1
|
Maejima I, Sato K. New aspects of a small GTPase RAB35 in brain development and function. Neural Regen Res 2025; 20:1971-1980. [PMID: 39254551 PMCID: PMC11691468 DOI: 10.4103/nrr.nrr-d-23-01543] [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: 09/12/2023] [Revised: 12/12/2023] [Accepted: 12/30/2023] [Indexed: 09/11/2024] Open
Abstract
In eukaryotic cells, organelles in the secretory, lysosomal, and endocytic pathways actively exchange biological materials with each other through intracellular membrane trafficking, which is the process of transporting the cargo of proteins, lipids, and other molecules to appropriate compartments via transport vesicles or intermediates. These processes are strictly regulated by various small GTPases such as the RAS-like in rat brain (RAB) protein family, which is the largest subfamily of the RAS superfamily. Dysfunction of membrane trafficking affects tissue homeostasis and leads to a wide range of diseases, including neurological disorders and neurodegenerative diseases. Therefore, it is important to understand the physiological and pathological roles of RAB proteins in brain function. RAB35, a member of the RAB family, is an evolutionarily conserved protein in metazoans. A wide range of studies using cultured mammalian cells and model organisms have revealed that RAB35 mediates various processes such as cytokinesis, endocytic recycling, actin bundling, and cell migration. RAB35 is also involved in neurite outgrowth and turnover of synaptic vesicles. We generated brain-specific Rab35 knockout mice to study the physiological roles of RAB35 in brain development and function. These mice exhibited defects in anxiety-related behaviors and spatial memory. Strikingly, RAB35 is required for the precise positioning of pyramidal neurons during hippocampal development, and thereby for normal hippocampal lamination. In contrast, layer formation in the cerebral cortex occurred superficially, even in the absence of RAB35, suggesting a predominant role for RAB35 in hippocampal development rather than in cerebral cortex development. Recent studies have suggested an association between RAB35 and neurodegenerative diseases, including Parkinson's disease and Alzheimer's disease. In this review, we provide an overview of the current understanding of subcellular functions of RAB35. We also provide insights into the physiological role of RAB35 in mammalian brain development and function, and discuss the involvement of RAB35 dysfunction in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ikuko Maejima
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Ken Sato
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| |
Collapse
|
2
|
Fernandes L, Kleene R, Congiu L, Freitag S, Kneussel M, Loers G, Schachner M. CHL1 depletion affects dopamine receptor D2-dependent modulation of mouse behavior. Front Behav Neurosci 2023; 17:1288509. [PMID: 38025382 PMCID: PMC10665519 DOI: 10.3389/fnbeh.2023.1288509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction The dopaminergic system plays a key role in the appropriate functioning of the central nervous system, where it is essential for emotional balance, arousal, reward, and motor control. The cell adhesion molecule close homolog of L1 (CHL1) contributes to dopaminergic system development, and CHL1 and the dopamine receptor D2 (D2R) are associated with mental disorders like schizophrenia, addiction, autism spectrum disorder and depression. Methods Here, we investigated how the interplay between CHL1 and D2R affects the behavior of young adult male and female wild-type (CHL+/+) and CHL1-deficient (CHL1-/-) mice, when D2R agonist quinpirole and antagonist sulpiride are applied. Results Low doses of quinpirole (0.02 mg/kg body weight) induced hypolocomotion of CHL1+/+ and CHL1-/- males and females, but led to a delayed response in CHL1-/- mice. Sulpiride (1 mg/kg body weight) affected locomotion of CHL1-/- females and social interaction of CHL1+/+ females as well as social interactions of CHL1-/- and CHL1+/+ males. Quinpirole increased novelty-seeking behavior of CHL1-/- males compared to CHL1+/+ males. Vehicle-treated CHL1-/- males and females showed enhanced working memory and reduced stress-related behavior. Discussion We propose that CHL1 regulates D2R-dependent functions in vivo. Deficiency of CHL1 leads to abnormal locomotor activity and emotionality, and to sex-dependent behavioral differences.
Collapse
Affiliation(s)
- Luciana Fernandes
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Ralf Kleene
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Ludovica Congiu
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Sandra Freitag
- Institut für Molekulare Neurogenetik, Zentrum für Molekulare Neurobiologie Hamburg, ZMNH, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Matthias Kneussel
- Institut für Molekulare Neurogenetik, Zentrum für Molekulare Neurobiologie Hamburg, ZMNH, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Melitta Schachner
- Department of Cell Biology and Neuroscience, Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ, United States
| |
Collapse
|
3
|
Maejima I, Hara T, Tsukamoto S, Koizumi H, Kawauchi T, Akuzawa T, Hirai R, Kobayashi H, Isobe I, Emoto K, Kosako H, Sato K. RAB35 is required for murine hippocampal development and functions by regulating neuronal cell distribution. Commun Biol 2023; 6:440. [PMID: 37085665 PMCID: PMC10121692 DOI: 10.1038/s42003-023-04826-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 04/07/2023] [Indexed: 04/23/2023] Open
Abstract
RAB35 is a multifunctional small GTPase that regulates endocytic recycling, cytoskeletal rearrangement, and cytokinesis. However, its physiological functions in mammalian development remain unclear. Here, we generated Rab35-knockout mice and found that RAB35 is essential for early embryogenesis. Interestingly, brain-specific Rab35-knockout mice displayed severe defects in hippocampal lamination owing to impaired distribution of pyramidal neurons, although defects in cerebral cortex formation were not evident. In addition, Rab35-knockout mice exhibited defects in spatial memory and anxiety-related behaviors. Quantitative proteomics indicated that the loss of RAB35 significantly affected the levels of other RAB proteins associated with endocytic trafficking, as well as some neural cell adhesion molecules, such as contactin-2. Collectively, our findings revealed that RAB35 is required for precise neuronal distribution in the developing hippocampus by regulating the expression of cell adhesion molecules, thereby influencing spatial memory.
Collapse
Affiliation(s)
- Ikuko Maejima
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, 371-8512, Japan
| | - Taichi Hara
- Laboratory of Food and Life Science, Faculty of Human Sciences, Waseda University, Tokorozawa, Saitama, 359-1192, Japan
| | - Satoshi Tsukamoto
- Laboratory Animal and Genome Sciences Section, National Institutes for Quantum and Radiological Science and Technology, Chiba, Chiba, 263-8555, Japan
| | - Hiroyuki Koizumi
- Department of Biological Sciences, School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
- Department of Molecular and Cellular Biology, School of Pharmaceutical Sciences, Ohu University, Koriyama, Fukushima, 963-8611, Japan
| | - Takeshi Kawauchi
- Department of Adaptive and Maladaptive Responses in Health and Diseases, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Tomoko Akuzawa
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, 371-8512, Japan
| | - Rika Hirai
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, 371-8512, Japan
| | - Hisae Kobayashi
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, 371-8512, Japan
| | - Inoya Isobe
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, 371-8512, Japan
| | - Kazuo Emoto
- Department of Biological Sciences, School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hidetaka Kosako
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima, Tokushima, 770-8503, Japan
| | - Ken Sato
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, 371-8512, Japan.
- Gunma University Initiative for Advanced Research (GIAR), Gunma University, Maebashi, Gunma, 371-8512, Japan.
| |
Collapse
|
4
|
Wiskott K, Gilardi F, Hainard A, Sanchez JC, Thomas A, Sajic T, Fracasso T. Blood proteome of acute intracranial hemorrhage in infant victims of abusive head trauma. Proteomics 2023; 23:e2200078. [PMID: 36576318 DOI: 10.1002/pmic.202200078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 12/02/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022]
Abstract
Abusive head trauma (AHT) is a leading cause of mortality and morbidity in infants. While the reported incidence is close to 40 cases per 100'000 births/year, misdiagnoses are commonly observed in cases with atypical, subacute, or chronic presentation. Currently, standard clinical evaluation of inflicted intracranial hemorrhagic injury (ICH) in infants urgently requires a screening test able to identify infants who need additional investigations. Blood biomarkers characteristic of AHT may assist in detecting these infants, improving prognosis through early medical care. To date, the application of innovative omics technologies in retrospective studies of AHT in infants is rare, due also to the blood serum and cerebrospinal fluid of AHT cases being scarce and not systematically accessible. Here, we explored the circulating blood proteomes of infants with severe AHT and their atraumatic controls. We discovered 165 circulating serum proteins that display differential changes in AHT cases compared with atraumatic controls. The peripheral blood proteomes of pediatric AHT commonly reflect: (i) potentially secreted proteome from injured brain, and (ii) proteome dysregulated in the system's circulation by successive biological events following acute ICH. This study opens up a novel opportunity for research efforts in clinical screening of AHT cases.
Collapse
Affiliation(s)
- Kim Wiskott
- Forensic medicine unit, University Center of Legal Medicine, Geneva 4, Switzerland
| | - Federica Gilardi
- Faculty Unit of Toxicology, University Center of Legal Medicine, Lausanne University Hospital, Lausanne 25, Switzerland.,Unit of Forensic Toxicology and Chemistry, CURML, Lausanne University Hospital and Geneva University Hospital, Geneva, Switzerland
| | - Alexandre Hainard
- Proteomics Core Facility, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jean-Charles Sanchez
- Translational Biomarker Group, Department of Internal Medicine, University of Geneva, Geneva, Switzerland
| | - Aurelien Thomas
- Faculty Unit of Toxicology, University Center of Legal Medicine, Lausanne University Hospital, Lausanne 25, Switzerland.,Unit of Forensic Toxicology and Chemistry, CURML, Lausanne University Hospital and Geneva University Hospital, Geneva, Switzerland
| | - Tatjana Sajic
- Faculty Unit of Toxicology, University Center of Legal Medicine, Lausanne University Hospital, Lausanne 25, Switzerland.,Unit of Forensic Toxicology and Chemistry, CURML, Lausanne University Hospital and Geneva University Hospital, Geneva, Switzerland
| | - Tony Fracasso
- Forensic medicine unit, University Center of Legal Medicine, Geneva 4, Switzerland
| |
Collapse
|
5
|
Onate-Quiroz KV, Nwosu BU, Salemi P. Novel duplication of the cell adhesion molecule L1-like gene in an individual with cognitive impairment, tall stature, and obesity: A case report. Front Neurol 2023; 14:1104649. [PMID: 37114233 PMCID: PMC10126371 DOI: 10.3389/fneur.2023.1104649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 03/23/2023] [Indexed: 04/29/2023] Open
Abstract
The gene that codes for the close homolog of L1 (CHL1 gene) is located in the 3p26.3 cytogenetic band in the distal portion of the 3p chromosome. This gene is highly expressed in the central nervous system and plays an important role in brain formation and plasticity. Complete or partial CHL 1 gene-deficient mice have demonstrated neurocognitive deficits. In humans, mutations of the CHL 1 gene are infrequent with most mutations described in the literature as deletions. This case report describes an individual with a duplication in the CHL 1 and a presentation consistent with a syndromic form of neurocognitive impairment. To the best of our knowledge, this mutation has not been previously described in the literature.
Collapse
|
6
|
Yoo M, Kayastha N, Kwon O, Man W, Cai L, Schachner M. Analysis of the functional sequences in the promoter region of the human adhesion molecule close homolog of L1. Int J Neurosci 2022; 132:483-489. [PMID: 33054469 DOI: 10.1080/00207454.2020.1822357] [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: 03/23/2020] [Revised: 07/13/2020] [Accepted: 09/04/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Close Homolog of L1 (CHL1) is a member of the L1 family of cell adhesion molecules. CHL1 gene is located on human chromosome 3 and has been linked to several pathologies, including 3p deletion syndrome, schizophrenia, and tumor growth and metastasis. OBJECTIVE The goal of the present study was to determine which region of the CHL1 promoter is most competent in driving CHL1 gene expression. Methods: Five candidate DNA fragments in the promoter regions were selected by screening across six species for evolutionary conserved sequences. The activity of these five promoter regions was quantitatively evaluated using a GFP reporter gene in transfection experiments, performed in C6 glioma cells. RESULTS Of the five promoter regions tested, three drove reporter GFP expression, with the conserved region 6 (CR6, Gene ID AC066595.5, 25851-26850) being the most active for transcription. CONCLUSION The identification of the CR6 activity provides a better understanding of the regulatory mechanisms underlying CHL1 expression. It may help future discovery of therapeutic strategies that involve influencing critical promoter regions to drive transcriptional regulation of the mammalian CHL1 gene.HIGHLIGHTSConserved regions of CHL1 promoter sequences were identified by in-silico analysis.Five conserved regions were tested for gene regulatory activity using a reporter assay.Conserved regions CR5, CR6 and CR7 show gene regulatory function in a reporter assay.Co-transfection of CR5 and CR6 yielded the highest reporter activity.The core region of CR6 (CR6core) was identified as a cis-acting element.In-tandem promoter CR5core-CR6core was the best in a reporter assay.
