1
|
Shanmugam I, Radhakrishnan S, Santosh S, Ramnath A, Anil M, Devarajan Y, Maheswaran S, Narayanan V, Pitchaimani A. Emerging role and translational potential of small extracellular vesicles in neuroscience. Life Sci 2024; 355:122987. [PMID: 39151884 DOI: 10.1016/j.lfs.2024.122987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Small extracellular vesicles (sEV) are endogenous lipid-bound membrane vesicles secreted by both prokaryotic and eukaryotic cells into the extracellular environment, performs several biological functions such as cell-cell communication, transfer of proteins, mRNA, and ncRNA to target cells in distant sites. Due to their role in molecular pathogenesis and its potential to deliver biological cargo to target cells, it has become a prominent area of interest in recent research in the field of Neuroscience. However, their role in neurological disorders, like neurodegenerative diseases is more complex and still unaddressed. Thus, this review focuses on the role of sEV in neurodegenerative and neurodevelopmental diseases, including their biogenesis, classification, and pathogenesis, with translational advantages and limitations in the area of neurobiology.
Collapse
Affiliation(s)
- Iswarya Shanmugam
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology, Vellore. TN, India; School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Sivani Radhakrishnan
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Shradha Santosh
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Akansha Ramnath
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Meghna Anil
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Yogesh Devarajan
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology, Vellore. TN, India; School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Saravanakumar Maheswaran
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology, Vellore. TN, India; School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Vaibav Narayanan
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology, Vellore. TN, India; School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Arunkumar Pitchaimani
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology, Vellore. TN, India; School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India.
| |
Collapse
|
2
|
Slapø NB, Jørgensen KN, Elvsåshagen T, Nerland S, Roelfs D, Valstad M, Timpe CMF, Richard G, Beck D, Sæther LS, Frogner Werner MC, Lagerberg TV, Andreassen OA, Melle I, Agartz I, Westlye LT, Moberget T, Jönsson EG. Relationship between function and structure in the visual cortex in healthy individuals and in patients with severe mental disorders. Psychiatry Res Neuroimaging 2023; 332:111633. [PMID: 37028226 DOI: 10.1016/j.pscychresns.2023.111633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/12/2023] [Accepted: 03/17/2023] [Indexed: 04/09/2023]
Abstract
Patients with schizophrenia spectrum disorders (SCZspect) and bipolar disorders (BD) show impaired function in the primary visual cortex (V1), indicated by altered visual evoked potential (VEP). While the neural substrate for altered VEP in these patients remains elusive, altered V1 structure may play a role. One previous study found a positive relationship between the amplitude of the P100 component of the VEP and V1 surface area, but not V1 thickness, in a small sample of healthy individuals. Here, we aimed to replicate these findings in a larger healthy control (HC) sample (n = 307) and to examine the same relationship in patients with SCZspect (n = 30) or BD (n = 45). We also compared the mean P100 amplitude, V1 surface area and V1 thickness between controls and patients and found no significant group differences. In HC only, we found a significant positive P100-V1 surface area association, while there were no significant P100-V1 thickness relationships in HC, SCZspect or BD. Together, our results confirm previous findings of a positive P100-V1 surface area association in HC, whereas larger patient samples are needed to further clarify the function-structure relationship in V1 in SCZspect and BD.
Collapse
Affiliation(s)
- Nora Berz Slapø
- NORMENT, Institute of Clinical Medicine, University of Oslo, Norway.
| | - Kjetil Nordbø Jørgensen
- NORMENT, Institute of Clinical Medicine, University of Oslo, Norway; Department of Psychiatry, Telemark Hospital, Skien, Norway
| | - Torbjørn Elvsåshagen
- NORMENT, Institute of Clinical Medicine, University of Oslo, Norway; Department of Neurology, Oslo University Hospital, Norway
| | - Stener Nerland
- NORMENT, Institute of Clinical Medicine, University of Oslo, Norway; Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Daniel Roelfs
- NORMENT, Institute of Clinical Medicine, University of Oslo, Norway
| | - Mathias Valstad
- Department of Mental Disorders, Norwegian Institute of Public Health, Norway
| | - Clara M F Timpe
- NORMENT, Institute of Clinical Medicine, University of Oslo, Norway; Department of Psychology, University of Oslo, Norway
| | | | - Dani Beck
- NORMENT, Institute of Clinical Medicine, University of Oslo, Norway; Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | | | | | - Trine Vik Lagerberg
- NORMENT, Division of Mental Health and Addiction, Oslo University hospital, Norway
| | - Ole A Andreassen
- NORMENT, Institute of Clinical Medicine, University of Oslo, Norway; NORMENT, Division of Mental Health and Addiction, Oslo University hospital, Norway
| | - Ingrid Melle
- NORMENT, Institute of Clinical Medicine, University of Oslo, Norway
| | - Ingrid Agartz
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; NORMENT, Division of Mental Health and Addiction, Oslo University hospital, Norway; Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Sciences, Stockholm Region, Stockholm, Sweden
| | - Lars T Westlye
- NORMENT, Institute of Clinical Medicine, University of Oslo, Norway; Department of Psychology, University of Oslo, Norway
| | - Torgeir Moberget
- NORMENT, Institute of Clinical Medicine, University of Oslo, Norway; Department of Behavioral Sciences, Faculty of Health Sciences, Oslo Metropolitan University, OsloMet, Oslo, Norway
| | - Erik G Jönsson
- NORMENT, Institute of Clinical Medicine, University of Oslo, Norway; Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Sciences, Stockholm Region, Stockholm, Sweden
| |
Collapse
|
3
|
Mısır E, Akay GG. Synaptic dysfunction in schizophrenia. Synapse 2023:e22276. [PMID: 37210696 DOI: 10.1002/syn.22276] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 04/25/2023] [Accepted: 05/07/2023] [Indexed: 05/22/2023]
Abstract
Schizophrenia is a chronic disease presented with psychotic symptoms, negative symptoms, impairment in the reward system, and widespread neurocognitive deterioration. Disruption of synaptic connections in neural circuits is responsible for the disease's development and progression. Because deterioration in synaptic connections results in the impaired effective processing of information. Although structural impairments of the synapse, such as a decrease in dendritic spine density, have been shown in previous studies, functional impairments have also been revealed with the development of genetic and molecular analysis methods. In addition to abnormalities in protein complexes regulating exocytosis in the presynaptic region and impaired vesicle release, especially, changes in proteins related to postsynaptic signaling have been reported. In particular, impairments in postsynaptic density elements, glutamate receptors, and ion channels have been shown. At the same time, effects on cellular adhesion molecular structures such as neurexin, neuroligin, and cadherin family proteins were detected. Of course, the confusing effect of antipsychotic use in schizophrenia research should also be considered. Although antipsychotics have positive and negative effects on synapses, studies indicate synaptic deterioration in schizophrenia independent of drug use. In this review, the deterioration in synapse structure and function and the effects of antipsychotics on the synapse in schizophrenia will be discussed.
Collapse
Affiliation(s)
- Emre Mısır
- Department of Psychiatry, Baskent University Faculty of Medicine, Ankara, Turkey
- Department of Interdisciplinary Neuroscience, Ankara University, Ankara, Turkey
| | - Güvem Gümüş Akay
- Department of Interdisciplinary Neuroscience, Ankara University, Ankara, Turkey
- Faculty of Medicine, Department of Physiology, Ankara University, Ankara, Turkey
- Brain Research Center (AÜBAUM), Ankara University, Ankara, Turkey
- Department of Cellular Neuroscience and Advanced Microscopic Neuroimaging, Neuroscience and Neurotechnology Center of Excellence (NÖROM), Ankara, Turkey
| |
Collapse
|
4
|
Ghanbarzehi A, Sepehrinezhad A, Hashemi N, Karimi M, Shahbazi A. Disclosing common biological signatures and predicting new therapeutic targets in schizophrenia and obsessive-compulsive disorder by integrated bioinformatics analysis. BMC Psychiatry 2023; 23:40. [PMID: 36641432 PMCID: PMC9840830 DOI: 10.1186/s12888-023-04543-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
Schizophrenia (SCZ) is a severe mental illness mainly characterized by a number of psychiatric symptoms. Obsessive-compulsive disorder (OCD) is a long-lasting and devastating mental disorder. SCZ has high co-occurrence with OCD resulting in the emergence of a concept entitled "schizo-obsessive disorder" as a new specific clinical entity with more severe psychiatric symptoms. Many studies have been done on SCZ and OCD, but the common pathogenesis between them is not clear yet. Therefore, this study aimed to identify shared genetic basis, potential biomarkers and therapeutic targets between these two disorders. Gene sets were extracted from the Geneweaver and Harmonizome databases for each disorder. Interestingly, the combination of both sets revealed 89 common genes between SCZ and OCD, the most important of which were BDNF, SLC6A4, GAD1, HTR2A, GRIN2B, DRD2, SLC6A3, COMT, TH and DLG4. Then, we conducted a comprehensive bioinformatics analysis of the common genes. Receptor activity as the molecular functions, neuron projection and synapse as the cellular components as well as serotonergic synapse, dopaminergic synapse and alcoholism as the pathways were the most significant commonalities in enrichment analyses. In addition, transcription factor (TFs) analysis predicted significant TFs such as HMGA1, MAPK14, HINFP and TEAD2. Hsa-miR-3121-3p and hsa-miR-495-3p were the most important microRNAs (miRNAs) associated with both disorders. Finally, our study predicted 19 existing drugs (importantly, Haloperidol, Fluoxetine and Melatonin) that may have a potential influence on this co-occurrence. To summarize, this study may help us to better understand and handle the co-occurrence of SCZ and OCD by identifying potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Abdolhakim Ghanbarzehi
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Sepehrinezhad
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Nazanin Hashemi
- Department of Biotechnology, Bangalore University, Bangalore, Karnataka, India
| | - Minoo Karimi
- Department of Audiology, School of Rehabilitation, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Shahbazi
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Schizophrenia and psychedelic state: Dysconnection versus hyper-connection. A perspective on two different models of psychosis stemming from dysfunctional integration processes. Mol Psychiatry 2023; 28:59-67. [PMID: 35931756 DOI: 10.1038/s41380-022-01721-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 07/15/2022] [Accepted: 07/22/2022] [Indexed: 01/07/2023]
Abstract
Psychotic symptoms are a cross-sectional dimension affecting multiple diagnostic categories, despite schizophrenia represents the prototype of psychoses. Initially, dopamine was considered the most involved molecule in the neurobiology of schizophrenia. Over the next years, several biological factors were added to the discussion helping to constitute the concept of schizophrenia as a disease marked by a deficit of functional integration, contributing to the formulation of the Dysconnection Hypothesis in 1995. Nowadays the notion of dysconnection persists in the conceptualization of schizophrenia enriched by neuroimaging findings which corroborate the hypothesis. At the same time, in recent years, psychedelics received a lot of attention by the scientific community and astonishing findings emerged about the rearrangement of brain networks under the effect of these compounds. Specifically, a global decrease in functional connectivity was found, highlighting the disintegration of preserved and functional circuits and an increase of overall connectivity in the brain. The aim of this paper is to compare the biological bases of dysconnection in schizophrenia with the alterations of neuronal cyto-architecture induced by psychedelics and the consequent state of cerebral hyper-connection. These two models of psychosis, despite diametrically opposed, imply a substantial deficit of integration of neural signaling reached through two opposite paths.
Collapse
|
6
|
Dubonyte U, Asenjo-Martinez A, Werge T, Lage K, Kirkeby A. Current advancements of modelling schizophrenia using patient-derived induced pluripotent stem cells. Acta Neuropathol Commun 2022; 10:183. [PMID: 36527106 PMCID: PMC9756764 DOI: 10.1186/s40478-022-01460-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/12/2022] [Indexed: 12/23/2022] Open
Abstract
Schizophrenia (SZ) is a severe psychiatric disorder, with a prevalence of 1-2% world-wide and substantial health- and social care costs. The pathology is influenced by both genetic and environmental factors, however the underlying cause still remains elusive. SZ has symptoms including delusions, hallucinations, confused thoughts, diminished emotional responses, social withdrawal and anhedonia. The onset of psychosis is usually in late adolescence or early adulthood. Multiple genome-wide association and whole exome sequencing studies have provided extraordinary insights into the genetic variants underlying familial as well as polygenic forms of the disease. Nonetheless, a major limitation in schizophrenia research remains the lack of clinically relevant animal models, which in turn hampers the development of novel effective therapies for the patients. The emergence of human induced pluripotent stem cell (hiPSC) technology has allowed researchers to work with SZ patient-derived neuronal and glial cell types in vitro and to investigate the molecular basis of the disorder in a human neuronal context. In this review, we summarise findings from available studies using hiPSC-based neural models and discuss how these have provided new insights into molecular and cellular pathways of SZ. Further, we highlight different examples of how these models have shown alterations in neurogenesis, neuronal maturation, neuronal connectivity and synaptic impairment as well as mitochondrial dysfunction and dysregulation of miRNAs in SZ patient-derived cultures compared to controls. We discuss the pros and cons of these models and describe the potential of using such models for deciphering the contribution of specific human neural cell types to the development of the disease.
