1
|
Klimczak P, Alcaide J, Gramuntell Y, Castillo-Gómez E, Varea E, Perez-Rando M, Nacher J. Long-term effects of a double hit murine model for schizophrenia on parvalbumin expressing cells and plasticity-related molecules in the thalamic reticular nucleus and the habenula. Transl Psychiatry 2024; 14:450. [PMID: 39448557 PMCID: PMC11502763 DOI: 10.1038/s41398-024-03166-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 10/04/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024] Open
Abstract
The exposure to aversive experiences during early-life affects brain maturation and induces changes in behavior. Additionally, when these experiences coincide with subtle neurodevelopmental alterations, they may contribute to the emergence of psychiatric disorders, such as schizophrenia. Studies in patients and animal models have identified changes in parvalbumin (PV) expressing inhibitory neurons, highlighting their significance in the etiology of this disorder. Most studies have been focused on the cortex, but PV+ neurons also provide inhibitory input to diencephalic regions, particularly to the thalamus (through cells in the thalamic reticular nucleus, TRN) and the habenula. Remarkably, alterations in both nuclei have been described in schizophrenia. Some of these changes in PV+ cells may be mediated by perineuronal nets (PNN), specialized regions of the extracellular matrix that often surround them and regulate their synaptic input and activity. Interestingly, the physiological maturation and integration of PV+ neurons, which involves the assembly of PNN, occurs during early postnatal life. Plasticity molecules associated to inhibitory neurons, such as PSA-NCAM, or NMDA receptors (NMDAR) can also influence the structure and function of these cells. Growing evidence also indicates that glial cells regulate the physiology of PV+ neurons by influencing their maturation and modulating their synaptic connectivity. To explore the impact of early-life aversive experiences and concomitant subtle neurodevelopmental alterations on diencephalic PV+ cells, we analyzed adult male mice subjected to a double-hit model (DHM) of schizophrenia, combining a single injection of an NMDAR antagonist at P7 and post-weaning social isolation. We observed that exploratory behavior, PV+ neurons and their associated PNN, as well as PSA-NCAM and NMDAR expression and glial cells, in the TRN and the habenula were affected by the DHM or one of its factors. To our knowledge, this is the first report on such alterations in these diencephalic structures in an animal model combining neurodevelopmental alterations and early-life stress during adolescence. Our findings complement previous work on PV+ neurons in cortical regions and underscore the importance of studying diencephalic inhibitory networks and their intricate interactions with aversive experiences and neurodevelopmental alterations during early life in the context of schizophrenia.
Collapse
Affiliation(s)
- Patrycja Klimczak
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010, Valencia, Spain
| | - Julia Alcaide
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010, Valencia, Spain
| | - Yaiza Gramuntell
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010, Valencia, Spain
| | - Esther Castillo-Gómez
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain
- Department of Medicine, School of Medical Sciences, Universitat Jaume I, Valencia, Spain
| | - Emilio Varea
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain
| | - Marta Perez-Rando
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain.
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain.
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010, Valencia, Spain.
| | - Juan Nacher
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain.
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain.
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010, Valencia, Spain.
| |
Collapse
|
2
|
Walters JM, Noblet HA, Chung HJ. An emerging role of STriatal-Enriched protein tyrosine Phosphatase in hyperexcitability-associated brain disorders. Neurobiol Dis 2024; 200:106641. [PMID: 39159894 DOI: 10.1016/j.nbd.2024.106641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024] Open
Abstract
STriatal-Enriched protein tyrosine Phosphatase (STEP) is a brain-specific tyrosine phosphatase that is associated with numerous neurological and neuropsychiatric disorders. STEP dephosphorylates and inactivates various kinases and phosphatases critical for neuronal function and health including Fyn, Pyk2, ERK1/2, p38, and PTPα. Importantly, STEP dephosphorylates NMDA and AMPA receptors, two major glutamate receptors that mediate fast excitatory synaptic transmission. This STEP-mediated dephosphorylation leads to their internalization and inhibits both Hebbian synaptic potentiation and homeostatic synaptic scaling. Hence, STEP has been widely accepted to weaken excitatory synaptic strength. However, emerging evidence implicates a novel role of STEP in neuronal hyperexcitability and seizure disorders. Genetic deletion and pharmacological blockade of STEP reduces seizure susceptibility in acute seizure mouse models and audiogenic seizures in a mouse model of Fragile X syndrome. Pharmacologic inhibition of STEP also decreases hippocampal activity and neuronal intrinsic excitability. Here, we will highlight the divergent roles of STEP in excitatory synaptic transmission and neuronal intrinsic excitability, present the potential underlying mechanisms, and discuss their impact on STEP-associated neurologic and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jennifer M Walters
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hayden A Noblet
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hee Jung Chung
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Institute of Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
3
|
Liu S, Yan Z, Peng Y, Liu Y, Li Y, Xu D, Gong Y, Cui Z, Wu Y, Zhang Y, Wang D, Pan W, Yang X. Lentinan has a beneficial effect on cognitive deficits induced by chronic Toxoplasma gondii infection in mice. Parasit Vectors 2023; 16:454. [PMID: 38093309 PMCID: PMC10717010 DOI: 10.1186/s13071-023-06023-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 10/19/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Toxoplasma gondii (T. gondii) is increasingly considered a risk factor for neurodegenerative diseases. However, there is only limited information on the development of drugs for T. gondii infection. Lentinan from Lentinula edodes is a bioactive ingredient with the potential to enhance anti-infective immunity. The present study aimed to investigate the neuroprotective effect of lentinan on T. gondii-associated cognitive deficits in mice. METHODS A chronic T. gondii infection mouse model was established by administering 10 cysts of T. gondii by gavage. Lentinan was intraperitoneally administered 2 weeks before infection. Behavioral tests, RNA sequencing, immunofluorescence, transmission electron microscopy and Golgi-Cox staining were performed to assess the effect of lentinan on cognitive deficits and neuropathology in vivo. In vitro, the direct and indirect effects of lentinan on the proliferation of T. gondii tachyzoites were evaluated in the absence and presence of BV-2 cells, respectively. RESULTS Lentinan prevented T. gondii-induced cognitive deficits and altered the transcriptome profile of genes related to neuroinflammation, microglial activation, synaptic function, neural development and cognitive behavior in the hippocampus of infected mice. Moreover, lentinan reduced the infection-induced accumulation of microglia and downregulated the mRNA expression of proinflammatory cytokines. In addition, the neurite and synaptic ultrastructural damage in the hippocampal CA1 region due to infection was ameliorated by lentinan administration. Lentinan decreased the cyst burden in the brains of infected mice, which was correlated with behavioral performance. In line with this finding, lentinan could significantly inhibit the proliferation of T. gondii tachyzoites in the microglial cell line BV2, although lentinan had no direct inhibitory effect on parasite growth. CONCLUSIONS Lentinan prevents cognitive deficits via the improvement of neurite impairment and synaptic loss induced by T. gondii infection, which may be associated with decreased cyst burden in the brain. Overall, our findings indicate that lentinan can ameliorate T. gondii-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Shuxi Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ziyi Yan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuan Peng
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yunqiu Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yiling Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Daxiang Xu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yuying Gong
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Zeyu Cui
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yongshui Wu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yumei Zhang
- Department of Pathogenic Biology, Binzhou Medical University, Binzhou, 256603, Shandong, China
| | - Dahui Wang
- Liangshan College (Li Shui) China, Lishui University, Lishui, 323000, Zhejiang, China.
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
4
|
Felipe Souza E Silva L, Siena Dos Santos A, Mayumi Yuzawa J, Luiz de Barros Torresi J, Ziroldo A, Rosado Rosenstock T. SIRTUINS MODULATORS COUNTERACT MITOCHONDRIAL DYSFUNCTION IN CELLULAR MODELS OF HYPOXIA: RELEVANCE TO SCHIZOPHRENIA. Neuroscience 2023:S0306-4522(23)00200-2. [PMID: 37169164 DOI: 10.1016/j.neuroscience.2023.04.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 04/16/2023] [Accepted: 04/27/2023] [Indexed: 05/13/2023]
Abstract
Schizophrenia (SZ) is a neurodevelopmental-associated disorder strongly related to environmental factors, such as hypoxia. Because there is no cure for SZ or any pharmacological approach that could revert hypoxia-induced cellular damages, we evaluated whether modulators of sirtuins could abrogate hypoxia-induced mitochondrial deregulation as a neuroprotective strategy. Firstly, astrocytes from control (Wistar) and Spontaneously Hypertensive Rats (SHR), a model of both SZ and neonatal hypoxia, were submitted to chemical hypoxia. Then, cells were exposed to different concentrations of Nicotinamide (NAM), Resveratrol (Resv), and Sirtinol (Sir) for 48hrs. Our data indicate that sirtuins modulation reduces cell death increasing the acetylation of histone 3. This outcome is related to the rescue of loss of mitochondrial membrane potential, changes in mitochondrial calcium buffering capacity, decreased O2-• levels and increased expression of metabolic regulators (Nrf-1 and Nfe2l2) and mitochondrial content. Such findings are relevant not only for hypoxia-associated conditions, named pre-eclampsia but also for SZ since prenatal hypoxia is a relevant environmental factor related to this burdensome neuropsychiatric disorder.
Collapse
Affiliation(s)
- Luiz Felipe Souza E Silva
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Amanda Siena Dos Santos
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Jessica Mayumi Yuzawa
- Department of Physiological Science, Santa Casa de São Paulo School of Medical Science, São Paulo, Brazil
| | | | - Alan Ziroldo
- Department of Physiological Science, Santa Casa de São Paulo School of Medical Science, São Paulo, Brazil
| | - Tatiana Rosado Rosenstock
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil; Dept. of Bioscience, In-vitro Neuroscience, Sygnature Discovery, Nottingham, United Kingdom.
| |
Collapse
|
5
|
Lv S, Yao K, Zhang Y, Zhu S. NMDA receptors as therapeutic targets for depression treatment: Evidence from clinical to basic research. Neuropharmacology 2023; 225:109378. [PMID: 36539011 DOI: 10.1016/j.neuropharm.2022.109378] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/08/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
Ketamine, functioning as a channel blocker of the excitatory glutamate-gated N-methyl-d-aspartate (NMDA) receptors, displays compelling fast-acting and sustained antidepressant effects for treatment-resistant depression. Over the past decades, clinical and preclinical studies have implied that the pathology of depression is associated with dysfunction of glutamatergic transmission. In particular, the discovery of antidepressant agents modulating NMDA receptor function has prompted breakthroughs for depression treatment compared with conventional antidepressants targeting the monoaminergic system. In this review, we first summarized the signalling pathway of the ketamine-mediated antidepressant effects, based on the glutamate hypothesis of depression. Second, we reviewed the hypotheses of the synaptic mechanism and network of ketamine antidepressant effects within different brain areas and distinct subcellular localizations, including NMDA receptor antagonism on GABAergic interneurons, extrasynaptic and synaptic NMDA receptor-mediated antagonism, and ketamine blocking bursting activities in the lateral habenula. Third, we reviewed the different roles of NMDA receptor subunits in ketamine-mediated cognitive and psychiatric behaviours in genetically-manipulated rodent models. Finally, we summarized the structural basis of NMDA receptor channel blockers and discussed NMDA receptor modulators that have been reported to exert potential antidepressant effects in animal models or in clinical trials. Integrating the cutting-edge technologies of cryo-EM and artificial intelligence-based drug design (AIDD), we expect that the next generation of first-in-class rapid antidepressants targeting NMDA receptors would be an emerging direction for depression therapeutics. This article is part of the Special Issue on 'Ketamine and its Metabolites'.
Collapse
Affiliation(s)
- Shiyun Lv
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China; University of Chinese Academy of Sciences, Beijing, China
| | - Kejie Yao
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China; University of Chinese Academy of Sciences, Beijing, China
| | - Youyi Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China; University of Chinese Academy of Sciences, Beijing, China
| | - Shujia Zhu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
6
|
Schizophrenia-associated LRRTM1 regulates cognitive behavior through controlling synaptic function in the mediodorsal thalamus. Mol Psychiatry 2021; 26:6912-6925. [PMID: 33981006 DOI: 10.1038/s41380-021-01146-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 01/08/2023]
Abstract
Reduced activity of the mediodorsal thalamus (MD) and abnormal functional connectivity of the MD with the prefrontal cortex (PFC) cause cognitive deficits in schizophrenia. However, the molecular basis of MD hypofunction in schizophrenia is not known. Here, we identified leucine-rich-repeat transmembrane neuronal protein 1 (LRRTM1), a postsynaptic cell-adhesion molecule, as a key regulator of excitatory synaptic function and excitation-inhibition balance in the MD. LRRTM1 is strongly associated with schizophrenia and is highly expressed in the thalamus. Conditional deletion of Lrrtm1 in the MD in adult mice reduced excitatory synaptic function and caused a parallel reduction in the afferent synaptic activity of the PFC, which was reversed by the reintroduction of LRRTM1 in the MD. Our results indicate that chronic reduction of synaptic strength in the MD by targeted deletion of Lrrtm1 functionally disengages the MD from the PFC and may account for cognitive, social, and sensorimotor gating deficits, reminiscent of schizophrenia.