Collapse
Affiliation(s)
- Myungsik Yoo
- W. M. Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Neha Kayastha
- W. M. Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Ohyoon Kwon
- W. M. Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Wai Man
- W. M. Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Li Cai
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Melitta Schachner
- W. M. Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
7
|
Loers G, Theis T, Baixia Hao H, Kleene R, Arsha S, Samuel N, Arsha N, Young W, Schachner M. Interplay in neural functions of cell adhesion molecule close homolog of L1 (CHL1) and Programmed Cell Death 6 (PDCD6). FASEB Bioadv 2022; 4:43-59. [PMID: 35024572 PMCID: PMC8728108 DOI: 10.1096/fba.2021-00027] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/16/2021] [Accepted: 07/27/2021] [Indexed: 11/11/2022] Open
Abstract
Close homolog of L1 (CHL1) is a cell adhesion molecule of the immunoglobulin superfamily. It promotes neuritogenesis and survival of neurons in vitro. In vivo, CHL1 promotes nervous system development, regeneration after trauma, and synaptic function and plasticity. We identified programmed cell death 6 (PDCD6) as a novel binding partner of the CHL1 intracellular domain (CHL1-ICD). Co-immunoprecipitation, pull-down assay with CHL1-ICD, and proximity ligation in cerebellum and pons of 3-day-old and 6-month-old mice, as well as in cultured cerebellar granule neurons and cortical astrocytes indicate an association between PDCD6 and CHL1. The Ca2+-chelator BAPTA-AM inhibited the association between CHL1 and PDCD6. The treatment of cerebellar granule neurons with a cell-penetrating peptide comprising the cell surface proximal 30 N-terminal amino acids of CHL1-ICD inhibited the association between CHL1 and PDCD6 and PDCD6- and CHL1-triggered neuronal survival. These results suggest that PDCD6 contributes to CHL1 functions in the nervous system.
Collapse
Affiliation(s)
- Gabriele Loers
- Zentrum für Molekulare NeurobiologieUniversitätsklinikum Hamburg‐EppendorfHamburgGermany
| | - Thomas Theis
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and NeuroscienceRutgers UniversityPiscatawayNJUSA
| | - Helen Baixia Hao
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and NeuroscienceRutgers UniversityPiscatawayNJUSA
| | - Ralf Kleene
- Zentrum für Molekulare NeurobiologieUniversitätsklinikum Hamburg‐EppendorfHamburgGermany
| | - Sanjana Arsha
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and NeuroscienceRutgers UniversityPiscatawayNJUSA
| | - Nina Samuel
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and NeuroscienceRutgers UniversityPiscatawayNJUSA
| | - Neha Arsha
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and NeuroscienceRutgers UniversityPiscatawayNJUSA
| | - Wise Young
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and NeuroscienceRutgers UniversityPiscatawayNJUSA
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and NeuroscienceRutgers UniversityPiscatawayNJUSA
| |
Collapse
|
8
|
Blazejewski SM, Bennison SA, Ha NT, Liu X, Smith TH, Dougherty KJ, Toyo-Oka K. Rpsa Signaling Regulates Cortical Neuronal Morphogenesis via Its Ligand, PEDF, and Plasma Membrane Interaction Partner, Itga6. Cereb Cortex 2021; 32:770-795. [PMID: 34347028 DOI: 10.1093/cercor/bhab242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 06/25/2021] [Accepted: 06/25/2021] [Indexed: 12/25/2022] Open
Abstract
Neuromorphological defects underlie neurodevelopmental disorders and functional defects. We identified a function for Rpsa in regulating neuromorphogenesis using in utero electroporation to knockdown Rpsa, resulting in apical dendrite misorientation, fewer/shorter extensions, and decreased spine density with altered spine morphology in upper neuronal layers and decreased arborization in upper/lower cortical layers. Rpsa knockdown disrupts multiple aspects of cortical development, including radial glial cell fiber morphology and neuronal layering. We investigated Rpsa's ligand, PEDF, and interacting partner on the plasma membrane, Itga6. Rpsa, PEDF, and Itga6 knockdown cause similar phenotypes, with Rpsa and Itga6 overexpression rescuing morphological defects in PEDF-deficient neurons in vivo. Additionally, Itga6 overexpression increases and stabilizes Rpsa expression on the plasma membrane. GCaMP6s was used to functionally analyze Rpsa knockdown via ex vivo calcium imaging. Rpsa-deficient neurons showed less fluctuation in fluorescence intensity, suggesting defective subthreshold calcium signaling. The Serpinf1 gene coding for PEDF is localized at chromosome 17p13.3, which is deleted in patients with the neurodevelopmental disorder Miller-Dieker syndrome. Our study identifies a role for Rpsa in early cortical development and for PEDF-Rpsa-Itga6 signaling in neuromorphogenesis, thus implicating these molecules in the etiology of neurodevelopmental disorders like Miller-Dieker syndrome and identifying them as potential therapeutics.
Collapse
Affiliation(s)
- Sara M Blazejewski
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Sarah A Bennison
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Ngoc T Ha
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Xiaonan Liu
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Trevor H Smith
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Kimberly J Dougherty
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Kazuhito Toyo-Oka
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| |
Collapse
|
9
|
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
|
10
|
Epifanova E, Salina V, Lajkó D, Textoris-Taube K, Naumann T, Bormuth O, Bormuth I, Horan S, Schaub T, Borisova E, Ambrozkiewicz MC, Tarabykin V, Rosário M. Adhesion dynamics in the neocortex determine the start of migration and the post-migratory orientation of neurons. SCIENCE ADVANCES 2021; 7:eabf1973. [PMID: 34215578 PMCID: PMC11060048 DOI: 10.1126/sciadv.abf1973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 05/19/2021] [Indexed: 06/13/2023]
Abstract
The neocortex is stereotypically organized into layers of excitatory neurons arranged in a precise parallel orientation. Here we show that dynamic adhesion both preceding and following radial migration is essential for this organization. Neuronal adhesion is regulated by the Mowat-Wilson syndrome-associated transcription factor Zeb2 (Sip1/Zfhx1b) through direct repression of independent adhesion pathways controlled by Neuropilin-1 (Nrp1) and Cadherin-6 (Cdh6). We reveal that to initiate radial migration, neurons must first suppress adhesion to the extracellular matrix. Zeb2 regulates the multipolar stage by transcriptional repression of Nrp1 and thereby downstream inhibition of integrin signaling. Upon completion of migration, neurons undergo an orientation process that is independent of migration. The parallel organization of neurons within the neocortex is controlled by Cdh6 through atypical regulation of integrin signaling via its RGD motif. Our data shed light on the mechanisms that regulate initiation of radial migration and the postmigratory orientation of neurons during neocortical development.
Collapse
Affiliation(s)
- Ekaterina Epifanova
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Cell and Neurobiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Valentina Salina
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Cell and Neurobiology, Charitéplatz 1, 10117 Berlin, Germany
- Institute of Neuroscience, Lobachevsky University of Nizhny Novgorod, Nizhny Novgorod 603950, Russian Federation
| | - Denis Lajkó
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Cell and Neurobiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Kathrin Textoris-Taube
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Biochemistry, Core Facility High-Throughput Mass Spectrometry, Charitéplatz 1, 10117 Berlin, Germany
| | - Thomas Naumann
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Functional Neuroanatomy, Charitéplatz 1, 10117 Berlin, Germany
| | - Olga Bormuth
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Cell and Neurobiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Ingo Bormuth
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Cell and Neurobiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Stephen Horan
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Cell and Neurobiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Theres Schaub
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Cell and Neurobiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Ekaterina Borisova
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Cell and Neurobiology, Charitéplatz 1, 10117 Berlin, Germany
- Institute of Neuroscience, Lobachevsky University of Nizhny Novgorod, Nizhny Novgorod 603950, Russian Federation
| | - Mateusz C Ambrozkiewicz
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Cell and Neurobiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Victor Tarabykin
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Cell and Neurobiology, Charitéplatz 1, 10117 Berlin, Germany
- Institute of Neuroscience, Lobachevsky University of Nizhny Novgorod, Nizhny Novgorod 603950, Russian Federation
| | - Marta Rosário
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Cell and Neurobiology, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
11
|
Gandawijaya J, Bamford RA, Burbach JPH, Oguro-Ando A. Cell Adhesion Molecules Involved in Neurodevelopmental Pathways Implicated in 3p-Deletion Syndrome and Autism Spectrum Disorder. Front Cell Neurosci 2021; 14:611379. [PMID: 33519384 PMCID: PMC7838543 DOI: 10.3389/fncel.2020.611379] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/15/2020] [Indexed: 01/06/2023] Open
Abstract
Autism spectrum disorder (ASD) is characterized by impaired social interaction, language delay and repetitive or restrictive behaviors. With increasing prevalence, ASD is currently estimated to affect 0.5–2.0% of the global population. However, its etiology remains unclear due to high genetic and phenotypic heterogeneity. Copy number variations (CNVs) are implicated in several forms of syndromic ASD and have been demonstrated to contribute toward ASD development by altering gene dosage and expression. Increasing evidence points toward the p-arm of chromosome 3 (chromosome 3p) as an ASD risk locus. Deletions occurring at chromosome 3p result in 3p-deletion syndrome (Del3p), a rare genetic disorder characterized by developmental delay, intellectual disability, facial dysmorphisms and often, ASD or ASD-associated behaviors. Therefore, we hypothesize that overlapping molecular mechanisms underlie the pathogenesis of Del3p and ASD. To investigate which genes encoded in chromosome 3p could contribute toward Del3p and ASD, we performed a comprehensive literature review and collated reports investigating the phenotypes of individuals with chromosome 3p CNVs. We observe that high frequencies of CNVs occur in the 3p26.3 region, the terminal cytoband of chromosome 3p. This suggests that CNVs disrupting genes encoded within the 3p26.3 region are likely to contribute toward the neurodevelopmental phenotypes observed in individuals affected by Del3p. The 3p26.3 region contains three consecutive genes encoding closely related neuronal immunoglobulin cell adhesion molecules (IgCAMs): Close Homolog of L1 (CHL1), Contactin-6 (CNTN6), and Contactin-4 (CNTN4). CNVs disrupting these neuronal IgCAMs may contribute toward ASD phenotypes as they have been associated with key roles in neurodevelopment. CHL1, CNTN6, and CNTN4 have been observed to promote neurogenesis and neuronal survival, and regulate neuritogenesis and synaptic function. Furthermore, there is evidence that these neuronal IgCAMs possess overlapping interactomes and participate in common signaling pathways regulating axon guidance. Notably, mouse models deficient for these neuronal IgCAMs do not display strong deficits in axonal migration or behavioral phenotypes, which is in contrast to the pronounced defects in neuritogenesis and axon guidance observed in vitro. This suggests that when CHL1, CNTN6, or CNTN4 function is disrupted by CNVs, other neuronal IgCAMs may suppress behavioral phenotypes by compensating for the loss of function.
Collapse
Affiliation(s)
- Josan Gandawijaya
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Rosemary A Bamford
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - J Peter H Burbach
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Asami Oguro-Ando
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
12
|
Lin WW, Ou GY, Lin JZ, Yi SJ, Yao WC, Pan HC, Zhao WJ. Neuregulin 1 enhances cell adhesion molecule L1 like expression levels and promotes malignancy in human glioma. Oncol Lett 2020; 20:326-336. [PMID: 32565959 PMCID: PMC7285836 DOI: 10.3892/ol.2020.11548] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 02/28/2020] [Indexed: 02/05/2023] Open
Abstract
Neural cell adhesion molecular L1-like protein (CHL1) is a member of the cell adhesion molecule L1 family and serves an important role in the development and progression of tumors. The cytokine neuregulin 1 (NRG1) has been indicated in the tumorigenesis and promotion of metastasis through the modulation of L1. However, the roles of NRG1 in regulating CHL1 in glioma have not been elucidated. The present study investigated the protein expression levels and roles of CHL1 and the possible correlation between NRG1 and CHL1 protein expression levels in human gliomas, both in vivo and in vitro. Using immunohistochemistry coupled with a human glioma tissue microarray, it was demonstrated that the percentage of CHL1-positive areas was the highest in grade II glioma tissues. Using immunofluorescence staining, a positive correlation was identified between the expression levels of CHL1 and proliferating cell nuclear antigen. In addition, CHL1 downregulation also resulted in increased senescence of U-87 MG human glioblastoma cells. In vitro, administration of NRG1α induced a significant increase in CHL1 protein expression levels in human glioma SHG-44 and U251 cells and in human glioblastoma U-87 MG cells, whereas NRG1β failed to increase CHL1 expression levels in U251 cells. These findings were further confirmed by the downregulation of NRG1 expression levels using small interfering RNA treatment, which resulted in the reduction of CHL1 protein expression levels in U-87 MG cells. These data indicate that NRG1 can regulate CHL1 protein expression levels in gliomas, that it is correlated with malignancy, and that NRG1 may contribute to malignancy by upregulating CHL1 protein expression levels in glioma/glioblastoma cells.
