1
|
Wang YJ, Zan GY, Xu C, Li XP, Shu X, Yao SY, Xu XS, Qiu X, Chen Y, Jin K, Zhou QX, Ye JY, Wang Y, Xu L, Chen Z, Liu JG. The claustrum-prelimbic cortex circuit through dynorphin/κ-opioid receptor signaling underlies depression-like behaviors associated with social stress etiology. Nat Commun 2023; 14:7903. [PMID: 38036497 PMCID: PMC10689794 DOI: 10.1038/s41467-023-43636-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 11/15/2023] [Indexed: 12/02/2023] Open
Abstract
Ample evidence has suggested the stress etiology of depression, but the underlying mechanism is not fully understood yet. Here, we report that chronic social defeat stress (CSDS) attenuates the excitatory output of the claustrum (CLA) to the prelimbic cortex (PL) through the dynorphin/κ-opioid receptor (KOR) signaling, being critical for depression-related behaviors in male mice. The CSDS preferentially impairs the excitatory output from the CLA onto the parvalbumin (PV) of the PL, leading to PL micronetwork dysfunction by disinhibiting pyramidal neurons (PNs). Optogenetic activation or inhibition of this circuit suppresses or promotes depressive-like behaviors, which is reversed by chemogenetic inhibition or activation of the PV neurons. Notably, manipulating the dynorphin/KOR signaling in the CLA-PL projecting terminals controls depressive-like behaviors that is suppressed or promoted by optogenetic activation or inhibition of CLA-PL circuit. Thus, this study reveals both mechanism of the stress etiology of depression and possibly therapeutic interventions by targeting CLA-PL circuit.
Collapse
Affiliation(s)
- Yu-Jun Wang
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19 A Yuquan Road, 100049, Beijing, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China
| | - Gui-Ying Zan
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19 A Yuquan Road, 100049, Beijing, China
| | - Cenglin Xu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xue-Ping Li
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Xuelian Shu
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19 A Yuquan Road, 100049, Beijing, China
| | - Song-Yu Yao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiao-Shan Xu
- Laboratory of Learning and Memory, Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Kunming, 650223, China
| | - Xiaoyun Qiu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yexiang Chen
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurobiology of Zhejiang Province, Hangzhou, 310053, China
| | - Kai Jin
- Laboratory of Learning and Memory, Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Kunming, 650223, China
| | - Qi-Xin Zhou
- Laboratory of Learning and Memory, Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Kunming, 650223, China
| | - Jia-Yu Ye
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurobiology of Zhejiang Province, Hangzhou, 310053, China
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lin Xu
- Laboratory of Learning and Memory, Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Kunming, 650223, China.
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Jing-Gen Liu
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, No. 19 A Yuquan Road, 100049, Beijing, China.
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurobiology of Zhejiang Province, Hangzhou, 310053, China.
| |
Collapse
|
2
|
Meng L, Gui S, Ouyang Z, Wu Y, Zhuang Y, Pang Q, Fan R. Low-dose bisphenols exposure sex-specifically induces neurodevelopmental toxicity in juvenile rats and the antagonism of EGCG. JOURNAL OF HAZARDOUS MATERIALS 2023; 459:132074. [PMID: 37473573 DOI: 10.1016/j.jhazmat.2023.132074] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 07/10/2023] [Accepted: 07/14/2023] [Indexed: 07/22/2023]
Abstract
Bisphenols (BPs) can negatively affect neurobehaviors in rats, whereas the mechanism remains unclear. Here, the mechanism of BPs-induced neurodevelopmental toxicity and its effective detoxification measures were investigated in vitro and in vivo. In in vitro experiments, primary hippocampal neurons from neonatal rats of different genders were treated with bisphenol A (BPA), bisphenol S (BPS) and bisphenol B (BPB) at 1 nM-100 μM, epigallocatechin gallate (EGCG) and G15, an antagonist of G protein-coupled estrogen receptor (GPER) for 7 d. Results indicated that BPs affected neuronal morphogenesis, impaired GABA synthesis and Glu/GABA homeostasis. Neuronal morphogenetic damage induced by low-doses BPA may be mediated by GPER. Neurotoxicity of BPS is weaker than BPA and BPB. In in vivo studies, exposure to BPA (0.5 μg/kg·bw/day) on PND 10-40 caused oxidative stress and inflammation in rat hippocampus, disrupted neuronal morphogenesis and neurotransmitter homeostasis, ultimately impaired spatial memory of rats. Males are more sensitive to BPA exposure than females. Both in vivo and in vitro studies indicated that EGCG, a phytoestrogen, can alleviate BPA-induced neurotoxicity. Taken together, low-doses BPA exposure sex-specifically disrupted neurodevelopment and further impaired learning and memory ability in rats, which may be mediated by GPER. Promisingly, EGCG effectively mitigated the BPA-induced neurodevelopmental toxicity.
Collapse
Affiliation(s)
- Lingxue Meng
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Shiheng Gui
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Zedong Ouyang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Yajuan Wu
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Youling Zhuang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Qihua Pang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Ruifang Fan
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
3
|
Barch DM, Boudewyn MA, Carter CC, Erickson M, Frank MJ, Gold JM, Luck SJ, MacDonald AW, Ragland JD, Ranganath C, Silverstein SM, Yonelinas A. Cognitive [Computational] Neuroscience Test Reliability and Clinical Applications for Serious Mental Illness (CNTRaCS) Consortium: Progress and Future Directions. Curr Top Behav Neurosci 2022; 63:19-60. [PMID: 36173600 DOI: 10.1007/7854_2022_391] [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] [Indexed: 11/25/2022]
Abstract
The development of treatments for impaired cognition in schizophrenia has been characterized as the most important challenge facing psychiatry at the beginning of the twenty-first century. The Cognitive Neuroscience Treatment Research to Improve Cognition in Schizophrenia (CNTRICS) project was designed to build on the potential benefits of using tasks and tools from cognitive neuroscience to better understanding and treat cognitive impairments in psychosis. These benefits include: (1) the use of fine-grained tasks that measure discrete cognitive processes; (2) the ability to design tasks that distinguish between specific cognitive domain deficits and poor performance due to generalized deficits resulting from sedation, low motivation, poor test taking skills, etc.; and (3) the ability to link cognitive deficits to specific neural systems, using animal models, neuropsychology, and functional imaging. CNTRICS convened a series of meetings to identify paradigms from cognitive neuroscience that maximize these benefits and identified the steps need for translation into use in clinical populations. The Cognitive Neuroscience Test Reliability and Clinical Applications for Schizophrenia (CNTRaCS) Consortium was developed to help carry out these steps. CNTRaCS consists of investigators at five different sites across the country with diverse expertise relevant to a wide range of the cognitive systems identified as critical as part of CNTRICs. This work reports on the progress and current directions in the evaluation and optimization carried out by CNTRaCS of the tasks identified as part of the original CNTRICs process, as well as subsequent extensions into the Positive Valence systems domain of Research Domain Criteria (RDoC). We also describe the current focus of CNTRaCS, which involves taking a computational psychiatry approach to measuring cognitive and motivational function across the spectrum of psychosis. Specifically, the current iteration of CNTRaCS is using computational modeling to isolate parameters reflecting potentially more specific cognitive and visual processes that may provide greater interpretability in understanding shared and distinct impairments across psychiatric disorders.
Collapse
Affiliation(s)
- Deanna M Barch
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA.
| | | | | | | | | | - James M Gold
- Maryland Psychiatric Research Center, Baltimore, MD, USA
| | | | | | | | | | | | | |
Collapse
|
4
|
Iwamura Y, Nakayama T, Matsumoto A, Ogi Y, Yamaguchi M, Kobayashi A, Matsumoto K, Katsura Y, Konoike N, Nakamura K, Ikeda K. Effect of dopamine receptor-related compounds on naive common marmosets for auditory steady state response. J Neurophysiol 2022; 128:229-238. [PMID: 35583977 DOI: 10.1152/jn.00147.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Abnormalities of auditory steady state responses (ASSR) and the effects of antipsychotic drugs on ASSR have been investigated in patients with schizophrenia. It is presumed that effects of drugs do not directly reflect on ASSR, because of ASSR abnormalities associated with schizophrenia. Therefore, to investigate the direct effect of drugs on ASSR, we established an ASSR evaluation system for common marmosets in a naïve state. Dopamine D1 receptor stimulation (SKF-81297, 2 mg/kg, intraperitoneal) significantly increased evoked power (EP) at 40 Hz. The phase locking factor (PLF) was increased significantly at 20, 30, 40, and 80 Hz. However, the administration of a dopamine D1 receptor antagonist (SCH-39166, 0.3 mg/kg intraperitoneal) resulted in a significant decrease in EP and PLF at 30 Hz. Dopamine D2 receptor stimulation (quinpirole, 1 mg/kg, intramuscular) tended to increase EP and induced power (IP) at all frequencies, and a significant difference was observed at 30 Hz IP. There was no change in PLF at all frequencies. In addition, dopamine D2 receptor blockade (raclopride, 3 mg/kg, intraperitoneal) reduced EP and PLF at 30 Hz. Subcutaneous administration of the serotonin dopamine antagonist, risperidone (0.3 mg/kg), tended to increase IP and decrease PLF, but not significantly. Taken together, it is possible to compare the differences in the mode of action of drugs on ASSR using naïve non-human primates.
Collapse
Affiliation(s)
- Yoshihiro Iwamura
- Platform Technology Research Unit, Drug Research Division, Sumitomo Pharma, Co., Osaka, Japan
| | - Tatsuo Nakayama
- Platform Technology Research Unit, Drug Research Division, Sumitomo Pharma, Co., Osaka, Japan
| | - Atsushi Matsumoto
- Platform Technology Research Unit, Drug Research Division, Sumitomo Pharma, Co., Osaka, Japan
| | - Yuji Ogi
- Platform Technology Research Unit, Drug Research Division, Sumitomo Pharma, Co., Osaka, Japan
| | - Masataka Yamaguchi
- Platform Technology Research Unit, Drug Research Division, Sumitomo Pharma, Co., Osaka, Japan
| | - Atsushi Kobayashi
- Platform Technology Research Unit, Drug Research Division, Sumitomo Pharma, Co., Osaka, Japan
| | - Kenji Matsumoto
- Platform Technology Research Unit, Drug Research Division, Sumitomo Pharma, Co., Osaka, Japan
| | - Yasunori Katsura
- Platform Technology Research Unit, Drug Research Division, Sumitomo Pharma, Co., Osaka, Japan
| | - Naho Konoike
- Cognitive Neuroscience Section, Primate Research Institute, Kyoto University, Aichi, Japan
| | - Katsuki Nakamura
- Cognitive Neuroscience Section, Primate Research Institute, Kyoto University, Aichi, Japan
| | - Kazuhito Ikeda
- Platform Technology Research Unit, Drug Research Division, Sumitomo Pharma, Co., Osaka, Japan
| |
Collapse
|
5
|
Vines L, Sotelo D, Johnson A, Dennis E, Manza P, Volkow ND, Wang GJ. Ketamine use disorder: preclinical, clinical, and neuroimaging evidence to support proposed mechanisms of actions. INTELLIGENT MEDICINE 2022; 2:61-68. [PMID: 35783539 PMCID: PMC9249268 DOI: 10.1016/j.imed.2022.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Ketamine, a noncompetitive NMDA receptor antagonist, has been exclusively used as an anesthetic in medicine and has led to new insights into the pathophysiology of neuropsychiatric disorders. Clinical studies have shown that low subanesthetic doses of ketamine produce antidepressant effects for individuals with depression. However, its use as a treatment for psychiatric disorders has been limited due to its reinforcing effects and high potential for diversion and misuse. Preclinical studies have focused on understanding the molecular mechanisms underlying ketamine's antidepressant effects, but a precise mechanism had yet to be elucidated. Here we review different hypotheses for ketamine's mechanism of action including the direct inhibition and disinhibition of NMDA receptors, AMPAR activation, and heightened activation of monoaminergic systems. The proposed mechanisms are not mutually exclusive, and their combined influence may exert the observed structural and functional neural impairments. Long term use of ketamine induces brain structural, functional impairments, and neurodevelopmental effects in both rodents and humans. Its misuse has increased rapidly in the past 20 years and is one of the most common addictive drugs used in Asia. The proposed mechanisms of action and supporting neuroimaging data allow for the development of tools to identify 'biotypes' of ketamine use disorder (KUD) using machine learning approaches, which could inform intervention and treatment.
Collapse
Affiliation(s)
| | | | - Allison Johnson
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States
| | - Evan Dennis
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States
| | - Peter Manza
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States
| | - Nora D. Volkow
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States
| | - Gene-Jack Wang
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
6
|
Maksymetz J, Byun NE, Luessen DJ, Li B, Barry RL, Gore JC, Niswender CM, Lindsley CW, Joffe ME, Conn PJ. mGlu 1 potentiation enhances prelimbic somatostatin interneuron activity to rescue schizophrenia-like physiological and cognitive deficits. Cell Rep 2021; 37:109950. [PMID: 34731619 PMCID: PMC8628371 DOI: 10.1016/j.celrep.2021.109950] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 08/09/2021] [Accepted: 10/14/2021] [Indexed: 01/03/2023] Open
Abstract
Evidence for prefrontal cortical (PFC) GABAergic dysfunction is one of the most consistent findings in schizophrenia and may contribute to cognitive deficits. Recent studies suggest that the mGlu1 subtype of metabotropic glutamate receptor regulates cortical inhibition; however, understanding the mechanisms through which mGlu1 positive allosteric modulators (PAMs) regulate PFC microcircuit function and cognition is essential for advancing these potential therapeutics toward the clinic. We report a series of electrophysiology, optogenetic, pharmacological magnetic resonance imaging, and animal behavior studies demonstrating that activation of mGlu1 receptors increases inhibitory transmission in the prelimbic PFC by selective excitation of somatostatin-expressing interneurons (SST-INs). An mGlu1 PAM reverses cortical hyperactivity and concomitant cognitive deficits induced by N-methyl-d-aspartate (NMDA) receptor antagonists. Using in vivo optogenetics, we show that prelimbic SST-INs are necessary for mGlu1 PAM efficacy. Collectively, these findings suggest that mGlu1 PAMs could reverse cortical GABAergic deficits and exhibit efficacy in treating cognitive dysfunction in schizophrenia.
