic and environmental risk factors that predispose to schizophrenia disrupt the development and normal functioning of the GABAergic system.
Collapse
Affiliation(s)
- Martin J Schmidt
- Department of Psychiatry, Vanderbilt University, Nashville, TN, USA
| | - Karoly Mirnics
- Department of Psychiatry, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN, USA
- Department of Psychiatry, University of Szeged, Szeged, Hungary
| |
Collapse
|
40
|
Greenberg Z, Ramshaw H, Schwarz Q. Time Windows of Interneuron Development: Implications to Our Understanding of the Aetiology and Treatment of Schizophrenia. AIMS Neurosci 2015. [DOI: 10.3934/neuroscience.2015.4.294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
41
|
Hoftman GD, Volk DW, Bazmi HH, Li S, Sampson AR, Lewis DA. Altered cortical expression of GABA-related genes in schizophrenia: illness progression vs developmental disturbance. Schizophr Bull 2015; 41:180-91. [PMID: 24361861 PMCID: PMC4266281 DOI: 10.1093/schbul/sbt178] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Schizophrenia is a neurodevelopmental disorder with altered expression of GABA-related genes in the prefrontal cortex (PFC). However, whether these gene expression abnormalities reflect disturbances in postnatal developmental processes before clinical onset or arise as a consequence of clinical illness remains unclear. METHODS Expression levels for 7 GABA-related transcripts (vesicular GABA transporter [vGAT], GABA membrane transporter [GAT1], GABAA receptor subunit α1 [GABRA1] [novel in human and monkey cohorts], glutamic acid decarboxylase 67 [GAD67], parvalbumin, calretinin, and somatostatin [previously reported in human cohort, but not in monkey cohort]) were quantified in the PFC from 42 matched pairs of schizophrenia and comparison subjects and from 49 rhesus monkeys ranging in age from 1 week postnatal to adulthood. RESULTS Levels of vGAT and GABRA1, but not of GAT1, messenger RNAs (mRNAs) were lower in the PFC of the schizophrenia subjects. As previously reported, levels of GAD67, parvalbumin, and somatostatin, but not of calretinin, mRNAs were also lower in these subjects. Neither illness duration nor age accounted for the levels of the transcripts with altered expression in schizophrenia. In monkey PFC, developmental changes in expression levels of many of these transcripts were in the opposite direction of the changes observed in schizophrenia. For example, mRNA levels for vGAT, GABRA1, GAD67, and parvalbumin all increased with age. CONCLUSIONS Together with published reports, these findings support the interpretation that the altered expression of GABA-related transcripts in schizophrenia reflects a blunting of normal postnatal development changes, but they cannot exclude a decline during the early stages of clinical illness.
Collapse
Affiliation(s)
- Gil D. Hoftman
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA;,Department of Neuroscience, Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, PA
| | - David W. Volk
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | - H. Holly Bazmi
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | - Siyu Li
- Department of Statistics, University of Pittsburgh, Pittsburgh, PA
| | - Allan R. Sampson
- Department of Statistics, University of Pittsburgh, Pittsburgh, PA
| | - David A. Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA;,Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA,*To whom correspondence should be addressed; University of Pittsburgh, 3811 O’Hara Street, Biomedical Science Tower W1654, Pittsburgh, PA 15213-2593, US; tel: +412-383-8548, fax: +412-624-9910,
| |
Collapse
|
42
|
Comenencia-Ortiz E, Moss SJ, Davies PA. Phosphorylation of GABAA receptors influences receptor trafficking and neurosteroid actions. Psychopharmacology (Berl) 2014; 231:3453-65. [PMID: 24847959 PMCID: PMC4135009 DOI: 10.1007/s00213-014-3617-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 05/02/2014] [Indexed: 01/06/2023]
Abstract
