1
|
Onwordi EC, Whitehurst T, Shatalina E, Mansur A, Arumuham A, Osugo M, Marques TR, Jauhar S, Gupta S, Mehrotra R, Rabiner EA, Gunn RN, Natesan S, Howes OD. Synaptic Terminal Density Early in the Course of Schizophrenia: An In Vivo UCB-J Positron Emission Tomographic Imaging Study of SV2A. Biol Psychiatry 2024; 95:639-646. [PMID: 37330164 PMCID: PMC10923626 DOI: 10.1016/j.biopsych.2023.05.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND The synaptic hypothesis is an influential theory of the pathoetiology of schizophrenia (SCZ), which is supported by the finding that there is lower uptake of the synaptic terminal density marker [11C]UCB-J in patients with chronic SCZ than in control participants. However, it is unclear whether these differences are present early in the illness. To address this, we investigated [11C]UCB-J volume of distribution (VT) in antipsychotic-naïve/free patients with SCZ who were recruited from first-episode services compared with healthy volunteers. METHODS Forty-two volunteers (SCZ n = 21, healthy volunteers n = 21) underwent [11C]UCB-J positron emission tomography to index [11C]UCB-J VT and distribution volume ratio in the anterior cingulate, frontal, and dorsolateral prefrontal cortices; the temporal, parietal and occipital lobes; and the hippocampus, thalamus, and amygdala. Symptom severity was assessed in the SCZ group using the Positive and Negative Syndrome Scale. RESULTS We found no significant effects of group on [11C]UCB-J VT or distribution volume ratio in most regions of interest (effect sizes from d = 0.0-0.7, p > .05), with two exceptions: we found lower distribution volume ratio in the temporal lobe (d = 0.7, uncorrected p < .05) and lower VT/fp in the anterior cingulate cortex in patients (d = 0.7, uncorrected p < .05). The Positive and Negative Syndrome Scale total score was negatively associated with [11C]UCB-J VT in the hippocampus in the SCZ group (r = -0.48, p = .03). CONCLUSIONS These findings indicate that large differences in synaptic terminal density are not present early in SCZ, although there may be more subtle effects. When taken together with previous evidence of lower [11C]UCB-J VT in patients with chronic illness, this may indicate synaptic density changes during the course of SCZ.
Collapse
Affiliation(s)
- Ellis Chika Onwordi
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, United Kingdom; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom; Centre for Psychiatry and Mental Health, Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom.
| | - Thomas Whitehurst
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, United Kingdom; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Ekaterina Shatalina
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, United Kingdom; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Ayla Mansur
- Department of Brain Sciences, Imperial College London, The Commonwealth Building, Hammersmith Hospital, London, United Kingdom; Invicro, Burlington Danes Building, London, United Kingdom
| | - Atheeshaan Arumuham
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, United Kingdom; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Martin Osugo
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, United Kingdom; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Tiago Reis Marques
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, United Kingdom; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Sameer Jauhar
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Susham Gupta
- Early Detection and Early Intervention, East London National Health Service Foundation Trust, London, United Kingdom
| | - Ravi Mehrotra
- Early Intervention in Psychosis Team, West Middlesex University Hospital, West London National Health Service Trust, Isleworth, London, United Kingdom
| | - Eugenii A Rabiner
- Invicro, Burlington Danes Building, London, United Kingdom; Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Roger N Gunn
- Department of Brain Sciences, Imperial College London, The Commonwealth Building, Hammersmith Hospital, London, United Kingdom; Invicro, Burlington Danes Building, London, United Kingdom
| | - Sridhar Natesan
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, United Kingdom; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Oliver D Howes
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, United Kingdom; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom.
| |
Collapse
|
2
|
Mahoney HL, Bloom CA, Justin HS, Capraro BM, Morris C, Gonzalez D, Sandefur E, Faulkner J, Reiss S, Valladares A, Ocampo A, Carter B, Lussier AL, Dinh LP, Weeber E, Gamsby J, Gulick D. DISC1 and reelin interact to alter cognition, inhibition, and neurogenesis in a novel mouse model of schizophrenia. Front Cell Neurosci 2024; 17:1321632. [PMID: 38283751 PMCID: PMC10813205 DOI: 10.3389/fncel.2023.1321632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 12/11/2023] [Indexed: 01/30/2024] Open
Abstract
The etiology of schizophrenia (SCZ) is multifactorial, and depending on a host of genetic and environmental factors. Two putative SCZ susceptibility genes, Disrupted-in-Schizophrenia-1 (DISC1) and reelin (RELN), interact at a molecular level, suggesting that combined disruption of both may lead to an intensified SCZ phenotype. To examine this gene-gene interaction, we produced a double mutant mouse line. Mice with heterozygous RELN haploinsufficiency were crossed with mice expressing dominant-negative c-terminal truncated human DISC1 to produce offspring with both mutations (HRM/DISC1 mice). We used an array of behavioral tests to generate a behavioral phenotype for these mice, then examined the prefrontal cortex and hippocampus using western blotting and immunohistochemistry to probe for SCZ-relevant molecular and cellular alterations. Compared to wild-type controls, HRM/DISC1 mice demonstrated impaired pre-pulse inhibition, altered cognition, and decreased activity. Diazepam failed to rescue anxiety-like behaviors, paradoxically increasing activity in HRM/DISC1 mice. At a cellular level, we found increased α1-subunit containing GABA receptors in the prefrontal cortex, and a reduction in fast-spiking parvalbumin positive neurons. Maturation of adult-born neurons in the hippocampus was also altered in HRM/DISC1 mice. While there was no difference in the total number proliferating cells, more of these cells were in immature stages of development. Homozygous DISC1 mutation combined with RELN haploinsufficiency produces a complex phenotype with neuropsychiatric characteristics relevant to SCZ and related disorders, expanding our understanding of how multiple genetic susceptibility factors might interact to influence the variable presentation of these disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Danielle Gulick
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
3
|
Sharma A, Hill KE, Schwarzbauer JE. Extracellular matrix composition affects outgrowth of dendrites and dendritic spines on cortical neurons. Front Cell Neurosci 2023; 17:1177663. [PMID: 37388410 PMCID: PMC10300442 DOI: 10.3389/fncel.2023.1177663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/29/2023] [Indexed: 07/01/2023] Open
Abstract
The composition of the extracellular matrix (ECM) in nervous tissue plays an important role in controlling neuronal outgrowth and synapse development. Changes in both protein and glycosaminoglycan components of the ECM occur with tissue injury and may affect neuron growth. To investigate neuron responses to alterations in fibronectin (FN), a major component of the wound ECM, we grew cortical neurons on cell-derived decellularized matrices composed of wild type FN (FN+/+) or of a mutant form of FN (FNΔ/+) from which the III13 heparin-binding site had been deleted by CRISPR-Cas 9 gene editing. The most significant effect of the mutant FN was a reduction in dendrite outgrowth. Not only were dendrites shorter on mutant FNΔ/+-collagen (COL) matrix than on wild type (FN+/+-COL) matrix, but the number of dendrites and dendritic spines per neuron and the spine densities were also dramatically reduced on FNΔ/+-COL matrices. Mass spectrometry and immunostaining identified a reduction in tenascin-C (TN-C) levels in the mutant matrix. TN-C is an ECM protein that binds to the III13 site of FN and modulates cell-matrix interactions and has been linked to dendrite development. We propose that TN-C binding to FN in the wound matrix supports dendrite and spine development during repair of damaged neural tissue. Overall, these results show that changes in ECM composition can dramatically affect elaboration of neurites and support the idea that the ECM microenvironment controls neuron morphology and connectivity.
Collapse
Affiliation(s)
| | | | - Jean E. Schwarzbauer
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States
| |
Collapse
|
4
|
Howes OD, Onwordi EC. The synaptic hypothesis of schizophrenia version III: a master mechanism. Mol Psychiatry 2023; 28:1843-1856. [PMID: 37041418 PMCID: PMC10575788 DOI: 10.1038/s41380-023-02043-w] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 04/13/2023]
Abstract
The synaptic hypothesis of schizophrenia has been highly influential. However, new approaches mean there has been a step-change in the evidence available, and some tenets of earlier versions are not supported by recent findings. Here, we review normal synaptic development and evidence from structural and functional imaging and post-mortem studies that this is abnormal in people at risk and with schizophrenia. We then consider the mechanism that could underlie synaptic changes and update the hypothesis. Genome-wide association studies have identified a number of schizophrenia risk variants converging on pathways regulating synaptic elimination, formation and plasticity, including complement factors and microglial-mediated synaptic pruning. Induced pluripotent stem cell studies have demonstrated that patient-derived neurons show pre- and post-synaptic deficits, synaptic signalling alterations, and elevated, complement-dependent elimination of synaptic structures compared to control-derived lines. Preclinical data show that environmental risk factors linked to schizophrenia, such as stress and immune activation, can lead to synapse loss. Longitudinal MRI studies in patients, including in the prodrome, show divergent trajectories in grey matter volume and cortical thickness compared to controls, and PET imaging shows in vivo evidence for lower synaptic density in patients with schizophrenia. Based on this evidence, we propose version III of the synaptic hypothesis. This is a multi-hit model, whereby genetic and/or environmental risk factors render synapses vulnerable to excessive glia-mediated elimination triggered by stress during later neurodevelopment. We propose the loss of synapses disrupts pyramidal neuron function in the cortex to contribute to negative and cognitive symptoms and disinhibits projections to mesostriatal regions to contribute to dopamine overactivity and psychosis. It accounts for the typical onset of schizophrenia in adolescence/early adulthood, its major risk factors, and symptoms, and identifies potential synaptic, microglial and immune targets for treatment.
Collapse
Affiliation(s)
- Oliver D Howes
- Faculty of Medicine, Institute of Clinical Sciences (ICS), Imperial College London, London, W12 0NN, UK.
- Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, W12 0NN, UK.
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 8AF, UK.
| | - Ellis Chika Onwordi
- Faculty of Medicine, Institute of Clinical Sciences (ICS), Imperial College London, London, W12 0NN, UK.
- Psychiatric Imaging Group, Medical Research Council, London Institute of Medical Sciences, Hammersmith Hospital, London, W12 0NN, UK.
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 8AF, UK.
- Centre for Psychiatry and Mental Health, Wolfson Institute of Population Health, Queen Mary University of London, London, E1 2AB, UK.
| |
Collapse
|
5
|
Lyu J, Nagarajan R, Kambali M, Wang M, Rudolph U. Selective inhibition of somatostatin-positive dentate hilar interneurons induces age-related cellular changes and cognitive dysfunction. PNAS NEXUS 2023; 2:pgad134. [PMID: 37168673 PMCID: PMC10165806 DOI: 10.1093/pnasnexus/pgad134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/19/2023] [Accepted: 04/04/2023] [Indexed: 05/13/2023]
Abstract
The cellular basis of age-related impairments of hippocampal function is not fully understood. In order to evaluate the role of somatostatin-positive (Sst+) interneurons in the dentate gyrus (DG) hilus in this process, we chemogenetically inhibited Sst+ interneurons in the DG hilus. Chronic chemogenetic inhibition (CCI) of these neurons resulted in increased c-Fos staining in the DG hilus, a decrease in the percentage of GAD67- and of Sst-expressing interneurons in the DG, and increased microglial activation in DG, CA3, and CA1. Total dendritic length and spine density were reduced in DG and CA1, suggesting reduced dendritic complexity. Behaviorally, the recognition index in an object recognition task and the percentage of spontaneous alternations in the Y-maze were decreased, while in both initial and reversal learning in the Morris water maze, the latencies to find the hidden platform were increased, suggesting cognitive dysfunction. Our findings establish a causal role for a reduced function of Sst+ interneurons in the DG hilus for cognitive decline and suggest that this reduced function may contribute to age-related impairments of learning and memory. Furthermore, our CCI mice may represent a cellularly defined model of hippocampal aging.
Collapse
Affiliation(s)
- Jinrui Lyu
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802-6178, USA
- Neuroscience Program, College of Liberal Arts and Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61802-6178, USA
| | - Rajasekar Nagarajan
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802-6178, USA
| | - Maltesh Kambali
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802-6178, USA
| | - Muxiao Wang
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802-6178, USA
- Neuroscience Program, College of Liberal Arts and Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61802-6178, USA
| | | |
Collapse
|
6
|
Extracellular matrix and synapse formation. Biosci Rep 2023; 43:232259. [PMID: 36503961 PMCID: PMC9829651 DOI: 10.1042/bsr20212411] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 11/08/2022] [Accepted: 11/24/2022] [Indexed: 12/14/2022] Open
Abstract
The extracellular matrix (ECM) is a complex molecular network distributed throughout the extracellular space of different tissues as well as the neuronal system. Previous studies have identified various ECM components that play important roles in neuronal maturation and signal transduction. ECM components are reported to be involved in neurogenesis, neuronal migration, and axonal growth by interacting or binding to specific receptors. In addition, the ECM is found to regulate synapse formation, the stability of the synaptic structure, and synaptic plasticity. Here, we mainly reviewed the effects of various ECM components on synapse formation and briefly described the related diseases caused by the abnormality of several ECM components.