Collapse
Affiliation(s)
- James Maksymetz
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Nellie E Byun
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Deborah J Luessen
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Brianna Li
- Vanderbilt University, Nashville, TN 37232, USA
| | - Robert L Barry
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Radiology & Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - John C Gore
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Radiology & Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Max E Joffe
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA; Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
7
|
Noronha C, Ribeiro AS, Taipa R, Castro DS, Reis J, Faria C, Paredes J. Cadherin Expression and EMT: A Focus on Gliomas. Biomedicines 2021; 9:biomedicines9101328. [PMID: 34680444 PMCID: PMC8533397 DOI: 10.3390/biomedicines9101328] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/17/2021] [Accepted: 09/19/2021] [Indexed: 12/13/2022] Open
Abstract
Cadherins are calcium-binding proteins with a pivotal role in cell adhesion and tissue homeostasis. The cadherin-dependent mechanisms of cell adhesion and migration are exploited by cancer cells, contributing to tumor invasiveness and dissemination. In particular, cadherin switch is a hallmark of epithelial to mesenchymal transition, a complex development process vastly described in the progression of most epithelial cancers. This is characterized by drastic changes in cell polarity, adhesion, and motility, which lead from an E-cadherin positive differentiated epithelial state into a dedifferentiated mesenchymal-like state, prone to metastization and defined by N-cadherin expression. Although vastly explored in epithelial cancers, how these mechanisms contribute to the pathogenesis of other non-epithelial tumor types is poorly understood. Herein, the current knowledge on cadherin expression in normal development in parallel to tumor pathogenesis is reviewed, focusing on epithelial to mesenchymal transition. Emphasis is taken in the unascertained cadherin expression in CNS tumors, particularly in gliomas, where the potential contribution of an epithelial-to-mesenchymal-like process to glioma genesis and how this may be associated with changes in cadherin expression is discussed.
Collapse
Affiliation(s)
- Carolina Noronha
- Neurosurgery Department, Hospital de Santo António, Centro Hospitalar Universitario do Porto, 4099-001 Porto, Portugal; (C.N.); (J.R.)
- Cancer Metastasis Group, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Ana Sofia Ribeiro
- Cancer Metastasis Group, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
| | - Ricardo Taipa
- Neuropathology Unit, Hospital de Santo António, Centro Hospitalar Universitario do Porto, 4099-001 Porto, Portugal;
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Diogo S. Castro
- Stem Cells & Neurogenesis Group, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
| | - Joaquim Reis
- Neurosurgery Department, Hospital de Santo António, Centro Hospitalar Universitario do Porto, 4099-001 Porto, Portugal; (C.N.); (J.R.)
- Anatomy Department, Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Cláudia Faria
- Neurosurgery Department, Hospital de Santa Maria, Centro Hospitalar Universitario Lisboa Norte, 1649-028 Lisboa, Portugal;
- IMM—Instituto de Medicina Molecular Joao Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Joana Paredes
- Cancer Metastasis Group, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Correspondence:
| |
Collapse
|
8
|
Jiménez-Balado J, Eich TS. GABAergic dysfunction, neural network hyperactivity and memory impairments in human aging and Alzheimer's disease. Semin Cell Dev Biol 2021; 116:146-159. [PMID: 33573856 PMCID: PMC8292162 DOI: 10.1016/j.semcdb.2021.01.005] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/25/2021] [Accepted: 01/30/2021] [Indexed: 02/07/2023]
Abstract
In this review, we focus on the potential role of the γ-aminobutyric acidergic (GABAergic) system in age-related episodic memory impairments in humans, with a particular focus on Alzheimer's disease (AD). Well-established animal models have shown that GABA plays a central role in regulating and synchronizing neuronal signaling in the hippocampus, a brain area critical for episodic memory that undergoes early and significant morphologic and functional changes in the course of AD. Neuroimaging research in humans has documented hyperactivity in the hippocampus and losses of resting state functional connectivity in the Default Mode Network, a network that itself prominently includes the hippocampus-presaging episodic memory decline in individuals at-risk for AD. Apolipoprotein ε4, the highest genetic risk factor for AD, is associated with GABAergic dysfunction in animal models, and episodic memory impairments in humans. In combination, these findings suggest that GABA may be the linchpin in a complex system of factors that eventually leads to the principal clinical hallmark of AD: episodic memory loss. Here, we will review the current state of literature supporting this hypothesis. First, we will focus on the molecular and cellular basis of the GABAergic system and its role in memory and cognition. Next, we report the evidence of GABA dysregulations in AD and normal aging, both in animal models and human studies. Finally, we outline a model of GABAergic dysfunction based on the results of functional neuroimaging studies in humans, which have shown hippocampal hyperactivity to episodic memory tasks concurrent with and even preceding AD diagnosis, along with factors that may modulate this association.
Collapse
Affiliation(s)
- Joan Jiménez-Balado
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Teal S Eich
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
9
|
Zhang W, Xiong BR, Zhang LQ, Huang X, Yuan X, Tian YK, Tian XB. The Role of the GABAergic System in Diseases of the Central Nervous System. Neuroscience 2021; 470:88-99. [PMID: 34242730 DOI: 10.1016/j.neuroscience.2021.06.037] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 12/20/2022]
Abstract
It is well known that the central nervous system (CNS) is a complex neuronal network and its function depends on the balance between excitatory and inhibitory neurons. Disruption of the excitatory/inhibitory (E/I) balance is the main cause for the majority of the CNS diseases. In this review, we will discuss roles of the inhibitory system in the CNS diseases. The GABAergic system as the main inhibitory system, is essential for the appropriate functioning of the CNS, especially as it is engaged in the formation of learning and memory. Many researchers have reported that the GABAergic system is involved in regulating synaptic plasticity, cognition and long-term potentiation. Some clinical manifestations (such as cognitive dysfunctions, attention deficits, etc.) have also been shown to emerge after abnormalities in the GABAergic system accompanied with concomitant diseases, that include Alzheimer's disease (AD), Parkinson's disease (PD), Autism spectrum disorder (ASD), Schizophrenia, etc. The GABAergic system consists of GABA, GABA transporters, GABAergic receptors and GABAergic neurons. Changes in any of these components may contribute to the dysfunctions of the CNS. In this review, we will synthesize studies which demonstrate how the GABAergic system participates in the pathogenesis of the CNS disorders, which may provide a new idea that might be used to treat the CNS diseases.
Collapse
Affiliation(s)
- Wen Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China
| | - Bing-Rui Xiong
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, 430071 Wuhan, Hubei, China
| | - Long-Qing Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China
| | - Xian Huang
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China
| | - Xiaoman Yuan
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China
| | - Yu-Ke Tian
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China
| | - Xue-Bi Tian
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China.
| |
Collapse
|
10
|
Cano JC, Huang W, Fénelon K. The amygdala modulates prepulse inhibition of the auditory startle reflex through excitatory inputs to the caudal pontine reticular nucleus. BMC Biol 2021; 19:116. [PMID: 34082731 PMCID: PMC8176709 DOI: 10.1186/s12915-021-01050-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 05/12/2021] [Indexed: 01/20/2023] Open
Abstract
Background Sensorimotor gating is a fundamental pre-attentive process that is defined as the inhibition of a motor response by a sensory event. Sensorimotor gating, commonly measured using the prepulse inhibition (PPI) of the auditory startle reflex task, is impaired in patients suffering from various neurological and psychiatric disorders. PPI deficits are a hallmark of schizophrenia, and they are often associated with attention and other cognitive impairments. Although the reversal of PPI deficits in animal models is widely used in pre-clinical research for antipsychotic drug screening, the neurotransmitter systems and synaptic mechanisms underlying PPI are still not resolved, even under physiological conditions. Recent evidence ruled out the longstanding hypothesis that PPI is mediated by midbrain cholinergic inputs to the caudal pontine reticular nucleus (PnC). Instead, glutamatergic, glycinergic, and GABAergic inhibitory mechanisms are now suggested to be crucial for PPI, at the PnC level. Since amygdalar dysfunctions alter PPI and are common to pathologies displaying sensorimotor gating deficits, the present study was designed to test that direct projections to the PnC originating from the amygdala contribute to PPI. Results Using wild type and transgenic mice expressing eGFP under the control of the glycine transporter type 2 promoter (GlyT2-eGFP mice), we first employed tract-tracing, morphological reconstructions, and immunohistochemical analyses to demonstrate that the central nucleus of the amygdala (CeA) sends glutamatergic inputs lateroventrally to PnC neurons, including GlyT2+ cells. Then, we showed the contribution of the CeA-PnC excitatory synapses to PPI in vivo by demonstrating that optogenetic inhibition of this connection decreases PPI, and optogenetic activation induces partial PPI. Finally, in GlyT2-Cre mice, whole-cell recordings of GlyT2+ PnC neurons in vitro paired with optogenetic stimulation of CeA fibers, as well as photo-inhibition of GlyT2+ PnC neurons in vivo, allowed us to implicate GlyT2+ neurons in the PPI pathway. Conclusions Our results uncover a feedforward inhibitory mechanism within the brainstem startle circuit by which amygdalar glutamatergic inputs and GlyT2+ PnC neurons contribute to PPI. We are providing new insights to the clinically relevant theoretical construct of PPI, which is disrupted in various neuropsychiatric and neurological diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01050-z.
Collapse
Affiliation(s)
- Jose Carlos Cano
- Department of Biological Sciences, University of Texas at El Paso, 500 West University Avenue, El Paso, TX, 79912, USA
| | - Wanyun Huang
- Biology Department, University of Massachusetts Amherst, Life Science Laboratories, 240 Thatcher Road, Amherst, MA, 01002, USA
| | - Karine Fénelon
- Biology Department, University of Massachusetts Amherst, Life Science Laboratories, 240 Thatcher Road, Amherst, MA, 01002, USA.
| |
Collapse
|
11
|
Jami SA, Cameron S, Wong JM, Daly ER, McAllister AK, Gray JA. Increased excitation-inhibition balance and loss of GABAergic synapses in the serine racemase knockout model of NMDA receptor hypofunction. J Neurophysiol 2021; 126:11-27. [PMID: 34038186 DOI: 10.1152/jn.00661.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
There is substantial evidence that both N-methyl-D-aspartate receptor (NMDAR) hypofunction and dysfunction of GABAergic neurotransmission contribute to schizophrenia, though the relationship between these pathophysiological processes remains largely unknown. Although models using cell-type-specific genetic deletion of NMDARs have been informative, they display overly pronounced phenotypes extending beyond those of schizophrenia. Here, we used the serine racemase knockout (SRKO) mice, a model of reduced NMDAR activity rather than complete receptor elimination, to examine the link between NMDAR hypofunction and decreased GABAergic inhibition. The SRKO mice, in which there is a >90% reduction in the NMDAR coagonist d-serine, exhibit many of the neurochemical and behavioral abnormalities observed in schizophrenia. We found a significant reduction in inhibitory synapses onto CA1 pyramidal neurons in the SRKO mice. This reduction increases the excitation/inhibition balance resulting in enhanced synaptically driven neuronal excitability without changes in intrinsic excitability. Consistently, significant reductions in inhibitory synapse density in CA1 were observed by immunohistochemistry. We further show, using a single-neuron genetic deletion approach, that the loss of GABAergic synapses onto pyramidal neurons observed in the SRKO mice is driven in a cell-autonomous manner following the deletion of SR in individual CA1 pyramidal cells. These results support a model whereby NMDAR hypofunction in pyramidal cells disrupts GABAergic synapses leading to disrupted feedback inhibition and impaired neuronal synchrony.NEW & NOTEWORTHY Recently, disruption of excitation/inhibition (E/I) balance has become an area of considerable interest for psychiatric research. Here, we report a reduction in inhibition in the serine racemase knockout mouse model of schizophrenia that increases E/I balance and enhances synaptically driven neuronal excitability. This reduced inhibition was driven cell-autonomously in pyramidal cells lacking serine racemase, suggesting a novel mechanism for how chronic NMDA receptor hypofunction can disrupt information processing in schizophrenia.
Collapse
Affiliation(s)
- Shekib A Jami
- Center for Neuroscience, University of California, Davis, California
| | - Scott Cameron
- Center for Neuroscience, University of California, Davis, California
| | - Jonathan M Wong
- Center for Neuroscience, University of California, Davis, California
| | - Emily R Daly
- Center for Neuroscience, University of California, Davis, California
| | - A Kimberley McAllister
- Center for Neuroscience, University of California, Davis, California.,Department of Neurobiology, Physiology, and Behavior, University of California, Davis, California.,Department of Neurology, University of California, Davis, California
| | - John A Gray
- Center for Neuroscience, University of California, Davis, California.,Department of Neurology, University of California, Davis, California
| |
Collapse
|
12
|
Huang J, Tong J, Zhang P, Zhou Y, Cui Y, Tan S, Wang Z, Yang F, Kochunov P, Chiappelli J, Tian B, Tian L, Tan Y, Hong LE. Effects of neuroactive metabolites of the tryptophan pathway on working memory and cortical thickness in schizophrenia. Transl Psychiatry 2021; 11:198. [PMID: 33795641 PMCID: PMC8016899 DOI: 10.1038/s41398-021-01311-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 02/19/2021] [Accepted: 03/11/2021] [Indexed: 01/10/2023] Open
Abstract
A number of tryptophan metabolites known to be neuroactive have been examined for their potential associations with cognitive deficits in schizophrenia. Among these metabolites, kynurenic acid (KYNA), 5-hydroxyindole (5-HI), and quinolinic acid (QUIN) are documented in their diverse effects on α-7 nicotinic acetylcholine receptor (α7nAChR) and/or N-methyl-D-aspartate receptor (NMDAR), two of the receptor types thought to contribute to cognitive impairment in schizophrenia. In this study, serum levels of KYNA, 5-HI, and QUIN were measured in 195 patients with schizophrenia and in 70 healthy controls using liquid chromatography-tandem mass spectrometry; cognitive performance in MATRICS Consensus Cognitive Battery and cortical thickness measured by magnetic resonance imaging were obtained. Patients with schizophrenia had significantly lower serum KYNA (p < 0.001) and QUIN (p = 0.02) levels, and increased 5-HI/KYNA (p < 0.001) and QUIN/KYNA ratios (p < 0.001) compared with healthy controls. Multiple linear regression showed that working memory was positively correlated with serum 5-HI levels (t = 2.10, p = 0.04), but inversely correlated with KYNA concentrations (t = -2.01, p = 0.05) in patients. Patients with high 5-HI and low KYNA had better working memory than other subgroups (p = 0.01). Higher 5-HI levels were associated with thicker left lateral orbitofrontal cortex (t = 3.71, p = 2.94 × 10-4) in patients. The different effects of 5-HI and KYNA on working memory may appear consistent with their opposite receptor level mechanisms. Our findings appear to provide a new insight into the dynamic roles of tryptophan pathway metabolites on cognition, which may benefit novel therapeutic development that targets cognitive impairment in schizophrenia.