Collapse
Affiliation(s)
- Ugne Dubonyte
- Department of Neuroscience and Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, Copenhagen, Denmark
| | - Andrea Asenjo-Martinez
- Department of Neuroscience and Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, Copenhagen, Denmark
| | - Thomas Werge
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine and Lundbeck Foundation Center for GeoGenetics, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
| | - Kasper Lage
- Institute of Biological Psychiatry, Mental Health Services, Copenhagen University Hospital, Copenhagen, Denmark
- Stanley Center for Psychiatric Research and The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Agnete Kirkeby
- Department of Neuroscience and Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, Copenhagen, Denmark.
- Department of Experimental Medical Science and Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
7
|
Yu L, Li Y. Involvement of Intestinal Enteroendocrine Cells in Neurological and Psychiatric Disorders. Biomedicines 2022; 10:biomedicines10102577. [PMID: 36289839 PMCID: PMC9599815 DOI: 10.3390/biomedicines10102577] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/21/2022] [Accepted: 09/21/2022] [Indexed: 11/24/2022] Open
Abstract
Neurological and psychiatric patients have increased dramatically in number in the past few decades. However, effective treatments for these diseases and disorders are limited due to heterogeneous and unclear pathogenic mechanisms. Therefore, further exploration of the biological aspects of the disease, and the identification of novel targets to develop alternative treatment strategies, is urgently required. Systems-level investigations have indicated the potential involvement of the brain–gut axis and intestinal microbiota in the pathogenesis and regulation of neurological and psychiatric disorders. While intestinal microbiota is crucial for maintaining host physiology, some important sensory and regulatory cells in the host should not be overlooked. Intestinal epithelial enteroendocrine cells (EECs) residing in the epithelium throughout intestine are the key regulators orchestrating the communication along the brain-gut-microbiota axis. On one hand, EECs sense changes in luminal microorganisms via microbial metabolites; on the other hand, they communicate with host body systems via neuroendocrine molecules. Therefore, EECs are believed to play important roles in neurological and psychiatric disorders. This review highlights the involvement of EECs and subtype cells, via secretion of endocrine molecules, in the development and regulation of neurological and psychiatric disorders, including Parkinson’s disease (PD), schizophrenia, visceral pain, neuropathic pain, and depression. Moreover, the current paper summarizes the potential mechanism of EECs in contributing to disease pathogenesis. Examination of these mechanisms may inspire and lead to the development of new aspects of treatment strategies for neurological and psychiatric disorders in the future.
Collapse
Affiliation(s)
- Liangen Yu
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19716, USA
| | - Yihang Li
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19716, USA
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
- Correspondence:
| |
Collapse
|
8
|
Eyolfson E, Carr T, Fraunberger E, Khan A, Clark I, Mychasiuk R, Lohman AW. Repeated mild traumatic brain injuries in mice cause age- and sex-specific alterations in dendritic spine density. Exp Neurol 2022; 357:114172. [PMID: 35863503 DOI: 10.1016/j.expneurol.2022.114172] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/08/2022] [Accepted: 07/14/2022] [Indexed: 11/25/2022]
Abstract
Mild traumatic brain injuries (mTBI) plague the human population and their prevalence is increasing annually. More so, repeated mTBIs (RmTBI) are known to manifest and compound neurological deficits in vulnerable populations. Age at injury and sex are two important factors influencing RmTBI pathophysiology, but we continue to know little about the specific effects of RmTBI in youth and females. In this study, we directly quantified the effects of RmTBI on adolescent and adult, male and female mice, with a closed-head lateral impact model. We report age- and sex-specific neurobehavioural deficits in motor function and working memory, microglia responses to injury, and the subsequent changes in dendritic spine density in select brain regions. Specifically, RmTBI caused increased footslips in adult male mice as assessed in a beam walk assay and significantly reduced the time spent with a novel object in adolescent male and female mice. RmTBIs caused a significant reduction in microglia density in male mice in the motor cortex, but not female mice. Finally, RmTBI significantly reduced dendritic spine density in the agranular insular cortex (a region of the prefrontal cortex in mice) and increased dendritic spine density in the adolescent male motor cortex. Together, the data provided in this study sheds new light on the heterogeneity in RmTBI-induced behavioural, glial, and neuronal architecture changes dependent on age and sex.
Collapse
Affiliation(s)
- Eric Eyolfson
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada.
| | - Thomas Carr
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada.
| | - Erik Fraunberger
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada.
| | - Asher Khan
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada.
| | - Isabel Clark
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada.
| | - Richelle Mychasiuk
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada; Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia.
| | - Alexander W Lohman
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
9
|
Multi-Omics Analysis Reveals Myelin, Presynaptic and Nicotinate Alterations in the Hippocampus of G72/G30 Transgenic Mice. J Pers Med 2022; 12:jpm12020244. [PMID: 35207732 PMCID: PMC8878587 DOI: 10.3390/jpm12020244] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 01/15/2023] Open
Abstract
The primate-specific G72/G30 gene locus has been associated with major psychiatric disorders, such as schizophrenia and bipolar disorder. We have previously generated transgenic mice which carry the G72/G30 locus and express the longest G72 splice variant (LG72) protein encoded by this locus with schizophrenia-related symptoms. Here, we used a multi-omics approach, including quantitative proteomics and metabolomics to investigate molecular alterations in the hippocampus of G72/G30 transgenic (G72Tg) mice. Our proteomics analysis revealed decreased expression of myelin-related proteins and NAD-dependent protein deacetylase sirtuin-2 (Sirt2) as well as increased expression of the scaffolding presynaptic proteins bassoon (Bsn) and piccolo (Pclo) and the cytoskeletal protein plectin (Plec1) in G72Tg compared to wild-type (WT) mice. Metabolomics analysis indicated decreased levels of nicotinate in G72Tg compared to WT hippocampi. Decreased hippocampal protein expression for selected proteins, namely myelin oligodentrocyte glycoprotein (Mog), Cldn11 and myelin proteolipid protein (Plp), was confirmed with Western blot in a larger population of G72Tg and WT mice. The identified molecular pathway alterations shed light on the hippocampal function of LG72 protein in the context of neuropsychiatric phenotypes.
Collapse
|
10
|
Julayanont P, Suryadevara U. Psychosis. Continuum (Minneap Minn) 2021; 27:1682-1711. [PMID: 34881732 DOI: 10.1212/con.0000000000001013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PURPOSE OF REVIEW Psychosis can manifest in primary psychotic disorders, neurologic diseases, and medical conditions. This article reviews the definition of psychotic symptoms and the evaluation and management of psychosis in primary psychiatric and neurologic disorders frequently seen in neurologic practice. RECENT FINDINGS Emerging evidence supports significant connections between psychosis and structural and functional brain changes in both primary psychotic and neurologic disorders. In addition to antidopaminergic activity, the mechanism of new-generation antipsychotics shifts to act on serotonin receptors, which potentially contributes to their benefits in the treatment of negative symptoms of psychosis and a lesser frequency of extrapyramidal side effects compared with typical antipsychotics. This is also helpful in the treatment of psychosis in patients who have neurodegenerative diseases and are vulnerable to developing extrapyramidal side effects from typical antipsychotics. SUMMARY Even with significant overlap, management of psychosis in primary psychotic disorders differs from the approach of psychosis in neurologic diseases. This article helps clinicians learn how to practically evaluate psychosis from both psychiatric and neurologic perspectives.
Collapse
|
11
|
In vivo evidence of lower synaptic vesicle density in schizophrenia. Mol Psychiatry 2021; 26:7690-7698. [PMID: 34135473 DOI: 10.1038/s41380-021-01184-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 05/20/2021] [Accepted: 05/28/2021] [Indexed: 02/05/2023]
Abstract
Decreased synaptic spine density has been the most consistently reported postmortem finding in schizophrenia (SCZ). A recently developed in vivo measure of synaptic vesicle density estimated using the novel positron emission tomography (PET) ligand [11C]UCB-J is a proxy measure of synaptic density. In this study we determined whether [11C]UCB-J binding, an in vivo measure of synaptic vesicle density, is altered in SCZ. SCZ patients (n = 13, 3 F) and age-, gender-matched healthy controls (HCs) (n = 15, 3 F) underwent PET imaging using [11C]UCB-J and high-resolution research tomography (HRRT). [11C]UCB-J distribution volume (VT) and binding potential (BPND) were estimated using a 1T model with centrum-semiovale as the reference region. Relative to HCs, SCZ patients, showed significantly lower [11C]UCB-J BPND with significant differences in the frontal cortex (-10%, Cohen's d = 1.01), anterior cingulate (-11%, Cohen's d = 1.24), hippocampus (-15%, Cohen's d = 1.29), occipital cortex (-14%, Cohen's d = 1.34), parietal cortex (-10%, p = 0.03, Cohen's d = 0.85) and temporal cortex (-11%, Cohen's d = 1.23). These differences remained significant after partial volume correction. [11C]UCB-J BPND did not correlate with cumulative antipsychotic exposure or gray-matter volume. Consistent with the postmortem and in vivo findings, synaptic vesicle density is lower across several brain regions in SCZ. Frontal synaptic vesicle density correlated with psychosis symptom severity and cognitive performance on social cognition and processing speed. These findings indicate that [11C]UCB-J PET is a sensitive tool to detect lower synaptic density in SCZ and holds promise for future studies of early detection and disease progression.
Collapse
|
12
|
Liu X, Ying J, Wang X, Zheng Q, Zhao T, Yoon S, Yu W, Yang D, Fang Y, Hua F. Astrocytes in Neural Circuits: Key Factors in Synaptic Regulation and Potential Targets for Neurodevelopmental Disorders. Front Mol Neurosci 2021; 14:729273. [PMID: 34658786 PMCID: PMC8515196 DOI: 10.3389/fnmol.2021.729273] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/02/2021] [Indexed: 12/14/2022] Open
Abstract
Astrocytes are the major glial cells in the brain, which play a supporting role in the energy and nutritional supply of neurons. They were initially regarded as passive space-filling cells, but the latest progress in the study of the development and function of astrocytes highlights their active roles in regulating synaptic transmission, formation, and plasticity. In the concept of "tripartite synapse," the bidirectional influence between astrocytes and neurons, in addition to their steady-state and supporting function, suggests that any negative changes in the structure or function of astrocytes will affect the activity of neurons, leading to neurodevelopmental disorders. The role of astrocytes in the pathophysiology of various neurological and psychiatric disorders caused by synaptic defects is increasingly appreciated. Understanding the roles of astrocytes in regulating synaptic development and the plasticity of neural circuits could help provide new treatments for these diseases.
