1
|
Fan S, Liu J, Chofflet N, Bailey AO, Russell WK, Zhang Z, Takahashi H, Ren G, Rudenko G. Molecular mechanism of contactin 2 homophilic interaction. Structure 2024; 32:1652-1666.e8. [PMID: 38968938 PMCID: PMC11455609 DOI: 10.1016/j.str.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/23/2024] [Accepted: 06/10/2024] [Indexed: 07/07/2024]
Abstract
Contactin 2 (CNTN2) is a cell adhesion molecule involved in axon guidance, neuronal migration, and fasciculation. The ectodomains of CNTN1-CNTN6 are composed of six Ig domains (Ig1-Ig6) and four FN domains. Here, we show that CNTN2 forms transient homophilic interactions (KD ∼200 nM). Cryo-EM structures of full-length CNTN2 and CNTN2_Ig1-Ig6 reveal a T-shaped homodimer formed by intertwined, parallel monomers. Unexpectedly, the horseshoe-shaped Ig1-Ig4 headpieces extend their Ig2-Ig3 tips outwards on either side of the homodimer, while Ig4, Ig5, Ig6, and the FN domains form a central stalk. Cross-linking mass spectrometry and cell-based binding assays confirm the 3D assembly of the CNTN2 homodimer. The interface mediating homodimer formation differs between CNTNs, as do the homophilic versus heterophilic interaction mechanisms. The CNTN family thus encodes a versatile molecular platform that supports a very diverse portfolio of protein interactions and that can be leveraged to strategically guide neural circuit development.
Collapse
Affiliation(s)
- Shanghua Fan
- Department of Pharmacology and Toxicology; University of Texas Medical Branch, Galveston, TX 77555, USA; Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jianfang Liu
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| | - Nicolas Chofflet
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 2B2, Canada
| | - Aaron O Bailey
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - William K Russell
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ziqi Zhang
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada
| | - Hideto Takahashi
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada; Division of Experimental Medicine, McGill University, Montreal, QC H3A 0G4, Canada.
| | - Gang Ren
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| | - Gabby Rudenko
- Department of Pharmacology and Toxicology; University of Texas Medical Branch, Galveston, TX 77555, USA; Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
2
|
Zhou H, Yu Q, Li Y, Fu F, Li R, Chen G, Wang D, Lu Y, Yang X, Li D, Liao C. Case Report: Two Novel L1CAM Mutations in Two Unrelated Chinese Families With X-Linked Hydrocephalus. Front Genet 2022; 13:810853. [PMID: 35571029 PMCID: PMC9099044 DOI: 10.3389/fgene.2022.810853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 04/04/2022] [Indexed: 11/23/2022] Open
Abstract
L1 cell adhesion molecule is a type I transmembrane glycoprotein belonging to the immunoglobulin superfamily. Pathogenic mutations of L1CAM can cause L1 syndrome, referred to as a variety of disease spectrums characterized by hydrocephalus. In the present study, we reported two novel variants of L1CAM in two unrelated Chinese families with fetal hydrocephalus history. The woman of family 1, with three consecutive adverse birth histories of male fetuses with hydrocephalus, was identified by an exome sequence with a heterozygous mutation in the L1CAM gene, NM_000425.4: c.1696_1703 + 14del (p. S566Vfs*35), which was predicted to be pathogenic. It is predicted to disrupt RNA splicing and likely leads to an absent or disrupted protein product. In family 2, the mother, previously with once a voluntary termination of pregnancy owning to the fetus with hydrocephalus, was pregnant with a fetus with hydrocephalus in her second pregnancy. After fetal blood sampling, a pathogenic deletion of 1511bp in L1CAM, chromosome X: 153131395-153132905(hg19/GRCh37)/NM_000425.4: c.2043_2432-121del1511 leading to deletion of fibronectin type-III repeats I-II, was identified in the fetus with hydrocephalus inherited from the mother by an exome sequence. On her third pregnancy, a healthy female fetus was born without the L1CAM variant by preimplantation genetic testing for the monogenic disorder. This study emphasizes the importance of ultrasonic manifestation and family history of fetal hydrocephalus for L1CAM diagnosis. Our study expands the genotypes of L1CAM and aids the genetic counseling of fetal hydrocephalus and even preimplantation genetic testing for the monogenic disorder.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Can Liao
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
3
|
Spead O, Weaver CJ, Moreland T, Poulain FE. Live imaging of retinotectal mapping reveals topographic map dynamics and a previously undescribed role for Contactin 2 in map sharpening. Development 2021; 148:272618. [PMID: 34698769 DOI: 10.1242/dev.199584] [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: 03/03/2021] [Accepted: 10/07/2021] [Indexed: 11/20/2022]
Abstract
Organization of neuronal connections into topographic maps is essential for processing information. Yet, our understanding of topographic mapping has remained limited by our inability to observe maps forming and refining directly in vivo. Here, we used Cre-mediated recombination of a new colorswitch reporter in zebrafish to generate the first transgenic model allowing the dynamic analysis of retinotectal mapping in vivo. We found that the antero-posterior retinotopic map forms early but remains dynamic, with nasal and temporal retinal axons expanding their projection domains over time. Nasal projections initially arborize in the anterior tectum but progressively refine their projection domain to the posterior tectum, leading to the sharpening of the retinotopic map along the antero-posterior axis. Finally, using a CRISPR-mediated mutagenesis approach, we demonstrate that the refinement of nasal retinal projections requires the adhesion molecule Contactin 2. Altogether, our study provides the first analysis of a topographic map maturing in real time in a live animal and opens new strategies for dissecting the molecular mechanisms underlying precise topographic mapping in vertebrates.
Collapse
Affiliation(s)
- Olivia Spead
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Cory J Weaver
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Trevor Moreland
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Fabienne E Poulain
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| |
Collapse
|
4
|
Desse VE, Blanchette CR, Nadour M, Perrat P, Rivollet L, Khandekar A, Bénard CY. Neuronal post-developmentally acting SAX-7S/L1CAM can function as cleaved fragments to maintain neuronal architecture in C. elegans. Genetics 2021; 218:6296841. [PMID: 34115111 DOI: 10.1093/genetics/iyab086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 05/24/2021] [Indexed: 01/09/2023] Open
Abstract
Whereas remarkable advances have uncovered mechanisms that drive nervous system assembly, the processes responsible for the lifelong maintenance of nervous system architecture remain poorly understood. Subsequent to its establishment during embryogenesis, neuronal architecture is maintained throughout life in the face of the animal's growth, maturation processes, the addition of new neurons, body movements, and aging. The C. elegans protein SAX-7, homologous to the vertebrate L1 protein family of neural adhesion molecules, is required for maintaining the organization of neuronal ganglia and fascicles after their successful initial embryonic development. To dissect the function of sax-7 in neuronal maintenance, we generated a null allele and sax-7S-isoform-specific alleles. We find that the null sax-7(qv30) is, in some contexts, more severe than previously described mutant alleles, and that the loss of sax-7S largely phenocopies the null, consistent with sax-7S being the key isoform in neuronal maintenance. Using a sfGFP::SAX-7S knock-in, we observe sax-7S to be predominantly expressed across the nervous system, from embryogenesis to adulthood. Yet, its role in maintaining neuronal organization is ensured by post-developmentally acting SAX-7S, as larval transgenic sax-7S(+) expression alone is sufficient to profoundly rescue the null mutants' neuronal maintenance defects. Moreover, the majority of the protein SAX-7 appears to be cleaved, and we show that these cleaved SAX-7S fragments together, not individually, can fully support neuronal maintenance. These findings contribute to our understanding of the role of the conserved protein SAX-7/L1CAM in long-term neuronal maintenance, and may help decipher processes that go awry in some neurodegenerative conditions.
Collapse
Affiliation(s)
- Virginie E Desse
- Department of Biological Sciences, CERMO-FC Research Center, Université du Québec à Montréal, Montréal, QC H2X 1Y4, Canada
| | - Cassandra R Blanchette
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Malika Nadour
- Department of Biological Sciences, CERMO-FC Research Center, Université du Québec à Montréal, Montréal, QC H2X 1Y4, Canada
| | - Paola Perrat
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Lise Rivollet
- Department of Biological Sciences, CERMO-FC Research Center, Université du Québec à Montréal, Montréal, QC H2X 1Y4, Canada
| | - Anagha Khandekar
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Claire Y Bénard
- Department of Biological Sciences, CERMO-FC Research Center, Université du Québec à Montréal, Montréal, QC H2X 1Y4, Canada
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|
5
|
Suter TACS, Blagburn SV, Fisher SE, Anderson-Keightly HM, D'Elia KP, Jaworski A. TAG-1 Multifunctionality Coordinates Neuronal Migration, Axon Guidance, and Fasciculation. Cell Rep 2020; 30:1164-1177.e7. [PMID: 31995756 PMCID: PMC7049094 DOI: 10.1016/j.celrep.2019.12.085] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 10/25/2019] [Accepted: 12/22/2019] [Indexed: 11/03/2022] Open
Abstract
Neuronal migration, axon fasciculation, and axon guidance need to be closely coordinated for neural circuit assembly. Spinal motor neurons (MNs) face unique challenges during development because their cell bodies reside within the central nervous system (CNS) and their axons project to various targets in the body periphery. The molecular mechanisms that contain MN somata within the spinal cord while allowing their axons to exit the CNS and navigate to their final destinations remain incompletely understood. We find that the MN cell surface protein TAG-1 anchors MN cell bodies in the spinal cord to prevent their emigration, mediates motor axon fasciculation during CNS exit, and guides motor axons past dorsal root ganglia. TAG-1 executes these varied functions in MN development independently of one another. Our results identify TAG-1 as a key multifunctional regulator of MN wiring that coordinates neuronal migration, axon fasciculation, and axon guidance. Suter et al. demonstrate that the motor neuron cell surface molecule TAG-1 confines motor neurons to the central nervous system, promotes motor axon fasciculation, and steers motor axons past inappropriate targets. This study highlights how a single cell adhesion molecule coordinates multiple steps in neuronal wiring through partially divergent mechanisms.
