1
|
Ruzicka WB, Mohammadi S, Fullard JF, Davila-Velderrain J, Subburaju S, Tso DR, Hourihan M, Jiang S, Lee HC, Bendl J, Voloudakis G, Haroutunian V, Hoffman GE, Roussos P, Kellis M. Single-cell multi-cohort dissection of the schizophrenia transcriptome. Science 2024; 384:eadg5136. [PMID: 38781388 DOI: 10.1126/science.adg5136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 07/21/2023] [Indexed: 05/25/2024]
Abstract
The complexity and heterogeneity of schizophrenia have hindered mechanistic elucidation and the development of more effective therapies. Here, we performed single-cell dissection of schizophrenia-associated transcriptomic changes in the human prefrontal cortex across 140 individuals in two independent cohorts. Excitatory neurons were the most affected cell group, with transcriptional changes converging on neurodevelopment and synapse-related molecular pathways. Transcriptional alterations included known genetic risk factors, suggesting convergence of rare and common genomic variants on neuronal population-specific alterations in schizophrenia. Based on the magnitude of schizophrenia-associated transcriptional change, we identified two populations of individuals with schizophrenia marked by expression of specific excitatory and inhibitory neuronal cell states. This single-cell atlas links transcriptomic changes to etiological genetic risk factors, contextualizing established knowledge within the human cortical cytoarchitecture and facilitating mechanistic understanding of schizophrenia pathophysiology and heterogeneity.
Collapse
Affiliation(s)
- W Brad Ruzicka
- Laboratory for Epigenomics in Human Psychopathology, McLean Hospital, Belmont, MA 02478, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Shahin Mohammadi
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - John F Fullard
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jose Davila-Velderrain
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Neurogenomics Research Center, Human Technopole, 20157 Milan, Italy
| | - Sivan Subburaju
- Laboratory for Epigenomics in Human Psychopathology, McLean Hospital, Belmont, MA 02478, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel Reed Tso
- Laboratory for Epigenomics in Human Psychopathology, McLean Hospital, Belmont, MA 02478, USA
| | - Makayla Hourihan
- Laboratory for Epigenomics in Human Psychopathology, McLean Hospital, Belmont, MA 02478, USA
| | - Shan Jiang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hao-Chih Lee
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jaroslav Bendl
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Georgios Voloudakis
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Vahram Haroutunian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mental Illness Research, Education and Clinical Centers, James J. Peters VA Medical Center, Bronx, NY 10468, USA
| | - Gabriel E Hoffman
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Panos Roussos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Neurogenomics Research Center, Human Technopole, 20157 Milan, Italy
| | - Manolis Kellis
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
2
|
Arime Y, Saitoh Y, Ishikawa M, Kamiyoshihara C, Uchida Y, Fujii K, Takao K, Akiyama K, Ohkawa N. Activation of prefrontal parvalbumin interneurons ameliorates working memory deficit even under clinically comparable antipsychotic treatment in a mouse model of schizophrenia. Neuropsychopharmacology 2024; 49:720-730. [PMID: 38049583 PMCID: PMC10876596 DOI: 10.1038/s41386-023-01769-z] [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: 05/01/2023] [Revised: 10/06/2023] [Accepted: 11/01/2023] [Indexed: 12/06/2023]
Abstract
One of the critical unmet medical needs in schizophrenia is the treatment for cognitive deficits. However, the neural circuit mechanisms of them remain unresolved. Previous studies utilizing animal models of schizophrenia did not consider the fact that patients with schizophrenia generally cannot discontinue antipsychotic medication due to the high risk of relapse. Here, we used multi-dimensional approaches, including histological analysis of the prelimbic cortex (PL), LC-MS/MS-based in vivo dopamine D2 receptor occupancy analysis for antipsychotics, in vivo calcium imaging, and behavioral analyses of mice using chemogenetics to investigate neural mechanisms and potential therapeutic strategies for working memory deficit in a chronic phencyclidine (PCP) mouse model of schizophrenia. Chronic PCP administration led to alterations in excitatory and inhibitory synapses, specifically in dendritic spines of pyramidal neurons, vesicular glutamate transporter 1 (VGLUT1) positive terminals, and parvalbumin (PV) positive GABAergic interneurons located in layer 2-3 of the PL. Continuous administration of olanzapine, which achieved a sustained therapeutic window of dopamine D2 receptor occupancy (60-80%) in the striatum, did not ameliorate these synaptic abnormalities and working memory deficit in the chronic PCP-treated mice. We demonstrated that chemogenetic activation of PV neurons in the PL, as confirmed by in vivo calcium imaging, ameliorated working memory deficit in this model even under clinically comparable olanzapine treatment which by itself inhibited only PCP-induced psychomotor hyperactivity. Our study suggests that targeting prefrontal PV neurons could be a promising therapeutic intervention for cognitive deficits in schizophrenia in combination with antipsychotic medication.
Collapse
Affiliation(s)
- Yosefu Arime
- Division for Memory and Cognitive Function, Research Center for Advanced Medical Science, Comprehensive Research Facilities for Advanced Medical Science, Dokkyo Medical University, Tochigi, Japan.
- Department of Biological Psychiatry and Neuroscience, Dokkyo Medical University School of Medicine, Tochigi, Japan.
| | - Yoshito Saitoh
- Division for Memory and Cognitive Function, Research Center for Advanced Medical Science, Comprehensive Research Facilities for Advanced Medical Science, Dokkyo Medical University, Tochigi, Japan
| | - Mikiko Ishikawa
- Division for Memory and Cognitive Function, Research Center for Advanced Medical Science, Comprehensive Research Facilities for Advanced Medical Science, Dokkyo Medical University, Tochigi, Japan
- Department of Biological Psychiatry and Neuroscience, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Chikako Kamiyoshihara
- Division for Memory and Cognitive Function, Research Center for Advanced Medical Science, Comprehensive Research Facilities for Advanced Medical Science, Dokkyo Medical University, Tochigi, Japan
| | - Yasuo Uchida
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Kazuki Fujii
- Department of Behavioral Physiology, Faculty of Medicine, University of Toyama, Toyama, Japan
- Life Science Research Center, University of Toyama, Toyama, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
| | - Keizo Takao
- Department of Behavioral Physiology, Faculty of Medicine, University of Toyama, Toyama, Japan
- Life Science Research Center, University of Toyama, Toyama, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
| | - Kazufumi Akiyama
- Department of Biological Psychiatry and Neuroscience, Dokkyo Medical University School of Medicine, Tochigi, Japan
- Kawada Hospital, Okayama, Japan
| | - Noriaki Ohkawa
- Division for Memory and Cognitive Function, Research Center for Advanced Medical Science, Comprehensive Research Facilities for Advanced Medical Science, Dokkyo Medical University, Tochigi, Japan.
| |
Collapse
|
3
|
Schoonover KE, Miller NE, Fish KN, Lewis DA. Scaling of smaller pyramidal neuron size and lower energy production in schizophrenia. Neurobiol Dis 2024; 191:106394. [PMID: 38176569 PMCID: PMC10898364 DOI: 10.1016/j.nbd.2023.106394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/21/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Dorsolateral prefrontal cortex (DLPFC) dysfunction in schizophrenia appears to reflect alterations in layer 3 pyramidal neurons (L3PNs), including smaller cell bodies and lower expression of mitochondrial energy production genes. However, prior somal size studies used biased strategies for identifying L3PNs, and somal size and levels of energy production markers have not been assessed in individual L3PNs. STUDY DESIGN We combined fluorescent in situ hybridization (FISH) of vesicular glutamate transporter 1 (VGLUT1) mRNA and immunohistochemical-labeling of NeuN to determine if the cytoplasmic distribution of VGLUT1 mRNA permits the unbiased identification and somal size quantification of L3PNs. Dual-label FISH for VGLUT1 mRNA and cytochrome C oxidase subunit 4I1 (COX4I1) mRNA, a marker of energy production, was used to assess somal size and COX4I1 transcript levels in individual DLPFC L3PNs from schizophrenia (12 males; 2 females) and unaffected comparison (13 males; 1 female) subjects. STUDY RESULTS Measures of L3PN somal size with NeuN immunohistochemistry or VGLUT1 mRNA provided nearly identical results (ICC = 0.96, p < 0.0001). Mean somal size of VGLUT1-identified L3PNs was 8.7% smaller (p = 0.004) and mean COX4I1 mRNA levels per L3PN were 16.7% lower (p = 0.01) in schizophrenia. These measures were correlated across individual L3PNs in both subject groups (rrm = 0.81-0.86). CONCLUSIONS This preliminary study presents a novel method for combining unbiased neuronal identification with quantitative assessments of somal size and mRNA levels. We replicated findings of smaller somal size and lower COX4I1 mRNA levels in DLPFC L3PNs in schizophrenia. The normal scaling of COX4I1 mRNA levels with somal size in schizophrenia suggests that lower markers of energy production are secondary to L3PN morphological alterations in the illness.
Collapse
Affiliation(s)
- Kirsten E Schoonover
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Department of Psychiatry Biomedical Science Tower, W1653 3811 O'Hara Street Pittsburgh, PA 15213, United States of America
| | - Nora E Miller
- Department of Neuroscience, Dietrich School of Arts and Sciences, University of Pittsburgh, Department of Psychiatry, Biomedical Science Tower W1653 3811 O'Hara Street Pittsburgh, PA 15213, United States of America
| | - Kenneth N Fish
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Department of Psychiatry Biomedical Science Tower, W1653 3811 O'Hara Street Pittsburgh, PA 15213, United States of America
| | - David A Lewis
- Translational Neuroscience Program, Department of Psychiatry, School of Medicine, University of Pittsburgh, Department of Psychiatry Biomedical Science Tower, W1653 3811 O'Hara Street Pittsburgh, PA 15213, United States of America.
| |
Collapse
|
4
|
Inhibition of Vesicular Glutamate Transporters (VGLUTs) with Chicago Sky Blue 6B Before Focal Cerebral Ischemia Offers Neuroprotection. Mol Neurobiol 2023; 60:3130-3146. [PMID: 36802054 PMCID: PMC10122628 DOI: 10.1007/s12035-023-03259-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 02/07/2023] [Indexed: 02/21/2023]
Abstract
Brain ischemia is one of the leading causes of death and long-term disability in the world. Interruption of the blood supply to the brain is a direct stimulus for many pathological events. The massive vesicular release of glutamate (Glu) after ischemia onset induces excitotoxicity, which is a potent stress on neurons. Loading of presynaptic vesicles with Glu is the first step of glutamatergic neurotransmission. Vesicular glutamate transporters 1, 2, and 3 (VGLUT1, 2, and 3) are the main players involved in filling presynaptic vesicles with Glu. VGLUT1 and VGLUT2 are expressed mainly in glutamatergic neurons. Therefore, the possibility of pharmacological modulation to prevent ischemia-related brain damage is attractive. In this study, we aimed to determine the effect of focal cerebral ischemia on the spatiotemporal expression of VGLUT1 and VGLUT2 in rats. Next, we investigated the influence of VGLUT inhibition with Chicago Sky Blue 6B (CSB6B) on Glu release and stroke outcome. The effect of CSB6B pretreatment on infarct volume and neurological deficit was compared with a reference model of ischemic preconditioning. The results of this study indicate that ischemia upregulated the expression of VGLUT1 in the cerebral cortex and in the dorsal striatum 3 days after ischemia onset. The expression of VGLUT2 was elevated in the dorsal striatum and in the cerebral cortex 24 h and 3 days after ischemia, respectively. Microdialysis revealed that pretreatment with CSB6B significantly reduced the extracellular Glu concentration. Altogether, this study shows that inhibition of VGLUTs might be a promising therapeutic strategy for the future.
Collapse
|
5
|
Bauminger H, Gaisler-Salomon I. Beyond NMDA Receptors: Homeostasis at the Glutamate Tripartite Synapse and Its Contributions to Cognitive Dysfunction in Schizophrenia. Int J Mol Sci 2022; 23:8617. [PMID: 35955750 PMCID: PMC9368772 DOI: 10.3390/ijms23158617] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/30/2022] [Accepted: 08/01/2022] [Indexed: 02/01/2023] Open
Abstract
Cognitive deficits are core symptoms of schizophrenia but remain poorly addressed by dopamine-based antipsychotic medications. Glutamate abnormalities are implicated in schizophrenia-related cognitive deficits. While the role of the NMDA receptor has been extensively studied, less attention was given to other components that control glutamate homeostasis. Glutamate dynamics at the tripartite synapse include presynaptic and postsynaptic components and are tightly regulated by neuron-astrocyte crosstalk. Here, we delineate the role of glutamate homeostasis at the tripartite synapse in schizophrenia-related cognitive dysfunction. We focus on cognitive domains that can be readily measured in humans and rodents, i.e., working memory, recognition memory, cognitive flexibility, and response inhibition. We describe tasks used to measure cognitive function in these domains in humans and rodents, and the relevance of glutamate alterations in these domains. Next, we delve into glutamate tripartite synaptic components and summarize findings that implicate the relevance of these components to specific cognitive domains. These collective findings indicate that neuron-astrocyte crosstalk at the tripartite synapse is essential for cognition, and that pre- and postsynaptic components play a critical role in maintaining glutamate homeostasis and cognitive well-being. The contribution of these components to cognitive function should be considered in order to better understand the role played by glutamate signaling in cognition and develop efficient pharmacological treatment avenues for schizophrenia treatment-resistant symptoms.