Collapse
|
7
|
Wang X, Li Y, Chen J, Li Z, Li J, Qin L. Aberrant Auditory Steady-State Response of Awake Mice After Single Application of the NMDA Receptor Antagonist MK-801 Into the Medial Geniculate Body. Int J Neuropsychopharmacol 2020; 23:459-468. [PMID: 32725129 PMCID: PMC7387767 DOI: 10.1093/ijnp/pyaa022] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/18/2020] [Accepted: 03/21/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Systemic administration of noncompetitive N-methyl-D-aspartate receptor (NMDAR) antagonists such as MK-801 is widely used to model psychosis of schizophrenia (SZ). Acute systemic MK-801 in rodents caused an increase of the auditory steady-state responses (ASSRs), the oscillatory neural responses to periodic auditory stimulation, while most studies in patients with SZ reported a decrease of ASSRs. This inconsistency may be attributable to the comprehensive effects of systemic administration of MK-801. Here, we examined how the ASSR is affected by selectively blocking NMDAR in the thalamus. METHODS We implanted multiple electrodes in the auditory cortex (AC) and prefrontal cortex to simultaneously record the local field potential and spike activity (SA) of multiple sites from awake mice. Click-trains at a 40-Hz repetition rate were used to evoke the ASSR. We compared the mean trial power and phase-locking factor and the firing rate of SA before and after microinjection of MK-801 (1.5 µg) into the medial geniculate body (MGB). RESULTS We found that both the AC and prefrontal cortex showed a transient local field potential response at the onset of click-train stimulus, which was less affected by the application of MK-801 in the MGB. Following the onset response, the AC also showed a response continuing throughout the stimulus period, corresponding to the ASSR, which was suppressed by the application of MK-801. CONCLUSION Our data suggest that the MGB is one of the generators of ASSR, and NMDAR hypofunction in the thalamocortical projection may account for the ASSR deficits in SZ.
Collapse
Affiliation(s)
- Xuejiao Wang
- Department of Physiology, China Medical University, Shenyang, People’s Republic of China
| | - Yingzhuo Li
- Department of Physiology, China Medical University, Shenyang, People’s Republic of China
| | - Jingyu Chen
- Department of Physiology, China Medical University, Shenyang, People’s Republic of China
| | - Zijie Li
- Department of Physiology, China Medical University, Shenyang, People’s Republic of China
| | - Jinhong Li
- Department of Physiology, China Medical University, Shenyang, People’s Republic of China
| | - Ling Qin
- Department of Physiology, China Medical University, Shenyang, People’s Republic of China,Correspondence: Ling Qin, MD, PhD, Department of Physiology, China Medical University, Shenyang, 110122, People’s Republic of China ()
| |
Collapse
|
8
|
Schoonover KE, Dienel SJ, Lewis DA. Prefrontal cortical alterations of glutamate and GABA neurotransmission in schizophrenia: Insights for rational biomarker development. Biomark Neuropsychiatry 2020; 3. [PMID: 32656540 PMCID: PMC7351254 DOI: 10.1016/j.bionps.2020.100015] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Certain cognitive deficits in schizophrenia, such as impaired working memory, are thought to reflect alterations in the neural circuitry of the dorsolateral prefrontal cortex (DLPFC). Gamma oscillations in the DLPFC appear to be a neural corollary of working memory function, and the power of these oscillations during working memory tasks is lower in individuals with schizophrenia. Thus, gamma oscillations represent a potentially useful biomarker to index dysfunction in the DLPFC circuitry responsible for working memory in schizophrenia. Postmortem studies, by identifying the cellular basis of DLPFC dysfunction, can help inform the utility of biomarker measures obtained in vivo. Given that gamma oscillations reflect network activity of excitatory pyramidal neurons and inhibitory GABA neurons, we review postmortem findings of alterations to both cell types in the DLPFC and discuss how these findings might inform future biomarker development and use.
Collapse
Affiliation(s)
- Kirsten E Schoonover
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, United States
| | - Samuel J Dienel
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, United States.,Medical Scientist Training Program, University of Pittsburgh, United States.,Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, United States.,Center for the Neural Basis of Cognition, Carnegie Mellon University, United States
| | - David A Lewis
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, United States.,Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, United States.,Center for the Neural Basis of Cognition, Carnegie Mellon University, United States
| |
Collapse
|
9
|
Xiao G, Llano DA. Hitting The Right Spot: NMDA Receptors in the Auditory Thalamus May Hold the Key to Understanding Schizophrenia. Int J Neuropsychopharmacol 2020; 23:578-580. [PMID: 32374824 PMCID: PMC7710913 DOI: 10.1093/ijnp/pyaa032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 05/01/2020] [Indexed: 12/18/2022] Open
Abstract
In this issue, Wang and colleagues solve an important puzzle in the understanding of schizophrenia. Previous work has linked N-methyl-D-aspartate (NMDA) receptor hypofunction to schizophrenia and shown that individuals with schizophrenia have a suppressed steady-state cortical response to 40-Hz repetitive auditory stimulation. However, systemic application of NMDA antagonists paradoxically increases this cortical response in rodents. Here, by specifically applying NMDA receptor blockade in the auditory thalamus while simultaneously measuring the acoustically driven response in 2 cortical regions, Wang and colleagues found the drop in the steady-state response that is seen in schizophrenia. These findings solve an important paradox in the field and suggest that specific thalamic neurochemical alterations may occur in the brain of individuals with schizophrenia. In addition, this work suggests that suppression of NMDA receptors in the thalamus may serve as a potential animal model for the disease.
Collapse
Affiliation(s)
- Gang Xiao
- Department of Molecular and Integrative Physiology, University of Illinois, Urbana-Champaign, Illinois,Beckman Institute for Advanced Science and Technology, Urbana, Illinois
| | - Daniel A Llano
- Department of Molecular and Integrative Physiology, University of Illinois, Urbana-Champaign, Illinois,Beckman Institute for Advanced Science and Technology, Urbana, Illinois,Corresponding Author: Daniel A. Llano MD, PhD, 2355 Beckman Institute, 405 N. Mathews Ave, Urbana, IL 61801 ()
| |
Collapse
|
10
|
E Silva LFS, Brito MD, Yuzawa JMC, Rosenstock TR. Mitochondrial Dysfunction and Changes in High-Energy Compounds in Different Cellular Models Associated to Hypoxia: Implication to Schizophrenia. Sci Rep 2019; 9:18049. [PMID: 31792231 PMCID: PMC6889309 DOI: 10.1038/s41598-019-53605-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 10/24/2019] [Indexed: 02/06/2023] Open
Abstract
Schizophrenia (SZ) is a multifactorial mental disorder, which has been associated with a number of environmental factors, such as hypoxia. Considering that numerous neural mechanisms depends on energetic supply (ATP synthesis), the maintenance of mitochondrial metabolism is essential to keep cellular balance and survival. Therefore, in the present work, we evaluated functional parameters related to mitochondrial function, namely calcium levels, mitochondrial membrane potential, redox homeostasis, high-energy compounds levels and oxygen consumption, in astrocytes from control (Wistar) and Spontaneously Hypertensive Rats (SHR) animals exposed both to chemical and gaseous hypoxia. We show that astrocytes after hypoxia presented depolarized mitochondria, disturbances in Ca2+ handling, destabilization in redox system and alterations in ATP, ADP, Pyruvate and Lactate levels, in addition to modification in NAD+/NADH ratio, and Nfe2l2 and Nrf1 expression. Interestingly, intrauterine hypoxia also induced augmentation in mitochondrial biogenesis and content. Altogether, our data suggest that hypoxia can induce mitochondrial deregulation and a decrease in energy metabolism in the most prevalent cell type in the brain, astrocytes. Since SHR are also considered an animal model of SZ, our results can likewise be related to their phenotypic alterations and, therefore, our work also allow an increase in the knowledge of this burdensome disorder.
Collapse
|
11
|
López Hill X, Richeri A, McGregor R, Acuña A, Scorza C. Neuro-behavioral effects after systemic administration of MK-801 and disinhibition of the anterior thalamic nucleus in rats: Potential relevance in schizophrenia. Brain Res 2019; 1718:176-185. [PMID: 31071305 DOI: 10.1016/j.brainres.2019.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/01/2019] [Accepted: 05/04/2019] [Indexed: 10/26/2022]
Abstract
Non-competitive N-methyl-d-aspartate receptor (NMDA-R) antagonists have been suggested to evoke psychotomimetic-like behaviors by selectively targeting GABAergic elements in cortical and thalamic circuits. In previous studies, we had reported the involvement of the reticular and anterior thalamic nuclei (ATN) in the MK-801-evoked hyperactivity and other motor alterations. Consistent with the possibility that these responses were mediated by thalamic disinhibition, we examined the participation of cortical and hippocampal areas innervated by ATN in the responses elicited by the systemic administration of MK-801 (0.2 mg/kg) and compared them to the effects produced by the microinjection of a subconvulsive dose of bicuculline (GABAA receptor antagonist) in the ATN. We used the expression of Fos related antigen 2 (Fra-2) as a neuronal activity marker in the ATN and its projection areas such as hippocampus (HPC), retrosplenial cortex (RS), entorhinal cortex (EC) and medial prefrontal cortex (mPFC). Dorsal (caudate-putamen, CPu) and ventral striatum (nucleus accumbens, core and shell, NAc,co and NAc,sh) were also studied. Behavioral and brain activation results suggest a partial overlap after the effect of MK-801 administration and ATN disinhibition. MK-801 and ATN disinhibition increases locomotor activity and disorganized movements, while ATN disinhibition also reduces rearing behavior. A significant increase in Fra-2 immunoreactivity (Fra-2-IR) in the ATN, mPFC (prelimbic area, PrL) and NAc,sh was observed after MK-801, while a different pattern of Fra-2-IR was detected following ATN disinhibition (e.g., increase in DG and NAc,sh, and decrease in PrL cortex). Overall, our data may contribute to the understanding of dysfunctional neural circuits involved in schizophrenia.
Collapse
Affiliation(s)
- Ximena López Hill
- Department of Experimental Neuropharmacology, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Analía Richeri
- Department of Experimental Neuropharmacology, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay; Laboratory of Cell Biology, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Ronald McGregor
- Veterans Administration Greater Los Angeles Healthcare System, Neurobiology Research (151A3), North Hills, CA 91343, United States; Department Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA 90095, United States; Brain Research Institute, University of California at Los Angeles, Los Angeles, CA 90095, United States
| | - Alejo Acuña
- Department of Experimental Neuropharmacology, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Cecilia Scorza
- Department of Experimental Neuropharmacology, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay.
| |
Collapse
|
12
|
Wang X, Luo C, Mao XY, Li X, Yin JY, Zhang W, Zhou HH, Liu ZQ. Metformin reverses the schizophrenia-like behaviors induced by MK-801 in rats. Brain Res 2019; 1719:30-39. [PMID: 31121159 DOI: 10.1016/j.brainres.2019.05.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 05/16/2019] [Accepted: 05/19/2019] [Indexed: 12/11/2022]
Abstract
Schizophrenia is known to be a complex and disabling psychiatric disorder. Dopamine receptor antagonists have a significant therapeutic effect in improving the positive symptoms that are associated with the illness. Therefore, dopamine receptor antagonists are commonly used in the treatment of schizophrenia; however, they do not achieve satisfactory results in improving negative symptoms and cognitive impairment. Metformin, widely known as an antidiabetic drug, has been found to enhance spatial memory formation and improve anxiety-like behaviors in rodents. Metformin's neuroprotective effect has been well documented in several neurological disorders including Alzheimer's disease, Parkinson's disease, strokes, Huntington's disease, and seizures. In the present study, we used a rat model to explore the effect of metformin on schizophrenia-like behaviors induced by MK-801 (dizocilpine), an N-methyl-D-aspartate (NMDA) receptor antagonist. We found that the pre-pulse inhibition (PPI) deficit caused by MK-801 could be alleviated by metformin. The hyperlocomotion in the open field test induced by chronic treatment of MK-801 was reversed by administration of metformin. Metformin has no effect on the baseline level of anxiety in normal naive rats, while metformin could relieve the anxiety-like behaviors in MK-801-treatment rats, though this effect is not reaching a significant level. Additionally, metformin could significantly ameliorate working memory impairments induced by MK-801. Moreover, the increased level of phosphorylation of Akt and GSK3β in the frontal cortex induced by MK-801 was normalized by metformin. In conclusion, our results demonstrate that metformin improved schizophrenia-like symptoms in rats, and is therefore a potential agent for the treatment of schizophrenia.