Collapse
Affiliation(s)
- Junchao Huang
- grid.414351.60000 0004 0530 7044Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| | - Jinghui Tong
- grid.414351.60000 0004 0530 7044Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| | - Ping Zhang
- grid.414351.60000 0004 0530 7044Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| | - Yanfang Zhou
- grid.414351.60000 0004 0530 7044Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| | - Yimin Cui
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Beijing, China
| | - Shuping Tan
- grid.414351.60000 0004 0530 7044Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| | - Zhiren Wang
- grid.414351.60000 0004 0530 7044Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| | - Fude Yang
- grid.414351.60000 0004 0530 7044Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| | - Peter Kochunov
- grid.411024.20000 0001 2175 4264Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD USA
| | - Joshua Chiappelli
- grid.411024.20000 0001 2175 4264Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD USA
| | - Baopeng Tian
- grid.414351.60000 0004 0530 7044Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China
| | - Li Tian
- grid.10939.320000 0001 0943 7661Faculty of Medicine, Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Yunlong Tan
- Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, China.
| | - L. Elliot Hong
- grid.411024.20000 0001 2175 4264Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD USA
| |
Collapse
|
13
|
Costanzi M, Cianfanelli B, Santirocchi A, Lasaponara S, Spataro P, Rossi-Arnaud C, Cestari V. Forgetting Unwanted Memories: Active Forgetting and Implications for the Development of Psychological Disorders. J Pers Med 2021; 11:jpm11040241. [PMID: 33810436 PMCID: PMC8066077 DOI: 10.3390/jpm11040241] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/23/2021] [Indexed: 11/16/2022] Open
Abstract
Intrusive memories are a common feature of many psychopathologies, and suppression-induced forgetting of unwanted memories appears as a critical ability to preserve mental health. In recent years, biological and cognitive studies converged in revealing that forgetting is due to active processes. Recent neurobiological studies provide evidence on the active role of main neurotransmitter systems in forgetting, suggesting that the brain actively works to suppress retrieval of unwanted memories. On the cognitive side, there is evidence that voluntary and involuntary processes (here termed "intentional" and "incidental" forgetting, respectively) contribute to active forgetting. In intentional forgetting, an inhibitory control mechanism suppresses awareness of unwanted memories at encoding or retrieval. In incidental forgetting, retrieval practice of some memories involuntarily suppresses the retrieval of other related memories. In this review we describe recent findings on deficits in active forgetting observed in psychopathologies, like post-traumatic stress disorder, depression, schizophrenia, and obsessive-compulsive disorder. Moreover, we report studies in which the role of neurotransmitter systems, known to be involved in the pathogenesis of mental disorders, has been investigated in active forgetting paradigms. The possibility that biological and cognitive mechanisms of active forgetting could be considered as hallmarks of the early onset of psychopathologies is also discussed.
Collapse
Affiliation(s)
- Marco Costanzi
- Department of Human Sciences, Lumsa University, 00193 Rome, Italy; (B.C.); (S.L.)
- Correspondence:
| | - Beatrice Cianfanelli
- Department of Human Sciences, Lumsa University, 00193 Rome, Italy; (B.C.); (S.L.)
| | - Alessandro Santirocchi
- Department of Psychology, Sapienza University, 00185 Rome, Italy; (A.S.); (C.R.-A.); (V.C.)
| | - Stefano Lasaponara
- Department of Human Sciences, Lumsa University, 00193 Rome, Italy; (B.C.); (S.L.)
- Department of Psychology, Sapienza University, 00185 Rome, Italy; (A.S.); (C.R.-A.); (V.C.)
| | - Pietro Spataro
- Department of Economy, Universitas Mercatorum, 00100 Rome, Italy;
| | - Clelia Rossi-Arnaud
- Department of Psychology, Sapienza University, 00185 Rome, Italy; (A.S.); (C.R.-A.); (V.C.)
| | - Vincenzo Cestari
- Department of Psychology, Sapienza University, 00185 Rome, Italy; (A.S.); (C.R.-A.); (V.C.)
| |
Collapse
|
14
|
Wang LJ, Mu LL, Ren ZX, Tang HJ, Wei YD, Wang WJ, Song PP, Zhu L, Ling Q, Gao H, Zhang L, Song X, Wei HF, Chang LX, Wei T, Wang YJ, Zhao W, Wang Y, Liu LY, Zhou YD, Zhou RD, Xu HS, Jiao DL. Predictive Role of Executive Function in the Efficacy of Intermittent Theta Burst Transcranial Magnetic Stimulation Modalities for Treating Methamphetamine Use Disorder-A Randomized Clinical Trial. Front Psychiatry 2021; 12:774192. [PMID: 34925101 PMCID: PMC8674464 DOI: 10.3389/fpsyt.2021.774192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/02/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Repetitive transcranial magnetic stimulation (rTMS) has therapeutic effects on craving in methamphetamine (METH) use disorder (MUD). The chronic abuse of METH causes impairments in executive function, and improving executive function reduces relapse and improves treatment outcomes for drug use disorder. The purpose of this study was to determine whether executive function helped predict patients' responses to rTMS treatment. Methods: This study employed intermittent theta burst stimulation (iTBS) rTMS modalities and observed their therapeutic effects on executive function and craving in MUD patients. MUD patients from an isolated Drug Rehabilitation Institute in China were chosen and randomly allocated to the iTBS group and sham-stimulation group. All participants underwent the Behavior Rating Inventory of Executive Function - Adult Version Scale (BRIEF-A) and Visual Analog Scales (VAS) measurements. Sixty-five healthy adults matched to the general condition of MUD patients were also recruited as healthy controls. Findings: Patients with MUD had significantly worse executive function. iTBS groups had better treatment effects on the MUD group than the sham-stimulation group. Further Spearman rank correlation and stepwise multivariate regression analysis revealed that reduction rates of the total score of the BRIEF-A and subscale scores of the inhibition factor and working memory factor in the iTBS group positively correlated with improvements in craving. ROC curve analysis showed that working memory (AUC = 87.4%; 95% CI = 0.220, 0.631) and GEC (AUC = 0.761%; 95% CI = 0.209, 0.659) had predictive power to iTBS therapeutic efficacy. The cutoff values are 13.393 and 59.804, respectively. Conclusions: The iTBS rTMS had a better therapeutic effect on the executive function of patients with MUD, and the improved executive function had the potential to become a predictor for the efficacy of iTBS modality for MUD treatment. Clinical Trial Registration: ClinicalTrials.gov, identifier: ChiCTR2100046954.
Collapse
Affiliation(s)
- Li-Jin Wang
- School of Mental Health, Bengbu Medical College, Bengbu, China
| | - Lin-Lin Mu
- School of Mental Health, Bengbu Medical College, Bengbu, China
| | - Zi-Xuan Ren
- School of Mental Health, Bengbu Medical College, Bengbu, China
| | - Hua-Jun Tang
- Compulsory Isolated Drug Rehabilitation Center, Bengbu, China
| | - Ya-Dong Wei
- Compulsory Isolated Drug Rehabilitation Center, Bengbu, China
| | - Wen-Juan Wang
- School of Mental Health, Bengbu Medical College, Bengbu, China
| | - Pei-Pei Song
- School of Mental Health, Bengbu Medical College, Bengbu, China
| | - Lin Zhu
- School of Mental Health, Bengbu Medical College, Bengbu, China
| | - Qiang Ling
- Compulsory Isolated Drug Rehabilitation Center, Bengbu, China
| | - He Gao
- Compulsory Isolated Drug Rehabilitation Center, Bengbu, China
| | - Lei Zhang
- School of Mental Health, Bengbu Medical College, Bengbu, China
| | - Xun Song
- School of Mental Health, Bengbu Medical College, Bengbu, China
| | - Hua-Feng Wei
- School of Mental Health, Bengbu Medical College, Bengbu, China
| | - Lei-Xin Chang
- School of Mental Health, Bengbu Medical College, Bengbu, China
| | - Tao Wei
- School of Mental Health, Bengbu Medical College, Bengbu, China
| | - Yu-Jing Wang
- School of Mental Health, Bengbu Medical College, Bengbu, China
| | - Wei Zhao
- School of Mental Health, Bengbu Medical College, Bengbu, China
| | - Yan Wang
- School of Mental Health, Bengbu Medical College, Bengbu, China
| | - Lu-Ying Liu
- School of Mental Health, Bengbu Medical College, Bengbu, China
| | - Yi-Ding Zhou
- School of Mental Health, Bengbu Medical College, Bengbu, China
| | - Rui-Dong Zhou
- School of Mental Health, Bengbu Medical College, Bengbu, China
| | - Hua-Shan Xu
- School of Mental Health, Bengbu Medical College, Bengbu, China
| | - Dong-Liang Jiao
- School of Mental Health, Bengbu Medical College, Bengbu, China
| |
Collapse
|
15
|
Linde J, Zimmer-Bensch G. DNA Methylation-Dependent Dysregulation of GABAergic Interneuron Functionality in Neuropsychiatric Diseases. Front Neurosci 2020; 14:586133. [PMID: 33041771 PMCID: PMC7525021 DOI: 10.3389/fnins.2020.586133] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 08/25/2020] [Indexed: 12/30/2022] Open
Abstract
Neuropsychiatric diseases, such as mood disorders, schizophrenia, and autism, represent multifactorial disorders, differing in causes, disease onset, severity, and symptoms. A common feature of numerous neuropsychiatric conditions are defects in the cortical inhibitory GABAergic system. The balance of excitation and inhibition is fundamental for proper and efficient information processing in the cerebral cortex. Thus, altered inhibition is suggested to account for pathological symptoms like cognitive impairments and dysfunctional multisensory integration. While it became apparent that most of these diseases have a clear genetic component, environmental influences emerged as an impact of disease manifestation, onset, and severity. Epigenetic mechanisms of transcriptional control, such as DNA methylation, are known to be responsive to external stimuli, and are suspected to be implicated in the functional impairments of GABAergic interneurons, and hence, the pathophysiology of neuropsychiatric diseases. Here, we provide an overview about the multifaceted functional implications of DNA methylation and DNA methyltransferases in cortical interneuron development and function in health and disease. Apart from the regulation of gamma-aminobutyric acid-related genes and genes relevant for interneuron development, we discuss the role of DNA methylation-dependent regulation of synaptic transmission by the modulation of endocytosis-related genes as potential pathophysiological mechanisms underlying neuropsychiatric conditions. Deciphering the hierarchy and mechanisms of changes in epigenetic signatures is crucial to develop effective strategies for treatment and prevention.
Collapse
Affiliation(s)
- Jenice Linde
- Division of Functional Epigenetics in the Animal Model, Institute for Biology II, RWTH Aachen University, Aachen, Germany.,Research Training Group 2416 MultiSenses - MultiScales, RWTH Aachen University, Aachen, Germany
| | - Geraldine Zimmer-Bensch
- Division of Functional Epigenetics in the Animal Model, Institute for Biology II, RWTH Aachen University, Aachen, Germany.,Research Training Group 2416 MultiSenses - MultiScales, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
16
|
Tonna M, Ponzi D, Palanza P, Marchesi C, Parmigiani S. Proximate and ultimate causes of ritual behavior. Behav Brain Res 2020; 393:112772. [PMID: 32544508 DOI: 10.1016/j.bbr.2020.112772] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/23/2020] [Accepted: 06/08/2020] [Indexed: 01/24/2023]
Abstract
Ritual behaviour, intended as a specific, repetitive and rigid form of action flow, appears both in social and non-social environmental contexts, representing an ubiquitous phenomenon in animal life including human individuals and cultures. The purpose of this contribution is to investigate an evolutionary continuum in proximate and ultimate causes of ritual behavior. A phylogenetic homology in proximal mechanisms can be found, based on the repetition of genetically programmed and/or epigenetically acquired action patterns of behavior. As far as its adaptive significance, ethological comparative studies show that the tendency to ritualization is driven by the unpredictability of social or ecological environmental stimuli. In this perspective, rituals may have a "homeostatic" function over unpredictable environments, as further highlighted by psychopathological compulsions. In humans, a circular loop may have occurred among ritual practices and symbolic activity to deal with a novel culturally-mediated world. However, we suggest that the compulsion to action patterns repetition, typical of all rituals, has a genetically inborn motor foundation, thus precognitive and pre-symbolic. Rooted in such phylogenetically conserved motor structure (proximate causes), the evolution of cognitive and symbolic capacities have generated the complexity of human rituals, though maintaining the original adaptive function (ultimate causes) to cope with unpredictable environments.