Collapse
Affiliation(s)
- Tracey A C S Suter
- Department of Neuroscience, Brown University, Providence, RI 02912, USA; Robert J. and Nancy D. Carney Institute for Brain Science, Providence, RI 02912, USA
| | - Sara V Blagburn
- Department of Neuroscience, Brown University, Providence, RI 02912, USA; Robert J. and Nancy D. Carney Institute for Brain Science, Providence, RI 02912, USA
| | - Sophie E Fisher
- Department of Neuroscience, Brown University, Providence, RI 02912, USA; Robert J. and Nancy D. Carney Institute for Brain Science, Providence, RI 02912, USA
| | | | - Kristen P D'Elia
- Department of Neuroscience, Brown University, Providence, RI 02912, USA; Department of Biology, Providence College, Providence, RI 02918, USA
| | - Alexander Jaworski
- Department of Neuroscience, Brown University, Providence, RI 02912, USA; Robert J. and Nancy D. Carney Institute for Brain Science, Providence, RI 02912, USA.
| |
Collapse
|
6
|
D’Amato AR, Puhl DL, Ziemba AM, Johnson CDL, Doedee J, Bao J, Gilbert RJ. Exploring the effects of electrospun fiber surface nanotopography on neurite outgrowth and branching in neuron cultures. PLoS One 2019; 14:e0211731. [PMID: 30716106 PMCID: PMC6361509 DOI: 10.1371/journal.pone.0211731] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/18/2019] [Indexed: 12/19/2022] Open
Abstract
Three aligned, electrospun fiber scaffolds with unique surface features were created from poly-L-lactic acid (PLLA). Fibers without surface nanotopography (smooth fibers), fibers with surface divots (shallow pits), and fibers with surface pits (deeper pits) were fabricated, and fiber alignment, diameter, and density were characterized using scanning electron microscopy (SEM). Whole dorsal root ganglia (DRG) were isolated from rats and placed onto uncoated fibers or fibers coated with laminin. On uncoated fibers, neurite outgrowth was restricted by fibers displaying divoted or pitted nanotopography when compared to neurite outgrowth on smooth fibers. However, neurites extending from whole DRG cultured on laminin-coated fibers were not restricted by divoted or pitted surface nanotopography. Thus, neurites extending on laminin-coated fibers were able to extend long neurites even in the presence of surface divots or pits. To further explore this result, individual neurons isolated from dissociated DRG were seeded onto laminin-coated smooth, pitted, or divoted fibers. Interestingly, neurons on pitted or divoted fibers exhibited a 1.5-fold increase in total neurite length, and a 2.3 or 2.7-fold increase in neurite branching compared to neurons on smooth fibers, respectively. Based on these findings, we conclude that fiber roughness in the form of pits or divots can promote extension and branching of long neurites along aligned electrospun fibers in the presence of an extracellular matrix protein coating. Thus, aligned, electrospun fibers can be crafted to not only direct the extension of axons but to induce unique branching morphologies.
Collapse
Affiliation(s)
- Anthony R. D’Amato
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York,United States of America
| | - Devan L. Puhl
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York,United States of America
| | - Alexis M. Ziemba
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York,United States of America
| | - Christopher D. L. Johnson
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York,United States of America
| | - Janneke Doedee
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York,United States of America
| | - Jonathan Bao
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York,United States of America
| | - Ryan J. Gilbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York,United States of America
| |
Collapse
|
7
|
Ducuing H, Gardette T, Pignata A, Tauszig-Delamasure S, Castellani V. Commissural axon navigation in the spinal cord: A repertoire of repulsive forces is in command. Semin Cell Dev Biol 2019; 85:3-12. [DOI: 10.1016/j.semcdb.2017.12.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 12/11/2017] [Accepted: 12/11/2017] [Indexed: 01/31/2023]
|
8
|
Gurung S, Asante E, Hummel D, Williams A, Feldman-Schultz O, Halloran MC, Sittaramane V, Chandrasekhar A. Distinct roles for the cell adhesion molecule Contactin2 in the development and function of neural circuits in zebrafish. Mech Dev 2018; 152:1-12. [PMID: 29777776 DOI: 10.1016/j.mod.2018.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 04/02/2018] [Accepted: 05/09/2018] [Indexed: 01/17/2023]
Abstract
Contactin2 (Cntn2)/Transient Axonal Glycoprotein 1 (Tag1), a neural cell adhesion molecule, has established roles in neuronal migration and axon fasciculation in chick and mouse. In zebrafish, antisense morpholino-based studies have indicated roles for cntn2 in the migration of facial branchiomotor (FBM) neurons, the guidance of the axons of the nucleus of the medial longitudinal fascicle (nucMLF), and the outgrowth of Rohon-Beard (RB) central axons. To study functions of Cntn2 in later stages of neuronal development, we generated cntn2 mutant zebrafish using CRISPR-Cas9. Using a null mutant allele, we detected genetic interactions between cntn2 and the planar cell polarity gene vangl2, as shown previously with cntn2 morphants, demonstrating a function for cntn2 during FBM neuron migration in a sensitized background of reduced planar cell polarity signaling. In addition, maternal-zygotic (MZ) cntn2 mutant larvae exhibited aberrant touch responses and swimming, suggestive of defects in sensorimotor circuits, consistent with studies in mice. However, the nucMLF axon convergence, FBM neuron migration, and RB outgrowth defects seen in morphants were not seen in the mutants, and we show here that they are likely off-target effects of morpholinos. However, MLF axons exhibited local defasciculation in MZcntn2 mutants, consistent with a role for Cntn2 in axon fasciculation. These data demonstrate distinct roles for zebrafish cntn2 in neuronal migration and axon fasciculation, and in the function of sensorimotor circuits.
Collapse
Affiliation(s)
- Suman Gurung
- Division of Biological Sciences and Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Emilia Asante
- Division of Biological Sciences and Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Devynn Hummel
- Division of Biological Sciences and Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Ashley Williams
- Department of Biology, Georgia Southern University, Statesboro, GA 30458, USA
| | - Oren Feldman-Schultz
- Department of Integrative Biology, Department of Neuroscience, University of Wisconsin, Madison, WI 53706, USA
| | - Mary C Halloran
- Department of Integrative Biology, Department of Neuroscience, University of Wisconsin, Madison, WI 53706, USA
| | - Vinoth Sittaramane
- Department of Biology, Georgia Southern University, Statesboro, GA 30458, USA
| | - Anand Chandrasekhar
- Division of Biological Sciences and Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA.
| |
Collapse
|
9
|
Moog NK, Entringer S, Heim C, Wadhwa PD, Kathmann N, Buss C. Influence of maternal thyroid hormones during gestation on fetal brain development. Neuroscience 2017; 342:68-100. [PMID: 26434624 PMCID: PMC4819012 DOI: 10.1016/j.neuroscience.2015.09.070] [Citation(s) in RCA: 253] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 09/22/2015] [Accepted: 09/25/2015] [Indexed: 01/09/2023]
Abstract
Thyroid hormones (THs) play an obligatory role in many fundamental processes underlying brain development and maturation. The developing embryo/fetus is dependent on maternal supply of TH. The fetal thyroid gland does not commence TH synthesis until mid gestation, and the adverse consequences of severe maternal TH deficiency on offspring neurodevelopment are well established. Recent evidence suggests that even more moderate forms of maternal thyroid dysfunction, particularly during early gestation, may have a long-lasting influence on child cognitive development and risk of neurodevelopmental disorders. Moreover, these observed alterations appear to be largely irreversible after birth. It is, therefore, important to gain a better understanding of the role of maternal thyroid dysfunction on offspring neurodevelopment in terms of the nature, magnitude, time-specificity, and context-specificity of its effects. With respect to the issue of context specificity, it is possible that maternal stress and stress-related biological processes during pregnancy may modulate maternal thyroid function. The possibility of an interaction between the thyroid and stress systems in the context of fetal brain development has, however, not been addressed to date. We begin this review with a brief overview of TH biology during pregnancy and a summary of the literature on its effect on the developing brain. Next, we consider and discuss whether and how processes related to maternal stress and stress biology may interact with and modify the effects of maternal thyroid function on offspring brain development. We synthesize several research areas and identify important knowledge gaps that may warrant further study. The scientific and public health relevance of this review relates to achieving a better understanding of the timing, mechanisms and contexts of thyroid programing of brain development, with implications for early identification of risk, primary prevention and intervention.
Collapse
Affiliation(s)
- N K Moog
- Department of Medical Psychology, Charité University Medicine Berlin, Luisenstrasse 57, 10117 Berlin, Germany
| | - S Entringer
- Department of Medical Psychology, Charité University Medicine Berlin, Luisenstrasse 57, 10117 Berlin, Germany; University of California, Irvine, Development, Health, and Disease Research Program, 333 The City Drive West, Suite 1200, Orange, CA 92868, USA; Department of Pediatrics, University of California, Irvine, School of Medicine, 505 South Main Street, Suite 525, Orange, CA 92868, USA
| | - C Heim
- Department of Medical Psychology, Charité University Medicine Berlin, Luisenstrasse 57, 10117 Berlin, Germany; Department of Biobehavioral Health, Pennsylvania State University, College of Health and Human Development, 219 Biobehavioral Health Building, University Park, PA 16802, USA
| | - P D Wadhwa
- University of California, Irvine, Development, Health, and Disease Research Program, 333 The City Drive West, Suite 1200, Orange, CA 92868, USA; Department of Pediatrics, University of California, Irvine, School of Medicine, 505 South Main Street, Suite 525, Orange, CA 92868, USA; Department of Psychiatry and Human Behavior, University of California, Irvine, School of Medicine, 3117 Gillespie Neuroscience Research Facility, 837 Health Sciences Drive, Irvine, CA 92697, USA; Department of Obstetrics and Gynecology, University of California, Irvine, School of Medicine, 3117 Gillespie Neuroscience Research Facility, 837 Health Sciences Drive, Irvine, CA 92697, USA; Department of Epidemiology, University of California, Irvine, School of Medicine, 3117 Gillespie Neuroscience Research Facility, 837 Health Sciences Drive, Irvine, CA 92697, USA
| | - N Kathmann
- Department of Clinical Psychology, Humboldt-Universität zu Berlin, Rudower Chaussee 18, 12489 Berlin, Germany
| | - C Buss
- Department of Medical Psychology, Charité University Medicine Berlin, Luisenstrasse 57, 10117 Berlin, Germany; University of California, Irvine, Development, Health, and Disease Research Program, 333 The City Drive West, Suite 1200, Orange, CA 92868, USA; Department of Pediatrics, University of California, Irvine, School of Medicine, 505 South Main Street, Suite 525, Orange, CA 92868, USA.
| |
Collapse
|
10
|
Abstract
Contactin-2/transiently expressed axonal surface glycoprotein-1 (TAG-1) is a cell adhesion molecule belonging to the immunoglobulin superfamily (IgSF). It has six immunoglobulin-like extracellular domains and four fibronectin III-like ones, with anchoring to the cell membrane through glycosylphosphatidyl inositol. Contactin-2/TAG-1 is expressed in specific neurons transiently on the axonal surface during the fetal period. In postnatal stages, Contactin-2/TAG-1 is expressed in cerebellar granule cells, hippocampal pyramidal cells, and the juxtaparanodal regions of myelinated nerve fibers. In the embryonic nervous system, Contactin-2/TAG-1 plays important roles in axonal elongation, axonal guidance, and cellular migration. In the postnatal nervous system, it also plays an essential role in the formation of myelinated nerve fibers. Moreover, Contactin-2/TAG-1 has been linked to autoimmune diseases of the human nervous system. Taken together, Contactin-2/TAG-1 plays a central role in a variety of functions from development to disease.