Collapse
Affiliation(s)
- Xu Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China
| | - Chao Luo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China; School of Life Sciences, Central South University, Changsha, Hunan 410078, PR China
| | - Xiao-Yuan Mao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China
| | - Xi Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China
| | - Ji-Ye Yin
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China.
| |
Collapse
|
13
|
Tecelão D, Mendes A, Martins D, Fu C, Chaddock CA, Picchioni MM, McDonald C, Kalidindi S, Murray R, Prata DP. The effect of psychosis associated CACNA1C, and its epistasis with ZNF804A, on brain function. GENES BRAIN AND BEHAVIOR 2018; 18:e12510. [PMID: 30079586 DOI: 10.1111/gbb.12510] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 07/23/2018] [Accepted: 08/02/2018] [Indexed: 12/11/2022]
Abstract
CACNA1C-rs1006737 and ZNF804A-rs1344706 polymorphisms are among the most robustly associated with schizophrenia (SCZ) and bipolar disorder (BD), and recently with brain phenotypes. As these patients show abnormal verbal fluency (VF) and related brain activation, we asked whether the latter was affected by these polymorphisms (alone and in interaction)-to better understand how they might induce risk. We recently reported effects on functional VF-related (for ZNF804A-rs1344706) and structural (for both) connectivity. We genotyped and fMRI-scanned 54 SCZ, 40 BD and 80 controls during VF. With SPM, we assessed the main effect of CACNA1C-rs1006737, and its interaction with ZNF804A-rs1344706, and their interaction with diagnosis, on regional brain activation and functional connectivity (psychophysiological interactions-PPI). Using public data, we reported effects of CACNA1C-rs1006737 and diagnosis on brain expression. The CACNA1C-rs1006737 risk allele was associated with increased activation, particularly in the bilateral prefronto-temporal cortex and thalamus; decreased PPI, especially in the left temporal cortex; and gene expression in white matter and the cerebellum. We also found unprecedented evidence for epistasis (interaction between genetic polymorphisms) in the caudate nucleus, thalamus, and cingulate and temporal cortical activation; and CACNA1C up-regulation in SCZ and BD parietal cortices. Some effects were dependent on BD/SCZ diagnosis. All imaging results were whole-brain, voxel-wise, and familywise-error corrected. Our results support evidence implicating CACNA1C and ZNF804A in BD and SCZ, adding novel imaging evidence in clinical populations, and of epistasis-which needs further replication. Further scrutiny of the inherent neurobiological mechanisms may disclose their potential as putative drug targets.
Collapse
Affiliation(s)
- Diogo Tecelão
- Departamento de Física, Faculdade de Ciências e Tecnologia da Universidade Nova de Lisboa, Lisbon, Portugal
| | - Ana Mendes
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa
| | - Daniel Martins
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Cynthia Fu
- School of Psychology, The University of East London, London, UK
| | - Christopher A Chaddock
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Marco M Picchioni
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.,St. Andrew's Academic Department, St Andrew's Healthcare, Northampton, UK
| | - Colm McDonald
- Centre for Neuroimaging and Cognitive Genomics (NICOG) & NCBES Galway Neuroscience Centre, College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Sridevi Kalidindi
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Robin Murray
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Diana P Prata
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa.,Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.,Instituto Universitário de Lisboa (ISCTE-IUL), Cis-IUL, Lisbon, Portugal
| |
Collapse
|
14
|
Wolf R, Dobrowolny H, Nullmeier S, Bogerts B, Schwegler H. Effects of neonatal excitotoxic lesions in ventral thalamus on social interaction in the rat. Eur Arch Psychiatry Clin Neurosci 2018; 268:461-470. [PMID: 28361258 DOI: 10.1007/s00406-017-0781-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 03/06/2017] [Indexed: 12/31/2022]
Abstract
The role of the thalamus in schizophrenia has increasingly been studied in recent years. Deficits in the ventral thalamus have been described in only few postmortem and neuroimaging studies. We utilised our previously introduced neurodevelopmental animal model, the neonatal excitotoxic lesion of the ventral thalamus of Sprague-Dawley rats (Wolf et al., Pharmacopsychiatry 43:99-109, 22). At postnatal day (PD7), male pubs received bilateral thalamic infusions with ibotenic acid (IBA) or artificial cerebrospinal fluid (control). In adulthood, social interaction of two animals not familiar to each other was studied by a computerised video tracking system. This study displays clear lesion effects on social interaction of adult male rats. The significant reduction of total contact time and the significant increase in distance between the animals in the IBA group compared to controls can be interpreted as social withdrawal modelling a negative symptom of schizophrenia. The significant increase of total distance travelled in the IBA group can be hypothesised as agitation modelling a positive symptom of schizophrenia. Using a triple concept of social interaction, the percentage of no social interaction (Non-SI%) was significantly larger, and inversely, the percentage of passive social interaction (SI-passive%) was significantly smaller in the IBA group when compared to controls. In conclusion, on the background of findings in schizophrenic patients, the effects of neonatal ventral thalamic IBA lesions in adult male rats support the hypothesis of face and construct validity as animal model of schizophrenia.
Collapse
Affiliation(s)
- Rainer Wolf
- Department of Psychiatry, Ruhr-University Bochum, Alexandrinenstr. 1, 44791, Bochum, Germany.
- Department of Psychiatry, Otto-von-Guericke University, Magdeburg, Germany.
| | - Henrik Dobrowolny
- Department of Psychiatry, Otto-von-Guericke University, Magdeburg, Germany
| | - Sven Nullmeier
- Institute of Anatomy, Otto-von-Guericke University, Magdeburg, Germany
| | - Bernhard Bogerts
- Department of Psychiatry, Otto-von-Guericke University, Magdeburg, Germany
| | - Herbert Schwegler
- Institute of Anatomy, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
15
|
Sullivan CR, O'Donovan SM, McCullumsmith RE, Ramsey A. Defects in Bioenergetic Coupling in Schizophrenia. Biol Psychiatry 2018; 83:739-750. [PMID: 29217297 PMCID: PMC5891385 DOI: 10.1016/j.biopsych.2017.10.014] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/18/2017] [Accepted: 10/09/2017] [Indexed: 02/06/2023]
Abstract
Synaptic neurotransmission relies on maintenance of the synapse and meeting the energy demands of neurons. Defects in excitatory and inhibitory synapses have been implicated in schizophrenia, likely contributing to positive and negative symptoms as well as impaired cognition. Recently, accumulating evidence has suggested that bioenergetic systems, important in both synaptic function and cognition, are abnormal in psychiatric illnesses such as schizophrenia. Animal models of synaptic dysfunction demonstrated endophenotypes of schizophrenia as well as bioenergetic abnormalities. We report findings on the bioenergetic interplay of astrocytes and neurons and discuss how dysregulation of these pathways may contribute to the pathogenesis of schizophrenia, highlighting metabolic systems as important therapeutic targets.
Collapse
Affiliation(s)
- Courtney R Sullivan
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, Ohio
| | - Sinead M O'Donovan
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, Ohio
| | - Robert E McCullumsmith
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, Ohio.
| | - Amy Ramsey
- Department of Pharmacology and Toxicology, University of Toronto, Ontario, Canada
| |
Collapse
|
16
|
Ravikrishnan A, Gandhi PJ, Shelkar GP, Liu J, Pavuluri R, Dravid SM. Region-specific Expression of NMDA Receptor GluN2C Subunit in Parvalbumin-Positive Neurons and Astrocytes: Analysis of GluN2C Expression using a Novel Reporter Model. Neuroscience 2018; 380:49-62. [PMID: 29559384 DOI: 10.1016/j.neuroscience.2018.03.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/26/2018] [Accepted: 03/09/2018] [Indexed: 12/12/2022]
Abstract
Hypofunction of NMDA receptors in parvalbumin (PV)-positive interneurons has been proposed as a potential mechanism for cortical abnormalities and symptoms in schizophrenia. GluN2C-containing receptors have been linked to this hypothesis due to the higher affinity of psychotomimetic doses of ketamine for GluN1/2C receptors. However, the precise cell-type expression of GluN2C subunit remains unknown. We describe the expression of the GluN2C subunit using a novel EGFP reporter model. We observed EGFP(GluN2C) localization in PV-positive neurons in the nucleus reticularis of the thalamus, globus pallidus externa and interna, ventral pallidum and substantia nigra. In contrast, EGFP(GluN2C)-expressing cells did not co-localize with PV-positive neurons in the cortex, striatum, hippocampus or amygdala. Instead, EGFP(GluN2C) expression in these regions co-localized with an astrocytic marker. We confirmed functional expression of GluN2C-containing receptors in the PV-neurons in substantia nigra and cortical astrocytes using electrophysiology. GluN2C was found to be enriched in several first-order and higher order thalamic nuclei. Interestingly, we found that a previous GluN2C β-gal reporter model excluded expression from PV-neurons and certain thalamic nuclei but exhibited expression in the retrosplenial cortex. GluN2C's unique distribution in neuronal and non-neuronal cells in a brain region-specific manner raises interesting questions regarding the role of GluN2C-containing receptors in the central nervous system.
Collapse
Affiliation(s)
| | - Pauravi J Gandhi
- Department of Pharmacology, Creighton University, Omaha, NE 68178, USA
| | - Gajanan P Shelkar
- Department of Pharmacology, Creighton University, Omaha, NE 68178, USA
| | - Jinxu Liu
- Department of Pharmacology, Creighton University, Omaha, NE 68178, USA
| | | | - Shashank M Dravid
- Department of Pharmacology, Creighton University, Omaha, NE 68178, USA.
| |
Collapse
|
17
|
Sherif MA, Cortes-Briones JA, Ranganathan M, Skosnik PD. Cannabinoid-glutamate interactions and neural oscillations: implications for psychosis. Eur J Neurosci 2018; 48:2890-2902. [PMID: 29247465 DOI: 10.1111/ejn.13800] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 12/06/2017] [Accepted: 12/07/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Mohamed A. Sherif
- Department of Psychiatry; Yale University School of Medicine; VA Connecticut Healthcare System Building 5, Suite C-214 950 Campbell Avenue West Haven CT 06516 USA
| | - Jose A. Cortes-Briones
- Department of Psychiatry; Yale University School of Medicine; VA Connecticut Healthcare System Building 5, Suite C-214 950 Campbell Avenue West Haven CT 06516 USA
| | - Mohini Ranganathan
- Department of Psychiatry; Yale University School of Medicine; VA Connecticut Healthcare System Building 5, Suite C-214 950 Campbell Avenue West Haven CT 06516 USA
| | - Patrick D. Skosnik
- Department of Psychiatry; Yale University School of Medicine; VA Connecticut Healthcare System Building 5, Suite C-214 950 Campbell Avenue West Haven CT 06516 USA
| |
Collapse
|
18
|
Yu Y, Lin Y, Takasaki Y, Wang C, Kimura H, Xing J, Ishizuka K, Toyama M, Kushima I, Mori D, Arioka Y, Uno Y, Shiino T, Nakamura Y, Okada T, Morikawa M, Ikeda M, Iwata N, Okahisa Y, Takaki M, Sakamoto S, Someya T, Egawa J, Usami M, Kodaira M, Yoshimi A, Oya-Ito T, Aleksic B, Ohno K, Ozaki N. Rare loss of function mutations in N-methyl-D-aspartate glutamate receptors and their contributions to schizophrenia susceptibility. Transl Psychiatry 2018; 8:12. [PMID: 29317596 PMCID: PMC5802496 DOI: 10.1038/s41398-017-0061-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/10/2017] [Accepted: 10/26/2017] [Indexed: 12/27/2022] Open
Abstract
In schizophrenia (SCZ) and autism spectrum disorder (ASD), the dysregulation of glutamate transmission through N-methyl-D-aspartate receptors (NMDARs) has been implicated as a potential etiological mechanism. Previous studies have accumulated evidence supporting NMDAR-encoding genes' role in etiology of SCZ and ASD. We performed a screening study for exonic regions of GRIN1, GRIN2A, GRIN2C, GRIN2D, GRIN3A, and GRIN3B, which encode NMDAR subunits, in 562 participates (370 SCZ and 192 ASD). Forty rare variants were identified including 38 missense, 1 frameshift mutation in GRIN2C and 1 splice site mutation in GRIN2D. We conducted in silico analysis for all variants and detected seven missense variants with deleterious prediction. De novo analysis was conducted if pedigree samples were available. The splice site mutation in GRIN2D is predicted to result in intron retention by minigene assay. Furthermore, the frameshift mutation in GRIN2C and splice site mutation in GRIN2D were genotyped in an independent sample set comprising 1877 SCZ cases, 382 ASD cases, and 2040 controls. Both of them were revealed to be singleton. Our study gives evidence in support of the view that ultra-rare variants with loss of function (frameshift, nonsense or splice site) in NMDARs genes may contribute to possible risk of SCZ.
