1
|
Katsuyama Y, Hattori M. REELIN ameliorates Alzheimer's disease, but how? Neurosci Res 2024; 208:8-14. [PMID: 39094979 DOI: 10.1016/j.neures.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent type of dementia; therefore, there is a high demand for therapeutic medication targeting it. In this context, extensive research has been conducted to identify molecular targets for drugs. AD manifests through two primary pathological signs: senile plaques and neurofibrillary tangles, caused by accumulations of amyloid-beta (Aβ) and phosphorylated tau, respectively. Thus, studies concerning the molecular mechanisms underlying AD etiology have primarily focused on Aβ generation and tau phosphorylation, with the anticipation of uncovering a signaling pathway impacting these molecular processes. Over the past two decades, studies using not only experimental model systems but also examining human brains have accumulated fragmentary evidences suggesting that REELIN signaling pathway is deeply involved in AD. Here, we explore REELIN signaling pathway and its involvement in memory function within the brain and review studies investigating molecular connections between REELIN signaling pathway and AD etiology. This review aims to understand how the manipulation (activation) of this pathway might ameliorate the disease's etiology.
Collapse
Affiliation(s)
- Yu Katsuyama
- Division of Neuroanatomy, Department of Anatomy, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan.
| | - Mitsuharu Hattori
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi 467-8603, Japan
| |
Collapse
|
2
|
Singh AA, Yadav D, Khan F, Song M. Indole-3-Carbinol and Its Derivatives as Neuroprotective Modulators. Brain Sci 2024; 14:674. [PMID: 39061415 PMCID: PMC11274471 DOI: 10.3390/brainsci14070674] [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: 06/06/2024] [Revised: 06/26/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and its downstream tropomyosin receptor kinase B (TrkB) signaling pathway play pivotal roles in the resilience and action of antidepressant drugs, making them prominent targets in psychiatric research. Oxidative stress (OS) contributes to various neurological disorders, including neurodegenerative diseases, stroke, and mental illnesses, and exacerbates the aging process. The nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant responsive element (ARE) serves as the primary cellular defense mechanism against OS-induced brain damage. Thus, Nrf2 activation may confer endogenous neuroprotection against OS-related cellular damage; notably, the TrkB/phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway, stimulated by BDNF-dependent TrkB signaling, activates Nrf2 and promotes its nuclear translocation. However, insufficient neurotrophin support often leads to the downregulation of the TrkB signaling pathway in brain diseases. Thus, targeting TrkB activation and the Nrf2-ARE system is a promising therapeutic strategy for treating neurodegenerative diseases. Phytochemicals, including indole-3-carbinol (I3C) and its metabolite, diindolylmethane (DIM), exhibit neuroprotective effects through BDNF's mimetic activity; Akt phosphorylation is induced, and the antioxidant defense mechanism is activated by blocking the Nrf2-kelch-like ECH-associated protein 1 (Keap1) complex. This review emphasizes the therapeutic potential of I3C and its derivatives for concurrently activating neuronal defense mechanisms in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Alka Ashok Singh
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (A.A.S.); (D.Y.)
| | - Dhananjay Yadav
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (A.A.S.); (D.Y.)
| | - Fazlurrahman Khan
- Institute of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea;
- International Graduate Program of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea
| | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (A.A.S.); (D.Y.)
| |
Collapse
|
3
|
Chen M, Xu L, Wu Y, Soba P, Hu C. The organization and function of the Golgi apparatus in dendrite development and neurological disorders. Genes Dis 2023; 10:2425-2442. [PMID: 37554209 PMCID: PMC10404969 DOI: 10.1016/j.gendis.2022.11.009] [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: 07/04/2022] [Revised: 09/13/2022] [Accepted: 11/05/2022] [Indexed: 12/24/2022] Open
Abstract
Dendrites are specialized neuronal compartments that sense, integrate and transfer information in the neural network. Their development is tightly controlled and abnormal dendrite morphogenesis is strongly linked to neurological disorders. While dendritic morphology ranges from relatively simple to extremely complex for a specified neuron, either requires a functional secretory pathway to continually replenish proteins and lipids to meet dendritic growth demands. The Golgi apparatus occupies the center of the secretory pathway and is regulating posttranslational modifications, sorting, transport, and signal transduction, as well as acting as a non-centrosomal microtubule organization center. The neuronal Golgi apparatus shares common features with Golgi in other eukaryotic cell types but also forms distinct structures known as Golgi outposts that specifically localize in dendrites. However, the organization and function of Golgi in dendrite development and its impact on neurological disorders is just emerging and so far lacks a systematic summary. We describe the organization of the Golgi apparatus in neurons, review the current understanding of Golgi function in dendritic morphogenesis, and discuss the current challenges and future directions.
Collapse
Affiliation(s)
- Meilan Chen
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education Institute for Brain, Science and Rehabilitation, South China Normal University, Guangzhou, Guangdong 510631, China
- Department of Ophthalmology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510320, China
| | - Lu Xu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education Institute for Brain, Science and Rehabilitation, South China Normal University, Guangzhou, Guangdong 510631, China
| | - Yi Wu
- Department of Ophthalmology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510320, China
| | - Peter Soba
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Bonn 53115, Germany
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Chun Hu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education Institute for Brain, Science and Rehabilitation, South China Normal University, Guangzhou, Guangdong 510631, China
| |
Collapse
|
4
|
Antonucci F, Bozzi Y. Action of Botulinum Neurotoxin E Type in Experimental Epilepsies. Toxins (Basel) 2023; 15:550. [PMID: 37755976 PMCID: PMC10536604 DOI: 10.3390/toxins15090550] [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: 07/21/2023] [Revised: 08/09/2023] [Accepted: 08/26/2023] [Indexed: 09/28/2023] Open
Abstract
Botulinum neurotoxins (BoNTs) are zinc endopeptidases produced by the Clostridium genus of anerobic bacteria, largely known for their ability to cleave synaptic proteins, leading to neuromuscular paralysis. In the central nervous system, BoNTs are known to block the release of glutamate neurotransmitter, and for this reason, researchers explored the possible therapeutic action in disorders characterized by neuronal hyperactivity, such as epilepsy. Thus, using multidisciplinary approaches and models of experimental epilepsy, we investigated the pharmacological potential of BoNT/E serotype. In this review, written in memory of Prof. Matteo Caleo, a pioneer in these studies, we go back over the hypotheses and experimental approaches that led us to the conclusion that intrahippocampal administration of BoNT/E (i) displays anticonvulsant effects if prophylactically delivered in a model of acute generalized seizures; (ii) does not have any antiepileptogenic action after the induction of status epilepticus; (iii) reduces frequency of spontaneous seizures in a model of recurrent seizures if delivered during the chronic phase but in a transient manner. Indeed, the control on spontaneous seizures stops when BoNT/E effects are off (few days), thus limiting its pharmacological potential in humans.
Collapse
Affiliation(s)
- Flavia Antonucci
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, via Fratelli Cervi 93, 20054 Milan, Italy
- CNR Institute of Neuroscience, via Raoul Follereau 3, 20854 Vedano al Lambro, Italy
- CIMeC-Center for Mind/Brain Sciences, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, Italy
| | - Yuri Bozzi
- CIMeC-Center for Mind/Brain Sciences, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, Italy
- CNR Institute of Neuroscience, via Giuseppe Moruzzi 1, 56124 Pisa, Italy
| |
Collapse
|
5
|
Enck JR, Olson EC. Calcium Signaling during Cortical Apical Dendrite Initiation: A Role for Cajal-Retzius Neurons. Int J Mol Sci 2023; 24:12965. [PMID: 37629145 PMCID: PMC10455361 DOI: 10.3390/ijms241612965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
The apical dendrite of a cortical projection neuron (CPN) is generated from the leading process of the migrating neuron as the neuron completes migration. This transformation occurs in the cortical marginal zone (MZ), a layer that contains the Cajal-Retzius neurons and their axonal projections. Cajal-Retzius neurons (CRNs) are well known for their critical role in secreting Reelin, a glycoprotein that controls dendritogenesis and cell positioning in many regions of the developing brain. In this study, we examine the possibility that CRNs in the MZ may provide additional signals to arriving CPNs, that may promote the maturation of CPNs and thus shape the development of the cortex. We use whole embryonic hemisphere explants and multiphoton microscopy to confirm that CRNs display intracellular calcium transients of <1-min duration and high amplitude during early corticogenesis. In contrast, developing CPNs do not show high-amplitude calcium transients, but instead show a steady increase in intracellular calcium that begins at the time of dendritic initiation, when the leading process of the migrating CPN is encountering the MZ. The possible existence of CRN to CPN communication was revealed by the application of veratridine, a sodium channel activator, which has been shown to preferentially stimulate more mature cells in the MZ at an early developmental time. Surprisingly, veratridine application also triggers large calcium transients in CPNs, which can be partially blocked by a cocktail of antagonists that block glutamate and glycine receptor activation. These findings outline a model in which CRN spontaneous activity triggers the release of glutamate and glycine, neurotransmitters that can trigger intracellular calcium elevations in CPNs. These elevations begin as CPNs initiate dendritogenesis and continue as waves in the post-migratory cells. Moreover, we show that the pharmacological blockade of glutamatergic signaling disrupts migration, while forced expression of a bacterial voltage-gated calcium channel (CavMr) in the migrating neurons promotes dendritic growth and migration arrest. The identification of CRN to CPN signaling during early development provides insight into the observation that many autism-linked genes encode synaptic proteins that, paradoxically, are expressed in the developing cortex well before the appearance of synapses and the establishment of functional circuits.
Collapse
Affiliation(s)
| | - Eric C. Olson
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, 505 Irving Ave., Syracuse, NY 13210, USA;
| |
Collapse
|
6
|
Di Donato N, Guerrini R, Billington CJ, Barkovich AJ, Dinkel P, Freri E, Heide M, Gershon ES, Gertler TS, Hopkin RJ, Jacob S, Keedy SK, Kooshavar D, Lockhart PJ, Lohmann DR, Mahmoud IG, Parrini E, Schrock E, Severi G, Timms AE, Webster RI, Willis MJH, Zaki MS, Gleeson JG, Leventer RJ, Dobyns WB. Monoallelic and biallelic mutations in RELN underlie a graded series of neurodevelopmental disorders. Brain 2022; 145:3274-3287. [PMID: 35769015 PMCID: PMC9989350 DOI: 10.1093/brain/awac164] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/02/2022] [Accepted: 04/19/2022] [Indexed: 11/14/2022] Open
Abstract
Reelin, a large extracellular protein, plays several critical roles in brain development and function. It is encoded by RELN, first identified as the gene disrupted in the reeler mouse, a classic neurological mutant exhibiting ataxia, tremors and a 'reeling' gait. In humans, biallelic variants in RELN have been associated with a recessive lissencephaly variant with cerebellar hypoplasia, which matches well with the homozygous mouse mutant that has abnormal cortical structure, small hippocampi and severe cerebellar hypoplasia. Despite the large size of the gene, only 11 individuals with RELN-related lissencephaly with cerebellar hypoplasia from six families have previously been reported. Heterozygous carriers in these families were briefly reported as unaffected, although putative loss-of-function variants are practically absent in the population (probability of loss of function intolerance = 1). Here we present data on seven individuals from four families with biallelic and 13 individuals from seven families with monoallelic (heterozygous) variants of RELN and frontotemporal or temporal-predominant lissencephaly variant. Some individuals with monoallelic variants have moderate frontotemporal lissencephaly, but with normal cerebellar structure and intellectual disability with severe behavioural dysfunction. However, one adult had abnormal MRI with normal intelligence and neurological profile. Thorough literature analysis supports a causal role for monoallelic RELN variants in four seemingly distinct phenotypes including frontotemporal lissencephaly, epilepsy, autism and probably schizophrenia. Notably, we observed a significantly higher proportion of loss-of-function variants in the biallelic compared to the monoallelic cohort, where the variant spectrum included missense and splice-site variants. We assessed the impact of two canonical splice-site variants observed as biallelic or monoallelic variants in individuals with moderately affected or normal cerebellum and demonstrated exon skipping causing in-frame loss of 46 or 52 amino acids in the central RELN domain. Previously reported functional studies demonstrated severe reduction in overall RELN secretion caused by heterozygous missense variants p.Cys539Arg and p.Arg3207Cys associated with lissencephaly suggesting a dominant-negative effect. We conclude that biallelic variants resulting in complete absence of RELN expression are associated with a consistent and severe phenotype that includes cerebellar hypoplasia. However, reduced expression of RELN remains sufficient to maintain nearly normal cerebellar structure. Monoallelic variants are associated with incomplete penetrance and variable expressivity even within the same family and may have dominant-negative effects. Reduced RELN secretion in heterozygous individuals affects only cortical structure whereas the cerebellum remains intact. Our data expand the spectrum of RELN-related neurodevelopmental disorders ranging from lethal brain malformations to adult phenotypes with normal brain imaging.