Collapse
Affiliation(s)
- Tomoyuki Masuda
- a Doctoral and Master's Programs in Kansei, Behavioral and Brain Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba , Ibaraki , Japan.,b Department of Neurology , Faculty of Medicine, University of Tsukuba , Ibaraki , Japan.,c Department of Neurobiology , Faculty of Medicine, University of Tsukuba , Ibaraki , Japan
| |
Collapse
|
11
|
de Ramon Francàs G, Zuñiga NR, Stoeckli ET. The spinal cord shows the way - How axons navigate intermediate targets. Dev Biol 2016; 432:43-52. [PMID: 27965053 DOI: 10.1016/j.ydbio.2016.12.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 11/26/2016] [Accepted: 12/01/2016] [Indexed: 12/13/2022]
Abstract
Functional neural circuits depend on the establishment of specific connections between neurons and their target cells. To this end, many axons have to travel long distances to reach their target cells during development. Studies addressing the molecular mechanisms of axon guidance have to overcome the complexity of subpopulation-specific requirements with respect to pathways, guidance cues, and target recognition. Compared to the brain, the relatively simple structure of the spinal cord provides an advantage for experimental studies of axon guidance mechanisms. Therefore, the so far best understood model for axon guidance is the dI1 population of dorsal interneurons of the spinal cord. They extend their axons ventrally towards the floor plate. After midline crossing, they turn rostrally along the contralateral floor-plate border. Despite the fact that the trajectory of dI1 axons seems to be rather simple, the number of axon guidance molecules involved in the decisions taken by these axons is bewildering. Because guidance molecules and mechanisms are conserved throughout the developing nervous system, we can generalize what we have learned about the navigation of the floor plate as an intermediate target for commissural axons to the brain.
Collapse
Affiliation(s)
- Gemma de Ramon Francàs
- University of Zurich, Department of Molecular Life Sciences and Neuroscience Center Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Nikole R Zuñiga
- University of Zurich, Department of Molecular Life Sciences and Neuroscience Center Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Esther T Stoeckli
- University of Zurich, Department of Molecular Life Sciences and Neuroscience Center Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| |
Collapse
|
12
|
Frei JA, Stoeckli ET. SynCAMs - From axon guidance to neurodevelopmental disorders. Mol Cell Neurosci 2016; 81:41-48. [PMID: 27594578 DOI: 10.1016/j.mcn.2016.08.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 08/28/2016] [Accepted: 08/31/2016] [Indexed: 12/22/2022] Open
Abstract
Many cell adhesion molecules are located at synapses but only few of them can be considered synaptic cell adhesion molecules in the strict sense. Besides the Neurexins and Neuroligins, the LRRTMs (leucine rich repeat transmembrane proteins) and the SynCAMs/CADMs can induce synapse formation when expressed in non-neuronal cells and therefore are true synaptic cell adhesion molecules. SynCAMs (synaptic cell adhesion molecules) are a subfamily of the immunoglobulin superfamily of cell adhesion molecules. As suggested by their name, they were first identified as cell adhesion molecules at the synapse which were sufficient to trigger synapse formation. They also contribute to myelination by mediating axon-glia cell contacts. More recently, their role in earlier stages of neural circuit formation was demonstrated, as they also guide axons both in the peripheral and in the central nervous system. Mutations in SynCAM genes were found in patients diagnosed with autism spectrum disorders. The diverse functions of SynCAMs during development suggest that neurodevelopmental disorders are not only due to defects in synaptic plasticity. Rather, early steps of neural circuit formation are likely to contribute.
Collapse
Affiliation(s)
- Jeannine A Frei
- Hussman Institute for Autism, 801 W Baltimore Street, Baltimore, MD 20201, United States
| | - Esther T Stoeckli
- Dept of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| |
Collapse
|
13
|
Huang TN, Hsueh YP. Brain-specific transcriptional regulator T-brain-1 controls brain wiring and neuronal activity in autism spectrum disorders. Front Neurosci 2015; 9:406. [PMID: 26578866 PMCID: PMC4630302 DOI: 10.3389/fnins.2015.00406] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 10/12/2015] [Indexed: 12/25/2022] Open
Abstract
T-brain-1 (TBR1) is a brain-specific T-box transcription factor. In 1995, Tbr1 was first identified from a subtractive hybridization that compared mouse embryonic and adult telencephalons. Previous studies of Tbr1−∕− mice have indicated critical roles for TBR1 in the development of the cerebral cortex, amygdala, and olfactory bulb. Neuronal migration and axonal projection are two important developmental features controlled by TBR1. Recently, recurrent de novo disruptive mutations in the TBR1 gene have been found in patients with autism spectrum disorders (ASDs). Human genetic studies have identified TBR1 as a high-confidence risk factor for ASDs. Because only one allele of the TBR1 gene is mutated in these patients, Tbr1+∕− mice serve as a good genetic mouse model to explore the mechanism by which de novo TBR1 mutation leads to ASDs. Although neuronal migration and axonal projection defects of cerebral cortex are the most prominent phenotypes in Tbr1−∕− mice, these features are not found in Tbr1+∕− mice. Instead, inter- and intra-amygdalar axonal projections and NMDAR expression and activity in amygdala are particularly susceptible to Tbr1 haploinsufficiency. The studies indicated that both abnormal brain wiring (abnormal amygdalar connections) and excitation/inhibition imbalance (NMDAR hypoactivity), two prominent models for ASD etiology, are present in Tbr1+∕− mice. Moreover, calcium/calmodulin-dependent serine protein kinase (CASK) was found to interact with TBR1. The CASK–TBR1 complex had been shown to directly bind the promoter of the Grin2b gene, which is also known as Nmdar2b, and upregulate Grin2b expression. This molecular function of TBR1 provides an explanation for NMDAR hypoactivity in Tbr1+∕− mice. In addition to Grin2b, cell adhesion molecules—including Ntng1, Cdh8, and Cntn2—are also regulated by TBR1 to control axonal projections of amygdala. Taken together, the studies of Tbr1 provide an integrated picture of ASD etiology at the cellular and circuit levels.
Collapse
Affiliation(s)
- Tzyy-Nan Huang
- Institute of Molecular Biology, Academia Sinica Taipei, Taiwan
| | - Yi-Ping Hsueh
- Institute of Molecular Biology, Academia Sinica Taipei, Taiwan
| |
Collapse
|
14
|
Frei JA, Andermatt I, Gesemann M, Stoeckli ET. The SynCAM synaptic cell adhesion molecules are involved in sensory axon pathfinding by regulating axon-axon contacts. J Cell Sci 2014; 127:5288-302. [PMID: 25335893 DOI: 10.1242/jcs.157032] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Synaptic cell adhesion molecules (SynCAMs) are crucial for synapse formation and plasticity. However, we have previously demonstrated that SynCAMs are also required during earlier stages of neural circuit formation because SynCAM1 and SynCAM2 (also known as CADM1 and CADM2, respectively) are important for the guidance of post-crossing commissural axons. In contrast to the exclusively homophilic cis-interactions reported by previous studies, our previous in vivo results suggested the existence of heterophilic cis-interactions between SynCAM1 and SynCAM2. Indeed, as we show here, the presence of homophilic and heterophilic cis-interactions modulates the interaction of SynCAMs with trans-binding partners, as observed previously for other immunoglobulin superfamily cell adhesion molecules. These in vitro findings are in agreement with results from in vivo studies, which demonstrate a role for SynCAMs in the formation of sensory neural circuits in the chicken embryo. In the absence of SynCAMs, selective axon-axon interactions are perturbed resulting in aberrant pathfinding of sensory axons.
Collapse
Affiliation(s)
- Jeannine A Frei
- Institute of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Irwin Andermatt
- Institute of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Matthias Gesemann
- Institute of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Esther T Stoeckli
- Institute of Molecular Life Sciences and Neuroscience Center Zurich, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
15
|
Frei JA, Stoeckli ET. SynCAMs extend their functions beyond the synapse. Eur J Neurosci 2014; 39:1752-60. [DOI: 10.1111/ejn.12544] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 01/17/2014] [Accepted: 02/03/2014] [Indexed: 02/06/2023]
Affiliation(s)
- Jeannine A. Frei
- Institute of Molecular Life Sciences and Neuroscience Center Zurich; University of Zurich; Winterthurerstrasse 190 8057 Zurich Switzerland
| | - Esther T. Stoeckli
- Institute of Molecular Life Sciences and Neuroscience Center Zurich; University of Zurich; Winterthurerstrasse 190 8057 Zurich Switzerland
| |
Collapse
|
16
|
Tbr1 haploinsufficiency impairs amygdalar axonal projections and results in cognitive abnormality. Nat Neurosci 2014; 17:240-7. [PMID: 24441682 DOI: 10.1038/nn.3626] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 12/11/2013] [Indexed: 12/12/2022]
Abstract
The neuron-specific transcription factor T-box brain 1 (TBR1) regulates brain development. Disruptive mutations in the TBR1 gene have been repeatedly identified in patients with autism spectrum disorders (ASDs). Here, we show that Tbr1 haploinsufficiency results in defective axonal projections of amygdalar neurons and the impairment of social interaction, ultrasonic vocalization, associative memory and cognitive flexibility in mice. Loss of a copy of the Tbr1 gene altered the expression of Ntng1, Cntn2 and Cdh8 and reduced both inter- and intra-amygdalar connections. These developmental defects likely impair neuronal activation upon behavioral stimulation, which is indicated by fewer c-FOS-positive neurons and lack of GRIN2B induction in Tbr1(+/-) amygdalae. We also show that upregulation of amygdalar neuronal activity by local infusion of a partial NMDA receptor agonist, d-cycloserine, ameliorates the behavioral defects of Tbr1(+/-) mice. Our study suggests that TBR1 is important in the regulation of amygdalar axonal connections and cognition.