Collapse
Affiliation(s)
- Yanjie Yu
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yingni Lin
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuto Takasaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Chenyao Wang
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroki Kimura
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Jingrui Xing
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Kanako Ishizuka
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Miho Toyama
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Itaru Kushima
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Institute for Advanced Research, Nagoya University, Nagoya, Japan
| | - Daisuke Mori
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Brain and Mind Research Center, Nagoya University, Nagoya, Japan
| | - Yuko Arioka
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Japan
| | - Yota Uno
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Laboratory for Psychiatric and Molecular Neuroscience, McLean Hospital, Belmont, MA, 02478, USA
| | - Tomoko Shiino
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukako Nakamura
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Okada
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mako Morikawa
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masashi Ikeda
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Nakao Iwata
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Yuko Okahisa
- Department of Neuropsychiatry Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Manabu Takaki
- Department of Neuropsychiatry Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinji Sakamoto
- Department of Neuropsychiatry Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toshiyuki Someya
- Department of Psychiatry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Jun Egawa
- Department of Psychiatry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Masahide Usami
- Department of Child and Adolescent Psychiatry Kohnodai Hospital, National Center for Global Health and Medicine, Tokyo, Japan
| | - Masaki Kodaira
- Department of Child and Adolescent Psychiatry Kohnodai Hospital, National Center for Global Health and Medicine, Tokyo, Japan
| | - Akira Yoshimi
- Division of Clinical Sciences and Neuropsychopharmacology, Faculty and Graduate School of Pharmacy, Meijo University, Nagoya, Japan
| | - Tomoko Oya-Ito
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Nutrition, Shubun University, Ichinomiya, Japan
| | - Branko Aleksic
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
19
|
Pratt JA, Morris B, Dawson N. Deconstructing Schizophrenia: Advances in Preclinical Models for Biomarker Identification. Curr Top Behav Neurosci 2018; 40:295-323. [PMID: 29721851 DOI: 10.1007/7854_2018_48] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Schizophrenia is considered to develop as a consequence of genetic and environmental factors impacting on brain neural systems and circuits during vulnerable neurodevelopmental periods, thereby resulting in symptoms in early adulthood. Understanding of the impact of schizophrenia risk factors on brain biology and behaviour can help in identifying biologically relevant pathways that are attractive for informing clinical studies and biomarker development. In this chapter, we emphasize the importance of adopting a reciprocal forward and reverse translation approach that is iteratively updated when additional new information is gained, either preclinically or clinically, for offering the greatest opportunity for discovering panels of biomarkers for the diagnosis, prognosis and treatment of schizophrenia. Importantly, biomarkers for identifying those at risk may inform early intervention strategies prior to the development of schizophrenia.Given the emerging nature of this approach in the field, this review will highlight recent research of preclinical biomarkers in schizophrenia that show the most promise for informing clinical needs with an emphasis on relevant imaging, electrophysiological, cognitive behavioural and biochemical modalities. The implementation of this reciprocal translational approach is exemplified firstly by the production and characterization of preclinical models based on the glutamate hypofunction hypothesis, genetic and environmental risk factors for schizophrenia (reverse translation), and then the recent clinical recognition of the thalamic reticular thalamus (TRN) as an important locus of brain dysfunction in schizophrenia as informed by preclinical findings (forward translation).
Collapse
Affiliation(s)
- Judith A Pratt
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK.
| | - Brian Morris
- Institute of Neuroscience and Psychology, College of Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Neil Dawson
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, UK
| |
Collapse
|
20
|
Fernandez LMJ, Pellegrini C, Vantomme G, Béard E, Lüthi A, Astori S. Cortical afferents onto the nucleus Reticularis thalami promote plasticity of low-threshold excitability through GluN2C-NMDARs. Sci Rep 2017; 7:12271. [PMID: 28947779 PMCID: PMC5612942 DOI: 10.1038/s41598-017-12552-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/11/2017] [Indexed: 12/22/2022] Open
Abstract
Thalamus and cortex represent a highly integrated processing unit that elaborates sensory representations. Interposed between cortex and thalamus, the nucleus Reticularis thalami (nRt) receives strong cortical glutamatergic input and mediates top-down inhibitory feedback to thalamus. Despite growing appreciation that the nRt is integral for thalamocortical functions from sleep to attentional wakefulness, we still face considerable gaps in the synaptic bases for cortico-nRt communication and plastic regulation. Here, we examined modulation of nRt excitability by cortical synaptic drive in Ntsr1-Cre x ChR2tg/+ mice expressing Channelrhodopsin2 in layer 6 corticothalamic cells. We found that cortico-nRt synapses express a major portion of NMDA receptors containing the GluN2C subunit (GluN2C-NMDARs). Upon repetitive photoactivation (10 Hz trains), GluN2C-NMDARs induced a long-term increase in nRt excitability involving a potentiated recruitment of T-type Ca2+ channels. In anaesthetized mice, analogous stimulation of cortical afferents onto nRt produced long-lasting changes in cortical local field potentials (LFPs), with delta oscillations being augmented at the expense of slow oscillations. This shift in LFP spectral composition was sensitive to NMDAR blockade in the nRt. Our data reveal a novel mechanism involving plastic modification of synaptically recruited T-type Ca2+ channels and nRt bursting and indicate a critical role for GluN2C-NMDARs in thalamocortical rhythmogenesis.
Collapse
Affiliation(s)
- Laura M J Fernandez
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
| | - Chiara Pellegrini
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
| | - Gil Vantomme
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
| | - Elidie Béard
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland
| | - Anita Lüthi
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland.
| | - Simone Astori
- Department of Fundamental Neurosciences, University of Lausanne, 1005, Lausanne, Switzerland. .,Brain Mind Institute, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland.
| |
Collapse
|
21
|
Noli B, Sanna F, Brancia C, D'Amato F, Manconi B, Vincenzoni F, Messana I, Melis MR, Argiolas A, Ferri GL, Cocco C. Profiles of VGF Peptides in the Rat Brain and Their Modulations after Phencyclidine Treatment. Front Cell Neurosci 2017. [PMID: 28626390 PMCID: PMC5454051 DOI: 10.3389/fncel.2017.00158] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
From the VGF precursor protein originate several low molecular weight peptides, whose distribution in the brain and blood circulation is not entirely known. Among the VGF peptides, those containing the N-terminus portion were altered in the cerebro-spinal fluid (CSF) and hypothalamus of schizophrenia patients. "Hence, we aimed to better investigate the involvement of the VGF peptides in schizophrenia by studying their localization in the brain regions relevant for the disease, and revealing their possible modulations in response to certain neuronal alterations occurring in schizophrenia". We produced antibodies against different VGF peptides encompassing the N-terminus, but also C-terminus-, TLQP-, GGGE- peptide sequences, and the so named NERP-3 and -4. These antibodies were used to carry out specific ELISA and immunolocalization studies while mass spectrometry (MS) analysis was also performed to recognize the intact brain VGF fragments. We used a schizophrenia rat model, in which alterations in the prepulse inhibition (PPI) of the acoustic startle response occurred after PCP treatment. In normal rats, all the VGF peptides studied were distributed in the brain areas examined including hypothalamus, prefrontal cortex, hippocampus, accumbens and amygdaloid nuclei and also in the plasma. By liquid chromatography-high resolution mass, we identified different intact VGF peptide fragments, including those encompassing the N-terminus and the NERPs. PCP treatment caused behavioral changes that closely mimic schizophrenia, estimated by us as a disruption of PPI of the acoustic startle response. The PCP treatment also induced selective changes in the VGF peptide levels within certain brain areas. Indeed, an increase in VGF C-terminus and TLQP peptides was revealed in the prefrontal cortex (p < 0.01) where they were localized within parvoalbumin and tyrosine hydroxylase (TH) containing neurons, respectively. Conversely, in the nucleus accumbens, PCP treatment produced a down-regulation in the levels of VGF C-terminus-, N-terminus- and GGGE- peptides (p < 0.01), expressed in GABAergic- (C-terminus/GGGE) and somatostatin- (N-terminus) neurons. These results confirm that VGF peptides are widely distributed in the brain and modulated in specific areas involved in schizophrenia.
Collapse
Affiliation(s)
- Barbara Noli
- Neuro-Endocrine-Fluorecence (NEF) Laboratory, Department of Biomedical Sciences, University of CagliariMonserrato, Italy
| | - Fabrizio Sanna
- Neuropsychobiology Laboratory, Department of Biomedical Sciences, University of CagliariMonserrato, Italy
| | - Carla Brancia
- Neuro-Endocrine-Fluorecence (NEF) Laboratory, Department of Biomedical Sciences, University of CagliariMonserrato, Italy
| | - Filomena D'Amato
- Neuro-Endocrine-Fluorecence (NEF) Laboratory, Department of Biomedical Sciences, University of CagliariMonserrato, Italy
| | - Barbara Manconi
- Department of Life and Environmental Sciences, University of CagliariMonserrato, Italy
| | - Federica Vincenzoni
- Institute of Biochemistry and Clinical Biochemistry, Catholic UniversityRome, Italy
| | - Irene Messana
- Institute of Chemistry of the Molecular Recognition, National Research Council (CNR)Rome, Italy
| | - Maria R Melis
- Neuropsychobiology Laboratory, Department of Biomedical Sciences, University of CagliariMonserrato, Italy
| | - Antonio Argiolas
- Neuropsychobiology Laboratory, Department of Biomedical Sciences, University of CagliariMonserrato, Italy
| | - Gian-Luca Ferri
- Neuro-Endocrine-Fluorecence (NEF) Laboratory, Department of Biomedical Sciences, University of CagliariMonserrato, Italy
| | - Cristina Cocco
- Neuro-Endocrine-Fluorecence (NEF) Laboratory, Department of Biomedical Sciences, University of CagliariMonserrato, Italy
| |
Collapse
|
22
|
Yasuda K, Hayashi Y, Yoshida T, Kashiwagi M, Nakagawa N, Michikawa T, Tanaka M, Ando R, Huang A, Hosoya T, McHugh TJ, Kuwahara M, Itohara S. Schizophrenia-like phenotypes in mice with NMDA receptor ablation in intralaminar thalamic nucleus cells and gene therapy-based reversal in adults. Transl Psychiatry 2017; 7:e1047. [PMID: 28244984 PMCID: PMC5545645 DOI: 10.1038/tp.2017.19] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 12/30/2016] [Indexed: 01/09/2023] Open
Abstract
In understanding the mechanism of schizophrenia pathogenesis, a significant finding is that drug abuse of phencyclidine or its analog ketamine causes symptoms similar to schizophrenia. Such drug effects are triggered even by administration at post-adolescent stages. Both drugs are N-methyl-d-aspartate receptor (NMDAR) antagonists, leading to a major hypothesis that glutamate hypofunction underlies schizophrenia pathogenesis. The precise region that depends on NMDAR function, however, is unclear. Here, we developed a mouse strain in which NMDARs in the intralaminar thalamic nuclei (ILN) were selectively disrupted. The mutant mice exhibited various schizophrenia-like phenotypes, including deficits in working memory, long-term spatial memory, and attention, as well as impulsivity, impaired prepulse inhibition, hyperlocomotion and hyperarousal. The electroencephalography analysis revealed that the mutant mice had a significantly reduced power in a wide range of frequencies including the alpha, beta and gamma bands, both during wake and rapid eye movement (REM) sleep, and a modest decrease of gamma power during non-REM sleep. Notably, restoring NMDARs in the adult ILN rescued some of the behavioral abnormalities. These findings suggest that NMDAR dysfunction in the ILN contributes to the pathophysiology of schizophrenia-related disorders. Furthermore, the reversal of inherent schizophrenia-like phenotypes in the adult mutant mice supports that ILN is a potential target site for a therapeutic strategy.