Collapse
|
7
|
Su Y, Yang X, Yang L, Liu X, She Z, Zhang Y, Dong Z. Thyroid hormones regulate reelin expression in neuropsychiatric disorders. Can J Physiol Pharmacol 2022; 100:1033-1044. [PMID: 36166833 DOI: 10.1139/cjpp-2022-0270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The incidence and prevalence of hypothyroidism in pregnancy have increased over the past two decades, leading to the occurrence of neuropsychiatric disorders. However, the underlying mechanisms of thyroid hormone (TH)-regulated gene expression and neuropsychiatric development during the postnatal period remain unknown. Recent achievements have shown that reelin, a large extracellular glycoprotein, plays a crucial role in neuronal migration and localization during the development of neocortex and cerebellar cortex, thereby participating in the development of neuropsychiatric diseases. Reelin-induced neuronal migration requires triiodothyronine (T3) from the deiodination of thyroxine (T4) by fetal brain deiodinases. Previous studies have reported decreased reelin levels and abnormal gene expression, which are the same as the pathological alternations in reelin-induced neuropsychiatric disorders including schizophrenia and autism. Low T3 in the fetal brain due to hypothyroxinemia during pregnancy may be detrimental to neuronal migration, leading to neuropsychiatric disorders. In this review, we focus on the reelin expression between hypothyroidism and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Yadi Su
- College of Stomatology, Chongqing Medical University, Chongqing, 401334, PR China
| | - Xiaoyu Yang
- College of Pediatrics, Chongqing Medical University, Chongqing, 401334, PR China
| | - Lu Yang
- College of Stomatology, Chongqing Medical University, Chongqing, 401334, PR China
| | - Xinjing Liu
- College of Public Health and Management, Chongqing Medical University, Chongqing, 401334, PR China
| | - Zhenghang She
- College of Pediatrics, Chongqing Medical University, Chongqing, 401334, PR China
| | - Youwen Zhang
- College of Pediatrics, Chongqing Medical University, Chongqing, 401334, PR China
| | - Zhifang Dong
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, PR China
| |
Collapse
|
8
|
Structure of Reelin repeat 8 and the adjacent C-terminal region. Biophys J 2022; 121:2526-2537. [DOI: 10.1016/j.bpj.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/15/2022] [Accepted: 05/31/2022] [Indexed: 11/02/2022] Open
|
9
|
Scala M, Grasso EA, Di Cara G, Riva A, Striano P, Verrotti A. The Pathophysiological Link Between Reelin and Autism: Overview and New Insights. Front Genet 2022; 13:869002. [PMID: 35422848 PMCID: PMC9002092 DOI: 10.3389/fgene.2022.869002] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Reelin is a secreted extracellular matrix protein playing pivotal roles in neuronal migration and cortical stratification during embryonal brain development. In the adult brain, its activity is crucial for synaptic plasticity, memory processing, and cognition. Genetic alterations in RELN have been variably reported as possible contributors to the pathogenesis of autism spectrum disorders (ASD). In particular, GCCs repeats in the 5′UTR, and single nucleotide polymorphysms (SNPs) in RELN have been suggested to affect brain development and predispose to autism. We reviewed pertinent literature on RELN expression and haplotypes transmission in children with ASD, critically analyzing available evidence in support of the pathophysiological association between Reelin deficiency and ASD.
Collapse
Affiliation(s)
- Marcello Scala
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal, and Child Health (DINOGMI), University of Genoa, Genoa, Italy.,Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, University of Genoa, Genoa, Italy
| | | | - Giuseppe Di Cara
- Department of Medicine and Surgery, Pediatric Clinic, University of Perugia, Perugia, Italy
| | - Antonella Riva
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal, and Child Health (DINOGMI), University of Genoa, Genoa, Italy.,Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, University of Genoa, Genoa, Italy
| | - Pasquale Striano
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal, and Child Health (DINOGMI), University of Genoa, Genoa, Italy.,Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto Giannina Gaslini, University of Genoa, Genoa, Italy
| | - Alberto Verrotti
- Department of Medicine and Surgery, Pediatric Clinic, University of Perugia, Perugia, Italy
| |
Collapse
|
10
|
Nelson MM, Hoff JD, Zeese ML, Corfas G. Poly (ADP-Ribose) Polymerase 1 Regulates Cajal-Retzius Cell Development and Neural Precursor Cell Adhesion. Front Cell Dev Biol 2021; 9:693595. [PMID: 34708032 PMCID: PMC8542860 DOI: 10.3389/fcell.2021.693595] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 09/16/2021] [Indexed: 11/13/2022] Open
Abstract
Poly (ADP-ribose) polymerase 1 (PARP1) is a ubiquitously expressed enzyme that regulates DNA damage repair, cell death, inflammation, and transcription. PARP1 functions by adding ADP-ribose polymers (PAR) to proteins including itself, using NAD+ as a donor. This post-translational modification known as PARylation results in changes in the activity of PARP1 and its substrate proteins and has been linked to the pathogenesis of various neurological diseases. PARP1 KO mice display schizophrenia-like behaviors, have impaired memory formation, and have defects in neuronal proliferation and survival, while mutations in genes that affect PARylation have been associated with intellectual disability, psychosis, neurodegeneration, and stroke in humans. Yet, the roles of PARP1 in brain development have not been extensively studied. We now find that loss of PARP1 leads to defects in brain development and increased neuronal density at birth. We further demonstrate that PARP1 loss increases the expression levels of genes associated with neuronal migration and adhesion in the E15.5 cerebral cortex, including Reln. This correlates with an increased number of Cajal–Retzius (CR) cells in vivo and in cultures of embryonic neural progenitor cells (NPCs) derived from the PARP1 KO cortex. Furthermore, PARP1 loss leads to increased NPC adhesion to N-cadherin, like that induced by experimental exposure to Reelin. Taken together, these results uncover a novel role for PARP1 in brain development, i.e., regulation of CR cells, neuronal density, and cell adhesion.
Collapse
Affiliation(s)
- Megan M Nelson
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, MI, United States.,Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, United States
| | - J Damon Hoff
- Single Molecule Analysis in Real-Time Center, Department of Biophysics, University of Michigan, Ann Arbor, MI, United States
| | - Mya L Zeese
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, MI, United States.,Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Gabriel Corfas
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, MI, United States.,Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
11
|
Nishibe M, Toyoda H, Hiraga SI, Yamashita T, Katsuyama Y. Synaptic and Genetic Bases of Impaired Motor Learning Associated with Modified Experience-Dependent Cortical Plasticity in Heterozygous Reeler Mutants. Cereb Cortex 2021; 32:504-519. [PMID: 34339488 DOI: 10.1093/cercor/bhab227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 06/11/2021] [Accepted: 06/13/2021] [Indexed: 11/12/2022] Open
Abstract
Patients with neurodevelopmental disorders show impaired motor skill learning. It is unclear how the effect of genetic variation on synaptic function and transcriptome profile may underlie experience-dependent cortical plasticity, which supports the development of fine motor skills. RELN (reelin) is one of the genes implicated in neurodevelopmental psychiatric vulnerability. Heterozygous reeler mutant (HRM) mice displayed impairments in reach-to-grasp learning, accompanied by less extensive cortical map reorganization compared with wild-type mice, examined after 10 days of training by intracortical microstimulation. Assessed by patch-clamp recordings after 3 days of training, the training induced synaptic potentiation and increased glutamatergic-transmission of cortical layer III pyramidal neurons in wild-type mice. In contrast, the basal excitatory and inhibitory synaptic functions were depressed, affected both by presynaptic and postsynaptic impairments in HRM mice; and thus, no further training-induced synaptic plasticity occurred. HRM exhibited downregulations of cortical synaptophysin, immediate-early gene expressions, and gene enrichment, in response to 3 days of training compared with trained wild-type mice, shown using quantitative reverse transcription polymerase chain reaction, immunohistochemisty, and RNA-sequencing. We demonstrated that motor learning impairments associated with modified experience-dependent cortical plasticity are at least partially attributed by the basal synaptic alternation as well as the aberrant early experience-induced gene enrichment in HRM.
Collapse
Affiliation(s)
- Mariko Nishibe
- Office of Strategic Innovative Dentistry, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan.,Department of Anatomy, Shiga University of Medical Science, Shiga 520-2192, Japan
| | - Hiroki Toyoda
- Department of Oral Physiology, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| | - Shin-Ichiro Hiraga
- Department of Neuromedical Science, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Toshihide Yamashita
- Department of Neuromedical Science, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.,Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.,Department of Molecular Neuroscience, WPI Immunology Frontier Research Center, Osaka 565-0871, Japan.,Graduate School of Frontier Biosciences, Osaka University, Osaka 565-0871, Japan
| | - Yu Katsuyama
- Department of Anatomy, Shiga University of Medical Science, Shiga 520-2192, Japan
| |
Collapse
|
12
|
Pahle J, Muhia M, Wagener RJ, Tippmann A, Bock HH, Graw J, Herz J, Staiger JF, Drakew A, Kneussel M, Rune GM, Frotscher M, Brunne B. Selective Inactivation of Reelin in Inhibitory Interneurons Leads to Subtle Changes in the Dentate Gyrus But Leaves Cortical Layering and Behavior Unaffected. Cereb Cortex 2021; 30:1688-1707. [PMID: 31667489 PMCID: PMC7132935 DOI: 10.1093/cercor/bhz196] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Reelin is an extracellular matrix protein, known for its dual role in neuronal migration during brain development and in synaptic plasticity at adult stages. During the perinatal phase, Reelin expression switches from Cajal-Retzius (CR) cells, its main source before birth, to inhibitory interneurons (IN), the main source of Reelin in the adult forebrain. IN-derived Reelin has been associated with schizophrenia and temporal lobe epilepsy; however, the functional role of Reelin from INs is presently unclear. In this study, we used conditional knockout mice, which lack Reelin expression specifically in inhibitory INs, leading to a substantial reduction in total Reelin expression in the neocortex and dentate gyrus. Our results show that IN-specific Reelin knockout mice exhibit normal neuronal layering and normal behavior, including spatial reference memory. Although INs are the major source of Reelin within the adult stem cell niche, Reelin from INs does not contribute substantially to normal adult neurogenesis. While a closer look at the dentate gyrus revealed some unexpected alterations at the cellular level, including an increase in the number of Reelin expressing CR cells, overall our data suggest that Reelin derived from INs is less critical for cortex development and function than Reelin expressed by CR cells.
Collapse
Affiliation(s)
- Jasmine Pahle
- Institute for Structural Neurobiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Mary Muhia
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Robin J Wagener
- Neurology Clinic, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Anja Tippmann
- Institute for Structural Neurobiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.,Department of Systems Neuroscience, Johann-Friedrich-Blumenbach Institute for Zoology and Anthropology, University of Göttingen, 37075 Göttingen, Germany
| | - Hans H Bock
- Clinic of Gastroenterology and Hepatology, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany
| | - Janice Graw
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jochen F Staiger
- Institute for Neuroanatomy, University Medical Center Göttingen, Georg-August-University Göttingen, 37075 Göttingen, Germany
| | - Alexander Drakew
- Institute for Structural Neurobiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.,Institute of Clinical Neuroanatomy, Faculty of Medicine, 60590 Frankfurt, Germany
| | - Matthias Kneussel
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Gabriele M Rune
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Michael Frotscher
- Institute for Structural Neurobiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Bianka Brunne
- Institute for Structural Neurobiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.,Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
13
|
Turk LS, Kuang X, Dal Pozzo V, Patel K, Chen M, Huynh K, Currie MJ, Mitchell D, Dobson RCJ, D'Arcangelo G, Dai W, Comoletti D. The structure-function relationship of a signaling-competent, dimeric Reelin fragment. Structure 2021; 29:1156-1170.e6. [PMID: 34089653 DOI: 10.1016/j.str.2021.05.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/26/2021] [Accepted: 05/14/2021] [Indexed: 01/21/2023]
Abstract
Reelin operates through canonical and non-canonical pathways that mediate several aspects of brain development and function. Reelin's dimeric central fragment (CF), generated through proteolytic cleavage, is required for the lipoprotein-receptor-dependent canonical pathway activation. Here, we analyze the signaling properties of a variety of Reelin fragments and measure the differential binding affinities of monomeric and dimeric CF fragments to lipoprotein receptors to investigate the mode of canonical signal activation. We also present the cryoelectron tomography-solved dimeric structure of Reelin CF and support it using several other biophysical techniques. Our findings suggest that Reelin CF forms a covalent parallel dimer with some degree of flexibility between the two protein chains. As a result of this conformation, Reelin binds to lipoprotein receptors in a manner inaccessible to its monomeric form and is capable of stimulating canonical pathway signaling.
Collapse
Affiliation(s)
- Liam S Turk
- Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA; Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA; School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Xuyuan Kuang
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Department of Hyperbaric Oxygen, Central South University, Changsha, Hunan Province, China
| | - Valentina Dal Pozzo
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Khush Patel
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Muyuan Chen
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kevin Huynh
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Michael J Currie
- Biomolecular Interactions Centre and School of Biological Sciences, University of Canterbury, Christchurch 8041, New Zealand
| | - Daniel Mitchell
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Renwick C J Dobson
- Biomolecular Interactions Centre and School of Biological Sciences, University of Canterbury, Christchurch 8041, New Zealand; Bio21 Molecular Science and Biotechnology Institute, Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Gabriella D'Arcangelo
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Wei Dai
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Institute for Quantitative Biomedicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.
| | - Davide Comoletti
- Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA; Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA; School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand.
| |
Collapse
|
14
|
Yang J, Yang X, Tang K. Interneuron development and dysfunction. FEBS J 2021; 289:2318-2336. [PMID: 33844440 DOI: 10.1111/febs.15872] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 04/09/2021] [Indexed: 12/17/2022]
Abstract
Understanding excitation and inhibition balance in the brain begins with the tale of two basic types of neurons, glutamatergic projection neurons and GABAergic interneurons. The diversity of cortical interneurons is contributed by multiple origins in the ventral forebrain, various tangential migration routes, and complicated regulations of intrinsic factors, extrinsic signals, and activities. Abnormalities of interneuron development lead to dysfunction of interneurons and inhibitory circuits, which are highly associated with neurodevelopmental disorders including schizophrenia, autism spectrum disorders, and intellectual disability. In this review, we mainly discuss recent findings on the development of cortical interneuron and on neurodevelopmental disorders related to interneuron dysfunction.