Collapse
Affiliation(s)
- Nataliya Di Donato
- Institute for Clinical Genetics, University Hospital, TU Dresden, 01307 Dresden, Germany
| | - Renzo Guerrini
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Meyer Children's Hospital, University of Florence, 50139 Florence, Italy
| | - Charles J Billington
- Department of Pediatrics, Division of Genetics and Metabolism, University of Minnesota, Minneapolis, MN 55454, USA
| | - A James Barkovich
- Departments of Radiology and Biomedical Imaging, Neurology, Pediatrics, and Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Philine Dinkel
- Institute for Clinical Genetics, University Hospital, TU Dresden, 01307 Dresden, Germany
| | - Elena Freri
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Michael Heide
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
- German Primate Center, Leibniz Institute for Primate Research, 37077 Goettingen, Germany
| | - Elliot S Gershon
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, IL 60637, USA
| | - Tracy S Gertler
- Division of Neurology, Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Robert J Hopkin
- Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Department of Pediatrics, Division of Human Genetics, Cincinnati, OH 45229, USA
| | - Suma Jacob
- Department of Psychiatry, University of Minnesota, Minneapolis, MN 55454, USA
| | - Sarah K Keedy
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, IL 60637, USA
| | - Daniz Kooshavar
- Bruce Lefory Centre, Murdoch Children's Research Institute and University of Melbourne Department of Pediatrics, Melbourne 3052, Australia
| | - Paul J Lockhart
- Bruce Lefory Centre, Murdoch Children's Research Institute and University of Melbourne Department of Pediatrics, Melbourne 3052, Australia
| | - Dietmar R Lohmann
- Institut fur Humangenetik, Universitatsklinikum Essen, 45147 Essen, Germany
| | - Iman G Mahmoud
- Pediatric Neurology Department, Cairo University Children's Hospital, Cairo, Egypt
| | - Elena Parrini
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Meyer Children's Hospital, University of Florence, 50139 Florence, Italy
| | - Evelin Schrock
- Institute for Clinical Genetics, University Hospital, TU Dresden, 01307 Dresden, Germany
| | - Giulia Severi
- Medical Genetics Unit, S. Orsola-Malpighi Hospital, 40138 Bologna, Italy
| | - Andrew E Timms
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Richard I Webster
- T. Y. Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, Sydney 2145, Australia
| | - Mary J H Willis
- Uniformed Services University School of Medicine and Naval Medical Center, Department of Pediatrics, San Diego, CA 92134, USA
| | - Maha S Zaki
- Pediatric Neurology Department, Cairo University Children's Hospital, Cairo, Egypt
- Clinical Genetics Department, Human Genetics and Genome Research Division, National Research Centre, Cairo Governorate 12622, Egypt
| | - Joseph G Gleeson
- Department of Neurosciences, Howard Hughes Medical Institute, University of California, San Diego, La Jolla, CA 92093, USA
| | - Richard J Leventer
- Department of Neurology, Royal Children's Hospital, Murdoch Children's Research Institute and University of Melbourne Department of Pediatrics, Melbourne 3052, Australia
| | - William B Dobyns
- Department of Pediatrics, Division of Genetics and Metabolism, University of Minnesota, Minneapolis, MN 55454, USA
| |
Collapse
|
7
|
Batzel GO, Moreno BK, Lopez LS, Nguyen CK, Livingston BT, Joester D, Lyons DC. Proteomic and Transcriptomic Analyses in the Slipper Snail Crepidula
fornicata Uncover Shell Matrix Genes Expressed During Adult and Larval Biomineralization. Integr Org Biol 2022; 4:obac023. [PMID: 35968217 PMCID: PMC9365450 DOI: 10.1093/iob/obac023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/18/2022] [Indexed: 11/12/2022] Open
Abstract
The gastropod shell is a composite composed of minerals and shell matrix proteins (SMPs). SMPs have been identified by proteomics in many molluscs, but few have been studied in detail. Open questions include (1) what gene regulatory networks regulate SMP expression, (2) what roles individual SMPs play in biomineralization, and (3) how the complement of SMPs changes over development. These questions are best addressed in a species in which gene perturbation studies are available; one such species is the slipper snail, Crepidula fornicata. Here, SEM and pXRD analysis demonstrated that the adult shell of C. fornicata exhibits crossed lamellar microstructure and is composed of aragonite. Using high-throughput proteomics we identified 185 SMPs occluded within the adult shell. Over half of the proteins in the shell proteome have known biomineralization domains, while at least 10% have no homologs in public databases. Differential gene expression analysis identified 20 SMP genes that are up-regulated in the shell-producing mantle tissue. Over half of these 20 SMPs are expressed during development with two, CfSMP1 and CfSMP2, expressed exclusively in the shell gland. Together, the description of the shell microstructure and a list of SMPs now sets the stage for studying the consequences of SMP gene knockdowns in molluscs.
Collapse
Affiliation(s)
- G O Batzel
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography , UCSD, La Jolla, CA 92037, USA
| | - B K Moreno
- Department of Materials Science and Engineering, Northwestern University , Evanston, IL 60208, USA
| | - L S Lopez
- Department of Biological Sciences, California State University , Long Beach, CA 90802, USA
| | - C K Nguyen
- Department of Biological Sciences, California State University , Long Beach, CA 90802, USA
| | - B T Livingston
- Department of Biological Sciences, California State University , Long Beach, CA 90802, USA
| | - D Joester
- Department of Materials Science and Engineering, Northwestern University , Evanston, IL 60208, USA
| | - D C Lyons
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography , UCSD, La Jolla, CA 92037, USA
| |
Collapse
|
8
|
Schaaf ZA, Tat L, Cannizzaro N, Panoutsopoulos AA, Green R, Rülicke T, Hippenmeyer S, Zarbalis KS. WDFY3 mutation alters laminar position and morphology of cortical neurons. Mol Autism 2022; 13:27. [PMID: 35733184 PMCID: PMC9219247 DOI: 10.1186/s13229-022-00508-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 06/09/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Proper cerebral cortical development depends on the tightly orchestrated migration of newly born neurons from the inner ventricular and subventricular zones to the outer cortical plate. Any disturbance in this process during prenatal stages may lead to neuronal migration disorders (NMDs), which can vary in extent from focal to global. Furthermore, NMDs show a substantial comorbidity with other neurodevelopmental disorders, notably autism spectrum disorders (ASDs). Our previous work demonstrated focal neuronal migration defects in mice carrying loss-of-function alleles of the recognized autism risk gene WDFY3. However, the cellular origins of these defects in Wdfy3 mutant mice remain elusive and uncovering it will provide critical insight into WDFY3-dependent disease pathology. METHODS Here, in an effort to untangle the origins of NMDs in Wdfy3lacZ mice, we employed mosaic analysis with double markers (MADM). MADM technology enabled us to genetically distinctly track and phenotypically analyze mutant and wild-type cells concomitantly in vivo using immunofluorescent techniques. RESULTS We revealed a cell autonomous requirement of WDFY3 for accurate laminar positioning of cortical projection neurons and elimination of mispositioned cells during early postnatal life. In addition, we identified significant deviations in dendritic arborization, as well as synaptic density and morphology between wild type, heterozygous, and homozygous Wdfy3 mutant neurons in Wdfy3-MADM reporter mice at postnatal stages. LIMITATIONS While Wdfy3 mutant mice have provided valuable insight into prenatal aspects of ASD pathology that remain inaccessible to investigation in humans, like most animal models, they do not a perfectly replicate all aspects of human ASD biology. The lack of human data makes it indeterminate whether morphological deviations described here apply to ASD patients or some of the other neurodevelopmental conditions associated with WDFY3 mutation. CONCLUSIONS Our genetic approach revealed several cell autonomous requirements of WDFY3 in neuronal development that could underlie the pathogenic mechanisms of WDFY3-related neurodevelopmental conditions. The results are also consistent with findings in other ASD animal models and patients and suggest an important role for WDFY3 in regulating neuronal function and interconnectivity in postnatal life.
Collapse
Affiliation(s)
- Zachary A Schaaf
- University of California at Davis, Department of Pathology and Laboratory Medicine, Sacramento, CA, 95817, USA
- Shriners Hospitals for Children Northern California, Sacramento, CA, 95817, USA
| | - Lyvin Tat
- University of California at Davis, Department of Pathology and Laboratory Medicine, Sacramento, CA, 95817, USA
| | - Noemi Cannizzaro
- University of California at Davis, Department of Pathology and Laboratory Medicine, Sacramento, CA, 95817, USA
| | - Alexios A Panoutsopoulos
- Shriners Hospitals for Children Northern California, Sacramento, CA, 95817, USA
- University of California at Davis, Department of Physiology and Membrane Biology, Sacramento, CA, 95817, USA
| | - Ralph Green
- University of California at Davis, Department of Pathology and Laboratory Medicine, Sacramento, CA, 95817, USA
| | - Thomas Rülicke
- Department of Biomedical Sciences, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Simon Hippenmeyer
- Institute of Science and Technology Austria, Am Campus 1, 3400, Klosterneuburg, Austria
| | - Konstantinos S Zarbalis
- University of California at Davis, Department of Pathology and Laboratory Medicine, Sacramento, CA, 95817, USA.
- Shriners Hospitals for Children Northern California, Sacramento, CA, 95817, USA.
- UC Davis MIND Institute, Sacramento, CA, 95817, USA.
| |
Collapse
|
9
|
Hu L, Zhang L. Adult neural stem cells and schizophrenia. World J Stem Cells 2022; 14:219-230. [PMID: 35432739 PMCID: PMC8968214 DOI: 10.4252/wjsc.v14.i3.219] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/18/2021] [Accepted: 03/07/2022] [Indexed: 02/06/2023] Open
Abstract
Schizophrenia (SCZ) is a devastating and complicated mental disorder accompanied by variable positive and negative symptoms and cognitive deficits. Although many genetic risk factors have been identified, SCZ is also considered as a neurodevelopmental disorder. Elucidation of the pathogenesis and the development of treatment is challenging because complex interactions occur between these genetic risk factors and environment in essential neurodevelopmental processes. Adult neural stem cells share a lot of similarities with embryonic neural stem cells and provide a promising model for studying neuronal development in adulthood. These adult neural stem cells also play an important role in cognitive functions including temporal and spatial memory encoding and context discrimination, which have been shown to be closely linked with many psychiatric disorders, such as SCZ. Here in this review, we focus on the SCZ risk genes and the key components in related signaling pathways in adult hippocampal neural stem cells and summarize their roles in adult neurogenesis and animal behaviors. We hope that this would be helpful for the understanding of the contribution of dysregulated adult neural stem cells in the pathogenesis of SCZ and for the identification of potential therapeutic targets, which could facilitate the development of novel medication and treatment.