Collapse
Affiliation(s)
- Hagar Bauminger
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa 3498838, Israel;
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa 3498838, Israel
| | - Inna Gaisler-Salomon
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa 3498838, Israel;
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa 3498838, Israel
| |
Collapse
|
6
|
Modified climbing fiber/Purkinje cell synaptic connectivity in the cerebellum of the neonatal phencyclidine model of schizophrenia. Proc Natl Acad Sci U S A 2022; 119:e2122544119. [PMID: 35588456 PMCID: PMC9173783 DOI: 10.1073/pnas.2122544119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Synaptogenesis and neural network remodeling are at their maximum during the perinatal period of human brain development. Perturbations of this highly sensitive stage might underlie the etiology of neurodevelopmental disorders. Subchronic neonatal administration of phencyclidine, a drug of abuse, has been used to model schizophrenia in rodents. In this model, we found specific long-term synaptic changes in Purkinje cells and transient gene expression changes in the cerebellum. While transient increased neuronal activity in the cerebellum, induced using chemogenetics, reproduces some phencyclidine-induced molecular changes, it is insufficient to reproduce the long-term synaptic effects. Our results show the complex mechanism of action of phencyclidine on the development of neuronal connectivity and further highlight the potential contribution of cerebellar defects in psychiatric diseases. Environmental perturbations during the first years of life are a major factor in psychiatric diseases. Phencyclidine (PCP), a drug of abuse, has psychomimetic effects, and neonatal subchronic administration of PCP in rodents leads to long-term behavioral changes relevant for schizophrenia. The cerebellum is increasingly recognized for its role in diverse cognitive functions. However, little is known about potential cerebellar changes in models of schizophrenia. Here, we analyzed the characteristics of the cerebellum in the neonatal subchronic PCP model. We found that, while the global cerebellar cytoarchitecture and Purkinje cell spontaneous spiking properties are unchanged, climbing fiber/Purkinje cell synaptic connectivity is increased in juvenile mice. Neonatal subchronic administration of PCP is accompanied by increased cFos expression, a marker of neuronal activity, and transient modification of the neuronal surfaceome in the cerebellum. The largest change observed is the overexpression of Ctgf, a gene previously suggested as a biomarker for schizophrenia. This neonatal increase in Ctgf can be reproduced by increasing neuronal activity in the cerebellum during the second postnatal week using chemogenetics. However, it does not lead to increased climbing fiber/Purkinje cell connectivity in juvenile mice, showing the complexity of PCP action. Overall, our study shows that administration of the drug of abuse PCP during the developmental period of intense cerebellar synaptogenesis and circuit remodeling has long-term and specific effects on Purkinje cell connectivity and warrants the search for this type of synaptic changes in psychiatric diseases.
Collapse
|
7
|
Wang SS, Zhou CX, Elsheikha HM, He JJ, Zou FC, Zheng WB, Zhu XQ, Zhao GH. Temporal transcriptomic changes in long non-coding RNAs and messenger RNAs involved in the host immune and metabolic response during Toxoplasma gondii lytic cycle. Parasit Vectors 2022; 15:22. [PMID: 35012632 PMCID: PMC8750853 DOI: 10.1186/s13071-021-05140-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/21/2021] [Indexed: 02/07/2023] Open
Abstract
Background Long non-coding RNAs (lncRNAs) are important regulators of various biological and pathological processes, in particular the inflammatory response by modulating the transcriptional control of inflammatory genes. However, the role of lncRNAs in regulating the immune and inflammatory responses during infection with the protozoan parasite Toxoplasma gondii remains largely unknown. Methods We performed a longitudinal RNA sequencing analysis of human foreskin fibroblast (HFF) cells infected by T. gondii to identify differentially expressed long non-coding RNAs (lncRNAs) and messenger RNAs (mRNAs), and dysregulated pathways over the course of T. gondii lytic cycle. The transcriptome data were validated by qRT-PCR. Results RNA sequencing revealed significant transcriptional changes in the infected HFFs. A total of 697, 1234, 1499, 873, 1466, 561, 676 and 716 differentially expressed lncRNAs (DElncRNAs), and 636, 1266, 1843, 2303, 3022, 1757, 3088 and 2531 differentially expressed mRNAs (DEmRNAs) were identified at 1.5, 3, 6, 9, 12, 24, 36 and 48 h post-infection, respectively. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis of DElncRNAs and DEmRNAs revealed that T. gondii infection altered the expression of genes involved in the regulation of host immune response (e.g., cytokine–cytokine receptor interaction), receptor signaling (e.g., NOD-like receptor signaling pathway), disease (e.g., Alzheimer's disease), and metabolism (e.g., fatty acid degradation). Conclusions These results provide novel information for further research on the role of lncRNAs in immune regulation of T. gondii infection. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13071-021-05140-3.
Collapse
Affiliation(s)
- Sha-Sha Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.,State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, Gansu, China
| | - Chun-Xue Zhou
- Department of Pathogen Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250100, Shandong, China
| | - Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Loughborough, LE12 5RD, UK
| | - Jun-Jun He
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, Gansu, China.,Key Laboratory of Veterinary Public Health of Higher Education of Yunnan Province, College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, Yunnan, China
| | - Feng-Cai Zou
- Key Laboratory of Veterinary Public Health of Higher Education of Yunnan Province, College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, Yunnan, China
| | - Wen-Bin Zheng
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Xing-Quan Zhu
- Key Laboratory of Veterinary Public Health of Higher Education of Yunnan Province, College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, Yunnan, China. .,College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China.
| | - Guang-Hui Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
8
|
Pathological oligodendrocyte precursor cells revealed in human schizophrenic brains and trigger schizophrenia-like behaviors and synaptic defects in genetic animal model. Mol Psychiatry 2022; 27:5154-5166. [PMID: 36131044 PMCID: PMC9763102 DOI: 10.1038/s41380-022-01777-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 01/19/2023]
Abstract
Although the link of white matter to pathophysiology of schizophrenia is documented, loss of myelin is not detected in patients at the early stages of the disease, suggesting that pathological evolution of schizophrenia may occur before significant myelin loss. Disrupted-in-schizophrenia-1 (DISC1) protein is highly expressed in oligodendrocyte precursor cells (OPCs) and regulates their maturation. Recently, DISC1-Δ3, a major DISC1 variant that lacks exon 3, has been identified in schizophrenia patients, although its pathological significance remains unknown. In this study, we detected in schizophrenia patients a previously unidentified pathological phenotype of OPCs exhibiting excessive branching. We replicated this phenotype by generating a mouse strain expressing DISC1-Δ3 gene in OPCs. We further demonstrated that pathological OPCs, rather than myelin defects, drive the onset of schizophrenic phenotype by hyperactivating OPCs' Wnt/β-catenin pathway, which consequently upregulates Wnt Inhibitory Factor 1 (Wif1), leading to the aberrant synaptic formation and neuronal activity. Suppressing Wif1 in OPCs rescues synaptic loss and behavioral disorders in DISC1-Δ3 mice. Our findings reveal the pathogenetic role of OPC-specific DISC1-Δ3 variant in the onset of schizophrenia and highlight the therapeutic potential of Wif1 as an alternative target for the treatment of this disease.
Collapse
|
9
|
Lindström SH, Sundberg SC, Larsson M, Andersson FK, Broman J, Granseth B. VGluT1 Deficiency Impairs Visual Attention and Reduces the Dynamic Range of Short-Term Plasticity at Corticothalamic Synapses. Cereb Cortex 2021; 30:1813-1829. [PMID: 31711131 PMCID: PMC7132919 DOI: 10.1093/cercor/bhz204] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/08/2019] [Accepted: 08/13/2019] [Indexed: 12/17/2022] Open
Abstract
The most common excitatory neurotransmitter in the central nervous system, glutamate, is loaded into synaptic vesicles by vesicular glutamate transporters (VGluTs). The primary isoforms, VGluT1 and 2, are expressed in complementary patterns throughout the brain and correlate with short-term synaptic plasticity. VGluT1 deficiency is observed in certain neurological disorders, and hemizygous (VGluT1+/−) mice display increased anxiety and depression, altered sensorimotor gating, and impairments in learning and memory. The synaptic mechanisms underlying these behavioral deficits are unknown. Here, we show that VGluT1+/− mice had decreased visual processing speeds during a sustained visual-spatial attention task. Furthermore, in vitro recordings of corticothalamic (CT) synapses revealed dramatic reductions in short-term facilitation, increased initial release probability, and earlier synaptic depression in VGluT1+/− mice. Our electron microscopy results show that VGluT1 concentration is reduced at CT synapses of hemizygous mice, but other features (such as vesicle number and active zone size) are unchanged. We conclude that VGluT1-haploinsuficiency decreases the dynamic range of gain modulation provided by CT feedback to the thalamus, and this deficiency contributes to the observed attentional processing deficit. We further hypothesize that VGluT1 concentration regulates release probability by applying a “brake” to an unidentified presynaptic protein that typically acts as a positive regulator of release.
Collapse
Affiliation(s)
- Sarah H Lindström
- Department of Clinical and Experimental Medicine, Division of Neurobiology, Linköping University, Linköping, 58185, Sweden
| | - Sofie C Sundberg
- Department of Clinical and Experimental Medicine, Division of Neurobiology, Linköping University, Linköping, 58185, Sweden
| | - Max Larsson
- Department of Clinical and Experimental Medicine, Division of Neurobiology, Linköping University, Linköping, 58185, Sweden
| | - Fredrik K Andersson
- Department of Clinical and Experimental Medicine, Division of Neurobiology, Linköping University, Linköping, 58185, Sweden
| | - Jonas Broman
- Department of Clinical and Experimental Medicine, Division of Neurobiology, Linköping University, Linköping, 58185, Sweden
| | - Björn Granseth
- Department of Clinical and Experimental Medicine, Division of Neurobiology, Linköping University, Linköping, 58185, Sweden
| |
Collapse
|
10
|
Schoonover KE, Kennedy WM, Roberts RC. Cortical copper transporter expression in schizophrenia: interactions of risk gene dysbindin-1. J Neural Transm (Vienna) 2021; 128:701-709. [PMID: 33890175 PMCID: PMC11000637 DOI: 10.1007/s00702-021-02333-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/28/2021] [Indexed: 10/21/2022]
Abstract
Schizophrenia susceptibility factor dysbindin-1 is associated with cognitive processes. Downregulated dysbindin-1 expression is associated with lower expression of copper transporters ATP7A and CTR1, required for copper transport to the central nervous system. We measured dysbindin-1 isoforms-1A and -1BC, CTR1, and ATP7A via Western blots of the postmortem dorsolateral prefrontal cortex (DLPFC) of schizophrenia subjects (n = 28) and matched controls (n = 14). In addition, we subdivided the schizophrenia group by treatment status and comorbidity of alcohol use disorder (AUD) and assessed the relationships between proteins. Schizophrenia subjects exhibited similar protein levels to that of controls, with no effect of antipsychotic treatment. We observed a shift towards more dysbindin-1A expression in schizophrenia, as revealed by the ratio of dysbindin-1 isoforms. Dysbindin-1A expression was negatively correlated with ATP7A in schizophrenia, with no correlation present in controls. AUD subjects exhibited less dysbindin-1BC and CTR1 than those without AUD. Our results, taken together with previous data, suggest that alterations in dysbindin-1 and copper transporters are brain-region specific. For example, protein levels of ATP7A, dysbindin 1BC, and CTR1 are lower in the substantia nigra in schizophrenia subjects. AUD in the DLPFC was associated with lower protein levels of dysbindin-1 and CTR1. Changes in dysbindin-1 isoform ratio and relationships appear to be prevalent in the disease, potentially impacting symptomology.
Collapse
Affiliation(s)
- Kirsten E Schoonover
- Department of Psychology and Behavioral Neuroscience, The University of Alabama at Birmingham, 3811 O'Hara Street BST W1651, Pittsburgh, PA, 15213, USA.
| | - William M Kennedy
- Department of Psychiatry and Behavioral Neurobiology, The University of Alabama at Birmingham, Pittsburgh, USA
| | - Rosalinda C Roberts
- Department of Psychiatry and Behavioral Neurobiology, The University of Alabama at Birmingham, Pittsburgh, USA
| |
Collapse
|
11
|
Tendilla-Beltrán H, Sanchez-Islas NDC, Marina-Ramos M, Leza JC, Flores G. The prefrontal cortex as a target for atypical antipsychotics in schizophrenia, lessons of neurodevelopmental animal models. Prog Neurobiol 2020; 199:101967. [PMID: 33271238 DOI: 10.1016/j.pneurobio.2020.101967] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 10/10/2020] [Accepted: 11/22/2020] [Indexed: 02/06/2023]
Abstract
Prefrontal cortex (PFC) inflammatory imbalance, oxidative/nitrosative stress (O/NS) and impaired neuroplasticity in schizophrenia are thought to have neurodevelopmental origins. Animal models are not only useful to test this hypothesis, they are also effective to establish a relationship among brain disturbances and behavior with the atypical antipsychotics (AAPs) effects. Here we review data of PFC post-mortem and in vivo neuroimaging, human induced pluripotent stem cells (hiPSC), and peripheral blood studies of inflammatory, O/NS, and neuroplasticity alterations in the disease as well as about their modulation by AAPs. Moreover, we reviewed the PFC alterations and the AAP mechanisms beyond their canonical antipsychotic action in four neurodevelopmental animal models relevant to the study of schizophrenia with a distinct approach in the generation of schizophrenia-like phenotypes, but all converge in O/NS and altered neuroplasticity in the PFC. These animal models not only reinforce the neurodevelopmental risk factor model of schizophrenia but also arouse some novel potential therapeutic targets for the disease including the reestablishment of the antioxidant response by the perineuronal nets (PNNs) and the nuclear factor erythroid 2-related factor (Nrf2) pathway, as well as the dendritic spine dynamics in the PFC pyramidal cells.