Collapse
Affiliation(s)
- Jiaxin Yang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, China
| | - Xiong Yang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, China
| | - Ke Tang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, China
| |
Collapse
|
15
|
Lu MH, Hsueh YP. Protein synthesis as a modifiable target for autism-related dendritic spine pathophysiologies. FEBS J 2021; 289:2282-2300. [PMID: 33511762 DOI: 10.1111/febs.15733] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/04/2021] [Accepted: 01/26/2021] [Indexed: 12/20/2022]
Abstract
Autism spectrum disorder (ASD) is increasingly recognized as a condition of altered brain connectivity. As synapses are fundamental subcellular structures for neuronal connectivity, synaptic pathophysiology has become one of central themes in autism research. Reports disagree upon whether the density of dendritic spines, namely excitatory synapses, is increased or decreased in ASD and whether the protein synthesis that is critical for dendritic spine formation and function is upregulated or downregulated. Here, we review recent evidence supporting a subgroup of ASD models with decreased dendritic spine density (hereafter ASD-DSD), including Nf1 and Vcp mutant mice. We discuss the relevance of branched-chain amino acid (BCAA) insufficiency in relation to unmet protein synthesis demand in ASD-DSD. In contrast to ASD-DSD, ASD models with hyperactive mammalian target of rapamycin (mTOR) may represent the opposite end of the disease spectrum, often characterized by increases in protein synthesis and dendritic spine density (denoted ASD-ISD). Finally, we propose personalized dietary leucine as a strategy tailored to balancing protein synthesis demand, thereby ameliorating dendritic spine pathophysiologies and autism-related phenotypes in susceptible patients, especially those with ASD-DSD.
Collapse
Affiliation(s)
- Ming-Hsuan Lu
- Department of Medical Education, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Yi-Ping Hsueh
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, ROC
| |
Collapse
|
16
|
Carroll SH, Macias Trevino C, Li EB, Kawasaki K, Myers N, Hallett SA, Alhazmi N, Cotney J, Carstens RP, Liao EC. An Irf6- Esrp1/2 regulatory axis controls midface morphogenesis in vertebrates. Development 2020; 147:dev194498. [PMID: 33234718 PMCID: PMC7774891 DOI: 10.1242/dev.194498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 11/02/2020] [Indexed: 12/25/2022]
Abstract
Irf6 and Esrp1 are important for palate development across vertebrates. In zebrafish, we found that irf6 regulates the expression of esrp1 We detailed overlapping Irf6 and Esrp1/2 expression in mouse orofacial epithelium. In zebrafish, irf6 and esrp1/2 share expression in periderm, frontonasal ectoderm and oral epithelium. Genetic disruption of irf6 and esrp1/2 in zebrafish resulted in cleft of the anterior neurocranium. The esrp1/2 mutant also developed cleft of the mouth opening. Lineage tracing of cranial neural crest cells revealed that the cleft resulted not from migration defect, but from impaired chondrogenesis. Analysis of aberrant cells within the cleft revealed expression of sox10, col1a1 and irf6, and these cells were adjacent to krt4+ and krt5+ cells. Breeding of mouse Irf6; Esrp1; Esrp2 compound mutants suggested genetic interaction, as the triple homozygote and the Irf6; Esrp1 double homozygote were not observed. Further, Irf6 heterozygosity reduced Esrp1/2 cleft severity. These studies highlight the complementary analysis of Irf6 and Esrp1/2 in mouse and zebrafish, and identify a unique aberrant cell population in zebrafish expressing sox10, col1a1 and irf6 Future work characterizing this cell population will yield additional insight into cleft pathogenesis.
Collapse
Affiliation(s)
- Shannon H. Carroll
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Shriners Hospital for Children, Boston, MA 02114, USA
| | - Claudio Macias Trevino
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | | | - Kenta Kawasaki
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Shriners Hospital for Children, Boston, MA 02114, USA
| | - Nikita Myers
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Shawn A. Hallett
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Nora Alhazmi
- Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Justin Cotney
- Department of Genetics and Genome Sciences, University of Connecticut Health, CT 06030, USA
| | - Russ P. Carstens
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eric C. Liao
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Shriners Hospital for Children, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
17
|
Faini G, Del Bene F, Albadri S. Reelin functions beyond neuronal migration: from synaptogenesis to network activity modulation. Curr Opin Neurobiol 2020; 66:135-143. [PMID: 33197872 DOI: 10.1016/j.conb.2020.10.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/08/2020] [Accepted: 10/11/2020] [Indexed: 01/05/2023]
Abstract
Reelin, a glycoprotein of the extracellular matrix, has been the focus of several studies over the years, mostly for its role in cell migration. Here we report the role of this molecule and of its downstream pathways in post-mitotic neurons and how they contribute to neural circuit assembly, refinement and function. Accumulating evidence has pointed at a major role for Reelin in axonal guidance, synaptogenesis and dendritic spine formation. In particular, new evidence points at a direct role in axonal targeting and refinement at the target site. In addition, recent advances highlight new functions of Reelin in the modulation of synaptic activity, plasticity and behavior and in the direct regulation of GABA receptors expression and stability. We discuss these findings in the context of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Giulia Faini
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Filippo Del Bene
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France.
| | - Shahad Albadri
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| |
Collapse
|
18
|
Jossin Y. Reelin Functions, Mechanisms of Action and Signaling Pathways During Brain Development and Maturation. Biomolecules 2020; 10:biom10060964. [PMID: 32604886 PMCID: PMC7355739 DOI: 10.3390/biom10060964] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 06/24/2020] [Accepted: 06/24/2020] [Indexed: 12/14/2022] Open
Abstract
During embryonic development and adulthood, Reelin exerts several important functions in the brain including the regulation of neuronal migration, dendritic growth and branching, dendritic spine formation, synaptogenesis and synaptic plasticity. As a consequence, the Reelin signaling pathway has been associated with several human brain disorders such as lissencephaly, autism, schizophrenia, bipolar disorder, depression, mental retardation, Alzheimer’s disease and epilepsy. Several elements of the signaling pathway are known. Core components, such as the Reelin receptors very low-density lipoprotein receptor (VLDLR) and Apolipoprotein E receptor 2 (ApoER2), Src family kinases Src and Fyn, and the intracellular adaptor Disabled-1 (Dab1), are common to most but not all Reelin functions. Other downstream effectors are, on the other hand, more specific to defined tasks. Reelin is a large extracellular protein, and some aspects of the signal are regulated by its processing into smaller fragments. Rather than being inhibitory, the processing at two major sites seems to be fulfilling important physiological functions. In this review, I describe the various cellular events regulated by Reelin and attempt to explain the current knowledge on the mechanisms of action. After discussing the shared and distinct elements of the Reelin signaling pathway involved in neuronal migration, dendritic growth, spine development and synaptic plasticity, I briefly outline the data revealing the importance of Reelin in human brain disorders.
Collapse
Affiliation(s)
- Yves Jossin
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, 1200 Brussels, Belgium
| |
Collapse
|
19
|
Wang L, Zhao D, Wang M, Wang Y, Vreugdenhil M, Lin J, Lu C. Modulation of Hippocampal Gamma Oscillations by Dopamine in Heterozygous Reeler Mice in vitro. Front Cell Neurosci 2020; 13:586. [PMID: 32116553 PMCID: PMC7026475 DOI: 10.3389/fncel.2019.00586] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/23/2019] [Indexed: 11/14/2022] Open
Abstract
The reelin haploinsufficient heterozygous reeler mice (HRM), an animal model of schizophrenia, have altered mesolimbic dopaminergic pathways and share similar neurochemical and behavioral properties with patients with schizophrenia. Dysfunctional neural circuitry with impaired gamma (γ) oscillation (30–80 Hz) has been implicated in abnormal cognition in patients with schizophrenia. However, the function of neural circuitry in terms of γ oscillation and its modulation by dopamine (DA) has not been reported in HRM. In this study, first, we recorded γ oscillations in CA3 from wild-type mice (WTM) and HRM hippocampal slices, and we studied the effects of DA on γ oscillations. We found that there was no difference in γ power between WTM and HRM and that DA increased γ power of WTM but not HRM, suggesting that DA modulations of network oscillations in HRM are impaired. Second, we found that N-methyl-D-aspartate receptor (NMDAR) antagonist MK-801 itself increased γ power and occluded DA-mediated enhancement of γ power in WTM but partially restored DA modulation of γ oscillations in HRM. Third, inhibition of phosphatidylinositol 3-kinase (PI3K), a downstream molecule of NMDAR, increased γ power and blocked the effects of DA on γ oscillation in WTM and had no significant effect on γ power but largely restored DA modulation of γ oscillations in HRM. Our results reveal that impaired DA function in HRM is associated with dysregulated NMDAR–PI3K signaling, a mechanism that may be relevant in the pathology of schizophrenia.
Collapse
Affiliation(s)
- Lu Wang
- The International-Joint Lab for Non-Invasive Neural Modulation, Xinxiang Medical University, Xinxiang, China.,Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical University, Xinxiang, China.,Department of Neurobiology and Physiology, Xinxiang Medical University, Xinxiang, China
| | - Dandan Zhao
- The International-Joint Lab for Non-Invasive Neural Modulation, Xinxiang Medical University, Xinxiang, China.,Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical University, Xinxiang, China.,Department of Neurobiology and Physiology, Xinxiang Medical University, Xinxiang, China
| | - Mengmeng Wang
- The International-Joint Lab for Non-Invasive Neural Modulation, Xinxiang Medical University, Xinxiang, China.,Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical University, Xinxiang, China.,Department of Neurobiology and Physiology, Xinxiang Medical University, Xinxiang, China
| | - Yuan Wang
- The International-Joint Lab for Non-Invasive Neural Modulation, Xinxiang Medical University, Xinxiang, China.,Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical University, Xinxiang, China
| | - Martin Vreugdenhil
- Department of Life Science, School of Health Sciences, Birmingham City University, Birmingham, United Kingdom
| | - Juntang Lin
- School of Biomedical Engineering, Xinxiang Medical University, Xinxiang, China
| | - Chengbiao Lu
- The International-Joint Lab for Non-Invasive Neural Modulation, Xinxiang Medical University, Xinxiang, China.,Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical University, Xinxiang, China.,Department of Neurobiology and Physiology, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
20
|
The Reeler Mouse: A Translational Model of Human Neurological Conditions, or Simply a Good Tool for Better Understanding Neurodevelopment? J Clin Med 2019; 8:jcm8122088. [PMID: 31805691 PMCID: PMC6947477 DOI: 10.3390/jcm8122088] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/26/2019] [Accepted: 11/28/2019] [Indexed: 12/25/2022] Open
Abstract
The first description of the Reeler mutation in mouse dates to more than fifty years ago, and later, its causative gene (reln) was discovered in mouse, and its human orthologue (RELN) was demonstrated to be causative of lissencephaly 2 (LIS2) and about 20% of the cases of autosomal-dominant lateral temporal epilepsy (ADLTE). In both human and mice, the gene encodes for a glycoprotein referred to as reelin (Reln) that plays a primary function in neuronal migration during development and synaptic stabilization in adulthood. Besides LIS2 and ADLTE, RELN and/or other genes coding for the proteins of the Reln intracellular cascade have been associated substantially to other conditions such as spinocerebellar ataxia type 7 and 37, VLDLR-associated cerebellar hypoplasia, PAFAH1B1-associated lissencephaly, autism, and schizophrenia. According to their modalities of inheritances and with significant differences among each other, these neuropsychiatric disorders can be modeled in the homozygous (reln−/−) or heterozygous (reln+/−) Reeler mouse. The worth of these mice as translational models is discussed, with focus on their construct and face validity. Description of face validity, i.e., the resemblance of phenotypes between the two species, centers onto the histological, neurochemical, and functional observations in the cerebral cortex, hippocampus, and cerebellum of Reeler mice and their human counterparts.
Collapse
|
21
|
Gaitanou M, Segklia K, Matsas R. Cend1, a Story with Many Tales: From Regulation of Cell Cycle Progression/Exit of Neural Stem Cells to Brain Structure and Function. Stem Cells Int 2019; 2019:2054783. [PMID: 31191667 PMCID: PMC6525816 DOI: 10.1155/2019/2054783] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/21/2019] [Accepted: 02/07/2019] [Indexed: 12/15/2022] Open
Abstract
Neural stem/precursor cells (NPCs) generate the large variety of neuronal phenotypes comprising the adult brain. The high diversity and complexity of this organ have its origin in embryonic life, during which NPCs undergo symmetric and asymmetric divisions and then exit the cell cycle and differentiate to acquire neuronal identities. During these processes, coordinated regulation of cell cycle progression/exit and differentiation is essential for generation of the appropriate number of neurons and formation of the correct structural and functional neuronal circuits in the adult brain. Cend1 is a neuronal lineage-specific modulator involved in synchronization of cell cycle exit and differentiation of neuronal precursors. It is expressed all along the neuronal lineage, from neural stem/progenitor cells to mature neurons, and is associated with the dynamics of neuron-generating divisions. Functional studies showed that Cend1 has a critical role during neurogenesis in promoting cell cycle exit and neuronal differentiation. Mechanistically, Cend1 acts via the p53-dependent/Cyclin D1/pRb signaling pathway as well as via a p53-independent route involving a tripartite interaction with RanBPM and Dyrk1B. Upon Cend1 function, Notch1 signaling is suppressed and proneural genes such as Mash1 and Neurogenins 1/2 are induced. Due to its neurogenic activity, Cend1 is a promising candidate therapeutic gene for brain repair, while the Cend1 minimal promoter is a valuable tool for neuron-specific gene delivery in the CNS. Mice with Cend1 genetic ablation display increased NPC proliferation, decreased migration, and higher levels of apoptosis during development. As a result, they show in the adult brain deficits in a range of motor and nonmotor behaviors arising from irregularities in cerebellar cortex lamination and impaired Purkinje cell differentiation as well as a paucity in GABAergic interneurons of the cerebral cortex, hippocampus, and amygdala. Taken together, these studies highlight the necessity for Cend1 expression in the formation of a structurally and functionally normal brain.