Collapse
Affiliation(s)
- Ling Hu
- Department of Laboratory Animal Science and Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Lei Zhang
- Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center) and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai 200092, China
| |
Collapse
|
10
|
Dab1-deficient deep layer neurons prevent Dab1-deficient superficial layer neurons from entering the cortical plate. Neurosci Res 2022; 180:23-35. [DOI: 10.1016/j.neures.2022.03.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 02/06/2023]
|
11
|
Patel PA, Hegert JV, Cristian I, Kerr A, LaConte LEW, Fox MA, Srivastava S, Mukherjee K. Complete loss of the X-linked gene CASK causes severe cerebellar degeneration. J Med Genet 2022; 59:1044-1057. [PMID: 35149592 DOI: 10.1136/jmedgenet-2021-108115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 01/13/2022] [Indexed: 01/19/2023]
Abstract
BACKGROUND Heterozygous loss of X-linked genes like CASK and MeCP2 (Rett syndrome) causes developmental delay in girls, while in boys, loss of the only allele of these genes leads to epileptic encephalopathy. The mechanism for these disorders remains unknown. CASK-linked cerebellar hypoplasia is presumed to result from defects in Tbr1-reelin-mediated neuronal migration. METHOD Here we report clinical and histopathological analyses of a deceased 2-month-old boy with a CASK-null mutation. We next generated a mouse line where CASK is completely deleted (hemizygous and homozygous) from postmigratory neurons in the cerebellum. RESULT The CASK-null human brain was smaller in size but exhibited normal lamination without defective neuronal differentiation, migration or axonal guidance. The hypoplastic cerebellum instead displayed astrogliosis and microgliosis, which are markers for neuronal loss. We therefore hypothesise that CASK loss-induced cerebellar hypoplasia is the result of early neurodegeneration. Data from the murine model confirmed that in CASK loss, a small cerebellum results from postdevelopmental degeneration of cerebellar granule neurons. Furthermore, at least in the cerebellum, functional loss from CASK deletion is secondary to degeneration of granule cells and not due to an acute molecular functional loss of CASK. Intriguingly, female mice with heterozygous deletion of CASK in the cerebellum do not display neurodegeneration. CONCLUSION We suggest that X-linked neurodevelopmental disorders like CASK mutation and Rett syndrome are pathologically neurodegenerative; random X-chromosome inactivation in heterozygous mutant girls, however, results in 50% of cells expressing the functional gene, resulting in a non-progressive pathology, whereas complete loss of the only allele in boys leads to unconstrained degeneration and encephalopathy.
Collapse
Affiliation(s)
- Paras A Patel
- Fralin Biomedical Research Institute at VTC, Roanoke, Virginia, USA
| | - Julia V Hegert
- Department of Pathology, Orlando Health, Orlando, Florida, USA
| | | | - Alicia Kerr
- Fralin Biomedical Research Institute at VTC, Roanoke, Virginia, USA
| | | | - Michael A Fox
- Fralin Biomedical Research Institute at VTC, Roanoke, Virginia, USA.,School of Neuroscience, Blacksburg, Virginia, USA
| | - Sarika Srivastava
- Fralin Biomedical Research Institute at VTC, Roanoke, Virginia, USA.,Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
| | - Konark Mukherjee
- Fralin Biomedical Research Institute at VTC, Roanoke, Virginia, USA .,Department of Psychiatry, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
| |
Collapse
|
12
|
Mohamadian M, Rastegar M, Pasamanesh N, Ghadiri A, Ghandil P, Naseri M. Clinical and Molecular Spectrum of Muscular Dystrophies (MDs) with Intellectual Disability (ID): a Comprehensive Overview. J Mol Neurosci 2022; 72:9-23. [PMID: 34727324 DOI: 10.1007/s12031-021-01933-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 10/18/2021] [Indexed: 12/22/2022]
Abstract
Muscular dystrophies encompass a wide and heterogeneous subset of hereditary myopathies that manifest by the structural or functional abnormalities in the skeletal muscle. Some pathogenic mutations induce a dysfunction or loss of proteins that are critical for the stability of muscle cells, leading to progressive muscle degradation and weakening. Several studies have well-established cognitive deficits in muscular dystrophies which are mainly due to the disruption of brain-specific expression of affected muscle proteins. We provide a comprehensive overview of the types of muscular dystrophies that are accompanied by intellectual disability by detailed consulting of the main libraries. The current paper focuses on the clinical and molecular evidence about Duchenne, congenital, limb-girdle, and facioscapulohumeral muscular dystrophies as well as myotonic dystrophies. Because these syndromes impose a heavy burden of psychological and financial problems on patients, their families, and the health care community, a thorough examination is necessary to perform timely psychological and medical interventions and thus improve the quality of life.
Collapse
Affiliation(s)
- Malihe Mohamadian
- Cancer Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran, 616476515.
| | - Mandana Rastegar
- Department of Molecular Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Negin Pasamanesh
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ata Ghadiri
- Department of Immunology, Medical School, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Pegah Ghandil
- Diabetes Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Medical Genetics, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohsen Naseri
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
13
|
Orcinha C, Kilias A, Paschen E, Follo M, Haas CA. Reelin Is Required for Maintenance of Granule Cell Lamination in the Healthy and Epileptic Hippocampus. Front Mol Neurosci 2021; 14:730811. [PMID: 34483838 PMCID: PMC8414139 DOI: 10.3389/fnmol.2021.730811] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 07/26/2021] [Indexed: 11/21/2022] Open
Abstract
One characteristic feature of mesial temporal lobe epilepsy is granule cell dispersion (GCD), a pathological widening of the granule cell layer in the dentate gyrus. The loss of the extracellular matrix protein Reelin, an important positional cue for neurons, correlates with GCD formation in MTLE patients and in rodent epilepsy models. Here, we used organotypic hippocampal slice cultures (OHSC) from transgenic mice expressing enhanced green fluorescent protein (eGFP) in differentiated granule cells (GCs) to monitor GCD formation dynamically by live cell video microscopy and to investigate the role of Reelin in this process. We present evidence that following treatment with the glutamate receptor agonist kainate (KA), eGFP-positive GCs migrated mainly toward the hilar region. In the hilus, Reelin-producing neurons were rapidly lost following KA treatment as shown in a detailed time series. Addition of recombinant Reelin fragments to the medium effectively prevented the KA-triggered movement of eGFP-positive GCs. Placement of Reelin-coated beads into the hilus of KA-treated cultures stopped the migration of GCs in a distance-dependent manner. In addition, quantitative Western blot analysis revealed that KA treatment affects the Reelin signal transduction pathway by increasing intracellular adaptor protein Disabled-1 synthesis and reducing the phosphorylation of cofilin, a downstream target of the Reelin pathway. Both events were normalized by addition of recombinant Reelin fragments. Finally, following neutralization of Reelin in healthy OHSC by incubation with the function-blocking CR-50 Reelin antibody, GCs started to migrate without any direction preference. Together, our findings demonstrate that normotopic position of Reelin is essential for the maintenance of GC lamination in the dentate gyrus and that GCD is the result of a local Reelin deficiency.
Collapse
Affiliation(s)
- Catarina Orcinha
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Antje Kilias
- Biomicrotechnology, Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg im Breisgau, Germany
| | - Enya Paschen
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany.,Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
| | - Marie Follo
- Lighthouse Core Facility, Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Carola A Haas
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany.,Center for Basics in NeuroModulation, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
14
|
Maternal Ethanol Exposure Acutely Elevates Src Family Kinase Activity in the Fetal Cortex. Mol Neurobiol 2021; 58:5210-5223. [PMID: 34272687 PMCID: PMC8497457 DOI: 10.1007/s12035-021-02467-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/20/2021] [Indexed: 11/24/2022]
Abstract
Fetal alcohol syndrome (FAS) is characterized by disrupted fetal brain development and postnatal cognitive impairment. The targets of alcohol are diverse, and it is not clear whether there are common underlying molecular mechanisms producing these disruptions. Prior work established that acute ethanol exposure causes a transient increase in tyrosine phosphorylation of multiple proteins in cultured embryonic cortical cells. In this study, we show that a similar tyrosine phosphorylation transient occurs in the fetal brain after maternal dosing with ethanol. Using phospho-specific antibodies and immunohistochemistry, we mapped regions of highest tyrosine phosphorylation in the fetal cerebral cortex and found that areas of dendritic and axonal growth showed elevated tyrosine phosphorylation 10 min after maternal ethanol exposure. These were also areas of Src expression and Src family kinase (SFK) activation loop phosphorylation (pY416) expression. Importantly, maternal pretreatment with the SFK inhibitor dasatinib completely prevents both the pY416 increase and the tyrosine phosphorylation response. The phosphorylation response was observed in the perisomatic region and neurites of immature migrating and differentiating primary neurons. Importantly, the initial phosphotyrosine transient (~ 30 min) targets both Src and Dab1, two critical elements in Reelin signaling, a pathway required for normal cortical development. This initial phosphorylation response is followed by sustained reduction in Ser3 phosphorylation of n-cofilin, a critical actin severing protein and an identified downstream effector of Reelin signaling. This biochemical disruption is associated with sustained reduction of F-actin content and disrupted Golgi apparatus morphology in developing cortical neurons. The finding outlines a model in which the initial activation of SFKs by ethanol has the potential to disrupt multiple developmentally important signaling systems for several hours after maternal exposure.
Collapse
|
15
|
Alzate-Correa D, Mei-Ling Liu J, Jones M, Silva TM, Alves MJ, Burke E, Zuñiga J, Kaya B, Zaza G, Aslan MT, Blackburn J, Shimada MY, Fernandes-Junior SA, Baer LA, Stanford KI, Kempton A, Smith S, Szujewski CC, Silbaugh A, Viemari JC, Takakura AC, Garcia AJ, Moreira TS, Czeisler CM, Otero JJ. Neonatal apneic phenotype in a murine congenital central hypoventilation syndrome model is induced through non-cell autonomous developmental mechanisms. Brain Pathol 2020; 31:84-102. [PMID: 32654284 PMCID: PMC7881415 DOI: 10.1111/bpa.12877] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 05/10/2020] [Accepted: 06/11/2020] [Indexed: 12/19/2022] Open
Abstract
Congenital central hypoventilation syndrome (CCHS) represents a rare genetic disorder usually caused by mutations in the homeodomain transcription factor PHOX2B. Some CCHS patients suffer mainly from deficiencies in CO2 and/or O2 respiratory chemoreflex, whereas other patients present with full apnea shortly after birth. Our goal was to identify the neuropathological mechanisms of apneic presentations in CCHS. In the developing murine neuroepithelium, Phox2b is expressed in three discrete progenitor domains across the dorsal-ventral axis, with different domains responsible for producing unique autonomic or visceral motor neurons. Restricting the expression of mutant Phox2b to the ventral visceral motor neuron domain induces marked newborn apnea together with a significant loss of visceral motor neurons, RTN ablation, and preBötzinger complex dysfunction. This finding suggests that the observed apnea develops through non-cell autonomous developmental mechanisms. Mutant Phox2b expression in dorsal rhombencephalic neurons did not generate significant respiratory dysfunction, but did result in subtle metabolic thermoregulatory deficiencies. We confirm the expression of a novel murine Phox2b splice variant which shares exons 1 and 2 with the more widely studied Phox2b splice variant, but which differs in exon 3 where most CCHS mutations occur. We also show that mutant Phox2b expression in the visceral motor neuron progenitor domain increases cell proliferation at the expense of visceral motor neuron development. We propose that visceral motor neurons may function as organizers of brainstem respiratory neuron development, and that disruptions in their development result in secondary/non-cell autonomous maldevelopment of key brainstem respiratory neurons.
