1
|
Ahammad I, Lamisa AB, Bhattacharjee A, Jamal TB, Arefin MS, Chowdhury ZM, Hossain MU, Das KC, Keya CA, Salimullah M. AITeQ: a machine learning framework for Alzheimer's prediction using a distinctive five-gene signature. Brief Bioinform 2024; 25:bbae291. [PMID: 38877887 PMCID: PMC11179120 DOI: 10.1093/bib/bbae291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 05/23/2024] [Accepted: 06/04/2024] [Indexed: 06/18/2024] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's disease, pose a significant global health challenge with their complex etiology and elusive biomarkers. In this study, we developed the Alzheimer's Identification Tool (AITeQ) using ribonucleic acid-sequencing (RNA-seq), a machine learning (ML) model based on an optimized ensemble algorithm for the identification of Alzheimer's from RNA-seq data. Analysis of RNA-seq data from several studies identified 87 differentially expressed genes. This was followed by a ML protocol involving feature selection, model training, performance evaluation, and hyperparameter tuning. The feature selection process undertaken in this study, employing a combination of four different methodologies, culminated in the identification of a compact yet impactful set of five genes. Twelve diverse ML models were trained and tested using these five genes (CNKSR1, EPHA2, CLSPN, OLFML3, and TARBP1). Performance metrics, including precision, recall, F1 score, accuracy, Matthew's correlation coefficient, and receiver operating characteristic area under the curve were assessed for the finally selected model. Overall, the ensemble model consisting of logistic regression, naive Bayes classifier, and support vector machine with optimized hyperparameters was identified as the best and was used to develop AITeQ. AITeQ is available at: https://github.com/ishtiaque-ahammad/AITeQ.
Collapse
Affiliation(s)
- Ishtiaque Ahammad
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka 1349, Bangladesh
| | - Anika Bushra Lamisa
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka 1349, Bangladesh
| | - Arittra Bhattacharjee
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka 1349, Bangladesh
| | - Tabassum Binte Jamal
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka 1349, Bangladesh
| | - Md Shamsul Arefin
- Department of Biochemistry and Microbiology, North South University, Bashundhara, Dhaka 1229, Bangladesh
| | - Zeshan Mahmud Chowdhury
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka 1349, Bangladesh
| | - Mohammad Uzzal Hossain
- Bioinformatics Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka 1349, Bangladesh
| | - Keshob Chandra Das
- Molecular Biotechnology Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka 1349, Bangladesh
| | - Chaman Ara Keya
- Department of Biochemistry and Microbiology, North South University, Bashundhara, Dhaka 1229, Bangladesh
| | - Md Salimullah
- Molecular Biotechnology Division, National Institute of Biotechnology, Ganakbari, Ashulia, Savar, Dhaka 1349, Bangladesh
| |
Collapse
|
2
|
Guidance landscapes unveiled by quantitative proteomics to control reinnervation in adult visual system. Nat Commun 2022; 13:6040. [PMID: 36229455 PMCID: PMC9561644 DOI: 10.1038/s41467-022-33799-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 09/30/2022] [Indexed: 12/24/2022] Open
Abstract
In the injured adult central nervous system (CNS), activation of pro-growth molecular pathways in neurons leads to long-distance regeneration. However, most regenerative fibers display guidance defects, which prevent reinnervation and functional recovery. Therefore, the molecular characterization of the proper target regions of regenerative axons is essential to uncover the modalities of adult reinnervation. In this study, we use mass spectrometry (MS)-based quantitative proteomics to address the proteomes of major nuclei of the adult visual system. These analyses reveal that guidance-associated molecules are expressed in adult visual targets. Moreover, we show that bilateral optic nerve injury modulates the expression of specific proteins. In contrast, the expression of guidance molecules remains steady. Finally, we show that regenerative axons are able to respond to guidance cues ex vivo, suggesting that these molecules possibly interfere with brain target reinnervation in adult. Using a long-distance regeneration model, we further demonstrate that the silencing of specific guidance signaling leads to rerouting of regenerative axons in vivo. Altogether, our results suggest ways to modulate axon guidance of regenerative neurons to achieve circuit repair in adult.
Collapse
|
3
|
Skarlatou S, Hérent C, Toscano E, Mendes CS, Bouvier J, Zampieri N. Afadin Signaling at the Spinal Neuroepithelium Regulates Central Canal Formation and Gait Selection. Cell Rep 2021; 31:107741. [PMID: 32521266 DOI: 10.1016/j.celrep.2020.107741] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 04/03/2020] [Accepted: 05/15/2020] [Indexed: 11/28/2022] Open
Abstract
Afadin, a scaffold protein controlling the activity of the nectin family of cell adhesion molecules, regulates important morphogenetic processes during development. In the central nervous system, afadin has critical roles in neuronal migration, axonal elongation, and synapse formation. Here we examine the role of afadin in development of spinal motor circuits. Afadin elimination in motor neuron progenitors results in striking locomotor behavior: left-right limb alternation is substituted by synchronous activation, characteristic of bound gait. We find that afadin function at the neuroepithelium is required for structural organization of the spinal midline and central canal morphogenesis. Perturbation of afadin results in formation of two central canals, aberrant contralateral wiring of different classes of spinal premotor interneurons, and loss of left-right limb alternation, highlighting important developmental principles controlling the assembly of spinal motor circuits.