Collapse
Affiliation(s)
- Xing Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Jun Ying
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Xifeng Wang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qingcui Zheng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Tiancheng Zhao
- Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Sungtae Yoon
- Helping Minds International Charitable Foundation, New York, NY, United States
| | - Wen Yu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Danying Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Yang Fang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, China
| |
Collapse
|
13
|
Kelly JR, Minuto C, Cryan JF, Clarke G, Dinan TG. The role of the gut microbiome in the development of schizophrenia. Schizophr Res 2021; 234:4-23. [PMID: 32336581 DOI: 10.1016/j.schres.2020.02.010] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 02/22/2020] [Accepted: 02/25/2020] [Indexed: 02/07/2023]
Abstract
Schizophrenia is a heterogeneous neurodevelopmental disorder involving the convergence of a complex and dynamic bidirectional interaction of genetic expression and the accumulation of prenatal and postnatal environmental risk factors. The development of the neural circuitry underlying social, cognitive and emotional domains requires precise regulation from molecular signalling pathways, especially during critical periods or "windows", when the brain is particularly sensitive to the influence of environmental input signalling. Many of the brain regions involved, and the molecular substrates sub-serving these domains are responsive to life-long microbiota-gut-brain (MGB) axis signalling. This intricate microbial signalling system communicates with the brain via the vagus nerve, immune system, enteric nervous system, enteroendocrine signalling and production of microbial metabolites, such as short-chain fatty acids. Preclinical data has demonstrated that MGB axis signalling influences neurotransmission, neurogenesis, myelination, dendrite formation and blood brain barrier development, and modulates cognitive function and behaviour patterns, such as, social interaction, stress management and locomotor activity. Furthermore, preliminary clinical studies suggest altered gut microbiota profiles in schizophrenia. Unravelling MGB axis signalling in the context of an evolving dimensional framework in schizophrenia may provide a more complete understanding of the neurobiological architecture of this complex condition and offers the possibility of translational interventions.
Collapse
Affiliation(s)
- John R Kelly
- Department of Psychiatry, Trinity College Dublin, Ireland
| | - Chiara Minuto
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland
| | - Timothy G Dinan
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland.
| |
Collapse
|
14
|
Mastrandrea R, Piras F, Gabrielli A, Banaj N, Caldarelli G, Spalletta G, Gili T. The unbalanced reorganization of weaker functional connections induces the altered brain network topology in schizophrenia. Sci Rep 2021; 11:15400. [PMID: 34321538 PMCID: PMC8319172 DOI: 10.1038/s41598-021-94825-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/08/2021] [Indexed: 01/10/2023] Open
Abstract
Network neuroscience shed some light on the functional and structural modifications occurring to the brain associated with the phenomenology of schizophrenia. In particular, resting-state functional networks have helped our understanding of the illness by highlighting the global and local alterations within the cerebral organization. We investigated the robustness of the brain functional architecture in 44 medicated schizophrenic patients and 40 healthy comparators through an advanced network analysis of resting-state functional magnetic resonance imaging data. The networks in patients showed more resistance to disconnection than in healthy controls, with an evident discrepancy between the two groups in the node degree distribution computed along a percolation process. Despite a substantial similarity of the basal functional organization between the two groups, the expected hierarchy of healthy brains' modular organization is crumbled in schizophrenia, showing a peculiar arrangement of the functional connections, characterized by several topologically equivalent backbones. Thus, the manifold nature of the functional organization's basal scheme, together with its altered hierarchical modularity, may be crucial in the pathogenesis of schizophrenia. This result fits the disconnection hypothesis that describes schizophrenia as a brain disorder characterized by an abnormal functional integration among brain regions.
Collapse
Affiliation(s)
| | - Fabrizio Piras
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, 00179, Rome, Italy
| | - Andrea Gabrielli
- Dipartimento di Ingegneria, Università Roma Tre, 00146, Rome, Italy.,Istituto dei Sistemi Complessi (ISC)-CNR, UoS Sapienza, Dipartimento di Fisica, Università "Sapienza", 00185, Rome, Italy
| | - Nerisa Banaj
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, 00179, Rome, Italy
| | - Guido Caldarelli
- Networks Unit, IMT School for Advanced Studies, 55100, Lucca, Italy.,Istituto dei Sistemi Complessi (ISC)-CNR, UoS Sapienza, Dipartimento di Fisica, Università "Sapienza", 00185, Rome, Italy
| | - Gianfranco Spalletta
- Laboratory of Neuropsychiatry, IRCCS Santa Lucia Foundation, 00179, Rome, Italy. .,Division of Neuropsychiatry, Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Tommaso Gili
- Networks Unit, IMT School for Advanced Studies, 55100, Lucca, Italy
| |
Collapse
|
15
|
Millán AP, Torres JJ, Johnson S, Marro J. Growth strategy determines the memory and structural properties of brain networks. Neural Netw 2021; 142:44-56. [PMID: 33984735 DOI: 10.1016/j.neunet.2021.04.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 03/04/2021] [Accepted: 04/20/2021] [Indexed: 11/18/2022]
Abstract
The interplay between structure and function affects the emerging properties of many natural systems. Here we use an adaptive neural network model that couples activity and topological dynamics and reproduces the experimental temporal profiles of synaptic density observed in the brain. We prove that the existence of a transient period of relatively high synaptic connectivity is critical for the development of the system under noise circumstances, such that the resulting network can recover stored memories. Moreover, we show that intermediate synaptic densities provide optimal developmental paths with minimum energy consumption, and that ultimately it is the transient heterogeneity in the network that determines its evolution. These results could explain why the pruning curves observed in actual brain areas present their characteristic temporal profiles and they also suggest new design strategies to build biologically inspired neural networks with particular information processing capabilities.
Collapse
Affiliation(s)
- Ana P Millán
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Clinical Neurophysiology and MEG Center, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, The Netherlands.
| | - Joaquín J Torres
- Institute 'Carlos I' for Theoretical and Computational Physics, University of Granada, Spain
| | - Samuel Johnson
- School of Mathematics, University of Birmingham, Edgbaston B15 2TT, UK; Alan Turing Institute, London NW1 2DB, UK
| | - J Marro
- Institute 'Carlos I' for Theoretical and Computational Physics, University of Granada, Spain
| |
Collapse
|
16
|
Briana DD, Malamitsi-Puchner A. Chorioamnionitis in utero, schizophrenia in adulthood: limited current evidence-future research focus? J Matern Fetal Neonatal Med 2021; 35:4782-4787. [PMID: 33435777 DOI: 10.1080/14767058.2020.1863370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Background: Developmental adaptive processes during gestation that are known to be involved in permanent changes in physiology and metabolism or "early life programming" can adversely affect fetal brain development, impacting both brain structure and function.Data: Emerging evidence strongly supports the developmental origin of schizophrenia, which may potentially be a result of prenatal exposure to a diversity of factors, especially infections, in genetically predisposed subjects. Structural and functional brain changes during development of schizophrenia are determined by genetic components, altered expression of schizophrenia risk genes and epigenetic dysregulation. However, the precise mechanisms underlying these relationships remain unclear. Findings from human and animal studies suggest that inflammatory-immune responses and activation of oxidative stress pathways are crucial in mediating intrauterine infection-induced neurodevelopmental and neuropsychiatric diseases.Aim: Considering the high prevalence of intrauterine inflammation in the context of chorioamnionitis during human pregnancy and the paucity of knowledge on fetal programming of schizophrenia, this mini review aims to exclusively consolidate the current evidence supporting a potential association between chorioamnionitis and schizophrenia.
Collapse
Affiliation(s)
- Despina D Briana
- NICU, 3rd Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ariadne Malamitsi-Puchner
- NICU, 3rd Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
17
|
Yatchenko Y, Ben-Shachar D. Update of Mitochondrial Network Analysis by Imaging: Proof of Technique in Schizophrenia. Methods Mol Biol 2021; 2277:187-201. [PMID: 34080153 DOI: 10.1007/978-1-0716-1270-5_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Mitochondria, similar to living cells and organelles, have a negative membrane potential, which ranges between (-108) and (150) mV as compared to (-70) and (-90) mV of the plasma membrane. Therefore, permeable lipophilic cations tend to accumulate in the mitochondria. Those cations which exhibit fluorescence activity after accumulation into energized systems are widely used to decipher changes in membrane potential by imaging techniques. Here we describe the use of two different dyes for labeling mitochondrial membrane potential (Δψm) in live cells. One is the lipophilic cation 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazol-carbocyanine iodide (JC-1), which alters reversibly its color from green (J-monomer, at its low concentration in the cytosol) to red (J-aggregates, at its high concentration in active mitochondria) with increasing mitochondrial membrane potential (Δψm). The other is MitoTracker® Orange, a mitochondrion-selective probe which passively diffuses across the plasma membrane and accumulates in active mitochondria depending on their Δψm. We show that in addition to changes in Δψm, these specific dyes can be used to follow alterations in mitochondrial distribution and mitochondrial network connectivity. We suggest that JC-1 is a preferable probe to compare between different cell types and cell state, as a red to green ratio of fluorescence intensities is used for analysis. This ratio depends only on the mitochondrial membrane potential and not on other cellular and/or mitochondrial-dependent or independent factors that may alter, for example, due to treatment or disease state. However, in cells labeled either with green or red fluorescence protein, JC-1 cannot be used. Therefore, other dyes are preferable. We demonstrate various applications of JC-1 and MitoTracker Orange staining to study mitochondrial abnormalities in different cell types derived from schizophrenia patients and healthy subjects.
Collapse
Affiliation(s)
- Yekaterina Yatchenko
- Laboratory of Psychobiology, Department of Neuroscience, B. Rappaport Faculty of Medicine, Technion IIT, Haifa, Israel
| | - Dorit Ben-Shachar
- Laboratory of Psychobiology, Department of Neuroscience, B. Rappaport Faculty of Medicine, Technion IIT, Haifa, Israel.
| |
Collapse
|
18
|
Tzioras M, Stevenson AJ, Boche D, Spires-Jones TL. Microglial contribution to synaptic uptake in the prefrontal cortex in schizophrenia. Neuropathol Appl Neurobiol 2020; 47:346-351. [PMID: 32892388 DOI: 10.1111/nan.12660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/22/2020] [Indexed: 01/19/2023]
Abstract
Microglia in human post-mortem tissue in schizophrenia patients' brains engulf synaptic material, but not differently to age-matched non-neurological control brains. Also, schizophrenia brains display similar levels of microgliosis to control brains.
Collapse
Affiliation(s)
- M Tzioras
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK.,Centre for Brain Discovery Sciences, The University of Edinburgh, Edinburgh, UK
| | - A J Stevenson
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK.,Centre for Brain Discovery Sciences, The University of Edinburgh, Edinburgh, UK
| | - D Boche
- Clinical Neurosciences, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - T L Spires-Jones
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK.,Centre for Brain Discovery Sciences, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
19
|
Cheadle L, Rivera SA, Phelps JS, Ennis KA, Stevens B, Burkly LC, Lee WCA, Greenberg ME. Sensory Experience Engages Microglia to Shape Neural Connectivity through a Non-Phagocytic Mechanism. Neuron 2020; 108:451-468.e9. [PMID: 32931754 DOI: 10.1016/j.neuron.2020.08.002] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 05/10/2020] [Accepted: 08/05/2020] [Indexed: 12/21/2022]
Abstract
Sensory experience remodels neural circuits in the early postnatal brain through mechanisms that remain to be elucidated. Applying a new method of ultrastructural analysis to the retinogeniculate circuit, we find that visual experience alters the number and structure of synapses between the retina and the thalamus. These changes require vision-dependent transcription of the receptor Fn14 in thalamic relay neurons and the induction of its ligand TWEAK in microglia. Fn14 functions to increase the number of bulbous spine-associated synapses at retinogeniculate connections, likely contributing to the strengthening of the circuit that occurs in response to visual experience. However, at retinogeniculate connections near TWEAK-expressing microglia, TWEAK signals via Fn14 to restrict the number of bulbous spines on relay neurons, leading to the elimination of a subset of connections. Thus, TWEAK and Fn14 represent an intercellular signaling axis through which microglia shape retinogeniculate connectivity in response to sensory experience.