Collapse
Affiliation(s)
- Diego Alzate-Correa
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Jillian Mei-Ling Liu
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Mikayla Jones
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Talita M Silva
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Michele Joana Alves
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Elizabeth Burke
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Jessica Zuñiga
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Behiye Kaya
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Giuliana Zaza
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Mehmet Tahir Aslan
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Jessica Blackburn
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Marina Y Shimada
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Silvio A Fernandes-Junior
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Lisa A Baer
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Kristin I Stanford
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Amber Kempton
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Sakima Smith
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Caroline C Szujewski
- Institute for Integrative Physiology, Grossman Institute for Neuroscience Quantitative Biology and Human Behavior, The Committee on Neurobiology, The University of Chicago, Chicago, IL, USA
| | - Abby Silbaugh
- Institute for Integrative Physiology, Grossman Institute for Neuroscience Quantitative Biology and Human Behavior, The Committee on Neurobiology, The University of Chicago, Chicago, IL, USA
| | - Jean-Charles Viemari
- P3M Team, Institut de Neurosciences de la Timone, UMR 7289 AMU-CNRS, Marseille, France
| | - Ana C Takakura
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Alfredo J Garcia
- Institute for Integrative Physiology, Grossman Institute for Neuroscience Quantitative Biology and Human Behavior, The Committee on Neurobiology, The University of Chicago, Chicago, IL, USA
| | - Thiago S Moreira
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Catherine M Czeisler
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - José J Otero
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
16
|
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: 122] [Impact Index Per Article: 24.4] [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
|
17
|
Sawahata M, Mori D, Arioka Y, Kubo H, Kushima I, Kitagawa K, Sobue A, Shishido E, Sekiguchi M, Kodama A, Ikeda R, Aleksic B, Kimura H, Ishizuka K, Nagai T, Kaibuchi K, Nabeshima T, Yamada K, Ozaki N. Generation and analysis of novel Reln-deleted mouse model corresponding to exonic Reln deletion in schizophrenia. Psychiatry Clin Neurosci 2020; 74:318-327. [PMID: 32065683 PMCID: PMC7318658 DOI: 10.1111/pcn.12993] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/30/2020] [Accepted: 02/03/2020] [Indexed: 12/14/2022]
Abstract
AIM A Japanese individual with schizophrenia harboring a novel exonic deletion in RELN was recently identified by genome-wide copy-number variation analysis. Thus, the present study aimed to generate and analyze a model mouse to clarify whether Reln deficiency is associated with the pathogenesis of schizophrenia. METHODS A mouse line with a novel RELN exonic deletion (Reln-del) was established using the CRISPR/Cas9 method to elucidate the underlying molecular mechanism. Subsequently, general behavioral tests and histopathological examinations of the model mice were conducted and phenotypic analysis of the cerebellar granule cell migration was performed. RESULTS The phenotype of homozygous Reln-del mice was similar to that of reeler mice with cerebellar atrophy, dysplasia of the cerebral layers, and abrogated protein levels of cerebral reelin. The expression of reelin in heterozygous Reln-del mice was approximately half of that in wild-type mice. Conversely, behavioral analyses in heterozygous Reln-del mice without cerebellar atrophy or dysplasia showed abnormal social novelty in the three-chamber social interaction test. In vitro reaggregation formation and neuronal migration were severely altered in the cerebellar cultures of homozygous Reln-del mice. CONCLUSION The present results in novel Reln-del mice modeled after our patient with a novel exonic deletion in RELN are expected to contribute to the development of reelin-based therapies for schizophrenia.
Collapse
Affiliation(s)
- Masahito Sawahata
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Japan
| | - Daisuke Mori
- Brain and Mind Research Center, Nagoya University, Nagoya, Japan.,Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuko Arioka
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Japan
| | - Hisako Kubo
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Itaru Kushima
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Institute for Advanced Research, Nagoya University, Nagoya, Japan
| | - Kanako Kitagawa
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Japan
| | - Akira Sobue
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Japan
| | - Emiko Shishido
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mariko Sekiguchi
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akiko Kodama
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ryosuke Ikeda
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Branko Aleksic
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroki Kimura
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kanako Ishizuka
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Taku Nagai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Japan
| | - Kozo Kaibuchi
- Department of Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Graduate School of Health Sciences, Fujita Health University, Toyoake, Japan
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
18
|
Lalonde R, Strazielle C. Motor Performances of Spontaneous and Genetically Modified Mutants with Cerebellar Atrophy. THE CEREBELLUM 2019; 18:615-634. [PMID: 30820866 DOI: 10.1007/s12311-019-01017-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Chance discovery of spontaneous mutants with atrophy of the cerebellar cortex has unearthed genes involved in optimizing motor coordination. Rotorod, stationary beam, and suspended wire tests are useful in delineating behavioral phenotypes of spontaneous mutants with cerebellar atrophy such as Grid2Lc, Grid2ho, Rorasg, Agtpbp1pcd, Relnrl, and Dab1scm. Likewise, transgenic or null mutants serving as experimental models of spinocerebellar ataxia (SCA) are phenotyped with the same tests. Among experimental models of autosomal dominant SCA, rotorod deficits were reported in SCA1 to 3, SCA5 to 8, SCA14, SCA17, and SCA27 and stationary beam deficits in SCA1 to 3, SCA5, SCA6, SCA13, SCA17, and SCA27. Beam tests are sensitive to experimental therapies of various kinds including molecules affecting glutamate signaling, mesenchymal stem cells, anti-oligomer antibodies, lentiviral vectors carrying genes, interfering RNAs, or neurotrophic factors, and interbreeding with other mutants.
Collapse
Affiliation(s)
- Robert Lalonde
- Department of Psychology, University of Rouen, 76821, Mont-Saint-Aignan Cedex, France.
| | - Catherine Strazielle
- Laboratory of Stress, Immunity, and Pathogens EA7300, and CHRU of Nancy, University of Lorraine, 54500, Vandoeuvre-les-Nancy, France
| |
Collapse
|
19
|
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
|
20
|
Canet-Pons J, Schubert R, Duecker RP, Schrewe R, Wölke S, Kieslich M, Schnölzer M, Chiocchetti A, Auburger G, Zielen S, Warnken U. Ataxia telangiectasia alters the ApoB and reelin pathway. Neurogenetics 2018; 19:237-255. [DOI: 10.1007/s10048-018-0557-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 10/09/2018] [Indexed: 02/07/2023]
|
21
|
Sun L, Chen R, Bai Y, Li J, Wu Q, Shen Q, Wang X. Morphological and Physiological Characteristics of Ebf2-EGFP-Expressing Cajal-Retzius Cells in Developing Mouse Neocortex. Cereb Cortex 2018; 29:3864-3878. [DOI: 10.1093/cercor/bhy265] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 09/10/2018] [Indexed: 12/22/2022] Open
Abstract
Abstract
Cajal-Retzius (CR) cells are one of the earliest populations of neurons in the cerebral cortex of rodents and primates, and they play a critical role in corticogenesis and cortical lamination during neocortical development. However, a comprehensive morphological and physiological profile of CR cells in the mouse neocortex has not yet been established. Here, we systematically investigated the dynamic development of CR cells in Tg(Ebf2-EGFP)58Gsat/Mmcd mice. The morphological complexity, membrane activities and presynaptic inputs of CR cells coordinately increase and reach a plateau at P5–P9 before regressing. Using 3D reconstruction, we delineated a parallel-stratification pattern of the axonal extension of CR cells. Furthermore, we found that the morphological structure and presynaptic inputs of CR cells were disturbed in Reelin-deficient mice. These findings confirm that CR cells undergo a transient maturation process in layer 1 before disappearing. Importantly, Reelin deficiency impairs the formation of synaptic connections onto CR cells. In conclusion, our results provide insights into the rapid maturation and axonal stratification of CR cells in layer 1. These findings suggest that both the electrophysiological activities and the morphology of CR cells provide vital guidance for the modulation of early circuits, in a Reelin-dependent manner.
Collapse
Affiliation(s)
- Le Sun
- Institute of Biophysics, State Key Laboratory of Brain and Cognitive Sciences, CAS Center for Excellence in Brain Science and Intelligence Technology; Chinese Academy of Sciences, Beijing, China
| | - Ruiguo Chen
- Institute of Biophysics, State Key Laboratory of Brain and Cognitive Sciences, CAS Center for Excellence in Brain Science and Intelligence Technology; Chinese Academy of Sciences, Beijing, China
- The College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Ye Bai
- Institute of Biophysics, State Key Laboratory of Brain and Cognitive Sciences, CAS Center for Excellence in Brain Science and Intelligence Technology; Chinese Academy of Sciences, Beijing, China
- The College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Jia Li
- PTN graduate program, School of Life Science, Peking University, Beijing, China
| | - Qian Wu
- Institute of Biophysics, State Key Laboratory of Brain and Cognitive Sciences, CAS Center for Excellence in Brain Science and Intelligence Technology; Chinese Academy of Sciences, Beijing, China
| | - Qin Shen
- Tongji Hospital, Brain and Spinal Cord Innovative Research Center, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiaoqun Wang
- Institute of Biophysics, State Key Laboratory of Brain and Cognitive Sciences, CAS Center for Excellence in Brain Science and Intelligence Technology; Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
22
|
Prume M, Rollenhagen A, Lübke JHR. Structural and Synaptic Organization of the Adult Reeler Mouse Somatosensory Neocortex: A Comparative Fine-Scale Electron Microscopic Study of Reeler With Wild Type Mice. Front Neuroanat 2018; 12:80. [PMID: 30344480 PMCID: PMC6182073 DOI: 10.3389/fnana.2018.00080] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 09/13/2018] [Indexed: 11/17/2022] Open
Abstract
The reeler mouse has been widely used to study various aspects of cortico- and synaptogenesis, but also as a model for several neurological and neurodegenerative disorders. In contrast to development, comparably little is known about the neuronal composition and synaptic organization of the adult reeler mouse neocortex, in particular at the fine-scale electron microscopic level, which was investigated here and compared with wild type (WT) mice. In this study, the “barrel field” of the adult reeler and WT mouse somatosensory neocortex is used as a model system. In reeler the characteristic six-layered structure is no longer existent, but replaced by a conglomerate of neurons organized in homologous clusters with maintained morphological identity and heterologous clusters between neurons and/or oligodendrocytes. These clusters are loosely scattered throughout the neocortical mass between the pial surface and the white matter. In contrast to WT, layer 1 (L1), if existent, seems to be diluted into the volume of the neocortical mass with no clear boundary. L1 also contains clusters of migrated or persistent neurons, oligodendro- and astrocytes. As in WT, myelinated and unmyelinated axons were found throughout the neocortical mass, but in reeler they were organized in massive fiber bundles with a high fiber packing density. A prominent and massive thalamocortical projection traverses through the neocortical mass, always accompanied by numerous “active” oligodendrocytes whereas in WT no such projections were found and “silent” oligodendrocytes were restricted to the white matter. In the adult reeler mouse neocortex, synaptic boutons terminate on somata, dendritic shafts, spines of different types and axon initial segments with no signs of structural distortion and/or degeneration, indicating a “normal” postsynaptic innervation pattern of neurons. In addition, synaptic complexes between boutons and their postsynaptic targets are tightly ensheathed by fine astrocytic processes, as in WT. In conclusion, the neuronal clusters may represent a possible alternative organization principle in adult reeler mice “replacing” layer formation. If so, these homologous clusters may represent individual “functional units” where neurons are highly interconnected and may function as the equivalent of neurons integrated in a cortical layer. The structural composition and postsynaptic innervation pattern of neurons by synaptic boutons provide the structural basis for the establishment of a functional although altered cortical network in the adult reeler mouse.
Collapse
Affiliation(s)
- Miriam Prume
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Jülich, Germany
| | - Astrid Rollenhagen
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Jülich, Germany
| | - Joachim H R Lübke
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Jülich, Germany.,Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH University Hospital Aachen, Aachen, Germany.,JARA Translational Brain Medicine, Jülich, Germany
| |
Collapse
|
23
|
Newell AJ, Lalitsasivimol D, Willing J, Gonzales K, Waters EM, Milner TA, McEwen BS, Wagner CK. Progesterone receptor expression in cajal-retzius cells of the developing rat dentate gyrus: Potential role in hippocampus-dependent memory. J Comp Neurol 2018; 526:2285-2300. [PMID: 30069875 PMCID: PMC6193812 DOI: 10.1002/cne.24485] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 05/03/2018] [Accepted: 05/23/2018] [Indexed: 12/16/2022]
Abstract
The development of medial temporal lobe circuits is critical for subsequent learning and memory functions later in life. The present study reports the expression of progesterone receptor (PR), a powerful transcription factor of the nuclear steroid receptor superfamily, in Cajal-Retzius cells of the molecular layer of the dentate gyrus of rats. PR was transiently expressed from the day of birth through postnatal day 21, but was absent thereafter. Although PR immunoreactive (PR-ir) cells did not clearly express typical markers of mature neurons, they possessed an ultrastructural morphology consistent with neurons. PRir cells did not express markers for GABAergic neurons, neuronal precursor cells, nor radial glia. However, virtually all PR cells co-expressed the calcium binding protein, calretinin, and the glycoprotein, reelin, both reliable markers for Cajal-Retzius neurons, a transient population of developmentally critical pioneer neurons that guide synaptogenesis of perforant path afferents and histogenesis of the dentate gyrus. Indeed, inhibition of PR activity during the first two weeks of life impaired adult performance on both the novel object recognition and object placement memory tasks, two behavioral tasks hypothesized to describe facets of episodic-like memory in rodents. These findings suggest that PR plays an unexplored and important role in the development of hippocampal circuitry and adult memory function.