Collapse
Affiliation(s)
- Sophie Skarlatou
- Cluster of Excellence NeuroCure, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Coralie Hérent
- Paris-Saclay Institute of Neuroscience, UMR 9197 CNRS & Université Paris-Saclay, Avenue de La Terrasse, 91190 Gif sur Yvette, France
| | - Elisa Toscano
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - César S Mendes
- CEDOC, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - Julien Bouvier
- Paris-Saclay Institute of Neuroscience, UMR 9197 CNRS & Université Paris-Saclay, Avenue de La Terrasse, 91190 Gif sur Yvette, France
| | - Niccolò Zampieri
- Cluster of Excellence NeuroCure, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany.
| |
Collapse
|
4
|
Clark IC, Gutiérrez-Vázquez C, Wheeler MA, Li Z, Rothhammer V, Linnerbauer M, Sanmarco LM, Guo L, Blain M, Zandee SEJ, Chao CC, Batterman KV, Schwabenland M, Lotfy P, Tejeda-Velarde A, Hewson P, Manganeli Polonio C, Shultis MW, Salem Y, Tjon EC, Fonseca-Castro PH, Borucki DM, Alves de Lima K, Plasencia A, Abate AR, Rosene DL, Hodgetts KJ, Prinz M, Antel JP, Prat A, Quintana FJ. Barcoded viral tracing of single-cell interactions in central nervous system inflammation. Science 2021; 372:372/6540/eabf1230. [PMID: 33888612 DOI: 10.1126/science.abf1230] [Citation(s) in RCA: 164] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/27/2021] [Accepted: 03/12/2021] [Indexed: 12/11/2022]
Abstract
Cell-cell interactions control the physiology and pathology of the central nervous system (CNS). To study astrocyte cell interactions in vivo, we developed rabies barcode interaction detection followed by sequencing (RABID-seq), which combines barcoded viral tracing and single-cell RNA sequencing (scRNA-seq). Using RABID-seq, we identified axon guidance molecules as candidate mediators of microglia-astrocyte interactions that promote CNS pathology in experimental autoimmune encephalomyelitis (EAE) and, potentially, multiple sclerosis (MS). In vivo cell-specific genetic perturbation EAE studies, in vitro systems, and the analysis of MS scRNA-seq datasets and CNS tissue established that Sema4D and Ephrin-B3 expressed in microglia control astrocyte responses via PlexinB2 and EphB3, respectively. Furthermore, a CNS-penetrant EphB3 inhibitor suppressed astrocyte and microglia proinflammatory responses and ameliorated EAE. In summary, RABID-seq identified microglia-astrocyte interactions and candidate therapeutic targets.
Collapse
Affiliation(s)
- Iain C Clark
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Department of Bioengineering, University of California, Berkeley, California Institute for Quantitative Biosciences, Berkeley, CA 94720, USA
| | - Cristina Gutiérrez-Vázquez
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Michael A Wheeler
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Zhaorong Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Veit Rothhammer
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Mathias Linnerbauer
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Liliana M Sanmarco
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lydia Guo
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Manon Blain
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Stephanie E J Zandee
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada
| | - Chun-Cheih Chao
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Katelyn V Batterman
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Marius Schwabenland
- Institute of Neuropathology, University of Freiburg, D-79106 Freiburg, Germany
| | - Peter Lotfy
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Amalia Tejeda-Velarde
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Patrick Hewson
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Carolina Manganeli Polonio
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Michael W Shultis
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yasmin Salem
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Emily C Tjon
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Pedro H Fonseca-Castro
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Davis M Borucki
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kalil Alves de Lima
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Agustin Plasencia
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Adam R Abate
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California Institute for Quantitative Biosciences, San Francisco, CA 94158, USA.,Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Douglas L Rosene
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Kevin J Hodgetts
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Marco Prinz
- Institute of Neuropathology, University of Freiburg, D-79106 Freiburg, Germany.,Signaling Research Centres BIOSS and CIBSS, University of Freiburg, D-79106 Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, D-79106 Freiburg, Germany
| | - Jack P Antel
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Alexandre Prat
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA. .,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
5
|
Alvarez S, Varadarajan SG, Butler SJ. Dorsal commissural axon guidance in the developing spinal cord. Curr Top Dev Biol 2020; 142:197-231. [PMID: 33706918 DOI: 10.1016/bs.ctdb.2020.10.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Commissural axons have been a key model system for identifying axon guidance signals in vertebrates. This review summarizes the current thinking about the molecular and cellular mechanisms that establish a specific commissural neural circuit: the dI1 neurons in the developing spinal cord. We assess the contribution of long- and short-range signaling while sequentially following the developmental timeline from the birth of dI1 neurons, to the extension of commissural axons first circumferentially and then contralaterally into the ventral funiculus.