Collapse
Affiliation(s)
- Wen-Wen Lin
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Guan-Yong Ou
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Jia-Zhe Lin
- Neurosurgical Department, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - San-Jun Yi
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Wei-Cheng Yao
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Hong-Chao Pan
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Wei-Jiang Zhao
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, P.R. China
- Correspondence to: Professor Wei-Jiang Zhao, Cell Biology Department, Wuxi School of Medicine, Jiangnan University, 1800 Lihu Dadao Road, Wuxi, Jiangsu 214122, P.R. China, E-mail:
| |
Collapse
|
13
|
Duchateau A, de Thonel A, El Fatimy R, Dubreuil V, Mezger V. The "HSF connection": Pleiotropic regulation and activities of Heat Shock Factors shape pathophysiological brain development. Neurosci Lett 2020; 725:134895. [PMID: 32147500 DOI: 10.1016/j.neulet.2020.134895] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 02/29/2020] [Accepted: 03/04/2020] [Indexed: 12/21/2022]
Abstract
The Heat Shock Factors (HSFs) have been historically identified as a family of transcription factors that are activated and work in a stress-responsive manner, after exposure to a large variety of stimuli. However, they are also critical in normal conditions, in a life long manner, in a number of physiological processes that encompass gametogenesis, embryonic development and the integrity of adult organs and organisms. The importance of such roles is emphasized by the devastating impact of their deregulation on health, ranging from reproductive failure, neurodevelopmental disorders, cancer, and aging pathologies, including neurodegenerative disorders. Here, we provide an overview of the delicate choreography of the regulation of HSFs during neurodevelopment, at prenatal and postnatal stages. The regulation of HSFs acts at multiple layers and steps, and comprises the control of (i) HSF mRNA and protein levels, (ii) HSF activity in terms of DNA-binding and transcription, (iii) HSF homo- and hetero-oligomerization capacities, and (iv) HSF combinatory set of post-translational modifications. We also describe how these regulatory mechanisms operate in the normal developing brain and how their perturbation impact neurodevelopment under prenatal or perinatal stress conditions. In addition, we put into perspective the possible role of HSFs in the evolution of the vertebrate brains and the importance of the HSF pathway in a large variety of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Agathe Duchateau
- Université de Paris, Epigenetics and Cell Fate, CNRS, F-75013, Paris, France; Département Hospitalo-Universitaire DHU PROTECT, Paris, France; ED 562 BioSPC, Université de Paris, F-75205, Paris Cedex 13, France
| | - Aurélie de Thonel
- Université de Paris, Epigenetics and Cell Fate, CNRS, F-75013, Paris, France; Département Hospitalo-Universitaire DHU PROTECT, Paris, France
| | - Rachid El Fatimy
- Université de Paris, Epigenetics and Cell Fate, CNRS, F-75013, Paris, France; Département Hospitalo-Universitaire DHU PROTECT, Paris, France
| | - Véronique Dubreuil
- Université de Paris, Epigenetics and Cell Fate, CNRS, F-75013, Paris, France; Département Hospitalo-Universitaire DHU PROTECT, Paris, France
| | - Valérie Mezger
- Université de Paris, Epigenetics and Cell Fate, CNRS, F-75013, Paris, France; Département Hospitalo-Universitaire DHU PROTECT, Paris, France.
| |
Collapse
|
14
|
Parcerisas A, Pujadas L, Ortega-Gascó A, Perelló-Amorós B, Viais R, Hino K, Figueiro-Silva J, La Torre A, Trullás R, Simó S, Lüders J, Soriano E. NCAM2 Regulates Dendritic and Axonal Differentiation through the Cytoskeletal Proteins MAP2 and 14-3-3. Cereb Cortex 2020; 30:3781-3799. [PMID: 32043120 DOI: 10.1093/cercor/bhz342] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/27/2019] [Accepted: 01/08/2020] [Indexed: 01/13/2023] Open
Abstract
Neural cell adhesion molecule 2 (NCAM2) is involved in the development and plasticity of the olfactory system. Genetic data have implicated the NCAM2 gene in neurodevelopmental disorders including Down syndrome and autism, although its role in cortical development is unknown. Here, we show that while overexpression of NCAM2 in hippocampal neurons leads to minor alterations, its downregulation severely compromises dendritic architecture, leading to an aberrant phenotype including shorter dendritic trees, retraction of dendrites, and emergence of numerous somatic neurites. Further, our data reveal alterations in the axonal tree and deficits in neuronal polarization. In vivo studies confirm the phenotype and reveal an unexpected role for NCAM2 in cortical migration. Proteomic and cell biology experiments show that NCAM2 molecules exert their functions through a protein complex with the cytoskeletal-associated proteins MAP2 and 14-3-3γ and ζ. We provide evidence that NCAM2 depletion results in destabilization of the microtubular network and reduced MAP2 signal. Our results demonstrate a role for NCAM2 in dendritic formation and maintenance, and in neural polarization and migration, through interaction of NCAM2 with microtubule-associated proteins.
Collapse
Affiliation(s)
- Antoni Parcerisas
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.,Vall d'Hebron Institut de Recerca (VHIR), 08035, Barcelona, Spain
| | - Lluís Pujadas
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.,Vall d'Hebron Institut de Recerca (VHIR), 08035, Barcelona, Spain
| | - Alba Ortega-Gascó
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.,Vall d'Hebron Institut de Recerca (VHIR), 08035, Barcelona, Spain
| | - Bartomeu Perelló-Amorós
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.,Vall d'Hebron Institut de Recerca (VHIR), 08035, Barcelona, Spain
| | - Ricardo Viais
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), 08028, Barcelona, Spain
| | - Keiko Hino
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA 95616, USA
| | - Joana Figueiro-Silva
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.,Neurobiology Unit, Institut d'Investigacions Biomèdiques de Barcelona, CSIC, IDIBAPS, 08036, Barcelona, Spain
| | - Anna La Torre
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA 95616, USA
| | - Ramón Trullás
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.,Neurobiology Unit, Institut d'Investigacions Biomèdiques de Barcelona, CSIC, IDIBAPS, 08036, Barcelona, Spain
| | - Sergi Simó
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA 95616, USA
| | - Jens Lüders
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), 08028, Barcelona, Spain
| | - Eduardo Soriano
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.,Vall d'Hebron Institut de Recerca (VHIR), 08035, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA) Academia, 08010, Barcelona, Spain
| |
Collapse
|
15
|
Mohan V, Wade SD, Sullivan CS, Kasten MR, Sweetman C, Stewart R, Truong Y, Schachner M, Manis PB, Maness PF. Close Homolog of L1 Regulates Dendritic Spine Density in the Mouse Cerebral Cortex Through Semaphorin 3B. J Neurosci 2019; 39:6233-6250. [PMID: 31182634 PMCID: PMC6687901 DOI: 10.1523/jneurosci.2984-18.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 04/30/2019] [Accepted: 05/30/2019] [Indexed: 02/05/2023] Open
Abstract
Dendritic spines in the developing mammalian neocortex are initially overproduced and then eliminated during adolescence to achieve appropriate levels of excitation in mature networks. We show here that the L1 family cell adhesion molecule Close Homolog of L1 (CHL1) and secreted repellent ligand Semaphorin 3B (Sema3B) function together to induce dendritic spine pruning in developing cortical pyramidal neurons. Loss of CHL1 in null mutant mice in both genders resulted in increased spine density and a greater proportion of immature spines on apical dendrites in the prefrontal and visual cortex. Electron microscopy showed that excitatory spine synapses with postsynaptic densities were increased in the CHL1-null cortex, and electrophysiological recording in prefrontal slices from mutant mice revealed deficiencies in excitatory synaptic transmission. Mechanistically, Sema3B protein induced elimination of spines on apical dendrites of cortical neurons cultured from wild-type but not CHL1-null embryos. Sema3B was secreted by the cortical neuron cultures, and its levels increased when cells were treated with the GABA antagonist gabazine. In vivo CHL1 was coexpressed with Sema3B in pyramidal neuron subpopulations and formed a complex with Sema3B receptor subunits Neuropilin-2 and PlexinA4. CHL1 and NrCAM, a closely related L1 adhesion molecule, localized primarily to distinct spines and promoted spine elimination to Sema3B or Sema3F, respectively. These results support a new concept in which selective spine elimination is achieved through different secreted semaphorins and L1 family adhesion molecules to sculpt functional neural circuits during postnatal maturation.SIGNIFICANCE STATEMENT Dendritic spines in the mammalian neocortex are initially overproduced and then pruned in adolescent life through unclear mechanisms to sculpt maturing cortical circuits. Here, we show that spine and excitatory synapse density of pyramidal neurons in the developing neocortex is regulated by the L1 adhesion molecule, Close Homolog of L1 (CHL1). CHL1 mediated spine pruning in response to the secreted repellent ligand Semaphorin 3B and associated with receptor subunits Neuropilin-2 and PlexinA4. CHL1 and related L1 adhesion molecule NrCAM localized to distinct spines, and promoted spine elimination to Semaphorin 3B and -3F, respectively. These results support a new concept in which selective elimination of individual spines and nascent synapses can be achieved through the action of distinct secreted semaphorins and L1 adhesion molecules.
Collapse
Affiliation(s)
| | | | | | - Michael R Kasten
- Department of Otolaryngology/Head and Neck Surgery
- Department of Cell Biology and Physiology
| | | | | | - Young Truong
- Department of Biostatistics, School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey 08854, and
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Paul B Manis
- Department of Otolaryngology/Head and Neck Surgery
- Department of Cell Biology and Physiology
| | | |
Collapse
|
16
|
Kotani N, Yamaguchi A, Ohnishi T, Kuwahara R, Nakano T, Nakano Y, Ida Y, Murakoshi T, Honke K. Proximity proteomics identifies cancer cell membrane cis-molecular complex as a potential cancer target. Cancer Sci 2019; 110:2607-2619. [PMID: 31228215 PMCID: PMC6676139 DOI: 10.1111/cas.14108] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/10/2019] [Accepted: 06/15/2019] [Indexed: 12/22/2022] Open
Abstract
Cancer‐specific antigens expressed in the cell membrane have been used as targets for several molecular targeted strategies in the last 20 years with remarkable success. To develop more effective cancer treatments, novel targets and strategies for targeted therapies are needed. Here, we examined the cancer cell membrane‐resident “cis‐bimolecular complex” as a possible cancer target (cis‐bimolecular cancer target: BiCAT) using proximity proteomics, a technique that has attracted attention in the last 10 years. BiCAT were detected using a previously developed method termed the enzyme‐mediated activation of radical source (EMARS), to label the components proximal to a given cell membrane molecule. EMARS analysis identified some BiCAT, such as close homolog of L1 (CHL1), fibroblast growth factor 3 (FGFR3) and α2 integrin, which are commonly expressed in mouse primary lung cancer cells and human lung squamous cell carcinoma cells. Analysis of cancer specimens from 55 lung cancer patients revealed that CHL1 and α2 integrin were highly co–expressed in almost all cancer tissues compared with normal lung tissues. As an example of BiCAT application, in vitro simulation of effective drug combinations used for multiple drug treatment strategies was performed using reagents targeted to BiCAT molecules. The combination treatment based on BiCAT information moderately suppressed cancer cell proliferation compared with single administration, suggesting that the information about BiCAT in cancer cells is useful for the appropriate selection of the combination among molecular targeted reagents. Thus, BiCAT has the potential to contribute to several molecular targeted strategies in future.