Collapse
Affiliation(s)
- Andrew J. Newell
- Department of Psychology, Center for Neuroscience Research’, 1400 Washington Ave., University at Albany, Albany, NY 12222
| | - Diana Lalitsasivimol
- Department of Psychology, Center for Neuroscience Research’, 1400 Washington Ave., University at Albany, Albany, NY 12222
| | - Jari Willing
- Department of Psychology, Behavioral Neuroscience Program, 603 E Daniel St., University of Illinois at Urbana-Champaign, Champaign, IL 61820
| | - Keith Gonzales
- Department of Psychology, Center for Neuroscience Research’, 1400 Washington Ave., University at Albany, Albany, NY 12222
| | - Elizabeth M. Waters
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065
| | - Teresa A. Milner
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61, St New York, NY 1006521
| | - Bruce S. McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065
| | - Christine K. Wagner
- Department of Psychology, Center for Neuroscience Research’, 1400 Washington Ave., University at Albany, Albany, NY 12222
| |
Collapse
|
24
|
Gilbert J, Man HY. Fundamental Elements in Autism: From Neurogenesis and Neurite Growth to Synaptic Plasticity. Front Cell Neurosci 2017; 11:359. [PMID: 29209173 PMCID: PMC5701944 DOI: 10.3389/fncel.2017.00359] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 10/31/2017] [Indexed: 01/12/2023] Open
Abstract
Autism spectrum disorder (ASD) is a set of neurodevelopmental disorders with a high prevalence and impact on society. ASDs are characterized by deficits in both social behavior and cognitive function. There is a strong genetic basis underlying ASDs that is highly heterogeneous; however, multiple studies have highlighted the involvement of key processes, including neurogenesis, neurite growth, synaptogenesis and synaptic plasticity in the pathophysiology of neurodevelopmental disorders. In this review article, we focus on the major genes and signaling pathways implicated in ASD and discuss the cellular, molecular and functional studies that have shed light on common dysregulated pathways using in vitro, in vivo and human evidence. HighlightsAutism spectrum disorder (ASD) has a prevalence of 1 in 68 children in the United States. ASDs are highly heterogeneous in their genetic basis. ASDs share common features at the cellular and molecular levels in the brain. Most ASD genes are implicated in neurogenesis, structural maturation, synaptogenesis and function.
Collapse
Affiliation(s)
- James Gilbert
- Department of Biology, Boston University, Boston, MA, United States
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA, United States.,Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
25
|
Blume M, Inoguchi F, Sugiyama T, Owada Y, Osumi N, Aimi Y, Taki K, Katsuyama Y. Dab1 contributes differently to the morphogenesis of the hippocampal subdivisions. Dev Growth Differ 2017; 59:657-673. [DOI: 10.1111/dgd.12393] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 07/31/2017] [Accepted: 08/16/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Marissa Blume
- School for Mental Health and Neuroscience; European Graduate School of Neuroscience; Maastricht University; Maastricht 6229 ER The Netherlands
- Department of Developmental Neuroscience; Graduate School of Medicine; Tohoku University; Sendai 980-8575 Japan
| | - Fuduki Inoguchi
- Department of Anatomy; Shiga University of Medical Science; Otsu 520-2192 Japan
| | - Taku Sugiyama
- Department of Developmental Neuroscience; Graduate School of Medicine; Tohoku University; Sendai 980-8575 Japan
| | - Yuji Owada
- Department of Organ Anatomy; Graduate School of Medicine; Tohoku University; Sendai 980-8575 Japan
| | - Noriko Osumi
- Department of Developmental Neuroscience; Graduate School of Medicine; Tohoku University; Sendai 980-8575 Japan
| | - Yoshinari Aimi
- Department of Anatomy; Shiga University of Medical Science; Otsu 520-2192 Japan
| | - Kosuke Taki
- Department of Anatomy; Shiga University of Medical Science; Otsu 520-2192 Japan
| | - Yu Katsuyama
- Department of Developmental Neuroscience; Graduate School of Medicine; Tohoku University; Sendai 980-8575 Japan
- Department of Anatomy; Shiga University of Medical Science; Otsu 520-2192 Japan
- Department of Organ Anatomy; Graduate School of Medicine; Tohoku University; Sendai 980-8575 Japan
| |
Collapse
|
26
|
C-Terminal Region Truncation of RELN Disrupts an Interaction with VLDLR, Causing Abnormal Development of the Cerebral Cortex and Hippocampus. J Neurosci 2017; 37:960-971. [PMID: 28123028 DOI: 10.1523/jneurosci.1826-16.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 11/03/2016] [Accepted: 11/19/2016] [Indexed: 11/21/2022] Open
Abstract
We discovered a hypomorphic reelin (Reln) mutant with abnormal cortical lamination and no cerebellar hypoplasia. This mutant, RelnCTRdel, carries a chemically induced splice-site mutation that truncates the C-terminal region (CTR) domain of RELN protein and displays remarkably distinct phenotypes from reeler The mutant does not have an inverted cortex, but cortical neurons overmigrate and invade the marginal zone, which are characteristics similar to a phenotype seen in the cerebral cortex of Vldlrnull mice. The dentate gyrus shows a novel phenotype: the infrapyramidal blade is absent, while the suprapyramidal blade is present and laminated. Genetic epistasis analysis showed that RelnCTRdel/Apoer2null double homozygotes have phenotypes akin to those of reeler mutants, while RelnCTRdel/Vldlrnull mice do not. Given that the receptor double knock-out mice resemble reeler mutants, we infer that RelnCTRdel/Apoer2null double homozygotes have both receptor pathways disrupted. This suggests that CTR-truncation disrupts an interaction with VLDLR (very low-density lipoprotein receptor), while the APOER2 signaling pathway remains active, which accounts for the hypomorphic phenotype in RelnCTRdel mice. A RELN-binding assay confirms that CTR truncation significantly decreases RELN binding to VLDLR, but not to APOER2. Together, the in vitro and in vivo results demonstrate that the CTR domain confers receptor-binding specificity of RELN. SIGNIFICANCE STATEMENT Reelin signaling is important for brain development and is associated with human type II lissencephaly. Reln mutations in mice and humans are usually associated with cerebellar hypoplasia. A new Reln mutant with a truncation of the C-terminal region (CTR) domain shows that Reln mutation can cause abnormal phenotypes in the cortex and hippocampus without cerebellar hypoplasia. Genetic analysis suggested that CTR truncation disrupts an interaction with the RELN receptor VLDLR (very low-density lipoprotein receptor); this was confirmed by a RELN-binding assay. This result provides a mechanistic explanation for the hypomorphic phenotype of the CTR-deletion mutant, and further suggests that Reln mutations may cause more subtle forms of human brain malformation than classic lissencephalies.
Collapse
|
27
|
Genetic and Molecular Approaches to Study Neuronal Migration in the Developing Cerebral Cortex. Brain Sci 2017; 7:brainsci7050053. [PMID: 28475113 PMCID: PMC5447935 DOI: 10.3390/brainsci7050053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 04/21/2017] [Accepted: 05/02/2017] [Indexed: 11/17/2022] Open
Abstract
The migration of neuronal cells in the developing cerebral cortex is essential for proper development of the brain and brain networks. Disturbances in this process, due to genetic abnormalities or exogenous factors, leads to aberrant brain formation, brain network formation, and brain function. In the last decade, there has been extensive research in the field of neuronal migration. In this review, we describe different methods and approaches to assess and study neuronal migration in the developing cerebral cortex. First, we discuss several genetic methods, techniques and genetic models that have been used to study neuronal migration in the developing cortex. Second, we describe several molecular approaches to study aberrant neuronal migration in the cortex which can be used to elucidate the underlying mechanisms of neuronal migration. Finally, we describe model systems to investigate and assess the potential toxicity effect of prenatal exposure to environmental chemicals on proper brain formation and neuronal migration.
Collapse
|
28
|
Hirota Y, Nakajima K. Control of Neuronal Migration and Aggregation by Reelin Signaling in the Developing Cerebral Cortex. Front Cell Dev Biol 2017; 5:40. [PMID: 28507985 PMCID: PMC5410752 DOI: 10.3389/fcell.2017.00040] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 03/31/2017] [Indexed: 11/13/2022] Open
Abstract
The mammalian cerebral neocortex has a well-organized laminar structure, achieved by the highly coordinated control of neuronal migration. During cortical development, excitatory neurons born near the lateral ventricle migrate radially to reach their final positions to form the cortical plate. During this process, dynamic changes are observed in the morphologies and migration modes, including multipolar migration, locomotion, and terminal translocation, of the newborn neurons. Disruption of these migration processes can result in neuronal disorders such as lissencephaly and periventricular heterotopia. The extracellular protein, Reelin, mainly secreted by the Cajal-Retzius neurons in the marginal zone during development, plays a crucial role in the neuronal migration and neocortical lamination. Reelin signaling, which exerts essential roles in the formation of the layered neocortex, is triggered by the binding of Reelin to its receptors, ApoER2 and VLDLR, followed by phosphorylation of the Dab1 adaptor protein. Accumulating evidence suggests that Reelin signaling controls multiple steps of neuronal migration, including the transition from multipolar to bipolar neurons, terminal translocation, and termination of migration beneath the marginal zone. In addition, it has been shown that ectopically expressed Reelin can cause neuronal aggregation via an N-cadherin-mediated manner. This review attempts to summarize our knowledge of the roles played by Reelin in neuronal migration and the underlying mechanisms.
Collapse
Affiliation(s)
- Yuki Hirota
- Department of Anatomy, Keio University School of MedicineTokyo, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of MedicineTokyo, Japan
| |
Collapse
|
29
|
Khialeeva E, Carpenter EM. Nonneuronal roles for the reelin signaling pathway. Dev Dyn 2016; 246:217-226. [PMID: 27739126 DOI: 10.1002/dvdy.24462] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 10/09/2016] [Accepted: 10/10/2016] [Indexed: 12/21/2022] Open
Abstract
The reelin signaling pathway has been established as an important regulator of cell migration during development of the central nervous system, and disruptions in reelin signaling alter the positioning of many types of neurons. Reelin is a large extracellular matrix glycoprotein and governs cell migration through activation of multiple intracellular signaling events by means of the receptors ApoE receptor 2 (ApoER2) and very low density lipoprotein receptor (VLDLR), and the intracellular adaptor protein Disabled-1 (Dab1). Earlier studies reported expression of reelin in nonneuronal tissues, but the functions of this signaling pathway outside of the nervous system have not been studied until recently. A large body of evidence now suggests that reelin functions during development and disease of multiple nonneuronal tissues. This review addresses recent advances in the field of nonneuronal reelin signaling. Developmental Dynamics 246:217-226, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Elvira Khialeeva
- Molecular Biology Interdepartmental Program, University of California Los Angeles, Los Angeles, California
| | - Ellen M Carpenter
- Department of Psychiatry and Biobehavioral Science, University of California Los Angeles School of Medicine, Los Angeles, California
| |
Collapse
|
30
|
Ishii K, Kubo KI, Nakajima K. Reelin and Neuropsychiatric Disorders. Front Cell Neurosci 2016; 10:229. [PMID: 27803648 PMCID: PMC5067484 DOI: 10.3389/fncel.2016.00229] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/22/2016] [Indexed: 12/22/2022] Open
Abstract
Proper neuronal migration and laminar formation during corticogenesis is essential for normal brain function. Disruption of these developmental processes is thought to be involved in the pathogenesis of some neuropsychiatric conditions. Especially, Reelin, a glycoprotein mainly secreted by the Cajal-Retzius cells and a subpopulation of GABAergic interneurons, has been shown to play a critical role, both during embryonic and postnatal periods. Indeed, animal studies have clearly revealed that Reelin is an essential molecule for proper migration of cortical neurons and finally regulates the cell positioning in the cortex during embryonic and early postnatal stages; by contrast, Reelin signaling is closely involved in synaptic function in adulthood. In humans, genetic studies have shown that the reelin gene (RELN) is associated with a number of psychiatric diseases, including Schizophrenia (SZ), bipolar disorder (BP) and autistic spectrum disorder. Indeed, Reln haploinsufficiency has been shown to cause cognitive impairment in rodents, suggesting the expression level of the Reelin protein is closely related to the higher brain functions. However, the molecular abnormalities in the Reelin pathway involved in the pathogenesis of psychiatric disorders are not yet fully understood. In this article, we review the current progress in the understanding of the Reelin functions that could be related to the pathogenesis of psychiatric disorders. Furthermore, we discuss the basis for selecting Reelin and molecules in its downstream signaling pathway as potential therapeutic targets for psychiatric illnesses.