Collapse
|
17
|
Stoeckli ET, Kilinc D, Kunz B, Kunz S, Lee GU, Martines E, Rader C, Suter D. Analysis of cell-cell contact mediated by Ig superfamily cell adhesion molecules. CURRENT PROTOCOLS IN CELL BIOLOGY 2013; 61:9.5.1-9.5.85. [PMID: 24510806 DOI: 10.1002/0471143030.cb0905s61] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Cell-cell adhesion is a fundamental requirement for all multicellular organisms. The calcium-independent cell adhesion molecules of the immunoglobulin superfamily (IgSF-CAMs) represent a major subgroup. They consist of immunoglobulin folds alone or in combination with other protein modules, often fibronectin type-III folds. More than 100 IgSF-CAMs have been identified in vertebrates and invertebrates. Most of the IgSF-CAMs are cell surface molecules that are membrane-anchored either by a single transmembrane segment or by a glycosylphosphatidylinositol (GPI) anchor. Some of the IgSF-CAMs also occur in soluble form, e.g., in the cerebrospinal fluid or in the vitreous fluid of the eye, due to naturally occurring cleavage of the GPI anchor or the membrane-proximal peptide segment. Some IgSF-CAMs, such as NCAM, occur in various forms that are generated by alternative splicing. This unit contains a series of protocols that have been used to study the function of IgSF-CAMs in vitro and in vivo.
Collapse
Affiliation(s)
- Esther T Stoeckli
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Devrim Kilinc
- School of Chemistry and Chemical Biology, University College Dublin, Belfield, Dublin, Ireland
| | - Beat Kunz
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Stefan Kunz
- Institute of Microbiology, University Hospital Center and University of Lausanne, Lausanne, Switzerland
| | - Gil U Lee
- School of Chemistry and Chemical Biology, University College Dublin, Belfield, Dublin, Ireland
| | - Elena Martines
- Nanomedicine Centre, School of Chemistry and Chemical Biology, University College Dublin, Dublin, Ireland
| | - Christoph Rader
- Department of Cancer Biology, Scripps Florida, Jupiter, Florida
| | - Daniel Suter
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana
| |
Collapse
|
18
|
Expanding the Ig superfamily code for laminar specificity in retina: expression and role of contactins. J Neurosci 2013; 32:14402-14. [PMID: 23055510 DOI: 10.1523/jneurosci.3193-12.2012] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Bipolar, amacrine, and retinal ganglion cells elaborate arbors and form synapses within the inner plexiform layer (IPL) of the vertebrate retina. Specific subsets of these neuronal types synapse in one or a few of the ≥10 sublaminae of the IPL. Four closely related Ig superfamily transmembrane adhesion molecules--Sidekick1 (Sdk1), Sdk2, Dscam, and DscamL--are expressed by non-overlapping subsets of chick retinal neurons and promote their lamina-specific arborization (Yamagata and Sanes, 2008). Here, we asked whether contactins (Cntns), six homologs of Sdks and Dscams, are expressed by and play roles in other subsets. In situ hybridization showed that cntn1-5 were differentially expressed by subsets of amacrine cells. Immunohistochemistry showed that each Cntn protein was concentrated in a subset of IPL sublaminae. To assess roles of Cntns in retinal development, we focused on Cntn2. Depletion of Cntn2 by RNA interference markedly reduced the ability of Cntn2-positive cells to restrict their arbors to appropriate sublaminae. Conversely, ectopic expression of cntn2 redirected neurites of transduced neurons to the Cntn2-positive sublaminae. Thus, both loss- and gain-of-function strategies implicate Cntn2 in lamina-specific neurite targeting. Studies in heterologous cells showed that Cntn2 mediates homophilic adhesion, but does not bind detectably to Sdks, Dscams, or other Cntns. Overexpression analysis showed that Cntns1 and 3 can also redirect neurites to appropriate sublaminae. We propose that Cntns, Sdks, and Dscams comprise an Ig superfamily code that uses homophilic interactions to promote lamina-specific targeting of retinal dendrites in IPL.
Collapse
|
19
|
Stoeckli ET. Neural circuit formation in the cerebellum is controlled by cell adhesion molecules of the Contactin family. Cell Adh Migr 2011; 4:523-6. [PMID: 20622526 DOI: 10.4161/cam.4.4.12733] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Cell adhesion molecules of the immunoglobulin superfamily (IgSF CAMs) have been implicated in neural circuit formation in both the peripheral and the central nervous system. Several recent studies highlight a role of the Contactin group of IgSF CAMs in cerebellar development, in particular in the development of granule cells. Granule cells are the most numerous type of neurons in the nervous system and by forming a secondary proliferative zone in the cerebellum they provide an exception to the rule that neuronal precursors proliferate in the ventricular zone. Granule cells express Contactin-2, Contactin-1, and Contactin-6 in a sequential manner. Contactins are required for axon guidance, fasciculation, and synaptogenesis, and thus affect multiple steps in neural circuit formation in the developing cerebellum.
Collapse
Affiliation(s)
- Esther T Stoeckli
- University of Zurich, Institute of Molecular Life Sciences, Zurich, Switzerland.
| |
Collapse
|
20
|
Itoh K, Fujisaki K, Watanabe M. Human L1CAM carrying the missense mutations of the fibronectin-like type III domains is localized in the endoplasmic reticulum and degraded by polyubiquitylation. J Neurosci Res 2011; 89:1637-45. [PMID: 21688291 DOI: 10.1002/jnr.22695] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 04/11/2011] [Accepted: 04/15/2011] [Indexed: 11/09/2022]
Abstract
Any mutations in the human neural cell adhesion molecule L1 (hL1CAM) gene might cause various types of serious neurological syndromes in humans, characterized by increased mortality, mental retardation, and various malformations of the nervous system. Such missense mutations often cause severe abnormalities or even fatalities, and the reason for this may be a disruption of the adhesive function of L1CAM resulting from a misdirection of the degradative pathway. Transfection studies using neuroblastoma N2a cells demonstrated that hL1CAM carrying the missense mutations in the fibronectin-like type III (FnIII) domains most likely is located within the endoplasmic reticulum (ER), but it is less well expressed on the cell surface. One mutant, L935P, in the fourth FnIII domain, was chosen from six mutants (K655 and G698 at Fn1, L935P and P941 at Fn4, W1036 and Y1070 at Fn5) in the FnIII domains to study in detail the functions of hL1CAM(200 kDa) , such as the intracellular traffic and degradation, because only a single band at 200 kDa was detected in the hL1CAM(L935P) -transfected cells. hL1CAM(200 kDa) is expressed predominantly in the ER but not on the cell surface. In addition, this missense mutated hL1CAM(200 kDa) is polyubiquitylated at some sites in the extracellular domain and thus becomes degraded by proteasomes via the ER-associated degradation pathway. These observations demonstrate that the missense mutations of hL1CAM in the FnIII domain may cause the resultant pathogenesis because of a loss of expression on the cell surface resulting from misrouting to the degradative pathway.
Collapse
Affiliation(s)
- Kouichi Itoh
- Laboratory of Molecular and Cellular Neurosciences, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki-city, Kagawa, Japan.
| | | | | |
Collapse
|
21
|
Expression of the IgSF protein Kirre in the rat central nervous system. Life Sci 2011; 88:590-7. [DOI: 10.1016/j.lfs.2011.01.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Revised: 12/17/2010] [Accepted: 01/14/2011] [Indexed: 11/23/2022]
|
22
|
The expression of TAG-1 in glial cells is sufficient for the formation of the juxtaparanodal complex and the phenotypic rescue of tag-1 homozygous mutants in the CNS. J Neurosci 2010; 30:13943-54. [PMID: 20962216 DOI: 10.1523/jneurosci.2574-10.2010] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Myelinated fibers are organized into specialized domains that ensure the rapid propagation of action potentials and are characterized by protein complexes underlying axoglial interactions. TAG-1 (Transient Axonal Glycoprotein-1), a cell adhesion molecule of the Ig superfamily, is expressed by neurons as well as by myelinating glia. It is essential for the molecular organization of myelinated fibers as it maintains the integrity of the juxtaparanodal region through its interactions with Caspr2 and the voltage-gated potassium channels (VGKCs) on the axolemma. Since TAG-1 is the only known component of the juxtaparanodal complex expressed by the glial cell, it is important to clarify its role in the molecular organization of juxtaparanodes. For this purpose, we generated transgenic mice that exclusively express TAG-1 in oligodendrocytes and lack endogenous gene expression (Tag-1(-/-);plp(Tg(rTag-1))). Phenotypic analysis clearly demonstrates that glial TAG-1 is sufficient for the proper organization and maintenance of the juxtaparanodal domain in the CNS. Biochemical analysis shows that glial TAG-1 physically interacts with Caspr2 and VGKCs. Ultrastructural and behavioral analysis of Tag-1(-/-);plp(Tg(rTag-1)) mice shows that the expression of glial TAG-1 is sufficient to restore the axonal and myelin deficits as well as the behavioral defects observed in Tag-1(-/-) animals. Together, these data highlight the pivotal role of myelinating glia on axonal domain differentiation and organization.
Collapse
|
23
|
Liu H, Focia PJ, He X. Homophilic adhesion mechanism of neurofascin, a member of the L1 family of neural cell adhesion molecules. J Biol Chem 2010; 286:797-805. [PMID: 21047790 DOI: 10.1074/jbc.m110.180281] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The L1 family neural cell adhesion molecules play key roles in specifying the formation and remodeling of the neural network, but their homophilic interaction that mediates adhesion is not well understood. We report two crystal structures of a dimeric form of the headpiece of neurofascin, an L1 family member. The four N-terminal Ig-like domains of neurofascin form a horseshoe shape, akin to several other immunoglobulin superfamily cell adhesion molecules such as hemolin, axonin, and Dscam. The neurofascin dimer, captured in two crystal forms with independent packing patterns, reveals a pair of horseshoes in trans-synaptic adhesion mode. The adhesion interaction is mediated mostly by the second Ig-like domain, which features an intermolecular β-sheet formed by the joining of two individual GFC β-sheets and a large but loosely packed hydrophobic cluster. Mutagenesis combined with gel filtration assays suggested that the side chain hydrogen bonds at the intermolecular β-sheet are essential for the homophilic interaction and that the residues at the hydrophobic cluster play supplementary roles. Our structures reveal a conserved homophilic adhesion mode for the L1 family and also shed light on how the pathological mutations of L1 affect its structure and function.