Collapse
Affiliation(s)
- Matteo Tonna
- Department of Mental Health, Local Health Service, Parma, Italy.
| | - Davide Ponzi
- Department of Medicine and Surgery, Neuroscience Unit, University of Parma, Italy
| | - Paola Palanza
- Department of Medicine and Surgery, Neuroscience Unit, University of Parma, Italy
| | - Carlo Marchesi
- Department of Medicine and Surgery, Neuroscience Unit, University of Parma, Italy
| | - Stefano Parmigiani
- Department of Chemistry, Life Sciences and Environmental Sustainaibility, Unit of Behavioral Biology, University of Parma, Italy
| |
Collapse
|
17
|
Blanco-Ayala T, Sathyasaikumar KV, Uys JD, Pérez-de-la-Cruz V, Pidugu LS, Schwarcz R. N-Acetylcysteine Inhibits Kynurenine Aminotransferase II. Neuroscience 2020; 444:160-169. [PMID: 32768617 DOI: 10.1016/j.neuroscience.2020.07.049] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 07/08/2020] [Accepted: 07/28/2020] [Indexed: 12/22/2022]
Abstract
The tryptophan metabolite kynurenic acid (KYNA) may play an important role in normal and abnormal cognitive processes, most likely by interfering with α7 nicotinic and NMDA receptor function. KYNA is formed from its immediate precursor kynurenine either by non-enzymatic oxidation or through irreversible transamination by kynurenine aminotransferases. In the mammalian brain, kynurenine aminotransferase II (KAT II) is the principal enzyme responsible for the neosynthesis of rapidly mobilizable KYNA, and therefore constitutes an attractive target for pro-cognitive interventions. N-acetylcysteine (NAC), a brain-penetrant drug with pro-cognitive efficacy in humans, has been proposed to exert its actions by increasing the levels of the anti-oxidant glutathione (GSH) in the brain. We report here that NAC, but not GSH, inhibits KAT II activity in brain tissue homogenates from rats and humans with IC50 values in the high micromolar to low millimolar range. With similar potency, the drug interfered with the de novo formation of KYNA in rat brain slices, and NAC was a competitive inhibitor of recombinant human KAT II (Ki: 450 μM). Furthermore, GSH failed to S-glutathionylate recombinant human KAT II treated with the dithiocarbamate drug disulfiram. Shown by microdialysis in the prefrontal cortex of rats treated with kynurenine (50 mg/kg, i.p.), peripheral administration of NAC (500 mg/kg, i.p., 120 and 60 min before the application of kynurenine) reduced KYNA neosynthesis by ∼50%. Together, these results suggest that NAC exerts its neurobiological effects at least in part by reducing cerebral KYNA formation via KAT II inhibition.
Collapse
Affiliation(s)
- T Blanco-Ayala
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - K V Sathyasaikumar
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - J D Uys
- Department of Cellular and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - V Pérez-de-la-Cruz
- Laboratorio de Neurobioquimica y Conducta, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, S.S.A. Ciudad de México, Mexico
| | - L S Pidugu
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - R Schwarcz
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
18
|
Gallo NB, Paul A, Van Aelst L. Shedding Light on Chandelier Cell Development, Connectivity, and Contribution to Neural Disorders. Trends Neurosci 2020; 43:565-580. [PMID: 32564887 PMCID: PMC7392791 DOI: 10.1016/j.tins.2020.05.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/06/2020] [Accepted: 05/07/2020] [Indexed: 02/04/2023]
Abstract
Chandelier cells (ChCs) are a unique type of GABAergic interneuron that selectively innervate the axon initial segment (AIS) of excitatory pyramidal neurons; the subcellular domain where action potentials are initiated. The proper genesis and maturation of ChCs is critical for regulating neural ensemble firing in the neocortex throughout development and adulthood. Recently, genetic and molecular studies have shed new light on the complex innerworkings of ChCs in health and disease. This review presents an overview of recent studies on the developmental origins, migratory properties, and morphology of ChCs. In addition, attention is given to newly identified molecules regulating ChC morphogenesis and connectivity as well as recent work linking ChC dysfunction to neural disorders, including schizophrenia, epilepsy, and autism spectrum disorder (ASD).
Collapse
Affiliation(s)
- Nicholas B Gallo
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 11724, USA; Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Anirban Paul
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Linda Van Aelst
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 11724, USA.
| |
Collapse
|
19
|
Yakovlev A, Manzhurtsev A, Menshchikov P, Ublinskiy M, Bozhko O, Akhadov T, Semenova N. The Effect of Visual Stimulation on GABA and Macromolecule Levels in the Human Brain in vivo. Biophysics (Nagoya-shi) 2020. [DOI: 10.1134/s0006350920010248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
20
|
|
21
|
El-Abassi R, Soliman MY, Villemarette-Pittman N, England JD. SPS: Understanding the complexity. J Neurol Sci 2019; 404:137-149. [PMID: 31377632 DOI: 10.1016/j.jns.2019.06.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 05/31/2019] [Accepted: 06/17/2019] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Stiff-person syndrome (SPS), first described in 1956 by Moersch and Woltman, is a progressive autoimmune disorder with core features of chronic fluctuating progressive truncal and limb rigidity and painful muscle spasms leading to gait difficulties, falls and an appearance that resembles tin soldiers. The syndrome is a rare, highly disabling disorder of the central nervous and frequently results in significant disability. Understanding of the etiology, clinical spectrum, diagnostic workup and therapeutic modalities for this painful and disabling disorder has vastly evolved over the past few years with more confidence in classifying and treating the patients. The purpose of this review is to increase the awareness, early detection, and treatment of this disabling disease. METHOD PubMed was searched, all date inclusive, using the following phrases: stiff person syndrome,anti-Glutamic acid decarboxylase (Anti-GAD) antibody syndrome, Progressive encephalomyelitis with rigidity and myoclonus (PERM), and Paraneoplastic Stiff Person syndrome. No filters or restrictions were used. A total of 888 articles were identified. RESULTS The results were narrowed to 190 citations after excluding non-English and duplicate reports. Clinical presentation, laboratory testing, treatment, and prognosis were categorized and summarized. DISCUSSION In this article we will discuss the epidemiology, presentation and classification. Explain the pathophysiology of SPS and the autoimmune mechanisms involved. Discuss the diagnostic approach and treatments available, as well as, the prognosis and outcome.
Collapse
Affiliation(s)
- Rima El-Abassi
- Department of Neurology, Louisiana State University School of medicine, New Orleans, USA.
| | - Michael Y Soliman
- Department of Neurology, Louisiana State University School of medicine, New Orleans, USA
| | | | - John D England
- Department of Neurology, Louisiana State University School of medicine, New Orleans, USA
| |
Collapse
|
22
|
Napoli E, Schneider A, Wang JY, Trivedi A, Carrillo NR, Tassone F, Rogawski M, Hagerman RJ, Giulivi C. Allopregnanolone Treatment Improves Plasma Metabolomic Profile Associated with GABA Metabolism in Fragile X-Associated Tremor/Ataxia Syndrome: a Pilot Study. Mol Neurobiol 2019; 56:3702-3713. [PMID: 30187385 PMCID: PMC6401336 DOI: 10.1007/s12035-018-1330-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 08/22/2018] [Indexed: 12/17/2022]
Abstract
Currently, there is no effective treatment for the fragile X-associated tremor/ataxia syndrome (FXTAS), a late-onset neurodegenerative disorder. In this pilot study, we evaluated whether allopregnanolone, a natural neurosteroid that exerts beneficial effects in neurodegenerative diseases, nervous system injury, and peripheral neuropathies, could improve lymphocytic bioenergetics and plasma pharmacometabolomics in six males with FXTAS (68 ± 3 years old; FMR1 CGG repeats 94 ± 4; FXTAS stages ranging from 3 to 5) enrolled in a 12-week open-label intervention study conducted at the University of California Davis from December 2015 through July 2016. Plasma pharmacometabolomics and lymphocytic mitochondria function were assessed at baseline (on the day of the first infusion) and at follow-up (within 48 h from the last infusion). In parallel, quantitative measurements of tremor and ataxia and neuropsychological evaluations of mental state, executive function, learning, memory, and psychological symptoms were assessed at the same time points. Allopregnanolone treatment impacted significantly GABA metabolism, oxidative stress, and some of the mitochondria-related outcomes. Notably, the magnitude of the individual metabolic response, as well as the correlation with some of the behavioral tests, was overwhelmingly carrier-specific. Based on this pilot study, allopregnanolone treatment has the potential for improving cognitive and GABA metabolism in FXTAS aligned with the concept of precision medicine.
Collapse
Affiliation(s)
- Eleonora Napoli
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA, 95616, USA
| | - Andrea Schneider
- Department of Pediatrics, School of Medicine, University of California Davis, Sacramento, CA, USA
- UC Davis Health, UC Davis MIND Institute, Sacramento, CA, USA
| | - Jun Yi Wang
- UC Davis Health, UC Davis MIND Institute, Sacramento, CA, USA
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Aditi Trivedi
- School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Nika Roa Carrillo
- School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Flora Tassone
- UC Davis Health, UC Davis MIND Institute, Sacramento, CA, USA
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Michael Rogawski
- Department of Neurology, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Randi J Hagerman
- Department of Pediatrics, School of Medicine, University of California Davis, Sacramento, CA, USA
- UC Davis Health, UC Davis MIND Institute, Sacramento, CA, USA
| | - Cecilia Giulivi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA, 95616, USA.
- UC Davis Health, UC Davis MIND Institute, Sacramento, CA, USA.
| |
Collapse
|
23
|
Tonna M, Marchesi C, Parmigiani S. The biological origins of rituals: An interdisciplinary perspective. Neurosci Biobehav Rev 2019; 98:95-106. [DOI: 10.1016/j.neubiorev.2018.12.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 12/31/2018] [Accepted: 12/31/2018] [Indexed: 12/31/2022]
|
24
|
Gilmour G, Porcelli S, Bertaina-Anglade V, Arce E, Dukart J, Hayen A, Lobo A, Lopez-Anton R, Merlo Pich E, Pemberton DJ, Havenith MN, Glennon JC, Harel BT, Dawson G, Marston H, Kozak R, Serretti A. Relating constructs of attention and working memory to social withdrawal in Alzheimer’s disease and schizophrenia: issues regarding paradigm selection. Neurosci Biobehav Rev 2019; 97:47-69. [DOI: 10.1016/j.neubiorev.2018.09.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 08/29/2018] [Accepted: 09/27/2018] [Indexed: 12/12/2022]
|
25
|
Zhang X, Green MV, Thayer SA. HIV gp120-induced neuroinflammation potentiates NMDA receptors to overcome basal suppression of inhibitory synapses by p38 MAPK. J Neurochem 2019; 148:499-515. [PMID: 30520043 DOI: 10.1111/jnc.14640] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 11/21/2018] [Accepted: 11/27/2018] [Indexed: 12/18/2022]
Abstract
HIV-associated neurocognitive disorder affects about half of HIV-infected patients. HIV impairs neuronal function through indirect mechanisms mainly mediated by inflammatory cytokines and neurotoxic viral proteins, such as the envelope protein gp120. HIV gp120 elicits a neuroinflammatory response that potentiates NMDA receptor function and induces the loss of excitatory synapses. How gp120 influences neuronal inhibition remains unknown. In this study, we expressed a green fluorescent protein (GFP)-tagged recombinant antibody-like protein that binds to the post-synaptic scaffolding protein gephyrin to label inhibitory synapses in living neurons. Treatment with 600 pM gp120 for 24 h increased the number of labeled inhibitory synapses. HIV gp120 evoked the release of interleukin-1β (IL-1β) from microglia to activate IL-1 receptors on neurons. Subsequent activation of the tyrosine kinase Src and GluN2A-containing NMDA receptors increased the number of inhibitory synapses via a process that required protein synthesis. In naïve cultures, inhibition of neuronal p38 mitogen-activated protein kinase (p38 MAPK) increased the number of inhibitory synapses suggesting that p38 MAPK produces a basal suppression of inhibitory synapses that is overcome in the presence of gp120. Direct activation of a mutant form of p38 MAPK expressed in neurons mimicked basal suppression of inhibitory synapses. This study shows for the first time that gp120-induced neuroinflammation increases the number of inhibitory synapses and that this increase overcomes a basal suppression of synaptic inhibition. Increased inhibition may be an adaptive mechanism enabling neurons to counteract excess excitatory input in order to maintain network homeostasis. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Xinwen Zhang
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Matthew V Green
- Graduate Program in Neuroscience, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Stanley A Thayer
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, USA.,Graduate Program in Neuroscience, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|
26
|
Kalemaki K, Konstantoudaki X, Tivodar S, Sidiropoulou K, Karagogeos D. Mice With Decreased Number of Interneurons Exhibit Aberrant Spontaneous and Oscillatory Activity in the Cortex. Front Neural Circuits 2018; 12:96. [PMID: 30429776 PMCID: PMC6220423 DOI: 10.3389/fncir.2018.00096] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 10/11/2018] [Indexed: 11/13/2022] Open
Abstract
GABAergic (γ-aminobutyric acid) neurons are inhibitory neurons and protect neural tissue from excessive excitation. Cortical GABAergic neurons play a pivotal role for the generation of synchronized cortical network oscillations. Imbalance between excitatory and inhibitory mechanisms underlies many neuropsychiatric disorders and is correlated with abnormalities in oscillatory activity, especially in the gamma frequency range (30–80 Hz). We investigated the functional changes in cortical network activity in response to developmentally reduced inhibition in the adult mouse barrel cortex (BC). We used a mouse model that displays ∼50% fewer cortical interneurons due to the loss of Rac1 protein from Nkx2.1/Cre-expressing cells [Rac1 conditional knockout (cKO) mice], to examine how this developmental loss of cortical interneurons may affect basal synaptic transmission, synaptic plasticity, spontaneous activity, and neuronal oscillations in the adult BC. The decrease in the number of interneurons increased basal synaptic transmission, as examined by recording field excitatory postsynaptic potentials (fEPSPs) from layer II networks in the Rac1 cKO mouse cortex, decreased long-term potentiation (LTP) in response to tetanic stimulation but did not alter the pair-pulse ratio (PPR). Furthermore, under spontaneous recording conditions, Rac1 cKO brain slices exhibit enhanced sensitivity and susceptibility to emergent spontaneous activity. We also find that this developmental decrease in the number of cortical interneurons results in local neuronal networks with alterations in neuronal oscillations, exhibiting decreased power in low frequencies (delta, theta, alpha) and gamma frequency range (30–80 Hz) with an extra aberrant peak in high gamma frequency range (80–150 Hz). Therefore, our data show that disruption in GABAergic inhibition alters synaptic properties and plasticity, while it additionally disrupts the cortical neuronal synchronization in the adult BC.