Collapse
Affiliation(s)
- Kazuhiro Ishii
- Department of Anatomy, Keio University School of Medicine Tokyo, Japan
| | - Ken-Ichiro Kubo
- Department of Anatomy, Keio University School of Medicine Tokyo, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine Tokyo, Japan
| |
Collapse
|
31
|
Orcinha C, Münzner G, Gerlach J, Kilias A, Follo M, Egert U, Haas CA. Seizure-Induced Motility of Differentiated Dentate Granule Cells Is Prevented by the Central Reelin Fragment. Front Cell Neurosci 2016; 10:183. [PMID: 27516734 PMCID: PMC4963407 DOI: 10.3389/fncel.2016.00183] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 07/08/2016] [Indexed: 12/01/2022] Open
Abstract
Granule cell dispersion (GCD) represents a pathological widening of the granule cell layer in the dentate gyrus and it is frequently observed in patients with mesial temporal lobe epilepsy (MTLE). Recent studies in human MTLE specimens and in animal epilepsy models have shown that a decreased expression and functional inactivation of the extracellular matrix protein Reelin correlates with GCD formation, but causal evidence is still lacking. Here, we used unilateral kainate (KA) injection into the mouse hippocampus, an established MTLE animal model, to precisely map the loss of reelin mRNA-synthesizing neurons in relation to GCD along the septotemporal axis of the epileptic hippocampus. We show that reelin mRNA-producing neurons are mainly lost in the hilus and that this loss precisely correlates with the occurrence of GCD. To monitor GCD formation in real time, we used organotypic hippocampal slice cultures (OHSCs) prepared from mice which express enhanced green fluorescent protein (eGFP) primarily in differentiated dentate granule cells. Using life cell microscopy we observed that increasing doses of KA resulted in an enhanced motility of eGFP-positive granule cells. Moreover, KA treatment of OHSC resulted in a rapid loss of Reelin-producing interneurons mainly in the hilus, as observed in vivo. A detailed analysis of the migration behavior of individual eGFP-positive granule cells revealed that the majority of these neurons actively migrate toward the hilar region, where Reelin-producing neurons are lost. Treatment with KA and subsequent addition of the recombinant R3–6 Reelin fragment significantly prevented the movement of eGFP-positive granule cells. Together, these findings suggest that GCD formation is indeed triggered by a loss of Reelin in hilar interneurons.
Collapse
Affiliation(s)
- Catarina Orcinha
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center, University of FreiburgFreiburg, Germany; Faculty of Medicine, University of FreiburgFreiburg, Germany; Faculty of Biology, University of FreiburgFreiburg, Germany; Bernstein Center Freiburg, University of FreiburgFreiburg, Germany
| | - Gert Münzner
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center, University of FreiburgFreiburg, Germany; Faculty of Medicine, University of FreiburgFreiburg, Germany; Faculty of Biology, University of FreiburgFreiburg, Germany
| | - Johannes Gerlach
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center, University of FreiburgFreiburg, Germany; Faculty of Medicine, University of FreiburgFreiburg, Germany; Faculty of Biology, University of FreiburgFreiburg, Germany
| | - Antje Kilias
- Faculty of Biology, University of FreiburgFreiburg, Germany; Bernstein Center Freiburg, University of FreiburgFreiburg, Germany; Laboratory for Biomicrotechnology, Department of Microsystems Engineering, University of FreiburgFreiburg, Germany
| | - Marie Follo
- Faculty of Medicine, University of FreiburgFreiburg, Germany; Lighthouse Core Facility, Department of Medicine I, Medical Center, University of FreiburgFreiburg, Germany
| | - Ulrich Egert
- Bernstein Center Freiburg, University of FreiburgFreiburg, Germany; Laboratory for Biomicrotechnology, Department of Microsystems Engineering, University of FreiburgFreiburg, Germany; BrainLinks-BrainTools, Cluster of Excellence, University of FreiburgFreiburg, Germany
| | - Carola A Haas
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center, University of FreiburgFreiburg, Germany; Faculty of Medicine, University of FreiburgFreiburg, Germany; Bernstein Center Freiburg, University of FreiburgFreiburg, Germany; BrainLinks-BrainTools, Cluster of Excellence, University of FreiburgFreiburg, Germany
| |
Collapse
|
32
|
Bock HH, May P. Canonical and Non-canonical Reelin Signaling. Front Cell Neurosci 2016; 10:166. [PMID: 27445693 PMCID: PMC4928174 DOI: 10.3389/fncel.2016.00166] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/08/2016] [Indexed: 12/11/2022] Open
Abstract
Reelin is a large secreted glycoprotein that is essential for correct neuronal positioning during neurodevelopment and is important for synaptic plasticity in the mature brain. Moreover, Reelin is expressed in many extraneuronal tissues; yet the roles of peripheral Reelin are largely unknown. In the brain, many of Reelin's functions are mediated by a molecular signaling cascade that involves two lipoprotein receptors, apolipoprotein E receptor-2 (Apoer2) and very low density-lipoprotein receptor (Vldlr), the neuronal phosphoprotein Disabled-1 (Dab1), and members of the Src family of protein tyrosine kinases as crucial elements. This core signaling pathway in turn modulates the activity of adaptor proteins and downstream protein kinase cascades, many of which target the neuronal cytoskeleton. However, additional Reelin-binding receptors have been postulated or described, either as coreceptors that are essential for the activation of the "canonical" Reelin signaling cascade involving Apoer2/Vldlr and Dab1, or as receptors that activate alternative or additional signaling pathways. Here we will give an overview of canonical and alternative Reelin signaling pathways, molecular mechanisms involved, and their potential physiological roles in the context of different biological settings.
Collapse
Affiliation(s)
- Hans H Bock
- Clinic of Gastroenterology and Hepatology, Heinrich-Heine-University Düsseldorf Düsseldorf, Germany
| | - Petra May
- Clinic of Gastroenterology and Hepatology, Heinrich-Heine-University Düsseldorf Düsseldorf, Germany
| |
Collapse
|
33
|
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
|
34
|
Lammert DB, Howell BW. RELN Mutations in Autism Spectrum Disorder. Front Cell Neurosci 2016; 10:84. [PMID: 27064498 PMCID: PMC4814460 DOI: 10.3389/fncel.2016.00084] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 03/18/2016] [Indexed: 11/13/2022] Open
Abstract
RELN encodes a large, secreted glycoprotein integral to proper neuronal positioning during development and regulation of synaptic function postnatally. Rare, homozygous, null mutations lead to lissencephaly with cerebellar hypoplasia (LCH), accompanied by developmental delay and epilepsy. Until recently, little was known about the frequency or consequences of heterozygous mutations. Several lines of evidence from multiple studies now implicate heterozygous mutations in RELN in autism spectrum disorders (ASD). RELN maps to the AUTS1 locus on 7q22, and at this time over 40 distinct mutations have been identified that would alter the protein sequence, four of which are de novo. The RELN mutations that are most clearly consequential are those that are predicted to inactivate the signaling function of the encoded protein and those that fall in a highly conserved RXR motif found at the core of the 16 Reelin subrepeats. Despite the growing evidence of RELN dysfunction in ASD, it appears that these mutations in isolation are insufficient and that secondary genetic or environmental factors are likely required for a diagnosis.
Collapse
Affiliation(s)
- Dawn B Lammert
- Department of Neuroscience and Physiology, SUNY Upstate Medical School Syracuse, NY, USA
| | - Brian W Howell
- Department of Neuroscience and Physiology, SUNY Upstate Medical School Syracuse, NY, USA
| |
Collapse
|
35
|
Cionni M, Menke C, Stottmann RW. Novel genetic tools facilitate the study of cortical neuron migration. Mamm Genome 2015; 27:8-16. [PMID: 26662625 DOI: 10.1007/s00335-015-9615-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 11/23/2015] [Indexed: 10/22/2022]
Abstract
Key facets of mammalian forebrain cortical development include the radial migration of projection neurons and subsequent cellular differentiation into layer-specific subtypes. Inappropriate regulation of these processes can lead to a number of congenital brain defects in both mouse and human, including lissencephaly and intellectual disability. The genes regulating these processes are still not all identified, suggesting genetic analyses will continue to be a powerful tool in mechanistically studying the development of the cerebral cortex. Reelin is a molecule which we have understood to be critical for proper cortical development for many years. The precise mechanism of Reelin, however, is not fully understood. To address both of these unresolved issues, we report here the creation of a novel conditional allele of the Reelin gene and showcase the use of an Etv1-GFP transgenic line highlighting a subpopulation of the cortex: layer V pyramidal neurons. Together, these represent genetic tools which may facilitate the study of cortical development in a number of different ways.
Collapse
Affiliation(s)
- Megan Cionni
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., MLC 7016, Cincinnati, OH, 45229, USA
| | - Chelsea Menke
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., MLC 7016, Cincinnati, OH, 45229, USA
| | - Rolf W Stottmann
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., MLC 7016, Cincinnati, OH, 45229, USA. .,Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
| |
Collapse
|
36
|
Reiner O, Karzbrun E, Kshirsagar A, Kaibuchi K. Regulation of neuronal migration, an emerging topic in autism spectrum disorders. J Neurochem 2015; 136:440-56. [PMID: 26485324 DOI: 10.1111/jnc.13403] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 09/04/2015] [Accepted: 10/09/2015] [Indexed: 12/14/2022]
Abstract
Autism spectrum disorders (ASD) encompass a group of neurodevelopmental diseases that demonstrate strong heritability, however, the inheritance is not simple and many genes have been associated with these disorders. ASD is regarded as a neurodevelopmental disorder, and abnormalities at different developmental stages are part of the disease etiology. This review provides a general background on neuronal migration during brain development and discusses recent advancements in the field connecting ASD and aberrant neuronal migration. We propose that neuronal migration impairment may be an important common pathophysiology in autism spectrum disorders (ASD). This review provides a general background on neuronal migration during brain development and discusses recent advancements in the field connecting ASD and aberrant neuronal migration.