RATIONALE Gamma-aminobutyric acid type A receptors (GABAARs) are the principal mediators of inhibitory transmission in the mammalian central nervous system. GABAARs can be localized at post-synaptic inhibitory specializations or at extrasynaptic sites. While synaptic GABAARs are activated transiently following the release of GABA from presynaptic vesicles, extrasynaptic GABAARs are typically activated continuously by ambient GABA concentrations and thus mediate tonic inhibition. The tonic inhibitory currents mediated by extrasynaptic GABAARs control neuronal excitability and the strength of synaptic transmission. However, the mechanisms by which neurons control the functional properties of extrasynaptic GABAARs had not yet been explored. OBJECTIVES We review GABAARs, how they are assembled and trafficked, and the role phosphorylation has on receptor insertion and membrane stabilization. Finally, we review the modulation of GABAARs by neurosteroids and how GABAAR phosphorylation can influence the actions of neurosteroids. CONCLUSIONS Trafficking and stability of functional channels to the membrane surface are critical for inhibitory efficacy. Phosphorylation of residues within GABAAR subunits plays an essential role in the assembly, trafficking, and cell surface stability of GABAARs. Neurosteroids are produced in the brain and are highly efficacious allosteric modulators of GABAAR-mediated current. This allosteric modulation by neurosteroids is influenced by the phosphorylated state of the GABAAR which is subunit dependent, adding temporal and regional variability to the neurosteroid response. Possible links between neurosteroid actions, phosphorylation, and GABAAR trafficking remain to be explored, but potential novel therapeutic targets may exist for numerous neurological and psychological disorders which are linked to fluctuations in neurosteroid levels and GABAA subunit expression.
Collapse
|
43
|
Altered expression of δGABAA receptors in health and disease. Neuropharmacology 2014; 88:24-35. [PMID: 25128850 DOI: 10.1016/j.neuropharm.2014.08.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Revised: 07/28/2014] [Accepted: 08/03/2014] [Indexed: 01/08/2023]
Abstract
γ-Aminobutyric acid type A receptors that contain the δ subunit (δGABAA receptors) are expressed in multiple types of neurons throughout the central nervous system, where they generate a tonic conductance that shapes neuronal excitability and synaptic plasticity. These receptors regulate a variety of important behavioral functions, including memory, nociception and anxiety, and may also modulate neurogenesis. Given their functional significance, δGABAA receptors are considered to be novel therapeutic targets for the treatment of memory dysfunction, pain, insomnia and mood disorders. These receptors are highly responsive to sedative-hypnotic drugs, general anesthetics and neuroactive steroids. A further remarkable feature of δGABAA receptors is that their expression levels are highly dynamic and fluctuate substantially during development and in response to physiological changes including stress and the reproductive cycle. Furthermore, the expression of these receptors varies in pathological conditions such as alcoholism, fragile X syndrome, epilepsy, depression, schizophrenia, mood disorders and traumatic brain injury. Such fluctuations in receptor expression have significant consequences for behavior and may alter responsiveness to therapeutic drugs. This review considers the alterations in the expression of δGABAA receptors associated with various states of health and disease and the implications of these changes.
Collapse
|
44
|
Datta D, Arion D, Lewis DA. Developmental Expression Patterns of GABAA Receptor Subunits in Layer 3 and 5 Pyramidal Cells of Monkey Prefrontal Cortex. Cereb Cortex 2014; 25:2295-305. [PMID: 24610118 DOI: 10.1093/cercor/bhu040] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Cortical pyramidal neuron activity is regulated in part through inhibitory inputs mediated by GABAA receptors. The subunit composition of these receptors confers distinct functional properties. Thus, developmental shifts in subunit expression will likely influence the characteristics of pyramidal cell firing and the functional maturation of processes that depend on these neurons. We used laser microdissection and PCR to quantify postnatal developmental changes in the expression of GABAA receptor subunits (α1, α2, α5, β2, γ2, and δ) in layer 3 pyramidal cells of monkey prefrontal cortex, which are critical for working memory. To determine the specificity of these changes, we examined glutamate receptor subunits (AMPA Glur1 and NMDA Grin1) and conducted the same analyses in layer 5 pyramidal cells. Expression of GABAA receptor subunit mRNAs changed substantially, whereas glutamate receptor subunit changes were modest over postnatal development. Some transcripts (e.g., GABAA α1) progressively increased from birth until adulthood, whereas others (e.g., GABAA α2) declined with age. Changes in some transcripts were present in only one layer (e.g., GABAA δ). The development of GABAA receptor subunit expression in primate prefrontal pyramidal neurons is protracted and subunit- and layer-specific. These trajectories might contribute to the molecular basis for the maturation of working memory.