Collapse
Affiliation(s)
- Hiram Tendilla-Beltrán
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico; Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), CDMX, Mexico
| | | | - Mauricio Marina-Ramos
- Departamento de Ciencias de la Salud, Universidad Popular Autónoma del Estado de Puebla, Puebla, Mexico
| | - Juan C Leza
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Instituto Universitario de Investigación en Neuroquímica (IUIN), UCM. Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Investigación Sanitaria Hospital, 12 de Octubre (Imas12), Madrid, Spain
| | - Gonzalo Flores
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico.
| |
Collapse
|
12
|
Kleine AD, Reuss B. Interactions of Antibodies to the Gram-Negative Gastric Bacterium Helicobacter pylori with the Synaptic Calcium Sensor Synaptotagmin 5, Correlate to Impaired Vesicle Recycling in SiMa Human Neuroblastoma Cells. J Mol Neurosci 2020; 71:481-505. [PMID: 32860155 PMCID: PMC7851109 DOI: 10.1007/s12031-020-01670-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 07/15/2020] [Indexed: 11/29/2022]
Abstract
Due to molecular mimicry, maternal antibacterial antibodies are suspected to promote neurodevelopmental changes in the offspring that finally can cause disorders like autism and schizophrenia. Using a human first trimester prenatal brain multiprotein array (MPA), we demonstrate here that antibodies to the digestive tract bacteria Helicobacter pylori (α-HPy) and Campylobacter jejuni (α-CJe) interact with different synaptic proteins, including the calcium sensor synaptotagmin 5 (Syt5). Interactions of both antisera with Syt5 were confirmed by Western blot with a HEK293-cells overexpression lysate of this protein. Immunofluorescence and Western blotting revealed SiMa cells to express Syt5, which also co-migrated with a band/spot labeled by either α-HPy or α-CJe. Functionally, a 12-h pretreatment of SiMa cells with 10 μg/ml of either α-HPy or α-CJe resulted in a significant reduction of acetylcholine(ACh)-dependent calcium signals as compared to controls. Also ACh-dependent vesicle recycling was significantly reduced in cells pretreated with either α-HPy or α-CJe. Similar effects were observed upon pretreatment of SiMa cells with Syt5-specific antibodies. In conclusion, the present study supports the view that prenatal maternal antibacterial immune responses towards HPy and by this to Syt5 are able to cause functional changes, which in the end might contribute also to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Aaron David Kleine
- Institute for Neuroanatomy, University Medicine Göttingen Kreuzbergring 36, 37075, Göttingen, Federal Republic of Germany
| | - Bernhard Reuss
- Institute for Neuroanatomy, University Medicine Göttingen Kreuzbergring 36, 37075, Göttingen, Federal Republic of Germany.
| |
Collapse
|
13
|
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: 17] [Impact Index Per Article: 3.4] [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
|
14
|
Shortall SE, Brown AM, Newton-Mann E, Dawe-Lane E, Evans C, Fowler M, King MV. Calbindin Deficits May Underlie Dissociable Effects of 5-HT 6 and mGlu 7 Antagonists on Glutamate and Cognition in a Dual-Hit Neurodevelopmental Model for Schizophrenia. Mol Neurobiol 2020; 57:3439-3457. [PMID: 32533466 PMCID: PMC7340678 DOI: 10.1007/s12035-020-01938-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/13/2020] [Indexed: 12/11/2022]
Abstract
Despite several compounds entering clinical trials for the negative and cognitive symptoms of schizophrenia, few have progressed beyond phase III. This is partly attributed to a need for improved preclinical models, to understand disease and enable predictive evaluation of novel therapeutics. To this end, one recent approach incorporates "dual-hit" neurodevelopmental insults like neonatal phencyclidine plus isolation rearing (PCP-Iso). Glutamatergic dysfunction contributes to schizophrenia pathophysiology and may represent a treatment target, so we used enzyme-based microsensors to evaluate basal- and drug-evoked glutamate release in hippocampal slices from rats that received neonatal PCP and/or isolation rearing. 5-HT6 antagonist-evoked glutamate release (thought to be mediated indirectly via GABAergic disinhibition) was reduced in PCP-Iso, as were cognitive effects of a 5-HT6 antagonist in a hippocampal glutamate-dependent novel object discrimination task. Yet mGlu7 antagonist-evoked glutamatergic and cognitive responses were spared. Immunohistochemical analyses suggest these findings (which mirror the apparent lack of clinical response to 5-HT6 antagonists in schizophrenia) are not due to reduced hippocampal 5-HT input in PCP-Iso, but may be explained by reduced calbindin expression. This calcium-binding protein is present in a subset of GABAergic interneurons receiving preferential 5-HT innervation and expressing 5-HT6 receptors. Its loss (in schizophrenia and PCP-Iso) would be expected to reduce interneuron firing and potentially prevent further 5-HT6 antagonist-mediated disinhibition, without impacting on responses of VIP-expressing interneurons to mGlu7 antagonism. This research highlights the importance of improved understanding for selection of appropriate preclinical models, especially where disease neurobiology impacts on cells mediating the effects of potential therapeutics.
Collapse
Affiliation(s)
- Sinead E Shortall
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Angus M Brown
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Eliot Newton-Mann
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Erin Dawe-Lane
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Chanelle Evans
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Maxine Fowler
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - Madeleine V King
- School of Life Sciences, Medical School, Queen's Medical Centre, The University of Nottingham, Nottingham, NG7 2UH, UK.
| |
Collapse
|
15
|
Mallya AP, Wang HD, Lee HNR, Deutch AY. Microglial Pruning of Synapses in the Prefrontal Cortex During Adolescence. Cereb Cortex 2020; 29:1634-1643. [PMID: 29668872 DOI: 10.1093/cercor/bhy061] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 02/19/2018] [Accepted: 02/22/2018] [Indexed: 12/20/2022] Open
Abstract
Exaggerated synaptic elimination in the prefrontal cortex (PFC) during adolescence has been suggested to contribute to the neuropathological changes of schizophrenia. Recent data indicate that microglia (MG) sculpt synapses during early postnatal development. However, it is not known if MG contribute to the structural maturation of the PFC, which has a protracted postnatal development. We determined if MG are involved in developmentally specific synapse elimination in the PFC, focusing on adolescence. Layer 5 PFC pyramidal cells (PCs) were intracellularly filled with Lucifer Yellow for dendritic spine measurements in postnatal day (P) 24, P30, P35, P39, and P50 rats. In the contralateral PFC we evaluated if MG engulfed presynaptic (glutamatergic) and postsynaptic (dendritic spines) elements. Dendritic spine density increased from P24 to P35, when spine density peaked. There was a significant increase in MG engulfment of spines at P39 relative to earlier ages; this subsided by P50. MG also phagocytosed presynaptic glutamatergic terminals. These data indicate that MG transiently prune synapses of PFC PCs during adolescence, when the symptoms of schizophrenia typically first appear. An increase in MG-mediated synaptic remodeling of PFC PCs may contribute to the structural changes observed in schizophrenia.
Collapse
Affiliation(s)
| | - Hui-Dong Wang
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Han Noo Ri Lee
- Neuroscience Program, Vanderbilt University, Nashville, TN, USA
| | - Ariel Y Deutch
- Neuroscience Program, Vanderbilt University, Nashville, TN, USA.,Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
16
|
Zheng XX, Zhang KY, Li YC, Chen YW, Yue YS, Xia SZ, Li Y, Deng HH, Jing HL, Cao YJ. Imperatorin ameliorates learning and memory deficits through
BDNF
/
TrkB
and
ERK
/
CaMKIIα
/
CREB
signaling in prenatally‐stressed female offspring. Phytother Res 2020; 34:2408-2418. [DOI: 10.1002/ptr.6692] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/04/2020] [Accepted: 03/20/2020] [Indexed: 12/30/2022]
Affiliation(s)
- Xing X. Zheng
- Shaanxi Province Biomedicine Key Laboratory, School of Pharmacy Northwest University Xi'an China
- Key Laboratory of Resource Biology and Biotechnology in Western China Northwest University, Ministry of Education Xi'an China
| | - Kai Y. Zhang
- School of Computer Science and Technology (SCST), Xidian University Xi'an China
| | - Ying C. Li
- Shaanxi Province Biomedicine Key Laboratory, School of Pharmacy Northwest University Xi'an China
- Key Laboratory of Resource Biology and Biotechnology in Western China Northwest University, Ministry of Education Xi'an China
| | - Yi W. Chen
- Shaanxi Province Biomedicine Key Laboratory, School of Pharmacy Northwest University Xi'an China
- Key Laboratory of Resource Biology and Biotechnology in Western China Northwest University, Ministry of Education Xi'an China
| | - Yi S. Yue
- Shaanxi Province Biomedicine Key Laboratory, School of Pharmacy Northwest University Xi'an China
- Key Laboratory of Resource Biology and Biotechnology in Western China Northwest University, Ministry of Education Xi'an China
| | - Si Z. Xia
- Shaanxi Province Biomedicine Key Laboratory, School of Pharmacy Northwest University Xi'an China
- Key Laboratory of Resource Biology and Biotechnology in Western China Northwest University, Ministry of Education Xi'an China
| | - Yang Li
- Shaanxi Province Biomedicine Key Laboratory, School of Pharmacy Northwest University Xi'an China
- Key Laboratory of Resource Biology and Biotechnology in Western China Northwest University, Ministry of Education Xi'an China
| | - Huan H. Deng
- Shaanxi Province Biomedicine Key Laboratory, School of Pharmacy Northwest University Xi'an China
- Key Laboratory of Resource Biology and Biotechnology in Western China Northwest University, Ministry of Education Xi'an China
| | - Hui L. Jing
- Department of Dermatology Xi'an Hospital of Traditional Chinese Medicine Xi'an China
| | - Yan J. Cao
- Shaanxi Province Biomedicine Key Laboratory, School of Pharmacy Northwest University Xi'an China
- Key Laboratory of Resource Biology and Biotechnology in Western China Northwest University, Ministry of Education Xi'an China
| |
Collapse
|
17
|
Bitanihirwe BKY, Woo TUW. A conceptualized model linking matrix metalloproteinase-9 to schizophrenia pathogenesis. Schizophr Res 2020; 218:28-35. [PMID: 32001079 DOI: 10.1016/j.schres.2019.12.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 12/18/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022]
Abstract
Matrix metalloproteinase 9 (MMP-9) is an extracellularly operating zinc-dependent endopeptidase that is commonly expressed in the brain, other tissues. It is synthesized in a latent zymogen form known as pro-MMP-9 that is subsequently converted to the active MMP-9 enzyme following cleavage of the pro-domain. Within the central nervous system, MMP-9 is localized and released from neurons, astrocytes and microglia where its expression levels are modulated by cytokines and growth factors during both normal and pathological conditions as well as by reactive oxygen species generated during oxidative stress. MMP-9 is involved in a number of key neurodevelopmental processes that are thought to be affected in schizophrenia, including maturation of the inhibitory neurons that contain the calcium-binding protein parvalbumin, developmental formation of the specialized extracellular matrix structure perineuronal net, synaptic pruning, and myelination. In this context, the present article provides a narrative synthesis of the existing evidence linking MMP-9 dysregulation to schizophrenia pathogenesis. We start by providing an overview of MMP-9 involvement in brain development and physiology. We then discuss the potential mechanisms through which MMP-9 dysregulation may affect neural circuitry maturation as well as how these anomalies may contribute to the disease process of schizophrenia. We conclude by articulating a comprehensive, cogent, and experimentally testable hypothesis linking MMP-9 to the developmental pathophysiologic cascade that triggers the onset and sustains the chronicity of the illness.
Collapse
Affiliation(s)
| | - Tsung-Ung W Woo
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA; Program in Cellular Neuropathology, McLean Hospital, Belmont, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
18
|
Bobilev AM, Perez JM, Tamminga CA. Molecular alterations in the medial temporal lobe in schizophrenia. Schizophr Res 2020; 217:71-85. [PMID: 31227207 DOI: 10.1016/j.schres.2019.06.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/29/2019] [Accepted: 06/01/2019] [Indexed: 11/30/2022]
Abstract
The medial temporal lobe (MTL) and its individual structures have been extensively implicated in schizophrenia pathophysiology, with considerable efforts aimed at identifying structural and functional differences in this brain region. The major structures of the MTL for which prominent differences have been revealed include the hippocampus, the amygdala and the superior temporal gyrus (STG). The different functions of each of these regions have been comprehensively characterized, and likely contribute differently to schizophrenia. While neuroimaging studies provide an essential framework for understanding the role of these MTL structures in various aspects of the disease, ongoing efforts have sought to employ molecular measurements in order to elucidate the biology underlying these macroscopic differences. This review provides a summary of the molecular findings in three major MTL structures, and discusses convergent findings in cellular architecture and inter-and intra-cellular networks. The findings of this effort have uncovered cell-type, network and gene-level specificity largely unique to each brain region, indicating distinct molecular origins of disease etiology. Future studies should test the functional implications of these molecular changes at the circuit level, and leverage new advances in sequencing technology to further refine our understanding of the differential contribution of MTL structures to schizophrenia.