Collapse
Affiliation(s)
- Norihiro Kotani
- Department of Biochemistry, Saitama Medical University, Saitama, Japan
| | - Arisa Yamaguchi
- Department of Biochemistry, Kochi University Medical School, Kochi, Japan
| | - Tomoko Ohnishi
- Department of Biochemistry, Kochi University Medical School, Kochi, Japan
| | - Ryusuke Kuwahara
- Quantum Wave Microscopy Unit, Okinawa Institute of Science and Technology Graduate University (OIST), Okinawa, Japan
| | - Takanari Nakano
- Department of Biochemistry, Saitama Medical University, Saitama, Japan
| | - Yuka Nakano
- Department of Biochemistry, Saitama Medical University, Saitama, Japan
| | - Yui Ida
- Department of Biochemistry, Saitama Medical University, Saitama, Japan
| | | | - Koichi Honke
- Department of Biochemistry, Kochi University Medical School, Kochi, Japan
| |
Collapse
|
17
|
Yang WK, Chueh YR, Cheng YJ, Siegenthaler D, Pielage J, Chien CT. Epidermis-Derived L1CAM Homolog Neuroglian Mediates Dendrite Enclosure and Blocks Heteroneuronal Dendrite Bundling. Curr Biol 2019; 29:1445-1459.e3. [PMID: 31006568 DOI: 10.1016/j.cub.2019.03.050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 01/31/2019] [Accepted: 03/23/2019] [Indexed: 12/31/2022]
Abstract
Building sensory dendritic arbors requires branching, growth, spacing, and substrate support. The conserved L1CAM family of cell-adhesion molecules generates neuronal isoforms to regulate neurite development in various aspects. However, whether non-neuronal isoforms participate in any of these aspects is unclear. In Drosophila, the L1CAM homolog Neuroglian (Nrg) is expressed as two isoforms: the neuronal isoform Nrg180 on dendritic surfaces of dendritic arborization (da) neurons and the non-neuronal isoform Nrg167 in epidermis innervated by dendrites. We found that epidermal Nrg167 encircles dendrites by interactions with dendritic Nrg180 to support dendrite growth, stabilization, and enclosure inside epidermis. Interestingly, whereas Nrg180 forms homophilic interactions to facilitate axonal bundling, heteroneuronal dendrites in the same innervating field avoid bundling through unknown mechanisms to maintain individual dendritic patterns. Here, we show that both epidermal Nrg167 depletion and neuronal Nrg180 overexpression can cause dendrite bundling, with genetic analyses suggesting that Nrg167-Nrg180 interactions antagonize Nrg180-Nrg180 homophilic interaction to prevent dendrite bundling. Furthermore, internalization of Nrg180 also participates in resolving dendrite bundling, as overexpression of endocytosis-defective Nrg180 and compromising endocytosis in neurons both exacerbated dendrite-bundling defects. Together, our study highlights the functional significance of substrate-derived Nrg167 in conferring dendrite stability, positioning, and avoidance.
Collapse
Affiliation(s)
- Wei-Kang Yang
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| | - Yi-Ru Chueh
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| | - Ying-Ju Cheng
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| | - Dominique Siegenthaler
- Department of Zoology and Neurobiology, University of Kaiserslautern, 67663 Kaiserslautern, Germany
| | - Jan Pielage
- Department of Zoology and Neurobiology, University of Kaiserslautern, 67663 Kaiserslautern, Germany
| | - Cheng-Ting Chien
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan.
| |
Collapse
|
18
|
Ou-Yang MH, Kurz JE, Nomura T, Popovic J, Rajapaksha TW, Dong H, Contractor A, Chetkovich DM, Tourtellotte WG, Vassar R. Axonal organization defects in the hippocampus of adult conditional BACE1 knockout mice. Sci Transl Med 2018; 10:eaao5620. [PMID: 30232227 PMCID: PMC11017370 DOI: 10.1126/scitranslmed.aao5620] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 03/09/2018] [Accepted: 05/21/2018] [Indexed: 12/14/2022]
Abstract
β-Site APP (amyloid precursor protein) cleaving enzyme 1 (BACE1) is the β-secretase enzyme that initiates production of the toxic amyloid-β peptide that accumulates in the brains of patients with Alzheimer's disease (AD). Hence, BACE1 is a prime therapeutic target, and several BACE1 inhibitor drugs are currently being tested in clinical trials for AD. However, the safety of BACE1 inhibition is unclear. Germline BACE1 knockout mice have multiple neurological phenotypes, although these could arise from BACE1 deficiency during development. To address this question, we report that tamoxifen-inducible conditional BACE1 knockout mice in which the Bace1 gene was ablated in the adult largely lacked the phenotypes observed in germline BACE1 knockout mice. However, one BACE1-null phenotype was induced after Bace1 gene deletion in the adult mouse brain. This phenotype showed reduced length and disorganization of the hippocampal mossy fiber infrapyramidal bundle, the axonal pathway of dentate gyrus granule cells that is maintained by neurogenesis in the mouse brain. This defect in axonal organization correlated with reduced BACE1-mediated cleavage of the neural cell adhesion protein close homolog of L1 (CHL1), which has previously been associated with axon guidance. Although our results indicate that BACE1 inhibition in the adult mouse brain may avoid phenotypes associated with BACE1 deficiency during embryonic and postnatal development, they also suggest that BACE1 inhibitor drugs developed for treating AD may disrupt the organization of an axonal pathway in the hippocampus, an important structure for learning and memory.
Collapse
Affiliation(s)
- Ming-Hsuan Ou-Yang
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jonathan E Kurz
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Toshihiro Nomura
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL 60208, USA
| | - Jelena Popovic
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Tharinda W Rajapaksha
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Anis Contractor
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL 60208, USA
| | - Dane M Chetkovich
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Neurology and Clinical Neurosciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Warren G Tourtellotte
- Department of Neurology and Clinical Neurosciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Division of Neuropathology, Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Robert Vassar
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
19
|
Kotani N, Ida Y, Nakano T, Sato I, Kuwahara R, Yamaguchi A, Tomita M, Honke K, Murakoshi T. Tumor-dependent secretion of close homolog of L1 results in elevation of its circulating level in mouse model for human lung tumor. Biochem Biophys Res Commun 2018; 501:982-987. [DOI: 10.1016/j.bbrc.2018.05.096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 05/14/2018] [Indexed: 01/01/2023]
|
20
|
Vulovic M, Divac N, Jakovcevski I. Confocal Synaptology: Synaptic Rearrangements in Neurodegenerative Disorders and upon Nervous System Injury. Front Neuroanat 2018; 12:11. [PMID: 29497366 PMCID: PMC5818405 DOI: 10.3389/fnana.2018.00011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 02/01/2018] [Indexed: 01/26/2023] Open
Abstract
The nervous system is a notable exception to the rule that the cell is the structural and functional unit of tissue systems and organs. The functional unit of the nervous system is the synapse, the contact between two nerve cells. As such, synapses are the foci of investigations of nervous system organization and function, as well as a potential readout for the progression of various disorders of the nervous system. In the past decade the development of antibodies specific to presynaptic terminals has enabled us to assess, at the optical, laser scanning microscopy level, these subcellular structures, and has provided a simple method for the quantification of various synapses. Indeed, excitatory (glutamatergic) and inhibitory synapses can be visualized using antibodies against the respective vesicular transporters, and choline-acetyl transferase (ChAT) immunoreactivity identifies cholinergic synapses throughout the central nervous system. Here we review the results of several studies in which these methods were used to estimate synaptic numbers as the structural equivalent of functional outcome measures in spinal cord and femoral nerve injuries, as well as in genetic mouse models of neurodegeneration, including Alzheimer's disease (AD). The results implicate disease- and brain region-specific changes in specific types of synapses, which correlate well with the degree of functional deficit caused by the disease process. Additionally, results are reproducible between various studies and experimental paradigms, supporting the reliability of the method. To conclude, this quantitative approach enables fast and reliable estimation of the degree of the progression of neurodegenerative changes and can be used as a parameter of recovery in experimental models.
Collapse
Affiliation(s)
- Maja Vulovic
- Department of Anatomy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nevena Divac
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Igor Jakovcevski
- Institute for Molecular and Behavioral Neuroscience, University Hospital Cologne, Center for Molecular Medicine Cologne, Cologne, Germany.,Experimental Neurophysiology, German Center for Neurodegenerative Diseases, Bonn, Germany
| |
Collapse
|
21
|
Super-Resolution Track-Density Imaging Reveals Fine Anatomical Features in Tree Shrew Primary Visual Cortex and Hippocampus. Neurosci Bull 2017; 34:438-448. [PMID: 29247318 DOI: 10.1007/s12264-017-0199-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/07/2017] [Indexed: 12/21/2022] Open
Abstract
Diffusion-weighted magnetic resonance imaging (dMRI) is widely used to study white and gray matter (GM) micro-organization and structural connectivity in the brain. Super-resolution track-density imaging (TDI) is an image reconstruction method for dMRI data, which is capable of providing spatial resolution beyond the acquired data, as well as novel and meaningful anatomical contrast that cannot be obtained with conventional reconstruction methods. TDI has been used to reveal anatomical features in human and animal brains. In this study, we used short track TDI (stTDI), a variation of TDI with enhanced contrast for GM structures, to reconstruct direction-encoded color maps of fixed tree shrew brain. The results were compared with those obtained with the traditional diffusion tensor imaging (DTI) method. We demonstrated that fine microstructures in the tree shrew brain, such as Baillarger bands in the primary visual cortex and the longitudinal component of the mossy fibers within the hippocampal CA3 subfield, were observable with stTDI, but not with DTI reconstructions from the same dMRI data. The possible mechanisms underlying the enhanced GM contrast are discussed.
Collapse
|
22
|
Oved K, Farberov L, Gilam A, Israel I, Haguel D, Gurwitz D, Shomron N. MicroRNA-Mediated Regulation of ITGB3 and CHL1 Is Implicated in SSRI Action. Front Mol Neurosci 2017; 10:355. [PMID: 29163031 PMCID: PMC5682014 DOI: 10.3389/fnmol.2017.00355] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 10/18/2017] [Indexed: 01/05/2023] Open
Abstract
Background: Selective serotonin reuptake inhibitor (SSRI) antidepressant drugs are the first-line of treatment for major depressive disorder (MDD) but are effective in <70% of patients. Our earlier genome-wide studies indicated that two genes encoding for cell adhesion proteins, close homolog of L1 (CHL1) and integrin beta-3 (ITGB3), and microRNAs, miR-151a-3p and miR-221/222, are implicated in the variable sensitivity and response of human lymphoblastoid cell lines (LCL) from unrelated individuals to SSRI drugs. Methods: The microRNAs miR-221, miR-222, and miR-151-a-3p, along with their target gene binding sites, were explored in silico using miRBase, TargetScan, microRNAviewer, and the UCSC Genome Browser. Luciferase reporter assays were conducted for demonstrating the direct functional regulation of ITGB3 and CHL1 expression by miR-221/222 and miR-151a-3p, respectively. A human LCL exhibiting low sensitivity to paroxetine was utilized for studying the phenotypic effect of CHL1 regulation by miR-151a-3p on SSRI response. Results: By showing direct regulation of CHL1 and ITGB3 by miR-151a-3p and miR-221/222, respectively, we link these microRNAs and genes with cellular SSRI sensitivity phenotypes. We report that miR-151a-3p increases cell sensitivity to paroxetine via down-regulating CHL1 expression. Conclusions: miR-151a-3p, miR-221/222 and their (here confirmed) respective target-genes, CHL1 and ITGB3, are implicated in SSRI responsiveness, and possibly in the clinical response to antidepressant drugs.
Collapse
Affiliation(s)
- Keren Oved
- Faculty of Medicine, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Luba Farberov
- Faculty of Medicine, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Avial Gilam
- Faculty of Medicine, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Ifat Israel
- Faculty of Medicine, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Danielle Haguel
- Faculty of Medicine, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - David Gurwitz
- Faculty of Medicine, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Noam Shomron
- Faculty of Medicine, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
23
|
Homophilic binding of the neural cell adhesion molecule CHL1 regulates development of ventral midbrain dopaminergic pathways. Sci Rep 2017; 7:9368. [PMID: 28839197 PMCID: PMC5570898 DOI: 10.1038/s41598-017-09599-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 07/26/2017] [Indexed: 11/09/2022] Open
Abstract
Abnormal development of ventral midbrain (VM) dopaminergic (DA) pathways, essential for motor and cognitive function, may underpin a number of neurological disorders and thereby highlight the importance of understanding the birth and connectivity of the associated neurons. While a number of regulators of VM DA neurogenesis are known, processes involved in later developmental events, including terminal differentiation and axon morphogenesis, are less well understood. Recent transcriptional analysis studies of the developing VM identified genes expressed during these stages, including the cell adhesion molecule with homology to L1 (Chl1). Here, we map the temporal and spatial expression of CHL1 and assess functional roles of substrate-bound and soluble-forms of the protein during VM DA development. Results showed early CHL1 in the VM, corresponding with roles in DA progenitor migration and differentiation. Subsequently, we demonstrated roles for CHL1 in both axonal extension and repulsion, selectively of DA neurons, suggestive of a role in guidance towards forebrain targets and away from hindbrain nuclei. In part, CHL1 mediates these roles through homophilic CHL1-CHL1 interactions. Collectively, these findings enhance our knowledge of VM DA pathways development, and may provide new insights into understanding DA developmental conditions such as autism spectrum disorders.