Collapse
Affiliation(s)
- Heli Liu
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | | | | |
Collapse
|
24
|
Barry J, Gu Y, Gu C. Polarized targeting of L1-CAM regulates axonal and dendritic bundling in vitro. Eur J Neurosci 2010; 32:1618-31. [PMID: 20964729 DOI: 10.1111/j.1460-9568.2010.07447.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Proper axonal and dendritic bundling is essential for the establishment of neuronal connections and the synchronization of synaptic inputs, respectively. Cell adhesion molecules of the L1-CAM (L1-cell adhesion molecule) family regulate axon guidance and fasciculation, neuron migration, dendrite morphology, and synaptic plasticity. It remains unclear how these molecules play so many different roles. Here we show that polarized axon-dendrite targeting of an avian L1-CAM protein, NgCAM (neuron-glia cell adhesion molecule), can regulate the switch of bundling of the two major compartments of rat hippocampal neurons. Using a new in-vitro model for studying neurite-neurite interactions, we found that expressed axonal NgCAM induced robust axonal bundling via the trans-homophilic interaction of immunoglobulin domains. Interestingly, dendritic bundling was induced by the dendritic targeting of NgCAM, caused by either deleting its fibronectin repeats or blocking activities of protein kinases. Consistent with the NgCAM results, expression of mouse L1-CAM also induced axonal bundling and blocking kinase activities disrupted its axonal targeting. Furthermore, the trans-homophilic interaction stabilized the bundle formation, probably through recruiting NgCAM proteins to contact sites and promoting guided axon outgrowth. Taken together, our results suggest that precise localization of L1-CAM is important for establishing proper cell-cell contacts in neural circuits.
Collapse
Affiliation(s)
- Joshua Barry
- Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, USA
| | | | | |
Collapse
|
25
|
Tapanes-Castillo A, Weaver EJ, Smith RP, Kamei Y, Caspary T, Hamilton-Nelson KL, Slifer SH, Martin ER, Bixby JL, Lemmon VP. A modifier locus on chromosome 5 contributes to L1 cell adhesion molecule X-linked hydrocephalus in mice. Neurogenetics 2010; 11:53-71. [PMID: 19565280 PMCID: PMC2863031 DOI: 10.1007/s10048-009-0203-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2009] [Accepted: 06/08/2009] [Indexed: 12/25/2022]
Abstract
Humans with L1 cell adhesion molecule (L1CAM) mutations exhibit X-linked hydrocephalus, as well as other severe neurological disorders. L1-6D mutant mice, which are homozygous for a deletion that removes the sixth immunoglobulin-like domain of L1cam, seldom display hydrocephalus on the 129/Sv background. However, the same L1-6D mutation produces severe hydrocephalus on the C57BL/6J background. To begin to understand how L1cam deficiencies result in hydrocephalus and to identify modifier loci that contribute to X-linked hydrocephalus by genetically interacting with L1cam, we conducted a genome-wide scan on F2 L1-6D mice, bred from L1-6D 129S2/SvPasCrlf and C57BL/6J mice. Linkage studies, utilizing chi-square tests and quantitative trait loci mapping techniques, were performed. Candidate modifier loci were further investigated in an extension study. Linkage was confirmed for a locus on chromosome 5, which we named L1cam hydrocephalus modifier 1 (L1hydro1), p = 4.04 X 10(-11).
Collapse
Affiliation(s)
- Alexis Tapanes-Castillo
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Lois Pope LIFE Center, Room 4-16, 1095 NW 14th Terrace, Miami, FL 33136, USA
| | - Eli J. Weaver
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Lois Pope LIFE Center, Room 4-16, 1095 NW 14th Terrace, Miami, FL 33136, USA, Department of Neuroscience, Case Western Reserve University, Cleveland, OH, USA
| | - Robin P. Smith
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Lois Pope LIFE Center, Room 4-16, 1095 NW 14th Terrace, Miami, FL 33136, USA, Neuroscience Program, University of Miami, Miami, FL, USA
| | - Yoshimasa Kamei
- Department of Neuroscience, Case Western Reserve University, Cleveland, OH, USA
| | - Tamara Caspary
- Department of Human Genetics, Emory University, Atlanta, GA, USA
| | - Kara L. Hamilton-Nelson
- Dr. John T. MacDonald Foundation, Department of Human Genetics, Miami Institute for Human Genomics, University of Miami, Miami, FL, USA
| | - Susan H. Slifer
- Dr. John T. MacDonald Foundation, Department of Human Genetics, Miami Institute for Human Genomics, University of Miami, Miami, FL, USA
| | - Eden R. Martin
- Dr. John T. MacDonald Foundation, Department of Human Genetics, Miami Institute for Human Genomics, University of Miami, Miami, FL, USA
| | - John L. Bixby
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Lois Pope LIFE Center, Room 4-16, 1095 NW 14th Terrace, Miami, FL 33136, USA, Neuroscience Program, University of Miami, Miami, FL, USA, Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA, Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Vance P. Lemmon
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Lois Pope LIFE Center, Room 4-16, 1095 NW 14th Terrace, Miami, FL 33136, USA, Department of Neuroscience, Case Western Reserve University, Cleveland, OH, USA, Neuroscience Program, University of Miami, Miami, FL, USA, Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
26
|
Nielsen J, Kulahin N, Walmod PS. Extracellular protein interactions mediated by the neural cell adhesion molecule, NCAM: heterophilic interactions between NCAM and cell adhesion molecules, extracellular matrix proteins, and viruses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 663:23-53. [PMID: 20017013 DOI: 10.1007/978-1-4419-1170-4_2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Janne Nielsen
- Protein Laboratory, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | | | | |
Collapse
|
27
|
Lewis(x) and alpha2,3-sialyl glycans and their receptors TAG-1, Contactin, and L1 mediate CD24-dependent neurite outgrowth. J Neurosci 2009; 29:6677-90. [PMID: 19458237 DOI: 10.1523/jneurosci.4361-08.2009] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Although carbohydrates have been implicated in cell interactions in the nervous system, the molecular bases of their functions have remained largely obscure. Here, we show that promotion or inhibition of neurite outgrowth of cerebellar or dorsal root ganglion neurons, respectively, induced by the mucin-type adhesion molecule CD24 depends on alpha2,3-linked sialic acid and Lewis(x) present on glia-specific CD24 glycoforms. Alpha2,3-sialyl residues of CD24 bind to a structural motif in the first fibronectin type III domain of the adhesion molecule L1. Following the observation that the adhesion molecules TAG-1 and Contactin show sequence homologies with fucose-specific lectins, we obtained evidence that TAG-1 and Contactin mediate Lewis(x)-dependent CD24-induced effects on neurite outgrowth. Thus, L1, TAG-1, and Contactin function as lectin-like neuronal receptors. Their cis interactions with neighboring adhesion molecules, e.g., Caspr1 and Caspr2, and with their triggered signal transduction pathways elicit cell type-specific promotion or inhibition of neurite outgrowth induced by glial CD24 in a glycan-dependent trans interaction.
Collapse
|
28
|
Shimoda Y, Watanabe K. Contactins: emerging key roles in the development and function of the nervous system. Cell Adh Migr 2009; 3:64-70. [PMID: 19262165 DOI: 10.4161/cam.3.1.7764] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Contactins are a subgroup of molecules belonging to the immunoglobulin superfamily that are expressed exclusively in the nervous system. The subgroup consists of six members: contactin, TAG-1, BIG-1, BIG-2, NB-2 and NB-3. Since their identification in the late 1980s, contactin and TAG-1 have been studied extensively. Axonal expression and the neurite extension activity of contactin and TAG-1 attracted researchers to study the function of these molecules in axon guidance during development. After the exciting discovery of the molecular function of contactin and TAG-1 in myelination earlier this decade, these two molecules have come to be known as the principal molecules in the function and maintenance of myelinated neurons. In contrast, the function of the other four members of this subgroup remained unknown until recently. Here, we will give an overview of contactin function, including recent progress on BIG-2, NB-2 and NB-3.
Collapse
Affiliation(s)
- Yasushi Shimoda
- Department of Bioengineering, Nagaoka University of Technology, Nagaoka, Niigata, Japan
| | | |
Collapse
|
29
|
Bizzoca A, Corsi P, Gennarini G. The mouse F3/contactin glycoprotein: structural features, functional properties and developmental significance of its regulated expression. Cell Adh Migr 2009; 3:53-63. [PMID: 19372728 PMCID: PMC2675150 DOI: 10.4161/cam.3.1.7462] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2008] [Accepted: 11/19/2008] [Indexed: 12/18/2022] Open
Abstract
F3/Contactin is an immunoglobulin superfamily component expressed in the nervous tissue of several species. Here we focus on the structural and functional properties of its mouse relative, on the mechanisms driving its regulated expression and on its developmental role. F3/Contactin is differentially expressed in distinct populations of central and peripheral neurons and in some non-neuronal cells. Accordingly, the regulatory region of the underlying gene includes promoter elements undergoing differential activation, associated with an intricate splicing profile, indicating that transcriptional and posttranscriptional mechanisms contribute to its expression. Transgenic models allowed to follow F3/Contactin promoter activation in vivo and to modify F3/Contactin gene expression under a heterologous promoter, which resulted in morphological and functional phenotypes. Besides axonal growth and pathfinding, these concerned earlier events, including precursor proliferation and commitment. This wide role in neural ontogenesis is consistent with the recognized interaction of F3/Contactin with developmental control genes belonging to the Notch pathway.