Collapse
Affiliation(s)
- Anastasia M Bobilev
- Department of Psychiatry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, United States of America.
| | - Jessica M Perez
- Department of Psychiatry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, United States of America.
| | - Carol A Tamminga
- Department of Psychiatry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, United States of America.
| |
Collapse
|
19
|
Zheng XX, Chen YW, Yue YS, Li YC, Xia SZ, Li Y, Deng HH, He J, Cao YJ. Icariin ameliorates learning and memory impairments through ERK/CaMKIIα/CREB signaling and HPA axis in prenatally stressed female offspring. Biomed Pharmacother 2019; 117:109077. [DOI: 10.1016/j.biopha.2019.109077] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/27/2019] [Accepted: 06/02/2019] [Indexed: 10/26/2022] Open
|
20
|
Nucifora LG, MacDonald ML, Lee BJ, Peters ME, Norris AL, Orsburn BC, Yang K, Gleason K, Margolis RL, Pevsner J, Tamminga CA, Sweet RA, Ross CA, Sawa A, Nucifora FC. Increased Protein Insolubility in Brains From a Subset of Patients With Schizophrenia. Am J Psychiatry 2019; 176:730-743. [PMID: 31055969 DOI: 10.1176/appi.ajp.2019.18070864] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The mechanisms leading to schizophrenia are likely to be diverse. However, there may be common pathophysiological pathways for subtypes of the disease. The authors tested the hypothesis that increased protein insolubility and ubiquitination underlie the pathophysiology for a subtype of schizophrenia. METHODS Prefrontal cortex and superior temporal gyrus from postmortem brains of individuals with and without schizophrenia were subjected to cold sarkosyl fractionation, separating proteins into soluble and insoluble fractions. Protein insolubility and ubiquitin levels were quantified for each insoluble fraction, with normalization to total homogenate protein. Mass spectrometry analysis was then performed to identify the protein contents of the insoluble fractions. The potential biological relevance of the detected proteins was assessed using Gene Ontology enrichment analysis and Ingenuity Pathway Analysis. RESULTS A subset of the schizophrenia brains showed an increase in protein insolubility and ubiquitination in the insoluble fraction. Mass spectrometry of the insoluble fraction revealed that brains with increased insolubility and ubiquitination exhibited a similar peptide expression by principal component analysis. The proteins that were significantly altered in the insoluble fraction were enriched for pathways relating to axon target recognition as well as nervous system development and function. CONCLUSIONS This study suggests a pathological process related to protein insolubility for a subset of patients with schizophrenia. Determining the molecular mechanism of this subtype of schizophrenia could lead to a better understanding of the pathways underlying the clinical phenotype in some patients with major mental illness as well as to improved nosology and identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Leslie G Nucifora
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Matthew L MacDonald
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Brian J Lee
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Matthew E Peters
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Alexis L Norris
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Benjamin C Orsburn
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Kun Yang
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Kelly Gleason
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Russell L Margolis
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Jonathan Pevsner
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Carol A Tamminga
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Robert A Sweet
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Christopher A Ross
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Akira Sawa
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| | - Frederick C Nucifora
- The Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore (L.G. Nucifora, Lee, Peters, Yang, Margolis, Pevsner, Ross, Sawa, F.C. Nucifora); the Departments of Psychiatry and Neurology, University of Pittsburgh, and the VISN 4 Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh (MacDonald, Sweet); the Department of Neurology, Kennedy Krieger Institute, Baltimore (Norris, Pevsner); the Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore (Norris, Pevsner, Ross, Sawa); Protein Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Md. (Orsburn); the Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas (Gleason, Tamminga); the Department of Neurology, Johns Hopkins University School of Medicine, Baltimore (Margolis, Ross, Sawa, F.C. Nucifora); Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore (Lee, Sawa); the Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore (Ross)
| |
Collapse
|
21
|
Lander SS, Khan U, Lewandowski N, Chakraborty D, Provenzano FA, Mingote S, Chornyy S, Frigerio F, Maechler P, Kaphzan H, Small SA, Rayport S, Gaisler-Salomon I. Glutamate Dehydrogenase-Deficient Mice Display Schizophrenia-Like Behavioral Abnormalities and CA1-Specific Hippocampal Dysfunction. Schizophr Bull 2019; 45:127-137. [PMID: 29471549 PMCID: PMC6293228 DOI: 10.1093/schbul/sby011] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Brain imaging has revealed that the CA1 subregion of the hippocampus is hyperactive in prodromal and diagnosed patients with schizophrenia (SCZ), and that glutamate is a driver of this hyperactivity. Strikingly, mice deficient in the glutamate synthetic enzyme glutaminase have CA1 hypoactivity and a SCZ-resilience profile, implicating glutamate-metabolizing enzymes. To address this further, we examined mice with a brain-wide deficit in the glutamate-metabolizing enzyme glutamate dehydrogenase (GDH), encoded by Glud1, which should lead to glutamate excess due to reduced glutamate metabolism in astrocytes. We found that Glud1-deficient mice have behavioral abnormalities in the 3 SCZ symptom domains, with increased baseline and amphetamine-induced hyperlocomotion as a positive symptom proxy, nest building and social preference as a negative symptom proxy, and reversal/extradimensional set shifting in the water T-maze and contextual fear conditioning as a cognitive symptom proxy. Neuroimaging of cerebral blood volume revealed hippocampal hyperactivity in CA1, which was associated with volume reduction. Parameters of hippocampal synaptic function revealed excess glutamate release and an elevated excitatory/inhibitory balance in CA1. Finally, in a direct clinical correlation using imaging-guided microarray, we found a significant SCZ-associated postmortem reduction in GLUD1 expression in CA1. These findings advance GLUD1 deficiency as a driver of excess hippocampal excitatory transmission and SCZ symptoms, and identify GDH as a target for glutamate modulation pharmacotherapy for SCZ. More broadly, these findings point to the likely involvement of alterations in glutamate metabolism in the pathophysiology of SCZ.
Collapse
Affiliation(s)
- Sharon S Lander
- Department of Psychology or Neurobiology, University of Haifa, Haifa, Israel
| | - Usman Khan
- Department of Neurology or Psychiatry, Columbia University, New York, NY
| | - Nicole Lewandowski
- Department of Neurology or Psychiatry, Columbia University, New York, NY
| | - Darpan Chakraborty
- Department of Psychology or Neurobiology, University of Haifa, Haifa, Israel
| | - Frank A Provenzano
- Department of Neurology or Psychiatry, Columbia University, New York, NY
| | - Susana Mingote
- Department of Neurology or Psychiatry, Columbia University, New York, NY,Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY
| | - Sergiy Chornyy
- Department of Psychology or Neurobiology, University of Haifa, Haifa, Israel
| | - Francesca Frigerio
- Department of Cell Physiology and Metabolism, Geneva University Medical Center, Geneva, Switzerl
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, Geneva University Medical Center, Geneva, Switzerl
| | - Hanoch Kaphzan
- Department of Psychology or Neurobiology, University of Haifa, Haifa, Israel
| | - Scott A Small
- Department of Neurology or Psychiatry, Columbia University, New York, NY
| | - Stephen Rayport
- Department of Neurology or Psychiatry, Columbia University, New York, NY,Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY
| | - Inna Gaisler-Salomon
- Department of Psychology or Neurobiology, University of Haifa, Haifa, Israel,To whom correspondence should be addressed; 199 Abba Khoushi Ave, Haifa, Israel; tel: +972-4-8249674, fax +972-4-8240966, email
| |
Collapse
|
22
|
Synaptic loss in schizophrenia: a meta-analysis and systematic review of synaptic protein and mRNA measures. Mol Psychiatry 2019; 24:549-561. [PMID: 29511299 PMCID: PMC6004314 DOI: 10.1038/s41380-018-0041-5] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 01/05/2018] [Accepted: 01/31/2018] [Indexed: 02/06/2023]
Abstract
Although synaptic loss is thought to be core to the pathophysiology of schizophrenia, the nature, consistency and magnitude of synaptic protein and mRNA changes has not been systematically appraised. Our objective was thus to systematically review and meta-analyse findings. The entire PubMed database was searched for studies from inception date to the 1st of July 2017. We selected case-control postmortem studies in schizophrenia quantifying synaptic protein or mRNA levels in brain tissue. The difference in protein and mRNA levels between cases and controls was extracted and meta-analysis conducted. Among the results, we found a significant reduction in synaptophysin in schizophrenia in the hippocampus (effect size: -0.65, p < 0.01), frontal (effect size: -0.36, p = 0.04), and cingulate cortices (effect size: -0.54, p = 0.02), but no significant changes for synaptophysin in occipital and temporal cortices, and no changes for SNAP-25, PSD-95, VAMP, and syntaxin in frontal cortex. There were insufficient studies for meta-analysis of complexins, synapsins, rab3A and synaptotagmin and mRNA measures. Findings are summarised for these, which generally show reductions in SNAP-25, PSD-95, synapsin and rab3A protein levels in the hippocampus but inconsistency in other regions. Our findings of moderate-large reductions in synaptophysin in hippocampus and frontal cortical regions, and a tendency for reductions in other pre- and postsynaptic proteins in the hippocampus are consistent with models that implicate synaptic loss in schizophrenia. However, they also identify potential differences between regions and proteins, suggesting synaptic loss is not uniform in nature or extent.
Collapse
|
23
|
Abstract
Contrary to the notion that neurology but not psychiatry is the domain of disorders evincing structural brain alterations, it is now clear that there are subtle but consistent neuropathological changes in schizophrenia. These range from increases in ventricular size to dystrophic changes in dendritic spines. A decrease in dendritic spine density in the prefrontal cortex (PFC) is among the most replicated of postmortem structural findings in schizophrenia. Examination of the mechanisms that account for the loss of dendritic spines has in large part focused on genes and molecules that regulate neuronal structure. But the simple question of what is the effector of spine loss, ie, where do the lost spines go, is unanswered. Recent data on glial cells suggest that microglia (MG), and perhaps astrocytes, play an important physiological role in synaptic remodeling of neurons during development. Synapses are added to the dendrites of pyramidal cells during the maturation of these neurons; excess synapses are subsequently phagocytosed by MG. In the PFC, this occurs during adolescence, when certain symptoms of schizophrenia emerge. This brief review discusses recent advances in our understanding of MG function and how these non-neuronal cells lead to structural changes in neurons in schizophrenia.
Collapse
Affiliation(s)
| | - Ariel Y Deutch
- Neuroscience Program, Vanderbilt University, Nashville, TN
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN
- Department of Pharmacology, Vanderbilt University, Nashville, TN
| |
Collapse
|
24
|
Sathyasaikumar KV, Tararina M, Wu HQ, Neale SA, Weisz F, Salt TE, Schwarcz R. Xanthurenic Acid Formation from 3-Hydroxykynurenine in the Mammalian Brain: Neurochemical Characterization and Physiological Effects. Neuroscience 2017; 367:85-97. [PMID: 29031603 PMCID: PMC5819007 DOI: 10.1016/j.neuroscience.2017.10.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/02/2017] [Accepted: 10/04/2017] [Indexed: 02/09/2023]
Abstract
Xanthurenic acid (XA), formed from 3-hydroxykynurenine (3-HK) in the kynurenine pathway of tryptophan degradation, may modulate glutamatergic neurotransmission by inhibiting the vesicular glutamate transporter and/or activating Group II metabotropic glutamate receptors. Here we examined the molecular and cellular mechanisms by which 3-HK controls the neosynthesis of XA in rat, mouse and human brain, and compared the physiological actions of 3-HK and XA in the rat brain. In tissue homogenates, XA formation from 3-HK was observed in all three species and traced to a major role of kynurenine aminotransferase II (KAT II). Transamination of 3-HK to XA was also demonstrated using human recombinant KAT II. Neosynthesis of XA was significantly increased in the quinolinate-lesioned rat striatum, indicating a non-neuronal localization of the process. Studies using rat cortical slices revealed that newly produced XA is rapidly released into the extracellular compartment, and that XA biosynthesis can be manipulated experimentally in the same way as the production of kynurenic acid from kynurenine (omission of Na+ or glucose, depolarizing conditions, or addition of 2-oxoacids). The synthesis of XA from 3-HK was confirmed in vivo by striatal microdialysis. In slices from the rat hippocampus, both 3-HK and XA reduced the slopes of dentate gyrus field EPSPs. The effect of 3-HK was reduced in the presence of the KAT inhibitor aminooxyacetic acid. Finally, both 3-HK and XA reduced the power of gamma-oscillatory activity recorded from the hippocampal CA3 region. Endogenous XA, newly formed from 3-HK, may therefore play a physiological role in attentional and cognitive processes.