Collapse
Affiliation(s)
- Katerina Kalemaki
- School of Medicine, University of Crete, Voutes University Campus, Heraklion, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas, Heraklion, Greece
| | | | - Simona Tivodar
- School of Medicine, University of Crete, Voutes University Campus, Heraklion, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas, Heraklion, Greece
| | - Kyriaki Sidiropoulou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas, Heraklion, Greece.,Department of Biology, University of Crete, Voutes University Campus, Heraklion, Greece
| | - Domna Karagogeos
- School of Medicine, University of Crete, Voutes University Campus, Heraklion, Greece.,Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas, Heraklion, Greece
| |
Collapse
|
27
|
Symmonds M, Moran CH, Leite MI, Buckley C, Irani SR, Stephan KE, Friston KJ, Moran RJ. Ion channels in EEG: isolating channel dysfunction in NMDA receptor antibody encephalitis. Brain 2018; 141:1691-1702. [PMID: 29718139 PMCID: PMC6207885 DOI: 10.1093/brain/awy107] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 01/31/2018] [Accepted: 02/22/2018] [Indexed: 12/15/2022] Open
Abstract
See Roberts and Breakspear (doi:10.1093/brain/awy136) for a scientific commentary on this article.Neurological and psychiatric practice frequently lack diagnostic probes that can assess mechanisms of neuronal communication non-invasively in humans. In N-methyl-d-aspartate (NMDA) receptor antibody encephalitis, functional molecular assays are particularly important given the presence of NMDA antibodies in healthy populations, the multifarious symptomology and the lack of radiological signs. Recent advances in biophysical modelling techniques suggest that inferring cellular-level properties of neural circuits from macroscopic measures of brain activity is possible. Here, we estimated receptor function from EEG in patients with NMDA receptor antibody encephalitis (n = 29) as well as from encephalopathic and neurological patient controls (n = 36). We show that the autoimmune patients exhibit distinct fronto-parietal network changes from which ion channel estimates can be obtained using a microcircuit model. Specifically, a dynamic causal model of EEG data applied to spontaneous brain responses identifies a selective deficit in signalling at NMDA receptors in patients with NMDA receptor antibody encephalitis but not at other ionotropic receptors. Moreover, though these changes are observed across brain regions, these effects predominate at the NMDA receptors of excitatory neurons rather than at inhibitory interneurons. Given that EEG is a ubiquitously available clinical method, our findings suggest a unique re-purposing of EEG data as an assay of brain network dysfunction at the molecular level.
Collapse
Affiliation(s)
- Mkael Symmonds
- Division of Clinical Neurology, John Radcliffe Hospital, Oxford, UK
- Department of Clinical Neurophysiology, John Radcliffe Hospital, Oxford, UK
- Epilepsy Research Group, Nuffield Department of Clinical Neurosciences, Oxford University, John Radcliffe Hospital, Oxford, Oxford, UK
| | - Catherine H Moran
- Department of Neurological Surgery, Beaumont Hospital, Dublin, Ireland
| | - M Isabel Leite
- Division of Clinical Neurology, John Radcliffe Hospital, Oxford, UK
- Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, Oxford University, John Radcliffe Hospital, Oxford, Oxford, UK
| | - Camilla Buckley
- Division of Clinical Neurology, John Radcliffe Hospital, Oxford, UK
| | - Sarosh R Irani
- Division of Clinical Neurology, John Radcliffe Hospital, Oxford, UK
- Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, Oxford University, John Radcliffe Hospital, Oxford, Oxford, UK
| | - Klaas Enno Stephan
- Translational Neuromodeling Unit, Institute for Biomedical Engineering, University of Zurich and ETH Zurich, 6 Wilfriedstrasse, Zurich, Switzerland
| | - Karl J Friston
- Wellcome Trust Centre for Neuroimaging, University College London, 12 Queen Square, London, UK
| | - Rosalyn J Moran
- Department of Engineering Mathematics, Merchant Venturers School of Engineering, University of Bristol, 75 Woodland Rd, Bristol, UK
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
28
|
Chiou LC, Lee HJ, Ernst M, Huang WJ, Chou JF, Chen HL, Mouri A, Chen LC, Treven M, Mamiya T, Fan PC, Knutson DE, Witzigmann C, Cook J, Sieghart W, Nabeshima T. Cerebellar α 6 -subunit-containing GABA A receptors: a novel therapeutic target for disrupted prepulse inhibition in neuropsychiatric disorders. Br J Pharmacol 2018. [PMID: 29518821 DOI: 10.1111/bph.14198] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE The pathophysiological role of α6 -subunit-containing GABAA receptors, which are mainly expressed in cerebellar granule cells, remains unclear. Recently, we demonstrated that hispidulin, a flavonoid isolated from a local herb that remitted a patient's intractable motor tics, attenuated methamphetamine-induced hyperlocomotion in mice as a positive allosteric modulator (PAM) of cerebellar α6 GABAA receptors. Here, using hispidulin and a selective α6 GABAA receptor PAM, the pyrazoloquinolinone Compound 6, we revealed an unprecedented role of cerebellar α6 GABAA receptors in disrupted prepulse inhibition of the startle response (PPI), which reflects sensorimotor gating deficits manifested in several neuropsychiatric disorders. EXPERIMENTAL APPROACH PPI disruptions were induced by methamphetamine and NMDA receptor antagonists in mice. Effects of the tested compounds were measured in Xenopus oocytes expressing recombinant α6 β3 γ2S GABAA receptors. KEY RESULTS Hispidulin given i.p. or by bilateral intracerebellar (i.cb.) injection rescued PPI disruptions induced by methamphetamine, ketamine, MK-801 and phencyclidine. Intracerebellar effects of hispidulin were mimicked by Ro15-4513 and loreclezole (two α6 GABAA receptor PAMs), but not by diazepam (an α6 GABAA receptor-inactive benzodiazepine) and were antagonized by furosemide (i.cb.), an α6 GABAA receptor antagonist. Importantly, Compound 6 (i.p.) also rescued methamphetamine-induced PPI disruption, an effect prevented by furosemide (i.cb.). Both hispidulin and Compound 6 potentiated α6 β3 γ2S GABAA receptor-mediated GABA currents. CONCLUSIONS AND IMPLICATIONS Positive allosteric modulation of cerebellar α6 GABAA receptors rescued disrupted PPI by attenuating granule cell activity. α6 GABAA receptor-selective PAMs are potential medicines for treating sensorimotor gating deficits in neuropsychiatric disorders. A mechanistic hypothesis is based on evidence for cerebellar contributions to cognitive functioning including sensorimotor gating.
Collapse
Affiliation(s)
- Lih-Chu Chiou
- Graduate Institute of Pharmacology, National Taiwan University, Taipei, Taiwan,Graduate Institute of Brain and Mind Sciences, National TaiwanUniversity, Taipei, Taiwan,Graduate Institute of Acupuncture Science, Taichung, Taiwan
| | - Hsin-Jung Lee
- Graduate Institute of Pharmacology, National Taiwan University, Taipei, Taiwan
| | - Margot Ernst
- Center for Brain Research, Department of Molecular Neurosciences, Medical University Vienna, Vienna, Austria
| | - Wei-Jan Huang
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
| | - Jui-Feng Chou
- Graduate Institute of Pharmacology, National Taiwan University, Taipei, Taiwan
| | - Hon-Lie Chen
- Graduate Institute of Pharmacology, National Taiwan University, Taipei, Taiwan
| | - Akihiro Mouri
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan,Advanced Diagnostic System Research Laboratory, Graduate School of Health Sciences, Fujita Health University, Toyoake, Japan
| | - Liang-Chieh Chen
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
| | - Marco Treven
- Center for Brain Research, Department of Molecular Neurosciences, Medical University Vienna, Vienna, Austria
| | - Takayoshi Mamiya
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Pi-Chuan Fan
- Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Daniel E Knutson
- Department of Chemistry and Biochemistry, University of Wisconsin – Milwaukee, Milwaukee, WI, USA
| | - Chris Witzigmann
- Department of Chemistry and Biochemistry, University of Wisconsin – Milwaukee, Milwaukee, WI, USA
| | - James Cook
- Department of Chemistry and Biochemistry, University of Wisconsin – Milwaukee, Milwaukee, WI, USA
| | - Werner Sieghart
- Center for Brain Research, Department of Molecular Neurosciences, Medical University Vienna, Vienna, Austria
| | - Toshitaka Nabeshima
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan,Advanced Diagnostic System Research Laboratory, Graduate School of Health Sciences, Fujita Health University, Toyoake, Japan,Aino University, Ibaraki, Japan
| |
Collapse
|
29
|
Xu MY, Wong AHC. GABAergic inhibitory neurons as therapeutic targets for cognitive impairment in schizophrenia. Acta Pharmacol Sin 2018; 39:733-753. [PMID: 29565038 DOI: 10.1038/aps.2017.172] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 12/25/2017] [Indexed: 12/24/2022]
Abstract
Schizophrenia is considered primarily as a cognitive disorder. However, functional outcomes in schizophrenia are limited by the lack of effective pharmacological and psychosocial interventions for cognitive impairment. GABA (gamma-aminobutyric acid) interneurons are the main inhibitory neurons in the central nervous system (CNS), and they play a critical role in a variety of pathophysiological processes including modulation of cortical and hippocampal neural circuitry and activity, cognitive function-related neural oscillations (eg, gamma oscillations) and information integration and processing. Dysfunctional GABA interneuron activity can disrupt the excitatory/inhibitory (E/I) balance in the cortex, which could represent a core pathophysiological mechanism underlying cognitive dysfunction in schizophrenia. Recent research suggests that selective modulation of the GABAergic system is a promising intervention for the treatment of schizophrenia-associated cognitive defects. In this review, we summarized evidence from postmortem and animal studies for abnormal GABAergic neurotransmission in schizophrenia, and how altered GABA interneurons could disrupt neuronal oscillations. Next, we systemically reviewed a variety of up-to-date subtype-selective agonists, antagonists, positive and negative allosteric modulators (including dual allosteric modulators) for α5/α3/α2 GABAA and GABAB receptors, and summarized their pro-cognitive effects in animal behavioral tests and clinical trials. Finally, we also discuss various representative histone deacetylases (HDAC) inhibitors that target GABA system through epigenetic modulations, GABA prodrug and presynaptic GABA transporter inhibitors. This review provides important information on current potential GABA-associated therapies and future insights for development of more effective treatments.
Collapse
|
30
|
Cai H, Zhou X, Dougherty GG, Reddy RD, Haas GL, Montrose DM, Keshavan M, Yao JK. Pregnenolone-progesterone-allopregnanolone pathway as a potential therapeutic target in first-episode antipsychotic-naïve patients with schizophrenia. Psychoneuroendocrinology 2018; 90:43-51. [PMID: 29433072 PMCID: PMC5864547 DOI: 10.1016/j.psyneuen.2018.02.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/29/2017] [Accepted: 02/04/2018] [Indexed: 11/23/2022]
Abstract
Neurosteroids are both endogenous and exogenous steroids that rapidly alter neuronal excitability through interactions with ligand-gated ion channels and other cell surface receptors. They are originated from cholesterol and have important implications for schizophrenia (SZ) pathophysiology and treatment strategies. Specifically, pregnenolone (PREG), progesterone (PROG) and allopregnanolone (ALLO) exhibit similar psychotropic properties. Using enzyme immunoassay, we compared the neurosteroids in PREG downstream pathways in plasma between healthy controls (HC, n = 43) and first-episode antipsychotic-naïve patients with SZ (FEAN-SZ, n = 53) before antipsychotic drug (APD) treatment. Comparisons were also made particularly along PREG-PROG-ALLO pathway in the same FEAN-SZ patients across multiple time points following initiation of treatment for 12 months (m). Firstly, at baseline, levels of PREG were significantly higher and those of ALLO were lower in FEAN-SZ than in HC, whereas PROG, cortisol, dehydroepiandrosterone (DHEA) and dehydroepiandrosterone sulfate (DHEAS) were not different. Consequently, the molar ratios of ALLO/PREG and ALLO/PROG in FEAN-SZ were significantly reduced. Secondly, in response to APD at 1 month, ALLO levels in FEAN-SZ were markedly elevated, whereas PREG and PROG levels decreased. Thirdly, among FEAN-SZ, lower levels of PROG (reflecting higher conversion to ALLO) at baseline may predict better therapeutic outcome after 1 month of APD treatment. These findings point to the perturbations of the PREG-PROG-ALLO pathway early in psychosis, and further study of this pathway may inform alternative and innovative therapeutic targets for SZ.
Collapse
Affiliation(s)
- HuaLin Cai
- Medical Research Service, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA; Departments of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15216, USA; The Second Xiangya Hospital and Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, China
| | - Xiang Zhou
- Medical Research Service, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA; Departments of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - George G Dougherty
- Medical Research Service, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Ravinder D Reddy
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Gretchen L Haas
- Medical Research Service, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Debra M Montrose
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Matcheri Keshavan
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Psychiatry, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - Jeffrey K Yao
- Medical Research Service, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA; Departments of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15216, USA; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
31
|
Noda Y, Barr MS, Zomorrodi R, Cash RFH, Rajji TK, Farzan F, Chen R, George TP, Daskalakis ZJ, Blumberger DM. Reduced Short-Latency Afferent Inhibition in Prefrontal but not Motor Cortex and Its Association With Executive Function in Schizophrenia: A Combined TMS-EEG Study. Schizophr Bull 2018; 44:193-202. [PMID: 28379529 PMCID: PMC5768054 DOI: 10.1093/schbul/sbx041] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND Cholinergic dysfunction is increasingly assumed to be involved in the pathophysiology of schizophrenia. Short-latency afferent inhibition (SAI) is a transcranial magnetic stimulation (TMS) paradigm that has been shown to assay central cholinergic activity from the motor cortex (M1). Recently, we established a method to index SAI from the dorsolateral prefrontal cortex (DLPFC), an area implicated in the pathophysiology of schizophrenia. We investigated SAI in M1 and DLPFC in schizophrenia. We hypothesized that modulation of N100 on TMS-evoked potentials (TEPs) from the DLPFC would be attenuated in patients with schizophrenia compared to healthy controls. METHODS SAI was examined in 12 patients, whose age was matched to controls, using TMS combined with electroencephalography (EEG). SAI was recorded with TMS applied to left M1 (M1-SAI) and DLPFC (DLPFC-SAI). For group comparison, we used the SAI data of healthy participants in our previous study. RESULTS In patients, N100 TEP was significantly attenuated with DLPFC-SAI, whereas P180 TEP was significantly increased with M1-SAI. Between patients and controls, there were significant differences in modulation of P180 TEP by M1-SAI (t22 = -2.748, P = .012; patients > controls) and N100 TEP by DLPFC-SAI (t22 = 5.456, P < .0001; patients < controls). Further, modulation of N100 TEP by DLPFC-SAI significantly correlated with executive function (r = -.740, P = .006, N = 12). CONCLUSION Our findings suggest that DLPFC-SAI but not M1-SAI were reduced in patients with schizophrenia and this was linked to deficits in cognition. This may reflect prefrontal cholinergic deficits and represent a biomarker for cholinergic and executive dysfunction in patients with schizophrenia.