Collapse
Affiliation(s)
- K Yasuda
- Laboratory for Behavioral Genetics, RIKEN Brain Science Institute, Saitama, Japan,Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Science, The University of Tokyo, Tokyo, Japan
| | - Y Hayashi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki, Japan
| | - T Yoshida
- Laboratory for Behavioral Genetics, RIKEN Brain Science Institute, Saitama, Japan
| | - M Kashiwagi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki, Japan
| | - N Nakagawa
- Laboratory for Local Neuronal Circuits, RIKEN Brain Science Institute, Saitama, Japan
| | - T Michikawa
- Biotechnological Optics Research Team, RIKEN Center for Advanced Photonics, Saitama, Japan
| | - M Tanaka
- Laboratory for Neuron-Glia Circuitry, RIKEN Brain Science Institute, Saitama, Japan
| | - R Ando
- Laboratory for Behavioral Genetics, RIKEN Brain Science Institute, Saitama, Japan
| | - A Huang
- Laboratory for Circuit and Behavioral Physiology, RIKEN Brain Science Institute, Saitama, Japan
| | - T Hosoya
- Laboratory for Local Neuronal Circuits, RIKEN Brain Science Institute, Saitama, Japan
| | - T J McHugh
- Laboratory for Circuit and Behavioral Physiology, RIKEN Brain Science Institute, Saitama, Japan
| | - M Kuwahara
- Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Science, The University of Tokyo, Tokyo, Japan
| | - S Itohara
- Laboratory for Behavioral Genetics, RIKEN Brain Science Institute, Saitama, Japan,Laboratory for Behavioral Genetics, RIKEN Brain Science Institute, Neural Circuit Genetics Research Building 102k, 2-1 Wako, Saitama 351-0198, Japan. E-mail;
| |
Collapse
|
23
|
Li X, Lu F, Li W, Qin L, Yao Y, Ge X, Yu Q, Liang X, Zhao D, Li X, Zhang J. Edaravone injection reverses learning and memory deficits in a rat model of vascular dementia. Acta Biochim Biophys Sin (Shanghai) 2017; 49:83-89. [PMID: 27864280 DOI: 10.1093/abbs/gmw116] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 10/25/2016] [Indexed: 12/16/2022] Open
Abstract
Edaravone is a novel free radical scavenger that exerts neuroprotective effects by inhibiting endothelial injury and by ameliorating neuronal damage in brain ischemia. Recently, it was reported that edaravone could alleviate the pathology and cognitive deficits of Alzheimer's disease patients. However, its relevance to vascular dementia (VaD) is not clear. In this study, we partially occluded the bilateral carotid arteries of rats surgically to induce chronic cerebral hypoperfusion (CCH), a well-known rat model of VaD. Water maze and step-down inhibitory test were used to evaluate the memory deficit. The activities of superoxide dismutase (SOD) and lactate dehydrogenase (LDH), the content of malondialdehyde (MDA) and total reactive oxygen species were measured to evaluate the oxidative stress level. Western blot analysis was used to evaluate the synaptic protein expression. It was found that treatment with edaravone for a 5-week period was able to reverse both spatial and fear-memory deficits in rats with CCH. Edaravone significantly reduced the level of oxidative stress in the brains of rats with CCH by increasing SOD activity and decreasing the content of MDA, LDH, and total reactive oxygen species. Furthermore, edaravone treatment also restored the levels of multiple synaptic proteins in the hippocampi of rats with CCH. Our data provide direct evidence supporting the neuroprotective effects of edaravone in VaD. We propose that the alleviation of oxidative stress and restoration of synaptic proteins play important roles in neuroprotection.
Collapse
Affiliation(s)
- Xu Li
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Henan Provincial Cancer Hospital, Zhengzhou 450008, China
| | - Fen Lu
- Department of Neurology, Affiliated People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou 450003, China
| | - Wei Li
- Department of Neurology, Affiliated People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou 450003, China
| | - Lingzhi Qin
- Department of Neurology, Affiliated People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou 450003, China
| | - Yong Yao
- Department of Neurology, Affiliated People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou 450003, China
| | - Xuerong Ge
- Department of Neurology, Affiliated People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou 450003, China
| | - Qingkai Yu
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Henan Provincial Cancer Hospital, Zhengzhou 450008, China
| | - Xinliang Liang
- Department of Medical Development, Affiliated People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou 450003, China
| | - Dongmei Zhao
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Henan Provincial Cancer Hospital, Zhengzhou 450008, China
| | - Xiaohong Li
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Henan Provincial Cancer Hospital, Zhengzhou 450008, China
| | - Jiewen Zhang
- Department of Neurology, Affiliated People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou 450003, China
| |
Collapse
|
24
|
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
|
25
|
Verkhratsky A, Steardo L, Parpura V, Montana V. Translational potential of astrocytes in brain disorders. Prog Neurobiol 2016; 144:188-205. [PMID: 26386136 PMCID: PMC4794425 DOI: 10.1016/j.pneurobio.2015.09.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 09/03/2015] [Accepted: 09/08/2015] [Indexed: 12/11/2022]
Abstract
Fundamentally, all brain disorders can be broadly defined as the homeostatic failure of this organ. As the brain is composed of many different cells types, including but not limited to neurons and glia, it is only logical that all the cell types/constituents could play a role in health and disease. Yet, for a long time the sole conceptualization of brain pathology was focused on the well-being of neurons. Here, we challenge this neuron-centric view and present neuroglia as a key element in neuropathology, a process that has a toll on astrocytes, which undergo complex morpho-functional changes that can in turn affect the course of the disorder. Such changes can be grossly identified as reactivity, atrophy with loss of function and pathological remodeling. We outline the pathogenic potential of astrocytes in variety of disorders, ranging from neurotrauma, infection, toxic damage, stroke, epilepsy, neurodevelopmental, neurodegenerative and psychiatric disorders, Alexander disease to neoplastic changes seen in gliomas. We hope that in near future we would witness glial-based translational medicine with generation of deliverables for the containment and cure of disorders. We point out that such as a task will require a holistic and multi-disciplinary approach that will take in consideration the concerted operation of all the cell types in the brain.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Life Science, The University of Manchester, Manchester, UK
- Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Luca Steardo
- Department of Psychiatry, University of Naples, SUN, Largo Madonna delle Grazie, Naples, Italy
| | - Vladimir Parpura
- Department of Neurobiology, Center for Glial Biology in Medicine and Atomic Force Microscopy & Nanotechnology Laboratories, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Vedrana Montana
- Department of Biotechnology, University of Rijeka, Rijeka, Croatia
| |
Collapse
|
26
|
Cohen SM, Tsien RW, Goff DC, Halassa MM. The impact of NMDA receptor hypofunction on GABAergic neurons in the pathophysiology of schizophrenia. Schizophr Res 2015; 167:98-107. [PMID: 25583246 PMCID: PMC4724170 DOI: 10.1016/j.schres.2014.12.026] [Citation(s) in RCA: 173] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 11/25/2014] [Accepted: 12/18/2014] [Indexed: 02/07/2023]
Abstract
While the dopamine hypothesis has dominated schizophrenia research for several decades, more recent studies have highlighted the role of fast synaptic transmitters and their receptors in schizophrenia etiology. Here we review evidence that schizophrenia is associated with a reduction in N-methyl-d-aspartate receptor (NMDAR) function. By highlighting postmortem, neuroimaging and electrophysiological studies, we provide evidence for preferential disruption of GABAergic circuits in the context of NMDAR hypo-activity states. The functional relationship between NMDARs and GABAergic neurons is realized at the molecular, cellular, microcircuit and systems levels. A synthesis of findings across these levels explains how NMDA-mediated inhibitory dysfunction may lead to aberrant interactions among brain regions, accounting for key clinical features of schizophrenia. This synthesis of schizophrenia unifies observations from diverse fields and may help chart pathways for developing novel diagnostics and therapeutics.
Collapse
Affiliation(s)
- Samuel M. Cohen
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY 10016, USA
| | - Richard W. Tsien
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY 10016, USA
| | - Donald C. Goff
- Department of Psychiatry, NYU Langone Medical Center, 550 First Avenue, New York City, NY 10016, USA
,Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, NY 10962, USA
| | - Michael M. Halassa
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY 10016, USA
,Department of Psychiatry, NYU Langone Medical Center, 550 First Avenue, New York City, NY 10016, USA
,To whom correspondence should be addressed:
| |
Collapse
|
27
|
The role of the thalamus in schizophrenia from a neuroimaging perspective. Neurosci Biobehav Rev 2015; 54:57-75. [DOI: 10.1016/j.neubiorev.2015.01.013] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 12/19/2014] [Accepted: 01/12/2015] [Indexed: 02/06/2023]
|
28
|
Parlog A, Schlüter D, Dunay IR. Toxoplasma gondii-induced neuronal alterations. Parasite Immunol 2015; 37:159-70. [PMID: 25376390 DOI: 10.1111/pim.12157] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 10/31/2014] [Indexed: 12/13/2022]
Abstract
The zoonotic pathogen Toxoplasma gondii infects over 30% of the human population. The intracellular parasite can persist lifelong in the CNS within neurons modifying their function and structure, thus leading to specific behavioural changes of the host. In recent years, several in vitro studies and murine models have focused on the elucidation of these modifications. Furthermore, investigations of the human population have correlated Toxoplasma seropositivity with changes in neurological functions; however, the complex underlying mechanisms of the subtle behavioural alteration are still not fully understood. The parasites are able to induce direct modifications in the infected cells, for example by altering dopamine metabolism, by functionally silencing neurons as well as by hindering apoptosis. Moreover, indirect effects of the peripheral immune system and alterations of the immune status of the CNS, observed during chronic infection, might also contribute to changes in neuronal connectivity and synaptic plasticity. In this review, we will provide an overview and highlight recent advances, which describe changes in the neuronal function and morphology upon T. gondii infection.
Collapse
Affiliation(s)
- A Parlog
- Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke University, Magdeburg, Germany
| | | | | |
Collapse
|
29
|
Höflich A, Hahn A, Küblböck M, Kranz GS, Vanicek T, Windischberger C, Saria A, Kasper S, Winkler D, Lanzenberger R. Ketamine-Induced Modulation of the Thalamo-Cortical Network in Healthy Volunteers As a Model for Schizophrenia. Int J Neuropsychopharmacol 2015; 18:pyv040. [PMID: 25896256 PMCID: PMC4576520 DOI: 10.1093/ijnp/pyv040] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 04/03/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Schizophrenia has been associated with disturbances of thalamic functioning. In light of recent evidence suggesting a significant impact of the glutamatergic system on key symptoms of schizophrenia, we assessed whether modulation of the glutamatergic system via blockage of the N-methyl-D-aspartate (NMDA)-receptor might lead to changes of thalamic functional connectivity. METHODS Based on the ketamine model of psychosis, we investigated changes in cortico-thalamic functional connectivity by intravenous ketamine challenge during a 55-minute resting-state scan. Thirty healthy volunteers were measured with pharmacological functional magnetic resonance imaging using a double-blind, randomized, placebo-controlled, crossover design. RESULTS Functional connectivity analysis revealed significant ketamine-specific changes within the thalamus hub network, more precisely, an increase of cortico-thalamic connectivity of the somatosensory and temporal cortex. CONCLUSIONS Our results indicate that changes of thalamic functioning as described for schizophrenia can be partly mimicked by NMDA-receptor blockage. This adds substantial knowledge about the neurobiological mechanisms underlying the profound changes of perception and behavior during the application of NMDA-receptor antagonists.
Collapse
Affiliation(s)
- Anna Höflich
- Department of Psychiatry and Psychotherapy (Drs Höflich, Hahn, Kranz, Vanicek, Kasper, Winkler, and Lanzenberger), and MR Center of Excellence and Center for Biomedical Engineering and Physics (Mr Küblböck and Dr Windischberger), Medical University of Vienna, Vienna, Austria; Experimental Psychiatry Unit, Center for Psychiatry and Psychotherapy, Medical University of Innsbruck, Innsbruck, Austria (Dr Saria)
| | - Andreas Hahn
- Department of Psychiatry and Psychotherapy (Drs Höflich, Hahn, Kranz, Vanicek, Kasper, Winkler, and Lanzenberger), and MR Center of Excellence and Center for Biomedical Engineering and Physics (Mr Küblböck and Dr Windischberger), Medical University of Vienna, Vienna, Austria; Experimental Psychiatry Unit, Center for Psychiatry and Psychotherapy, Medical University of Innsbruck, Innsbruck, Austria (Dr Saria)
| | - Martin Küblböck
- Department of Psychiatry and Psychotherapy (Drs Höflich, Hahn, Kranz, Vanicek, Kasper, Winkler, and Lanzenberger), and MR Center of Excellence and Center for Biomedical Engineering and Physics (Mr Küblböck and Dr Windischberger), Medical University of Vienna, Vienna, Austria; Experimental Psychiatry Unit, Center for Psychiatry and Psychotherapy, Medical University of Innsbruck, Innsbruck, Austria (Dr Saria)
| | - Georg S Kranz
- Department of Psychiatry and Psychotherapy (Drs Höflich, Hahn, Kranz, Vanicek, Kasper, Winkler, and Lanzenberger), and MR Center of Excellence and Center for Biomedical Engineering and Physics (Mr Küblböck and Dr Windischberger), Medical University of Vienna, Vienna, Austria; Experimental Psychiatry Unit, Center for Psychiatry and Psychotherapy, Medical University of Innsbruck, Innsbruck, Austria (Dr Saria)
| | - Thomas Vanicek
- Department of Psychiatry and Psychotherapy (Drs Höflich, Hahn, Kranz, Vanicek, Kasper, Winkler, and Lanzenberger), and MR Center of Excellence and Center for Biomedical Engineering and Physics (Mr Küblböck and Dr Windischberger), Medical University of Vienna, Vienna, Austria; Experimental Psychiatry Unit, Center for Psychiatry and Psychotherapy, Medical University of Innsbruck, Innsbruck, Austria (Dr Saria)
| | - Christian Windischberger
- Department of Psychiatry and Psychotherapy (Drs Höflich, Hahn, Kranz, Vanicek, Kasper, Winkler, and Lanzenberger), and MR Center of Excellence and Center for Biomedical Engineering and Physics (Mr Küblböck and Dr Windischberger), Medical University of Vienna, Vienna, Austria; Experimental Psychiatry Unit, Center for Psychiatry and Psychotherapy, Medical University of Innsbruck, Innsbruck, Austria (Dr Saria)
| | - Alois Saria
- Department of Psychiatry and Psychotherapy (Drs Höflich, Hahn, Kranz, Vanicek, Kasper, Winkler, and Lanzenberger), and MR Center of Excellence and Center for Biomedical Engineering and Physics (Mr Küblböck and Dr Windischberger), Medical University of Vienna, Vienna, Austria; Experimental Psychiatry Unit, Center for Psychiatry and Psychotherapy, Medical University of Innsbruck, Innsbruck, Austria (Dr Saria)
| | - Siegfried Kasper
- Department of Psychiatry and Psychotherapy (Drs Höflich, Hahn, Kranz, Vanicek, Kasper, Winkler, and Lanzenberger), and MR Center of Excellence and Center for Biomedical Engineering and Physics (Mr Küblböck and Dr Windischberger), Medical University of Vienna, Vienna, Austria; Experimental Psychiatry Unit, Center for Psychiatry and Psychotherapy, Medical University of Innsbruck, Innsbruck, Austria (Dr Saria)
| | - Dietmar Winkler
- Department of Psychiatry and Psychotherapy (Drs Höflich, Hahn, Kranz, Vanicek, Kasper, Winkler, and Lanzenberger), and MR Center of Excellence and Center for Biomedical Engineering and Physics (Mr Küblböck and Dr Windischberger), Medical University of Vienna, Vienna, Austria; Experimental Psychiatry Unit, Center for Psychiatry and Psychotherapy, Medical University of Innsbruck, Innsbruck, Austria (Dr Saria)
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy (Drs Höflich, Hahn, Kranz, Vanicek, Kasper, Winkler, and Lanzenberger), and MR Center of Excellence and Center for Biomedical Engineering and Physics (Mr Küblböck and Dr Windischberger), Medical University of Vienna, Vienna, Austria; Experimental Psychiatry Unit, Center for Psychiatry and Psychotherapy, Medical University of Innsbruck, Innsbruck, Austria (Dr Saria).