Collapse
Affiliation(s)
- K V Sathyasaikumar
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21228, USA
| | - M Tararina
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21228, USA
| | - H-Q Wu
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21228, USA
| | - S A Neale
- Institute of Ophthalmology, University College London, London, UK; Neurexpert Ltd., London, UK
| | - F Weisz
- Institute of Ophthalmology, University College London, London, UK
| | - T E Salt
- Institute of Ophthalmology, University College London, London, UK; Neurexpert Ltd., London, UK
| | - R Schwarcz
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21228, USA.
| |
Collapse
|
25
|
Martineau M, Guzman RE, Fahlke C, Klingauf J. VGLUT1 functions as a glutamate/proton exchanger with chloride channel activity in hippocampal glutamatergic synapses. Nat Commun 2017; 8:2279. [PMID: 29273736 PMCID: PMC5741633 DOI: 10.1038/s41467-017-02367-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 11/24/2017] [Indexed: 12/18/2022] Open
Abstract
Glutamate is the major excitatory transmitter in the vertebrate nervous system. To maintain synaptic efficacy, recycling synaptic vesicles (SV) are refilled with glutamate by vesicular glutamate transporters (VGLUTs). The dynamics and mechanism of glutamate uptake in intact neurons are still largely unknown. Here, we show by live-cell imaging with pH- and chloride-sensitive fluorescent probes in cultured hippocampal neurons of wild-type and VGLUT1-deficient mice that in SVs VGLUT functions as a glutamate/proton exchanger associated with a channel-like chloride conductance. After endocytosis most internalized Cl− is substituted by glutamate in an electrically, and presumably osmotically, neutral manner, and this process is driven by both the Cl− gradient itself and the proton motive force provided by the vacuolar H+-ATPase. Our results shed light on the transport mechanism of VGLUT under physiological conditions and provide a framework for how modulation of glutamate transport via Cl− and pH can change synaptic strength. During neurotransmission synaptic vesicles are filled with glutamate by vesicular glutamate transporters (VGLUTs). Here, authors image intact neurons and show that in synaptic vesicles VGLUT functions as a glutamate/proton exchanger associated with a channel-like chloride conductance.
Collapse
Affiliation(s)
- Magalie Martineau
- Department of Cellular Biophysics, Institute for Medical Physics and Biophysics, University of Muenster, 48149, Muenster, Germany. .,University of Bordeaux and Centre National de la Recherche Scientifique, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France.
| | - Raul E Guzman
- Institute of Complex Systems, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Christoph Fahlke
- Institute of Complex Systems, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Jürgen Klingauf
- Department of Cellular Biophysics, Institute for Medical Physics and Biophysics, University of Muenster, 48149, Muenster, Germany. .,IZKF Münster and Cluster of Excellence EXC 1003, Cells in Motion (CiM), 48149, Muenster, Germany.
| |
Collapse
|
26
|
Chen LJ, Wang YJ, Tseng GF. Cortical compression rapidly trimmed transcallosal projections and altered axonal anterograde transport machinery. Neuroscience 2017; 362:79-94. [PMID: 28827177 DOI: 10.1016/j.neuroscience.2017.08.020] [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: 04/13/2017] [Revised: 08/02/2017] [Accepted: 08/09/2017] [Indexed: 11/26/2022]
Abstract
Trauma and tumor compressing the brain distort underlying cortical neurons. Compressed cortical neurons remodel their dendrites instantly. The effects on axons however remain unclear. Using a rat epidural bead implantation model, we studied the effects of unilateral somatosensory cortical compression on its transcallosal projection and the reversibility of the changes following decompression. Compression reduced the density, branching profuseness and boutons of the projection axons in the contralateral homotopic cortex 1week and 1month post-compression. Projection fiber density was higher 1-month than 1-week post-compression, suggesting adaptive temporal changes. Compression reduced contralateral cortical synaptophysin, vesicular glutamate transporter 1 (VGLUT1) and postsynaptic density protein-95 (PSD95) expressions in a week and the first two marker proteins further by 1month. βIII-tubulin and kinesin light chain (KLC) expressions in the corpus callosum (CC) where transcallosal axons traveled were also decreased. Kinesin heavy chain (KHC) level in CC was temporarily increased 1week after compression. Decompression increased transcallosal axon density and branching profuseness to higher than sham while bouton density returned to sham levels. This was accompanied by restoration of synaptophysin, VGLUT1 and PSD95 expressions in the contralateral cortex of the 1-week, but not the 1-month, compression rats. Decompression restored βIII-tubulin, but not KLC and KHC expressions in CC. However, KLC and KHC expressions in the cell bodies of the layer II/III pyramidal neurons partially recovered. Our results show cerebral compression compromised cortical axonal outputs and reduced transcallosal projection. Some of these changes did not recover in long-term decompression.
Collapse
Affiliation(s)
- Li-Jin Chen
- Department of Anatomy, College of Medicine, Tzu-Chi University, Hualien, Taiwan.
| | - Yueh-Jan Wang
- Department of Anatomy, College of Medicine, Tzu-Chi University, Hualien, Taiwan.
| | - Guo-Fang Tseng
- Department of Anatomy, College of Medicine, Tzu-Chi University, Hualien, Taiwan.
| |
Collapse
|
27
|
Schenck S, Kunz L, Sahlender D, Pardon E, Geertsma ER, Savtchouk I, Suzuki T, Neldner Y, Štefanić S, Steyaert J, Volterra A, Dutzler R. Generation and Characterization of Anti-VGLUT Nanobodies Acting as Inhibitors of Transport. Biochemistry 2017; 56:3962-3971. [DOI: 10.1021/acs.biochem.7b00436] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Stephan Schenck
- Department
of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Laura Kunz
- Department
of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Daniela Sahlender
- Department
of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, Switzerland
| | - Els Pardon
- VIB
Center for Structural Biology, VIB, 1050 Brussels, Belgium
- Structural
Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Eric R. Geertsma
- Department
of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
- Institute
of Biochemistry, Biocenter, Goethe-University Frankfurt, Max-von-Laue-Straβe
9, 60438 Frankfurt
am Main, Germany
| | - Iaroslav Savtchouk
- Department
of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, Switzerland
| | - Toshiharu Suzuki
- Department
of Applied Chemistry, School of Engineering, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Yvonne Neldner
- Department
of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Saša Štefanić
- Institute
of Parasitology, University of Zurich, Winterthurerstrasse 266a, 8057 Zurich, Switzerland
| | - Jan Steyaert
- VIB
Center for Structural Biology, VIB, 1050 Brussels, Belgium
- Structural
Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Andrea Volterra
- Department
of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, Switzerland
| | - Raimund Dutzler
- Department
of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
28
|
Sapkota K, Irvine MW, Fang G, Burnell ES, Bannister N, Volianskis A, Culley GR, Dravid SM, Collingridge GL, Jane DE, Monaghan DT. Mechanism and properties of positive allosteric modulation of N-methyl-d-aspartate receptors by 6-alkyl 2-naphthoic acid derivatives. Neuropharmacology 2017; 125:64-79. [PMID: 28709671 DOI: 10.1016/j.neuropharm.2017.07.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/14/2017] [Accepted: 07/09/2017] [Indexed: 01/27/2023]
Abstract
The theory that N-methyl-d-aspartate receptor (NMDAR) hypofunction is responsible for the symptoms of schizophrenia is well supported by many pharmacological and genetic studies. Accordingly, positive allosteric modulators (PAMs) that augment NMDAR signaling may be useful for treating schizophrenia. Previously we have identified several NMDAR PAMs containing a carboxylic acid attached to naphthalene, phenanthrene, or coumarin ring systems. In this study, we describe several functional and mechanistic properties of UBP684, a 2-naphthoic acid derivative, which robustly potentiates agonist responses at each of the four GluN1a/GluN2 receptors and at neuronal NMDARs. UBP684 increases the maximal l-glutamate/glycine response while having minor subunit-specific effects on agonist potency. PAM binding is independent of agonist binding, and PAM activity is independent of membrane voltage, redox state, and the GluN1 exon 5 N-terminal insert. UBP684 activity is, however, markedly pH-dependent, with greater potentiation occurring at lower pHs and inhibitory activity at pH 8.4. UBP684 increases channel open probability (Po) and slows receptor deactivation time upon removal of l-glutamate, but not glycine. The structurally related PAM, UBP753, reproduced most of these findings, but did not prolong agonist removal deactivation time. Studies using cysteine mutants to lock the GluN1 and GluN2 ligand-binding domains (LBDs) in the agonist-bound states indicate that PAM potentiation requires GluN2 LBD conformational flexibility. Together, these findings suggest that UBP684 and UBP753 stabilize the GluN2 LBD in an active conformation and thereby increase Po. Thus, UBP684 and UBP753 may serve as lead compounds for developing agents to enhance NMDAR activity in disorders associated with NMDAR hypofunction.
Collapse
Affiliation(s)
- Kiran Sapkota
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Mark W Irvine
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Guangyu Fang
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Erica S Burnell
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Neil Bannister
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Arturas Volianskis
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK; Centre for Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Georgia R Culley
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | | | - Graham L Collingridge
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - David E Jane
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Daniel T Monaghan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA.
| |
Collapse
|
29
|
Keller A, Ambert N, Legendre A, Bedez M, Bouteiller JM, Bischoff S, Baudry M, Moussaoui S. Impact of synaptic localization and subunit composition of ionotropic glutamate receptors on synaptic function: modeling and simulation studies. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2017; 14:892-904. [PMID: 27164603 DOI: 10.1109/tcbb.2016.2561932] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Ionotropic NMDA and AMPA glutamate receptors (iGluRs) play important roles in synaptic function under physiological and pathological conditions. iGluRs sub-synaptic localization and subunit composition are dynamically regulated by activity-dependent insertion and internalization. However, understanding the impact on synaptic transmission of changes in composition and localization of iGluRs is difficult to address experimentally. To address this question, we developed a detailed computational model of glutamatergic synapses, including spine and dendritic compartments, elementary models of subtypes of NMDA and AMPA receptors, glial glutamate transporters, intracellular calcium and a calcium-dependent signaling cascade underlying the development of long-term potentiation (LTP). These synapses were distributed on a neuron model and numerical simulations were performed to assess the impact of changes in composition and localization (synaptic vs extrasynaptic) of iGluRs on synaptic transmission and plasticity following various patterns of presynaptic stimulation. In addition, the effects of various pharmacological compounds targeting NMDARs or AMPARs were determined. Our results showed that changes in NMDAR localization have a greater impact on synaptic plasticity than changes in AMPARs. Moreover, the results suggest that modulators of AMPA and NMDA receptors have differential effects on restoring synaptic plasticity under different experimental situations mimicking various human diseases.
Collapse
|
30
|
System-based proteomic and metabonomic analysis of the Df(16)A +/- mouse identifies potential miR-185 targets and molecular pathway alterations. Mol Psychiatry 2017; 22:384-395. [PMID: 27001617 PMCID: PMC5322275 DOI: 10.1038/mp.2016.27] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 01/24/2016] [Accepted: 01/28/2016] [Indexed: 12/25/2022]
Abstract
Deletions on chromosome 22q11.2 are a strong genetic risk factor for development of schizophrenia and cognitive dysfunction. We employed shotgun liquid chromatography-mass spectrometry (LC-MS) proteomic and metabonomic profiling approaches on prefrontal cortex (PFC) and hippocampal (HPC) tissue from Df(16)A+/- mice, a model of the 22q11.2 deletion syndrome. Proteomic results were compared with previous transcriptomic profiling studies of the same brain regions. The aim was to investigate how the combined effect of the 22q11.2 deletion and the corresponding miRNA dysregulation affects the cell biology at the systems level. The proteomic brain profiling analysis revealed PFC and HPC changes in various molecular pathways associated with chromatin remodelling and RNA transcription, indicative of an epigenetic component of the 22q11.2DS. Further, alterations in glycolysis/gluconeogenesis, mitochondrial function and lipid biosynthesis were identified. Metabonomic profiling substantiated the proteomic findings by identifying changes in 22q11.2 deletion syndrome (22q11.2DS)-related pathways, such as changes in ceramide phosphoethanolamines, sphingomyelin, carnitines, tyrosine derivates and panthothenic acid. The proteomic findings were confirmed using selected reaction monitoring mass spectrometry, validating decreased levels of several proteins encoded on 22q11.2, increased levels of the computationally predicted putative miR-185 targets UDP-N-acetylglucosamine-peptide N-acetylglucosaminyltransferase 110 kDa subunit (OGT1) and kinesin heavy chain isoform 5A and alterations in the non-miR-185 targets serine/threonine-protein phosphatase 2B catalytic subunit gamma isoform, neurofilament light chain and vesicular glutamate transporter 1. Furthermore, alterations in the proteins associated with mammalian target of rapamycin signalling were detected in the PFC and with glutamatergic signalling in the hippocampus. Based on the proteomic and metabonomic findings, we were able to develop a schematic model summarizing the most prominent molecular network findings in the Df(16)A+/- mouse. Interestingly, the implicated pathways can be linked to one of the most consistent and strongest proteomic candidates, (OGT1), which is a predicted miR-185 target. Our results provide novel insights into system-biological mechanisms associated with the 22q11DS, which may be linked to cognitive dysfunction and an increased risk to develop schizophrenia. Further investigation of these pathways could help to identify novel drug targets for the treatment of schizophrenia.