Collapse
Affiliation(s)
- Sandy Alvarez
- Department of Neurobiology, University of California, Los Angeles, CA, United States; Molecular Biology Interdepartmental Doctoral Program, University of California, Los Angeles, CA, United States
| | | | - Samantha J Butler
- Department of Neurobiology, University of California, Los Angeles, CA, United States; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, United States.
| |
Collapse
|
6
|
Abstract
The spinal cord receives, relays and processes sensory information from the periphery and integrates this information with descending inputs from supraspinal centres to elicit precise and appropriate behavioural responses and orchestrate body movements. Understanding how the spinal cord circuits that achieve this integration are wired during development is the focus of much research interest. Several families of proteins have well-established roles in guiding developing spinal cord axons, and recent findings have identified new axon guidance molecules. Nevertheless, an integrated view of spinal cord network development is lacking, and many current models have neglected the cellular and functional diversity of spinal cord circuits. Recent advances challenge the existing spinal cord axon guidance dogmas and have provided a more complex, but more faithful, picture of the ontogenesis of vertebrate spinal cord circuits.
Collapse
|
7
|
KIF20A/MKLP2 regulates the division modes of neural progenitor cells during cortical development. Nat Commun 2018; 9:2707. [PMID: 30006548 PMCID: PMC6045631 DOI: 10.1038/s41467-018-05152-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 06/14/2018] [Indexed: 12/17/2022] Open
Abstract
Balanced symmetric and asymmetric divisions of neural progenitor cells (NPCs) are crucial for brain development, but the underlying mechanisms are not fully understood. Here we report that mitotic kinesin KIF20A/MKLP2 interacts with RGS3 and plays a crucial role in controlling the division modes of NPCs during cortical neurogenesis. Knockdown of KIF20A in NPCs causes dislocation of RGS3 from the intercellular bridge (ICB), impairs the function of Ephrin-B–RGS cell fate signaling complex, and leads to a transition from proliferative to differentiative divisions. Germline and inducible knockout of KIF20A causes a loss of progenitor cells and neurons and results in thinner cortex and ventriculomegaly. Interestingly, loss of function of KIF20A induces early cell cycle exit and precocious neuronal differentiation without causing substantial cytokinesis defect or apoptosis. Our results identify a RGS–KIF20A axis in the regulation of cell division and suggest a potential link of the ICB to regulation of cell fate determination. The division of neural progenitors is closely regulated but how is unclear. Here, the authors show that mitotic kinesin KIF20A/MKLP2 interacts with a regulator of G protein signaling RGS3 in neural progenitor cells, dislodging it from the intercellular bridge of dividing cortical cells.
Collapse
|
8
|
Noraz N, Jaaoini I, Charoy C, Watrin C, Chounlamountri N, Benon A, Malleval C, Boudin H, Honnorat J, Castellani V, Pellier-Monnin V. Syk kinases are required for spinal commissural axon repulsion at the midline via the ephrin/Eph pathway. Development 2016; 143:2183-93. [PMID: 27122172 DOI: 10.1242/dev.128629] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 04/15/2016] [Indexed: 12/26/2022]
Abstract
In the hematopoietic system, Syk family tyrosine kinases are essential components of immunoreceptor ITAM-based signaling. While there is increasing data indicating the involvement of immunoreceptors in neural functions, the contribution of Syk kinases remains obscure. Previously, we identified phosphorylated forms of Syk kinases in specialized populations of migrating neurons or projecting axons. Moreover, we identified ephrin/Eph as guidance molecules utilizing the ITAM-bearing CD3zeta (Cd247) and associated Syk kinases for the growth cone collapse response induced in vitro Here, we show that in the developing spinal cord, Syk is phosphorylated in navigating commissural axons. By analyzing axon trajectories in open-book preparations of Syk(-/-); Zap70(-/-) mouse embryos, we show that Syk kinases are dispensable for attraction towards the midline but confer growth cone responsiveness to repulsive signals that expel commissural axons from the midline. Known to serve a repulsive function at the midline, ephrin B3/EphB2 are obvious candidates for driving the Syk-dependent repulsive response. Indeed, Syk kinases were found to be required for ephrin B3-induced growth cone collapse in cultured commissural neurons. In fragments of commissural neuron-enriched tissues, Syk is in a constitutively phosphorylated state and ephrin B3 decreased its level of phosphorylation. Direct pharmacological inhibition of Syk kinase activity was sufficient to induce growth cone collapse. In conclusion, Syk kinases act as a molecular switch of growth cone adhesive and repulsive responses.