Collapse
Affiliation(s)
- Lucas Cheadle
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Samuel A Rivera
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Jasper S Phelps
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Program in Neuroscience, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Katelin A Ennis
- Research and Early Development, Biogen, 115 Broadway, Cambridge, MA 04142, USA
| | - Beth Stevens
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Linda C Burkly
- Research and Early Development, Biogen, 115 Broadway, Cambridge, MA 04142, USA
| | - Wei-Chung Allen Lee
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Michael E Greenberg
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
20
|
Woo JJ, Pouget JG, Zai CC, Kennedy JL. The complement system in schizophrenia: where are we now and what's next? Mol Psychiatry 2020; 25:114-130. [PMID: 31439935 DOI: 10.1038/s41380-019-0479-0] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 02/06/2019] [Accepted: 02/11/2019] [Indexed: 12/24/2022]
Abstract
The complement system is a set of immune proteins involved in first-line defense against pathogens and removal of waste materials. Recent evidence has implicated the complement cascade in diseases involving the central nervous system, including schizophrenia. Here, we provide an up-to-date narrative review and critique of the literature on the relationship between schizophrenia and complement gene polymorphisms, gene expression, protein concentration, and pathway activity. A literature search identified 23 new studies since the first review on this topic in 2008. Overall complement pathway activity appears to be elevated in schizophrenia. Recent studies have identified complement component 4 (C4) and CUB and Sushi Multiple Domains 1 (CSMD1) as potential genetic markers of schizophrenia. In particular, there is some evidence of higher rates of C4B/C4S deficiency, reduced peripheral C4B concentration, and elevated brain C4A mRNA expression in schizophrenia patients compared to controls. To better elucidate the additive effects of multiple complement genotypes, we also conducted gene- and gene-set analysis through MAGMA which supported the role of Human Leukocyte Antigen class (HLA) III genes and, to a lesser extent, CSMD1 in schizophrenia; however, the HLA-schizophrenia association was likely driven by the C4 gene. Lastly, we identified several limitations of the literature on the complement system and schizophrenia, including: small sample sizes, inconsistent methodologies, limited measurements of neural concentrations of complement proteins, little exploration of the link between complement and schizophrenia phenotype, and lack of studies exploring schizophrenia treatment response. Overall, recent findings highlight complement components-in particular, C4 and CSMD1-as potential novel drug targets in schizophrenia. Given the growing availability of complement-targeted therapies, future clinical studies evaluating their efficacy in schizophrenia hold the potential to accelerate treatment advances.
Collapse
Affiliation(s)
- Julia J Woo
- Tanenbaum Centre for Pharmacogenetics, Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, 250 College Street, Toronto, ON, M5T 1R8, Canada
| | - Jennie G Pouget
- Tanenbaum Centre for Pharmacogenetics, Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, 250 College Street, Toronto, ON, M5T 1R8, Canada
| | - Clement C Zai
- Tanenbaum Centre for Pharmacogenetics, Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, 250 College Street, Toronto, ON, M5T 1R8, Canada
| | - James L Kennedy
- Tanenbaum Centre for Pharmacogenetics, Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, 250 College Street, Toronto, ON, M5T 1R8, Canada.
| |
Collapse
|
21
|
Rodrigues-Amorim D, Rivera-Baltanás T, Vallejo-Curto MDC, Rodriguez-Jamardo C, de las Heras E, Barreiro-Villar C, Blanco-Formoso M, Fernández-Palleiro P, Álvarez-Ariza M, López M, García-Caballero A, Olivares JM, Spuch C. Proteomics in Schizophrenia: A Gateway to Discover Potential Biomarkers of Psychoneuroimmune Pathways. Front Psychiatry 2019; 10:885. [PMID: 31849731 PMCID: PMC6897280 DOI: 10.3389/fpsyt.2019.00885] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 11/11/2019] [Indexed: 12/17/2022] Open
Abstract
Schizophrenia is a severe and disabling psychiatric disorder with a complex and multifactorial etiology. The lack of consensus regarding the multifaceted dysfunction of this ailment has increased the need to explore new research lines. This research makes use of proteomics data to discover possible analytes associated with psychoneuroimmune signaling pathways in schizophrenia. Thus, we analyze plasma of 45 patients [10 patients with first-episode schizophrenia (FES) and 35 patients with chronic schizophrenia] and 43 healthy subjects by label-free liquid chromatography-tandem mass spectrometry. The analysis revealed a significant reduction in the levels of glia maturation factor beta (GMF-β), the brain-derived neurotrophic factor (BDNF), and the 115-kDa isoform of the Rab3 GTPase-activating protein catalytic subunit (RAB3GAP1) in patients with schizophrenia as compared to healthy volunteers. In conclusion, GMF-β, BDNF, and 115-kDa isoform of RAB3GAP1 showed significantly reduced levels in plasma of patients with schizophrenia, thus making them potential biomarkers in schizophrenia.
Collapse
Affiliation(s)
- Daniela Rodrigues-Amorim
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - Tania Rivera-Baltanás
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - María del Carmen Vallejo-Curto
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - Cynthia Rodriguez-Jamardo
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - Elena de las Heras
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - Carolina Barreiro-Villar
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - María Blanco-Formoso
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - Patricia Fernández-Palleiro
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - María Álvarez-Ariza
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - Marta López
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - Alejandro García-Caballero
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
- Department of Psychiatry, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - José Manuel Olivares
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| | - Carlos Spuch
- Translational Neuroscience Research Group, Galicia Sur Health Research Institute, University of Vigo, CIBERSAM, Vigo, Spain
| |
Collapse
|
22
|
Further delineation of neuropsychiatric findings in Tatton-Brown-Rahman syndrome due to disease-causing variants in DNMT3A: seven new patients. Eur J Hum Genet 2019; 28:469-479. [PMID: 31685998 DOI: 10.1038/s41431-019-0485-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 06/20/2019] [Accepted: 07/02/2019] [Indexed: 12/14/2022] Open
Abstract
Tatton-Brown-Rahman (TBRS) syndrome is a recently described overgrowth syndrome caused by loss of function variants in the DNMT3A gene. This gene encodes for a DNA methyltransferase 3 alpha, which is involved in epigenetic regulation, especially during embryonic development. Somatic variants in DNMT3A have been widely studied in different types of tumors, including acute myeloid leukemia, hematopoietic, and lymphoid cancers. Germline gain-of-function variants in this gene have been recently implicated in microcephalic dwarfism. Common clinical features of patients with TBRS include tall stature, macrocephaly, intellectual disability (ID), and a distinctive facial appearance. Differential diagnosis of TBRS comprises Sotos, Weaver, and Malan Syndromes. The majority of these disorders present other clinical features with a high clinical overlap, making necessary a molecular confirmation of the clinical diagnosis. We here describe seven new patients with variants in DNMT3A, four of them with neuropsychiatric disorders, including schizophrenia and psychotic behavior. In addition, one of the patients has developed a brain tumor in adulthood. This patient has also cerebral atrophy, aggressive behavior, ID, and abnormal facial features. Clinical evaluation of this group of patients should include a complete neuropsychiatric assessment together with psychological support in order to detect and manage abnormal behaviors such as aggressiveness, impulsivity, and attention deficit-hyperactivity disorder. TBRS should be suspected in patients with overgrowth, ID, tall stature, and macrocephaly, who also have some neuropsychiatric disorders without any genetic defects in the commonest overgrowth disorders. Molecular confirmation in these patients is mandatory.
Collapse
|
23
|
Applying vinpocetine to reverse synaptic ultrastructure by regulating BDNF-related PSD-95 in alleviating schizophrenia-like deficits in rat. Compr Psychiatry 2019; 94:152122. [PMID: 31473552 DOI: 10.1016/j.comppsych.2019.152122] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 08/08/2019] [Accepted: 08/16/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Schizophrenia is a mental disorder characterized by hyperlocomotion, cognitive symptoms, and social withdrawal. Brain-derived neurotrophic factor (BDNF) and postsynaptic density (PSD)-95 are related to schizophrenia-like deficits via regulating the synaptic ultrastructure, and play a role in drug therapy. Vinpocetine is a nootropic phosphodiesterase-1 (PDE-1) inhibitor that can reverse ketamine-induced schizophrenia-like deficits by increasing BDNF expression. However, the effects of vinpocetine on alleviating schizophrenia-like deficits via reversing the synaptic ultrastructure by regulating BDNF-related PSD-95 have not been sufficiently studied. METHODS In this study, the schizophrenic model was built using ketamine (30 mg/kg) for 14 consecutive days. The effect of vinpocetine on reversing schizophrenia-like behaviors was examined via behavioral testing followed by treatment with certain doses of vinpocetine (20 mg/kg, i.p.). The BDNF and PSD-95 levels in the posterior cingulate cortex (PCC) were measured using biochemical assessments. In addition, the synaptic ultrastructure was observed using transmission electron microscopy (TEM). RESULTS Ketamine induced drastic schizophrenia-like behaviors, lower protein levels of BDNF and PSD-95, and a change in the synaptic ultrastructure in the PCC. After treatment, the vinpocetine revealed a marked amendment in schizophrenia-like behaviors induced by ketamine, including higher locomotor behavior, lower cognitive behavior, and social withdrawal defects. Vinpocetine could increase the PSD-95 protein level by up-regulating the expression of BDNF. In addition, the synaptic ultrastructure was changed after vinpocetine administration, including a reduction in the thickness and curvature of the synaptic interface, as well as an increase in synaptic cleft width in the PCC. CONCLUSION Vinpocetine can reverse the synaptic ultrastructure by regulating BDNF-related PSD-95 to alleviate schizophrenia-like deficits induced by ketamine in rats.
Collapse
|
24
|
Negr1 controls adult hippocampal neurogenesis and affective behaviors. Mol Psychiatry 2019; 24:1189-1205. [PMID: 30651602 DOI: 10.1038/s41380-018-0347-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 12/03/2018] [Accepted: 12/10/2018] [Indexed: 01/08/2023]
Abstract
Recent genome-wide association studies on major depressive disorder have implicated neuronal growth regulator 1 (Negr1), a GPI-anchored cell adhesion molecule in the immunoglobulin LON family. Although Negr1 has been shown to regulate neurite outgrowth and synapse formation, the mechanism through which this protein affects mood disorders is still largely unknown. In this research, we characterized Negr1-deficient (negr1-/-) mice to elucidate the function of Negr1 in anxiety and depression. We found that anxiety- and depression-like behaviors increased in negr1-/- mice compared with wild-type mice. In addition, negr1-/- mice had decreased adult hippocampal neurogenesis compared to wild-type mice. Concurrently, both LTP and mEPSC in the dentate gyrus (DG) region were severely compromised in negr1-/- mice. In our effort to elucidate the underlying molecular mechanisms, we found that lipocalin-2 (Lcn2) expression was decreased in the hippocampus of negr1-/- mice compared to wild-type mice. Heterologous Lcn2 expression in the hippocampal DG of negr1-/- mice rescued anxiety- and depression-like behaviors and restored neurogenesis and mEPSC frequency to their normal levels in these mice. Furthermore, we discovered that Negr1 interacts with leukemia inhibitory factor receptor (LIFR) and modulates LIF-induced Lcn2 expression. Taken together, our data uncovered a novel mechanism of mood regulation by Negr1 involving an interaction between Negr1 and LIFR along with Lcn2 expression.
Collapse
|
25
|
Lee G, Zhou Y. NMDAR Hypofunction Animal Models of Schizophrenia. Front Mol Neurosci 2019; 12:185. [PMID: 31417356 PMCID: PMC6685005 DOI: 10.3389/fnmol.2019.00185] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/17/2019] [Indexed: 12/20/2022] Open
Abstract
The N-methyl-d-aspartate receptor (NMDAR) hypofunction hypothesis has been proposed to help understand the etiology and pathophysiology of schizophrenia. This hypothesis was based on early observations that NMDAR antagonists could induce a full range of symptoms of schizophrenia in normal human subjects. Accumulating evidence in humans and animal studies points to NMDAR hypofunctionality as a convergence point for various symptoms of schizophrenia. Here we review animal models of NMDAR hypofunction generated by pharmacological and genetic approaches, and how they relate to the pathophysiology of schizophrenia. In addition, we discuss the limitations of animal models of NMDAR hypofunction and their potential utility for therapeutic applications.
Collapse
Affiliation(s)
| | - Yi Zhou
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| |
Collapse
|
26
|
Shimozono M, Osaka J, Kato Y, Araki T, Kawamura H, Takechi H, Hakeda-Suzuki S, Suzuki T. Cell surface molecule, Klingon, mediates the refinement of synaptic specificity in the Drosophila visual system. Genes Cells 2019; 24:496-510. [PMID: 31124270 DOI: 10.1111/gtc.12703] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 05/04/2019] [Accepted: 05/19/2019] [Indexed: 11/29/2022]
Abstract
In the Drosophila brain, neurons form genetically specified synaptic connections with defined neuronal targets. It is proposed that each central nervous system neuron expresses specific cell surface proteins, which act as identification tags. Through an RNAi screen of cell surface molecules in the Drosophila visual system, we found that the cell adhesion molecule Klingon (Klg) plays an important role in repressing the ectopic formation of extended axons, preventing the formation of excessive synapses. Cell-specific manipulation of klg showed that Klg is required in both photoreceptors and the glia, suggesting that the balanced homophilic interaction between photoreceptor axons and the glia is required for normal synapse formation. Previous studies suggested that Klg binds to cDIP and our genetic analyses indicate that cDIP is required in glia for ectopic synaptic repression. These data suggest that Klg play a critical role together with cDIP in refining synaptic specificity and preventing unnecessary connections in the brain.