Collapse
Affiliation(s)
- Antonella Bizzoca
- Department of Pharmacology and Human Physiology, Medical School, University of Bari, Bari, Italy
| | | | | |
Collapse
|
30
|
Kamiguchi H. The role of cell adhesion molecules in axon growth and guidance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 621:95-103. [PMID: 18269213 DOI: 10.1007/978-0-387-76715-4_7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
During development, axons elongate along the correct path toward their final targets. Growing axons maintain adhesive interactions with specific environmental cues via cell adhesion molecules (CAMs). The axon-environment adhesion must be dynamically controlled, both temporally and spatially, to enable the axons to navigate and migrate correctly. In this way, CAMs play a central role in mediating contact-dependent regulation of motile behavior of the axons. This chapter examines the mechanisms underlying how CAMs control axon growth and guidance, with a particular focus on intracellular signaling, trafficking, and interactions with the actin cytoskeleton.
Collapse
Affiliation(s)
- Hiroyuki Kamiguchi
- Laboratory for Neuronal Growth Mechanisms, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan.
| |
Collapse
|
31
|
Structural requirement of TAG-1 for retinal ganglion cell axons and myelin in the mouse optic nerve. J Neurosci 2008; 28:7624-36. [PMID: 18650339 DOI: 10.1523/jneurosci.1103-08.2008] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
White matter axons organize into fascicles that grow over long distances and traverse very diverse environments. The molecular mechanisms preserving this structure of white matter axonal tracts are not well known. Here, we used the optic nerve as a model and investigated the role of TAG-1, a cell adhesion molecule expressed by retinal axons. TAG-1 was first expressed in the embryonic retinal ganglion cells (RGCs) and later in the postnatal myelin-forming cells in the optic nerve. We describe the consequences of genetic loss of Tag-1 on the developing and adult retinogeniculate tract. Tag-1-null embryos display anomalies in the caliber of RGC axons, associated with an abnormal organization of the astroglial network in the optic nerve. The contralateral projections in the lateral geniculate nucleus are expanded postnatally. In the adult, Tag-1-null mice show a loss of RGC axons, with persistent abnormalities of axonal caliber and additional cytoskeleton and myelination defects. Therefore, TAG-1 is an essential regulator of the structure of RGC axons and their surrounding glial cells in the optic nerve.
Collapse
|
32
|
Savvaki M, Panagiotaropoulos T, Stamatakis A, Sargiannidou I, Karatzioula P, Watanabe K, Stylianopoulou F, Karagogeos D, Kleopa KA. Impairment of learning and memory in TAG-1 deficient mice associated with shorter CNS internodes and disrupted juxtaparanodes. Mol Cell Neurosci 2008; 39:478-90. [PMID: 18760366 DOI: 10.1016/j.mcn.2008.07.025] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2008] [Revised: 07/29/2008] [Accepted: 07/31/2008] [Indexed: 10/21/2022] Open
Abstract
The cell adhesion molecule TAG-1 is expressed by neurons and glial cells and plays a role in axon outgrowth, migration and fasciculation during development. TAG-1 is also required for the clustering of Kv1.1/1.2 potassium channels and Caspr2 at the juxtaparanodes of myelinated fibers. Behavioral examination of TAG-1 deficient mice (Tag-1(-/-)) showed cognitive impairments in the Morris water maze and novel object recognition tests, reduced spontaneous motor activity, abnormal gait coordination and increased response latency to noxious stimulation. Investigation at the molecular level revealed impaired juxtaparanodal clustering of Caspr2 and Kv1.1/1.2 in the hippocampus, entorhinal cortex, cerebellum and olfactory bulb, with diffusion into the internode. Caspr2 and Kv1.1 levels were reduced in the cerebellum and olfactory bulb. Moreover, Tag-1(-/-) mice had shorter internodes in the cerebral and cerebellar white matter. The detected molecular alterations may account for the behavioural deficits and hyperexcitability in these animals.
Collapse
Affiliation(s)
- Maria Savvaki
- Department of Basic Science, University of Crete Medical School, and Institute of Molecular Biology and Biotechnology, Heraklion, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Law CO, Kirby RJ, Aghamohammadzadeh S, Furley AJW. The neural adhesion molecule TAG-1 modulates responses of sensory axons to diffusible guidance signals. Development 2008; 135:2361-71. [DOI: 10.1242/dev.009019] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
When the axons of primary sensory neurons project into the embryonic mammalian spinal cord, they bifurcate and extend rostrocaudally before sending collaterals to specific laminae according to neuronal subclass. The specificity of this innervation has been suggested to be the result both of differential sensitivity to chemorepellants expressed in the ventral spinal cord and of the function of Ig-like neural cell adhesion molecules in the dorsal horn. The relationship between these mechanisms has not been addressed. Focussing on the pathfinding of TrkA+ NGF-dependent axons, we demonstrate for the first time that their axons project prematurely into the dorsal horn of both L1 and TAG-1 knockout mice. We show that axons lacking TAG-1, similar to those lacking L1, are insensitive to wild-type ventral spinal cord(VSC)-derived chemorepellants, indicating that adhesion molecule function is required in the axons, and that this loss of response is explained in part by loss of response to Sema3A. We present evidence that TAG-1 affects sensitivity to Sema3A by binding to L1 and modulating the endocytosis of the L1/neuropilin 1 Sema3A receptor complex. However, TAG-1 appears to affect sensitivity to other VSC-derived chemorepellants via an L1-independent mechanism. We suggest that this dependence of chemorepellant sensitivity on the functions of combinations of adhesion molecules is important to ensure that axons project via specific pathways before extending to their final targets.
Collapse
Affiliation(s)
- Chris O. Law
- Department of Biomedical Science, University of Sheffield, Western Bank,Sheffield S10 2TN, UK
| | - Rebecca J. Kirby
- Department of Biomedical Science, University of Sheffield, Western Bank,Sheffield S10 2TN, UK
| | | | - Andrew J. W. Furley
- Department of Biomedical Science, University of Sheffield, Western Bank,Sheffield S10 2TN, UK
| |
Collapse
|
34
|
Nielsen J, Kulahin N, Walmod PS. Extracellular Protein Interactions Mediated by the Neural Cell Adhesion Molecule, NCAM: Heterophilic Interactions Between NCAM and Cell Adhesion Molecules, Extracellular Matrix Proteins, and Viruses. Neurochem Res 2008. [DOI: 10.1007/s11064-008-9761-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
35
|
Seabold GK, Wang PY, Chang K, Wang CY, Wang YX, Petralia RS, Wenthold RJ. The SALM family of adhesion-like molecules forms heteromeric and homomeric complexes. J Biol Chem 2008; 283:8395-405. [PMID: 18227064 DOI: 10.1074/jbc.m709456200] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Synaptic adhesion-like molecules (SALMs) are a newly discovered family of adhesion molecules that play roles in synapse formation and neurite outgrowth. The SALM family is comprised of five homologous molecules that are expressed largely in the central nervous system. SALMs 1-3 contain PDZ-binding domains, whereas SALMs 4 and 5 do not. We are interested in characterizing the interactions of the SALMs both among the individual members and with other binding partners. In the present study, we focused on the interactions formed by the five SALM members in rat brain and heterologous cells. In brain, we found that SALMs 1-3 strongly co-immunoprecipitated with each other, whereas SALMs 4 and 5 did not, suggesting that SALMs 4 and 5 mainly form homomeric complexes. In heterologous cells transfected with SALMs, co-immunoprecipitation studies showed that all five SALMs form heteromeric and homomeric complexes. We also determined if SALMs could form trans-cellular associations between transfected heterologous cells. Both SALMs 4 and 5 formed homophilic, but not heterophilic associations, whereas no trans associations were formed by the other SALMs. The ability of SALM4 to form trans interactions is due to its extracellular N terminus because chimeras of SALM4 N terminus and SALM2 C terminus can form trans interactions, whereas chimeras of SALM2 N terminus and SALM4 C terminus cannot. Co-culture experiments using HeLa cells and rat hippocampal neurons expressing the SALMs showed that SALM4 is recruited to points of contact between the cells. In neurons, these points of contact were seen in both axons and dendrites.
Collapse
Affiliation(s)
- Gail K Seabold
- Laboratory of Neurochemistry, NIDCD, NIH, Bethesda, MD 20892-8027, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Functional dissection of the C. elegans cell adhesion molecule SAX-7, a homologue of human L1. Mol Cell Neurosci 2008; 37:56-68. [DOI: 10.1016/j.mcn.2007.08.014] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Revised: 08/08/2007] [Accepted: 08/21/2007] [Indexed: 11/24/2022] Open
|
37
|
Kulahin N, Li S, Hinsby A, Kiselyov V, Berezin V, Bock E. Fibronectin type III (FN3) modules of the neuronal cell adhesion molecule L1 interact directly with the fibroblast growth factor (FGF) receptor. Mol Cell Neurosci 2007; 37:528-36. [PMID: 18222703 DOI: 10.1016/j.mcn.2007.12.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2007] [Revised: 11/30/2007] [Accepted: 12/04/2007] [Indexed: 12/18/2022] Open
Abstract
The neuronal cell adhesion molecule (CAM) L1 promotes axonal outgrowth, presumably through an interaction with the fibroblast growth factor receptor (FGFR). The present study demonstrates a direct interaction between L1 fibronectin type III (FN3) modules I-V and FGFR1 immunoglobulin (Ig) modules II and III by surface plasmon resonance analysis. Binding of L1 to FGFR1 was enhanced by adenosine 5'-triphosphate (ATP), adenylylmethylenediphosphonate (AMP-PCP), and guanosine-5'-triphosphate (GTP), but not adenosine monophosphate (AMP). The L1-FN3 modules were capable of activating FGFR1, reflected by receptor phosphorylation, and this resulted in the induction of differentiation of primary neurons, reflected by neurite outgrowth. Furthermore, ATP modulated L1-induced neuronal differentiation and FGFR1 phosphorylation through regulation of the L1-FGFR1 interaction.
Collapse
Affiliation(s)
- Nikolaj Kulahin
- Protein Laboratory, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark.
| | | | | | | | | | | |
Collapse
|
38
|
Shapiro L, Love J, Colman DR. Adhesion molecules in the nervous system: structural insights into function and diversity. Annu Rev Neurosci 2007; 30:451-74. [PMID: 17600523 DOI: 10.1146/annurev.neuro.29.051605.113034] [Citation(s) in RCA: 153] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The unparalleled complexity of intercellular connections in the nervous system presents requirements for high levels of both specificity and diversity for the proteins that mediate cell adhesion. Here we describe recent advances toward understanding the molecular mechanisms that underlie adhesive binding, specificity, and diversity for several well-characterized families of adhesion molecules in the nervous system. Although many families of adhesion proteins, including cadherins and immunoglobulin superfamily members, are utilized in neural and nonneural contexts, nervous system-specific diversification mechanisms, such as precisely regulated alternative splicing, provide an important means to enable their function in the complex context of the nervous system.