Collapse
|
24
|
Buhusi M, Obray D, Guercio B, Bartlett MJ, Buhusi CV. Chronic mild stress impairs latent inhibition and induces region-specific neural activation in CHL1-deficient mice, a mouse model of schizophrenia. Behav Brain Res 2017. [PMID: 28647594 DOI: 10.1016/j.bbr.2017.06.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Schizophrenia is a neurodevelopmental disorder characterized by abnormal processing of information and attentional deficits. Schizophrenia has a high genetic component but is precipitated by environmental factors, as proposed by the 'two-hit' theory of schizophrenia. Here we compared latent inhibition as a measure of learning and attention, in CHL1-deficient mice, an animal model of schizophrenia, and their wild-type littermates, under no-stress and chronic mild stress conditions. All unstressed mice as well as the stressed wild-type mice showed latent inhibition. In contrast, CHL1-deficient mice did not show latent inhibition after exposure to chronic stress. Differences in neuronal activation (c-Fos-positive cell counts) were noted in brain regions associated with latent inhibition: Neuronal activation in the prelimbic/infralimbic cortices and the nucleus accumbens shell was affected solely by stress. Neuronal activation in basolateral amygdala and ventral hippocampus was affected independently by stress and genotype. Most importantly, neural activation in nucleus accumbens core was affected by the interaction between stress and genotype. These results provide strong support for a 'two-hit' (genes x environment) effect on latent inhibition in CHL1-deficient mice, and identify CHL1-deficient mice as a model of schizophrenia-like learning and attention impairments.
Collapse
Affiliation(s)
- Mona Buhusi
- Interdisciplinary Program in Neuroscience, USTAR BioInnovations Center, Dept. Psychology, Utah State University, Logan UT, United States.
| | - Daniel Obray
- Interdisciplinary Program in Neuroscience, USTAR BioInnovations Center, Dept. Psychology, Utah State University, Logan UT, United States
| | - Bret Guercio
- Interdisciplinary Program in Neuroscience, USTAR BioInnovations Center, Dept. Psychology, Utah State University, Logan UT, United States
| | - Mitchell J Bartlett
- Interdisciplinary Program in Neuroscience, USTAR BioInnovations Center, Dept. Psychology, Utah State University, Logan UT, United States
| | - Catalin V Buhusi
- Interdisciplinary Program in Neuroscience, USTAR BioInnovations Center, Dept. Psychology, Utah State University, Logan UT, United States
| |
Collapse
|
25
|
Katic J, Loers G, Tosic J, Schachner M, Kleene R. The cell adhesion molecule CHL1 interacts with patched-1 to regulate apoptosis during postnatal cerebellar development. J Cell Sci 2017. [PMID: 28630165 DOI: 10.1242/jcs.194563] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The immunoglobulin superfamily adhesion molecule close homolog of L1 (CHL1) plays important roles during nervous system development. Here, we identified the hedgehog receptor patched-1 (PTCH1) as a novel CHL1-binding protein and showed that CHL1 interacts with the first extracellular loop of PTCH1 via its extracellular domain. Colocalization and co-immunoprecipitation of CHL1 with PTCH1 suggest an association of CHL1 with this major component of the hedgehog signaling pathway. The trans-interaction of CHL1 with PTCH1 promotes neuronal survival in cultures of dissociated cerebellar granule cells and of organotypic cerebellar slices. An inhibitor of the PTCH1-regulated hedgehog signal transducer, smoothened (SMO), and inhibitors of RhoA and Rho-associated kinase (ROCK) 1 and 2 prevent CHL1-dependent survival of cultured cerebellar granule cells and survival of cerebellar granule and Purkinje cells in organotypic cultures. In histological sections from 10- and 14-day-old CHL1-deficient mice, enhanced apoptosis of granule, but not Purkinje, cells was observed. The results of the present study indicate that CHL1 triggers PTCH1-, SMO-, RhoA- and ROCK-dependent signal transduction pathways to promote neuronal survival after cessation of the major morphogenetic events during mouse cerebellar development.
Collapse
Affiliation(s)
- Jelena Katic
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Jelena Tosic
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA .,Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA.,Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong 515041, China
| | - Ralf Kleene
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| |
Collapse
|
26
|
Yang Z, Xie Q, Hu CL, Jiang Q, Shen HF, Schachner M, Zhao WJ. CHL1 Is Expressed and Functions as a Malignancy Promoter in Glioma Cells. Front Mol Neurosci 2017; 10:324. [PMID: 29089868 PMCID: PMC5650976 DOI: 10.3389/fnmol.2017.00324] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 09/25/2017] [Indexed: 02/06/2023] Open
Abstract
The cell adhesion molecule with homology to L1CAM (close homolog of L1) (CHL1) is a member of the cell adhesion molecule L1 (L1CAM) gene family. Although CHL1 expression and function have been reported in several tumors, the roles of CHL1 in the development of glioma remain unclear. In the present study, we investigated the effects of CHL1 on proliferation indexes and activation of Akt1 and Erk signaling by siRNA in U-87 MG human glioblastoma and human U251 and SHG-44 glioma cells. We found that siRNA targeting CHL1 significantly down-regulated the expression of CHL1 mRNA and protein accompanied by reduced cell proliferation and transmigration invasion in all three cell lines. Down-regulating CHL1 expression also reduced cell survival, as measured by the Bax/Bcl-2 ratio, and increased activation of caspase-3. In subcutaneous U-87 MG cell xenograft tumors in nude mice, intratumoral administration of siRNA targeting CHL1 treatment significantly down-regulated CHL1 expression in vivo, accompanied by increased levels of activated caspase-3. Our combined results confirmed for the first time that in contrast to findings about CHL1 in most other cancer types, CHL1 functions in promoting cell proliferation, metastasis and migration in human glioma cells both in vitro and in vivo. These results indicate that CHL1 is a therapeutic target in the clinical management of glioma/glioblastoma.
Collapse
Affiliation(s)
- Zhai Yang
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| | - Qing Xie
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| | - Cheng-Liang Hu
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| | - Qiong Jiang
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| | - Hui-Fan Shen
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, China
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- *Correspondence: Melitta Schachner Wei-Jiang Zhao
| | - Wei-Jiang Zhao
- Center for Neuroscience, Shantou University Medical College, Shantou, China
- *Correspondence: Melitta Schachner Wei-Jiang Zhao
| |
Collapse
|
27
|
Interaction between DISC1 and CHL1 in regulation of neurite outgrowth. Brain Res 2016; 1648:290-297. [DOI: 10.1016/j.brainres.2016.06.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 06/14/2016] [Accepted: 06/22/2016] [Indexed: 01/31/2023]
|
28
|
Singh S, Howell D, Trivedi N, Kessler K, Ong T, Rosmaninho P, Raposo AA, Robinson G, Roussel MF, Castro DS, Solecki DJ. Zeb1 controls neuron differentiation and germinal zone exit by a mesenchymal-epithelial-like transition. eLife 2016; 5. [PMID: 27178982 PMCID: PMC4891180 DOI: 10.7554/elife.12717] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 05/03/2016] [Indexed: 12/13/2022] Open
Abstract
In the developing mammalian brain, differentiating neurons mature morphologically via neuronal polarity programs. Despite discovery of polarity pathways acting concurrently with differentiation, it's unclear how neurons traverse complex polarity transitions or how neuronal progenitors delay polarization during development. We report that zinc finger and homeobox transcription factor-1 (Zeb1), a master regulator of epithelial polarity, controls neuronal differentiation by transcriptionally repressing polarity genes in neuronal progenitors. Necessity-sufficiency testing and functional target screening in cerebellar granule neuron progenitors (GNPs) reveal that Zeb1 inhibits polarization and retains progenitors in their germinal zone (GZ). Zeb1 expression is elevated in the Sonic Hedgehog (SHH) medulloblastoma subgroup originating from GNPs with persistent SHH activation. Restored polarity signaling promotes differentiation and rescues GZ exit, suggesting a model for future differentiative therapies. These results reveal unexpected parallels between neuronal differentiation and mesenchymal-to-epithelial transition and suggest that active polarity inhibition contributes to altered GZ exit in pediatric brain cancers. DOI:http://dx.doi.org/10.7554/eLife.12717.001 During the formation of the brain, developing neurons are faced with a logistical problem. After newborn neurons form they must change in shape and move to their final location in the brain. Despite much speculation, little is known about these processes. Neurons mature via the activity of several pathways that control the activity, or expression, of the neuron’s genes. One way of controlling such gene expression is through proteins called transcription factors. At the same time, the developing neurons go through a process called polarization, where different regions of the cell develop different characteristics. However, it was not known how the maturation and polarization processes are linked, or how the developing neurons actively regulate polarization. By studying the developing mouse brain, Singh et al. found that a transcription factor called Zeb1 keeps neurons in a immature state, stopping them from becoming polarized. Further investigation revealed that Zeb1 does this by preventing the production of a group of proteins that helps to polarize the cells. The most common type of malignant brain tumour in children is called a medulloblastoma. Singh et al. analyzed the genes expressed in mice that have a type of medulloblastoma that results from the constant activity of a gene called Sonic Hedgehog in developing neurons. This revealed that these tumour cells contain abnormally high levels of Zeb1, and so do not take on a polarized form. However, artificially restoring other factors that encourage the cells to polarize caused the neurons to mature normally. Further investigation is now needed to find out whether the activity of the Sonic Hedgehog gene regulates Zeb1 activity, and to discover whether inhibiting Zeb1 could prevent brain tumours from developing. DOI:http://dx.doi.org/10.7554/eLife.12717.002
Collapse
Affiliation(s)
- Shalini Singh
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, United States
| | - Danielle Howell
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, United States
| | - Niraj Trivedi
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, United States
| | | | - Taren Ong
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, United States
| | - Pedro Rosmaninho
- Department of Molecular Neurobiology, Instituto Gulbenkian de Ciência Oeiras, Oeiras, Portugal
| | - Alexandre Asf Raposo
- Department of Molecular Neurobiology, Instituto Gulbenkian de Ciência Oeiras, Oeiras, Portugal
| | - Giles Robinson
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, United States
| | - Martine F Roussel
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Diogo S Castro
- Department of Molecular Neurobiology, Instituto Gulbenkian de Ciência Oeiras, Oeiras, Portugal
| | - David J Solecki
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, United States
| |
Collapse
|
29
|
Zuko A, Oguro-Ando A, van Dijk R, Gregorio-Jordan S, van der Zwaag B, Burbach JPH. Developmental role of the cell adhesion molecule Contactin-6 in the cerebral cortex and hippocampus. Cell Adh Migr 2016; 10:378-92. [PMID: 26939565 DOI: 10.1080/19336918.2016.1155018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The gene encoding the neural cell adhesion molecule Contactin-6 (Cntn6 a.k.a. NB-3) has been implicated as an autism risk gene, suggesting that its mutation is deleterious to brain development. Due to its GPI-anchor at Cntn6 may exert cell adhesion/receptor functions in complex with other membrane proteins, or serve as a ligand. We aimed to uncover novel phenotypes related to Cntn6 functions during development in the cerebral cortex of adult Cntn6(-/-) mice. We first determined Cntn6 protein and mRNA expression in the cortex, thalamic nuclei and the hippocampus at P14, which decreased specifically in the cortex at adult stages. Neuroanatomical analysis demonstrated a significant decrease of Cux1+ projection neurons in layers II-IV and an increase of FoxP2+ projection neurons in layer VI in the visual cortex of adult Cntn6(-/-) mice compared to wild-type controls. Furthermore, the number of parvalbumin+ (PV) interneurons was decreased in Cntn6(-/-) mice, while the amount of NPY+ interneurons remained unchanged. In the hippocampus the delineation and outgrowth of mossy fibers remained largely unchanged, except for the observation of a larger suprapyramidal bundle. The observed abnormalities in the cerebral cortex and hippocampus of Cntn6(-/-) mice suggests that Cntn6 serves developmental functions involving cell survival, migration and fasciculation. Furthermore, these data suggest that Cntn6 engages in both trans- and cis-interactions and may be involved in larger protein interaction networks.