Collapse
Affiliation(s)
- Nelly Noraz
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Iness Jaaoini
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Camille Charoy
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Chantal Watrin
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Naura Chounlamountri
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Aurélien Benon
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Céline Malleval
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Hélène Boudin
- INSERM U1064, Institut de Transplantation Urologie-Néphrologie, Nantes F-44035, France
| | - Jérôme Honnorat
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France Hospices Civils de Lyon, Lyon F-69000, France
| | - Valérie Castellani
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| | - Véronique Pellier-Monnin
- INSERM U1217, Institut NeuroMyoGène, Lyon F-69000, France CNRS UMR5310, Institut NeuroMyoGène, Lyon F-69000, France University Claude Bernard Lyon 1, Lyon F-69000, France
| |
Collapse
|
9
|
Comer JD, Pan FC, Willet SG, Haldipur P, Millen KJ, Wright CVE, Kaltschmidt JA. Sensory and spinal inhibitory dorsal midline crossing is independent of Robo3. Front Neural Circuits 2015; 9:36. [PMID: 26257608 PMCID: PMC4511845 DOI: 10.3389/fncir.2015.00036] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 07/02/2015] [Indexed: 11/25/2022] Open
Abstract
Commissural neurons project across the midline at all levels of the central nervous system (CNS), providing bilateral communication critical for the coordination of motor activity and sensory perception. Midline crossing at the spinal ventral midline has been extensively studied and has revealed that multiple developmental lineages contribute to this commissural neuron population. Ventral midline crossing occurs in a manner dependent on Robo3 regulation of Robo/Slit signaling and the ventral commissure is absent in the spinal cord and hindbrain of Robo3 mutants. Midline crossing in the spinal cord is not limited to the ventral midline, however. While prior anatomical studies provide evidence that commissural axons also cross the midline dorsally, little is known of the genetic and molecular properties of dorsally-crossing neurons or of the mechanisms that regulate dorsal midline crossing. In this study, we describe a commissural neuron population that crosses the spinal dorsal midline during the last quarter of embryogenesis in discrete fiber bundles present throughout the rostrocaudal extent of the spinal cord. Using immunohistochemistry, neurotracing, and mouse genetics, we show that this commissural neuron population includes spinal inhibitory neurons and sensory nociceptors. While the floor plate and roof plate are dispensable for dorsal midline crossing, we show that this population depends on Robo/Slit signaling yet crosses the dorsal midline in a Robo3-independent manner. The dorsally-crossing commissural neuron population we describe suggests a substrate circuitry for pain processing in the dorsal spinal cord.
Collapse
Affiliation(s)
- John D Comer
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences New York, NY, USA ; Developmental Biology Program, Sloan-Kettering Institute New York, NY, USA ; Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program New York, NY, USA
| | - Fong Cheng Pan
- Vanderbilt University Program in Developmental Biology, Department of Cell and Developmental Biology, Vanderbilt Center for Stem Cell Biology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Spencer G Willet
- Vanderbilt University Program in Developmental Biology, Department of Cell and Developmental Biology, Vanderbilt Center for Stem Cell Biology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Parthiv Haldipur
- Seattle Children's Research Institute, Center for Integrative Brain Research Seattle, WA, USA
| | - Kathleen J Millen
- Seattle Children's Research Institute, Center for Integrative Brain Research Seattle, WA, USA ; Department of Pediatrics, Genetics Division, University of Washington Seattle, WA, USA
| | - Christopher V E Wright
- Vanderbilt University Program in Developmental Biology, Department of Cell and Developmental Biology, Vanderbilt Center for Stem Cell Biology, Vanderbilt University Medical Center Nashville, TN, USA
| | - Julia A Kaltschmidt
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences New York, NY, USA ; Developmental Biology Program, Sloan-Kettering Institute New York, NY, USA
| |
Collapse
|
10
|
Martinez E, Tran TS. Vertebrate spinal commissural neurons: a model system for studying axon guidance beyond the midline. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2015; 4:283-97. [PMID: 25619385 DOI: 10.1002/wdev.173] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 11/27/2014] [Accepted: 12/04/2014] [Indexed: 12/21/2022]
Abstract
For bilaterally symmetric organisms, the transfer of information between the left and right side of the nervous system is mediated by commissures formed by neurons that project their axons across the body midline to the contralateral side of the central nervous system (CNS). After crossing the midline, many of these axons must travel long distances to reach their targets, including those that extend from spinal commissural neurons. Owing to the highly stereotyped trajectories of spinal commissural neurons that can be divided into several segments as these axons project to their targets, it is an ideal system for investigators to ask fundamental questions related to mechanisms of short- and long-range axon guidance, fasciculation, and choice point decisions at the midline intermediate target. In addition, studies of patterning genes of the nervous system have revealed complex transcription factor codes that function in a combinatorial fashion to specify individual classes of spinal neurons including commissural neurons. Despite these advances and the functional importance of spinal commissural neurons in mediating the transfer of external sensory information from the peripheral nervous system (PNS) to the CNS, only a handful of studies have begun to elucidate the mechanistic logic underlying their long-range pathfinding and the characterization of their synaptic targets. Using in vitro assays, in vivo labeling methodologies, in combination with both loss- and gain-of-function experiments, several studies have revealed that the molecular mechanisms of long-range spinal commissural axon pathfinding involve an interplay between classical axon guidance cues, morphogens and cell adhesion molecules. For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Edward Martinez
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
| | | |
Collapse
|
11
|
Paixão S, Balijepalli A, Serradj N, Niu J, Luo W, Martin JH, Klein R. EphrinB3/EphA4-mediated guidance of ascending and descending spinal tracts. Neuron 2014; 80:1407-20. [PMID: 24360544 DOI: 10.1016/j.neuron.2013.10.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2013] [Indexed: 11/27/2022]
Abstract
The spinal cord contains many descending and ascending longitudinal tracts whose development appears to be controlled by distinct guidance systems. We identified a population of dorsal spinal neurons marked by coexpression of the transcription factor Zic2 and the guidance receptor EphA4. Zic2+;EphA4+ neurons are surrounded by mechanosensory terminals, suggesting innervation by mechanoreceptor afferents. Their axons form an ipsilateral ascending pathway that develops during embryogenesis and projects within the ventral aspect of the dorsal funiculus, the same location as the descending corticospinal tract (CST), which develops postnatally. Interestingly, the same guidance mechanism, namely, ephrinB3-induced EphA4 forward signaling, is required for the guidance of both ascending and descending axon tracts. Our analysis of conditional EphA4 mutant mice also revealed that the development of the dorsal funiculus occurs independently of EphA4 expression in descending CST axons and is linked to the distribution of Zic2+;EphA4+ spinal neurons and the formation of the ascending pathway.