Collapse
Affiliation(s)
- Maria Gaitanou
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, Vas. Sofias Avenue, 11521 Athens, Greece
| | - Katerina Segklia
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, Vas. Sofias Avenue, 11521 Athens, Greece
| | - Rebecca Matsas
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, Vas. Sofias Avenue, 11521 Athens, Greece
| |
Collapse
|
22
|
Loss-of-Huntingtin in Medial and Lateral Ganglionic Lineages Differentially Disrupts Regional Interneuron and Projection Neuron Subtypes and Promotes Huntington's Disease-Associated Behavioral, Cellular, and Pathological Hallmarks. J Neurosci 2019; 39:1892-1909. [PMID: 30626701 DOI: 10.1523/jneurosci.2443-18.2018] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/21/2018] [Accepted: 12/29/2018] [Indexed: 12/23/2022] Open
Abstract
Emerging studies are providing compelling evidence that the pathogenesis of Huntington's disease (HD), a neurodegenerative disorder with frequent midlife onset, encompasses developmental components. Moreover, our previous studies using a hypomorphic model targeting huntingtin during the neurodevelopmental period indicated that loss-of-function mechanisms account for this pathogenic developmental component (Arteaga-Bracho et al., 2016). In the present study, we specifically ascertained the roles of subpallial lineage species in eliciting the previously observed HD-like phenotypes. Accordingly, we used the Cre-loxP system to conditionally ablate the murine huntingtin gene (Httflx) in cells expressing the subpallial patterning markers Gsx2 (Gsx2-Cre) or Nkx2.1 (Nkx2.1-Cre) in Httflx mice of both sexes. These genetic manipulations elicited anxiety-like behaviors, hyperkinetic locomotion, age-dependent motor deficits, and weight loss in both Httflx;Gsx2-Cre and Httflx;Nkx2.1-Cre mice. In addition, these strains displayed unique but complementary spatial patterns of basal ganglia degeneration that are strikingly reminiscent of those seen in human cases of HD. Furthermore, we observed early deficits of somatostatin-positive and Reelin-positive interneurons in both Htt subpallial null strains, as well as early increases of cholinergic interneurons, Foxp2+ arkypallidal neurons, and incipient deficits with age-dependent loss of parvalbumin-positive neurons in Httflx;Nkx2.1-Cre mice. Overall, our findings indicate that selective loss-of-huntingtin function in subpallial lineages differentially disrupts the number, complement, and survival of forebrain interneurons and globus pallidus GABAergic neurons, thereby leading to the development of key neurological hallmarks of HD during adult life. Our findings have important implications for the establishment and deployment of neural circuitries and the integrity of network reserve in health and disease.SIGNIFICANCE STATEMENT Huntington's disease (HD) is a progressive degenerative disorder caused by aberrant trinucleotide expansion in the huntingtin gene. Mechanistically, this mutation involves both loss- and gain-of-function mechanisms affecting a broad array of cellular and molecular processes. Although huntingtin is widely expressed during adult life, the mutant protein only causes the demise of selective neuronal subtypes. The mechanisms accounting for this differential vulnerability remain elusive. In this study, we have demonstrated that loss-of-huntingtin function in subpallial lineages not only differentially disrupts distinct interneuron species early in life, but also leads to a pattern of neurological deficits that are reminiscent of HD. This work suggests that early disruption of selective neuronal subtypes may account for the profiles of enhanced regional cellular vulnerability to death in HD.
Collapse
|
23
|
Winship IR, Dursun SM, Baker GB, Balista PA, Kandratavicius L, Maia-de-Oliveira JP, Hallak J, Howland JG. An Overview of Animal Models Related to Schizophrenia. CANADIAN JOURNAL OF PSYCHIATRY. REVUE CANADIENNE DE PSYCHIATRIE 2019; 64:5-17. [PMID: 29742910 PMCID: PMC6364139 DOI: 10.1177/0706743718773728] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Schizophrenia is a heterogeneous psychiatric disorder that is poorly treated with current therapies. In this brief review, we provide an update regarding the use of animal models to study schizophrenia in an attempt to understand its aetiology and develop novel therapeutic strategies. Tremendous progress has been made developing and validating rodent models that replicate the aetiologies, brain pathologies, and behavioural abnormalities associated with schizophrenia in humans. Here, models are grouped into 3 categories-developmental, drug induced, and genetic-to reflect the heterogeneous risk factors associated with schizophrenia. Each of these models is associated with varied but overlapping pathophysiology, endophenotypes, behavioural abnormalities, and cognitive impairments. Studying schizophrenia using multiple models will permit an understanding of the core features of the disease, thereby facilitating preclinical research aimed at the development and validation of better pharmacotherapies to alter the progression of schizophrenia or alleviate its debilitating symptoms.
Collapse
Affiliation(s)
- Ian R Winship
- 1 Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta
| | - Serdar M Dursun
- 2 Department of Psychiatry, Neurochemical Research Unit and Bebensee Schizophrenia Research Unit, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta.,3 National Institute of Science and Technology-Translational Science, Brazil
| | - Glen B Baker
- 2 Department of Psychiatry, Neurochemical Research Unit and Bebensee Schizophrenia Research Unit, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta.,3 National Institute of Science and Technology-Translational Science, Brazil
| | - Priscila A Balista
- 4 Department of Pharmacy, Centro Universitario das Faculdades Metropolitanas Unidas, São Paulo, Brazil
| | - Ludmyla Kandratavicius
- 5 Department of Neuroscience and Behavior, Faculty of Medicine of Ribeirao Preto, University of São Paulo, Ribeirao Preto, Brazil
| | - Joao Paulo Maia-de-Oliveira
- 3 National Institute of Science and Technology-Translational Science, Brazil.,6 Department of Clinical Medicine, Rio Grande do Norte Federal University, Natal, Brazil
| | - Jaime Hallak
- 3 National Institute of Science and Technology-Translational Science, Brazil.,5 Department of Neuroscience and Behavior, Faculty of Medicine of Ribeirao Preto, University of São Paulo, Ribeirao Preto, Brazil.,7 Department of Psychiatry (NRU), University of Alberta, Edmonton, Alberta
| | - John G Howland
- 8 Department of Physiology, University of Saskatchewan, Saskatoon, Saskatchewan
| |
Collapse
|
24
|
Sobue A, Kushima I, Nagai T, Shan W, Kohno T, Aleksic B, Aoyama Y, Mori D, Arioka Y, Kawano N, Yamamoto M, Hattori M, Nabeshima T, Yamada K, Ozaki N. Genetic and animal model analyses reveal the pathogenic role of a novel deletion of RELN in schizophrenia. Sci Rep 2018; 8:13046. [PMID: 30158644 PMCID: PMC6115412 DOI: 10.1038/s41598-018-31390-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/23/2018] [Indexed: 12/30/2022] Open
Abstract
Reelin protein (RELN), an extracellular matrix protein, plays multiple roles that range from embryonic neuronal migration to spine formation in the adult brain. Results from genetic studies have suggested that RELN is associated with the risk of psychiatric disorders, including schizophrenia (SCZ). We previously identified a novel exonic deletion of RELN in a patient with SCZ. High-resolution copy number variation analysis revealed that this deletion included exons 52 to 58, which truncated the RELN in a similar manner to the Reln Orleans mutation (Relnrl-Orl). We examined the clinical features of this patient and confirmed a decreased serum level of RELN. To elucidate the pathophysiological role of the exonic deletion of RELN in SCZ, we conducted behavioral and neurochemical analyses using heterozygous Relnrl-Orl/+ mice. These mice exhibited abnormalities in anxiety, social behavior, and motor learning; the deficits in motor learning were ameliorated by antipsychotics. Methamphetamine-induced hyperactivity and dopamine release were significantly reduced in the Relnrl-Orl/+ mice. In addition, the levels of GABAergic markers were decreased in the brain of these mice. Taken together, our results suggest that the exonic deletion of RELN plays a pathological role, implicating functional changes in the dopaminergic and GABAergic systems, in the pathophysiology of SCZ.
Collapse
Affiliation(s)
- Akira Sobue
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Itaru Kushima
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan.,Institute for Advanced Research, Nagoya University, Nagoya, Aichi, Japan
| | - Taku Nagai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Wei Shan
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Takao Kohno
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Branko Aleksic
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yuki Aoyama
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Daisuke Mori
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan.,Brain and Mind Research Center, Nagoya University, Nagoya, Aichi, Japan
| | - Yuko Arioka
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan.,Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Aichi, Japan
| | - Naoko Kawano
- Institutes of Innovation for Future Society, Nagoya University, Nagoya, Aichi, Japan
| | - Maeri Yamamoto
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Mitsuharu Hattori
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory Fujita Health University, Graduate School of Health Sciences, Toyoake, Aichi, Japan.,Aino University, Ibaraki, Osaka, Japan
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan.
| |
Collapse
|
25
|
Brietzke E, Trevizol AP, Fries GR, Subramaniapillai M, Kapczinski F, McIntyre RS, Mansur RB. The impact of body mass index in gene expression of reelin pathway mediators in individuals with schizophrenia and mood disorders: A post-mortem study. J Psychiatr Res 2018; 102:186-191. [PMID: 29680575 DOI: 10.1016/j.jpsychires.2018.04.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/09/2018] [Accepted: 04/12/2018] [Indexed: 12/19/2022]
Abstract
The objective of this study was to compare the expression of genes involved in the reelin pathway, in the post-mortem brain of individuals with schizophrenia (SZ) and mood disorders (MD) with a healthy control (HC) group; and to investigate the role f body mass index (BMI) as a potential mediator. The "Gene Expression in Postmortem dlPFC and Hippocampus from Schizophrenia and Mood Disorders" study holds microarray data on individuals with SZ, MD and HCs (from whom 849 specimens are from the dlPFC and 579 from the hippocampus). mRNA data was obtained using HumanHT-12 v4 BeadChip arrays (Illumina). Multivariate analysis of covariance were used to investigate the main effects of group and relevant covariates on RELNm, NOTCH1, GRIN1m, GRIN3A, CAMK2Gm, CAMK2A, CAMK2Bm, CAMK2N2, GRIN2Bm, GRIN2A, CREBBPm, APOE, LDLR and DAB1 gene expression. In the dlPFC, individuals with SZ had higher expression, relative to HCs, of APOE. Individuals with MD had higher expression, relative to HCs, of CAMK2A, CAMK2N2, and GRIN2Bm. Moreover, individuals with MD had higher expression, relative to SZ patients, of CAMK2N2. There were significant group by BMI effects for expression of RELN, CAMK2A, CAMK2N2, and GRIN2A. In the hippocampus, individuals with MD had lower expression, relative to HCs, of APOE. The results of this study suggest that the expression of genes related to the reelin pathway could be different between individuals with SZ and MD and healthy controls, with a greater vulnerability associated with greater BMI.
Collapse
Affiliation(s)
- Elisa Brietzke
- Mood Disorders Psychopharmacology Unit (MDPU), University Health Network, Toronto, Canada; Department of Psychiatry, Universidade Federal de São Paulo, São Paulo, Brazil; Research Group in Molecular and Behavioral Neuroscience of Bipolar Disorder, Department of Psychiatry, Universidade Federal de São Paulo, SP, Brazil; University of Toronto, Toronto, Canada
| | - Alisson P Trevizol
- Mood Disorders Psychopharmacology Unit (MDPU), University Health Network, Toronto, Canada; University of Toronto, Toronto, Canada
| | - Gabriel R Fries
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, USA
| | - Mehala Subramaniapillai
- Mood Disorders Psychopharmacology Unit (MDPU), University Health Network, Toronto, Canada; Department of Psychiatry, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Flavio Kapczinski
- Department of Psychiatry & Behavioral Neurosciences, Mcmaster University, Hamilton, Canada
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit (MDPU), University Health Network, Toronto, Canada; University of Toronto, Toronto, Canada
| | - Rodrigo B Mansur
- Mood Disorders Psychopharmacology Unit (MDPU), University Health Network, Toronto, Canada; University of Toronto, Toronto, Canada.
| |
Collapse
|
26
|
Guy J, Sachkova A, Möck M, Witte M, Wagener RJ, Staiger JF. Intracortical Network Effects Preserve Thalamocortical Input Efficacy in a Cortex Without Layers. Cereb Cortex 2018; 27:4851-4866. [PMID: 27620977 DOI: 10.1093/cercor/bhw281] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 08/17/2016] [Indexed: 12/11/2022] Open
Abstract
Layer IV (LIV) of the rodent somatosensory cortex contains the somatotopic barrel field. Barrels receive much of the sensory input to the cortex through innervation by thalamocortical axons from the ventral posteromedial nucleus. In the reeler mouse, the absence of cortical layers results in the formation of mispositioned barrel-equivalent clusters of LIV fated neurons. Although functional imaging suggests that sensory input activates the cortex, little is known about the cellular and synaptic properties of identified excitatory neurons of the reeler cortex. We examined the properties of thalamic input to spiny stellate (SpS) neurons in the reeler cortex with in vitro electrophysiology, optogenetics, and subcellular channelrhodopsin-2-assisted circuit mapping (sCRACM). Our results indicate that reeler SpS neurons receive direct but weakened input from the thalamus, with a dispersed spatial distribution along the somatodendritic arbor. These results further document subtle alterations in functional connectivity concomitant of absent layering in the reeler mutant. We suggest that intracortical amplification mechanisms compensate for this weakening in order to allow reliable sensory transmission to the mutant neocortex.