Collapse
Affiliation(s)
- Amila Zuko
- a Brain Center Rudolf Magnus , Department of Translational Neuroscience , University Medical Center Utrecht , Utrecht , The Netherlands
| | - Asami Oguro-Ando
- a Brain Center Rudolf Magnus , Department of Translational Neuroscience , University Medical Center Utrecht , Utrecht , The Netherlands
| | - Roland van Dijk
- a Brain Center Rudolf Magnus , Department of Translational Neuroscience , University Medical Center Utrecht , Utrecht , The Netherlands
| | - Sara Gregorio-Jordan
- a Brain Center Rudolf Magnus , Department of Translational Neuroscience , University Medical Center Utrecht , Utrecht , The Netherlands
| | - Bert van der Zwaag
- b Department of Genetics , University Medical Center Utrecht , Utrecht , The Netherlands
| | - J Peter H Burbach
- a Brain Center Rudolf Magnus , Department of Translational Neuroscience , University Medical Center Utrecht , Utrecht , The Netherlands
| |
Collapse
|
30
|
Kleene R, Chaudhary H, Karl N, Katic J, Kotarska A, Guitart K, Loers G, Schachner M. Interaction between CHL1 and serotonin receptor 2c regulates signal transduction and behavior in mice. J Cell Sci 2015; 128:4642-52. [PMID: 26527397 DOI: 10.1242/jcs.176941] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 10/28/2015] [Indexed: 02/05/2023] Open
Abstract
The serotonergic system plays important roles in multiple functions of the nervous system and its malfunctioning leads to neurological and psychiatric disorders. Here, we show that the cell adhesion molecule close homolog of L1 (CHL1), which has been linked to mental disorders, binds to a peptide stretch in the third intracellular loop of the serotonin 2c (5-HT2c) receptor through its intracellular domain. Moreover, we provide evidence that CHL1 deficiency in mice leads to 5-HT2c-receptor-related reduction in locomotor activity and reactivity to novelty, and that CHL1 regulates signaling pathways triggered by constitutively active isoforms of the 5-HT2c receptor. Furthermore, we found that the 5-HT2c receptor and CHL1 colocalize in striatal and hippocampal GABAergic neurons, and that 5-HT2c receptor phosphorylation and its association with phosphatase and tensin homolog (PTEN) and β-arrestin 2 is regulated by CHL1. Our results demonstrate that CHL1 regulates signal transduction pathways through constitutively active 5-HT2c receptor isoforms, thereby altering 5-HT2c receptor functions and implicating CHL1 as a new modulator of the serotonergic system.
Collapse
Affiliation(s)
- Ralf Kleene
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, Hamburg 20246, Germany
| | - Harshita Chaudhary
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, Hamburg 20246, Germany
| | - Nicole Karl
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, Hamburg 20246, Germany
| | - Jelena Katic
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, Hamburg 20246, Germany
| | - Agnieszka Kotarska
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, Hamburg 20246, Germany
| | - Kathrin Guitart
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, Hamburg 20246, Germany
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, Hamburg 20246, Germany
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong 515041, China
| |
Collapse
|
31
|
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
|
32
|
Schmalbach B, Lepsveridze E, Djogo N, Papashvili G, Kuang F, Leshchyns'ka I, Sytnyk V, Nikonenko AG, Dityatev A, Jakovcevski I, Schachner M. Age-dependent loss of parvalbumin-expressing hippocampal interneurons in mice deficient in CHL1, a mental retardation and schizophrenia susceptibility gene. J Neurochem 2015; 135:830-44. [PMID: 26285062 DOI: 10.1111/jnc.13284] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 08/05/2015] [Accepted: 08/07/2015] [Indexed: 02/05/2023]
Abstract
In humans, deletions/mutations in the CHL1/CALL gene are associated with mental retardation and schizophrenia. Juvenile CHL1-deficient (CHL1(-/-) ) mice have been shown to display abnormally high numbers of parvalbumin-expressing (PV(+) ) hippocampal interneurons and, as adults, display behavioral traits observed in neuropsychiatric disorders. Here, we addressed the question whether inhibitory interneurons and synaptic plasticity in the CHL1(-/-) mouse are affected during brain maturation and in adulthood. We found that hippocampal, but not neocortical, PV(+) interneurons were reduced with age in CHL1(-/-) mice, from a surplus of +27% at 1 month to a deficit of -20% in adulthood compared with wild-type littermates. This loss occurred during brain maturation, correlating with microgliosis and enhanced interleukin-6 expression. In parallel with the loss of PV(+) interneurons, the inhibitory input to adult CA1 pyramidal cells was reduced and a deficit in short- and long-term potentiation developed at CA3-CA1 excitatory synapses between 2 and 9 months of age in CHL1(-/-) mice. This deficit could be abrogated by a GABAA receptor agonist. We propose that region-specific aberrant GABAergic synaptic connectivity resulting from the mutation and a subsequently enhanced synaptic elimination during brain maturation lead to microgliosis, increase in pro-inflammatory cytokine levels, loss of interneurons, and impaired synaptic plasticity. Close homolog of L1-deficient (CHL1(-/-) ) mice have abnormally high numbers of parvalbumin (PV)-expressing hippocampal interneurons in juvenile animals, but in adult animals a loss of these cells is observed. This loss correlates with an increased density of microglia (M), enhanced interleukin-6 (IL6) production and a deficit in short- and long-term potentiation at CA3-CA1 excitatory synapses. Furthermore, adult CHL1(-/-) mice display behavioral traits similar to those observed in neuropsychiatric disorders of humans.
Collapse
Affiliation(s)
- Barbara Schmalbach
- Zentrum für Molekulare Neurobiologie, Universität Hamburg, Hamburg, Germany
| | - Eka Lepsveridze
- Zentrum für Molekulare Neurobiologie, Universität Hamburg, Hamburg, Germany
- Ilia State University, Tbilisi, Georgia
| | - Nevena Djogo
- Zentrum für Molekulare Neurobiologie, Universität Hamburg, Hamburg, Germany
| | - Giorgi Papashvili
- Zentrum für Molekulare Neurobiologie, Universität Hamburg, Hamburg, Germany
| | - Fang Kuang
- Zentrum für Molekulare Neurobiologie, Universität Hamburg, Hamburg, Germany
| | - Iryna Leshchyns'ka
- Zentrum für Molekulare Neurobiologie, Universität Hamburg, Hamburg, Germany
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Vladimir Sytnyk
- Zentrum für Molekulare Neurobiologie, Universität Hamburg, Hamburg, Germany
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Alexander G Nikonenko
- Zentrum für Molekulare Neurobiologie, Universität Hamburg, Hamburg, Germany
- Department of Cytology, Bogomoletz Institute of Physiology, Kiev, Ukraine
| | - Alexander Dityatev
- Zentrum für Molekulare Neurobiologie, Universität Hamburg, Hamburg, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen, Magdeburg, Germany
| | - Igor Jakovcevski
- Zentrum für Molekulare Neurobiologie, Universität Hamburg, Hamburg, Germany
- Experimental Neurophysiology, University Hospital Cologne, Köln, Germany
- German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Melitta Schachner
- Zentrum für Molekulare Neurobiologie, Universität Hamburg, Hamburg, Germany
- Department of Cell Biology and Neuroscience, Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, New Jersey, USA
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| |
Collapse
|
33
|
Tassano E, Biancheri R, Denegri L, Porta S, Novara F, Zuffardi O, Gimelli G, Cuoco C. Heterozygous deletion of CHL1 gene: detailed array-CGH and clinical characterization of a new case and review of the literature. Eur J Med Genet 2015; 57:626-9. [PMID: 25451713 DOI: 10.1016/j.ejmg.2014.09.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 09/22/2014] [Indexed: 11/28/2022]
Abstract
CHL1 gene maps at 3p26.3 and encodes a cell adhesion molecule of the immunoglobulin superfamily highly expressed in the brain. CHL1 regulates neuronal migration and neurite overgrowth in the developing brain, while in mature neurons it accumulates in the axonal membrane and regulates synapse function via the clathrin-dependent pathways. To our knowledge, to date only three familial cases presenting heterozygous deletion of chromosome 3 at band p26.3, including only the CHL1 gene, have been reported. All the patients presented cognitive impairment characterized by learning and language difficulties. Here, we describe a six-year-old boy in which array-CGH analysis disclosed a terminal 3p26.3 deletion. The deletion was transmitted from his normal mother and included only the CHL1 gene. Our patient presented microcephaly, short stature, mild mental retardation, learning and language delay, and strabismus. In our study we compare the phenotypic and molecular cytogenetic features of CHL1 gene deletion cases. Verbal function developmental delay seems to be a common key finding. The concomitance of the genetic and phenotypic alterations could be a good evidence of a new emerging syndrome associated with the deletion of CHL1 gene alone, although the identification of new cases is required.
Collapse
|
34
|
Taylor-Weiner H, Ravi N, Engler AJ. Traction forces mediated by integrin signaling are necessary for definitive endoderm specification. J Cell Sci 2015; 128:1961-8. [PMID: 25908864 DOI: 10.1242/jcs.166157] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 03/24/2015] [Indexed: 12/28/2022] Open
Abstract
Pluripotent embryonic stem cells (ESCs) exert low-traction forces on their niche in vitro whereas specification to definitive endoderm in vivo coincides with force-mediated motility, suggesting a differentiation-mediated switch. However, the onset of contractility and extent to which force-mediated integrin signaling regulates fate choices is not understood. To address the requirement of tractions forces for differentiation, we examined mouse embryonic stem cell (ESC) specification towards definitive endoderm on fibrillar fibronectin containing a deformation-sensitive FRET probe. Inhibiting contractility resulted in an increase in the observed fibronectin FRET intensity ratio but also decreased the amount of phosphorylated nuclear SMAD2, leading to reduced expression of the definitive endoderm marker SOX17. By contrast ESCs maintained in pluripotency medium did not exert significant tractions against the fibronectin matrix. When laminin-111 was added to fibrillar matrices to improve the efficiency of definitive endoderm induction, ESCs decreased their fibronectin traction forces in a laminin-dependent manner; blocking the laminin-binding α3-integrin restored fibronectin matrix deformation and reduced SOX17 expression and SMAD2 phosphorylation, probably because of compensation of inhibitory signaling from SMAD7 after 5 days in culture. These data imply that traction forces and integrin signaling are important regulators of early fate decisions in ESCs.
Collapse
Affiliation(s)
- Hermes Taylor-Weiner
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Neeraja Ravi
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Adam J Engler
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA Biomedical Sciences Program, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
35
|
El Fatimy R, Miozzo F, Le Mouël A, Abane R, Schwendimann L, Sabéran-Djoneidi D, de Thonel A, Massaoudi I, Paslaru L, Hashimoto-Torii K, Christians E, Rakic P, Gressens P, Mezger V. Heat shock factor 2 is a stress-responsive mediator of neuronal migration defects in models of fetal alcohol syndrome. EMBO Mol Med 2015; 6:1043-61. [PMID: 25027850 PMCID: PMC4154132 DOI: 10.15252/emmm.201303311] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Fetal alcohol spectrum disorder (FASD) is a frequent cause of mental retardation. However, the molecular mechanisms underlying brain development defects induced by maternal alcohol consumption during pregnancy are unclear. We used normal and Hsf2-deficient mice and cell systems to uncover a pivotal role for heat shock factor 2 (HSF2) in radial neuronal migration defects in the cortex, a hallmark of fetal alcohol exposure. Upon fetal alcohol exposure, HSF2 is essential for the triggering of HSF1 activation, which is accompanied by distinctive post-translational modifications, and HSF2 steers the formation of atypical alcohol-specific HSF1-HSF2 heterocomplexes. This perturbs the in vivo binding of HSF2 to heat shock elements (HSEs) in genes that control neuronal migration in normal conditions, such as p35 or the MAPs (microtubule-associated proteins, such as Dclk1 and Dcx), and alters their expression. In the absence of HSF2, migration defects as well as alterations in gene expression are reduced. Thus, HSF2, as a sensor for alcohol stress in the fetal brain, acts as a mediator of the neuronal migration defects associated with FASD.