Collapse
Affiliation(s)
- Mai Shimozono
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokahama, Kanagawa, Japan
| | - Jiro Osaka
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokahama, Kanagawa, Japan
| | - Yuya Kato
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokahama, Kanagawa, Japan
| | - Tomohiro Araki
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokahama, Kanagawa, Japan
| | - Hinata Kawamura
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokahama, Kanagawa, Japan
| | - Hiroki Takechi
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokahama, Kanagawa, Japan
| | - Satoko Hakeda-Suzuki
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokahama, Kanagawa, Japan
| | - Takashi Suzuki
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokahama, Kanagawa, Japan
| |
Collapse
|
27
|
Millán AP, Torres JJ, Marro J. How Memory Conforms to Brain Development. Front Comput Neurosci 2019; 13:22. [PMID: 31057385 PMCID: PMC6477510 DOI: 10.3389/fncom.2019.00022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 03/26/2019] [Indexed: 12/20/2022] Open
Abstract
Nature exhibits countless examples of adaptive networks, whose topology evolves constantly coupled with the activity due to its function. The brain is an illustrative example of a system in which a dynamic complex network develops by the generation and pruning of synaptic contacts between neurons while memories are acquired and consolidated. Here, we consider a recently proposed brain developing model to study how mechanisms responsible for the evolution of brain structure affect and are affected by memory storage processes. Following recent experimental observations, we assume that the basic rules for adding and removing synapses depend on local synaptic currents at the respective neurons in addition to global mechanisms depending on the mean connectivity. In this way a feedback loop between "form" and "function" spontaneously emerges that influences the ability of the system to optimally store and retrieve sensory information in patterns of brain activity or memories. In particular, we report here that, as a consequence of such a feedback-loop, oscillations in the activity of the system among the memorized patterns can occur, depending on parameters, reminding mind dynamical processes. Such oscillations have their origin in the destabilization of memory attractors due to the pruning dynamics, which induces a kind of structural disorder or noise in the system at a long-term scale. This constantly modifies the synaptic disorder induced by the interference among the many patterns of activity memorized in the system. Such new intriguing oscillatory behavior is to be associated only to long-term synaptic mechanisms during the network evolution dynamics, and it does not depend on short-term synaptic processes, as assumed in other studies, that are not present in our model.
Collapse
Affiliation(s)
| | - Joaquín J. Torres
- Institute “Carlos I” for Theoretical and Computational Physics, University of Granada, Granada, Spain
| | | |
Collapse
|
28
|
Laskaris L, Zalesky A, Weickert CS, Di Biase MA, Chana G, Baune BT, Bousman C, Nelson B, McGorry P, Everall I, Pantelis C, Cropley V. Investigation of peripheral complement factors across stages of psychosis. Schizophr Res 2019; 204:30-37. [PMID: 30527272 DOI: 10.1016/j.schres.2018.11.035] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 10/02/2018] [Accepted: 11/30/2018] [Indexed: 12/21/2022]
Abstract
The complement cascade has been proposed to contribute to the pathogenesis of schizophrenia. However, it remains unclear whether peripheral complement levels differ in cases compared to controls, change over the course of illness and whether they are associated with current symptomatology. This study aimed to: i) investigate whether peripheral complement protein levels are altered at different stages of illness, and ii) identify patterns among complement protein levels that predict clinical symptoms. Complement factors C1q, C3 and C4 were quantified in 183 participants [n = 83 Healthy Controls (HC), n = 10 Ultra-High Risk (UHR) for psychosis, n = 40 First Episode Psychosis (FEP), n = 50 Chronic schizophrenia] using Multiplex ELISA. Permutation-based t-tests were used to assess between-group differences in complement protein levels at each of the three illness stages, relative to age- and gender-matched healthy controls. Canonical correlation analysis was used to identify patterns of complement protein levels that correlated with clinical symptoms. C4 was significantly increased in chronic schizophrenia patients, while C3 and C4 were significantly increased in UHR patients. There were no differences in C1q, C3 and C4 in FEP patients when adjusting for BMI. A molecular pattern of increased C4 and decreased C3 was associated with positive and negative symptom severity in the pooled patient sample. Our findings indicate that peripheral complement concentration is increased across specific stages of psychosis and its imbalance may be associated with symptom severity. Given the small sample size of the UHR group, these findings should be regarded as exploratory, requiring replication.
Collapse
Affiliation(s)
- Liliana Laskaris
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia; Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia; Centre for Neural Engineering, Department of Electrical and Electronic Engineering, University of Melbourne, Carlton South, VIC, Australia
| | - Andrew Zalesky
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia; Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia; Melbourne School of Engineering, The University of Melbourne, Parkville, VIC, Australia
| | - Cynthia Shannon Weickert
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia; Neuroscience Research Australia, Randwick, NSW, Australia; Schizophrenia Research Institute, Randwick, NSW, Australia; School of Psychiatry, University of New South Wales, Sydney, NSW, Australia; Brain & Mind Centre, The University of Sydney, NSW, Australia
| | - Maria A Di Biase
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia; Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia
| | - Gursharan Chana
- Melbourne School of Engineering, The University of Melbourne, Parkville, VIC, Australia; Centre for Neural Engineering, Department of Electrical and Electronic Engineering, University of Melbourne, Carlton South, VIC, Australia
| | - Bernhard T Baune
- Discipline of Psychiatry, The University of Adelaide, SA, Australia
| | - Chad Bousman
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia; Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia; Cooperative Research Centre for Mental Health, Carlton, VIC, Australia; Departments of Medical Genetics, Psychiatry, Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Barnaby Nelson
- Orygen, The National Centre of Excellence in Youth Mental Health, VIC, Australia; Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Patrick McGorry
- Orygen, The National Centre of Excellence in Youth Mental Health, VIC, Australia; Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Ian Everall
- Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia; Cooperative Research Centre for Mental Health, Carlton, VIC, Australia; Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia; Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia; North Western Mental Health, Melbourne Health, Parkville, VIC, Australia; Florey Institute for Neurosciences and Mental Health, Parkville, VIC, Australia; Centre for Neural Engineering, Department of Electrical and Electronic Engineering, University of Melbourne, Carlton South, VIC, Australia; Cooperative Research Centre for Mental Health, Carlton, VIC, Australia
| | - Vanessa Cropley
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia; Department of Psychiatry, The University of Melbourne, Parkville, VIC, Australia; Centre for Mental Health, Faculty of Health, Arts and Design, School of Health Sciences, Swinburne University, VIC, Australia.
| |
Collapse
|
29
|
The Astrocyte-Neuron Interface: An Overview on Molecular and Cellular Dynamics Controlling Formation and Maintenance of the Tripartite Synapse. Methods Mol Biol 2019; 1938:3-18. [PMID: 30617969 DOI: 10.1007/978-1-4939-9068-9_1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Astrocytes are known to provide trophic support to neurons and were originally thought to be passive space-filling cells in the brain. However, recent advances in astrocyte development and functions have highlighted their active roles in controlling brain functions by modulating synaptic transmission. A bidirectional cross talk between astrocytic processes and neuronal synapses define the concept of tripartite synapse. Any change in astrocytic structure/function influences neuronal activity which could lead to neurodevelopmental and neurodegenerative disorders. In this chapter, we briefly overview the methodologies used in deciphering the mechanisms of dynamic interplay between astrocytes and neurons.
Collapse
|
30
|
Benrimoh D, Parr T, Vincent P, Adams RA, Friston K. Active Inference and Auditory Hallucinations. COMPUTATIONAL PSYCHIATRY (CAMBRIDGE, MASS.) 2018; 2:183-204. [PMID: 30627670 PMCID: PMC6317754 DOI: 10.1162/cpsy_a_00022] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 10/03/2018] [Indexed: 01/27/2023]
Abstract
Auditory verbal hallucinations (AVH) are often distressing symptoms of several neuropsychiatric conditions, including schizophrenia. Using a Markov decision process formulation of active inference, we develop a novel model of AVH as false (positive) inference. Active inference treats perception as a process of hypothesis testing, in which sensory data are used to disambiguate between alternative hypotheses about the world. Crucially, this depends upon a delicate balance between prior beliefs about unobserved (hidden) variables and the sensations they cause. A false inference that a voice is present, even in the absence of auditory sensations, suggests that prior beliefs dominate perceptual inference. Here we consider the computational mechanisms that could cause this imbalance in perception. Through simulation, we show that the content of (and confidence in) prior beliefs depends on beliefs about policies (here sequences of listening and talking) and on beliefs about the reliability of sensory data. We demonstrate several ways in which hallucinatory percepts could occur when an agent expects to hear a voice in the presence of imprecise sensory data. This model expresses, in formal terms, alternative computational mechanisms that underwrite AVH and, speculatively, can be mapped onto neurobiological changes associated with schizophrenia. The interaction of action and perception is important in modeling AVH, given that speech is a fundamentally enactive and interactive process-and that hallucinators often actively engage with their voices.
Collapse
Affiliation(s)
| | - Thomas Parr
- Wellcome Trust Centre for Neuroimaging, Institute of Neurology, University College London, London, UK
| | - Peter Vincent
- Wellcome Trust Centre for Neuroimaging, Institute of Neurology, University College London, London, UK
| | - Rick A. Adams
- Division of Psychiatry, University College London, London, UK,Institute of Cognitive Neuroscience, University College London, London, UK
| | - Karl Friston
- Wellcome Trust Centre for Neuroimaging, Institute of Neurology, University College London, London, UK
| |
Collapse
|
31
|
Benrimoh D, Parr T, Vincent P, Adams RA, Friston K. Active Inference and Auditory Hallucinations. COMPUTATIONAL PSYCHIATRY (CAMBRIDGE, MASS.) 2018. [PMID: 30627670 DOI: 10.1162/cpsy˙a˙00022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Auditory verbal hallucinations (AVH) are often distressing symptoms of several neuropsychiatric conditions, including schizophrenia. Using a Markov decision process formulation of active inference, we develop a novel model of AVH as false (positive) inference. Active inference treats perception as a process of hypothesis testing, in which sensory data are used to disambiguate between alternative hypotheses about the world. Crucially, this depends upon a delicate balance between prior beliefs about unobserved (hidden) variables and the sensations they cause. A false inference that a voice is present, even in the absence of auditory sensations, suggests that prior beliefs dominate perceptual inference. Here we consider the computational mechanisms that could cause this imbalance in perception. Through simulation, we show that the content of (and confidence in) prior beliefs depends on beliefs about policies (here sequences of listening and talking) and on beliefs about the reliability of sensory data. We demonstrate several ways in which hallucinatory percepts could occur when an agent expects to hear a voice in the presence of imprecise sensory data. This model expresses, in formal terms, alternative computational mechanisms that underwrite AVH and, speculatively, can be mapped onto neurobiological changes associated with schizophrenia. The interaction of action and perception is important in modeling AVH, given that speech is a fundamentally enactive and interactive process-and that hallucinators often actively engage with their voices.
Collapse
Affiliation(s)
- David Benrimoh
- Wellcome Trust Centre for Neuroimaging, Institute of Neurology, University College London, London, UK
| | - Thomas Parr
- Wellcome Trust Centre for Neuroimaging, Institute of Neurology, University College London, London, UK
| | - Peter Vincent
- Wellcome Trust Centre for Neuroimaging, Institute of Neurology, University College London, London, UK
| | - Rick A Adams
- Division of Psychiatry, University College London, London, UK
| | - Karl Friston
- Wellcome Trust Centre for Neuroimaging, Institute of Neurology, University College London, London, UK
| |
Collapse
|
32
|
Ilgın C, Topuzoğlu A. Extracellular Vesicles in Psychiatry Research in the Context of RDoC Criteria. Psychiatry Investig 2018; 15:1011-1018. [PMID: 30380817 PMCID: PMC6259002 DOI: 10.30773/pi.2018.09.17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/06/2018] [Accepted: 09/17/2018] [Indexed: 12/18/2022] Open
Abstract
The analysis of extracellular vesicles has been accelerated because of the technological advancements in omics methods in recent decades. Extracellular vesicles provide multifaceted information regarding the functional status of the cells. This information would be critical in case of central nervous system cells, which are confined in a relatively sealed biological compartment. This obstacle is more dramatic in psychiatric disorders since their diagnosis primarily depend on the symptoms and signs of the patients. In this paper, we reviewed this rapidly advancing field by discussing definition of extracellular vesicles, their biogenesis and potential use as clinical biomarkers. Then we focused on their potential use in psychiatric disorders in the context of diagnosis and treatment of these disorders. Finally, we tried to combine the RDoC (Research Domain Criteria) with the use of extracellular vesicles in psychiatry research and practice. This review may offer new insights in both basic and translational research focusing on psychiatric disorders.