| |
Collapse
|
30
|
Wijtenburg SA, Yang S, Fischer BA, Rowland LM. In vivo assessment of neurotransmitters and modulators with magnetic resonance spectroscopy: application to schizophrenia. Neurosci Biobehav Rev 2015; 51:276-95. [PMID: 25614132 PMCID: PMC4427237 DOI: 10.1016/j.neubiorev.2015.01.007] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 01/02/2015] [Accepted: 01/08/2015] [Indexed: 12/28/2022]
Abstract
In vivo measurement of neurotransmitters and modulators is now feasible with advanced proton magnetic resonance spectroscopy ((1)H MRS) techniques. This review provides a basic tutorial of MRS, describes the methods available to measure brain glutamate, glutamine, γ-aminobutyric acid, glutathione, N-acetylaspartylglutamate, glycine, and serine at magnetic field strengths of 3T or higher, and summarizes the neurochemical findings in schizophrenia. Overall, (1)H MRS holds great promise for producing biomarkers that can serve as treatment targets, prediction of disease onset, or illness exacerbation in schizophrenia and other brain diseases.
Collapse
Affiliation(s)
- S Andrea Wijtenburg
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, PO Box 21247, Baltimore, MD 21228, USA.
| | - Shaolin Yang
- Department of Psychiatry, University of Illinois at Chicago, 1601 W. Taylor Street, Suite 512, Chicago, IL 60612, USA; Department of Radiology, University of Illinois at Chicago, 1601 W. Taylor Street, Suite 512, Chicago, IL 60612, USA; Department of Bioengineering, University of Illinois at Chicago, 1601 W. Taylor Street, Suite 512, Chicago, IL 60612, USA
| | - Bernard A Fischer
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, PO Box 21247, Baltimore, MD 21228, USA; Veterans Affairs Capital Network (VISN 5) Mental Illness Research, Education, and Clinical Center (MIRECC), Department of Veterans Affairs, 10 N. Greene Street, Baltimore, MD 21201, USA
| | - Laura M Rowland
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, PO Box 21247, Baltimore, MD 21228, USA; Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, USA; Department of Psychology, University of Maryland, Baltimore County, Baltimore, MD 21228, USA
| |
Collapse
|
31
|
Dietrich-Muszalska A, Bartosz G, Sadowska-Bartosz I. The Role of Nitric Oxide and Nitrosative Stress in Schizophrenia. ACTA ACUST UNITED AC 2014. [DOI: 10.1007/978-1-4939-0440-2_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
32
|
Hadley JA, Nenert R, Kraguljac NV, Bolding MS, White DM, Skidmore FM, Visscher KM, Lahti AC. Ventral tegmental area/midbrain functional connectivity and response to antipsychotic medication in schizophrenia. Neuropsychopharmacology 2014; 39:1020-30. [PMID: 24165885 PMCID: PMC3924537 DOI: 10.1038/npp.2013.305] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 10/18/2013] [Accepted: 10/21/2013] [Indexed: 01/31/2023]
Abstract
Medication management in schizophrenia is a lengthy process, as the lack of clinical response can only be confirmed after at least 4 weeks of antipsychotic treatment at a therapeutic dose. Thus, there is a clear need for the discovery of biomarkers that have the potential to accelerate the management of treatment. Using resting-state functional MRI, we examined the functional connectivity of the ventral tegmental area (VTA), the origin of the mesocorticolimbic dopamine projections, in 21 healthy controls and 21 unmedicated patients with schizophrenia at baseline (pre-treatment) and after 1 week of treatment with the antipsychotic drug risperidone (1-week post-treatment). Group-level functional connectivity maps were obtained and group differences in connectivity were assessed on the groups' participant-level functional connectivity maps. We also examined the relationship between pre-treatment/1-week post-treatment functional connectivity and treatment response. Compared with controls, patients exhibited significantly reduced pre-treatment VTA/midbrain connectivity to multiple cortical and subcortical regions, including the dorsal anterior cingulate cortex (dACC) and thalamus. After 1 week of treatment, VTA/midbrain connectivity to bilateral regions of the thalamus was re-established. Pre-treatment VTA/midbrain connectivity strength to dACC was positively correlated with good response to a 6-week course of risperidone, whereas pre-treatment VTA/midbrain connectivity strength to the default mode network was negatively correlated. Our findings suggest that VTA/midbrain resting-state connectivity may be a useful biomarker for the prediction of treatment response.
Collapse
Affiliation(s)
- Jennifer A Hadley
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rodolphe Nenert
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nina V Kraguljac
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mark S Bolding
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David M White
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Frank M Skidmore
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kristina M Visscher
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Adrienne C Lahti
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA,Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, SC 501, 1530 3rd Ave South, Birmingham, AL 35294-0017, USA, Tel: +205 996 6776, Fax: +205 975 4879, E-mail:
| |
Collapse
|
33
|
McCullumsmith RE, Hammond JH, Shan D, Meador-Woodruff JH. Postmortem brain: an underutilized substrate for studying severe mental illness. Neuropsychopharmacology 2014; 39:65-87. [PMID: 24091486 PMCID: PMC3857666 DOI: 10.1038/npp.2013.239] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 07/30/2013] [Accepted: 08/02/2013] [Indexed: 02/08/2023]
Abstract
We propose that postmortem tissue is an underutilized substrate that may be used to translate genetic and/or preclinical studies, particularly for neuropsychiatric illnesses with complex etiologies. Postmortem brain tissues from subjects with schizophrenia have been extensively studied, and thus serve as a useful vehicle for illustrating the challenges associated with this biological substrate. Schizophrenia is likely caused by a combination of genetic risk and environmental factors that combine to create a disease phenotype that is typically not apparent until late adolescence. The complexity of this illness creates challenges for hypothesis testing aimed at understanding the pathophysiology of the illness, as postmortem brain tissues collected from individuals with schizophrenia reflect neuroplastic changes from a lifetime of severe mental illness, as well as treatment with antipsychotic medications. While there are significant challenges with studying postmortem brain, such as the postmortem interval, it confers a translational element that is difficult to recapitulate in animal models. On the other hand, data derived from animal models typically provide specific mechanistic and behavioral measures that cannot be generated using human subjects. Convergence of these two approaches has led to important insights for understanding molecular deficits and their causes in this illness. In this review, we discuss the problem of schizophrenia, review the common challenges related to postmortem studies, discuss the application of biochemical approaches to this substrate, and present examples of postmortem schizophrenia studies that illustrate the role of the postmortem approach for generating important new leads for understanding the pathophysiology of severe mental illness.
Collapse
Affiliation(s)
| | - John H Hammond
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama-Birmingham, Birmingham, AL, USA
| | - Dan Shan
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama-Birmingham, Birmingham, AL, USA
| | - James H Meador-Woodruff
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama-Birmingham, Birmingham, AL, USA
| |
Collapse
|
34
|
Rial D, Lara DR, Cunha RA. The Adenosine Neuromodulation System in Schizophrenia. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2014; 119:395-449. [DOI: 10.1016/b978-0-12-801022-8.00016-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
35
|
Abstract
Freely available automated MR image analysis techniques are being increasingly used to investigate neuroanatomical abnormalities in patients with neurological disorders. It is important to assess the specificity and validity of automated measurements of structure volumes with respect to reliable manual methods that rely on human anatomical expertise. The thalamus is widely investigated in many neurological and neuropsychiatric disorders using MRI, but thalamic volumes are notoriously difficult to quantify given the poor between-tissue contrast at the thalamic gray-white matter interface. In the present study we investigated the reliability of automatically determined thalamic volume measurements obtained using FreeSurfer software with respect to a manual stereological technique on 3D T1-weighted MR images obtained from a 3 T MR system. Further to demonstrating impressive consistency between stereological and FreeSurfer volume estimates of the thalamus in healthy subjects and neurological patients, we demonstrate that the extent of agreeability between stereology and FreeSurfer is equal to the agreeability between two human anatomists estimating thalamic volume using stereological methods. Using patients with juvenile myoclonic epilepsy as a model for thalamic atrophy, we also show that both automated and manual methods provide very similar ratios of thalamic volume loss in patients. This work promotes the use of FreeSurfer for reliable estimation of global volume in healthy and diseased thalami.
Collapse
|
36
|
The application of nonlinear Dynamic Causal Modelling for fMRI in subjects at high genetic risk of schizophrenia. Neuroimage 2013; 73:16-29. [PMID: 23384525 DOI: 10.1016/j.neuroimage.2013.01.063] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 01/17/2013] [Accepted: 01/22/2013] [Indexed: 01/22/2023] Open
Abstract
Nonlinear Dynamic Causal Modelling (DCM) for fMRI provides computational modelling of gating mechanisms at the neuronal population level. It allows for estimations of connection strengths with nonlinear modulation within task-dependent networks. This paper presents an application of nonlinear DCM in subjects at high familial risk of schizophrenia performing the Hayling Sentence Completion Task (HSCT). We analysed scans of 19 healthy controls and 46 subjects at high familial risk of schizophrenia, which included 26 high risk subjects without psychotic symptoms and 20 subjects with psychotic symptoms. The activity-dependent network consists of the intra parietal cortex (IPS), inferior frontal gyrus (IFG), middle temporal gyrus (MTG), anterior cingulate cortex (ACC) and the mediodorsal (MD) thalamus. The connections between the MD thalamus and the IFG were gated by the MD thalamus. We used DCM to investigate altered connection strength of these connections. Bayesian Model Selection (BMS) at the group and family level was used to compare the optimal bilinear and nonlinear models. Bayesian Model Averaging (BMA) was used to assess the connection strengths with the gating from the MD thalamus and the IFG. The nonlinear models provided the better explanation of the data. Furthermore, the BMA analysis showed significantly lower connection strength of the thalamocortical connection with nonlinear modulation from the MD thalamus in high risk subjects with psychotic symptoms and those who subsequently developed schizophrenia. These findings demonstrate that nonlinear DCM provides a method to investigate altered connectivity at the level of neural circuits. The reduced connection strength with thalamic gating may be a neurobiomarker implicated in the development of psychotic symptoms. This study suggests that nonlinear DCM could lead to new insights into functional and effective dysconnection at the network level in subjects at high familial risk.