Collapse
Affiliation(s)
- Julien Guy
- Department of Neuroanatomy, Institute for Anatomy, University Medical Center, Georg-August-University, D-37075 Göttingen, Germany
| | - Alexandra Sachkova
- Department of Neuroanatomy, Institute for Anatomy, University Medical Center, Georg-August-University, D-37075 Göttingen, Germany
| | - Martin Möck
- Department of Neuroanatomy, Institute for Anatomy, University Medical Center, Georg-August-University, D-37075 Göttingen, Germany
| | - Mirko Witte
- Department of Neuroanatomy, Institute for Anatomy, University Medical Center, Georg-August-University, D-37075 Göttingen, Germany
| | - Robin J Wagener
- Department of Basic Neurosciences, University of Geneva, CH-1211, Geneva, Switzerland
| | - Jochen F Staiger
- Department of Neuroanatomy, Institute for Anatomy, University Medical Center, Georg-August-University, D-37075 Göttingen, Germany.,DFG Center for Nanoscale Microscopy & Molecular Physiology of the Brain (CNMPB), 37075 Göttingen, Germany
| |
Collapse
|
27
|
Luoni A, Gass P, Brambilla P, Ruggeri M, Riva MA, Inta D. Altered expression of schizophrenia-related genes in mice lacking mGlu5 receptors. Eur Arch Psychiatry Clin Neurosci 2018; 268:77-87. [PMID: 27581816 DOI: 10.1007/s00406-016-0728-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 08/22/2016] [Indexed: 12/15/2022]
Abstract
The evidence underlying the so-called glutamatergic hypothesis ranges from NMDA receptor hypofunction to an imbalance between excitatory and inhibitory circuits in specific brain structures. Among all glutamatergic system components, metabotropic receptors play a main role in regulating neuronal excitability and synaptic plasticity. Here, we investigated, using qRT-PCR and western blot, consequences in the hippocampus and prefrontal/frontal cortex (PFC/FC) of mice with a genetic deletion of the metabotropic glutamate receptor 5 (mGlu5), addressing key components of the GABAergic and glutamatergic systems. We found that mGlu5 knockout (KO) mice showed a significant reduction of reelin, GAD65, GAD67 and parvalbumin mRNA levels, which is specific for the PFC/FC, and that is paralleled by a significant reduction of protein levels in male KO mice. We next analyzed the main NMDA and AMPA receptor subunits, namely GluN1, GluN2A, GluN2B and GluA1, and we found that mGlu5 deletion determined a significant reduction of their mRNA levels, also within the hippocampus, with differences between the two genders. Our data suggest that neurochemical abnormalities impinging the glutamatergic and GABAergic systems may be responsible for the behavioral phenotype associated with mGlu5 KO animals and point to the close interaction of these molecular players for the development of neuropsychiatric disorders such as schizophrenia. These data could contribute to a better understanding of the involvement of mGlu5 alterations in the molecular imbalance between excitation and inhibition underlying the emergence of a schizophrenic-like phenotype and to understand the potential of mGlu5 modulators in reversing the deficits characterizing the schizophrenic pathology.
Collapse
Affiliation(s)
- Alessia Luoni
- Department of Pharmacological and Biomolecular Sciences, Center of Neuropharmacology, Università degli Studi di Milano, Milan, Italy
| | - Peter Gass
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health Mannheim, Medical Faculty Mannheim, University of Heidelberg, J 5, 68159, Mannheim, Germany
| | - Paolo Brambilla
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Mirella Ruggeri
- Section of Psychiatry, Department of Neurological, Biomedical and Movement Sciences, University of Verona, Verona, Italy
| | - Marco A Riva
- Department of Pharmacological and Biomolecular Sciences, Center of Neuropharmacology, Università degli Studi di Milano, Milan, Italy
| | - Dragos Inta
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health Mannheim, Medical Faculty Mannheim, University of Heidelberg, J 5, 68159, Mannheim, Germany. .,Department of Psychiatry (UPK), University of Basel, Wilhelm Klein-Str. 27, 4012, Basel, Switzerland.
| |
Collapse
|
28
|
Bosch C, Masachs N, Exposito-Alonso D, Martínez A, Teixeira CM, Fernaud I, Pujadas L, Ulloa F, Comella JX, DeFelipe J, Merchán-Pérez A, Soriano E. Reelin Regulates the Maturation of Dendritic Spines, Synaptogenesis and Glial Ensheathment of Newborn Granule Cells. Cereb Cortex 2018; 26:4282-4298. [PMID: 27624722 PMCID: PMC5066826 DOI: 10.1093/cercor/bhw216] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 06/20/2016] [Indexed: 02/02/2023] Open
Abstract
Significance Statement The extracellular protein Reelin has an important role in neurological diseases, including epilepsy, Alzheimer's disease and psychiatric diseases, targeting hippocampal circuits. Here we address the role of Reelin in the development of synaptic contacts in adult-generated granule cells (GCs), a neuronal population that is crucial for learning and memory and implicated in neurological and psychiatric diseases. We found that the Reelin pathway controls the shapes, sizes, and types of dendritic spines, the complexity of multisynaptic innervations and the degree of the perisynaptic astroglial ensheathment that controls synaptic homeostasis. These findings show a pivotal role of Reelin in GC synaptogenesis and provide a foundation for structural circuit alterations caused by Reelin deregulation that may occur in neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Carles Bosch
- Developmental Neurobiology and Regeneration Unit, Department of Cell Biology, Parc Científic de Barcelona and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain.,Institut de Recerca de l'Hospital Universitari de la Vall d'Hebron (VHIR), Barcelona 08023, Spain
| | - Nuria Masachs
- Developmental Neurobiology and Regeneration Unit, Department of Cell Biology, Parc Científic de Barcelona and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain
| | - David Exposito-Alonso
- Developmental Neurobiology and Regeneration Unit, Department of Cell Biology, Parc Científic de Barcelona and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain
| | - Albert Martínez
- Developmental Neurobiology and Regeneration Unit, Department of Cell Biology, Parc Científic de Barcelona and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain
| | - Cátia M Teixeira
- Developmental Neurobiology and Regeneration Unit, Department of Cell Biology, Parc Científic de Barcelona and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain
| | - Isabel Fernaud
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain.,Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Campus de Montegancedo, Madrid 28223, Spain.,Instituto Cajal (Consejo Superior de Investigaciones Científicas), Madrid 28002, Spain
| | - Lluís Pujadas
- Developmental Neurobiology and Regeneration Unit, Department of Cell Biology, Parc Científic de Barcelona and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain.,Institut de Recerca de l'Hospital Universitari de la Vall d'Hebron (VHIR), Barcelona 08023, Spain
| | - Fausto Ulloa
- Developmental Neurobiology and Regeneration Unit, Department of Cell Biology, Parc Científic de Barcelona and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain
| | - Joan X Comella
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain.,Institut de Recerca de l'Hospital Universitari de la Vall d'Hebron (VHIR), Barcelona 08023, Spain.,Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Javier DeFelipe
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain.,Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Campus de Montegancedo, Madrid 28223, Spain.,Instituto Cajal (Consejo Superior de Investigaciones Científicas), Madrid 28002, Spain
| | - Angel Merchán-Pérez
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain.,Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Campus de Montegancedo, Madrid 28223, Spain.,Departamento de Arquitectura y Tecnología de Sistemas Informáticos, Escuela Técnica Superior de Ingenieros Informáticos, Universidad Politécnica de Madrid, Madrid 28660, Spain
| | - Eduardo Soriano
- Developmental Neurobiology and Regeneration Unit, Department of Cell Biology, Parc Científic de Barcelona and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain.,Institut de Recerca de l'Hospital Universitari de la Vall d'Hebron (VHIR), Barcelona 08023, Spain.,Institució Catalana de Recerca i Estudis Avançats Academia, Barcelona 08010, Spain
| |
Collapse
|
29
|
Fu K, Miyamoto Y, Sumi K, Saika E, Muramatsu SI, Uno K, Nitta A. Overexpression of transmembrane protein 168 in the mouse nucleus accumbens induces anxiety and sensorimotor gating deficit. PLoS One 2017; 12:e0189006. [PMID: 29211814 PMCID: PMC5718521 DOI: 10.1371/journal.pone.0189006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 11/16/2017] [Indexed: 01/12/2023] Open
Abstract
Transmembrane protein 168 (TMEM168) comprises 697 amino acid residues, including some putative transmembrane domains. It is reported that TMEM168 controls methamphetamine (METH) dependence in the nucleus accumbens (NAc) of mice. Moreover, a strong link between METH dependence-induced adaptive changes in the brain and mood disorders has been evaluated. In the present study, we investigated the effects of accumbal TMEM168 in a battery of behavioral paradigms. The adeno-associated virus (AAV) Tmem168 vector was injected into the NAc of C57BL/6J mice (NAc-TMEM mice). Subsequently, the accumbal TMEM168 mRNA was increased approximately by seven-fold when compared with the NAc-Mock mice (controls). The NAc-TMEM mice reported no change in the locomotor activity, cognitive ability, social interaction, and depression-like behaviors; however, TMEM168 overexpression enhanced anxiety in the elevated-plus maze and light/dark box test. The increased anxiety was reversed by pretreatment with the antianxiety drug diazepam (0.3 mg/kg i.p.). Moreover, the NAc-TMEM mice exhibited decreased prepulse inhibition (PPI) in the startle response test, and the induced schizophrenia-like behavior was reversed by pretreatment with the antipsychotic drug risperidone (0.01 mg/kg i.p.). Furthermore, accumbal TMEM168 overexpression decreased the basal levels of extracellular GABA in the NAc and the high K+ (100 mM)-stimulated GABA elevation; however, the total contents of GABA in the NAc remained unaffected. These results suggest that the TMEM168-regulated GABAergic neuronal system in the NAc might become a novel target while studying the etiology of anxiety and sensorimotor gating deficits.
Collapse
Affiliation(s)
- Kequan Fu
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Toyama, Japan
| | - Yoshiaki Miyamoto
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Toyama, Japan
| | - Kazuyuki Sumi
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Toyama, Japan
| | - Eriko Saika
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Toyama, Japan
| | - Shin-ichi Muramatsu
- Division of Neurology, Department of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
- Center for Gene & Cell Therapy, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kyosuke Uno
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Toyama, Japan
| | - Atsumi Nitta
- Department of Pharmaceutical Therapy and Neuropharmacology, Faculty of Pharmaceutical Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Toyama, Japan
- * E-mail:
| |
Collapse
|
30
|
Bozzi Y, Provenzano G, Casarosa S. Neurobiological bases of autism-epilepsy comorbidity: a focus on excitation/inhibition imbalance. Eur J Neurosci 2017; 47:534-548. [PMID: 28452083 DOI: 10.1111/ejn.13595] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/18/2017] [Accepted: 04/21/2017] [Indexed: 12/13/2022]
Abstract
Autism spectrum disorders (ASD) and epilepsy are common neurological diseases of childhood, with an estimated incidence of approximately 0.5-1% of the worldwide population. Several genetic, neuroimaging and neuropathological studies clearly showed that both ASD and epilepsy have developmental origins and a substantial degree of heritability. Most importantly, ASD and epilepsy frequently coexist in the same individual, suggesting a common neurodevelopmental basis for these disorders. Genome-wide association studies recently allowed for the identification of a substantial number of genes involved in ASD and epilepsy, some of which are mutated in syndromes presenting both ASD and epilepsy clinical features. At the cellular level, both preclinical and clinical studies indicate that the different genetic causes of ASD and epilepsy may converge to perturb the excitation/inhibition (E/I) balance, due to the dysfunction of excitatory and inhibitory circuits in various brain regions. Metabolic and immune dysfunctions, as well as environmental causes also contribute to ASD pathogenesis. Thus, an E/I imbalance resulting from neurodevelopmental deficits of multiple origins might represent a common pathogenic mechanism for both diseases. Here, we will review the most significant studies supporting these hypotheses. A deeper understanding of the molecular and cellular determinants of autism-epilepsy comorbidity will pave the way to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Yuri Bozzi
- Neurodevelopmental Disorders Research Group, Centre for Mind/Brain Sciences, University of Trento, via Sommarive 9, 38123, Povo, Trento, Italy.,CNR Neuroscience Institute, Pisa, Italy
| | - Giovanni Provenzano
- Laboratory of Molecular Neuropathology, Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Simona Casarosa
- CNR Neuroscience Institute, Pisa, Italy.,Laboratory of Neural Development and Regeneration, Centre for Integrative Biology, University of Trento, Trento, Italy
| |
Collapse
|
31
|
Ampuero E, Jury N, Härtel S, Marzolo MP, van Zundert B. Interfering of the Reelin/ApoER2/PSD95 Signaling Axis Reactivates Dendritogenesis of Mature Hippocampal Neurons. J Cell Physiol 2016; 232:1187-1199. [PMID: 27653801 DOI: 10.1002/jcp.25605] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 09/12/2016] [Indexed: 12/21/2022]
Abstract
Reelin, an extracellular glycoprotein secreted in embryonic and adult brain, participates in neuronal migration and neuronal plasticity. Extensive evidence shows that reelin via activation of the ApoER2 and VLDLR receptors promotes dendrite and spine formation during early development. Further evidence suggests that reelin signaling is needed to maintain a stable architecture in mature neurons, but, direct evidence is lacking. During activity-dependent maturation of the neuronal circuitry, the synaptic protein PSD95 is inserted into the postsynaptic membrane to induce structural refinement and stability of spines and dendrites. Given that ApoER2 interacts with PSD95, we tested if reelin signaling interference in adult neurons reactivates the dendritic architecture. Unlike findings in developing cultures, the presently obtained in vitro and in vivo data show, for the first time, that reelin signaling interference robustly increase dendritogenesis and reduce spine density in mature hippocampal neurons. In particular, the expression of a mutant ApoER2 form (ApoER2-tailless), which is unable to interact with PSD95 and hence cannot transduce reelin signaling, resulted in robust dendritogenesis in mature hippocampal neurons in vitro. These results indicate that reelin/ApoER2/PSD95 signaling is important for neuronal structure maintenance in mature neurons. Mechanistically, obtained immunofluorescent data indicate that reelin signaling impairment reduced synaptic PSD95 levels, consequently leading to synaptic re-insertion of NR2B-NMDARs. Our findings underscore the importance of reelin in maintaining adult network stability and reveal a new mode for reactivating dendritogenesis in neurological disorders where dendritic arbor complexity is limited, such as in depression, Alzheimer's disease, and stroke. J. Cell. Physiol. 232: 1187-1199, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Estibaliz Ampuero
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago, Chile
| | - Nur Jury
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago, Chile
| | - Steffen Härtel
- SCIAN-Lab, CIMT, Bomedical Neuroscience Institute (BNI), ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - María-Paz Marzolo
- Laboratorio de Tráfico Intracelular y Señalización, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica, Santiago, Chile
| | - Brigitte van Zundert
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago, Chile
| |
Collapse
|
32
|
Devi U, Kumar V, Gupta PS, Dubey S, Singh M, Gautam S, Rawat JK, Roy S, Yadav RK, Ansari MN, Saeedan AS, Kaithwas G. Experimental Models for Autism Spectrum Disorder Follow-Up for the Validity. REVIEW JOURNAL OF AUTISM AND DEVELOPMENTAL DISORDERS 2016. [DOI: 10.1007/s40489-016-0088-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
33
|
Modeling psychiatric disorders: from genomic findings to cellular phenotypes. Mol Psychiatry 2016; 21:1167-79. [PMID: 27240529 PMCID: PMC4995546 DOI: 10.1038/mp.2016.89] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 04/20/2016] [Accepted: 04/21/2016] [Indexed: 12/15/2022]
Abstract
Major programs in psychiatric genetics have identified >150 risk loci for psychiatric disorders. These loci converge on a small number of functional pathways, which span conventional diagnostic criteria, suggesting a partly common biology underlying schizophrenia, autism and other psychiatric disorders. Nevertheless, the cellular phenotypes that capture the fundamental features of psychiatric disorders have not yet been determined. Recent advances in genetics and stem cell biology offer new prospects for cell-based modeling of psychiatric disorders. The advent of cell reprogramming and induced pluripotent stem cells (iPSC) provides an opportunity to translate genetic findings into patient-specific in vitro models. iPSC technology is less than a decade old but holds great promise for bridging the gaps between patients, genetics and biology. Despite many obvious advantages, iPSC studies still present multiple challenges. In this expert review, we critically review the challenges for modeling of psychiatric disorders, potential solutions and how iPSC technology can be used to develop an analytical framework for the evaluation and therapeutic manipulation of fundamental disease processes.