Collapse
Affiliation(s)
- Yoshihiro Noda
- Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Mera S Barr
- Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Addictions Division, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Reza Zomorrodi
- Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Robin F H Cash
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada
- Division of Brain, Imaging and Behaviour—Systems Neuroscience, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Monash Alfred Psychiatry Research Centre, Monash University Central Clinical School and The Alfred, Melbourne, Australia
| | - Tarek K Rajji
- Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Faranak Farzan
- Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Robert Chen
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada
- Division of Brain, Imaging and Behaviour—Systems Neuroscience, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Tony P George
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Addictions Division, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Zafiris J Daskalakis
- Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Daniel M Blumberger
- Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| |
Collapse
|
32
|
Castro-Zaballa S, Cavelli ML, Gonzalez J, Nardi AE, Machado S, Scorza C, Torterolo P. EEG 40 Hz Coherence Decreases in REM Sleep and Ketamine Model of Psychosis. Front Psychiatry 2018; 9:766. [PMID: 30705645 PMCID: PMC6345101 DOI: 10.3389/fpsyt.2018.00766] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 12/21/2018] [Indexed: 01/04/2023] Open
Abstract
Cognitive processes are carried out during wakefulness by means of extensive interactions between cortical and subcortical areas. In psychiatric conditions, such as psychosis, these processes are altered. Interestingly, REM sleep where most dreams occurs, shares electrophysiological, pharmacological, and neurochemical features with psychosis. Because of this fact, REM sleep is considered a natural model of psychosis. Ketamine is a non-competitive N-methyl-D-aspartate (NMDA) receptor antagonist that at sub-anesthetic dose induces psychotomimetic-like effects in humans and animals, and is employed as a pharmacological model of psychosis. Oscillations in the gamma frequency band of the electroencephalogram (EEG), mainly at about 40 Hz, have been involved in cognitive functions. Hence, the present study was conducted to analyze the EEG low gamma (30-45 Hz) band power and coherence of the cat, in natural (REM sleep) and pharmacological (sub-anesthetic doses of ketamine) models of psychosis. These results were compared with the gamma activity during alert (AW) and quiet wakefulness (QW), as well as during non-REM (NREM) sleep. Five cats were chronically prepared for polysomnographic recordings, with electrodes in different cortical areas. Basal recordings were obtained and ketamine (5, 10, and 15 mg/kg, i.m.) was administrated. Gamma activity (power and coherence) was analyzed in the abovementioned conditions. Compared to wakefulness and NREM sleep, following ketamine administration gamma coherence decreased among all cortical regions studied; the same coherence profile was observed during REM sleep. On the contrary, gamma power was relatively high under ketamine, and similar to QW and REM sleep. We conclude that functional interactions between cortical areas in the gamma frequency band decrease in both experimental models of psychosis. This uncoupling of gamma frequency activity may be involved in the cognitive features shared by dreaming and psychosis.
Collapse
Affiliation(s)
- Santiago Castro-Zaballa
- Laboratorio de Neurobiología del Sueño, Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Matías Lorenzo Cavelli
- Laboratorio de Neurobiología del Sueño, Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Joaquin Gonzalez
- Laboratorio de Neurobiología del Sueño, Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Antonio Egidio Nardi
- Laboratório de Pânico e Respiração, Instituto de Psiquiatria da Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratório de Neurociência da Atividade Física, Universidade Salgado de Oliveira, Rio de Janeiro, Brazil
| | - Sergio Machado
- Laboratório de Pânico e Respiração, Instituto de Psiquiatria da Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratório de Neurociência da Atividade Física, Universidade Salgado de Oliveira, Rio de Janeiro, Brazil.,The Intercontinental Neuroscience Research Group, Merida, Mexico
| | - Cecilia Scorza
- Departamento de Neurofarmacología Experimental, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Pablo Torterolo
- Laboratorio de Neurobiología del Sueño, Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.,The Intercontinental Neuroscience Research Group, Merida, Mexico
| |
Collapse
|
33
|
Dobberthien BJ, Tessier AG, Yahya A. Improved resolution of glutamate, glutamine and γ-aminobutyric acid with optimized point-resolved spectroscopy sequence timings for their simultaneous quantification at 9.4 T. NMR IN BIOMEDICINE 2018; 31:e3851. [PMID: 29105187 DOI: 10.1002/nbm.3851] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/15/2017] [Accepted: 09/25/2017] [Indexed: 06/07/2023]
Abstract
Glutamine (Gln), glutamate (Glu) and γ-aminobutyric acid (GABA) are relevant brain metabolites that can be measured with magnetic resonance spectroscopy (MRS). This work optimizes the point-resolved spectroscopy (PRESS) sequence echo times, TE1 and TE2 , for improved simultaneous quantification of the three metabolites at 9.4 T. Quantification was based on the proton resonances of Gln, Glu and GABA at ≈2.45, ≈2.35 and ≈2.28 ppm, respectively. Glu exhibits overlap with both Gln and GABA; in addition, the Gln peak is contaminated by signal from the strongly coupled protons of N-acetylaspartate (NAA), which resonate at about 2.49 ppm. J-coupling evolution of the protons was characterized numerically and verified experimentally. A {TE1 , TE2 } combination of {106 ms, 16 ms} minimized the NAA signal in the Gln spectral region, whilst retaining Gln, Glu and GABA peaks. The efficacy of the technique was verified on phantom solutions and on rat brain in vivo. LCModel was employed to analyze the in vivo spectra. The average T2 -corrected Gln, Glu and GABA concentrations were found to be 3.39, 11.43 and 2.20 mM, respectively, assuming a total creatine concentration of 8.5 mM. LCModel Cramér-Rao lower bounds (CRLBs) for Gln, Glu and GABA were in the ranges 14-17%, 4-6% and 16-19%, respectively. The optimal TE resulted in concentrations for Gln and GABA that agreed more closely with literature concentrations compared with concentrations obtained from short-TE spectra acquired with a {TE1 , TE2 } combination of {12 ms, 9 ms}. LCModel estimations were also evaluated with short-TE PRESS and with the optimized long TE of {106 ms, 16 ms}, using phantom solutions of known metabolite concentrations. It was shown that concentrations estimated with LCModel can be inaccurate when combined with short-TE PRESS, where there is peak overlap, even when low (<20%) CRLBs are reported.
Collapse
Affiliation(s)
| | - Anthony G Tessier
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
- Department of Medical Physics, Cross Cancer Institute, Edmonton, AB, Canada
| | - Atiyah Yahya
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
- Department of Medical Physics, Cross Cancer Institute, Edmonton, AB, Canada
| |
Collapse
|
34
|
Cortical high gamma network oscillations and connectivity: a translational index for antipsychotics to normalize aberrant neurophysiological activity. Transl Psychiatry 2017; 7:1285. [PMID: 29249806 PMCID: PMC5802558 DOI: 10.1038/s41398-017-0002-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/31/2017] [Accepted: 08/04/2017] [Indexed: 12/30/2022] Open
Abstract
Oscillatory activity in the gamma frequency range is a critical mechanism, which integrates neural networks within and across brain structures during cognitive processes. In schizophrenia, abnormalities in high gamma oscillations are ubiquitous and most likely reflect dysfunction in neuronal networks. In conscious rats, disturbed network oscillations associated with positive symptoms and cognitive deficits were modeled in different cortical areas by the dopaminergic agonist (amphetamine) and the N-methyl-D-aspartate (NMDA) receptor antagonists (PCP and MK801). Subsequently, the efficacies of marketed atypical antipsychotics (olanzapine, risperidone, and clozapine) to normalize dysfunctional oscillations and network connectivity were examined. Acute NMDA antagonists elicited aberrant synchrony in the gamma frequency oscillations. In addition, coherent slow alpha network activity was observed with MK801 and amphetamine, both of whose oscillatory rhythms were correlated with pronounced locomotor activity. All antipsychotics commonly decreased slow alpha and high gamma network oscillations in different cortical regions as well as motion behavior. In the combined treatments, antipsychotics attenuated NMDA antagonist-induced abnormalities in functional network oscillations and connectivity, whose effects on motor behavior is mechanistically related. These results suggest that pharmacologically induced disruption of cortical gamma oscillations and network connectivity in rats is a candidate model to study dysfunctional oscillatory patterns described in positive and negative symptoms of schizophrenia. The efficacy of antipsychotics to rescue cortical network oscillatory patterns is in line with the idea that glutamatergic and dopaminergic systems play a role in maintaining the integrity of cortical circuits. Thus, gamma oscillations could provide a powerful translational index to assess the integrity of neural networks and to evaluate the efficacy of drugs with potential antipsychotic properties.
Collapse
|
35
|
McGuire JL, Depasquale EA, Funk AJ, O'Donnovan SM, Hasselfeld K, Marwaha S, Hammond JH, Hartounian V, Meador-Woodruff JH, Meller J, McCullumsmith RE. Abnormalities of signal transduction networks in chronic schizophrenia. NPJ SCHIZOPHRENIA 2017; 3:30. [PMID: 28900113 PMCID: PMC5595970 DOI: 10.1038/s41537-017-0032-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 07/07/2017] [Accepted: 07/31/2017] [Indexed: 12/11/2022]
Abstract
Schizophrenia is a serious neuropsychiatric disorder characterized by disruptions of brain cell metabolism, microstructure, and neurotransmission. All of these processes require coordination of multiple kinase-mediated signaling events. We hypothesize that imbalances in kinase activity propagate through an interconnected network of intracellular signaling with potential to simultaneously contribute to many or all of the observed deficits in schizophrenia. We established a workflow distinguishing schizophrenia-altered kinases in anterior cingulate cortex using a previously published kinome array data set. We compared schizophrenia-altered kinases to haloperidol-altered kinases, and identified systems, functions, and regulators predicted using pathway analyses. We used kinase inhibitors with the kinome array to test hypotheses about imbalance in signaling and conducted preliminary studies of kinase proteins, phosphoproteins, and activity for kinases of interest. We investigated schizophrenia-associated single nucleotide polymorphisms in one of these kinases, AKT, for genotype-dependent changes in AKT protein or activity. Kinome analyses identified new kinases as well as some previously implicated in schizophrenia. These results were not explained by chronic antipsychotic treatment. Kinases identified in our analyses aligned with cytoskeletal arrangement and molecular trafficking. Of the kinases we investigated further, AKT and (unexpectedly) JNK, showed the most dysregulation in the anterior cingulate cortex of schizophrenia subjects. Changes in kinase activity did not correspond to protein or phosphoprotein levels. We also show that AKT single nucleotide polymorphism rs1130214, previously associated with schizophrenia, influenced enzyme activity but not protein or phosphoprotein levels. Our data indicate subtle changes in kinase activity and regulation across an interlinked kinase network, suggesting signaling imbalances underlie the core symptoms of schizophrenia. A study by US scientists indicates that changes in the activity of key signaling proteins may underlie core symptoms of schizophrenia. Protein kinases mediate the activation of intracellular signaling events and analyses of the kinome, the complete set of protein kinases encoded in the genome, previously revealed significant changes in phosphorylation patterns in postmortem brain tissue from patients with schizophrenia. Based on these findings, Jennifer McGuire at the University of Cincinnati and colleagues investigated the upstream regulation of these proteins. They identified both established and novel proteins associated with schizophrenia in the anterior cingulate cortex, with JNK and AKT activity being the most disrupted in schizophrenia patients. Their findings highlight how subtle changes in the activity of a small number of signaling proteins can propagate and have major consequences for mental health.
Collapse
Affiliation(s)
- Jennifer L McGuire
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA.
| | - Erica A Depasquale
- Department of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Adam J Funk
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA
| | - Sinead M O'Donnovan
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA
| | - Kathryn Hasselfeld
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA
| | - Shruti Marwaha
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - John H Hammond
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama-Birmingham, Birmingham, AL, USA
| | - Vahram Hartounian
- Psychiatry & Neuroscience, The Icahn School of Medicine at Mount Sinai, New York, NY, USA.,James J. Peters VA Medical Center, Mental Illness Research Education and Clinical Center (MIRECC), Bronx, NY, USA
| | - James H Meador-Woodruff
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama-Birmingham, Birmingham, AL, USA
| | - Jarek Meller
- Department of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Departments of Environmental Health, Electrical Engineering & Computing Systems and Biomedical Informatics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Robert E McCullumsmith
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
36
|
Radhu N, Dominguez LG, Greenwood TA, Farzan F, Semeralul MO, Richter MA, Kennedy JL, Blumberger DM, Chen R, Fitzgerald PB, Daskalakis ZJ. Investigating Cortical Inhibition in First-Degree Relatives and Probands in Schizophrenia. Sci Rep 2017; 7:43629. [PMID: 28240740 PMCID: PMC5378912 DOI: 10.1038/srep43629] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 01/26/2017] [Indexed: 01/16/2023] Open
Abstract
Deficits in GABAergic inhibitory neurotransmission are a reliable finding in schizophrenia (SCZ) patients. Previous studies have reported that unaffected first-degree relatives of patients with SCZ demonstrate neurophysiological abnormalities that are intermediate between probands and healthy controls. In this study, first-degree relatives of patients with SCZ and their related probands were investigated to assess frontal cortical inhibition. Long-interval cortical inhibition (LICI) was measured from the dorsolateral prefrontal cortex (DLPFC) using combined transcranial magnetic stimulation (TMS) and electroencephalography (EEG). The study presents an extended sample of 129 subjects (66 subjects have been previously reported): 19 patients with SCZ or schizoaffective disorder, 30 unaffected first-degree relatives of these SCZ patients, 13 obsessive-compulsive disorder (OCD) patients, 18 unaffected first-degree relatives of these OCD patients and 49 healthy subjects. In the DLPFC, cortical inhibition was significantly decreased in patients with SCZ compared to healthy subjects. First-degree relatives of patients with SCZ showed significantly more cortical inhibition than their SCZ probands. No differences were demonstrated between first-degree relatives of SCZ patients and healthy subjects. Taken together, these findings show that more studies are needed to establish an objective biological marker for potential diagnostic usage in severe psychiatric disorders.