Collapse
Affiliation(s)
- Sónia Paixão
- Department 'Molecules - Signals - Development,' Max Planck Institute of Neurobiology, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Aarathi Balijepalli
- Department 'Molecules - Signals - Development,' Max Planck Institute of Neurobiology, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Najet Serradj
- Department of Physiology, Pharmacology, and Neuroscience, City College of the City University of New York, New York, NY 10031, USA
| | - Jingwen Niu
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wenqin Luo
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John H Martin
- Department of Physiology, Pharmacology, and Neuroscience, City College of the City University of New York, New York, NY 10031, USA
| | - Rüdiger Klein
- Department 'Molecules - Signals - Development,' Max Planck Institute of Neurobiology, Am Klopferspitz 18, 82152 Martinsried, Germany; Munich Cluster for Systems Neurology (Synergy), 80336 Munich, Germany.
| |
Collapse
|
12
|
The small GTPase RhoA is required for proper locomotor circuit assembly. PLoS One 2013; 8:e67015. [PMID: 23825607 PMCID: PMC3692541 DOI: 10.1371/journal.pone.0067015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 05/13/2013] [Indexed: 11/19/2022] Open
Abstract
The assembly of neuronal circuits during development requires the precise navigation of axons, which is controlled by attractive and repulsive guidance cues. In the developing spinal cord, ephrinB3 functions as a short-range repulsive cue that prevents EphA4 receptor-expressing corticospinal tract and spinal interneuron axons from crossing the midline, ensuring proper formation of locomotor circuits. Here we report that the small GTPase RhoA, a key regulator of cytoskeletal dynamics, is also required for ephrinB3/EphA4-dependent locomotor circuit formation. Deletion of RhoA from neural progenitor cells results in mice that exhibit a rabbit-like hopping gait, which phenocopies mice lacking ephrinB3 or EphA4. Consistent with this locomotor defect, we found that corticospinal tract axons and spinal interneuron projections from RhoA-deficient mice aberrantly cross the spinal cord midline. Furthermore, we determined that loss of RhoA blocks ephrinB3-induced growth cone collapse of cortical axons and disrupts ephrinB3 expression at the spinal cord midline. Collectively, our results demonstrate that RhoA is essential for the ephrinB3/EphA4-dependent assembly of cortical and spinal motor circuits that control normal locomotor behavior.
Collapse
|
13
|
Bravo-Ambrosio A, Mastick G, Kaprielian Z. Motor axon exit from the mammalian spinal cord is controlled by the homeodomain protein Nkx2.9 via Robo-Slit signaling. Development 2012; 139:1435-46. [PMID: 22399681 PMCID: PMC3308178 DOI: 10.1242/dev.072256] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2012] [Indexed: 01/11/2023]
Abstract
Mammalian motor circuits control voluntary movements by transmitting signals from the central nervous system (CNS) to muscle targets. To form these circuits, motor neurons (MNs) must extend their axons out of the CNS. Although exit from the CNS is an indispensable phase of motor axon pathfinding, the underlying molecular mechanisms remain obscure. Here, we present the first identification of a genetic pathway that regulates motor axon exit from the vertebrate spinal cord, utilizing spinal accessory motor neurons (SACMNs) as a model system. SACMNs are a homogeneous population of spinal MNs with axons that leave the CNS through a discrete lateral exit point (LEP) and can be visualized by the expression of the cell surface protein BEN. We show that the homeodomain transcription factor Nkx2.9 is selectively required for SACMN axon exit and identify the Robo2 guidance receptor as a likely downstream effector of Nkx2.9; loss of Nkx2.9 leads to a reduction in Robo2 mRNA and protein within SACMNs and SACMN axons fail to exit the spinal cord in Robo2-deficient mice. Consistent with short-range interactions between Robo2 and Slit ligands regulating SACMN axon exit, Robo2-expressing SACMN axons normally navigate through LEP-associated Slits as they emerge from the spinal cord, and fail to exit in Slit-deficient mice. Our studies support the view that Nkx2.9 controls SACMN axon exit from the mammalian spinal cord by regulating Robo-Slit signaling.