Collapse
Affiliation(s)
- Orly Reiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal Karzbrun
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Aditya Kshirsagar
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Showa, Nagoya, Japan
| |
Collapse
|
37
|
Abstract
UNLABELLED The mechanisms controlling cortical dendrite initiation and targeting are poorly understood. Multiphoton imaging of developing mouse cortex reveals that apical dendrites emerge by direct transformation of the neuron's leading process during the terminal phase of neuronal migration. During this ∼110 min period, the dendritic arbor increases ∼2.5-fold in size and migration arrest occurs below the first stable branch point in the developing arbor. This dendritic outgrowth is triggered at the time of leading process contact with the marginal zone (MZ) and occurs primarily by neurite extension into the extracellular matrix of the MZ. In reeler cortices that lack the secreted glycoprotein Reelin, a subset of neurons completed migration but then retracted and reorganized their arbor in a tangential direction away from the MZ soon after migration arrest. For these reeler neurons, the tangential oriented primary neurites were longer lived than the radially oriented primary neurites, whereas the opposite was true of wild-type (WT) neurons. Application of Reelin protein to reeler cortices destabilized tangential neurites while stabilizing radial neurites and stimulating dendritic growth in the MZ. Therefore, Reelin functions as part of a polarity signaling system that links dendritogenesis in the MZ with cellular positioning and cortical lamination. SIGNIFICANCE STATEMENT Whether the apical dendrite emerges by transformation of the leading process of the migrating neuron or emerges de novo after migration is completed is unclear. Similarly, it is not clear whether the secreted glycoprotein Reelin controls migration and dendritic growth as related or separate processes. Here, multiphoton microscopy reveals the direct transformation of the leading process into the apical dendrite. This transformation is coupled to the successful completion of migration and neuronal soma arrest occurs below the first stable branch point of the nascent dendrite. Deficiency in Reelin causes the forming dendrite to avoid its normal target area and branch aberrantly, leading to improper cellular positioning. Therefore, this study links Reelin-dependent dendritogenesis with migration arrest and cortical lamination.
Collapse
|
38
|
He S, Li Z, Ge S, Yu YC, Shi SH. Inside-Out Radial Migration Facilitates Lineage-Dependent Neocortical Microcircuit Assembly. Neuron 2015; 86:1159-66. [PMID: 26050035 DOI: 10.1016/j.neuron.2015.05.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 03/21/2015] [Accepted: 04/27/2015] [Indexed: 01/24/2023]
Abstract
Neocortical excitatory neurons migrate radially along the glial fibers of mother radial glial progenitors (RGPs) in a birth-date-dependent inside-out manner. However, the precise functional significance of this well-established orderly neuronal migration remains largely unclear. Here, we show that strong electrical synapses selectively form between RGPs and their newborn progeny and between sister excitatory neurons in ontogenetic radial clones at the embryonic stage. Interestingly, the preferential electrical coupling between sister excitatory neurons, but not that between RGP and newborn progeny, is eliminated in mice lacking REELIN or upon clonal depletion of DISABLED-1, which compromises the inside-out radial neuronal migration pattern in the developing neocortex. Moreover, increased levels of Ephrin-A ligand or receptor that laterally disperse sister excitatory neurons also disrupt preferential electrical coupling between radially aligned sister excitatory neurons. These results suggest that RGP-guided inside-out radial neuronal migration facilitates the initial assembly of lineage-dependent precise columnar microcircuits in the neocortex.
Collapse
Affiliation(s)
- Shuijin He
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Zhizhong Li
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Shaoyu Ge
- Department of Neurobiology and Behavior, SUNY at Stony Brook, Stony Brook, NY 11794, USA
| | - Yong-Chun Yu
- Institute of Neurobiology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, P.R. China
| | - Song-Hai Shi
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
39
|
Squarzoni P, Thion MS, Garel S. Neuronal and microglial regulators of cortical wiring: usual and novel guideposts. Front Neurosci 2015; 9:248. [PMID: 26236185 PMCID: PMC4505395 DOI: 10.3389/fnins.2015.00248] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 06/30/2015] [Indexed: 12/17/2022] Open
Abstract
Neocortex functioning relies on the formation of complex networks that begin to be assembled during embryogenesis by highly stereotyped processes of cell migration and axonal navigation. The guidance of cells and axons is driven by extracellular cues, released along by final targets or intermediate targets located along specific pathways. In particular, guidepost cells, originally described in the grasshopper, are considered discrete, specialized cell populations located at crucial decision points along axonal trajectories that regulate tract formation. These cells are usually early-born, transient and act at short-range or via cell-cell contact. The vast majority of guidepost cells initially identified were glial cells, which play a role in the formation of important axonal tracts in the forebrain, such as the corpus callosum, anterior, and post-optic commissures as well as optic chiasm. In the last decades, tangential migrating neurons have also been found to participate in the guidance of principal axonal tracts in the forebrain. This is the case for several examples such as guideposts for the lateral olfactory tract (LOT), corridor cells, which open an internal path for thalamo-cortical axons and Cajal-Retzius cells that have been involved in the formation of the entorhino-hippocampal connections. More recently, microglia, the resident macrophages of the brain, were specifically observed at the crossroads of important neuronal migratory routes and axonal tract pathways during forebrain development. We furthermore found that microglia participate to the shaping of prenatal forebrain circuits, thereby opening novel perspectives on forebrain development and wiring. Here we will review the last findings on already known guidepost cell populations and will discuss the role of microglia as a potentially new class of atypical guidepost cells.
Collapse
Affiliation(s)
- Paola Squarzoni
- Centre National de la Recherche Scientifique UMR8197, Ecole Normale Supérieure, Institut de Biologie, Institut National de la Santé et de la Recherche Médicale U1024 Paris, France
| | - Morgane S Thion
- Centre National de la Recherche Scientifique UMR8197, Ecole Normale Supérieure, Institut de Biologie, Institut National de la Santé et de la Recherche Médicale U1024 Paris, France
| | - Sonia Garel
- Centre National de la Recherche Scientifique UMR8197, Ecole Normale Supérieure, Institut de Biologie, Institut National de la Santé et de la Recherche Médicale U1024 Paris, France
| |
Collapse
|
40
|
Hayashi K, Kubo KI, Kitazawa A, Nakajima K. Cellular dynamics of neuronal migration in the hippocampus. Front Neurosci 2015; 9:135. [PMID: 25964735 PMCID: PMC4408843 DOI: 10.3389/fnins.2015.00135] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 04/02/2015] [Indexed: 12/30/2022] Open
Abstract
A fine structure of the hippocampus is required for proper functions, and disruption of this formation by neuronal migration defects during development may play a role in some psychiatric illnesses. During hippocampal development in rodents, pyramidal neurons in the Ammon's horn are mostly generated in the ventricular zone (VZ), spent as multipolar cells just above the VZ, and then migrate radially toward the pial surface, ultimately settling into the hippocampal plate. Although this process is similar to that of neocortical projection neurons, these are not identical. In addition to numerous histological studies, the development of novel techniques gives a clear picture of the cellular dynamics of hippocampal neurons, as well as neocortical neurons. In this article, we provide an overview of the cellular mechanisms of rodent hippocampal neuronal migration including those of dentate granule cells, especially focusing on the differences of migration modes between hippocampal neurons and neocortical neurons. The unique migration mode of hippocampal pyramidal neurons might enable clonally related cells in the Ammon's horn to distribute in a horizontal fashion.
Collapse
Affiliation(s)
- Kanehiro Hayashi
- Department of Anatomy, Keio University School of Medicine Tokyo, Japan
| | - Ken-Ichiro Kubo
- Department of Anatomy, Keio University School of Medicine Tokyo, Japan
| | - Ayako Kitazawa
- Department of Anatomy, Keio University School of Medicine Tokyo, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine Tokyo, Japan
| |
Collapse
|
41
|
Skorput AGJ, Yeh HH. Effects of ethanol exposure in utero on Cajal-Retzius cells in the developing cortex. Alcohol Clin Exp Res 2015; 39:853-62. [PMID: 25845402 DOI: 10.1111/acer.12696] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/31/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND Prenatal exposure to ethanol exerts teratogenic effects on the developing brain. Here, we tested the hypothesis that exposure to ethanol in utero alters the disposition of Cajal-Retzius cells that play a key role in orchestrating proliferation, migration, and laminar integration of cortical neurons in the embryonic cortex. METHODS Pregnant Ebf2-EGFP mice, harboring EGFP-fluorescent Cajal-Retzius cells, were subjected to a 2% w/w ethanol consumption regimen starting at neural tube closure and lasting throughout gestation. Genesis of Cajal-Retzius cells was assessed by means of 5-bromo-2-deoxyuridine (BrdU) immunofluorescence at embryonic day 12.5, their counts and distribution were determined between postnatal day (P)0 and P4, patch clamp electrophysiology was performed between P2 and P3 to analyze GABA-mediated synaptic activity, and open-field behavioral testing was conducted in P45-P50 adolescents. RESULTS In Ebf2-EGFP embryos exposed to ethanol in utero, we found increased BrdU labeling and expanded distribution of Cajal-Retzius cells in the cortical hem, pointing to increased genesis and proliferation. Postnatally, we found an increase in Cajal-Retzius cell number in cortical layer I. In addition, they displayed altered patterning of spontaneous GABA-mediated synaptic barrages and enhanced GABA-mediated synaptic activity, suggesting enhanced GABAergic tone. CONCLUSIONS These findings, together, underscore that Cajal-Retzius cells contribute to the ethanol-induced aberration of cortical development and abnormal GABAergic neurotransmission at the impactful time when intracortical circuits form.
Collapse
Affiliation(s)
- Alexander G J Skorput
- Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | | |
Collapse
|
42
|
Extracellular proteolysis of reelin by tissue plasminogen activator following synaptic potentiation. Neuroscience 2014; 274:299-307. [PMID: 24892761 DOI: 10.1016/j.neuroscience.2014.05.046] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 05/21/2014] [Indexed: 01/19/2023]
Abstract
The secreted glycoprotein reelin plays an indispensable role in neuronal migration during development and in regulating adult synaptic functions. The upstream mechanisms responsible for initiating and regulating the duration and magnitude of reelin signaling are largely unknown. Here we report that reelin is cleaved between EGF-like repeats 6-7 (R6-7) by tissue plasminogen activator (tPA) under cell-free conditions. No changes were detected in the level of reelin and its fragments in the brains of tPA knockouts, implying that other unknown proteases are responsible for generating reelin fragments found constitutively in the adult brain. Induction of NMDAR-independent long-term potentiation with the potassium channel blocker tetraethylammonium chloride (TEA-Cl) led to a specific up-regulation of reelin processing at R6-7 in wild-type mice. In contrast, no changes in reelin expression and processing were observed in tPA knockouts following TEA-Cl treatment. These results demonstrate that synaptic potentiation results in tPA-dependent reelin processing and suggest that extracellular proteolysis of reelin may regulate reelin signaling in the adult brain.
Collapse
|
43
|
Hawthorne AL. Repurposing Reelin: the new role of radial glia, Reelin and Notch in motor neuron migration. Exp Neurol 2014; 256:17-20. [PMID: 24607503 DOI: 10.1016/j.expneurol.2014.02.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 02/20/2014] [Accepted: 02/24/2014] [Indexed: 01/10/2023]
Abstract
The role of Reelin during cerebral cortical neuron migration has long been studied, but the Reelin signaling pathway and its possible interactions are just beginning to be unraveled. Reelin is not only important in cerebral cortical migration, but has recently been shown to interact with the Notch signaling pathway and to be critical for radial glial cell number and morphology. Lee and Song (2013) show a new Notch- and Reelin-dependent role for radial glia in the mouse spinal cord: to act as a fine filter that allows somatic motor neuron axons but not cell bodies to traverse out of the CNS. Here, the types of neuronal migration are discussed, focusing on motor neurons and cues for proper localization. The interaction of Reelin signaling with the Notch pathway is reviewed, which dictates the proper formation of radial glia in the spinal cord in order to prevent ectopic motor neuron migration (Lee and Song, 2013). Future studies may reveal novel interactions and further insights as to how Reelin functions throughout the developing nervous system.