Collapse
|
31
|
Alural B, Genc S, Haggarty SJ. Diagnostic and therapeutic potential of microRNAs in neuropsychiatric disorders: Past, present, and future. Prog Neuropsychopharmacol Biol Psychiatry 2017; 73:87-103. [PMID: 27072377 PMCID: PMC5292013 DOI: 10.1016/j.pnpbp.2016.03.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 03/28/2016] [Accepted: 03/30/2016] [Indexed: 12/12/2022]
Abstract
Neuropsychiatric disorders are common health problems affecting approximately 1% of the population. Twin, adoption, and family studies have displayed a strong genetic component for many of these disorders; however, the underlying pathophysiological mechanisms and neural substrates remain largely unknown. Given the critical need for new diagnostic markers and disease-modifying treatments, expanding the focus of genomic studies of neuropsychiatric disorders to include the role of non-coding RNAs (ncRNAs) is of growing interest. Of known types of ncRNAs, microRNAs (miRNAs) are 20-25-nucleotide, single-stranded, molecules that regulate gene expression through post-transcriptional mechanisms and have the potential to coordinately regulate complex regulatory networks. In this review, we summarize the current knowledge on miRNA alteration/dysregulation in neuropsychiatric disorders, with a special emphasis on schizophrenia (SCZ), bipolar disorder (BD), and major depressive disorder (MDD). With an eye toward the future, we also discuss the diagnostic and prognostic potential of miRNAs for neuropsychiatric disorders in the context of personalized treatments and network medicine.
Collapse
Affiliation(s)
- Begum Alural
- Department of Neuroscience, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey; Izmir Biomedicine and Genome Center, Dokuz Eylul University, Izmir, Turkey
| | - Sermin Genc
- Department of Neuroscience, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey; Izmir Biomedicine and Genome Center, Dokuz Eylul University, Izmir, Turkey
| | - Stephen J Haggarty
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA; Chemical Neurobiology Laboratory, Departments of Neurology and Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
32
|
McDevitt J, Krynetskiy E. Genetic findings in sport-related concussions: potential for individualized medicine? Concussion 2017; 2:CNC26. [PMID: 30202567 PMCID: PMC6096436 DOI: 10.2217/cnc-2016-0020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 11/28/2016] [Indexed: 11/24/2022] Open
Abstract
Concussion is a traumatic transient disturbance of the brain. In sport, the initial time and severity of concussion is known giving an opportunity for subsequent analysis. Variability in susceptibility and recovery between individual athletes depends, among other parameters, on genetic factors. The genes-encoding polypeptides that determine incidence, severity and prognosis for concussion are the primary candidates for genetic analysis. Genetic polymorphisms in the genes contributing to plasticity and repair (APOE), synaptic connectivity (GRIN2A), calcium influx (CACNA1E), uptake and deposit of glutamate (SLC17A7) are potential biomarkers of concussion incidence and recovery rate. With catalogued genetic variants, prospective genotyping of athletes at the beginning of their career will allow medical professionals to improve concussion management and return-to-play decisions.
Collapse
Affiliation(s)
- Jane McDevitt
- East Stroudsburg University, Athletic Training Department, East Stroudsburg, PA 18301, USA.,East Stroudsburg University, Athletic Training Department, East Stroudsburg, PA 18301, USA
| | - Evgeny Krynetskiy
- Temple University School of Pharmacy, Pharmaceutical Sciences Department, Philadelphia, PA 19140, USA.,Temple University School of Pharmacy, Pharmaceutical Sciences Department, Philadelphia, PA 19140, USA
| |
Collapse
|
33
|
Piyabhan P, Wannasiri S, Naowaboot J. Bacopa monnieri(Brahmi) improved novel object recognition task and increased cerebral vesicular glutamate transporter type 3 in sub-chronic phencyclidine rat model of schizophrenia. Clin Exp Pharmacol Physiol 2016; 43:1234-1242. [DOI: 10.1111/1440-1681.12658] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 08/01/2016] [Accepted: 08/17/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Pritsana Piyabhan
- Division of Physiology; Department of Preclinical Science; Faculty of Medicine; Thammasat University; Klongluang Pathumthani Thailand
| | - Supaporn Wannasiri
- Division of Physiology; Department of Preclinical Science; Faculty of Medicine; Thammasat University; Klongluang Pathumthani Thailand
| | - Jarinyaporn Naowaboot
- Division of Pharmacology; Department of Preclinical Science; Faculty of Medicine; Thammasat University; Klongluang Pathumthani Thailand
| |
Collapse
|
34
|
García-Bea A, Walker MA, Hyde TM, Kleinman JE, Harrison PJ, Lane TA. Metabotropic glutamate receptor 3 (mGlu3; mGluR3; GRM3) in schizophrenia: Antibody characterisation and a semi-quantitative western blot study. Schizophr Res 2016; 177:18-27. [PMID: 27130562 PMCID: PMC5145804 DOI: 10.1016/j.schres.2016.04.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 04/06/2016] [Accepted: 04/11/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND Metabotropic glutamate receptor 3 (mGlu3, mGluR3), encoded by GRM3, is a risk gene for schizophrenia and a therapeutic target. It is unclear whether expression of the receptor is altered in the disorder or related to GRM3 risk genotype. Antibodies used to date to assess mGlu3 in schizophrenia have not been well validated. OBJECTIVE To characterise six commercially available anti-mGlu3 antibodies for use in human brain, and then conduct a semi-quantitative study of mGlu3 immunoreactivity in schizophrenia. METHODS Antibodies tested using Grm3-/- and Grm2-/-/3-/- mice and transfected HEK293T/17 cells. Western blotting on membrane protein isolated from superior temporal cortex of 70 patients with schizophrenia and 87 healthy comparison subjects, genotyped for GRM3 SNP rs10234440. RESULTS One (out of six) anti-mGlu3 antibodies was fully validated, a C-terminal antibody which detected monomeric (~100kDa) and dimeric (~200kDa) mGlu3. A second, N-terminal, antibody detected the 200kDa band but also produced non-specific bands. Using the C-terminal antibody for western blotting in human brain, mGlu3 immunoreactivity was found to decline with age, and was affected by pH and post mortem interval. There were no differences in monomeric or dimeric mGlu3 immunoreactivity in schizophrenia or in relation to GRM3 genotype. The antibody was not suitable for immunohistochemistry. INTERPRETATION These data highlight the value of knockout mouse tissue for antibody validation, and the need for careful antibody characterisation. The schizophrenia data show that involvement of GRM3 in the disorder and its genetic risk architecture is not reflected in total membrane mGlu3 immunoreactivity in superior temporal cortex.
Collapse
Affiliation(s)
| | - Mary A Walker
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Baltimore, USA; Department of Neurology, Johns Hopkins School of Medicine, Baltimore, USA; Department of Psychiatry & Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, USA
| | | | - Paul J Harrison
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom; Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, United Kingdom
| | - Tracy A Lane
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
35
|
Kentner AC, Khoury A, Lima Queiroz E, MacRae M. Environmental enrichment rescues the effects of early life inflammation on markers of synaptic transmission and plasticity. Brain Behav Immun 2016; 57:151-160. [PMID: 27002704 DOI: 10.1016/j.bbi.2016.03.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 03/04/2016] [Accepted: 03/17/2016] [Indexed: 12/13/2022] Open
Abstract
Environmental enrichment (EE) has been successful at rescuing the brain from a variety of early-life psychogenic stressors. However, its ability to reverse the behavioral and neural alterations induced by a prenatal maternal infection model of schizophrenia is less clear. Moreover, the specific interactions between the components (i.e. social enhancement, novelty, physical activity) of EE that lead to its success as a supportive intervention have not been adequately identified. In the current study, standard housed female Sprague-Dawley rats were administered either the inflammatory endotoxin lipopolysaccharide (LPS; 100μg/kg) or pyrogen-free saline (equivolume) on gestational day 15. On postnatal day 50, offspring were randomized into one of three conditions: EE (group housed in a large multi-level cage with novel toys, tubes and ramps), Colony Nesting (CN; socially-housed in a larger style cage), or Standard Care (SC; pair-housed in standard cages). Six weeks later we scored social engagement and performance in the object-in-place task. Afterwards hippocampus and prefrontal cortex (n=7-9) were collected and evaluated for excitatory amino acid transporter (EAAT) 1-3, brain-derived neurotrophic factor (BDNF), and neurotrophic tyrosine kinase, receptor type 2 (TrkB) gene expression (normalized to GAPDH) using qPCR methods. Overall, we show that gestational inflammation downregulates genes critical to synaptic transmission and plasticity, which may underlie the pathogenesis of neurodevelopmental disorders such as schizophrenia and autism. Additionally, we observed disruptions in both social engagement and spatial discrimination. Importantly, behavioral and neurophysiological effects were rescued in an experience dependent manner. Given the evidence that schizophrenia and autism may be associated with infection during pregnancy, these data have compelling implications for the prevention and reversibility of the consequences that follow immune activation in early in life.
Collapse
Affiliation(s)
- Amanda C Kentner
- School of Arts & Sciences, Health Psychology Program, MCPHS University (formerly the Massachusetts College of Pharmacy & Health Sciences), Boston, MA 02115, United States.
| | - Antoine Khoury
- School of Pharmacy, MCPHS University, Boston, MA 02115, United States
| | | | - Molly MacRae
- School of Arts & Sciences, Health Psychology Program, MCPHS University (formerly the Massachusetts College of Pharmacy & Health Sciences), Boston, MA 02115, United States
| |
Collapse
|
36
|
Camkurt MA, Karababa F, Erdal ME, Bayazıt H, Kandemir SB, Ay ME, Kandemir H, Ay Öİ, Çiçek E, Selek S, Taşdelen B. Investigation of Dysregulation of Several MicroRNAs in Peripheral Blood of Schizophrenia Patients. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2016; 14:256-60. [PMID: 27489379 PMCID: PMC4977811 DOI: 10.9758/cpn.2016.14.3.256] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 12/26/2015] [Accepted: 01/05/2016] [Indexed: 11/24/2022]
Abstract
Objective The prevalence of schizophrenia is 1%, and it is a debilitating disorder that often results in a shortened lifespan. Peripheral blood samples are good candidates to investigate because they can be easily drawn, and they are widely studied in psychiatric disorders. MicroRNAs are small non-coding RNA transcripts. They regulate the expression of genes by binding to the 3′-untranslated region (UTR) of mRNAs and pointing them to degrade. In this study, we aimed to investigate the expression of miR-9-5p, miR-29a-3p, miR-106-5p, miR-106b-5p, miR-107, miR-125a-3p, and miR-125b-3p in schizophrenia patients and healthy controls. Methods We collected blood samples from 16 patients with schizophrenia and 16 healthy controls. MicroRNAs were measured with reverse transcriptase polymerase chain reaction. Results Schizophrenia patients showed statistically significant upregulation of five microRNAs: miR9-5p (p=0.002), miR29a-3p (p<0.001), miR106b-5p (p=0.002), miR125a-3p (p<0.001), and miR125b-3p (p=0.018). Conclusion Our results increased the value of the miR106 and miR29 families as potentially and consistently dysregulated in psychiatric disorders. Our results should be considered preliminary, and they need confirmation in future studies with larger sample sizes.
Collapse
Affiliation(s)
| | - Fatih Karababa
- Psychiatry Clinic, Harran University Teaching Hospital, Şanlıurfa, Turkey
| | - Mehmet Emin Erdal
- Department of Medical Biology, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Hüseyin Bayazıt
- Psychiatry Clinic, Siverek State Hospital, Şanlıurfa, Turkey
| | | | - Mustafa Ertan Ay
- Department of Medical Biology, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Hasan Kandemir
- Department of Child and Adolescent Psychiatry, Harran University Teaching Hospital, Şanlıurfa, Turkey
| | - Özlem İzci Ay
- Department of Medical Biology, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Erdinç Çiçek
- Psychiatry Clinic, Çumra State Hospital, Konya, Turkey
| | - Salih Selek
- Department of Psychiatry and Behavioral Sciences, Harris County Psychiatric Center, University of Texas, Houston, TX, USA
| | - Bahar Taşdelen
- Department of Biostatistics, Mersin University, Mersin, Turkey
| |
Collapse
|
37
|
Hollins SL, Zavitsanou K, Walker FR, Cairns MJ. Alteration of transcriptional networks in the entorhinal cortex after maternal immune activation and adolescent cannabinoid exposure. Brain Behav Immun 2016; 56:187-96. [PMID: 26923065 DOI: 10.1016/j.bbi.2016.02.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 02/16/2016] [Accepted: 02/24/2016] [Indexed: 02/04/2023] Open
Abstract
Maternal immune activation (MIA) and adolescent cannabinoid exposure (ACE) have both been identified as major environmental risk factors for schizophrenia. We examined the effects of these two risk factors alone, and in combination, on gene expression during late adolescence. Pregnant rats were exposed to the viral infection mimic polyriboinosinic-polyribocytidylic acid (poly I:C) on gestational day (GD) 15. Adolescent offspring received daily injections of the cannabinoid HU210 for 14days starting on postnatal day (PND) 35. Gene expression was examined in the left entorhinal cortex (EC) using mRNA microarrays. We found prenatal treatment with poly I:C alone, or HU210 alone, produced relatively minor changes in gene expression. However, following combined treatments, offspring displayed significant changes in transcription. This dramatic and persistent alteration of transcriptional networks enriched with genes involved in neurotransmission, cellular signalling and schizophrenia, was associated with a corresponding perturbation in the expression of small non-coding microRNA (miRNA). These results suggest that a combination of environmental exposures during development leads to significant genomic remodeling that disrupts maturation of the EC and its associated circuitry with important implications as the potential antecedents of memory and learning deficits in schizophrenia and other neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sharon L Hollins
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia; Schizophrenia Research Institute, Sydney, NSW, Australia; Centre for Brain and Mental Health Research, Hunter Medical Research Institute, Newcastle, NSW 2305, Australia
| | - Katerina Zavitsanou
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
| | - Frederick Rohan Walker
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia; Centre for Brain and Mental Health Research, Hunter Medical Research Institute, Newcastle, NSW 2305, Australia
| | - Murray J Cairns
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia; Schizophrenia Research Institute, Sydney, NSW, Australia; Centre for Brain and Mental Health Research, Hunter Medical Research Institute, Newcastle, NSW 2305, Australia.