Collapse
Affiliation(s)
- Arlene Bravo-Ambrosio
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Grant Mastick
- Department of Biology, University of Nevada, Reno, NV 89557, USA
| | - Zaven Kaprielian
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
14
|
Kao TJ, Kania A. Ephrin-mediated cis-attenuation of Eph receptor signaling is essential for spinal motor axon guidance. Neuron 2011; 71:76-91. [PMID: 21745639 DOI: 10.1016/j.neuron.2011.05.031] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2011] [Indexed: 02/01/2023]
Abstract
Axon guidance receptors guide neuronal growth cones by binding in trans to axon guidance ligands in the developing nervous system. Some ligands are coexpressed in cis with their receptors, raising the question of the relative contribution of cis and trans interactions to axon guidance. Spinal motor axons use Eph receptors to select a limb trajectory in response to trans ephrins, while expressing ephrins in cis. We show that changes in motor neuron ephrin expression result in trajectory selection defects mirrored by changes in growth cone sensitivity to ephrins in vitro, arguing for ephrin cis-attenuation of Eph function. Furthermore, the relative contribution of trans-signaling and cis-attenuation is influenced by the subcellular distribution of ephrins to membrane patches containing Eph receptors. Thus, growth cone ephrins are essential for axon guidance in vivo and the balance between cis and trans modes of axon guidance ligand-receptor interaction contributes to the diversity of axon guidance signaling responses.
Collapse
Affiliation(s)
- Tzu-Jen Kao
- Institut de recherches cliniques de Montréal, Montréal, QC H2W1R7, Canada
| | | |
Collapse
|
15
|
Katsuki T, Joshi R, Ailani D, Hiromi Y. Compartmentalization within neurites: its mechanisms and implications. Dev Neurobiol 2011; 71:458-73. [PMID: 21557500 DOI: 10.1002/dneu.20859] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Neurons are morphologically characterized by long processes extending from a cell body. These processes, the dendrites and axon, are major sub-cellular compartments defined by morphological, molecular, and functional differences. However, evidence from vertebrates and invertebrates suggests that, based on molecular distribution, individual axons and dendrites are further divided into distinct compartments; many membrane molecules involved in axon guidance and synapse formation are localized to specific segments of axons or dendrites that share a boundary of localization. In this review, we describe recent progress in understanding the mechanisms of intra-neurite patterning, and discuss its potential roles in the development and function of the nervous system. Each protein employs different ways to achieve compartment-specific localization; some membrane molecules localize via cell-autonomous ability of neurons, while others require extrinsic signals for localization. The underlying regulatory mechanisms include transcriptional regulation, local translation, diffusion barrier, endocytosis, and selective membrane targeting. We propose that intra-neurite compartmentalization could provide platforms for structural and functional diversification of individual neurons.
Collapse
Affiliation(s)
- Takeo Katsuki
- Department of Developmental Genetics, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka, Japan
| | | | | | | |
Collapse
|
16
|
Su Y, Naser IB, Islam SM, Zhang S, Ahmed G, Chen S, Shinmyo Y, Kawakami M, Yamamura KI, Tanaka H. Draxin, an axon guidance protein, affects chick trunk neural crest migration. Dev Growth Differ 2009; 51:787-96. [PMID: 19824897 DOI: 10.1111/j.1440-169x.2009.01137.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The neural crest is a multipotent population of migratory cells that arises in the central nervous system and subsequently migrates along defined stereotypic pathways. In the present work, we analyzed the role of a repulsive axon guidance protein, draxin, in the migration of neural crest cells. Draxin is expressed in the roof plate of the chick trunk spinal cord and around the early migration pathway of neural crest cells. Draxin modulates chick neural crest cell migration in vitro by reducing the polarization of these cells. When exposed to draxin, the velocity of migrating neural crest cells was reduced, and the cells changed direction so frequently that the net migration distance was also reduced. Overexpression of draxin also caused some early migrating neural crest cells to change direction to the dorsolateral pathway in the chick trunk region, presumably due to draxin's inhibitory activity. These results demonstrate that draxin, an axon guidance protein, can also affect trunk neural crest migration in the chick embryo.
Collapse
Affiliation(s)
- Yuhong Su
- Division of Developmental Neurobiology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Reeber SL, Kaprielian Z. Leaving the midline: how Robo receptors regulate the guidance of post-crossing spinal commissural axons. Cell Adh Migr 2009; 3:300-4. [PMID: 19556886 DOI: 10.4161/cam.3.3.9156] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
In the developing nervous system, pathfinding axons navigate through a series of intermediate targets in order to form synaptic connections. Vertebrate spinal commissural axons extend toward and across the floor plate (FP), a key intermediate target located at the ventral midline (VM). Subsequently, post-crossing commissural axons grow either alongside or significant distances away from the floor plate (FP), but never re-cross the VM. Consistent with this behavior, post-crossing commissural axons lose responsiveness to the FP-associated chemoattractants, Netrin-1 and SHH, and gain responsiveness to Slits, which are potent midline repellents, in vitro. In addition, the results of several in vivo studies suggest that the upregulation of Slit-binding repulsive Robo receptors, Robo1/2, alters the responsiveness of decussated commissural axons to midline guidance cues. Nevertheless, in vertebrates, it is unclear whether Robo1/2 are the sole or major repellent receptors responsible for driving these commissural axons away from the VM and preventing their re-entry into the FP. We recently re-visited these issues in the chick spinal cord by assessing the consequences of manipulating Robo expression on commissural axons in ovo. Our findings suggest that, at least in chick embryos, the upregulation of repulsive Robos on post-crossing axons alters the responsiveness of these axons to midline repellents and facilitates their expulsion from, but is not likely to have a significant role in preventing their re-entry into the VM.