Collapse
Affiliation(s)
- Alicia L Hawthorne
- Dept. of Cell Biology, Emory University School of Medicine, 615 Michael St., WBRB 415, Atlanta, GA 30322, USA.
| |
Collapse
|
44
|
Qian T, Chen R, Nakamura M, Furukawa T, Kumada T, Akita T, Kilb W, Luhmann HJ, Nakahara D, Fukuda A. Activity-dependent endogenous taurine release facilitates excitatory neurotransmission in the neocortical marginal zone of neonatal rats. Front Cell Neurosci 2014; 8:33. [PMID: 24574969 PMCID: PMC3918584 DOI: 10.3389/fncel.2014.00033] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 01/22/2014] [Indexed: 12/27/2022] Open
Abstract
In the developing cerebral cortex, the marginal zone (MZ), consisting of early-generated neurons such as Cajal-Retzius cells, plays an important role in cell migration and lamination. There is accumulating evidence of widespread excitatory neurotransmission mediated by γ-aminobutyric acid (GABA) in the MZ. Cajal-Retzius cells express not only GABAA receptors but also α2/β subunits of glycine receptors, and exhibit glycine receptor-mediated depolarization due to high [Cl−]i. However, the physiological roles of glycine receptors and their endogenous agonists during neurotransmission in the MZ are yet to be elucidated. To address this question, we performed optical imaging from the MZ using the voltage-sensitive dye JPW1114 on tangential neocortical slices of neonatal rats. A single electrical stimulus evoked an action-potential-dependent optical signal that spread radially over the MZ. The amplitude of the signal was not affected by glutamate receptor blockers, but was suppressed by either GABAA or glycine receptor antagonists. Combined application of both antagonists nearly abolished the signal. Inhibition of Na+, K+-2Cl− cotransporter by 20 µM bumetanide reduced the signal, indicating that this transporter contributes to excitation. Analysis of the interstitial fluid obtained by microdialysis from tangential neocortical slices with high-performance liquid chromatography revealed that GABA and taurine, but not glycine or glutamate, were released in the MZ in response to the electrical stimulation. The ambient release of taurine was reduced by the addition of a voltage-sensitive Na+ channel blocker. Immunohistochemistry and immunoelectron microscopy indicated that taurine was stored both in Cajal-Retzius and non-Cajal-Retzius cells in the MZ, but was not localized in presynaptic structures. Our results suggest that activity-dependent non-synaptic release of endogenous taurine facilitates excitatory neurotransmission through activation of glycine receptors in the MZ.
Collapse
Affiliation(s)
- Taizhe Qian
- Department of Neurophysiology, Hamamatsu University School of Medicine Hamamatsu, Japan
| | - Rongqing Chen
- Institute of Physiology, University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Masato Nakamura
- Department of Psychology and Behavioral Neuroscience, Hamamatsu University School of Medicine Hamamatsu, Japan
| | - Tomonori Furukawa
- Department of Neurophysiology, Hamamatsu University School of Medicine Hamamatsu, Japan
| | - Tatsuro Kumada
- Department of Neurophysiology, Hamamatsu University School of Medicine Hamamatsu, Japan ; Department of Occupational Therapy, Tokoha University Hamamatsu, Japan
| | - Tenpei Akita
- Department of Neurophysiology, Hamamatsu University School of Medicine Hamamatsu, Japan
| | - Werner Kilb
- Institute of Physiology, University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg-University Mainz, Germany
| | - Daiichiro Nakahara
- Department of Psychology and Behavioral Neuroscience, Hamamatsu University School of Medicine Hamamatsu, Japan
| | - Atsuo Fukuda
- Department of Neurophysiology, Hamamatsu University School of Medicine Hamamatsu, Japan
| |
Collapse
|
45
|
Reelin in the Years: Controlling Neuronal Migration and Maturation in the Mammalian Brain. ACTA ACUST UNITED AC 2014. [DOI: 10.1155/2014/597395] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The extracellular protein Reelin was initially identified as an essential factor in the control of neuronal migration and layer formation in the developing mammalian brain. In the years following its discovery, however, it became clear that Reelin is a multifunctional protein that controls not only the positioning of neurons in the developing brain, but also their growth, maturation, and synaptic activity in the adult brain. In this review, we will highlight the major discoveries of the biological activities of Reelin and the underlying molecular mechanisms that affect the development and function of the mammalian brain, from embryonic ages to adulthood.
Collapse
|
46
|
Olson EC. Analysis of preplate splitting and early cortical development illuminates the biology of neurological disease. Front Pediatr 2014; 2:121. [PMID: 25426475 PMCID: PMC4227491 DOI: 10.3389/fped.2014.00121] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 10/23/2014] [Indexed: 12/31/2022] Open
Abstract
The development of the layered cerebral cortex starts with a process called preplate splitting. Preplate splitting involves the establishment of prospective cortical layer 6 (L6) neurons within a plexus of pioneer neurons called the preplate. The forming layer 6 splits the preplate into a superficial layer of pioneer neurons called the marginal zone and a deeper layer of pioneer neurons called the subplate. Disruptions of this early developmental event by toxin exposure or mutation are associated with neurological disease including severe intellectual disability. This review explores recent findings that reveal the dynamism of gene expression and morphological differentiation during this early developmental period. Over 1000 genes show expression increases of ≥2-fold during this period in differentiating mouse L6 neurons. Surprisingly, 88% of previously identified non-syndromic intellectual-disability (NS-ID) genes are expressed at this time and show an average expression increase of 1.6-fold in these differentiating L6 neurons. This changing genetic program must, in part, support the dramatic cellular reorganizations that occur during preplate splitting. While different models have been proposed for the formation of a layer of L6 cortical neurons within the preplate, original histological studies and more recent work exploiting transgenic mice suggest that the process is largely driven by the coordinated polarization and coalescence of L6 neurons rather than by cellular translocation or migration. The observation that genes associated with forms of NS-ID are expressed during very early cortical development raises the possibility of studying the relevant biological events at a time point when the cortex is small, contains relatively few cell types, and few functional circuits. This review then outlines how explant models may prove particularly useful in studying the consequence of toxin and mutation on the etiology of some forms of NS-ID.
Collapse
Affiliation(s)
- Eric C Olson
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University , Syracuse, NY , USA ; Developmental Exposure Alcohol Research Center (DEARC), Binghamton University , Binghamton, NY , USA
| |
Collapse
|
47
|
Kania A. Spinal motor neuron migration and the significance of topographic organization in the nervous system. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 800:133-48. [PMID: 24243104 DOI: 10.1007/978-94-007-7687-6_8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The nervous system displays a high degree of topographic organisation such that neuronal soma position is closely correlated to axonal trajectory. One example of such order is the myotopic organisation of the motor system where spinal motor neuron position parallels that of target muscles. This chapter will discuss the molecular mechanisms underlying motor neuron soma positioning, which include transcriptional control of Reelin signaling and cadherin expression. As the same transcription factors have been shown to control motor axon innervation of target muscles, a simple mechanism of topographic organisation specification is becoming evident raising the question of how coordinating soma position with axon trajectory might be important for nervous system wiring and its function.
Collapse
Affiliation(s)
- Artur Kania
- Institut de recherches cliniques de Montréal (IRCM), 110, ave. des Pins Ouest, Montréal, QC, H2W 1R7, Canada,
| |
Collapse
|
48
|
Martinez-Galan JR, Moncho-Bogani J, Caminos E. Expression of calcium-binding proteins in layer 1 reelin-immunoreactive cells during rat and mouse neocortical development. J Histochem Cytochem 2013; 62:60-9. [PMID: 24134921 DOI: 10.1369/0022155413509381] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cajal-Retzius cells in layer 1 of the developing cerebral cortex and their product of secretion, reelin, an extracellular matrix protein, play a crucial role in establishing the correct lamination pattern in this tissue. As many studies into reelin signaling routes and pathological alterations are conducted in murine models, we used double-labeling and confocal microscopy to compare the distribution of the cell-specific markers, calretinin and calbindin, in reelin-immunoreactive cells during postnatal rat and mouse neocortical development. In the rat, neither calretinin nor calbindin colocalized with reelin in Cajal-Retzius cells at P0-P2. From P5 to P14, the colocalization of reelin and calretinin was commonly found in presumptive rat subpial piriform cells. These cells progressively lacked calretinin expression and persisted into adulthood as part of the pool of layer 1 reelin-positive interneurons. Conversely, in the mouse, reelin-immunoreactive Cajal-Retzius cells colocalized with calretinin and/or calbindin. Subpial piriform cells containing reelin and calretinin were identified at P5-P7, but lacked calretinin expression at P14. In adult mice, as in the rat, reelin-immunoreactive cells did not colocalize with calcium-binding proteins. Our results reveal a complex neurochemical profile of layer 1 cells in the rat neocortex, which makes using a single calcium-binding protein as a marker of rodent reelin-immunoreactive cells difficult.
Collapse
Affiliation(s)
- Juan R Martinez-Galan
- Instituto de Investigación en Discapacidades Neurológicas/Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain (JRMG, JVMB, EC)
| | | | | |
Collapse
|
49
|
Ha S, Stottmann RW, Furley AJ, Beier DR. A forward genetic screen in mice identifies mutants with abnormal cortical patterning. ACTA ACUST UNITED AC 2013; 25:167-79. [PMID: 23968836 DOI: 10.1093/cercor/bht209] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Formation of a 6-layered cortical plate and axon tract patterning are key features of cerebral cortex development. Abnormalities of these processes may be the underlying cause for a range of functional disabilities seen in human neurodevelopmental disorders. To identify mouse mutants with defects in cortical lamination or corticofugal axon guidance, N-ethyl-N-nitrosourea (ENU) mutagenesis was performed using mice expressing LacZ reporter genes in layers II/III and V of the cortex (Rgs4-lacZ) or in corticofugal axons (TAG1-tau-lacZ). Four lines with abnormal cortical lamination have been identified. One of these was a splice site mutation in reelin (Reln) that results in a premature stop codon and the truncation of the C-terminal region (CTR) domain of reelin. Interestingly, this novel allele of Reln did not display cerebellar malformation or ataxia, and this is the first report of a Reln mutant without a cerebellar defect. Four lines with abnormal cortical axon development were also identified, one of which was found by whole-genome resequencing to carry a mutation in Lrp2. These findings demonstrated that the application of ENU mutagenesis to mice carrying transgenic reporters marking cortical anatomy is a sensitive and specific method to identify mutations that disrupt patterning of the developing brain.
Collapse
Affiliation(s)
- Seungshin Ha
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, University of Washington School of Medicine, Seattle, WA 98101, USA
| | - Rolf W Stottmann
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA Divisions of Human Genetics and Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA and
| | - Andrew J Furley
- Department of Biomedical Science, The University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - David R Beier
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, University of Washington School of Medicine, Seattle, WA 98101, USA
| |
Collapse
|
50
|
Pappas GD, Kriho V, Liu WS, Tremolizzo L, Lugli G, Larson J. Immunocytochemical localization of reelin in the olfactory bulb of the heterozygous reeler mouse: An animal model for schizophrenia. Neurol Res 2013; 25:819-30. [PMID: 14669525 DOI: 10.1179/016164103771953916] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Because heterozygous reeler (HR) mice share some abnormal traits with schizophrenic patients, and schizophrenia is often accompanied by impairment of olfactory function, this study examines reelin in the olfactory bulb of the HR mouse. In the WT mouse, reelin immunoreactivity is found in the extracellular matrix, and in the cytoplasm of olfactory nerve fibers, GABAergic interneurons, and glutamatergic mitral cells. Western blot analysis reveals that reelin immunoreactivity in the HR mouse is reduced by 45% compared to WT mouse. This is especially evident in the glomerular GABAergic interneurons. In WT mitral cells, reelin is found in discrete clumps near the axon hillock and within the axon. In the HR mouse, reelin axonal staining is diffuse and densely packed. In the rostral migratory stream of the HR mouse, immunolabeling shows an accumulation of reelin-containing neuronal precursors, apparently unable to shift from tangential to radial migration. These observations indicate that there is a downregulation of reelin in the HR mouse and suggest that secretion of reelin may be compromised. Further studies of the HR mouse may provide a new basis for understanding the role of reelin in the adult CNS, especially as it may relate to schizophrenia.
Collapse
Affiliation(s)
- George D Pappas
- Department of Psychiatry, Psychiatric Institute, Department of Anatomy & Cell Biology, University of Illinois at Chicago, m/c 912, 1601 W. Taylor St., Chicago, IL 60612, USA.
| | | | | | | | | | | |
Collapse
|