Collapse
|
37
|
Snyder MA, Adelman AE, Gao WJ. Gestational methylazoxymethanol exposure leads to NMDAR dysfunction in hippocampus during early development and lasting deficits in learning. Neuropsychopharmacology 2013; 38:328-40. [PMID: 22968815 PMCID: PMC3527110 DOI: 10.1038/npp.2012.180] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The N-methyl-D-aspartate (NMDA) receptor has long been associated with learning and memory processes as well as diseased states, particularly in schizophrenia (SZ). Additionally, SZ is increasingly recognized as a neurodevelopmental disorder with cognitive impairments often preceding the onset of psychosis. However, the cause of these cognitive deficits and what initiates the pathological process is unknown. Growing evidence has implicated the glutamate system and, in particular, N-methyl-D-aspartate receptor (NMDAR) dysfunction in the pathophysiology of SZ. Yet, the vast majority of SZ-related research has focused on NMDAR function in adults leaving the role of NMDARs during development uncharacterized. We used the prenatal methylazoxymethanol acetate (MAM, E17) exposure model to determine the alterations of NMDAR protein levels and function, as well as associated cognitive deficits during development. We found that MAM-exposed animals have significantly altered NMDAR protein levels and function in the juvenile and adolescent hippocampus. Furthermore, these changes are associated with learning and memory deficits in the Morris Water Maze. Thus, in the prenatal MAM-exposure SZ model, NMDAR expression and function is altered during the critical period of hippocampal development. These changes may be involved in disease initiation and cognitive impairment in the early stage of SZ.
Collapse
Affiliation(s)
- Melissa A Snyder
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Alicia E Adelman
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Wen-Jun Gao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA,Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA, Tel: +215 991 8907, Fax: +215 843 9802, E-mail:
| |
Collapse
|
38
|
McCullumsmith RE, Hammond J, Funk A, Meador-Woodruff JH. Recent advances in targeting the ionotropic glutamate receptors in treating schizophrenia. Curr Pharm Biotechnol 2012; 13:1535-42. [PMID: 22283761 DOI: 10.2174/138920112800784899] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 05/08/2011] [Accepted: 05/24/2011] [Indexed: 12/28/2022]
Abstract
The treatment of schizophrenia has been focused on modulation of dopamine receptors for over 50 years. Recent developments have implicated other neurotransmitter systems in the pathophysiology of this illness. The discovery and characterization of glutamate receptors and their roles in the brain has lead to novel approaches for the treatment of schizophrenia. In this article, we review drugs that modulate ionotropic gluamate receptors and discuss their efficacy for the treatment of this often debilitating severe mental illness.
Collapse
Affiliation(s)
- Robert E McCullumsmith
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | | | | | | |
Collapse
|
39
|
Scaffolding proteins of the post-synaptic density contribute to synaptic plasticity by regulating receptor localization and distribution: relevance for neuropsychiatric diseases. Neurochem Res 2012; 38:1-22. [PMID: 22991141 DOI: 10.1007/s11064-012-0886-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 08/16/2012] [Accepted: 09/10/2012] [Indexed: 10/27/2022]
Abstract
Synaptic plasticity represents the long lasting activity-related strengthening or weakening of synaptic transmission, whose well-characterized types are the long term potentiation and depression. Despite this classical definition, however, the molecular mechanisms by which synaptic plasticity may occur appear to be extremely complex and various. The post-synaptic density (PSD) of glutamatergic synapses is a major site for synaptic plasticity processes and alterations of PSD members have been recently implicated in neuropsychiatric diseases where an impairment of synaptic plasticity has also been reported. Among PSD members, scaffolding proteins have been demonstrated to bridge surface receptors with their intracellular effectors and to regulate receptors distribution and localization both at surface membranes and within the PSD. This review will focus on the molecular physiology and pathophysiology of synaptic plasticity processes, which are tuned by scaffolding PSD proteins and their close related partners, through the modulation of receptor localization and distribution at post-synaptic sites. We suggest that, by regulating both the compartmentalization of receptors along surface membrane and their degradation as well as by modulating receptor trafficking into the PSD, postsynaptic scaffolding proteins may contribute to form distinct signaling micro-domains, whose efficacy in transmitting synaptic signals depends on the dynamic stability of the scaffold, which in turn is provided by relative amounts and post-translational modifications of scaffolding members. The putative relevance for neuropsychiatric diseases and possible pathophysiological mechanisms are discussed in the last part of this work.
Collapse
|
40
|
Carty NC, Xu J, Kurup P, Brouillette J, Goebel-Goody SM, Austin DR, Yuan P, Chen G, Correa PR, Haroutunian V, Pittenger C, Lombroso PJ. The tyrosine phosphatase STEP: implications in schizophrenia and the molecular mechanism underlying antipsychotic medications. Transl Psychiatry 2012; 2:e137. [PMID: 22781170 PMCID: PMC3410627 DOI: 10.1038/tp.2012.63] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Glutamatergic signaling through N-methyl-D-aspartate receptors (NMDARs) is required for synaptic plasticity. Disruptions in glutamatergic signaling are proposed to contribute to the behavioral and cognitive deficits observed in schizophrenia (SZ). One possible source of compromised glutamatergic function in SZ is decreased surface expression of GluN2B-containing NMDARs. STEP(61) is a brain-enriched protein tyrosine phosphatase that dephosphorylates a regulatory tyrosine on GluN2B, thereby promoting its internalization. Here, we report that STEP(61) levels are significantly higher in the postmortem anterior cingulate cortex and dorsolateral prefrontal cortex of SZ patients, as well as in mice treated with the psychotomimetics MK-801 and phencyclidine (PCP). Accumulation of STEP(61) after MK-801 treatment is due to a disruption in the ubiquitin proteasome system that normally degrades STEP(61). STEP knockout mice are less sensitive to both the locomotor and cognitive effects of acute and chronic administration of PCP, supporting the functional relevance of increased STEP(61) levels in SZ. In addition, chronic treatment of mice with both typical and atypical antipsychotic medications results in a protein kinase A-mediated phosphorylation and inactivation of STEP(61) and, consequently, increased surface expression of GluN1/GluN2B receptors. Taken together, our findings suggest that STEP(61) accumulation may contribute to the pathophysiology of SZ. Moreover, we show a mechanistic link between neuroleptic treatment, STEP(61) inactivation and increased surface expression of NMDARs, consistent with the glutamate hypothesis of SZ.
Collapse
Affiliation(s)
- N C Carty
- Child Study Center, Yale University School of Medicine, New Haven, CT, USA
| | - J Xu
- Child Study Center, Yale University School of Medicine, New Haven, CT, USA
| | - P Kurup
- Child Study Center, Yale University School of Medicine, New Haven, CT, USA
| | - J Brouillette
- Child Study Center, Yale University School of Medicine, New Haven, CT, USA
| | - S M Goebel-Goody
- Child Study Center, Yale University School of Medicine, New Haven, CT, USA
| | - D R Austin
- Laboratory of Molecular Pathophysiology, Mood and Anxiety Disorders Research Program, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - P Yuan
- Laboratory of Molecular Pathophysiology, Mood and Anxiety Disorders Research Program, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - G Chen
- Laboratory of Molecular Pathophysiology, Mood and Anxiety Disorders Research Program, National Institute of Mental Health, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - P R Correa
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - V Haroutunian
- Department of Psychiatry, The Mount Sinai School of Medicine, New York, NY, USA
| | - C Pittenger
- Child Study Center, Yale University School of Medicine, New Haven, CT, USA,Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA,Department of Psychology, Yale University School of Medicine, New Haven, CT, USA
| | - P J Lombroso
- Child Study Center, Yale University School of Medicine, New Haven, CT, USA,Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA,Department of Neurobiology, Yale University School of Medicine, New Haven, CT, USA,Child Study Center, Yale University School of Medicine, P.O. Box 207900, New Haven, CT 06520-7900, USA. E-mail:
| |
Collapse
|
41
|
Guilarte TR, Opler M, Pletnikov M. Is lead exposure in early life an environmental risk factor for Schizophrenia? Neurobiological connections and testable hypotheses. Neurotoxicology 2012; 33:560-74. [PMID: 22178136 PMCID: PMC3647679 DOI: 10.1016/j.neuro.2011.11.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Revised: 11/30/2011] [Accepted: 11/30/2011] [Indexed: 02/06/2023]
Abstract
Schizophrenia is a devastating neuropsychiatric disorder of unknown etiology. There is general agreement in the scientific community that schizophrenia is a disorder of neurodevelopmental origin in which both genes and environmental factors come together to produce a schizophrenia phenotype later in life. The challenging questions have been which genes and what environmental factors? Although there is evidence that different chromosome loci and several genes impart susceptibility for schizophrenia; and epidemiological studies point to broad aspects of the environment, only recently there has been an interest in studying gene × environment interactions. Recent evidence of a potential association between prenatal lead (Pb(2+)) exposure and schizophrenia precipitated the search for plausible neurobiological connections. The most promising connection is that in schizophrenia and in developmental Pb(2+) exposure there is strong evidence for hypoactivity of the N-methyl-d-aspartate (NMDA) subtype of excitatory amino acid receptors as an underlying neurobiological mechanism in both conditions. A hypofunction of the NMDA receptor (NMDAR) complex during critical periods of development may alter neurobiological processes that are essential for brain growth and wiring, synaptic plasticity and cognitive and behavioral outcomes associated with schizophrenia. We also describe on-going proof of concept gene-environment interaction studies of early life Pb(2+) exposure in mice expressing the human mutant form of the disrupted in schizophrenia 1 (DISC-1) gene, a gene that is strongly associated with schizophrenia and allied mental disorders.
Collapse
Affiliation(s)
- Tomás R Guilarte
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY 10032, United States.
| | | | | |
Collapse
|
42
|
Demontis D, Nyegaard M, Buttenschøn HN, Hedemand A, Pedersen CB, Grove J, Flint TJ, Nordentoft M, Werge T, Hougaard DM, Sørensen KM, Yolken RH, Mors O, Børglum AD, Mortensen PB. Association of GRIN1 and GRIN2A-D with schizophrenia and genetic interaction with maternal herpes simplex virus-2 infection affecting disease risk. Am J Med Genet B Neuropsychiatr Genet 2011; 156B:913-22. [PMID: 21919190 DOI: 10.1002/ajmg.b.31234] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Accepted: 08/04/2011] [Indexed: 02/05/2023]
Abstract
N-methyl-D-aspartate (NMDA) receptors are very important for proper brain development and several lines of evidence support that hypofunction of the NMDA receptors are involved in the pathophysiology of schizophrenia. Gene variation and gene-environmental interactions involving the genes encoding the NMDA receptors are therefore likely to influence the risk of schizophrenia. The aim of this study was to determine (1) whether SNP variation in the genes (GRIN1, GRIN2A, GRIN2B, GRIN2C, and GRIN2D) encoding the NMDA receptor were associated with schizophrenia; (2) whether GRIN gene variation in the offspring interacted with maternal herpes simplex virus-2 (HSV-2) seropositivity during pregnancy influencing the risk of schizophrenia later in life. Individuals from three independently collected Danish case control samples were genotyped for 81 tagSNPs (in total 984 individuals diagnosed with schizophrenia and 1,500 control persons) and antibodies against maternal HSV-2 infection were measured in one of the samples (365 cases and 365 controls). Nine SNPs out of 30 in GRIN2B were significantly associated with schizophrenia. One SNP remained significant after Bonferroni correction (rs1806194, P(nominal) = 0.0008). Significant interaction between maternal HSV-2 seropositivity and GRIN2B genetic variation in the offspring were observed for seven SNPs and two remained significant after Bonferroni correction (rs1805539, P(nominal) = 0.0001 and rs1806205, P(nominal) = 0.0008). The significant associations and interactions were located at the 3' region of GRIN2B suggesting that genetic variation in this part of the gene may be involved in the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Ditte Demontis
- Department of Human Genetics, Aarhus University, Aarhus, Denmark.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Excitation, inhibition, local oscillations, or large-scale loops: what causes the symptoms of schizophrenia? Curr Opin Neurobiol 2011; 22:537-44. [PMID: 22079494 DOI: 10.1016/j.conb.2011.10.018] [Citation(s) in RCA: 199] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Revised: 10/13/2011] [Accepted: 10/20/2011] [Indexed: 01/23/2023]
Abstract
What causes the positive, negative, and cognitive symptoms of schizophrenia? The importance of circuits is underscored by the finding that no single gene contributes strongly to the disease. Thus, some circuit abnormality to which many proteins can contribute is the likely cause. There are several major hypotheses regarding the circuitry involved: first, a change in the balance of excitation/inhibition in the prefrontal cortex (PFC); second, abnormal EEG oscillations in the gamma range; third, an increase in theta/delta EEG power related to changes in the thalamus (particularly midline nuclei); fourth, hyperactivity in the hippocampus and consequent dopamine hyperfunction; and fifth, deficits in corollary discharge. Evidence for these hypotheses will be reviewed.