Collapse
Affiliation(s)
- Stacey L Reeber
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | | |
Collapse
|
18
|
Canty A, Murphy M. Molecular mechanisms of axon guidance in the developing corticospinal tract. Prog Neurobiol 2008; 85:214-35. [DOI: 10.1016/j.pneurobio.2008.02.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2007] [Revised: 12/11/2007] [Accepted: 02/08/2008] [Indexed: 02/04/2023]
|
19
|
Sandoval-Minero T, Varela-Echavarría A. Cross-midline interactions between mouse commissural hindbrain axons contribute to their efficient decussation. Dev Neurobiol 2008; 68:349-64. [PMID: 18085564 DOI: 10.1002/dneu.20586] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Information from both sides of the brain is integrated by axons that project across the midline of the central nervous system via numerous commissures present at all axial levels. Despite the accumulated experimental evidence, questions remain regarding the formation of commissures in the presence of strong repulsive signals in the ventral midline. Studies from invertebrates suggest that interaction at the midline between homologous axons of specific decussating neurons contributes to efficient midline crossing, but such evidence is lacking in vertebrate systems. We performed experiments to determine whether commissural axons of the caudal region of the hindbrain interact with their contralateral counterparts at the ventral midline and to evaluate the relevance of this reciprocal interaction. Double anterograde axon labeling with lipophilic tracers revealed close apposition between growth cones of contralateral pioneer decussating axons at the midline. Later, we detected fasciculation between contralateral axons that is maintained even after they have crossed the midline. Blocking axon projections unilaterally with a solid mechanical barrier decreased dramatically the midline crossing of the equivalent population from the contralateral side. Decussation was also blocked by a unilateral barrier permeable to diffusible molecules but not by an axon-permeable barrier. These results suggest that in the caudal region of the hindbrain, midline crossing is facilitated by interactions between decussating contralateral axon partners.
Collapse
Affiliation(s)
- Teresa Sandoval-Minero
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Qro., C.P. 76230, México
| | | |
Collapse
|
20
|
Wegmeyer H, Egea J, Rabe N, Gezelius H, Filosa A, Enjin A, Varoqueaux F, Deininger K, Schnütgen F, Brose N, Klein R, Kullander K, Betz A. EphA4-Dependent Axon Guidance Is Mediated by the RacGAP α2-Chimaerin. Neuron 2007; 55:756-67. [PMID: 17785182 DOI: 10.1016/j.neuron.2007.07.038] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2007] [Revised: 07/20/2007] [Accepted: 07/30/2007] [Indexed: 01/19/2023]
Abstract
Neuronal network formation in the developing nervous system is dependent on the accurate navigation of nerve cell axons and dendrites, which is controlled by attractive and repulsive guidance cues. Ephrins and their cognate Eph receptors mediate many repulsive axonal guidance decisions by intercellular interactions resulting in growth cone collapse and axon retraction of the Eph-presenting neuron. We show that the Rac-specific GTPase-activating protein alpha2-chimaerin binds activated EphA4 and mediates EphA4-triggered axonal growth cone collapse. alpha-Chimaerin mutant mice display a phenotype similar to that of EphA4 mutant mice, including aberrant midline axon guidance and defective spinal cord central pattern generator activity. Our results reveal an alpha-chimaerin-dependent signaling pathway downstream of EphA4, which is essential for axon guidance decisions and neuronal circuit formation in vivo.
Collapse
Affiliation(s)
- Heike Wegmeyer
- Department of Molecular Neurobiology and DFG Center for Molecular Physiology of the Brain, Max Planck Institute of Experimental Medicine, D-37075 Göttingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Jevince AR, Kadison SR, Pittman AJ, Chien CB, Kaprielian Z. Distribution of EphB receptors and ephrin-B1 in the developing vertebrate spinal cord. J Comp Neurol 2006; 497:734-50. [PMID: 16786562 PMCID: PMC2637817 DOI: 10.1002/cne.21001] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Contact-dependent interactions between EphB receptors and ephrin-B ligands mediate a variety of cell-cell communication events in the developing and mature central nervous system (CNS). These predominantly repulsive interactions occur at the interface between what are considered to be mutually exclusive EphB and ephrin-B expression domains. We previously used receptor and ligand affinity probes to show that ephrin-B ligands are expressed in the floor plate and within a dorsal region of the embryonic mouse spinal cord, while EphB receptors are present on decussated segments of commissural axons that navigate between these ephrin-B domains. Here we present the generation and characterization of two new monoclonal antibodies, mAb EfB1-3, which recognizes EphB1, EphB2, and EphB3, and mAb efrnB1, which is specific for ephrin-B1. We use these reagents and polyclonal antibodies specific for EphB1, EphB2, EphB3, or ephrin-B1 to describe the spatiotemporal expression patterns of EphB receptors and ephrin-B1 in the vertebrate spinal cord. Consistent with affinity probe binding, we show that EphB1, EphB2, and EphB3 are each preferentially expressed on decussated segments of commissural axons in vivo and in vitro, and that ephrin-B1 is expressed in a dorsal domain of the spinal cord that includes the roof plate. In contrast to affinity probe binding profiles, we show here that EphB1, EphB2, and EphB3 are present on the ventral commissure, and that EphB1 and EphB3 are expressed on axons that compose the dorsal funiculus. In addition, we unexpectedly find that mesenchymal cells, which surround the spinal cord and dorsal root ganglion, express ephrin-B1.