Collapse
Affiliation(s)
- Natasha Radhu
- Novartis Pharmaceuticals Canada Inc., Dorval, Quebec, Canada
| | - Luis Garcia Dominguez
- Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada
| | - Tiffany A Greenwood
- Department of Psychiatry, School of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Faranak Farzan
- Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada
| | - Mawahib O Semeralul
- Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada
| | - Margaret A Richter
- Frederick W. Thompson Anxiety Disorders Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - James L Kennedy
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario Canada
| | - Daniel M Blumberger
- Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada
| | - Robert Chen
- Division of Neurology, Krembil Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Paul B Fitzgerald
- Monash Alfred Psychiatry Research Centre, The Alfred and Monash University Central Clinical School, Victoria, Australia
| | - Zafiris J Daskalakis
- Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
37
|
Spiros A, Roberts P, Geerts H. Semi-mechanistic computer simulation of psychotic symptoms in schizophrenia with a model of a humanized cortico-striatal-thalamocortical loop. Eur Neuropsychopharmacol 2017; 27:107-119. [PMID: 28062203 DOI: 10.1016/j.euroneuro.2016.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 11/20/2016] [Accepted: 12/24/2016] [Indexed: 12/13/2022]
Abstract
Despite new insights into the pathophysiology of schizophrenia and clinical trials with highly selective drugs, no new therapeutic breakthroughs have been identified. We present a semi-mechanistic Quantitative Systems Pharmacology (QSP) computer model of a biophysically realistic cortical-striatal-thalamo-cortical loop. The model incorporates the direct, indirect and hyperdirect pathway of the basal ganglia and CNS drug targets that modulate neuronal firing, based on preclinical data about their localization and coupling to voltage-gated ion channels. Schizophrenia pathology is introduced using quantitative human imaging data on striatal hyperdopaminergic activity and cortical dysfunction. We identified an entropy measure of neuronal firing in the thalamus, related to the bandwidth of information processing that correlates well with reported historical clinical changes on PANSS Total with antipsychotics after introduction of their pharmacology (42 drug-dose combinations, r2=0.62). This entropy measure is further validated by predicting the clinical outcome of 28 other novel stand-alone interventions, 14 of them with non-dopamine D2R pharmacology, in addition to 8 augmentation trials (correlation between actual and predicted clinical scores r2=0.61). The platform predicts that most combinations of antipsychotics have a lower efficacy over what can be achieved by either one; negative pharmacodynamical interactions are prominent for aripiprazole added to risperidone, haloperidol, quetiapine and paliperidone. The model also recapitulates the increased probability for psychotic breakdown in a supersensitive environment and the effect of ketamine in healthy volunteers. This QSP platform, combined with similar readouts for motor symptoms, negative symptoms and cognitive impairment has the potential to improve our understanding of drug effects in schizophrenia patients.
Collapse
Affiliation(s)
- Athan Spiros
- In Silico Biosciences, Berwyn, PA, United States
| | - Patrick Roberts
- In Silico Biosciences, Berwyn, PA, United States; Washington State University, Vancouver, WA, United States
| | - Hugo Geerts
- In Silico Biosciences, Berwyn, PA, United States; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
38
|
Trulioff A, Ermakov A, Malashichev Y. Primary Cilia as a Possible Link between Left-Right Asymmetry and Neurodevelopmental Diseases. Genes (Basel) 2017; 8:genes8020048. [PMID: 28125008 PMCID: PMC5333037 DOI: 10.3390/genes8020048] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/21/2016] [Accepted: 01/19/2017] [Indexed: 12/11/2022] Open
Abstract
Cilia have multiple functions in the development of the entire organism, and participate in the development and functioning of the central nervous system. In the last decade, studies have shown that they are implicated in the development of the visceral left-right asymmetry in different vertebrates. At the same time, some neuropsychiatric disorders, such as schizophrenia, autism, bipolar disorder, and dyslexia, are known to be associated with lateralization failure. In this review, we consider possible links in the mechanisms of determination of visceral asymmetry and brain lateralization, through cilia. We review the functions of seven genes associated with both cilia, and with neurodevelopmental diseases, keeping in mind their possible role in the establishment of the left-right brain asymmetry.
Collapse
Affiliation(s)
- Andrey Trulioff
- Department of Vertebrate Zoology, Faculty of Biology, Saint Petersburg State University, Universitetskaya nab., 7/9, Saint Petersburg 199034, Russia.
| | - Alexander Ermakov
- Department of Vertebrate Zoology, Faculty of Biology, Saint Petersburg State University, Universitetskaya nab., 7/9, Saint Petersburg 199034, Russia.
- Laboratory of Molecular Neurobiology, Department of Ecological Physiology, Institute of Experimental Medicine, ul. Akad. Pavlov, 12, Saint Petersburg 197376, Russia.
| | - Yegor Malashichev
- Department of Vertebrate Zoology, Faculty of Biology, Saint Petersburg State University, Universitetskaya nab., 7/9, Saint Petersburg 199034, Russia.
- Laboratory of Molecular Neurobiology, Department of Ecological Physiology, Institute of Experimental Medicine, ul. Akad. Pavlov, 12, Saint Petersburg 197376, Russia.
| |
Collapse
|
39
|
Haussleiter IS, Wandinger KP, Juckel G. A case of GABAR antibodies in schizophrenia. BMC Psychiatry 2017; 17:9. [PMID: 28068943 PMCID: PMC5223378 DOI: 10.1186/s12888-016-1157-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 12/06/2016] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND In the last couple of years, schizophrenia was often discussed as autoimmune disease. Several antibodies were suspected, but so far there has been no proof of Gamma-aminobutyric acid (GABA) receptor antibodies in patients with schizophrenia. CASE PRESENTATION In this case report we present a 21-year old woman with schizophrenic symptoms, who showed anti-GABAB1 antibodies when screened by a vast recombinant neurology mosaic on Human Embryonic Kidney Cells 293 (HEK293) cells. The young woman presented with various psychotic symptoms as well as speech and motor ataxia, with the neurological signs starting in childhood. CONCLUSION A hypofunction of the GABAergic system is a possible cause of severe schizophrenic symptoms. Postmortem studies proved this hypothesis by showing dysfunctional GABAergic interneurons in various brain areas. Therefore one should always think of an immune-mediated pathogenesis as well memory impairment and behavioral changes co-occur with frequent seizures.
Collapse
Affiliation(s)
- Ida S. Haussleiter
- Department of Psychiatry, LWL University Hospital, Ruhr-University Bochum, Alexandrinen Str. 1, 44791 Bochum, Germany
| | | | - Georg Juckel
- Department of Psychiatry, LWL University Hospital, Ruhr-University Bochum, Alexandrinen Str. 1, 44791, Bochum, Germany.
| |
Collapse
|
40
|
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: 122] [Impact Index Per Article: 15.3] [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
|
41
|
Anticevic A, Schleifer C, Youngsun TC. Emotional and cognitive dysregulation in schizophrenia and depression: understanding common and distinct behavioral and neural mechanisms. DIALOGUES IN CLINICAL NEUROSCIENCE 2016. [PMID: 26869843 PMCID: PMC4734880 DOI: 10.31887/dcns.2015.17.4/aanticevic] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Emerging behavioral and neuroimaging studies in schizophrenia (SCZ) and major depressive disorder (MD) are mapping mechanisms of co-occurring and distinct affective disturbances across these disorders. This constitutes a critical goal towards developing rationally guided therapies for upstream neural pathways that contribute to comorbid symptoms across disorders. We highlight the current state of the art in our understanding of emotional dysregulation in SCZ versus MD by focusing on broad domains of behavioral function that can map onto underlying neural systems, namely deficits in hedonics, anticipatory behaviors, computations underlying value and effort, and effortful goal-directed behaviors needed to pursue rewarding outcomes. We highlight unique disturbances in each disorder that may involve dissociable neural systems, but also possible interactions between affect and cognition in MD versus SCZ. Finally, we review computational and translational approaches that offer mechanistic insight into how cellular-level disruptions can lead to complex affective disturbances, informing development of therapies across MD and SCZ.
Collapse
Affiliation(s)
- Alan Anticevic
- Department of Psychiatry, Yale University School of Medicine; Interdepartmental Neuroscience Program, Yale University; NIAAA Center for the Translational Neuroscience of Alcoholism; Department of Psychology, Yale University; Division of Neurocognition, Neurogenetics & Neurocomputation, Yale University School of Medicine (Alan Anticevic) - New Haven, Connecticut, USA
| | | | | |
Collapse
|
42
|
The NMDA receptor GluN2C subunit controls cortical excitatory-inhibitory balance, neuronal oscillations and cognitive function. Sci Rep 2016; 6:38321. [PMID: 27922130 PMCID: PMC5138829 DOI: 10.1038/srep38321] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 11/09/2016] [Indexed: 01/11/2023] Open
Abstract
Despite strong evidence for NMDA receptor (NMDAR) hypofunction as an underlying factor for cognitive disorders, the precise roles of various NMDAR subtypes remains unknown. The GluN2C-containing NMDARs exhibit unique biophysical properties and expression pattern, and lower expression of GluN2C subunit has been reported in postmortem brains from schizophrenia patients. We found that loss of GluN2C subunit leads to a shift in cortical excitatory-inhibitory balance towards greater inhibition. Specifically, pyramidal neurons in the medial prefrontal cortex (mPFC) of GluN2C knockout mice have reduced mEPSC frequency and dendritic spine density and a contrasting higher frequency of mIPSCs. In addition a greater number of perisomatic GAD67 puncta was observed suggesting a potential increase in parvalbumin interneuron inputs. At a network level the GluN2C knockout mice were found to have a more robust increase in power of oscillations in response to NMDAR blocker MK-801. Furthermore, GluN2C heterozygous and knockout mice exhibited abnormalities in cognition and sensorimotor gating. Our results demonstrate that loss of GluN2C subunit leads to cortical excitatory-inhibitory imbalance and abnormal neuronal oscillations associated with neurodevelopmental disorders.
Collapse
|
43
|
McKlveen JM, Morano RL, Fitzgerald M, Zoubovsky S, Cassella SN, Scheimann JR, Ghosal S, Mahbod P, Packard BA, Myers B, Baccei ML, Herman JP. Chronic Stress Increases Prefrontal Inhibition: A Mechanism for Stress-Induced Prefrontal Dysfunction. Biol Psychiatry 2016; 80:754-764. [PMID: 27241140 PMCID: PMC5629635 DOI: 10.1016/j.biopsych.2016.03.2101] [Citation(s) in RCA: 164] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 03/06/2016] [Accepted: 03/10/2016] [Indexed: 12/30/2022]
Abstract
BACKGROUND Multiple neuropsychiatric disorders, e.g., depression, are linked to imbalances in excitatory and inhibitory neurotransmission and prefrontal cortical dysfunction, and are concomitant with chronic stress. METHODS We used electrophysiologic (n = 5-6 animals, 21-25 cells/group), neuroanatomic (n = 6-8/group), and behavioral (n = 12/group) techniques to test the hypothesis that chronic stress increases inhibition of medial prefrontal cortex (mPFC) glutamatergic output neurons. RESULTS Using patch clamp recordings from infralimbic mPFC pyramidal neurons, we found that chronic stress selectively increases the frequency of miniature inhibitory postsynaptic currents with no effect on amplitude, which suggests that chronic stress increases presynaptic gamma-aminobutyric acid release. Elevated gamma-aminobutyric acid release under chronic stress is accompanied by increased inhibitory appositions and terminals onto glutamatergic cells, as assessed by both immunohistochemistry and electron microscopy. Furthermore, chronic stress decreases glucocorticoid receptor immunoreactivity specifically in a subset of inhibitory neurons, which suggests that increased inhibitory tone in the mPFC after chronic stress may be caused by loss of a glucocorticoid receptor-mediated brake on interneuron activity. These neuroanatomic and functional changes are associated with impairment of a prefrontal-mediated behavior. During chronic stress, rats initially make significantly more errors in the delayed spatial win-shift task, an mPFC-mediated behavior, which suggests a diminished impact of the mPFC on decision making. CONCLUSIONS Taken together, the data suggest that chronic stress increases synaptic inhibition onto prefrontal glutamatergic output neurons, limiting the influence of the prefrontal cortex in control of stress reactivity and behavior. Thus, these data provide a mechanistic link among chronic stress, prefrontal cortical hypofunction, and behavioral dysfunction.