Collapse
Affiliation(s)
- Can Ilgın
- Department of Public Health, Marmara University School of Medicine, Istanbul, Turkey
| | - Ahmet Topuzoğlu
- Department of Public Health, Marmara University School of Medicine, Istanbul, Turkey
| |
Collapse
|
33
|
Larsen B, Luna B. Adolescence as a neurobiological critical period for the development of higher-order cognition. Neurosci Biobehav Rev 2018; 94:179-195. [PMID: 30201220 PMCID: PMC6526538 DOI: 10.1016/j.neubiorev.2018.09.005] [Citation(s) in RCA: 403] [Impact Index Per Article: 57.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/29/2018] [Accepted: 09/06/2018] [Indexed: 01/08/2023]
Abstract
The transition from adolescence to adulthood is characterized by improvements in higher-order cognitive abilities and corresponding refinements of the structure and function of the brain regions that support them. Whereas the neurobiological mechanisms that govern early development of sensory systems are well-understood, the mechanisms that drive developmental plasticity of association cortices, such as prefrontal cortex (PFC), during adolescence remain to be explained. In this review, we synthesize neurodevelopmental findings at the cellular, circuit, and systems levels in PFC and evaluate them through the lens of established critical period (CP) mechanisms that guide early sensory development. We find remarkable correspondence between these neurodevelopmental processes and the mechanisms driving CP plasticity, supporting the hypothesis that adolescent development is driven by CP mechanisms that guide the rapid development of neurobiology and cognitive ability during adolescence and their subsequent stability in adulthood. Critically, understanding adolescence as a CP not only provides a mechanism for normative adolescent development, it provides a framework for understanding the role of experience and neurobiology in the emergence of psychopathology that occurs during this developmental period.
Collapse
Affiliation(s)
- Bart Larsen
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, 15213, United States; Center for the Neural Basis of Cognition, Pittsburgh, PA, 15213, United States.
| | - Beatriz Luna
- Center for the Neural Basis of Cognition, Pittsburgh, PA, 15213, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, United States
| |
Collapse
|
34
|
Oestreich LKL, Randeniya R, Garrido MI. White matter connectivity reductions in the pre-clinical continuum of psychosis: A connectome study. Hum Brain Mapp 2018; 40:529-537. [PMID: 30251761 DOI: 10.1002/hbm.24392] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 08/20/2018] [Accepted: 08/30/2018] [Indexed: 12/23/2022] Open
Abstract
Widespread white matter connectivity disruptions have commonly been reported in schizophrenia. However, it is questionable whether structural connectivity decline is specifically associated with schizophrenia or whether it extends along a continuum of psychosis into the healthy population. Elucidating brain structure changes associated with psychotic-like experiences in healthy individuals is insofar important as it is a necessary first step towards our understanding of brain pathology preceding florid psychosis. High resolution, multishell diffusion-weighted magnetic resonance images (MRI) were acquired from 89 healthy individuals. Whole-brain white matter fibre tracking was performed to quantify the strength of white matter connections. Network-based statistics were applied to white matter connections in a regression model in order to test for a linear relationship between streamline count and psychotic-like experiences. A significant subnetwork was identified whereby streamline count declined with increasing psychotic-like experiences. This network of structural connectivity reductions affected all cortical lobes, subcortical structures and the cerebellum and spanned along prominent association and commissural white matter pathways. A widespread network of linearly declining connectivity strength with increasing number of psychotic-like experiences was identified in healthy individuals. This finding is in line with white matter connectivity reductions reported from early to chronic stages of schizophrenia and might therefore aid the development of tools to identify individuals at risk of transitioning to psychosis.
Collapse
Affiliation(s)
- Lena K L Oestreich
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, Australia.,Queensland Brain Institute, The University of Queensland, Brisbane, Australia.,Centre for Advanced Imaging, The University of Queensland, Brisbane, Australia
| | - Roshini Randeniya
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia.,Australian Centre of Excellence for Integrative Brain Function, The University of Queensland, Brisbane, Australia
| | - Marta I Garrido
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia.,Centre for Advanced Imaging, The University of Queensland, Brisbane, Australia.,Australian Centre of Excellence for Integrative Brain Function, The University of Queensland, Brisbane, Australia.,School of Mathematics and Physics, The University of Queensland, Brisbane, Australia
| |
Collapse
|
35
|
Schoonover KE, Queern SL, Lapi SE, Roberts RC. Impaired copper transport in schizophrenia results in a copper-deficient brain state: A new side to the dysbindin story. World J Biol Psychiatry 2018; 21:13-28. [PMID: 30230404 PMCID: PMC6424639 DOI: 10.1080/15622975.2018.1523562] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Objectives: Several schizophrenia brain regions exhibit decreased dysbindin. Dysbindin modulates copper transport crucial for myelination, monoamine metabolism and cellular homeostasis. Schizophrenia patients (SZP) exhibit increased plasma copper, while copper-decreasing agents produce schizophrenia-like behavioural and pathological abnormalities. Therefore, we sought to determine dysbindin and copper transporter protein expression and copper content in SZP.Methods: We studied the copper-rich substantia nigra (SN) using Western blot and inductively-coupled plasma mass spectrometry. We characterised specific protein domains of copper transporters ATP7A, CTR1, ATP7B and dysbindin isoforms 1 A and 1B/C in SZP (n = 15) and matched controls (n = 11), and SN copper content in SZP (n = 14) and matched controls (n = 11). As a preliminary investigation, we compared medicated (ON; n = 11) versus unmedicated SZP (OFF; n = 4).Results: SZP exhibited increased C terminus, but not N terminus, ATP7A. SZP expressed less transmembrane CTR1 and dysbindin 1B/C than controls. ON exhibited increased C terminus ATP7A protein versus controls. OFF exhibited less N terminus ATP7A protein than controls and ON, suggesting medication-induced rescue of the ATP7A N terminus. SZP exhibited less SN copper content than controls.Conclusions: These results provide the first evidence of disrupted copper transport in schizophrenia SN that appears to result in a copper-deficient state. Furthermore, copper homeostasis may be modulated by specific dysbindin isoforms and antipsychotic treatment.
Collapse
Affiliation(s)
- Kirsten E. Schoonover
- Department of Psychology and Behavioral Neuroscience, University of Alabama at Birmingham
| | - Stacy L. Queern
- Department of Radiology, University of Alabama at Birmingham,Department of Chemistry, Washington University in St. Louis
| | - Suzanne E. Lapi
- Department of Radiology, University of Alabama at Birmingham,Department of Chemistry, Washington University in St. Louis
| | - Rosalinda C. Roberts
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham
| |
Collapse
|
36
|
Mohammadi A, Rashidi E, Amooeian VG. Brain, blood, cerebrospinal fluid, and serum biomarkers in schizophrenia. Psychiatry Res 2018; 265:25-38. [PMID: 29680514 DOI: 10.1016/j.psychres.2018.04.036] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/20/2018] [Accepted: 04/11/2018] [Indexed: 12/29/2022]
Abstract
Over the last decade, finding a reliable biomarker for the early detection of schizophrenia (Scz) has been a topic of interest. The main goal of the current review is to provide a comprehensive view of the brain, blood, cerebrospinal fluid (CSF), and serum biomarkers of Scz disease. Imaging studies have demonstrated that the volumes of the corpus callosum, thalamus, hippocampal formation, subiculum, parahippocampal gyrus, superior temporal gyrus, prefrontal and orbitofrontal cortices, and amygdala-hippocampal complex were reduced in patients diagnosed with Scz. It has been revealed that the levels of interleukin 1β (IL-1β), IL-6, IL-8, and TNF-α were increased in patients with Scz. Decreased mRNA levels of brain-derived neurotrophic factor (BDNF), tropomyosin receptor kinase B (TrkB), neurotrophin-3 (NT-3), nerve growth factor (NGF), and vascular endothelial growth factor (VEGF) genes have also been reported in Scz patients. Genes with known strong relationships with this disease include BDNF, catechol-O-methyltransferase (COMT), regulator of G-protein signaling 4 (RGS4), dystrobrevin-binding protein 1 (DTNBP1), neuregulin 1 (NRG1), Reelin (RELN), Selenium-binding protein 1 (SELENBP1), glutamic acid decarboxylase 67 (GAD 67), and disrupted in schizophrenia 1 (DISC1). The levels of dopamine, tyrosine hydroxylase (TH), serotonin or 5-hydroxytryptamine (5-HT) receptor 1A and B (5-HTR1A and 5-HTR1B), and 5-HT1B were significantly increased in Scz patients, while the levels of gamma-aminobutyric acid (GABA), 5-HT transporter (5-HTT), and 5-HT receptor 2A (5-HTR2A) were decreased. The increased levels of SELENBP1 and Glycogen synthase kinase 3 subunit α (GSK3α) genes in contrast with reduced levels of B-cell translocation gene 1 (BTG1), human leukocyte antigen DRB1 (HLA-DRB1), heterogeneous nuclear ribonucleoprotein A3 (HNRPA3), and serine/arginine-rich splicing factor 1 (SFRS1) genes have also been reported. This review covers various dysregulation of neurotransmitters and also highlights the strengths and weaknesses of studies attempting to identify candidate biomarkers.
Collapse
Affiliation(s)
- Alireza Mohammadi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Ehsan Rashidi
- Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Ghasem Amooeian
- Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
37
|
Mahabir S, Chatterjee D, Misquitta K, Chatterjee D, Gerlai R. Lasting changes induced by mild alcohol exposure during embryonic development in BDNF, NCAM and synaptophysin-positive neurons quantified in adult zebrafish. Eur J Neurosci 2018; 47:1457-1473. [PMID: 29846983 DOI: 10.1111/ejn.13975] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 05/15/2018] [Accepted: 05/18/2018] [Indexed: 01/17/2023]
Abstract
Fetal alcohol spectrum disorder is one of the leading causes of mental health issues worldwide. Analysis of zebrafish exposed to alcohol during embryonic development confirmed that even low concentrations of alcohol for a short period of time may have lasting behavioral consequences at the adult or old age. The mechanism of this alteration has not been studied. Here, we immersed zebrafish embryos into 1% alcohol solution (vol/vol%) at 24 hr post-fertilization (hpf) for 2 hr and analyzed potential changes using immunohistochemistry. We measured the number of BDNF (brain-derived neurotrophic factor) and NCAM (neuronal cell adhesion molecule)-positive neurons and the intensity of synaptophysin staining in eight brain regions: lateral zone of the dorsal telencephalic area, medial zone of the dorsal telencephalic area, dorsal nucleus of the ventral telencephalic area, ventral nucleus of the ventral telencephalic area, parvocellular preoptic nucleus, ventral habenular nucleus, corpus cerebella and inferior reticular formation. We found embryonic alcohol exposure to significantly reduce the number of BDNF- and NCAM-positive cells in all brain areas studied as compared to control. We also found alcohol to significantly reduce the intensity of synaptophysin staining in all brain areas except the cerebellum and preoptic area. These neuroanatomical changes correlated with previously demonstrated reduction of social behavior in embryonic alcohol-exposed zebrafish, raising the possibility of a causal link. Given the evolutionary conservation across fish and mammals, we emphasize the implication of our current study for human health: even small amount of alcohol consumption may be unsafe during pregnancy.