Collapse
Affiliation(s)
- Rachid El Fatimy
- CNRS UMR7216 Épigénétique et Destin Cellulaire, Paris Cedex 13, France Univ Paris Diderot Sorbonne Paris Cité, Paris Cedex 13, France ED 387 iViv UPMC Univ Paris 06, Paris, France Univ Paris Diderot, Paris Cedex 13, France
| | - Federico Miozzo
- CNRS UMR7216 Épigénétique et Destin Cellulaire, Paris Cedex 13, France Univ Paris Diderot Sorbonne Paris Cité, Paris Cedex 13, France ED 387 iViv UPMC Univ Paris 06, Paris, France Univ Paris Diderot, Paris Cedex 13, France
| | - Anne Le Mouël
- CNRS UMR7216 Épigénétique et Destin Cellulaire, Paris Cedex 13, France Univ Paris Diderot Sorbonne Paris Cité, Paris Cedex 13, France
| | - Ryma Abane
- CNRS UMR7216 Épigénétique et Destin Cellulaire, Paris Cedex 13, France Univ Paris Diderot Sorbonne Paris Cité, Paris Cedex 13, France ED 387 iViv UPMC Univ Paris 06, Paris, France Univ Paris Diderot, Paris Cedex 13, France
| | - Leslie Schwendimann
- INSERM U1141, Hôpital Robert Debré, Paris, France Faculté de Médecine Denis Diderot, Univ Paris Diderot Sorbonne Paris Cité, Paris, France
| | - Délara Sabéran-Djoneidi
- CNRS UMR7216 Épigénétique et Destin Cellulaire, Paris Cedex 13, France Univ Paris Diderot Sorbonne Paris Cité, Paris Cedex 13, France
| | - Aurélie de Thonel
- INSERM UMR 866, Dijon, France Faculty of Medicine and Pharmacy, Univ Burgundy, Dijon, France
| | - Illiasse Massaoudi
- CNRS UMR7216 Épigénétique et Destin Cellulaire, Paris Cedex 13, France Univ Paris Diderot Sorbonne Paris Cité, Paris Cedex 13, France
| | - Liliana Paslaru
- Carol Davila University of Medicine and Pharmacy Fundeni Hospital, Bucharest, Romania
| | - Kazue Hashimoto-Torii
- Department of Neurobiology and Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Elisabeth Christians
- Laboratoire de Biologie du Développement de Villefranche-sur-mer, Observatoire Océanologique, CNRS, Villefranche-sur-mer, France Sorbonne Universités UPMC Univ Paris 06, Villefranche-sur-mer, France
| | - Pasko Rakic
- Department of Neurobiology and Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Pierre Gressens
- INSERM U1141, Hôpital Robert Debré, Paris, France Faculté de Médecine Denis Diderot, Univ Paris Diderot Sorbonne Paris Cité, Paris, France
| | - Valérie Mezger
- CNRS UMR7216 Épigénétique et Destin Cellulaire, Paris Cedex 13, France Univ Paris Diderot Sorbonne Paris Cité, Paris Cedex 13, France
| |
Collapse
|
36
|
Katic J, Loers G, Kleene R, Karl N, Schmidt C, Buck F, Zmijewski JW, Jakovcevski I, Preissner KT, Schachner M. Interaction of the cell adhesion molecule CHL1 with vitronectin, integrins, and the plasminogen activator inhibitor-2 promotes CHL1-induced neurite outgrowth and neuronal migration. J Neurosci 2014; 34:14606-23. [PMID: 25355214 PMCID: PMC6608427 DOI: 10.1523/jneurosci.3280-13.2014] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 08/28/2014] [Accepted: 09/03/2014] [Indexed: 02/05/2023] Open
Abstract
The cell adhesion molecule close homolog of L1 (CHL1) plays important functional roles in the developing and adult nervous system. In search of the binding partners that mediate the diverse and sometimes opposing functions of CHL1, the extracellular matrix-associated proteins vitronectin and plasminogen activator inhibitor-2 (PAI-2) were identified as novel CHL1 interaction partners and tested for involvement in CHL1-dependent functions during mouse cerebellar development. CHL1-induced cerebellar neurite outgrowth and cell migration at postnatal days 6-8 were inhibited by a CHL1-derived peptide comprising the integrin binding RGD motif, and by antibodies against vitronectin or several integrins, indicating a vitronectin-dependent integrin-mediated pathway. A PAI-2-derived peptide, or antibodies against PAI-2, urokinase type plasminogen activator (uPA), uPA receptor, and several integrins reduced cell migration. CHL1 colocalized with vitronectin, PAI-2, and several integrins in cerebellar granule cells, suggesting an association among these proteins. Interestingly, at the slightly earlier age of 4-5 d, cerebellar neurons did not depend on CHL1 for neuritogenesis and cell migration. However, differentiation of progenitor cells into neurons at this stage was dependent on homophilic CHL1-CHL1 interactions. These observations indicate that homophilic CHL1 trans-interactions regulate differentiation of neuronal progenitor cells at early postnatal stages, while heterophilic trans-interactions of CHL1 with vitronectin, integrins, and the plasminogen activator system regulate neuritogenesis and neuronal cell migration at a later postnatal stage of cerebellar morphogenesis. Thus, within very narrow time windows in postnatal cerebellar development, distinct types of molecular interactions mediated by CHL1 underlie the diverse functions of this protein.
Collapse
Affiliation(s)
| | | | | | | | | | - Friedrich Buck
- Institut für Klinische Chemie, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Jaroslaw W Zmijewski
- Division of Pulmonary, Allergy & Critical Care Medicine, University of Alabama at Birmingham, BMRII-304, Birmingham, Alabama 35294
| | | | - Klaus T Preissner
- Department of Biochemistry, Medical School, Justus-Liebig-University, 35392 Giessen, Germany
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey 08854, Center for Neuroscience, Shantou University Medical College, Shantou 515041, People's Republic of China, and
| |
Collapse
|
37
|
Taylor-Weiner H, Schwarzbauer JE, Engler AJ. Defined extracellular matrix components are necessary for definitive endoderm induction. Stem Cells 2014; 31:2084-94. [PMID: 23766144 DOI: 10.1002/stem.1453] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 04/30/2013] [Accepted: 05/25/2013] [Indexed: 01/25/2023]
Abstract
Differentiation methods often rely exclusively on growth factors to direct mouse embryonic stem cell (ESC) fate, but the niche also contains fibrillar extracellular matrix (ECM) proteins, including fibronectin (FN) and laminin, which could also direct cell fate. Soluble differentiation factors are known to increase ECM expression, yet ECM's ability to direct ESC fate is not well understood. To address the extent to which these proteins regulate differentiation when assembled into a matrix, we examined mouse ESC embryoid bodies (EBs) and found that their ability to maintain pluripotency marker expression was impaired by soluble serum FN. EBs also showed a spatiotemporal correlation between expression of FN and GATA4, a marker of definitive endoderm (DE), and an inverse correlation between FN and Nanog, a pluripotency marker. Maintenance of mouse ESC pluripotency prevented fibrillar matrix production, but induction medium created lineage-specific ECM containing varying amounts of FN and laminin. Mouse ESC-derived matrix was unlike conventional fibroblast-derived matrix, which did not contain laminin. Naïve mouse ESCs plated onto ESC- and fibroblast-derived matrix exhibited composition-specific differentiation. With exogenously added laminin, fibroblast-derived matrix is more similar in composition to mouse ESC-derived matrix and lacks residual growth factors that mouse ESC matrix may contain. Naïve mouse ESCs in DE induction medium exhibited dose-dependent DE differentiation as a function of the amount of exogenous laminin in the matrix in an α3 integrin-dependent mechanism. These data imply that fibrillar FN is necessary for loss of pluripotency and that laminin within a FN matrix improves DE differentiation.
Collapse
Affiliation(s)
- Hermes Taylor-Weiner
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA; Sanford Consortium for Regenerative Medicine, La Jolla, California, USA
| | | | | |
Collapse
|
38
|
Nagaraj K, Mualla R, Hortsch M. The L1 Family of Cell Adhesion Molecules: A Sickening Number of Mutations and Protein Functions. ADVANCES IN NEUROBIOLOGY 2014; 8:195-229. [DOI: 10.1007/978-1-4614-8090-7_9] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
39
|
McLachlan IG, Heiman MG. Shaping dendrites with machinery borrowed from epithelia. Curr Opin Neurobiol 2013; 23:1005-10. [DOI: 10.1016/j.conb.2013.06.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 06/27/2013] [Indexed: 10/26/2022]
|
40
|
Oved K, Morag A, Pasmanik-Chor M, Rehavi M, Shomron N, Gurwitz D. Genome-wide expression profiling of human lymphoblastoid cell lines implicates integrin beta-3 in the mode of action of antidepressants. Transl Psychiatry 2013; 3:e313. [PMID: 24129413 PMCID: PMC3818017 DOI: 10.1038/tp.2013.86] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 08/15/2013] [Accepted: 09/08/2013] [Indexed: 01/10/2023] Open
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are the first-line treatment for major depression. However, the link between inhibition of serotonin reuptake and remission from depression remains controversial: in spite of the rapid onset of serotonin reuptake inhibition, remission from depression takes several weeks, presumably reflecting synaptogenesis/neurogenesis and neuronal rewiring. We compared genome-wide expression profiles of human lymphoblastoid cell lines from unrelated individuals following treatment with 1 μM paroxetine for 21 days with untreated control cells and examined which genes and microRNAs (miRNAs) showed the most profound and consistent expression changes. ITGB3, coding for integrin beta-3, showed the most consistent altered expression (1.92-fold increase, P=7.5 × 10(-8)) following chronic paroxetine exposure. Using genome-wide miRNA arrays, we observed a corresponding decrease in the expression of two miRNAs, miR-221 and miR-222, both predicted to target ITGB3. ITGB3 is crucial for the activity of the serotonin transporter (SERT), the drug target of SSRIs. Moreover, it is presumably required for the neuronal guidance activity of CHL1, whose expression was formerly identified as a tentative SSRI response biomarker. Further genes whose expression was significantly modulated by chronic paroxetine are also implicated in neurogenesis. Surprisingly, the expression of SERT or serotonin receptors was not modified. Our findings implicate ITGB3 in the mode of action of SSRI antidepressants and provide a novel link between CHL1 and the SERT. Our observations suggest that SSRIs may relieve depression primarily by promoting neuronal synaptogenesis/neurogenesis rather than by modulating serotonin neurotransmission per se.
Collapse
Affiliation(s)
- K Oved
- Department of Human Molecular Genetics and
Biochemistry, Sackler Faculty of Medicine, Tel-Aviv University,
Tel-Aviv, Israel
- Department of Cell and Developmental Biology,
Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv,
Israel
| | - A Morag
- Department of Human Molecular Genetics and
Biochemistry, Sackler Faculty of Medicine, Tel-Aviv University,
Tel-Aviv, Israel
- Department of Physiology and Pharmacology,
Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv,
Israel
| | - M Pasmanik-Chor
- Bioinformatics Unit, George Wise Faculty of
Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - M Rehavi
- Department of Physiology and Pharmacology,
Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv,
Israel
| | - N Shomron
- Department of Cell and Developmental Biology,
Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv,
Israel
- Sagol School of Neuroscience, Tel-Aviv
University, Tel-Aviv, Israel
| | - D Gurwitz
- Department of Human Molecular Genetics and
Biochemistry, Sackler Faculty of Medicine, Tel-Aviv University,
Tel-Aviv, Israel
| |
Collapse
|
41
|
N-acetyl-cysteine prevents pyramidal cell disarray and reelin-immunoreactive neuron deficiency in CA3 after prenatal immune challenge in rats. Pediatr Res 2013; 73:750-5. [PMID: 23478644 DOI: 10.1038/pr.2013.40] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Prenatal infection is a major risk factor for the occurrence of neuropsychiatric disorders. These have been associated with hippocampal neuroanatomical and functional abnormalities. In the present study, we evaluated the occurrence of pyramidal cell disarray and reelin neuronal deficit in the hippocampus, and the protective role of N-acetyl-cysteine (NAC) in a rodent experimental model of prenatal immune challenge. METHODS Sprague-Dawley rats received either 500 μg/kg of endotoxin (lipopolysaccharide, LPS) or 2 ml/kg of isotonic saline by i.p. injection on day 19 of gestation. After LPS injection, rats were or were not maintained on a preventive treatment of NAC (5 g/l in tap water), up to delivery. The pyramidal cell orientation and the number and type of reelin-expressing neurons were determined in male offspring. RESULTS Prenatal LPS challenge led to permanent pyramidal cell disarray and to an early and transient decreased density of reelin-immunoreactive neurons. These disorders, more pronounced in the CA3 area, were prevented by NAC. CONCLUSION Hippocampal cytoarchitectural alterations and reelin deficiency may be involved in the development of remote cognitive impairments in this model. The antioxidant NAC is an efficient neuroprotective drug that underlines the role of oxidative stress in prenatal infection and associated neurodevelopmental damage.
Collapse
|
42
|
Kershaw J, Leuze C, Aoki I, Obata T, Kanno I, Ito H, Yamaguchi Y, Handa H. Systematic changes to the apparent diffusion tensor of in vivo rat brain measured with an oscillating-gradient spin-echo sequence. Neuroimage 2013; 70:10-20. [DOI: 10.1016/j.neuroimage.2012.12.036] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 11/22/2012] [Accepted: 12/12/2012] [Indexed: 10/27/2022] Open
|
43
|
Using C. elegans to Decipher the Cellular and Molecular Mechanisms Underlying Neurodevelopmental Disorders. Mol Neurobiol 2013; 48:465-89. [DOI: 10.1007/s12035-013-8434-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Accepted: 02/26/2013] [Indexed: 10/27/2022]
|
44
|
The Angelman syndrome protein Ube3a is required for polarized dendrite morphogenesis in pyramidal neurons. J Neurosci 2013; 33:327-33. [PMID: 23283345 DOI: 10.1523/jneurosci.2509-12.2013] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Pyramidal neurons have a highly polarized dendritic morphology, characterized by one long apical dendrite and multiple short basal dendrites. They function as the primary excitatory cells of the mammalian prefrontal cortex and the corticospinal tract. However, the molecular mechanisms underlying the development of polarized dendrite morphology in pyramidal neurons remain poorly understood. Here, we report that the Angelman syndrome (AS) protein ubiquitin-protein ligase E3A (Ube3a) plays an important role in specifying the polarization of pyramidal neuron dendritic arbors in mice. shRNA-mediated downregulation of Ube3a selectively inhibited apical dendrite outgrowth and resulted in impaired dendrite polarity, which could be rescued by coexpressing mouse Ube3a isoform 2, but not isoform 1 or 3. Ube3a knockdown also disrupted the polarized distribution of the Golgi apparatus, a well established cellular mechanism for asymmetric dendritic growth in pyramidal neurons. Furthermore, downregulation of Ube3a completely blocked Reelin-induced rapid deployment of Golgi into dendrite. Consistently, we also observed selective inhibition of apical dendrite outgrowth in pyramidal neurons in a mouse model of AS. Overall, these results show that Ube3a is required for the specification of the apical dendrites and dendrite polarization in pyramidal neurons, and suggest a novel pathological mechanism for AS.