Collapse
|
34
|
Ranaivoson FM, von Daake S, Comoletti D. Structural Insights into Reelin Function: Present and Future. Front Cell Neurosci 2016; 10:137. [PMID: 27303268 PMCID: PMC4882317 DOI: 10.3389/fncel.2016.00137] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 05/10/2016] [Indexed: 12/20/2022] Open
Abstract
Reelin is a neuronal glycoprotein secreted by the Cajal-Retzius cells in marginal regions of the cerebral cortex and the hippocampus where it plays important roles in the control of neuronal migration and the formation of cellular layers during brain development. This 3461 residue-long protein is composed of a signal peptide, an F-spondin-like domain, eight Reelin repeats (RR1-8), and a positively charged sequence at the C-terminus. Biochemical data indicate that the central region of Reelin binds to the low-density lipoprotein receptors apolipoprotein E receptor 2 (ApoER2) and the very-low-density lipoprotein receptor (VLDLR), leading to the phosphorylation of the intracellular adaptor protein Dab1. After secretion, Reelin is rapidly degraded in three major fragments, but the functional significance of this degradation is poorly understood. Probably due to its large mass and the complexity of its architecture, the high-resolution, three-dimensional structure of Reelin has never been determined. However, the crystal structures of some of the RRs have been solved, providing important insights into their fold and the interaction with the ApoER2 receptor. This review discusses the current findings on the structure of Reelin and its binding to the ApoER2 and VLDLR receptors, and we discuss some areas where proteomics and structural biology can help understanding Reelin function in brain development and human health.
Collapse
Affiliation(s)
- Fanomezana M Ranaivoson
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers UniversityNew Brunswick, NJ, USA; Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers UniversityNew Brunswick, NJ, USA
| | - Sventja von Daake
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers UniversityNew Brunswick, NJ, USA; Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers UniversityNew Brunswick, NJ, USA
| | - Davide Comoletti
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers UniversityNew Brunswick, NJ, USA; Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers UniversityNew Brunswick, NJ, USA; Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers UniversityNew Brunswick, NJ, USA
| |
Collapse
|
35
|
Fraley ER, Burkett ZD, Day NF, Schwartz BA, Phelps PE, White SA. Mice with Dab1 or Vldlr insufficiency exhibit abnormal neonatal vocalization patterns. Sci Rep 2016; 6:25807. [PMID: 27184477 PMCID: PMC4868998 DOI: 10.1038/srep25807] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 04/22/2016] [Indexed: 11/27/2022] Open
Abstract
Genetic and epigenetic changes in components of the Reelin-signaling pathway (RELN, DAB1) are associated with autism spectrum disorder (ASD) risk. Social communication deficits are a key component of the ASD diagnostic criteria, but the underlying neurogenetic mechanisms remain unknown. Reln insufficient mice exhibit ASD-like behavioral phenotypes including altered neonatal vocalization patterns. Reelin affects multiple pathways including through the receptors, Very low-density lipoprotein receptor (Vldlr), Apolipoprotein receptor 2 (Apoer2), and intracellular signaling molecule Disabled-1 (Dab1). As Vldlr was previously implicated in avian vocalization, here we investigate vocalizations of neonatal mice with a reduction or absence of these components of the Reelin-signaling pathway. Mice with low or no Dab1 expression exhibited reduced calling rates, altered call-type usage, and differential vocal development trajectories. Mice lacking Vldlr expression also had altered call repertoires, and this effect was exacerbated by deficiency in Apoer2. Together with previous findings, these observations 1) solidify a role for Reelin in vocal communication of multiple species, 2) point to the canonical Reelin-signaling pathway as critical for development of normal neonatal calling patterns in mice, and 3) suggest that mutants in this pathway could be used as murine models for Reelin-associated vocal deficits in humans.
Collapse
Affiliation(s)
- E R Fraley
- Molecular, Cellular and Integrative Physiology Graduate Program, University of California, Los Angeles, USA.,Department of Integrative Biology and Physiology, University of California, Los Angeles, USA
| | - Z D Burkett
- Molecular, Cellular and Integrative Physiology Graduate Program, University of California, Los Angeles, USA.,Department of Integrative Biology and Physiology, University of California, Los Angeles, USA
| | - N F Day
- Department of Integrative Biology and Physiology, University of California, Los Angeles, USA
| | - B A Schwartz
- Undergraduate Interdepartmental Program in Neuroscience, University of California, Los Angeles, USA
| | - P E Phelps
- Molecular, Cellular and Integrative Physiology Graduate Program, University of California, Los Angeles, USA.,Department of Integrative Biology and Physiology, University of California, Los Angeles, USA
| | - S A White
- Molecular, Cellular and Integrative Physiology Graduate Program, University of California, Los Angeles, USA.,Department of Integrative Biology and Physiology, University of California, Los Angeles, USA
| |
Collapse
|
36
|
Kim KC, Gonzales EL, Lázaro MT, Choi CS, Bahn GH, Yoo HJ, Shin CY. Clinical and Neurobiological Relevance of Current Animal Models of Autism Spectrum Disorders. Biomol Ther (Seoul) 2016; 24:207-43. [PMID: 27133257 PMCID: PMC4859786 DOI: 10.4062/biomolther.2016.061] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 04/05/2016] [Indexed: 12/24/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social and communication impairments, as well as repetitive and restrictive behaviors. The phenotypic heterogeneity of ASD has made it overwhelmingly difficult to determine the exact etiology and pathophysiology underlying the core symptoms, which are often accompanied by comorbidities such as hyperactivity, seizures, and sensorimotor abnormalities. To our benefit, the advent of animal models has allowed us to assess and test diverse risk factors of ASD, both genetic and environmental, and measure their contribution to the manifestation of autistic symptoms. At a broader scale, rodent models have helped consolidate molecular pathways and unify the neurophysiological mechanisms underlying each one of the various etiologies. This approach will potentially enable the stratification of ASD into clinical, molecular, and neurophenotypic subgroups, further proving their translational utility. It is henceforth paramount to establish a common ground of mechanistic theories from complementing results in preclinical research. In this review, we cluster the ASD animal models into lesion and genetic models and further classify them based on the corresponding environmental, epigenetic and genetic factors. Finally, we summarize the symptoms and neuropathological highlights for each model and make critical comparisons that elucidate their clinical and neurobiological relevance.
Collapse
Affiliation(s)
- Ki Chan Kim
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Edson Luck Gonzales
- Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University, Seoul 05029, Republic of Korea.,School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - María T Lázaro
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chang Soon Choi
- Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University, Seoul 05029, Republic of Korea.,School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Geon Ho Bahn
- Department of Neuropsychiatry, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hee Jeong Yoo
- Department of Neuropsychiatry, Seoul National University Bungdang Hospital, Seongnam 13620, Republic of Korea
| | - Chan Young Shin
- Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University, Seoul 05029, Republic of Korea.,School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
37
|
Impairments in dendrite morphogenesis as etiology for neurodevelopmental disorders and implications for therapeutic treatments. Neurosci Biobehav Rev 2016; 68:946-978. [PMID: 27143622 DOI: 10.1016/j.neubiorev.2016.04.008] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 04/13/2016] [Accepted: 04/13/2016] [Indexed: 02/08/2023]
Abstract
Dendrite morphology is pivotal for neural circuitry functioning. While the causative relationship between small-scale dendrite morphological abnormalities (shape, density of dendritic spines) and neurodevelopmental disorders is well established, such relationship remains elusive for larger-scale dendrite morphological impairments (size, shape, branching pattern of dendritic trees). Here, we summarize published data on dendrite morphological irregularities in human patients and animal models for neurodevelopmental disorders, with focus on autism and schizophrenia. We next discuss high-risk genes for these disorders and their role in dendrite morphogenesis. We finally overview recent developments in therapeutic attempts and we discuss how they relate to dendrite morphology. We find that both autism and schizophrenia are accompanied by dendritic arbor morphological irregularities, and that majority of their high-risk genes regulate dendrite morphogenesis. Thus, we present a compelling argument that, along with smaller-scale morphological impairments in dendrites (spines and synapse), irregularities in larger-scale dendrite morphology (arbor shape, size) may be an important part of neurodevelopmental disorders' etiology. We suggest that this should not be ignored when developing future therapeutic treatments.
Collapse
|
38
|
Guidotti A, Grayson DR, Caruncho HJ. Epigenetic RELN Dysfunction in Schizophrenia and Related Neuropsychiatric Disorders. Front Cell Neurosci 2016; 10:89. [PMID: 27092053 PMCID: PMC4820443 DOI: 10.3389/fncel.2016.00089] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 03/21/2016] [Indexed: 01/02/2023] Open
Abstract
REELIN (RELN) is a large (420 kDa) glycoprotein that in adulthood is mostly synthesized in GABAergic neurons of corticolimbic structures. Upon secretion in the extracellular matrix (ECM), RELN binds to VLDL, APOE2, and α3β2 Integrin receptors located on dendritic shafts and spines of postsynaptic pyramidal neurons. Reduced levels of RELN expression in the adult brain induce cognitive impairment and dendritic spine density deficits. RELN supplementation recovers these deficits suggesting a trophic action for RELN in synaptic plasticity. We and others have shown that altered RELN expression in schizophrenia (SZ) and bipolar (BP) disorder patients is difficult to reconcile with classical Mendelian genetic disorders and it is instead plausible to associate these disorders with altered epigenetic homeostasis. Support for the contribution of altered epigenetic mechanisms in the down-regulation of RELN expression in corticolimbic structures of psychotic patients includes the concomitant increase of DNA-methyltransferases and the increased levels of the methyl donor S-adenosylmethionine (SAM). It is hypothesized that these conditions lead to RELN promoter hypermethylation and a reduction in RELN protein amounts in psychotic patients. The decreased synthesis and release of RELN from GABAergic corticolimbic neurons could serve as a model to elucidate the epigenetic pathophysiological mechanisms acting at pyramidal neuron dendrites that regulate synaptic plasticity and cognition in psychotic and non-psychotic subjects.
Collapse
Affiliation(s)
- Alessandro Guidotti
- Department of Psychiatry, The Psychiatric Institute, College of Medicine, University of Illinois at Chicago Chicago, IL, USA
| | - Dennis R Grayson
- Department of Psychiatry, The Psychiatric Institute, College of Medicine, University of Illinois at Chicago Chicago, IL, USA
| | - Hector J Caruncho
- College of Pharmacy and Nutrition, University of Saskatchewan Saskatoon, SK, Canada
| |
Collapse
|
39
|
Magliaro C, Cocito C, Bagatella S, Merighi A, Ahluwalia A, Lossi L. The number of Purkinje neurons and their topology in the cerebellar vermis of normal and reln haplodeficient mouse. Ann Anat 2016; 207:68-75. [PMID: 26996540 DOI: 10.1016/j.aanat.2016.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/02/2016] [Accepted: 02/25/2016] [Indexed: 12/14/2022]
Abstract
The Reeler heterozygous mice (reln(+/-)) are haplodeficient in the gene (reln) encoding for the reelin glycoprotein (RELN) and display reductions in brain/peripheral RELN similar to autistic or schizophrenic patients. Cytoarchitectonic alterations of the reln(+/-) brain may be subtle, and are difficult to demonstrate by current histological approaches. We analyzed the number and topological organization of the Purkinje neurons (PNs) in five vermal lobules - central (II-III), culmen (IV-V), tuber (VIIb), uvula (IX), and nodulus (X) - that process different types of afferent functional inputs in reln(+/+) and reln(+/-) adult mice (P60) of both sexes (n=24). Animals were crossed with L7GFP mice so that the GFP-tagged PNs could be directly identified in cryosections. Digital images from these sections were processed with different open source software for quantitative topological and statistical analyses. Diversity indices calculated were: maximum caliper, density, area of soma, dispersion along the XZ axis, and dispersion along the YZ axis. We demonstrate: i. reduction in density of PNs in reln(+/-) males (14.37%) and reln(+/-) females (17.73%) compared to reln(+/+) males; ii. that reln(+/-) males have larger PNs than other genotypes, and females (irrespective of the reln genetic background) have smaller PNs than reln(+/+) males; iii. PNs are more chaotically arranged along the YZ axis in reln(+/-) males than in reln(+/+) males and, except in central lobulus, reln(+/-) females. Therefore, image processing and statistics reveal previously unforeseen gender and genotype-related structural differences in cerebellum that may be clues for the definition of novel biomarkers in human psychiatric disorders.