Collapse
Affiliation(s)
- Samantha Mahabir
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Dipashree Chatterjee
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Keith Misquitta
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Diptendu Chatterjee
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Robert Gerlai
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada.,Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| |
Collapse
|
38
|
Concurrence of form and function in developing networks and its role in synaptic pruning. Nat Commun 2018; 9:2236. [PMID: 29884799 PMCID: PMC5993834 DOI: 10.1038/s41467-018-04537-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/03/2018] [Indexed: 02/07/2023] Open
Abstract
A fundamental question in neuroscience is how structure and function of neural systems are related. We study this interplay by combining a familiar auto-associative neural network with an evolving mechanism for the birth and death of synapses. A feedback loop then arises leading to two qualitatively different types of behaviour. In one, the network structure becomes heterogeneous and dissasortative, and the system displays good memory performance; furthermore, the structure is optimised for the particular memory patterns stored during the process. In the other, the structure remains homogeneous and incapable of pattern retrieval. These findings provide an inspiring picture of brain structure and dynamics that is compatible with experimental results on early brain development, and may help to explain synaptic pruning. Other evolving networks—such as those of protein interactions—might share the basic ingredients for this feedback loop and other questions, and indeed many of their structural features are as predicted by our model. How structure and function coevolve in developing brains is little understood. Here, the authors study a coupled model of network development and memory, and find that due to the feedback networks with some initial memory capacity evolve into heterogeneous structures with high memory performance.
Collapse
|
39
|
Wojtalik JA, Eack SM, Smith MJ, Keshavan MS. Using Cognitive Neuroscience to Improve Mental Health Treatment: A Comprehensive Review. JOURNAL OF THE SOCIETY FOR SOCIAL WORK AND RESEARCH 2018; 9:223-260. [PMID: 30505392 PMCID: PMC6258037 DOI: 10.1086/697566] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Mental health interventions do not yet offer complete, client-defined functional recovery, and novel directions in treatment research are needed to improve the efficacy of available interventions. One promising direction is the integration of social work and cognitive neuroscience methods, which provides new opportunities for clinical intervention research that will guide development of more effective mental health treatments that holistically attend to the biological, social, and environmental contributors to disability and recovery. This article reviews emerging trends in cognitive neuroscience and provides examples of how these advances can be used by social workers and allied professions to improve mental health treatment. We discuss neuroplasticity, which is the dynamic and malleable nature of the brain. We also review the use of risk and resiliency biomarkers and novel treatment targets based on neuroimaging findings to prevent disability, personalize treatment, and make interventions more targeted and effective. The potential of treatment research to contribute to neuroscience discoveries regarding brain change is considered from the experimental-medicine approach adopted by the National Institute of Mental Health. Finally, we provide resources and recommendations to facilitate the integration of cognitive neuroscience into mental health research in social work.
Collapse
Affiliation(s)
- Jessica A Wojtalik
- Doctoral candidate at the University of Pittsburgh School of Social Work
| | - Shaun M Eack
- Professor at the University of Pittsburgh School of Social Work and Department of Psychiatry
| | - Matthew J Smith
- Associate professor at the University of Michigan School of Social Work
| | | |
Collapse
|
40
|
Garrett L, Ung M, Heermann T, Niedermeier KM, Hölter S. Analysis of Neuropsychiatric Disease‐Related Functional Neuroanatomical Markers in Mice. ACTA ACUST UNITED AC 2018; 8:79-128. [DOI: 10.1002/cpmo.37] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Lillian Garrett
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health Neuherberg Germany
- German Mouse Clinic, Helmholtz Zentrum München; German Research Centre for Environmental Health Neuherberg Germany
| | - Marie‐Claire Ung
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health Neuherberg Germany
| | - Tamara Heermann
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health Neuherberg Germany
| | - Kristina M. Niedermeier
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health Neuherberg Germany
| | - Sabine Hölter
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health Neuherberg Germany
- German Mouse Clinic, Helmholtz Zentrum München; German Research Centre for Environmental Health Neuherberg Germany
| |
Collapse
|
41
|
Dong H, Zhu M, Meng L, Ding Y, Yang D, Zhang S, Qiang W, Fisher DW, Xu EY. Pumilio2 regulates synaptic plasticity via translational repression of synaptic receptors in mice. Oncotarget 2018; 9:32134-32148. [PMID: 30181804 PMCID: PMC6114944 DOI: 10.18632/oncotarget.24345] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 01/13/2018] [Indexed: 11/25/2022] Open
Abstract
PUMILIO 2 (PUM2) is a member of Pumilio and FBF (PUF) family, an RNA binding protein family with phylogenetically conserved roles in germ cell development. The Drosophila Pumilio homolog is also required for dendrite morphogenesis and synaptic function via translational control of synaptic proteins, such as glutamate receptors, and recent mammalian studies demonstrated a similar role in neuronal culture with associated motor and memory abnormalities in vivo. Importantly, transgenic mice with PUM2 knockout show prominent epileptiform activity, and patients with intractable temporal lobe epilepsy and mice with pilocarpine-induced seizures have decreased neuronal PUM2, possibly leading to further seizure susceptibility. However, how PUM2 influences synaptic function in vivo and, subsequently, seizures is not known. We found that PUM2 is highly expressed in the brain, especially in the temporal lobe, and knockout of Pum2 (Pum2-/- ) resulted in significantly increased pyramidal cell dendrite spine and synapse density. In addition, multiple proteins associated with excitatory synaptic function, including glutamate receptor 2 (GLUR2), are up-regulated in Pum2-/- mice. The expression of GLUR2 protein but not mRNA is increased in the Pum2-/- mutant hippocampus, Glur2 transcripts are increased in mutant polysome fractions, and overexpression of PUM2 led to repression of reporter expression containing the 3'Untranslated Region (3'UTR) of Glur2, suggesting translation of GLUR2 was increased in the absence of Pum2. Overall, these studies provide a molecular mechanism for the increased temporal lobe excitability observed with PUM2 loss and suggest PUM2 might contribute to intractable temporal lobe epilepsy.
Collapse
Affiliation(s)
- Hongxin Dong
- Departments of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Mengyi Zhu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Liping Meng
- Departments of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yan Ding
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Ding Yang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Shanshan Zhang
- Departments of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Wenan Qiang
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Daniel W Fisher
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Eugene Yujun Xu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, P. R. China
| |
Collapse
|
42
|
Prytkova I, Brennand KJ. Prospects for Modeling Abnormal Neuronal Function in Schizophrenia Using Human Induced Pluripotent Stem Cells. Front Cell Neurosci 2017; 11:360. [PMID: 29217999 PMCID: PMC5703699 DOI: 10.3389/fncel.2017.00360] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 11/03/2017] [Indexed: 01/21/2023] Open
Abstract
Excitatory dopaminergic neurons, inhibitory GABAergic neurons, microglia, and oligodendrocytes have all been implicated in schizophrenia (SZ) network pathology. Still, SZ has been a difficult disorder to study, not only because of the limitations of animal models in capturing the complexity of the human mind, but also because it is greatly polygenic, with high rates of variability across the population. The advent of patient-derived pluripotent stem cells and induced neural and glial cultures has brought hope for modeling the molecular dysfunction underlying SZ pathology in a patient-specific manner. Here I review the successes of the patient-specific induced cultures in generating different cell types for the study of SZ, with special emphasis on the utility of co-culture techniques, both two- and three-dimensional, for modeling network dysfunction in disease.
Collapse
Affiliation(s)
- Iya Prytkova
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, line>New York, NY, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kristen J Brennand
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, line>New York, NY, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
43
|
Ivakhnitskaia E, Lin RW, Hamada K, Chang C. Timing of neuronal plasticity in development and aging. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2017; 7. [PMID: 29139210 DOI: 10.1002/wdev.305] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 08/21/2017] [Accepted: 09/11/2017] [Indexed: 01/21/2023]
Abstract
Molecular oscillators are well known for their roles in temporal control of some biological processes like cell proliferation, but molecular mechanisms that provide temporal control of differentiation and postdifferentiation events in cells are less understood. In the nervous system, establishment of neuronal connectivity during development and decline in neuronal plasticity during aging are regulated with temporal precision, but the timing mechanisms are largely unknown. Caenorhabditis elegans has been a preferred model for aging research and recently emerges as a new model for the study of developmental and postdevelopmental plasticity in neurons. In this review we discuss the emerging mechanisms in timing of developmental lineage progression, axon growth and pathfinding, synapse formation, and reorganization, and neuronal plasticity in development and aging. We also provide a current view on the conserved core axon regeneration molecules with the intention to point out potential regulatory points of temporal controls. We highlight recent progress in understanding timing mechanisms that regulate decline in regenerative capacity, including progressive changes of intrinsic timers and co-opting the aging pathway molecules. WIREs Dev Biol 2018, 7:e305. doi: 10.1002/wdev.305 This article is categorized under: Invertebrate Organogenesis > Worms Establishment of Spatial and Temporal Patterns > Regulation of Size, Proportion, and Timing Nervous System Development > Worms Gene Expression and Transcriptional Hierarchies > Regulatory RNA.
Collapse
Affiliation(s)
- Evguenia Ivakhnitskaia
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA.,Medical Scientist Training Program, University of Illinois at Chicago, Chicago, IL, USA.,Graduate Program in Neuroscience, University of Illinois at Chicago, Chicago, IL, USA
| | - Ryan Weihsiang Lin
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Kana Hamada
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Chieh Chang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA.,Graduate Program in Neuroscience, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
44
|
Virdee JK, Saro G, Fouillet A, Findlay J, Ferreira F, Eversden S, O'Neill MJ, Wolak J, Ursu D. A high-throughput model for investigating neuronal function and synaptic transmission in cultured neuronal networks. Sci Rep 2017; 7:14498. [PMID: 29101377 PMCID: PMC5670206 DOI: 10.1038/s41598-017-15171-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 10/23/2017] [Indexed: 12/30/2022] Open
Abstract
Loss of synapses or alteration of synaptic activity is associated with cognitive impairment observed in a number of psychiatric and neurological disorders, such as schizophrenia and Alzheimer’s disease. Therefore successful development of in vitro methods that can investigate synaptic function in a high-throughput format could be highly impactful for neuroscience drug discovery. We present here the development, characterisation and validation of a novel high-throughput in vitro model for assessing neuronal function and synaptic transmission in primary rodent neurons. The novelty of our approach resides in the combination of the electrical field stimulation (EFS) with data acquisition in spatially separated areas of an interconnected neuronal network. We integrated our methodology with state of the art drug discovery instrumentation (FLIPR Tetra) and used selective tool compounds to perform a systematic pharmacological validation of the model. We investigated pharmacological modulators targeting pre- and post-synaptic receptors (AMPA, NMDA, GABA-A, mGluR2/3 receptors and Nav, Cav voltage-gated ion channels) and demonstrated the ability of our model to discriminate and measure synaptic transmission in cultured neuronal networks. Application of the model described here as an unbiased phenotypic screening approach will help with our long term goals of discovering novel therapeutic strategies for treating neurological disorders.
Collapse
Affiliation(s)
- Jasmeet K Virdee
- Eli Lilly and Company, Lilly Research Centre, Windlesham, GU20 6PH, UK
| | - Gabriella Saro
- Eli Lilly and Company, Lilly Research Centre, Windlesham, GU20 6PH, UK
| | - Antoine Fouillet
- Eli Lilly and Company, Lilly Research Centre, Windlesham, GU20 6PH, UK
| | - Jeremy Findlay
- Eli Lilly and Company, Lilly Research Centre, Windlesham, GU20 6PH, UK
| | - Filipa Ferreira
- Eli Lilly and Company, Lilly Research Centre, Windlesham, GU20 6PH, UK
| | - Sarah Eversden
- Eli Lilly and Company, Lilly Research Centre, Windlesham, GU20 6PH, UK
| | - Michael J O'Neill
- Eli Lilly and Company, Lilly Research Centre, Windlesham, GU20 6PH, UK
| | - Joanna Wolak
- Eli Lilly and Company, Lilly Research Centre, Windlesham, GU20 6PH, UK
| | - Daniel Ursu
- Eli Lilly and Company, Lilly Research Centre, Windlesham, GU20 6PH, UK.
| |
Collapse
|
45
|
Wojtalik JA, Smith MJ, Keshavan MS, Eack SM. A Systematic and Meta-analytic Review of Neural Correlates of Functional Outcome in Schizophrenia. Schizophr Bull 2017; 43:1329-1347. [PMID: 28204755 PMCID: PMC5737663 DOI: 10.1093/schbul/sbx008] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Individuals with schizophrenia are burdened with impairments in functional outcome, despite existing interventions. The lack of understanding of the neurobiological correlates supporting adaptive function in the disorder is a significant barrier to developing more effective treatments. This research conducted a systematic and meta-analytic review of all peer-reviewed studies examining brain-functional outcome relationships in schizophrenia. A total of 53 (37 structural and 16 functional) brain imaging studies examining the neural correlates of functional outcome across 1631 individuals with schizophrenia were identified from literature searches in relevant databases occurring between January, 1968 and December, 2016. Study characteristics and results representing brain-functional outcome relationships were systematically extracted, reviewed, and meta-analyzed. Results indicated that better functional outcome was associated with greater fronto-limbic and whole brain volumes, smaller ventricles, and greater activation, especially during social cognitive processing. Thematic observations revealed that the dorsolateral prefrontal cortex, anterior cingulate, posterior cingulate, parahippocampal gyrus, superior temporal sulcus, and cerebellum may have role in functioning. The neural basis of functional outcome and disability is infrequently studied in schizophrenia. While existing evidence is limited and heterogeneous, these findings suggest that the structural and functional integrity of fronto-limbic brain regions is consistently related to functional outcome in individuals with schizophrenia. Further research is needed to understand the mechanisms and directionality of these relationships, and the potential for identifying neural targets to support functional improvement.