Collapse
Affiliation(s)
- Dibyadeep Datta
- Department of Neuroscience Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dominique Arion
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - David A Lewis
- Department of Neuroscience Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
45
|
Ye Z, McGee TP, Houston CM, Brickley SG. The contribution of δ subunit-containing GABAA receptors to phasic and tonic conductance changes in cerebellum, thalamus and neocortex. Front Neural Circuits 2013; 7:203. [PMID: 24391550 PMCID: PMC3870274 DOI: 10.3389/fncir.2013.00203] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 12/09/2013] [Indexed: 12/13/2022] Open
Abstract
We have made use of the δ subunit-selective allosteric modulator DS2 (4-chloro-N-[2-(2-thienyl)imidazo[1,2-a]pyridine-3-yl benzamide) to assay the contribution of δ-GABAARs to tonic and phasic conductance changes in the cerebellum, thalamus and neocortex. In cerebellar granule cells, an enhancement of the tonic conductance was observed for DS2 and the orthosteric agonist THIP (4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridin-3-ol). As expected, DS2 did not alter the properties of GABAA receptor-mediated inhibitory postsynaptic synaptic conductances (IPSCs) supporting a purely extrasynaptic role for δ-GABAARs in cerebellar granule cells. DS2 also enhanced the tonic conductance recorded from thalamic relay neurons of the visual thalamus with no alteration in IPSC properties. However, in addition to enhancing the tonic conductance DS2 also slowed the decay of IPSCs recorded from layer II/III neocortical neurons. A slowing of the IPSC decay also occurred in the presence of the voltage-gated sodium channel blocker TTX. Moreover, under conditions of reduced GABA release the ability of DS2 to enhance the tonic conductance was attenuated. These results indicate that δ-GABAARs can be activated following vesicular GABA release onto neocortical neurons and that the actions of DS2 on the tonic conductance may be influenced by the ambient GABA levels present in particular brain regions.
Collapse
Affiliation(s)
- Zhiwen Ye
- Biophysics Section, Department of Life Sciences, Imperial College London London, UK
| | - Thomas P McGee
- Department of Neuroscience, Physiology and Pharmacology, University College London London, UK
| | - Catriona M Houston
- Biophysics Section, Department of Life Sciences, Imperial College London London, UK
| | - Stephen G Brickley
- Biophysics Section, Department of Life Sciences, Imperial College London London, UK
| |
Collapse
|
46
|
Egawa K, Fukuda A. Pathophysiological power of improper tonic GABA(A) conductances in mature and immature models. Front Neural Circuits 2013; 7:170. [PMID: 24167475 PMCID: PMC3807051 DOI: 10.3389/fncir.2013.00170] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 09/28/2013] [Indexed: 11/25/2022] Open
Abstract
High-affinity extrasynaptic gamma-aminobutyric acid A (GABAA) receptors are tonically activated by low and consistent levels of ambient GABA, mediating chronic inhibition against neuronal excitability (tonic inhibition) and the modulation of neural development. Synaptic (phasic) inhibition is spatially and temporally precise compared with tonic inhibition, which provides blunt yet strong integral inhibitory force by shunting electrical signaling. Although effects of acute modification of tonic inhibition are known, its pathophysiological significance remains unclear because homeostatic regulation of neuronal excitability can compensate for long-term deficit of extrasynaptic GABAA receptor activation. Nevertheless, tonic inhibition is of great interest for its pathophysiological involvement in central nervous system (CNS) diseases and thus as a therapeutic target. Together with the development of experimental models for various pathological states, recent evidence demonstrates such pathological involvements of tonic inhibition in neuronal dysfunction. This review focuses on the recent progress of tonic activation of GABAA conductance on the development and pathology of the CNS. Findings indicate that neuronal function in various brain regions are exacerbated with a gain or loss of function of tonic inhibition by GABA spillover. Disturbance of tonic GABAA conductance mediated by non-synaptic ambient GABA may result in brain mal-development. Therefore, various pathological states (epilepsy, motor dysfunctions, psychiatric disorders, and neurodevelopmental disorders) may be partly attributable to abnormal tonic GABAA conductances. Thus, the tone of tonic conductance and level of ambient GABA may be precisely tuned to maintain the regular function and development of the CNS. Therefore, receptor expression and factors for regulating the ambient GABA concentration are highlighted to gain a deeper understanding of pathology and therapeutic strategy for CNS diseases.