Collapse
Affiliation(s)
- Lawrence Shapiro
- Department of Biochemistry and Molecular Biophysics, 2Edward S. Harkness Eye Institute, Columbia University, New York, New York 10032 USA.
| | | | | |
Collapse
|
39
|
Mörtl M, Sonderegger P, Diederichs K, Welte W. The crystal structure of the ligand-binding module of human TAG-1 suggests a new mode of homophilic interaction. Protein Sci 2007; 16:2174-83. [PMID: 17766378 PMCID: PMC2204121 DOI: 10.1110/ps.072802707] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Human TAG-1 is a neural cell adhesion molecule that is crucial for the development of the nervous system during embryogenesis. It consists of six immunoglobulin-like and four fibronectin III-like domains and is anchored to the membrane by glycosylphosphatidylinositol. Herein we present the crystal structure of the four N-terminal immunoglobulin-like domains of TAG-1 (TAG-1(Ig1-4)), known to be important in heterophilic and homophilic macromolecular interactions. The contacts of neighboring molecules within the crystal were investigated. A comparison with the structure of the chicken ortholog resulted in an alternative mode for the molecular mechanism of homophilic TAG-1 interaction. This mode of TAG-1 homophilic interaction is based on dimer formation rather than formation of a molecular zipper as proposed for the chicken ortholog.
Collapse
Affiliation(s)
- Mario Mörtl
- University of Konstanz, Department of Biology, Konstanz, Germany
| | | | | | | |
Collapse
|
40
|
Yamanaka H, Obata K, Kobayashi K, Dai Y, Fukuoka T, Noguchi K. Alteration of the cell adhesion molecule L1 expression in a specific subset of primary afferent neurons contributes to neuropathic pain. Eur J Neurosci 2007; 25:1097-111. [PMID: 17331206 PMCID: PMC1891330 DOI: 10.1111/j.1460-9568.2007.05344.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The cell adhesion molecule L1 (L1-CAM) plays important functional roles in the developing and adult nervous systems. Here we show that peripheral nerve injury induced dynamic post-transcriptional alteration of L1-CAM in the rat dorsal root ganglia (DRGs) and spinal cord. Sciatic nerve transection (SCNT) changed the expression of L1-CAM protein but not L1-CAM mRNA. In DRGs, SCNT induced accumulation of the L1-CAM into the surface of somata, which resulted in the formation of immunoreactive ring structures in a number of unmyelinated C-fiber neurons. These neurons with L1-CAM-immunoreactive ring structures were heavily colocalized with phosphorylated p38 MAPK. Western blot analysis revealed the increase of full-length L1-CAM and decrease of fragments of L1-CAM after SCNT in DRGs. Following SCNT, L1-CAM-immunoreactive profiles in the dorsal horn showed an increase mainly in pre-synaptic areas of laminae I–II with a delayed onset and colocalized with growth-associated protein 43. In contrast to DRGs, SCNT increased the proteolytic 80-kDa fragment of L1-CAM and decreased full-length L1-CAM in the spinal cord. The intrathecal injection of L1-CAM antibody for the extracellular domain of L1-CAM inhibited activation of p38 MAPK and emergence of ring structures of L1-CAM immunoreactivity in injured DRG neurons. Moreover, inhibition of extracellular L1-CAM binding by intrathecal administration of antibody suppressed the mechanical allodynia and thermal hyperalgesia induced by partial SCNT. Collectively, these data suggest that the modification of L1-CAM in nociceptive pathways might be an important pathomechanism of neuropathic pain.
Collapse
Affiliation(s)
- Hiroki Yamanaka
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan
| | | | | | | | | | | |
Collapse
|
41
|
Schultheis M, Diestel S, Schmitz B. The role of cytoplasmic serine residues of the cell adhesion molecule L1 in neurite outgrowth, endocytosis, and cell migration. Cell Mol Neurobiol 2007; 27:11-31. [PMID: 17151951 DOI: 10.1007/s10571-006-9113-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2005] [Accepted: 07/14/2006] [Indexed: 10/23/2022]
Abstract
1. The cell adhesion molecule L1 has been implicated in adhesion and migration of cells, in axon growth, guidance, and fasciculation, in myelination and synaptic plasticity. The cytoplasmic domain of neuronal L1 is highly conserved between species and has been shown to be phosphorylated at serine and tyrosine residues. 2. To investigate the significance of L1 serine phosphorylation, mutants of L1 were generated in which ser-1152, ser-1181, ser-1204, and ser-1248 were exchanged for leucine and rat B35 neuroblastoma cells were stably transfected with the L1-cDNA constructs. 3. Neurite outgrowth on poly-L-lysine (PLL) as substrate was determined either with or without differentiation into a neuronal phenotype with dbcAMP. In addition, antibody-induced endocytosis and cell migration were examined. 4. Our observations indicate that phosphorylation of single serine residues of the cytoplasmic domain of L1 contributes to neurite outgrowth through different mechanisms. Neurite growth is increased when ser-1152 or ser-1181 is replaced by a non-phosphorylatable leucine and decreased when ser-1204 or ser-1248 is mutated to leucine. Furthermore, mutation of ser-1181 to leucine results in strongly enhanced antibody-induced endocytosis of L1 and also in enhanced cell migration.
Collapse
Affiliation(s)
- M Schultheis
- Department of Biochemistry, Institute of Animal Sciences, University of Bonn, Katzenburgweg 9a, 53115, Bonn, Germany
| | | | | |
Collapse
|
42
|
Zhuang S, Kelo L, Nardi JB, Kanost MR. Neuroglian on hemocyte surfaces is involved in homophilic and heterophilic interactions of the innate immune system of Manduca sexta. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2007; 31:1159-67. [PMID: 17448535 DOI: 10.1016/j.dci.2007.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2006] [Revised: 02/16/2007] [Accepted: 03/02/2007] [Indexed: 05/15/2023]
Abstract
Neuroglian, a member of the L1 family of cell adhesion molecules (L1-CAMs), is expressed on surfaces of granular cells and a subset of large plasmatocytes of Manduca sexta that act as foci for hemocyte aggregation during the innate immune response. Neuroglian expressed on surfaces of transfected Sf9 cells induced their homophilic aggregation, with the aggregation being abolished in the presence of recombinant immunoglobulin (Ig) domains of neuroglian. Neuroglian and its Ig domains also can interact with hemocyte-specific integrin (HS integrin) as demonstrated with an enzyme-linked immunoassay and a surface plasmon resonance (SPR) assay. Neuroglian double-stranded (ds) RNA not only depresses expression of neuroglian in hemocytes but also depresses the cell-mediated encapsulation response of these hemocytes to foreign surfaces. After injection of a monoclonal antibody (MAb 3B11) into M. sexta larvae that recognizes the Ig domains of neuroglian, the cell-mediated encapsulation response of hemocytes was likewise inhibited. The Ig domains of neuroglian are involved in both homophilic and heterophilic interactions, and subsets of these six different Ig domains may affect different functions of neuroglian.
Collapse
Affiliation(s)
- Shufei Zhuang
- Department of Biochemistry, Kansas State University, Manhattan, KS 66506, USA
| | | | | | | |
Collapse
|
43
|
Triana-Baltzer GB, Liu Z, Berg DK. Pre- and postsynaptic actions of L1-CAM in nicotinic pathways. Mol Cell Neurosci 2006; 33:214-26. [PMID: 16952465 DOI: 10.1016/j.mcn.2006.07.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2006] [Revised: 07/25/2006] [Accepted: 07/27/2006] [Indexed: 01/05/2023] Open
Abstract
Cell adhesion molecules (CAMs) have long been known to guide axon outgrowth and pathfinding. More recent evidence indicates they contribute to synapse formation as well. The L1 family of IgCAMs has been implicated in long-term potentiation, learning, and some features of synaptic structure. We show here that L1 is localized in nicotinic pathways at both pre- and postsynaptic sites. In the chick ciliary ganglion, postsynaptic L1 is associated with nicotinic receptors and potentiates their downstream signaling. Postsynaptic L1 is also important for aligning presynaptic structures over the postsynaptic cell. Dominant negative experiments suggest this latter effect depends on homophilic interactions with presynaptic L1. At the neuromuscular junction L1 is also found presynaptically where dominant negative experiments again indicate a role in aligning presynaptic structures over postsynaptic receptors, both in culture and in vivo. These findings identify new roles for L1 at nicotinic synapses and underscore the multipotency of L1-CAMs.
Collapse
Affiliation(s)
- Gallen B Triana-Baltzer
- Neurobiology Section, Division of Biology, 0357, University of California, San Diego, 9500 Gilman Drive, La Jolla, 92093-0357, USA.
| | | | | |
Collapse
|
44
|
Pruss T, Kranz EU, Niere M, Volkmer H. A regulated switch of chick neurofascin isoforms modulates ligand recognition and neurite extension. Mol Cell Neurosci 2006; 31:354-65. [PMID: 16314110 DOI: 10.1016/j.mcn.2005.10.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2005] [Revised: 09/30/2005] [Accepted: 10/12/2005] [Indexed: 10/25/2022] Open
Abstract
Neural cell adhesion molecule neurofascin regulates the induction of neurite outgrowth, the establishment of synaptic connectivity and myelination. Neurofascin isoforms are generated by spatially and temporally controlled alternative splicing. Isoform NF166 is predominantly expressed in dorsal root ganglia from embryonal day 5 (E5) to E8, and a further neurofascin isoform NF185 appears at E9. Expression of neurofascin and its binding partner axonin-1 on sensory fibers implies functional interactions for neurite outgrowth. E7 sensory neurons require NF166-axonin-1 interactions for neurite extension, accordingly. The contribution of NF166-axonin-1 interaction for neurite outgrowth decreases in parallel with the appearance of NF185 on sensory neurons at E9. This finding may be explained by (1) alleviated intrinsic capability to use axonin-1 as a cellular receptor and (2) reduced binding of axonin-1 to NF185. Finally, NF166, but not NF185, serves as a cellular receptor for neurite induction via homophilic interactions with a neurofascin substrate.