Collapse
Affiliation(s)
- Jessica A Wojtalik
- School of Social Work, University of Pittsburgh, Pittsburgh, PA,To whom correspondence should be addressed; School of Social Work, University of Pittsburgh, 2117 Cathedral of Learning, Pittsburgh, PA 15260, US; tel: 412-624-6304, e-mail:
| | - Matthew J Smith
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | - Shaun M Eack
- School of Social Work, University of Pittsburgh, Pittsburgh, PA,Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
46
|
Kelly JR, Minuto C, Cryan JF, Clarke G, Dinan TG. Cross Talk: The Microbiota and Neurodevelopmental Disorders. Front Neurosci 2017; 11:490. [PMID: 28966571 PMCID: PMC5605633 DOI: 10.3389/fnins.2017.00490] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 08/17/2017] [Indexed: 12/11/2022] Open
Abstract
Humans evolved within a microbial ecosystem resulting in an interlinked physiology. The gut microbiota can signal to the brain via the immune system, the vagus nerve or other host-microbe interactions facilitated by gut hormones, regulation of tryptophan metabolism and microbial metabolites such as short chain fatty acids (SCFA), to influence brain development, function and behavior. Emerging evidence suggests that the gut microbiota may play a role in shaping cognitive networks encompassing emotional and social domains in neurodevelopmental disorders. Drawing upon pre-clinical and clinical evidence, we review the potential role of the gut microbiota in the origins and development of social and emotional domains related to Autism spectrum disorders (ASD) and schizophrenia. Small preliminary clinical studies have demonstrated gut microbiota alterations in both ASD and schizophrenia compared to healthy controls. However, we await the further development of mechanistic insights, together with large scale longitudinal clinical trials, that encompass a systems level dimensional approach, to investigate whether promising pre-clinical and initial clinical findings lead to clinical relevance.
Collapse
Affiliation(s)
- John R Kelly
- Department of Psychiatry and Neurobehavioural Science, University College CorkCork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| | - Chiara Minuto
- Department of Psychiatry and Neurobehavioural Science, University College CorkCork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| | - John F Cryan
- APC Microbiome Institute, University College CorkCork, Ireland.,Department of Anatomy and Neuroscience, University College CorkCork, Ireland
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioural Science, University College CorkCork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| | - Timothy G Dinan
- Department of Psychiatry and Neurobehavioural Science, University College CorkCork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| |
Collapse
|
47
|
Regulatory Mechanisms of Metamorphic Neuronal Remodeling Revealed Through a Genome-Wide Modifier Screen in Drosophila melanogaster. Genetics 2017; 206:1429-1443. [PMID: 28476867 PMCID: PMC5500141 DOI: 10.1534/genetics.117.200378] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 04/28/2017] [Indexed: 02/01/2023] Open
Abstract
During development, neuronal remodeling shapes neuronal connections to establish fully mature and functional nervous systems. Our previous studies have shown that the RNA-binding factor alan shepard (shep) is an important regulator of neuronal remodeling during metamorphosis in Drosophila melanogaster, and loss of shep leads to smaller soma size and fewer neurites in a stage-dependent manner. To shed light on the mechanisms by which shep regulates neuronal remodeling, we conducted a genetic modifier screen for suppressors of shep-dependent wing expansion defects and cellular morphological defects in a set of peptidergic neurons, the bursicon neurons, that promote posteclosion wing expansion. Out of 702 screened deficiencies that covered 86% of euchromatic genes, we isolated 24 deficiencies as candidate suppressors, and 12 of them at least partially suppressed morphological defects in shep mutant bursicon neurons. With RNA interference and mutant alleles of individual genes, we identified Daughters against dpp (Dad) and Olig family (Oli) as shep suppressor genes, and both of them restored the adult cellular morphology of shep-depleted bursicon neurons. Dad encodes an inhibitory Smad protein that inhibits bone morphogenetic protein (BMP) signaling, raising the possibility that shep interacted with BMP signaling through antagonism of Dad. By manipulating expression of the BMP receptor tkv, we found that activated BMP signaling was sufficient to rescue loss-of-shep phenotypes. These findings reveal mechanisms of shep regulation during neuronal development, and they highlight a novel genetic shep interaction with the BMP signaling pathway that controls morphogenesis in mature, terminally differentiated neurons during metamorphosis.
Collapse
|
48
|
Romme IAC, de Reus MA, Ophoff RA, Kahn RS, van den Heuvel MP. Connectome Disconnectivity and Cortical Gene Expression in Patients With Schizophrenia. Biol Psychiatry 2017; 81:495-502. [PMID: 27720199 DOI: 10.1016/j.biopsych.2016.07.012] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 07/16/2016] [Accepted: 07/18/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Genome-wide association studies have identified several common risk loci for schizophrenia (SCZ). In parallel, neuroimaging studies have shown consistent findings of widespread white matter disconnectivity in patients with SCZ. METHODS We examined the role of genes in brain connectivity in patients with SCZ by combining transcriptional profiles of 43 SCZ risk genes identified by the recent genome-wide association study of the Schizophrenia Working Group of the Psychiatric Genomics Consortium with data on macroscale connectivity reductions in patients with SCZ. Expression profiles of 43 Psychiatric Genomics Consortium SCZ risk genes were extracted from the Allen Human Brain Atlas, and their average profile across the cortex was correlated to the pattern of cortical disconnectivity as derived from diffusion-weighted magnetic resonance imaging data of patients with SCZ (n = 48) and matched healthy controls (n = 43). RESULTS The expression profile of SCZ risk genes across cortical regions was significantly correlated with the regional macroscale disconnectivity (r = .588; p = .017). In addition, effects were found to be potentially specific to SCZ, with transcriptional profiles not related to cortical disconnectivity in patients with bipolar I disorder (diffusion-weighted magnetic resonance imaging data; 216 patients, 144 controls). Further examination of correlations across all 20,737 genes present in the Allen Human Brain Atlas showed the set of top 100 strongest correlating genes to display significant enrichment for the disorder, potentially identifying new genes involved in the pathophysiology of SCZ. CONCLUSIONS Our results suggest that under disease conditions, cortical areas with pronounced expression of risk genes implicated in SCZ form central areas for white matter disconnectivity.
Collapse
Affiliation(s)
- Ingrid A C Romme
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marcel A de Reus
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roel A Ophoff
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands; Center for Neurobehavioral Genetics and Department of Human Genetics , University of California Los Angeles, Los Angeles, California
| | - René S Kahn
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Martijn P van den Heuvel
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
49
|
Sebel LE, Graves SM, Chan CS, Surmeier DJ. Haloperidol Selectively Remodels Striatal Indirect Pathway Circuits. Neuropsychopharmacology 2017; 42:963-973. [PMID: 27577602 PMCID: PMC5312058 DOI: 10.1038/npp.2016.173] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 08/08/2016] [Accepted: 08/10/2016] [Indexed: 01/20/2023]
Abstract
Typical antipsychotic drugs are widely thought to alleviate the positive symptoms of schizophrenia by antagonizing dopamine D2 receptors expressed by striatal spiny projection neurons (SPNs). What is less clear is why antipsychotics have a therapeutic latency of weeks. Using a combination of physiological and anatomical approaches in ex vivo brain slices from transgenic mice, it was found that 2 weeks of haloperidol treatment induced both intrinsic and synaptic adaptations specifically within indirect pathway SPNs (iSPNs). Perphenazine treatment had similar effects. Some of these adaptations were homeostatic, including a drop in intrinsic excitability and pruning of excitatory corticostriatal glutamatergic synapses. However, haloperidol treatment also led to strengthening of a subset of excitatory corticostriatal synapses. This slow remodeling of corticostriatal iSPN circuitry is likely to play a role in mediating the delayed therapeutic action of neuroleptics.
Collapse
Affiliation(s)
- Luke E Sebel
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Steven M Graves
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - C Savio Chan
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - D James Surmeier
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA,Department of Physiology, Feinberg School of Medicine, Northwestern University, 303 E Superior, Chicago, IL 60611, USA, Tel: +1 312 503 4904, Fax: +1 312 503 5101, E-mail:
| |
Collapse
|
50
|
da Silveira VT, Medeiros DDC, Ropke J, Guidine PA, Rezende GH, Moraes MFD, Mendes EMAM, Macedo D, Moreira FA, de Oliveira ACP. Effects of early or late prenatal immune activation in mice on behavioral and neuroanatomical abnormalities relevant to schizophrenia in the adulthood. Int J Dev Neurosci 2017; 58:1-8. [PMID: 28122258 DOI: 10.1016/j.ijdevneu.2017.01.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/19/2017] [Accepted: 01/19/2017] [Indexed: 12/20/2022] Open
Abstract
Maternal immune activation (MIA) during pregnancy in rodents increases the risk of the offspring to develop schizophrenia-related behaviors, suggesting a relationship between the immune system and the brain development. Here we tested the hypothesis that MIA induced by the viral mimetic polyinosinic-polycytidylic acid (poly I:C) in early or late gestation of mice leads to behavioral and neuroanatomical disorders in the adulthood. On gestational days (GDs) 9 or 17 pregnant dams were treated with poly I:C or saline via intravenous route and the offspring behaviors were measured during adulthood. Considering the progressive structural neuroanatomical alterations in the brain of individuals with schizophrenia, we used magnetic resonance imaging (MRI) to perform brain morphometric analysis of the offspring aged one year. MIA on GD9 or GD17 led to increased basal locomotor activity, enhanced motor responses to ketamine, a psychotomimetic drug, and reduced time spent in the center of the arena, suggesting an increased anxiety-like behavior. In addition, MIA on GD17 reduced glucose preference in the offspring. None of the treatments altered the relative volume of the lateral ventricles. However, a decrease in brain volume, especially for posterior structures, was observed for one-year-old animals treated with poly I:C compared with control groups. Thus, activation of the maternal immune system at different GDs lead to neuroanatomical and behavioral alterations possibly related to the positive and negative symptoms of schizophrenia. These results provide insights on neuroimmunonological and neurodevelopmental aspects of certain psychopathologies, such as schizophrenia.
Collapse
Affiliation(s)
- Vivian T da Silveira
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| | - Daniel de Castro Medeiros
- Center for Technology and Research in Magneto-Resonance (CTPMAG), Graduate Program in Electrical Engineering - Universidade Federal de Minas Gerais, Brazil
| | - Jivago Ropke
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| | - Patricia A Guidine
- Center for Technology and Research in Magneto-Resonance (CTPMAG), Graduate Program in Electrical Engineering - Universidade Federal de Minas Gerais, Brazil
| | - Gustavo H Rezende
- Center for Technology and Research in Magneto-Resonance (CTPMAG), Graduate Program in Electrical Engineering - Universidade Federal de Minas Gerais, Brazil
| | - Marcio Flavio D Moraes
- Center for Technology and Research in Magneto-Resonance (CTPMAG), Graduate Program in Electrical Engineering - Universidade Federal de Minas Gerais, Brazil
| | - Eduardo Mazoni A M Mendes
- Center for Technology and Research in Magneto-Resonance (CTPMAG), Graduate Program in Electrical Engineering - Universidade Federal de Minas Gerais, Brazil
| | - Danielle Macedo
- Drug Research and Development Center, Department of Physiology and Pharmacology, Medical School, Universidade Federal do Ceará, Brazil
| | - Fabricio A Moreira
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| | | |
Collapse
|