Collapse
Affiliation(s)
- Chiara Magliaro
- University of Pisa, Research Center E. Piaggio, Faculty of Engineering, Pisa, Italy
| | - Carolina Cocito
- University of Turin, Department of Veterinary Sciences, Turin, Italy
| | - Stefano Bagatella
- University of Turin, Department of Veterinary Sciences, Turin, Italy
| | - Adalberto Merighi
- University of Turin, Department of Veterinary Sciences, Turin, Italy
| | - Arti Ahluwalia
- University of Pisa, Research Center E. Piaggio, Faculty of Engineering, Pisa, Italy
| | - Laura Lossi
- University of Turin, Department of Veterinary Sciences, Turin, Italy.
| |
Collapse
|
40
|
Negrón-Oyarzo I, Lara-Vásquez A, Palacios-García I, Fuentealba P, Aboitiz F. Schizophrenia and reelin: a model based on prenatal stress to study epigenetics, brain development and behavior. Biol Res 2016; 49:16. [PMID: 26968981 PMCID: PMC4787713 DOI: 10.1186/s40659-016-0076-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 02/22/2016] [Indexed: 11/20/2022] Open
Abstract
Schizophrenia is a severe psychiatric disorder that results in a significant disability for the patient. The disorder is characterized by impairment of the adaptive orchestration of actions, a cognitive function that is mainly dependent on the prefrontal cortex. This behavioral deficit, together with cellular and neurophysiological alterations in the prefrontal cortex, as well as reduced density of GABAergic cells and aberrant oscillatory activity, all indicate structural and functional deficits of the prefrontal cortex in schizophrenia. Among the several risk factors for the development of schizophrenia, stress during the prenatal period has been identified as crucial. Thus, it is proposed that prenatal stress induces neurodevelopmental alterations in the prefrontal cortex that are expressed as cognitive impairment observed in schizophrenia. However, the precise mechanisms that link prenatal stress with the impairment of prefrontal cortex function is largely unknown. Reelin is an extracellular matrix protein involved in the development of cortical neural connectivity at embryonic stages, and in synaptic plasticity at postnatal stages. Interestingly, down-regulation of reelin expression has been associated with epigenetic changes in the reelin gene of the prefrontal cortex of schizophrenic patients. We recently showed that, similar to schizophrenic patients, prenatal stress induces down-expression of reelin associated with the methylation of its promoter in the rodent prefrontal cortex. These alterations were paralleled with altered prefrontal cortex functional connectivity and impairment in prefrontal cortex-dependent behavioral tasks. Therefore, considering molecular, cellular, physiological and behavioral evidence, we propose a unifying framework that links prenatal stress and prefrontal malfunction through epigenetic alterations of the reelin gene.
Collapse
Affiliation(s)
- Ignacio Negrón-Oyarzo
- Departamento de Psiquiatría, Escuela de Medicina, and Centro Interdisciplinario de Neurociencia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ariel Lara-Vásquez
- Departamento de Psiquiatría, Escuela de Medicina, and Centro Interdisciplinario de Neurociencia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ismael Palacios-García
- Departamento de Psiquiatría, Escuela de Medicina, and Centro Interdisciplinario de Neurociencia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo Fuentealba
- Departamento de Psiquiatría, Escuela de Medicina, and Centro Interdisciplinario de Neurociencia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco Aboitiz
- Departamento de Psiquiatría, Escuela de Medicina, and Centro Interdisciplinario de Neurociencia, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
41
|
Caruncho HJ, Brymer K, Romay-Tallón R, Mitchell MA, Rivera-Baltanás T, Botterill J, Olivares JM, Kalynchuk LE. Reelin-Related Disturbances in Depression: Implications for Translational Studies. Front Cell Neurosci 2016; 10:48. [PMID: 26941609 PMCID: PMC4766281 DOI: 10.3389/fncel.2016.00048] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/11/2016] [Indexed: 02/02/2023] Open
Abstract
The finding that reelin expression is significantly decreased in mood and psychotic disorders, together with evidence that reelin can regulate key aspects of hippocampal plasticity in the adult brain, brought our research group and others to study the possible role of reelin in the pathogenesis of depression. This review describes recent progress on this topic using an animal model of depression that makes use of repeated corticosterone (CORT) injections. This methodology produces depression-like symptoms in both rats and mice that are reversed by antidepressant treatment. We have reported that CORT causes a decrease in the number of reelin-immunopositive cells in the dentate gyrus subgranular zone (SGZ), where adult hippocampal neurogenesis takes place; that down-regulation of the number of reelin-positive cells closely parallels the development of a depression-like phenotype during repeated CORT treatment; that reelin downregulation alters the co-expression of reelin with neuronal nitric oxide synthase (nNOS); that deficits in reelin might also create imbalances in glutamatergic and GABAergic circuits within the hippocampus and other limbic structures; and that co-treatment with antidepressant drugs prevents both reelin deficits and the development of a depression-like phenotype. We also observed alterations in the pattern of membrane protein clustering in peripheral lymphocytes in animals with low levels of reelin. Importantly, we found parallel changes in membrane protein clustering in depression patients, which differentiated two subpopulations of naïve depression patients that showed a different therapeutic response to antidepressant treatment. Here, we review these findings and develop the hypothesis that restoring reelin-related function could represent a novel approach for antidepressant therapies.
Collapse
Affiliation(s)
- Hector J Caruncho
- Neuroscience Cluster, College of Pharmacy and Nutrition, University of Saskatchewan Saskatoon, SK, Canada
| | - Kyle Brymer
- Department of Psychology, University of Saskatchewan Saskatoon, SK, Canada
| | | | - Milann A Mitchell
- Department of Psychology, University of Saskatchewan Saskatoon, SK, Canada
| | - Tania Rivera-Baltanás
- Department of Psychiatry, Alvaro Cunqueiro Hospital, Biomedical Research Institute of Vigo Galicia, Spain
| | - Justin Botterill
- Department of Psychology, University of Saskatchewan Saskatoon, SK, Canada
| | - Jose M Olivares
- Department of Psychiatry, Alvaro Cunqueiro Hospital, Biomedical Research Institute of Vigo Galicia, Spain
| | - Lisa E Kalynchuk
- Department of Medicine, University of Saskatchewan Saskatoon, SK, Canada
| |
Collapse
|
42
|
AKT-independent Reelin signaling requires interactions of heterotrimeric Go and Src. Biochem Biophys Res Commun 2015; 467:1063-9. [PMID: 26441085 DOI: 10.1016/j.bbrc.2015.09.167] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 09/30/2015] [Indexed: 11/23/2022]
Abstract
Reelin, a large secreted extracellular matrix glycoprotein, plays a key role in neuronal migration during cortical development and promotes neuronal maturation. The signaling pathway regulating neuronal maturation in the postnatal period are relatively less well understood. In this study, we demonstrated that a heterotrimeric G protein, Go, is a novel target of Reelin-induced signaling to promote neurite outgrowth. In primary hippocampal neurons of Reelin-deficient reeler mice, neurite outgrowth was significantly reduced and rescued upon addition of Reelin. Pertussis toxin (PTX) treatment or transfection with Gαo-siRNA suppressed Reelin-mediated neurite outgrowth in wild-type neurons. Additionally, Reelin treatment led to increased phosphorylation of AKT, GSK3β, and JNK, which were all effectively blocked by the PI3K inhibitor, LY294002. By comparison, PTX specifically blocked JNK activation, but not AKT and GSK3β. Immunoprecipitation assays disclosed that Reelin increases the active forms of both Src and Gαo and promotes their direct association. Notably, Dab1, a cytoplasmic adaptor molecule that mediates Reelin signaling, did not interact with Gαo. Neurite outgrowth by Reelin was induced via activating Src kinase, which directly stimulated Gαo, activity, leading to JNK activation. Based on the collective findings, we suggest that Reelin-dependent signaling mechanisms may be split into Src-AKT-dependent and Src-Go-dependent pathways. Our results additionally provide evidence that Reelin receptors cross-communicate with heterologous G protein-coupled receptors (GPCR) independently of the cognate ligands of GPCR.
Collapse
|
43
|
Müller I, Çalışkan G, Stork O. The GAD65 knock out mouse - a model for GABAergic processes in fear- and stress-induced psychopathology. GENES BRAIN AND BEHAVIOR 2015; 14:37-45. [PMID: 25470336 DOI: 10.1111/gbb.12188] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 11/11/2014] [Accepted: 11/13/2014] [Indexed: 12/16/2022]
Abstract
The γ-amino butyric acid (GABA) synthetic enzyme glutamic acid decarboxylase (GAD)65 is critically involved in the activity-dependent regulation of GABAergic inhibition in the central nervous system. It is also required for the maturation of the GABAergic system during adolescence, a phase that is critical for the development of several neuropsychiatric diseases. Mice bearing a null mutation of the GAD65 gene develop hyperexcitability of the amygdala and hippocampus, and a phenotype of increased anxiety and pathological fear memory reminiscent of posttraumatic stress disorder. Although genetic association of GAD65 in human has not yet been reported, these findings are in line with observations of reduced GABAergic function in these brain regions of anxiety disorder patients. The particular value of GAD65(-/-) mice thus lies in modeling the effects of reduced GABAergic function in the mature nervous system. The expression of GAD65 and a second GAD isozyme, GAD67, are differentially regulated in response to stress in limbic brain areas suggesting that by controlling GABAergic inhibition these enzymes determine the vulnerability for the development of pathological anxiety and other stress-induced phenotypes. In fact, we could recently show that GAD65 haplodeficiency, which results in delayed postnatal increase of GABA levels, provides resilience to juvenile-stress-induced anxiety to GAD65(+/-) mice thus foiling the increased fear and anxiety in homozygous GAD65(-/-) mice.
Collapse
Affiliation(s)
- Iris Müller
- Department of Genetics & Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | | | | |
Collapse
|
44
|
Fujihara K, Miwa H, Kakizaki T, Kaneko R, Mikuni M, Tanahira C, Tamamaki N, Yanagawa Y. Glutamate Decarboxylase 67 Deficiency in a Subset of GABAergic Neurons Induces Schizophrenia-Related Phenotypes. Neuropsychopharmacology 2015; 40:2475-86. [PMID: 25904362 PMCID: PMC4538341 DOI: 10.1038/npp.2015.117] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 04/05/2015] [Accepted: 04/07/2015] [Indexed: 02/08/2023]
Abstract
Decreased expression of the GABA synthetic enzyme glutamate decarboxylase 67 (GAD67) in a subset of GABAergic neurons, including parvalbumin (PV)-expressing neurons, has been observed in postmortem brain studies of schizophrenics and in animal models of schizophrenia. However, it is unclear whether and how the perturbations of GAD67-mediated GABA synthesis and signaling contribute to the pathogenesis of schizophrenia. To address this issue, we generated the mice lacking GAD67 primarily in PV neurons and characterized them with focus on schizophrenia-related parameters. We found that heterozygous mutant mice exhibited schizophrenia-related behavioral abnormalities such as deficits in prepulse inhibition, MK-801 sensitivity, and social memory. Furthermore, we observed reduced inhibitory synaptic transmission, altered properties of NMDA receptor-mediated synaptic responses in pyramidal neurons, and increased spine density in hippocampal CA1 apical dendrites, suggesting a possible link between GAD67 deficiency and disturbed glutamatergic excitatory synaptic functions in schizophrenia. Thus, our results indicate that the mice heterozygous for GAD67 deficiency primarily in PV neurons share several neurochemical and behavioral abnormalities with schizophrenia, offering a novel tool for addressing the underlying pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Kazuyuki Fujihara
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Japan,Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo, Japan,Department of Psychiatry and Human Behavior, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hideki Miwa
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Japan,Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo, Japan,Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan, Tel: +81 27 220 8044, Fax: +81 27 220 8046, E-mail:
| | - Toshikazu Kakizaki
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Japan,Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo, Japan
| | - Ryosuke Kaneko
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Japan,Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo, Japan,Institute of Experimental Animal Research, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Masahiko Mikuni
- Department of Psychiatry and Human Behavior, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Chiyoko Tanahira
- Department of Morphological Neural Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Nobuaki Tamamaki
- Department of Morphological Neural Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuchio Yanagawa
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Japan,Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo, Japan
| |
Collapse
|
45
|
Stouffer MA, Golden JA, Francis F. Neuronal migration disorders: Focus on the cytoskeleton and epilepsy. Neurobiol Dis 2015; 92:18-45. [PMID: 26299390 DOI: 10.1016/j.nbd.2015.08.003] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 08/05/2015] [Accepted: 08/12/2015] [Indexed: 01/28/2023] Open
Abstract
A wide spectrum of focal, regional, or diffuse structural brain abnormalities, collectively known as malformations of cortical development (MCDs), frequently manifest with intellectual disability (ID), epilepsy, and/or autistic spectrum disorder (ASD). As the acronym suggests, MCDs are perturbations of the normal architecture of the cerebral cortex and hippocampus. The pathogenesis of these disorders remains incompletely understood; however, one area that has provided important insights has been the study of neuronal migration. The amalgamation of human genetics and experimental studies in animal models has led to the recognition that common genetic causes of neurodevelopmental disorders, including many severe epilepsy syndromes, are due to mutations in genes regulating the migration of newly born post-mitotic neurons. Neuronal migration genes often, though not exclusively, code for proteins involved in the function of the cytoskeleton. Other cellular processes, such as cell division and axon/dendrite formation, which similarly depend on cytoskeletal functions, may also be affected. We focus here on how the susceptibility of the highly organized neocortex and hippocampus may be due to their laminar organization, which involves the tight regulation, both temporally and spatially, of gene expression, specialized progenitor cells, the migration of neurons over large distances and a birthdate-specific layering of neurons. Perturbations in neuronal migration result in abnormal lamination, neuronal differentiation defects, abnormal cellular morphology and circuit formation. Ultimately this results in disorganized excitatory and inhibitory activity leading to the symptoms observed in individuals with these disorders.