| |
Collapse
|
38
|
Abstract
Schizophrenia is a serious psychiatric illness which is experienced by about 1 % of individuals worldwide and has a debilitating impact on perception, cognition, and social function. Over the years, several models/hypotheses have been developed which link schizophrenia to dysregulations of the dopamine, glutamate, and serotonin receptor pathways. An important segment of these pathways that have been extensively studied for the pathophysiology of schizophrenia is the presynaptic neurotransmitter release mechanism. This set of molecular events is an evolutionarily well-conserved process that involves vesicle recruitment, docking, membrane fusion, and recycling, leading to efficient neurotransmitter delivery at the synapse. Accumulated evidence indicate dysregulation of this mechanism impacting postsynaptic signal transduction via different neurotransmitters in key brain regions implicated in schizophrenia. In recent years, after ground-breaking work that elucidated the operations of this mechanism, research efforts have focused on the alterations in the messenger RNA (mRNA) and protein expression of presynaptic neurotransmitter release molecules in schizophrenia and other neuropsychiatric conditions. In this review article, we present recent evidence from schizophrenia human postmortem studies that key proteins involved in the presynaptic release mechanism are dysregulated in the disorder. We also discuss the potential impact of dysfunctional presynaptic neurotransmitter release on the various neurotransmitter systems implicated in schizophrenia.
Collapse
Affiliation(s)
- Chijioke N Egbujo
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - Duncan Sinclair
- Neuroscience Research Australia, Barker St, Randwick, NSW, 2031, Australia
| | - Chang-Gyu Hahn
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
39
|
McMeekin LJ, Lucas EK, Meador-Woodruff JH, McCullumsmith RE, Hendrickson RC, Gamble KL, Cowell RM. Cortical PGC-1α-Dependent Transcripts Are Reduced in Postmortem Tissue From Patients With Schizophrenia. Schizophr Bull 2016; 42:1009-17. [PMID: 26683626 PMCID: PMC4903048 DOI: 10.1093/schbul/sbv184] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The transcriptional coactivator peroxisome proliferator-activated receptor-gamma coactivator 1-alpha (PGC-1α) has been linked to multiple neurological and psychiatric disorders including schizophrenia, but its involvement in the pathophysiology of these disorders is unclear. Experiments in mice have revealed a set of developmentally-regulated cortical PGC-1α-dependent transcripts involved in calcium buffering (parvalbumin, PV), synchronous neurotransmitter release (synaptotagmin 2, Syt2; complexin 1, Cplx1) and axonal integrity (neurofilamaent heavy chain, Nefh). We measured the mRNA expression of PGC-1α and these transcripts in postmortem cortical tissue from control and schizophrenia patients and found a reduction in PGC-1α-dependent transcripts without a change in PGC-1α. While control subjects with high PGC-1α expression exhibited high PV and Nefh expression, schizophrenia subjects with high PGC-1α expression did not, suggesting dissociation between PGC-1α expression and these targets in schizophrenia. Unbiased analyses of the promoter regions for PGC-1α-dependent transcripts revealed enrichment of binding sites for the PGC-1α-interacting transcription factor nuclear respiratory factor 1 (NRF-1). NRF-1 mRNA expression was reduced in schizophrenia, and its transcript levels predicted that of PGC-1α-dependent targets in schizophrenia. Interestingly, the positive correlation between PGC-1α and PV, Syt2, or Cplx1 expression was lost in schizophrenia patients with low NRF-1 expression, suggesting that NRF-1 is a critical predictor of these genes in disease. These data suggest that schizophrenia involves a disruption in PGC-1α and/or NRF-1-associated transcriptional programs in the cortex and that approaches to enhance the activity of PGC-1α or transcriptional regulators like NRF-1 should be considered with the goal of restoring normal gene programs and improving cortical function.
Collapse
Affiliation(s)
- Laura J McMeekin
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL
| | - Elizabeth K Lucas
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - James H Meador-Woodruff
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL
| | | | | | - Karen L Gamble
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL
| | - Rita M Cowell
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL;
| |
Collapse
|
40
|
|
41
|
Lisek M, Boczek T, Ferenc B, Zylinska L. Regional brain dysregulation of Ca(2+)-handling systems in ketamine-induced rat model of experimental psychosis. Cell Tissue Res 2015; 363:609-20. [PMID: 26685921 PMCID: PMC4761010 DOI: 10.1007/s00441-015-2332-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 11/11/2015] [Indexed: 01/21/2023]
Abstract
Chronic N-methyl-D-aspartate receptor (NMDAR) antagonist treatment can provide valuable neurochemical and neuroanatomical models of experimental psychosis. One such antagonist, ketamine, with its short half-time and well-documented psychotomimetic action, has cognitive effects resembling various aspects of schizophrenia-like symptoms. In order to obtain insights into possible relationships between Ca(2+) homeostasis and schizophrenia-related symptoms, we investigate the effects of chronic ketamine administration on intracellular Ca(2+) levels in various brain regions and on the expression level of key members of the neuronal Ca(2+)-handling system in rats. We show increased intracellular [Ca(2+)] in all of the examined brain regions following ketamine treatment but an altered cytosolic Ca(2+) level correlated with hyperlocomotor activity was only established for the cortex and striatum. Our findings also suggest that an imbalance in the expression between the calcium "on" and "off" systems contributes to the deregulation of brain Ca(2+) homeostasis in our ketamine-induced model of experimental psychosis. Identification of the genes whose expression is affected by ketamine treatment indicates their involvement as putative etiological factors in schizophrenia.
Collapse
Affiliation(s)
- Malwina Lisek
- Department of Molecular Neurochemistry, Faculty of Health Sciences, Medical University of Lodz, 6/8 Mazowiecka Street, 92215, Lodz, Poland.
| | - Tomasz Boczek
- Department of Molecular Neurochemistry, Faculty of Health Sciences, Medical University of Lodz, 6/8 Mazowiecka Street, 92215, Lodz, Poland
| | - Bozena Ferenc
- Department of Molecular Neurochemistry, Faculty of Health Sciences, Medical University of Lodz, 6/8 Mazowiecka Street, 92215, Lodz, Poland
| | - Ludmila Zylinska
- Department of Molecular Neurochemistry, Faculty of Health Sciences, Medical University of Lodz, 6/8 Mazowiecka Street, 92215, Lodz, Poland
| |
Collapse
|
42
|
Tamminga CA, Zukin RS. Schizophrenia: Evidence implicating hippocampal GluN2B protein and REST epigenetics in psychosis pathophysiology. Neuroscience 2015. [PMID: 26211447 DOI: 10.1016/j.neuroscience.2015.07.038] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The hippocampus is strongly implicated in the psychotic symptoms of schizophrenia. Functionally, basal hippocampal activity (perfusion) is elevated in schizophrenic psychosis, as measured with positron emission tomography (PET) and with magnetic resonance (MR) perfusion techniques, while hippocampal activation to memory tasks is reduced. Subfield-specific hippocampal molecular pathology exists in human psychosis tissue which could underlie this neuronal hyperactivity, including increased GluN2B-containing NMDA receptors in hippocampal CA3, along with increased postsynaptic density protein-95 (PSD-95) along with augmented dendritic spines on the pyramidal neuron apical dendrites. We interpret these observations to implicate a reduction in the influence of a ubiquitous gene repressor, repressor element-1 silencing transcription factor (REST) in psychosis; REST is involved in the age-related maturation of the NMDA receptor from GluN2B- to GluN2A-containing NMDA receptors through epigenetic remodeling. These CA3 changes in psychosis leave the hippocampus liable to pathological increases in neuronal activity, feedforward excitation and false memory formation, sometimes with psychotic content.
Collapse
Affiliation(s)
- C A Tamminga
- UT Southwestern Medical School, Dallas, TX, United States.
| | - R S Zukin
- Albert Einstein School of Medicine, New York, NY, United States
| |
Collapse
|
43
|
Granseth B, Andersson FK, Lindström SH. The initial stage of reversal learning is impaired in mice hemizygous for the vesicular glutamate transporter (VGluT1). GENES BRAIN AND BEHAVIOR 2015; 14:477-85. [PMID: 26113146 DOI: 10.1111/gbb.12230] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 06/18/2015] [Accepted: 06/23/2015] [Indexed: 01/08/2023]
Abstract
Behavioral flexibility is a complex cognitive function that is necessary for survival in changeable environments. Patients with schizophrenia or Parkinson's disease often suffer from cognitive rigidity, reducing their capacity to function in society. Patients and rodent models with focal lesions in the prefrontal cortex (PFC) show similar rigidity, owing to the loss of PFC regulation of subcortical reward circuits involved in behavioral flexibility. The vesicular glutamate transporter (VGluT1) is preferentially expressed at modulatory synapses, including PFC neurons that project to components of the reward circuit (such as the nucleus accumbens, NAc). VGluT1(+/-) mice display behavioral phenotypes matching many symptoms of schizophrenia, and VGluT1 expression is reduced in the PFC of patients with schizophrenia and Parkinson's disease. Thus, it appears likely that VGluT1-expressing synapses from PFC play a key role in behavioral flexibility. To examine this hypothesis, we studied behavioral flexibility in VGluT1(+/-) mice by testing reversal learning in a visual discrimination task. Here, we show that VGluT1(+/-) mice acquired the initial visual discrimination at the same rate as controls. However, they failed to suppress responses to the previously rewarded stimulus following reversal of reward contingencies. Thus, our genetic disruption of modulatory glutamatergic signaling, including that arising from PFC, appears to have impaired the first stage of reversal learning (extinguishing responses to previously rewarded stimuli). Our data show that this deficit stems from a preservative phenotype. These findings suggest that glutamatergic regulation from the cortex is important for behavioral flexibility and the disruption of this pathway may be relevant in diseases such as schizophrenia.
Collapse
Affiliation(s)
- B Granseth
- Department of Clinical and Experimental Medicine, Division of Cell Biology, Linköping University, Linköping, Sweden
| | - F K Andersson
- Department of Clinical and Experimental Medicine, Division of Cell Biology, Linköping University, Linköping, Sweden
| | - S H Lindström
- Department of Clinical and Experimental Medicine, Division of Cell Biology, Linköping University, Linköping, Sweden
| |
Collapse
|
44
|
Sullivan CR, Funk AJ, Shan D, Haroutunian V, McCullumsmith RE. Decreased chloride channel expression in the dorsolateral prefrontal cortex in schizophrenia. PLoS One 2015; 10:e0123158. [PMID: 25826365 PMCID: PMC4380350 DOI: 10.1371/journal.pone.0123158] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 02/17/2015] [Indexed: 12/17/2022] Open
Abstract
Alterations in GABAergic neurotransmission are implicated in several psychiatric illnesses, including schizophrenia. The Na-K-Cl and K-Cl cotransporters regulate intracellular chloride levels. Abnormalities in cotransporter expression levels could shift the chloride electrochemical gradient and impair GABAergic transmission. In this study, we performed Western blot analysis to investigate whether the Na-K-Cl and K-Cl cotransporter protein is abnormally expressed in the dorsal lateral prefrontal cortex and the anterior cingulate cortex in patients with schizophrenia versus a control group. We found decreased K-Cl cotransporter protein expression in the dorsal lateral prefrontal cortex, but not the anterior cingulate cortex, in subjects with schizophrenia, supporting the hypothesis of region level abnormal GABAergic function in the pathophysiology of schizophrenia. Subjects with schizophrenia off antipsychotic medication at the time of death had decreased K-Cl cotransporter protein expression compared to both normal controls and subjects with schizophrenia on antipsychotics. Our results provide evidence for KCC2 protein abnormalities in schizophrenia and suggest that antipsychotic medications might reverse deficits of this protein in the illness.