Collapse
Affiliation(s)
- Kiyoshi Egawa
- Department of Neurology, Massachusetts General Hospital Charlestown, MA, USA ; Department of Pediatrics, Hokkaido University Graduate School of Medicine Sapporo, Japan
| | | |
Collapse
|
47
|
Smith SS. α4βδ GABAA receptors and tonic inhibitory current during adolescence: effects on mood and synaptic plasticity. Front Neural Circuits 2013; 7:135. [PMID: 24027497 PMCID: PMC3759753 DOI: 10.3389/fncir.2013.00135] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 07/28/2013] [Indexed: 11/13/2022] Open
Abstract
The onset of puberty is associated with alterations in mood as well as changes in cognitive function, which can be more pronounced in females. Puberty onset in female mice is associated with increased expression of α4βδ γ-amino-butyric acid-A (GABAA) receptors (GABARs) in CA1 hippocampus. These receptors, which normally have low expression in this central nervous system (CNS) site, emerge along the apical dendrites as well as on the dendritic spines of pyramidal neurons, adjacent to excitatory synapses where they underlie a tonic inhibition that shunts excitatory current and impairs activation of N-methyl-D-aspartate (NMDA) receptors, the trigger for synaptic plasticity. As would be expected, α4βδ expression at puberty also prevents long-term potentiation (LTP), an in vitro model of learning which is a function of network activity, induced by theta burst stimulation of the Schaffer collaterals to the CA1 hippocampus. The expression of these receptors also impairs spatial learning in a hippocampal-dependent task. These impairments are not seen in δ knock-out (-/-) mice, implicating α4βδ GABARs. α4βδ GABARs are also a sensitive target for steroids such as THP ([allo]pregnanolone or 3α-OH-5α[β]-pregnan-20-one), which are dependent upon the polarity of GABAergic current. It is well-known that THP can increase depolarizing current gated by α4βδ GABARs, but more recent data suggest that THP can reduce hyperpolarizing current by accelerating receptor desensitization. At puberty, THP reduces the hyperpolarizing GABAergic current, which removes the shunting inhibition that impairs synaptic plasticity and learning at this time. However, THP, a stress steroid, also increases anxiety, via its action at α4βδ GABARs because it is not seen in δ(-/-) mice. These findings will be discussed as well as their relevance to changes in mood and cognition at puberty, which can be a critical period for certain types of learning and when anxiety disorders and mood swings can emerge.
Collapse
Affiliation(s)
- Sheryl S Smith
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center Brooklyn, NY 11203, USA.