Collapse
Affiliation(s)
- Thomas Pruss
- NMI Naturwissenschaftliches und Medizinisches Institut an der Universität Tübingen, NMI, Markwiesenstr. 55, 72770 Reutlingen, Germany
| | | | | | | |
Collapse
|
45
|
Yang T, Yin W, Derevyanny VD, Moore LA, Longo FM. Identification of an ectodomain within the LAR protein tyrosine phosphatase receptor that binds homophilically and activates signalling pathways promoting neurite outgrowth. Eur J Neurosci 2006; 22:2159-70. [PMID: 16262654 DOI: 10.1111/j.1460-9568.2005.04403.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Elucidation of mechanisms by which receptor protein tyrosine phosphatases (PTPs) regulate neurite outgrowth will require characterization of ligand-receptor interactions and identification of ligand-induced signalling components mediating neurite outgrowth. The first identified ligand of the leucocyte common antigen-related (LAR) receptor PTP consists of a 99-residue ectodomain isoform, termed LARFN5C, which undergoes homophilic binding to LAR and promotes neurite outgrowth. We employed peptide mapping of LARFN5C to identify an active neurite-promoting domain of LAR. A peptide mimetic consisting of 37 residues (L59) and corresponding to the fifth LAR fibronectin type III (FNIII) domain prevented LARFN5C homophilic binding, demonstrated homophilic binding to itself and promoted neurite outgrowth of mouse E16-17 hippocampal neurons and of dorsal root ganglia explants. Response to L59 was partially lost when using neurons derived from LAR-deficient (-/-) mice or neurons treated with LAR siRNA, consistent with homophilic interaction of L59 with LAR. L59 neurite-promoting activity was decreased in the presence of inhibitors of Src, Trk, PLCgamma, PKC, PI3K and MAPK. L59 activated Src (a known substrate of LAR), FAK and TrkB and also activated downstream signalling intermediates including PKC, ERK, AKT and CREB. BDNF augmented the maximal neurite-promoting activity of L59, a finding consistent with the presence of shared and distinct signalling pathways activated by L59 with BDNF and L59 with TrkB. These studies are the first to identify an ectodomain of LAR (located within the fifth FNIII domain) capable of promoting neurite outgrowth and point to novel approaches for promotion of neurite outgrowth.
Collapse
Affiliation(s)
- Tao Yang
- Department of Neurology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | |
Collapse
|
46
|
Triana-Baltzer GB, Blank M. Cytoplasmic domain of protocadherin-α enhances homophilic interactions and recognizes cytoskeletal elements. ACTA ACUST UNITED AC 2006; 66:393-407. [PMID: 16408303 DOI: 10.1002/neu.20228] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cell adhesion molecules of the protocadherin-alpha (pcdh-alpha), -beta, and -gamma families have been proposed to be synaptic specifiers. Pcdh-alpha and -gamma family members localize in part to synapses, and deletion of all pcdh-gammas in mouse affects synaptogenesis. Little is known, however, about the binding specificities and intracellular signaling of protocadherins. Using heterologous expression of tagged constructs, immunostaining, and biotinylation of surface components followed by Western blots we demonstrate that pcdh-alphas undergo homophilic interactions that are significantly enhanced by the cytoplasmic domain. Pcdh-alphas cloned from chick ciliary ganglion have one of two cytoplasmic constant regions (A- and B-types). Screening a yeast two-hybrid library of ciliary ganglion cDNA with the A-type domain yielded a fragment of neurofilament M (NFM); screening with B-type domain yielded a fragment of the actin-bundling protein fascin. Cotransfection of HEK cells with the constructs indicated that the NFM and A-type fragments codistributed as did the fascin and B-type fragments, and the latter could be coimmunoprecipitated. Antibody-induced clustering of full-length pcdh-alphas on the surface of transfected HEK cells induced coclustering of the interacting NFM fragment. Native full-length NFM in tissue extracts bound specifically to the A-type domain on beads, while native full-length fascin in tissue extracts specifically coimmunoprecipitated with pcdh-alpha. Immunostaining neurons demonstrated codistribution of full-length pcdh-alpha with both NFM and actin filaments. These findings suggest cytoskeletal links for pcdh-alphas and identify candidate targets. They also demonstrate homophilic interactions for pcdh-alphas as described for classical cadherins.
Collapse
Affiliation(s)
- Gallen B Triana-Baltzer
- Neurobiology Section, Division of Biology, University of California, San Diego, La Jolla, California 92093-0357, USA
| | | |
Collapse
|
47
|
Kulahin N, Kasper C, Kristensen O, Kastrup JS, Berezin V, Bock E, Gajhede M. Expression, crystallization and preliminary X-ray analysis of the extracellular Ig modules I-IV and F3 modules I-III of the neural cell-adhesion molecule L1. Acta Crystallogr Sect F Struct Biol Cryst Commun 2005; 61:858-60. [PMID: 16511179 PMCID: PMC1978111 DOI: 10.1107/s1744309105026783] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2005] [Accepted: 08/23/2005] [Indexed: 11/10/2022]
Abstract
Four amino-terminal immunoglobulin (Ig) modules and three fibronectin type III (F3) modules of the mouse neural cell-adhesion molecule L1 have been expressed in Drosophila S2 cells. The Ig modules I-IV of L1 crystallized in a trigonal space group, with unit-cell parameters a = b = 239.6, c = 99.3 A, and the crystals diffracted X-rays to a resolution of about 3.5 A. The F3 modules I-III of L1 crystallized in a tetragonal space group, with unit-cell parameters a = b = 80.1, c = 131 A, and the crystals diffracted X-rays to 2.8 A resolution. This is a step towards the structure determination of the multimodular constructs of the neural cell-adhesion molecule L1 in order to understand the function of L1 on a structural basis.
Collapse
Affiliation(s)
- Nikolaj Kulahin
- Protein Laboratory, Institute of Molecular Pathology, Panum Institute, Blegdamsvej 3C, DK-2200 Copenhagen, Denmark.
| | | | | | | | | | | | | |
Collapse
|
48
|
Cheng L, Itoh K, Lemmon V. L1-mediated branching is regulated by two ezrin-radixin-moesin (ERM)-binding sites, the RSLE region and a novel juxtamembrane ERM-binding region. J Neurosci 2005; 25:395-403. [PMID: 15647482 PMCID: PMC2860578 DOI: 10.1523/jneurosci.4097-04.2005] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
We investigated how the neural cell adhesion molecule L1 mediates neurite outgrowth through L1-L1 homophilic interactions. Wild-type L1 and L1 with mutations in the cytoplasmic domain (CD) were introduced into L1 knock-out neurons, and transfected neurons were grown on an L1 substrate. Neurite length and branching were compared between wild-type L1 and L1CD mutations. Surprisingly, the L1CD is not required for L1-mediated neurite outgrowth but plays a critical role in neurite branching, through both the juxtamembrane region and the RSLE region. We demonstrate that both regions serve as ezrin-moesin-radixin-binding sites. A truncation mutant that deletes 110 of 114 amino acids of the L1CD still supports neurite outgrowth on an L1 substrate, suggesting that a coreceptor binds to L1 in cis and mediates neurite outgrowth and that L1-ankyrin interactions are not essential for neurite initiation or outgrowth. These data are consistent with a model in which L1 can influence L1-mediated neurite outgrowth and branching through both the L1CD and a coreceptor.
Collapse
Affiliation(s)
- Ling Cheng
- Department of Neurosciences, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | |
Collapse
|
49
|
Cheng L, Lemmon S, Lemmon V. RanBPM is an L1-interacting protein that regulates L1-mediated mitogen-activated protein kinase activation. J Neurochem 2005; 94:1102-10. [PMID: 16000162 PMCID: PMC2424128 DOI: 10.1111/j.1471-4159.2005.03254.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
A yeast two-hybrid screen using the last 28 amino acids of the cytoplasmic domain of the neural cell adhesion molecule L1 identified RanBPM as an L1-interacting protein. RanBPM associates with L1 in vivo and the N-terminal region of RanBPM (N-RanBPM), containing the SPRY domain, is sufficient for the interaction with L1 in a glutathione S-transferase pull-down assay. L1 antibody patching dramatically changes the subcellular localization of N-RanBPM in transfected COS cells. Overexpression of N-RanBPM in COS cells reduces L1-triggered extracellular signal-regulated kinase 1/2 activation by 50% and overexpression of N-RanBPM in primary neurons inhibits L1-mediated neurite outgrowth and branching. These data suggest that RanBPM is an adaptor protein that links L1 to the extracellular signal-regulated kinase/MAPK pathway.
Collapse
Affiliation(s)
- Ling Cheng
- Department of Neurosciences, Case Western Reserve University, Cleveland, Ohio, USA
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
| | - Sandra Lemmon
- Department of Molecular and Cellular Pharmacology, University of Miami, Miami, Florida, USA
| | - Vance Lemmon
- Department of Neurosciences, Case Western Reserve University, Cleveland, Ohio, USA
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida, USA
| |
Collapse
|
50
|
Cheng L, Lemmon V. Pathological missense mutations of neural cell adhesion molecule L1 affect neurite outgrowth and branching on an L1 substrate. Mol Cell Neurosci 2005; 27:522-30. [PMID: 15555929 DOI: 10.1016/j.mcn.2004.08.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2004] [Revised: 08/10/2004] [Accepted: 08/11/2004] [Indexed: 11/22/2022] Open
Abstract
A number of pathological missense mutations of L1CAM have been shown to disrupt L1-L1 homophilic binding and/or affect surface expression. To investigate whether these mutations disrupt L1-mediated neurite outgrowth, cerebellar neurons from L1 knockout mice are transfected with WT human L1 or L1 mutant constructs, and grown on an L1 substrate. Various parameters of neurite growth are quantified. Most L1 mutations do not affect neurite length significantly but several mutations cause a significant decrease in branching. Comparison of these data with data on L1 expression levels and homophilic binding strength show that changes in neurite growth cannot be simply explained by reductions in either of these parameters. Our results suggest that a coreceptor is involved in L1-mediated neurite outgrowth. Some pathological mutations have little effect on L1 mediated neurite growth, so it is unlikely that a failure of L1-mediated neurite outgrowth is the principle cause of brain defects in patients with L1 mutations.
Collapse
Affiliation(s)
- Ling Cheng
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| | | |
Collapse
|