Collapse
|
44
|
Synthesis of novel non-natural conformationally restricted cyclopropane amino acids of spirane series. Russ Chem Bull 2016. [DOI: 10.1007/s11172-015-1135-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
45
|
Uehara T, Sumiyoshi T, Kurachi M. New Pharmacotherapy Targeting Cognitive Dysfunction of Schizophrenia via Modulation of GABA Neuronal Function. Curr Neuropharmacol 2016; 13:793-801. [PMID: 26630957 PMCID: PMC4759318 DOI: 10.2174/1570159x13666151009120153] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 02/24/2015] [Accepted: 02/26/2015] [Indexed: 11/22/2022] Open
Abstract
Schizophrenia is considered a neurodevelopmental and neurodegenerative disorder. Cognitive impairment is a core symptom in patients with the illness, and has been suggested a major predictor of functional outcomes. Reduction of parvalbumin (PV)-positive γ-aminobutyric acid (GABA) interneurons has been associated with the pathophysiology of schizophrenia, in view of the link between the abnormality of GABA neurons and cognitive impairments of the disease. It is assumed that an imbalance of excitatory and inhibitory (E-I) activity induced by low activity of glutamatergic projections and PV-positive GABA interneurons in the prefrontal cortex resulted in sustained neural firing and gamma oscillation, leading to impaired cognitive function. Therefore, it is important to develop novel pharmacotherapy targeting GABA neurons and their activities. Clinical evidence suggests serotonin (5-HT) 1A receptor agonist improves cognitive disturbances of schizophrenia, consistent with results from preclinical studies, through mechanism that corrects E-I imbalance via the suppression of GABA neural function. On the other hand, T-817MA, a novel neurotrophic agent, ameliorated loss of PV-positive GABA neurons in the medial prefrontal cortex and reduction of gamma-band activity, as well as cognitive dysfunction in animal model of schizophrenia. In conclusion, a pharmacotherapy to alleviate abnormalities in GABA neurons through 5-HT1A agonists and T-817MA is expected to prevent the onset and/or progression of schizophrenia.
Collapse
Affiliation(s)
- Takashi Uehara
- Department of Neuropsychiatry, Kanazawa Medical University, 1-1 Daigaku, Uchinada-cho, Ishikawa 920-0293, Japan.
| | | | | |
Collapse
|
46
|
Shungu DC, Mao X, Gonzales R, Soones TN, Dyke JP, van der Veen JW, Kegeles LS. Brain γ-aminobutyric acid (GABA) detection in vivo with the J-editing (1) H MRS technique: a comprehensive methodological evaluation of sensitivity enhancement, macromolecule contamination and test-retest reliability. NMR IN BIOMEDICINE 2016; 29:932-42. [PMID: 27173449 PMCID: PMC4909570 DOI: 10.1002/nbm.3539] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 03/17/2016] [Accepted: 03/21/2016] [Indexed: 05/21/2023]
Abstract
Abnormalities in brain γ-aminobutyric acid (GABA) have been implicated in various neuropsychiatric and neurological disorders. However, in vivo GABA detection by (1) H MRS presents significant challenges arising from the low brain concentration, overlap by much stronger resonances and contamination by mobile macromolecule (MM) signals. This study addresses these impediments to reliable brain GABA detection with the J-editing difference technique on a 3-T MR system in healthy human subjects by: (i) assessing the sensitivity gains attainable with an eight-channel phased-array head coil; (ii) determining the magnitude and anatomic variation of the contamination of GABA by MM; and (iii) estimating the test-retest reliability of the measurement of GABA with this method. Sensitivity gains and test-retest reliability were examined in the dorsolateral prefrontal cortex (DLPFC), whereas MM levels were compared across three cortical regions: DLPFC, the medial prefrontal cortex (MPFC) and the occipital cortex (OCC). A three-fold higher GABA detection sensitivity was attained with the eight-channel head coil compared with the standard single-channel head coil in DLPFC. Despite significant anatomical variation in GABA + MM and MM across the three brain regions (p < 0.05), the contribution of MM to GABA + MM was relatively stable across the three voxels, ranging from 41% to 49%, a non-significant regional variation (p = 0.58). The test-retest reliability of GABA measurement, expressed as either the ratio to voxel tissue water (W) or to total creatine, was found to be very high for both the single-channel coil and the eight-channel phased-array coil. For the eight-channel coil, for example, Pearson's correlation coefficient of test vs. retest for GABA/W was 0.98 (R(2) = 0.96, p = 0.0007), the percentage coefficient of variation (CV) was 1.25% and the intraclass correlation coefficient (ICC) was 0.98. Similar reliability was also found for the co-edited resonance of combined glutamate and glutamine (Glx) for both coils. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Dikoma C. Shungu
- Department of Radiology, Weill Cornell Medical College, New York, NY
| | - Xiangling Mao
- Department of Radiology, Weill Cornell Medical College, New York, NY
| | - Robyn Gonzales
- Department of Psychiatry, Columbia University, New York, NY
| | | | - Jonathan P. Dyke
- Department of Radiology, Weill Cornell Medical College, New York, NY
| | | | - Lawrence S. Kegeles
- Department of Psychiatry, Columbia University, New York, NY
- Department of Radiology, Columbia University, New York, NY
| |
Collapse
|
47
|
Alshammari TK, Alshammari MA, Nenov MN, Hoxha E, Cambiaghi M, Marcinno A, James TF, Singh P, Labate D, Li J, Meltzer HY, Sacchetti B, Tempia F, Laezza F. Genetic deletion of fibroblast growth factor 14 recapitulates phenotypic alterations underlying cognitive impairment associated with schizophrenia. Transl Psychiatry 2016; 6:e806. [PMID: 27163207 PMCID: PMC5070049 DOI: 10.1038/tp.2016.66] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 02/25/2016] [Accepted: 03/05/2016] [Indexed: 12/14/2022] Open
Abstract
Cognitive processing is highly dependent on the functional integrity of gamma-amino-butyric acid (GABA) interneurons in the brain. These cells regulate excitability and synaptic plasticity of principal neurons balancing the excitatory/inhibitory tone of cortical networks. Reduced function of parvalbumin (PV) interneurons and disruption of GABAergic synapses in the cortical circuitry result in desynchronized network activity associated with cognitive impairment across many psychiatric disorders, including schizophrenia. However, the mechanisms underlying these complex phenotypes are still poorly understood. Here we show that in animal models, genetic deletion of fibroblast growth factor 14 (Fgf14), a regulator of neuronal excitability and synaptic transmission, leads to loss of PV interneurons in the CA1 hippocampal region, a critical area for cognitive function. Strikingly, this cellular phenotype associates with decreased expression of glutamic acid decarboxylase 67 (GAD67) and vesicular GABA transporter (VGAT) and also coincides with disrupted CA1 inhibitory circuitry, reduced in vivo gamma frequency oscillations and impaired working memory. Bioinformatics analysis of schizophrenia transcriptomics revealed functional co-clustering of FGF14 and genes enriched within the GABAergic pathway along with correlatively decreased expression of FGF14, PVALB, GAD67 and VGAT in the disease context. These results indicate that Fgf14(-/-) mice recapitulate salient molecular, cellular, functional and behavioral features associated with human cognitive impairment, and FGF14 loss of function might be associated with the biology of complex brain disorders such as schizophrenia.
Collapse
Affiliation(s)
- T K Alshammari
- Pharmacology and Toxicology Graduate Program, University of Texas Medical Branch, Galveston, TX, USA
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
- King Saud University Graduate Studies Abroad Program, King Saud University, Riyadh, Saudi Arabia
| | - M A Alshammari
- Pharmacology and Toxicology Graduate Program, University of Texas Medical Branch, Galveston, TX, USA
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
- King Saud University Graduate Studies Abroad Program, King Saud University, Riyadh, Saudi Arabia
| | - M N Nenov
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - E Hoxha
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience, University of Torino, Turin, Italy
| | - M Cambiaghi
- Department of Neuroscience, University of Torino, Turin, Italy
| | - A Marcinno
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
| | - T F James
- Department of Neuroscience, University of Torino, Turin, Italy
| | - P Singh
- Department of Mathematics, University of Houston, Houston, TX, USA
| | - D Labate
- Department of Mathematics, University of Houston, Houston, TX, USA
| | - J Li
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, USA
| | - H Y Meltzer
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - B Sacchetti
- Department of Neuroscience, University of Torino, Turin, Italy
| | - F Tempia
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience, University of Torino, Turin, Italy
| | - F Laezza
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
- Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX, USA
- Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
48
|
Pan B, Lian J, Huang XF, Deng C. Aripiprazole Increases the PKA Signalling and Expression of the GABAA Receptor and CREB1 in the Nucleus Accumbens of Rats. J Mol Neurosci 2016; 59:36-47. [PMID: 26894264 DOI: 10.1007/s12031-016-0730-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 02/10/2016] [Indexed: 12/24/2022]
Abstract
The GABAA receptor is implicated in the pathophysiology of schizophrenia and regulated by PKA signalling. Current antipsychotics bind with D2-like receptors, but not the GABAA receptor. The cAMP-responsive element-binding protein 1 (CREB1) is also associated with PKA signalling and may be related to the positive symptoms of schizophrenia. This study investigated the effects of antipsychotics in modulating D2-mediated PKA signalling and its downstream GABAA receptors and CREB1. Rats were treated orally with aripiprazole (0.75 mg/kg, ter in die (t.i.d.)), bifeprunox (0.8 mg/kg, t.i.d.), haloperidol (0.1 mg/kg, t.i.d.) or vehicle for 1 week. The levels of PKA-Cα and p-PKA in the prefrontal cortex (PFC), nucleus accumbens (NAc) and caudate putamen (CPu) were detected by Western blots. The mRNA levels of Gabrb1, Gabrb2, Gabrb3 and Creb1, and their protein expression were measured by qRT-PCR and Western blots, respectively. Aripiprazole elevated the levels of p-PKA and the ratio of p-PKA/PKA in the NAc, but not the PFC and CPu. Correlated with this elevated PKA signalling, aripiprazole elevated the mRNA and protein expression of the GABAA (β-1) receptor and CREB1 in the NAc. While haloperidol elevated the levels of p-PKA and the ratio of p-PKA/PKA in both NAc and CPu, it only tended to increase the expression of the GABAA (β-1) receptor and CREB1 in the NAc, but not the CPu. Bifeprunox had no effects on PKA signalling in these brain regions. These results suggest that aripiprazole has selective effects on upregulating the GABAA (β-1) receptor and CREB1 in the NAc, probably via activating PKA signalling.
Collapse
Affiliation(s)
- Bo Pan
- Antipsychotic Research Laboratory, Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia
- Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Jiamei Lian
- Antipsychotic Research Laboratory, Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia
- Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Xu-Feng Huang
- Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Chao Deng
- Antipsychotic Research Laboratory, Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia.
- Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, NSW, 2522, Australia.
| |
Collapse
|
49
|
Luo P, Chen C, Lu Y, Fu T, Lu Q, Xu X, Li C, He Z, Guo L. Baclofen ameliorates spatial working memory impairments induced by chronic cerebral hypoperfusion via up-regulation of HCN2 expression in the PFC in rats. Behav Brain Res 2016; 308:6-13. [PMID: 27085590 DOI: 10.1016/j.bbr.2016.04.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/19/2016] [Accepted: 04/12/2016] [Indexed: 12/26/2022]
Abstract
Chronic cerebral hypoperfusion (CCH) causes memory deficits and increases the risk of vascular dementia (VD) through several biologically plausible pathways. However, whether CCH causes prefrontal cortex (PFC)-dependent spatial working memory impairments and Baclofen, a GABAB receptor agonist, could ameliorate the impairments is still not clear especially the mechanisms underlying the process. In this study, rats were subjected to permanent bilateral occlusion of the common carotid arteries (two-vessel occlusion, 2VO) to induce CCH. Two weeks later, rats were treated with 25mg/kg Baclofen (intraperitioneal injection, i.p.) for 3 weeks. Spatial working memory was evaluated in a Morris water maze using a modified delayed matching-to-place (DMP) procedure. Western blotting and immunohistochemistry were used to quantify the protein levels and protein localization. Our results showed that 2VO caused striking spatial working memory impairments, accompanied with a decreased HCN2 expression in PFC, but the protein levels of protein gene product 9.5 (PGP9.5, a neuron specific protein), glial fibrillary acidic protein (GFAP), synaptophysin (SYP), brain-derived neurotrophic factor (BDNF), parvalbumin (PV) and HCN1 were not distinguishably changed as compared with sham-operated rats. Baclofen treatment significantly improved the spatial working memory impairments caused by 2VO, accompanied with a reversion of 2VO-induced down-regulation of HCN2. Furthermore, there was a co-localization of HCN2 subunits and parvalbumin-positive neurons in PFC. Therefore, HCN2 may target inhibitory interneurons that is implicated in working memory processes, which may be a possible mechanism of the up-regulation of HCN2 by Baclofen treatment that reliefs spatial working memory deficits in rats with CCH.
Collapse
Affiliation(s)
- Pan Luo
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Cheng Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yun Lu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - TianLi Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qing Lu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xulin Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Changjun Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhi He
- Department of Neuropsychopharmacology, Medical School of China Three Gorges University, Yichang 443002, China.
| | - Lianjun Guo
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
50
|
Obsessive-Compulsive Symptoms in Schizophrenia and in Obsessive-Compulsive Disorder: Differences and Similarities. J Psychiatr Pract 2016; 22:111-6. [PMID: 27138079 DOI: 10.1097/pra.0000000000000131] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION A growing literature suggests that obsessive-compulsive (OC) phenomena represent a distinct dimension in schizophrenia, independent of nuclear psychotic symptoms. Nevertheless, the OC psychopathologic profile in schizophrenia, compared with "pure" obsessive-compulsive disorder (OCD), has not yet been investigated extensively. This study investigated the clinical features of the OC dimension in patients with schizophrenia compared with patients with pure OCD. METHODS The main psychopathologic features of obsessions and compulsions were rated in 35 patients with schizophrenia and 31 patients with OCD, using the Structured Clinical Interview for DSM-IV Axis I Disorders, the Positive and Negative Syndrome Scale, the Yale-Brown Obsessive-Compulsive Scale, and the Leyton Obsessional Inventory. RESULTS OC phenomena were indistinguishable in terms of their severity, resistance, interference, and control in both groups. However, patients with OCD showed higher rates of aggressive, contamination-related, sexual, and somatic themes; moreover, in the group with schizophrenia, a positive relationship was found between washing compulsions and delusions and between hoarding obsessions and delusions. CONCLUSIONS These results indicate that patients with schizophrenia exhibit a narrower range of obsessive content compared with patients with OCD; in addition, OC and delusional themes tend to be related in schizophrenia as a unique symptomatic phenomenon.
Collapse
|