| |
Collapse
|
48
|
Fatemi SH, Folsom TD, Rooney RJ, Thuras PD. Expression of GABAA α2-, β1- and ε-receptors are altered significantly in the lateral cerebellum of subjects with schizophrenia, major depression and bipolar disorder. Transl Psychiatry 2013; 3:e303. [PMID: 24022508 PMCID: PMC3784760 DOI: 10.1038/tp.2013.64] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 06/17/2013] [Accepted: 07/10/2013] [Indexed: 12/15/2022] Open
Abstract
There is abundant evidence that dysfunction of the γ-aminobutyric acid (GABA)ergic signaling system is implicated in the pathology of schizophrenia and mood disorders. Less is known about the alterations in protein expression of GABA receptor subunits in brains of subjects with schizophrenia and mood disorders. We have previously demonstrated reduced expression of GABA(B) receptor subunits 1 and 2 (GABBR1 and GABBR2) in the lateral cerebella of subjects with schizophrenia, bipolar disorder and major depressive disorder. In the current study, we have expanded these studies to examine the mRNA and protein expression of 12 GABA(A) subunit proteins (α1, α2, α3, α5, α6, β1, β2, β3, δ, ε, γ2 and γ3) in the lateral cerebella from the same set of subjects with schizophrenia (N=9-15), bipolar disorder (N=10-15) and major depression (N=12-15) versus healthy controls (N=10-15). We found significant group effects for protein levels of the α2-, β1- and ε-subunits across treatment groups. We also found a significant group effect for mRNA levels of the α1-subunit across treatment groups. New avenues for treatment, such as the use of neurosteroids to promote GABA modulation, could potentially ameliorate GABAergic dysfunction in these disorders.
Collapse
Affiliation(s)
- S H Fatemi
- Division of Neuroscience Research, Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, USA,Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, USA,Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN, USA,Division of Neuroscience Research, Department of Psychiatry, University of Minnesota Medical School, 420 Delaware St SE, MMC 392, Minneapolis, MN 55455, USA. E-mail:
| | - T D Folsom
- Division of Neuroscience Research, Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, USA
| | | | - P D Thuras
- Department of Psychiatry, VA Medical Center Minneapolis, MN, USA
| |
Collapse
|
49
|
Lee JS, Lee JD, Park HJ, Oh MK, Chun JW, Kim SJ, Kim E, Kim JJ. Is the GABA System Related to the Social Competence Improvement Effect of Aripiprazole? An (18)F-Fluoroflumazenil PET Study. Psychiatry Investig 2013; 10:75-80. [PMID: 23482902 PMCID: PMC3590434 DOI: 10.4306/pi.2013.10.1.75] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 07/24/2012] [Accepted: 08/03/2012] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Patients with schizophrenia who are treated with aripiprazole experience some benefits including an improvement of social competence, but the underlying mechanism of this improvement has not been investigated yet. This study aimed to provide preliminary evidence that the GABA system may be involved in the effect of aripiprazole on social competence. METHODS Seventeen outpatients with schizophrenia (9 taking aripiprazole and 8 taking risperidone) and 18 healthy controls underwent (18)F-fluoroflumazenil PET, and GABAA receptor binding potential was compared between the three groups. RESULTS Voxelwise one-way ANOVA showed that GABAA receptor binding potentials in the right medial prefrontal cortex (p=0.04) and right dorsolateral prefrontal cortex (p=0.02) were significantly lower in the aripiprazole group than the risperidone group, and those in the left frontopolar cortex (p=0.03) and right premotor cortex (p=0.02) were significantly lower in the aripiprazole group than the risperidone and control groups. CONCLUSION Our results suggest that aripiprazole administration results in increased GABA transmission in the prefrontal regions, and that these increases may be a neural basis of aripiprazole's clinical benefits on an improvement of social competence.
Collapse
Affiliation(s)
- Jung Suk Lee
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Psychiatry, Bundang Jesaeng Hospital, Seongnam, Republic of Korea
| | - Jong Doo Lee
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hae-Jeong Park
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Maeng-Keun Oh
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ji Won Chun
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Se-Joo Kim
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Psychiatry, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eosu Kim
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Psychiatry, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae-Jin Kim
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Psychiatry, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
50
|
Gavin DP, Akbarian S. Epigenetic and post-transcriptional dysregulation of gene expression in schizophrenia and related disease. Neurobiol Dis 2012; 46:255-62. [DOI: 10.1016/j.nbd.2011.12.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 11/10/2011] [Accepted: 12/04/2011] [Indexed: 12/22/2022] Open
|