Collapse
Affiliation(s)
- Courtney R. Sullivan
- University of Cincinnati College of Medicine, Neuroscience Graduate Program, Cincinnati, Ohio, United States of America
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Adam J. Funk
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Dan Shan
- Department of Nephrology, University of Alabama Birmingham, Birmingham, Alabama, United States of America
| | - Vahram Haroutunian
- Department of Psychiatry, Mount Sinai School of Medicine, New York, New York, United States of America
- James J Peters Veterans Affairs Medical Center, New York, New York, United States of America
| | - Robert E. McCullumsmith
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
45
|
Hass J, Walton E, Kirsten H, Turner J, Wolthusen R, Roessner V, Sponheim SR, Holt D, Gollub R, Calhoun VD, Ehrlich S. Complexin2 modulates working memory-related neural activity in patients with schizophrenia. Eur Arch Psychiatry Clin Neurosci 2015; 265:137-45. [PMID: 25297695 PMCID: PMC4342303 DOI: 10.1007/s00406-014-0550-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 09/30/2014] [Indexed: 12/11/2022]
Abstract
The specific contribution of risk or candidate gene variants to the complex phenotype of schizophrenia is largely unknown. Studying the effects of such variants on brain function can provide insight into disease-associated mechanisms on a neural systems level. Previous studies found common variants in the complexin2 (CPLX2) gene to be highly associated with cognitive dysfunction in schizophrenia patients. Similarly, cognitive functioning was found to be impaired in Cplx2 gene-deficient mice if they were subjected to maternal deprivation or mild brain trauma during puberty. Here, we aimed to study seven common CPLX2 single-nucleotide polymorphisms (SNPs) and their neurogenetic risk mechanisms by investigating their relationship to a schizophrenia-related functional neuroimaging intermediate phenotype. We examined functional MRI and genotype data collected from 104 patients with DSM-IV-diagnosed schizophrenia and 122 healthy controls who participated in the Mind Clinical Imaging Consortium study of schizophrenia. Seven SNPs distributed over the whole CPLX2 gene were tested for association with working memory-elicited neural activity in a frontoparietal neural network. Three CPLX2 SNPs were significantly associated with increased neural activity in the dorsolateral prefrontal cortex and intraparietal sulcus in the schizophrenia sample, but showed no association in healthy controls. Since increased working memory-related neural activity in individuals with or at risk for schizophrenia has been interpreted as 'neural inefficiency,' these findings suggest that certain variants of CPLX2 may contribute to impaired brain function in schizophrenia, possibly combined with other deleterious genetic variants, adverse environmental events, or developmental insults.
Collapse
Affiliation(s)
- Johanna Hass
- Department of Child and Adolescent Psychiatry, School of Medicine, TU Dresden, Dresden, Germany
| | - Esther Walton
- Department of Child and Adolescent Psychiatry, School of Medicine, TU Dresden, Dresden, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), University of Leipzig, Leipzig, Germany,LIFE (Leipzig Interdisciplinary Research Cluster of Genetic Factors, Phenotypes and Environment), University of Leipzig, Leipzig, Germany
| | | | - Rick Wolthusen
- Department of Child and Adolescent Psychiatry, School of Medicine, TU Dresden, Dresden, Germany,MGH/MIT/HMS Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA USA,Department of Psychiatry, Massachusetts General Hospital, Boston, MA USA
| | - Veit Roessner
- Department of Child and Adolescent Psychiatry, School of Medicine, TU Dresden, Dresden, Germany
| | - Scott R Sponheim
- Department of Psychiatry and the Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN USA
| | - Daphne Holt
- MGH/MIT/HMS Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA USA,Department of Psychiatry, Massachusetts General Hospital, Boston, MA USA
| | - Randy Gollub
- MGH/MIT/HMS Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA USA,Department of Psychiatry, Massachusetts General Hospital, Boston, MA USA
| | - Vince D Calhoun
- The MIND Research Network, Albuquerque, NM USA,Department of Electrical and Computer Engineering, University of New Mexico, Albuquerque, NM USA
| | - Stefan Ehrlich
- Department of Child and Adolescent Psychiatry, School of Medicine, TU Dresden, Dresden, Germany,MGH/MIT/HMS Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA USA,Department of Psychiatry, Massachusetts General Hospital, Boston, MA USA
| |
Collapse
|
46
|
Saba L, Viscomi MT, Caioli S, Pignataro A, Bisicchia E, Pieri M, Molinari M, Ammassari-Teule M, Zona C. Altered Functionality, Morphology, and Vesicular Glutamate Transporter Expression of Cortical Motor Neurons from a Presymptomatic Mouse Model of Amyotrophic Lateral Sclerosis. Cereb Cortex 2015; 26:1512-28. [DOI: 10.1093/cercor/bhu317] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
47
|
Sahu A, Kumar S, Sreenivasamurthy SK, Selvan LDN, Madugundu AK, Yelamanchi SD, Puttamallesh VN, Dey G, Anil AK, Srinivasan A, Mukherjee KK, Gowda H, Satishchandra P, Mahadevan A, Pandey A, Prasad TSK, Shankar SK. Host response profile of human brain proteome in toxoplasma encephalitis co-infected with HIV. Clin Proteomics 2014; 11:39. [PMID: 25404878 PMCID: PMC4232683 DOI: 10.1186/1559-0275-11-39] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 09/02/2014] [Indexed: 01/27/2023] Open
Abstract
Background Toxoplasma encephalitis is caused by the opportunistic protozoan parasite Toxoplasma gondii. Primary infection with T. gondii in immunocompetent individuals remains largely asymptomatic. In contrast, in immunocompromised individuals, reactivation of the parasite results in severe complications and mortality. Molecular changes at the protein level in the host central nervous system and proteins associated with pathogenesis of toxoplasma encephalitis are largely unexplored. We used a global quantitative proteomic strategy to identify differentially regulated proteins and affected molecular networks in the human host during T. gondii infection with HIV co-infection. Results We identified 3,496 proteins out of which 607 proteins were differentially expressed (≥1.5-fold) when frontal lobe of the brain from patients diagnosed with toxoplasma encephalitis was compared to control brain tissues. We validated differential expression of 3 proteins through immunohistochemistry, which was confirmed to be consistent with mass spectrometry analysis. Pathway analysis of differentially expressed proteins indicated deregulation of several pathways involved in antigen processing, immune response, neuronal growth, neurotransmitter transport and energy metabolism. Conclusions Global quantitative proteomic approach adopted in this study generated a comparative proteome profile of brain tissues from toxoplasma encephalitis patients co-infected with HIV. Differentially expressed proteins include previously reported and several new proteins in the context of T. gondii and HIV infection, which can be further investigated. Molecular pathways identified to be associated with the disease should enhance our understanding of pathogenesis in toxoplasma encephalitis. Electronic supplementary material The online version of this article (doi:10.1186/1559-0275-11-39) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Apeksha Sahu
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India ; Bioinformatics Centre, School of Life Sciences, Pondicherry University, Puducherry, 605014 India
| | - Satwant Kumar
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India
| | - Sreelakshmi K Sreenivasamurthy
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India ; Manipal University, Madhav Nagar, Manipal, 576104 India
| | - Lakshmi Dhevi N Selvan
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India ; Amrita School of Biotechnology, Amrita University, Kollam, 690525 India
| | - Anil K Madugundu
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India ; Bioinformatics Centre, School of Life Sciences, Pondicherry University, Puducherry, 605014 India
| | - Soujanya D Yelamanchi
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India ; School of Biotechnology, KIIT University, Bhubaneswar, 751024 India
| | | | - Gourav Dey
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India ; Manipal University, Madhav Nagar, Manipal, 576104 India
| | | | - Anand Srinivasan
- Department of Pharmacology, Postgraduate Institute of Medical Education & Research, Chandigarh, 160012 India
| | - Kanchan K Mukherjee
- Department of Neurosurgery, Postgraduate Institute of Medical Education & Research, Chandigarh, 160012 India
| | - Harsha Gowda
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India
| | | | - Anita Mahadevan
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bangalore, 560029 India ; Human Brain Tissue Repository, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, Bangalore, 560029 India
| | - Akhilesh Pandey
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA ; Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 1205 USA ; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA ; The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Thottethodi Subrahmanya Keshava Prasad
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India ; Bioinformatics Centre, School of Life Sciences, Pondicherry University, Puducherry, 605014 India ; Manipal University, Madhav Nagar, Manipal, 576104 India ; Amrita School of Biotechnology, Amrita University, Kollam, 690525 India ; NIMHANS-IOB Proteomics and Bioinformatics Laboratory, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, Bangalore, 560029 India
| | - Susarla Krishna Shankar
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bangalore, 560029 India ; Human Brain Tissue Repository, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, Bangalore, 560029 India
| |
Collapse
|
48
|
Antipsychotic treatment modulates glutamate transport and NMDA receptor expression. Eur Arch Psychiatry Clin Neurosci 2014; 264 Suppl 1:S67-82. [PMID: 25214389 DOI: 10.1007/s00406-014-0534-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 09/02/2014] [Indexed: 12/21/2022]
Abstract
Schizophrenia patients often suffer from treatment-resistant cognitive and negative symptoms, both of which are influenced by glutamate neurotransmission. Innovative therapeutic strategies such as agonists at metabotropic glutamate receptors or glycin reuptake inhibitors try to modulate the brain's glutamate network. Interactions of amino acids with monoamines have been described on several levels, and first- and second-generation antipsychotic agents (FGAs, SGAs) are known to exert modulatory effects on the glutamatergic system. This review summarizes the current knowledge on effects of FGAs and SGAs on glutamate transport and receptor expression derived from pharmacological studies. Such studies serve as a control for molecular findings in schizophrenia brain tissue and are clinically relevant. Moreover, they may validate animal models for psychosis, foster basic research on antipsychotic substances and finally lead to a better understanding of how monoaminergic and amino acid neurotransmissions are intertwined. In the light of these results, important differences dependent on antipsychotic substances, dosage and duration of treatment became obvious. While some post-mortem findings might be confounded with multifold drug effects, others are unlikely to be influenced by antipsychotic treatment and could represent important markers of schizophrenia pathophysiology. In similarity to the convergence of toxic and psychotomimetic effects of dopaminergic, serotonergic and anti-glutamatergic substances, the therapeutic mechanisms of SGAs might merge on a yet to be defined molecular level. In particular, serotonergic effects of SGAs, such as an agonism at 5HT1A receptors, represent important targets for further clinical research.
Collapse
|
49
|
Hu W, MacDonald ML, Elswick DE, Sweet RA. The glutamate hypothesis of schizophrenia: evidence from human brain tissue studies. Ann N Y Acad Sci 2014; 1338:38-57. [PMID: 25315318 DOI: 10.1111/nyas.12547] [Citation(s) in RCA: 182] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A number of studies have indicated that antagonists of the N-methyl-d-aspartate subtypes of glutamate receptors can cause schizophrenia-like symptoms in healthy individuals and exacerbate symptoms in individuals with schizophrenia. These findings have led to the glutamate hypothesis of schizophrenia. Here we review the evidence for this hypothesis in postmortem studies of brain tissue from individuals affected by schizophrenia, summarizing studies of glutamate neuron morphology, of expression of glutamate receptors and transporters, and of the synthesizing and metabolizing enzymes for glutamate and its co-agonists. We found consistent evidence of morphological alterations of dendrites of glutamatergic neurons in the cerebral cortex of subjects with schizophrenia and of reduced levels of the axon bouton marker synaptophysin. There were no consistent alterations of mRNA expression of glutamate receptors, although there has been limited study of the corresponding proteins. Studies of the glutamate metabolic pathway have been limited, although there is some evidence that excitatory amino acid transporter-2, glutamine synthetase, and glutaminase have altered expression in schizophrenia. Future studies would benefit from additional direct examination of glutamatergic proteins. Further advances, such as selective testing of synaptic microdomains, cortical layers, and neuronal subtypes, may also be required to elucidate the nature of glutamate signaling impairments in schizophrenia.
Collapse
Affiliation(s)
- Wei Hu
- Department of Behavioral Medicine and Psychiatry, West Virginia University Health Sciences Center, Morgantown, West Virginia
| | | | | | | |
Collapse
|
50
|
Abazyan S, Yang EJ, Abazyan B, Xia M, Yang C, Rojas C, Slusher B, Sattler R, Pletnikov M. Mutant disrupted-in-schizophrenia 1 in astrocytes: focus on glutamate metabolism. J Neurosci Res 2014; 92:1659-68. [PMID: 25131692 DOI: 10.1002/jnr.23459] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 06/18/2014] [Accepted: 06/30/2014] [Indexed: 01/05/2023]
Abstract
Disrupted-in-schizophrenia 1 (DISC1) is a genetic risk factor that has been implicated in major mental disorders. DISC1 binds to and stabilizes serine racemase to regulate production of D-serine by astrocytes, contributing to glutamate (GLU) neurotransmission. However, the possible involvement of astrocytic DISC1 in synthesis, metabolism, reuptake, or secretion of GLU remains unexplored. Therefore, we studied the effects of dominant-negative mutant DISC1 on various aspects of GLU metabolism by using primary astrocyte cultures and hippocampal tissue from transgenic mice with astrocyte-restricted expression of mutant DISC1. Although mutant DISC1 had no significant effects on astrocyte proliferation, GLU reuptake, glutaminase, or glutamate carboxypeptidase II activity, expression of mutant DISC1 was associated with increased levels of alanine-serine-cysteine transporter 2, vesicular glutamate transporters 1 and 3 in primary astrocytes and in the hippocampus, and elevated expression of the NR1 subunit and diminished expression of the NR2A subunit of N-methyl-D-aspartate (NMDA) receptors in the hippocampus, at postnatal day 21. Our findings indicate that decreased D-serine production by astrocytic mutant DISC1 might lead to compensatory changes in levels of the amino acid transporters and NMDA receptors in the context of tripartite synapse.
Collapse
Affiliation(s)
- Sofya Abazyan
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | | | | | | | | | | | | | | |
Collapse
|