Collapse
|
45
|
Tian N, Leshchyns'ka I, Welch JH, Diakowski W, Yang H, Schachner M, Sytnyk V. Lipid raft-dependent endocytosis of close homolog of adhesion molecule L1 (CHL1) promotes neuritogenesis. J Biol Chem 2012; 287:44447-63. [PMID: 23144456 DOI: 10.1074/jbc.m112.394973] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
CHL1 plays a dual role by either promoting or inhibiting neuritogenesis. We report here that neuritogenesis-promoting ligand-dependent cell surface clustering of CHL1 induces palmitoylation and lipid raft-dependent endocytosis of CHL1. We identify βII spectrin as a binding partner of CHL1, and we show that partial disruption of the complex between CHL1 and βII spectrin accompanies CHL1 endocytosis. Inhibition of the association of CHL1 with lipid rafts by pharmacological disruption of lipid rafts or by mutation of cysteine 1102 within the intracellular domain of CHL1 reduces endocytosis of CHL1. Endocytosis of CHL1 is also reduced by nifedipine, an inhibitor of the L-type voltage-dependent Ca(2+) channels. CHL1-dependent neurite outgrowth is reduced by inhibitors of lipid raft assembly, inhibitors of voltage-dependent Ca(2+) channels, and overexpression of CHL1 with mutated cysteine Cys-1102. Our results suggest that ligand-induced and lipid raft-dependent regulation of CHL1 adhesion via Ca(2+)-dependent remodeling of the CHL1-βII spectrin complex and CHL1 endocytosis are required for CHL1-dependent neurite outgrowth.
Collapse
Affiliation(s)
- Nan Tian
- Zentrum für Molekulare Neurobiologie, Universitätskrankenhaus Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | | | | | | | | | | | | |
Collapse
|
46
|
Leuze CWU, Anwander A, Bazin PL, Dhital B, Stüber C, Reimann K, Geyer S, Turner R. Layer-specific intracortical connectivity revealed with diffusion MRI. Cereb Cortex 2012; 24:328-39. [PMID: 23099298 PMCID: PMC3888365 DOI: 10.1093/cercor/bhs311] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
In this work, we show for the first time that the tangential diffusion component is orientationally coherent at the human cortical surface. Using diffusion magnetic resonance imaging (dMRI), we have succeeded in tracking intracortical fiber pathways running tangentially within the cortex. In contrast with histological methods, which reveal little regarding 3-dimensional organization in the human brain, dMRI delivers additional understanding of the layer dependence of the fiber orientation. A postmortem brain block was measured at very high angular and spatial resolution. The dMRI data had adequate resolution to allow analysis of the fiber orientation within 4 notional cortical laminae. We distinguished a lamina at the cortical surface where diffusion was tangential along the surface, a lamina below the surface where diffusion was mainly radial, an internal lamina covering the Stria of Gennari, where both strong radial and tangential diffusion could be observed, and a deep lamina near the white matter, which also showed mainly radial diffusion with a few tangential compartments. The measurement of the organization of the tangential diffusion component revealed a strong orientational coherence at the cortical surface.
Collapse
Affiliation(s)
- Christoph W U Leuze
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Zhou L, Barão S, Laga M, Bockstael K, Borgers M, Gijsen H, Annaert W, Moechars D, Mercken M, Gevaert K, Gevaer K, De Strooper B. The neural cell adhesion molecules L1 and CHL1 are cleaved by BACE1 protease in vivo. J Biol Chem 2012; 287:25927-40. [PMID: 22692213 DOI: 10.1074/jbc.m112.377465] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The β-site amyloid precursor protein-cleaving enzyme BACE1 is a prime drug target for Alzheimer disease. However, the function and the physiological substrates of BACE1 remain largely unknown. In this work, we took a quantitative proteomic approach to analyze the secretome of primary neurons after acute BACE1 inhibition, and we identified several novel substrate candidates for BACE1. Many of these molecules are involved in neuronal network formation in the developing nervous system. We selected the adhesion molecules L1 and CHL1, which are crucial for axonal guidance and maintenance of neural circuits, for further validation as BACE1 substrates. Using both genetic BACE1 knock-out and acute pharmacological BACE1 inhibition in mice and cell cultures, we show that L1 and CHL1 are cleaved by BACE1 under physiological conditions. The BACE1 cleavage sites at the membrane-proximal regions of L1 (between Tyr(1086) and Glu(1087)) and CHL1 (between Gln(1061) and Asp(1062)) were determined by mass spectrometry. This work provides molecular insights into the function and the pathways in which BACE1 is involved, and it will help to predict or interpret possible side effects of BACE1 inhibitor drugs in current clinical trials.
Collapse
Affiliation(s)
- Lujia Zhou
- VIB Center for the Biology of Disease, KULeuven, 3000 Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Solecki DJ. Sticky situations: recent advances in control of cell adhesion during neuronal migration. Curr Opin Neurobiol 2012; 22:791-8. [PMID: 22560352 DOI: 10.1016/j.conb.2012.04.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 03/26/2012] [Accepted: 04/12/2012] [Indexed: 11/30/2022]
Abstract
The migration of neurons along glial fibers from a germinal zone (GZ) to their final laminar positions is essential for morphogenesis of the developing brain; aberrations in this process are linked to profound neurodevelopmental and cognitive disorders. During this critical morphogenic movement, neurons must navigate complex migration paths, propelling their cell bodies through the dense cellular environment of the developing nervous system to their final destinations. It is not understood how neurons can successfully migrate along their glial guides through the myriad processes and cell bodies of neighboring neurons. Although much progress has been made in understanding the substrates (Fishell G, Hatten ME: Astrotactin provides a receptor system for CNS neuronal migration. Development 1991, 113:755; Elias LA, Wang DD, Kriegstein AR: Gap junction adhesion is necessary for radial migration in the neocortex. Nature 2007, 448:901; Anton ES, Kreidberg JA, Rakic P: Distinct functions of alpha3 and alpha. (v) integrin receptors in neuronal migration and laminar organization of the cerebral cortex. Neuron 1999, 22:277; Anton ES, Marchionni MA, Lee KF, Rakic P: Role of GGF/neuregulin signaling in interactions between migrating neurons and radial glia in the developing cerebral cortex. Development 1997, 124:3501), guidance mechanisms (Polleux F, Whitford KL, Dijkhuizen PA, Vitalis T, Ghosh A: Control of cortical interneuron migration by neurotrophins and PI3-kinase signaling. Development 2002, 129:3147; Zhou P, et al.: Polarized signaling endosomes coordinate BDNF-induced chemotaxis of cerebellar precursors. Neuron 2007, 55:53; Renaud J, et al.: Plexin-A2 and its ligand, Sema6A, control nucleus-centrosome coupling in migrating granule cells. Nat Neurosci 2008, 11:440), cytoskeletal elements (Schaar BT, McConnell SK: Cytoskeletal coordination during neuronal migration. Proc Natl Acad Sci U S A 2005, 102:13652; Tsai JW, Bremner KH, Vallee RB: Dual subcellular roles for LIS1 and dynein in radial neuronal migration in live brain tissue. Nat Neurosci 2007, 10:970; Solecki DJ, et al.: Myosin II motors and F-actin dynamics drive the coordinated movement of the centrosome and soma during CNS glial-guided neuronal migration. Neuron 2009, 63:63), and post-translational modifications (Patrick GN, Zhou P, Kwon YT, Howley PM, Tsai LH: p35, the neuronal-specific activator of cyclin-dependent kinase 5 (Cdk5) is degraded by the ubiquitin-proteasome pathway. J Biol Chem 1998, 273:24057; Suetsugu S, et al.: Regulation of actin cytoskeleton by mDab1 through N-WASP and ubiquitination of mDab1. Biochem J 2004, 384:1; Karakuzu O, Wang DP, Cameron S: MIG-32 and SPAT-3A are PRC1 homologs that control neuronal migration inCaenorhabditis elegans. Development 2009, 136:943) required for neuronal migration, we have yet to elucidate how neurons regulate their cellular interactions and adhesive specificity to follow the appropriate migratory pathways. Here I will examine recent developments in our understanding of the mechanisms controlling neuronal cell adhesion and how these mechanisms interact with crucial neurodevelopmental events, such as GZ exit, migration pathway selection, multipolar-to-radial transition, and final lamination.
Collapse
Affiliation(s)
- David J Solecki
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, United States.
| |
Collapse
|
49
|
Loss of function studies in mice and genetic association link receptor protein tyrosine phosphatase α to schizophrenia. Biol Psychiatry 2011; 70:626-35. [PMID: 21831360 PMCID: PMC3176920 DOI: 10.1016/j.biopsych.2011.06.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Revised: 05/17/2011] [Accepted: 06/02/2011] [Indexed: 01/06/2023]
Abstract
BACKGROUND Solid evidence links schizophrenia (SZ) susceptibility to neurodevelopmental processes involving tyrosine phosphorylation-mediated signaling. Mouse studies implicate the Ptpra gene, encoding protein tyrosine phosphatase RPTPα, in the control of radial neuronal migration, cortical cytoarchitecture, and oligodendrocyte differentiation. The human gene encoding RPTPα, PTPRA, maps to a chromosomal region (20p13) associated with susceptibility to psychotic illness. METHODS We characterized neurobehavioral parameters, as well as gene expression in the central nervous system, of mice with a null mutation in the Ptpra gene. We searched for genetic association between polymorphisms in PTPRA and schizophrenia risk (two independent cohorts, 1420 cases and 1377 controls), and we monitored PTPRA expression in prefrontal dorsolateral cortex of SZ patients (35 cases, 2 control groups of 35 cases). RESULTS We found that Ptpra⁻/⁻ mice reproduce neurobehavioral endophenotypes of human SZ: sensitization to methamphetamine-induced hyperactivity, defective sensorimotor gating, and defective habituation to a startle response. Ptpra loss of function also leads to reduced expression of multiple myelination genes, mimicking the hypomyelination-associated changes in gene expression observed in postmortem patient brains. We further report that a polymorphism at the PTPRA locus is genetically associated with SZ, and that PTPRA mRNA levels are reduced in postmortem dorsolateral prefrontal cortex of subjects with SZ. CONCLUSIONS The implication of this well-studied signaling protein in SZ risk and endophenotype manifestation provides novel entry points into the etiopathology of this disease.
Collapse
|
50
|
Ye H, Zhao T, Tan YLJ, Liu J, Pallen CJ, Xiao ZC. Receptor-like protein-tyrosine phosphatase α enhances cell surface expression of neural adhesion molecule NB-3. J Biol Chem 2011; 286:26071-80. [PMID: 21622556 DOI: 10.1074/jbc.m110.214080] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Neural adhesion molecule NB-3 plays an important role in the apical dendrite development of layer V pyramidal neurons in the visual cortex, and receptor-like protein-tyrosine phosphatase α (PTPα) mediates NB-3 signaling in this process. Here we investigated the role of PTPα in regulating cell surface expression of NB-3. We found that cortical neurons from PTPα knock-out mice exhibited a lower level of NB-3 at the cell surface. When expressed in COS1 cells, NB-3 was enriched in the Golgi apparatus with a low level of cell surface expression. However, co-expression of PTPα increased the cell surface distribution of NB-3. Further analysis showed that PTPα facilitated Golgi exit of NB-3 and stabilized NB-3 protein at the cell surface by preventing its release from the plasma membrane. The extracellular region of PTPα but not its catalytic activity is necessary for its effect on NB-3 expression. Thus, the PTPα-mediated increase of NB-3 level at the cell surface represents a novel function of PTPα in NB-3 signaling in neural development.
Collapse
Affiliation(s)
- Haihong Ye
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| | | | | | | | | | | |
Collapse
|