Collapse
|
44
|
Sodhi MS, Simmons M, McCullumsmith R, Haroutunian V, Meador-Woodruff JH. Glutamatergic gene expression is specifically reduced in thalamocortical projecting relay neurons in schizophrenia. Biol Psychiatry 2011; 70:646-54. [PMID: 21549355 PMCID: PMC3176961 DOI: 10.1016/j.biopsych.2011.02.022] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 01/28/2011] [Accepted: 02/22/2011] [Indexed: 11/28/2022]
Abstract
BACKGROUND Impairment of glutamate neurons that relay sensory and cognitive information from the medial dorsal thalamus to the dorsolateral prefrontal cortex and other cortical regions may contribute to the pathophysiology of schizophrenia. In this study, we have assessed the cell-specific expression of glutamatergic transcripts in the medial dorsal thalamus. METHODS We used laser capture microdissection to harvest two populations of medial dorsal thalamic cells, one enriched with glutamatergic relay neurons and the other with gamma-aminobutyric acidergic neurons and astroglia, from postmortem brains of subjects with schizophrenia (n = 14) and a comparison group (n = 20). Quantitative polymerase chain reaction of extracted RNA was used to assay gene expression in the different cell populations. RESULTS The transcripts encoding the ionotropic glutamate receptor subunits NR2D, GluR3, GluR6, GluR7, and the intracellular proteins GRIP1 and SynGAP1 were significantly decreased in relay neurons but not in the mixed glial and interneuron population in schizophrenia. CONCLUSIONS Our data suggest that reduced ionotropic glutamatergic expression occurs selectively in neurons, which give rise to the cortical projections of the medial dorsal thalamus in schizophrenia, rather than in thalamic cells that function locally. Our findings indicate that glutamatergic innervation is dysfunctional in the circuitry between the medial dorsal thalamus and cortex.
Collapse
Affiliation(s)
- Monsheel S. Sodhi
- Dept. Psychiatry and Behavioral Neurobiology, UAB, 1720 7 Avenue Sth. Birmingham, Alabama 35294-0019, USA,Dept. Pharmacy Practice and Center for Pharmaceutical Biotechnology, College of Pharmacy, 900 S Ashland Ave, Chicago, Illinois 60607-4067, USA,Correspondence should be addressed to: Monsheel Sodhi Ph.D., 900 S. Ashland Ave. Chicago IL 60607-4067, USA, Tel: +1-312-355-5949, Fax: +1-312-413-9303,
| | - Micah Simmons
- Dept. Psychiatry and Behavioral Neurobiology, UAB, 1720 7 Avenue Sth. Birmingham, Alabama 35294-0019, USA
| | - Robert McCullumsmith
- Dept. Psychiatry and Behavioral Neurobiology, UAB, 1720 7 Avenue Sth. Birmingham, Alabama 35294-0019, USA
| | - Vahram Haroutunian
- Dept. Psychiatry, The Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, New York 10029-6574, USA
| | - James H. Meador-Woodruff
- Dept. Psychiatry and Behavioral Neurobiology, UAB, 1720 7 Avenue Sth. Birmingham, Alabama 35294-0019, USA
| |
Collapse
|
45
|
Faludi G, Mirnics K. Synaptic changes in the brain of subjects with schizophrenia. Int J Dev Neurosci 2011; 29:305-9. [PMID: 21382468 DOI: 10.1016/j.ijdevneu.2011.02.013] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Accepted: 02/26/2011] [Indexed: 01/16/2023] Open
Abstract
Clinical, epidemiological, neuroimaging and postmortem data all suggest schizophrenia is a neurodevelopmental disorder, and that synaptic disturbances might play a critical role in developing the disease. In 1982, Feinberg proposed that the schizophrenia might arise as a result of abnormal synaptic pruning. His hypothesis has survived 40years of accumulated data, and we review the critical findings related to synaptic dysfunction of schizophrenia. While it is clear that synaptic disturbances are integral and important for understanding the pathophysiology of schizophrenia, it has also become obvious that synaptic disturbances cannot be studied and understood as an independent disease hallmark, but only as a part of a complex network of homeostatic events. Development, glial-neural interaction, changes in energy homeostasis, diverse genetic predisposition, neuroimmune processes and environmental influences all can tip the delicate homeostatic balance of the synaptic morphology and connectivity in a uniquely individual fashion, thus contributing to the emergence of the various symptoms of this devastating disorder. Finally, we argue that based on a predominant change in gene expression pattern we can broadly sub-stratify schizophrenia into "synaptic" "oligodendroglial", "metabolic" and "inflammatory" subclasses.
Collapse
Affiliation(s)
- Gábor Faludi
- Department of Psychiatry, Kútvölgyi Clinical Centre, Semmelweis University, Budapest, Hungary
| | | |
Collapse
|
46
|
Wang L, Kisaalita WS. Administration of BDNF/ginsenosides combination enhanced synaptic development in human neural stem cells. J Neurosci Methods 2011; 194:274-82. [DOI: 10.1016/j.jneumeth.2010.10.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2010] [Revised: 10/07/2010] [Accepted: 10/28/2010] [Indexed: 01/06/2023]
|
47
|
Kantrowitz JT, Javitt DC. N-methyl-d-aspartate (NMDA) receptor dysfunction or dysregulation: the final common pathway on the road to schizophrenia? Brain Res Bull 2010; 83:108-21. [PMID: 20417696 PMCID: PMC2941541 DOI: 10.1016/j.brainresbull.2010.04.006] [Citation(s) in RCA: 312] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Revised: 03/12/2010] [Accepted: 04/18/2010] [Indexed: 11/19/2022]
Abstract
Schizophrenia is a severe mental disorder associated with a characteristic constellation of symptoms and neurocognitive deficits. At present, etiological mechanisms remain relatively unknown, although multiple points of convergence have been identified over recent years. One of the primary convergence points is dysfunction of N-methyl-d-aspartate (NMDAR)-type glutamate receptors. Antagonists of NMDAR produce a clinical syndrome that closely resembles, and uniquely incorporates negative and cognitive symptoms of schizophrenia, along with the specific pattern of neurocognitive dysfunction seen in schizophrenia. Genetic polymorphisms involving NMDAR subunits, particularly the GRIN2B subunit have been described. In addition, polymorphisms have been described in modulatory systems involving the NMDAR, including the enzymes serine racemase and d-amino acid oxidase/G72 that regulate brain d-serine synthesis. Reductions in plasma and brain glycine, d-serine and glutathione levels have been described as well, providing potential mechanisms underlying NMDAR dysfunction. Unique characteristics of the NMDAR are described that may explain the characteristic pattern of symptoms and neurocognitive deficits observed in schizophrenia. Finally, the NMDAR complex represents a convergence point for potential new treatment approaches in schizophrenia aimed at correcting underlying abnormalities in synthesis and regulation of allosteric modulators, as well as more general potentiation of pre- and post-synaptic glutamatergic and NMDAR function.
Collapse
Affiliation(s)
- Joshua T Kantrowitz
- Schizophrenia Research Center, Nathan Kline Institute for Psychiatric Research/New York University School of Medicine, 140 Old Orangeburg Road, Orangeburg, NY 10962, United States
| | | |
Collapse
|
48
|
Scorza FA, Schmitt A, Cysneiros RM, Arida RM, Cavalheiro EA, Gattaz WF. Thalamic nuclear abnormalities as a contributory factor in sudden cardiac deaths among patients with schizophrenia. Clinics (Sao Paulo) 2010; 65:539-46. [PMID: 20535373 PMCID: PMC2882549 DOI: 10.1590/s1807-59322010000500012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2010] [Revised: 01/31/2010] [Accepted: 02/09/2010] [Indexed: 11/21/2022] Open
Abstract
Patients with schizophrenia have a two- to three-fold increased risk of premature death as compared to patients without this disease. It has been established that patients with schizophrenia are at a high risk of developing cardiovascular disease. Moreover, an important issue that has not yet been explored is a possible existence of a "cerebral" focus that could trigger sudden cardiac death in patients with schizophrenia. Along these lines, several structural and functional alterations in the thalamic complex are evident in patients with schizophrenia and have been correlated with the symptoms manifested by these patients. With regard to abnormalities on the cellular and molecular level, previous studies have shown that schizophrenic patients have fewer neuronal projections from the thalamus to the prefrontal cortex as well as a reduced number of neurons, a reduced volume of either the entire thalamus or its subnuclei, and abnormal glutamate signaling. According to the glutamate hypothesis of schizophrenia, hypofunctional corticostriatal and striatothalamic projections are directly involved in the pathophysiology of the disease. Animal and post-mortem studies have provided a large amount of evidence that links the sudden unexpected death in epilepsy (SUDEP) that occurs in patients with schizophrenia and epilepsy to thalamic changes. Based on the results of these prior studies, it is clear that further research regarding the relationship between the thalamus and sudden cardiac death is of vital importance.
Collapse
Affiliation(s)
- Fulvio A. Scorza
- Disciplina de Neurologia Experimental, Universidade Federal de São Paulo (UNIFESP) - São Paulo/SP, Brasil
| | - Andrea Schmitt
- Department of Psychiatry, University of Goettingen - Germany
| | - Roberta M. Cysneiros
- Programa de Pós-Graduação em Distúrbios do Desenvolvimento do Centro de Ciências Biológicas e da Saúde da Universidade Presbiteriana Mackenzie - São Paulo/SP, Brasil
| | - Ricardo M. Arida
- Departamento de Fisiologia, Universidade Federal de São Paulo (UNIFESP) - São Paulo/SP, Brasil
| | - Esper A. Cavalheiro
- Centro de Cirurgia de Epilepsia (CIREP), Departamento de Neurociências e Ciências do Comportamento, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo - Ribeirão Preto/SP, Brasil
| | - Wagner F. Gattaz
- Laboratório de Neurociência (LIM-27), Instituto de Psiquiatria, Faculdade de Medicina da Universidade de São Paulo - São Paulo/SP, Brasil.,
, Tel: 0049 551 39 10366
| |
Collapse
|
49
|
Araud T, Wonnacott S, Bertrand D. Associated proteins: The universal toolbox controlling ligand gated ion channel function. Biochem Pharmacol 2010; 80:160-9. [PMID: 20346921 DOI: 10.1016/j.bcp.2010.03.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 03/02/2010] [Accepted: 03/15/2010] [Indexed: 02/06/2023]
Abstract
Ligand gated ion channels are integral multimeric membrane proteins that can detect with high sensitivity the presence of a specific transmitter in the extracellular space and transduce this signal into an ion flux. While these receptors are widely expressed in the nervous system, their expression is not limited to neurons or their postsynaptic targets but extends to non-neuronal cells where they participate in many physiological responses. Cells have developed complex regulatory mechanisms allowing for the precise control and modulation of ligand gated ion channels. In this overview the roles of accessory subunits and associated proteins in these regulatory mechanisms are reviewed and their relevance illustrated by examples at different ligand gated ion channel types, with emphasis on nicotinic acetylcholine receptors. Dysfunction of ligand gated ion channels can result in neuromuscular, neurological or psychiatric disorders. A better understanding of the precise function of associated proteins and how they impact on ligand gated ion channels will provide new therapeutic opportunities for clinical intervention.
Collapse
|
50
|
Pakkenberg B, Scheel-Krüger J, Kristiansen LV. Schizophrenia; from structure to function with special focus on the mediodorsal thalamic prefrontal loop. Acta Psychiatr Scand 2009; 120:345-54. [PMID: 19807715 DOI: 10.1111/j.1600-0447.2009.01447.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To describe structural and biochemical evidence from postmortem brains that implicates the reciprocal connections between the mediodorsal thalamic nucleus and the prefrontal cortex in cognitive symptoms of schizophrenia. METHOD The estimation of the regional volumes and cell numbers was obtained using stereological methods. The biochemical analyses of molecular expression in postmortem brain involve quantitative measurement of transcripts and proteins by in-situ (RNA) or Western blot/autoradiography in brains from patients with schizophrenia and comparison subjects. RESULTS Stereological studies in postmortem brain from patients with schizophrenia have reported divergent and often opposing findings in the total number of neurons and volume of the mediodorsal (MD) thalamic nucleus, and to a lesser degree in its reciprocally associated areas of the prefrontal cortex. Similarly, quantitative molecular postmortem studies have found large inter-subject and between-study variance at both the transcript and protein levels for receptors and their interacting molecules of several neurotransmitter systems in these interconnected anatomical regions. Combined, large variation in stereological and molecular studies indicates a complex and heterogeneous involvement of the MD thalamic-prefrontal loop in schizophrenia. CONCLUSION Based on a considerable heterogeneity in patients suffering from schizophrenia, large variation in postmortem studies, including stereological and molecular postmortem studies of the MD thalamus and frontal cortex, might be expected and may in fact partly help to explain the variable endophenotypic traits associated with this severe psychiatric illness.
Collapse
Affiliation(s)
- B Pakkenberg
- Research Laboratory for Stereology and Neuroscience, Bispebjerg University Hospital, Copenhagen, Denmark.
| | | | | |
Collapse
|