Collapse
Affiliation(s)
- Melissa A Stouffer
- INSERM UMRS 839, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie, Paris, France; Institut du Fer à Moulin, Paris, France
| | - Jeffrey A Golden
- Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Fiona Francis
- INSERM UMRS 839, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie, Paris, France; Institut du Fer à Moulin, Paris, France.
| |
Collapse
|
46
|
Ogden KK, Ozkan ED, Rumbaugh G. Prioritizing the development of mouse models for childhood brain disorders. Neuropharmacology 2015; 100:2-16. [PMID: 26231830 DOI: 10.1016/j.neuropharm.2015.07.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 07/18/2015] [Accepted: 07/22/2015] [Indexed: 12/20/2022]
Abstract
Mutations in hundreds of genes contribute to cognitive and behavioral dysfunction associated with developmental brain disorders (DBDs). Due to the sheer number of risk factors available for study combined with the cost of developing new animal models, it remains an open question how genes should be prioritized for in-depth neurobiological investigations. Recent reviews have argued that priority should be given to frequently mutated genes commonly found in sporadic DBD patients. Intrigued by this idea, we explored to what extent "high priority" risk factors have been studied in animals in an effort to assess their potential for generating valuable preclinical models capable of advancing the neurobiological understanding of DBDs. We found that in-depth whole animal studies are lacking for many high priority genes, with relatively few neurobiological studies performed in construct valid animal models aimed at understanding the pathological substrates associated with disease phenotypes. However, some high priority risk factors have been extensively studied in animal models and they have generated novel insights into DBD patho-neurobiology while also advancing early pre-clinical therapeutic treatment strategies. We suggest that prioritizing model development toward genes frequently mutated in non-specific DBD populations will accelerate the understanding of DBD patho-neurobiology and drive novel therapeutic strategies. This article is part of the Special Issue entitled 'Synaptopathy--from Biology to Therapy'.
Collapse
Affiliation(s)
- Kevin K Ogden
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Emin D Ozkan
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Gavin Rumbaugh
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| |
Collapse
|
47
|
Poser R, Dokter M, von Bohlen Und Halbach V, Berger SM, Busch R, Baldus M, Unsicker K, von Bohlen Und Halbach O. Impact of a deletion of the full-length and short isoform of p75NTR on cholinergic innervation and the population of postmitotic doublecortin positive cells in the dentate gyrus. Front Neuroanat 2015; 9:63. [PMID: 26074780 PMCID: PMC4444824 DOI: 10.3389/fnana.2015.00063] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 05/06/2015] [Indexed: 12/19/2022] Open
Abstract
Analyses of mice carrying a deletion of the pan-neurotrophin receptor p75NTR have allowed identifying p75NTR as an important structural regulator of the hippocampus. Most of the previous analyses were done using p75NTR (ExIII) knockout mice which still express the short isoform of p75NTR. To scrutinize the role of p75NTR in the hippocampus, we analyzed adult and aged p75NTR (ExIV) knockout mice, in which both, the short and the full-length isoform are deleted. Deletion of these isoforms induced morphological alterations in the adult dentate gyrus (DG), leading to an increase in the thickness of the molecular and granular layer. Based on these observations, we next determined the morphological substrates that might contribute to this phenotype. The cholinergic innervation of the molecular and granular layer of the DG was found to be significantly increased in the knockout mice. Furthermore, adult neurogenesis in the DG was found to be significantly altered with increased numbers of doublecortin (DCX) positive cells and reduced numbers of apoptotic cells in p75NTR (ExIV) knockout mice. However, cell proliferation as measured by phosphohiston H3 (PH3) positive cell numbers was not affected. These morphological alterations (number of DCX-positive cells and increased cholinergic fiber densities) as well as reduced cell death in the DG are likely to contribute to the observed thickening of the granular layer in p75NTR (ExIV) knockout mice. In addition, Sholl-analysis of DCX-positive neurons revealed a higher dendritic complexity and could thus be a possible morphological correlate for the increased thickness of the molecular layer in p75NTR deficient animals. Our data clearly demonstrate that deletion of both, the short and the full-length isoform of p75NTR affects DG morphology, due to alterations of the cholinergic system and an imbalance between neurogenesis and programmed cell death within the subgranular zone.
Collapse
Affiliation(s)
- Robert Poser
- Institute of Anatomy and Cell Biology, Universitätsmedizin Greifswald Greifswald, Germany
| | - Martin Dokter
- Institute of Anatomy and Cell Biology, Universitätsmedizin Greifswald Greifswald, Germany
| | | | - Stefan M Berger
- Department of Molecular Biology, Central Institute of Mental Health and Medical Faculty Mannheim, Heidelberg University Mannheim, Germany
| | - Ruben Busch
- Institute of Anatomy and Cell Biology, Universitätsmedizin Greifswald Greifswald, Germany
| | - Marian Baldus
- Institute of Anatomy and Cell Biology, Universitätsmedizin Greifswald Greifswald, Germany
| | - Klaus Unsicker
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, University of Freiburg Freiburg, Germany
| | | |
Collapse
|
48
|
Kim M, Jeong Y, Chang YC. Extracellular matrix protein reelin regulate dendritic spine density through CaMKIIβ. Neurosci Lett 2015; 599:97-101. [PMID: 26003447 DOI: 10.1016/j.neulet.2015.05.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/15/2015] [Accepted: 05/17/2015] [Indexed: 01/17/2023]
Abstract
Reelin, an extracellular matrix protein, plays an important role in brain development as well as synaptic plasticity. Interestingly, several recent studies have found that Reelin is important for dendritic spine formation in vitro and in vivo. However, the molecular mechanism by which Reelin regulates the dendritic spine density has not been studied well yet. In this study, we found that exogenous Reelin treatment was significantly increased the dendritic spine density in the primary hippocampal neurons. In addition, Reelin was increased the puncta numbers of synaptophysin and PSD-95. Moreover, we found that Reelin modulated the levels of CaMKIIβ, and CaMKIIβ siRNA prevented Reelin's effect on the dendritic spine density. Overall, our results are the first to demonstrate that CaMKIIβ might be required to enable Reelin to alter the dendritic spine density.
Collapse
Affiliation(s)
- Mihyun Kim
- Department of Physical Therapy, Inje University, Gimhae, 621-749, Republic of Korea
| | - Yun Jeong
- Department of Physical Therapy, Inje University, Gimhae, 621-749, Republic of Korea; Research Institute of Biomedical Engineering and Department of Medicine, Catholic University of Daegu School of Medicine, Daegu 705-718, Republic of Korea
| | - Young-Chae Chang
- Research Institute of Biomedical Engineering and Department of Medicine, Catholic University of Daegu School of Medicine, Daegu 705-718, Republic of Korea.
| |
Collapse
|
49
|
Palacios-García I, Lara-Vásquez A, Montiel JF, Díaz-Véliz GF, Sepúlveda H, Utreras E, Montecino M, González-Billault C, Aboitiz F. Prenatal stress down-regulates Reelin expression by methylation of its promoter and induces adult behavioral impairments in rats. PLoS One 2015; 10:e0117680. [PMID: 25679528 PMCID: PMC4332679 DOI: 10.1371/journal.pone.0117680] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 12/30/2014] [Indexed: 01/03/2023] Open
Abstract
Prenatal stress causes predisposition to cognitive and emotional disturbances and is a risk factor towards the development of neuropsychiatric conditions like depression, bipolar disorders and schizophrenia. The extracellular protein Reelin, expressed by Cajal-Retzius cells during cortical development, plays critical roles on cortical lamination and synaptic maturation, and its deregulation has been associated with maladaptive conditions. In the present study, we address the effect of prenatal restraint stress (PNS) upon Reelin expression and signaling in pregnant rats during the last 10 days of pregnancy. Animals from one group, including control and PNS exposed fetuses, were sacrificed and analyzed using immunohistochemical, biochemical, cell biology and molecular biology approaches. We scored changes in the expression of Reelin, its signaling pathway and in the methylation of its promoter. A second group included control and PNS exposed animals maintained until young adulthood for behavioral studies. Using the optical dissector, we show decreased numbers of Reelin-positive neurons in cortical layer I of PNS exposed animals. In addition, neurons from PNS exposed animals display decreased Reelin expression that is paralleled by changes in components of the Reelin-signaling cascade, both in vivo and in vitro. Furthermore, PNS induced changes in the DNA methylation levels of the Reelin promoter in culture and in histological samples. PNS adult rats display excessive spontaneous locomotor activity, high anxiety levels and problems of learning and memory consolidation. No significant visuo-spatial memory impairment was detected on the Morris water maze. These results highlight the effects of prenatal stress on the Cajal-Retzius neuronal population, and the persistence of behavioral consequences using this treatment in adults, thereby supporting a relevant role of PNS in the genesis of neuropsychiatric diseases. We also propose an in vitro model that can yield new insights on the molecular mechanisms behind the effects of prenatal stress.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antigens, Nuclear/genetics
- Antigens, Nuclear/metabolism
- Behavior, Animal
- Cell Adhesion Molecules, Neuronal/genetics
- Cell Adhesion Molecules, Neuronal/metabolism
- Cerebral Cortex/metabolism
- Cyclin-Dependent Kinase 5/metabolism
- DNA Methylation
- Disease Models, Animal
- Extracellular Matrix Proteins/genetics
- Extracellular Matrix Proteins/metabolism
- Female
- Maternal Exposure
- Mental Disorders/etiology
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Neurons/metabolism
- Pregnancy
- Prenatal Exposure Delayed Effects
- Promoter Regions, Genetic
- Rats
- Reelin Protein
- Serine Endopeptidases/genetics
- Serine Endopeptidases/metabolism
- Signal Transduction
- Stress, Physiological
- Stress, Psychological
Collapse
Affiliation(s)
- Ismael Palacios-García
- Departamento de Psiquiatría, Escuela de Medicina, and Centro Interdisciplinario de Neurociencia, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratory of Cell and Neuronal Dynamics (Cenedyn), Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| | - Ariel Lara-Vásquez
- Departamento de Psiquiatría, Escuela de Medicina, and Centro Interdisciplinario de Neurociencia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan F. Montiel
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad Diego Portales, Santiago, Chile
| | - Gabriela F. Díaz-Véliz
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Hugo Sepúlveda
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, and Fondo de Áreas Prioritarias (FONDAP) “Center for Genome Regulation”, Universidad Andrés Bello, Santiago, Chile
| | - Elías Utreras
- Laboratory of Cell and Neuronal Dynamics (Cenedyn), Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| | - Martín Montecino
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, and Fondo de Áreas Prioritarias (FONDAP) “Center for Genome Regulation”, Universidad Andrés Bello, Santiago, Chile
| | - Christian González-Billault
- Laboratory of Cell and Neuronal Dynamics (Cenedyn), Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| | - Francisco Aboitiz
- Departamento de Psiquiatría, Escuela de Medicina, and Centro Interdisciplinario de Neurociencia, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
50
|
Varela MJ, Lage S, Caruncho HJ, Cadavid MI, Loza MI, Brea J. Reelin influences the expression and function of dopamine D2 and serotonin 5-HT2A receptors: a comparative study. Neuroscience 2015; 290:165-74. [PMID: 25637489 DOI: 10.1016/j.neuroscience.2015.01.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 12/04/2014] [Accepted: 01/09/2015] [Indexed: 01/01/2023]
Abstract
Reelin is an extracellular matrix protein that plays a critical role in neuronal guidance during brain neurodevelopment and in synaptic plasticity in adults and has been associated with schizophrenia. Reelin mRNA and protein levels are reduced in various structures of post-mortem schizophrenic brains, in a similar way to those found in heterozygous reeler mice (HRM). Reelin is involved in protein expression in dendritic spines that are the major location where synaptic connections are established. Thus, we hypothesized that a genetic deficit in reelin would affect the expression and function of dopamine D2 and serotonin 5-HT2A receptors that are associated with the action of current antipsychotic drugs. In this study, D2 and 5-HT2A receptor expression and function were quantitated by using radioligand binding studies in the frontal cortex and striatum of HRM and wild-type mice (WTM). We observed increased expression (p<0.05) in striatum membranes and decreased expression (p<0.05) in frontal cortex membranes for both dopamine D2 and serotonin 5-HT2A receptors from HRM compared to WTM. Our results show parallel alterations of D2 and 5-HT2A receptors that are compatible with a possible hetero-oligomeric nature of these receptors. These changes are similar to changes described in schizophrenic patients and provide further support for the suitability of using HRM as a model for studying this disease and the effects of antipsychotic drugs.
Collapse
Affiliation(s)
- M J Varela
- BioFarma Research Group, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - S Lage
- BioFarma Research Group, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - H J Caruncho
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - M I Cadavid
- BioFarma Research Group, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - M I Loza
- BioFarma Research Group, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - J Brea
- BioFarma Research Group, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain.
| |
Collapse
|