Collapse
Affiliation(s)
- Angela R Jevince
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | | | |
Collapse
|
22
|
Goldshmit Y, McLenachan S, Turnley A. Roles of Eph receptors and ephrins in the normal and damaged adult CNS. ACTA ACUST UNITED AC 2006; 52:327-45. [PMID: 16774788 DOI: 10.1016/j.brainresrev.2006.04.006] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2005] [Revised: 04/21/2006] [Accepted: 04/23/2006] [Indexed: 12/19/2022]
Abstract
Injury to the central nervous system (CNS) usually results in very limited regeneration of lesioned axons, which are inhibited by the environment of the injury site. Factors that have been implicated in inhibition of axonal regeneration include myelin proteins, astrocytic gliosis and cell surface molecules that are involved in axon guidance during development. This review examines the contribution of one such family of developmental guidance molecules, the Eph receptor tyrosine kinases and their ligands, the ephrins in normal adult CNS and following injury or disease. Eph/ephrin signaling regulates axon guidance through contact repulsion during development of the CNS, inducing collapse of neuronal growth cones. Eph receptors and ephrins continue to be expressed in the adult CNS, although usually at lower levels, but are upregulated following neural injury on different cell types, including reactive astrocytes, neurons and oligodendrocytes. This upregulated expression may directly inhibit regrowth of regenerating axons; however, in addition, Eph expression also regulates astrocytic gliosis and formation of the glial scar. Therefore, Eph/ephrin signaling may inhibit regeneration by more than one mechanism and modulation of Eph receptor expression or signaling could prove pivotal in determining the outcome of injury in the adult CNS.
Collapse
Affiliation(s)
- Yona Goldshmit
- Centre for Neuroscience, The University of Melbourne, Melbourne, Vic 3010, Australia
| | | | | |
Collapse
|
23
|
Kadison SR, Murakami F, Matise MP, Kaprielian Z. The role of floor plate contact in the elaboration of contralateral commissural projections within the embryonic mouse spinal cord. Dev Biol 2006; 296:499-513. [PMID: 16854408 DOI: 10.1016/j.ydbio.2006.06.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2006] [Revised: 06/13/2006] [Accepted: 06/13/2006] [Indexed: 10/24/2022]
Abstract
In vertebrate embryos, commissural axons extend toward and across the floor plate (FP), an intermediate target at the ventral midline (VM) of the spinal cord. After decussating, many commissural axons turn into the longitudinal plane and elaborate diverse projections. FP contact is thought to alter the responsiveness of these axons so that they can exit the FP and adopt new trajectories. However, a requirement for the FP in shaping contralateral commissural projections has not been established in higher vertebrates. Here we further analyze to what extent FP contact is necessary for the elaboration of decussated commissural projections both in cultured, FP-excised spinal cord preparations and in gli2-deficient mice, which lack a FP. In FP-lacking spinal cords, we observe a large number of appropriately projecting contralateral commissural projections in vivo and in vitro. Surprisingly, even though gli2 mutants lack a FP, slit1-3 mRNA and their receptors (Robo1/2) are expressed in a wild-type-like manner. In addition, blocking Robo-Slit interactions in FP-lacking spinal cord explants prevents commissural axons from leaving the VM and turning longitudinally. Thus, compared to FP contact, Slit-Robo interactions are more critical for driving commissural axons out of the VM and facilitating the elaboration of a subset of contralateral commissural projections.
Collapse
Affiliation(s)
- Stephanie R Kadison
- Department of Pathology and Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Kennedy Center Rm. 624, 1410 Pelham Parkway South, Bronx, NY 10461, USA.
| | | | | | | |
Collapse
|
24
|
Cramer KS, Cerretti DP, Siddiqui SA. EphB2 regulates axonal growth at the midline in the developing auditory brainstem. Dev Biol 2006; 295:76-89. [PMID: 16626680 DOI: 10.1016/j.ydbio.2006.03.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2005] [Revised: 03/07/2006] [Accepted: 03/10/2006] [Indexed: 10/24/2022]
Abstract
Eph receptors play important roles in axon guidance at the midline. In the auditory system, growth of axons across the midline is an important determinant of auditory function. The avian cochlear nucleus, n. magnocellularis (NM), makes bilateral projections to its target, n. laminaris (NL). We examined the time course of NM axon growth toward the midline, the expression of Eph proteins at the midline during this growth, and the effects of Eph receptor misexpression on axonal growth across the midline. We found that NM axons reach the midline at E4. At this age, EphB receptors are expressed at the ventral floor plate. Expression extends dorsally to the ventricular zone beginning at E5. NM axons thus grow across the midline at a time when EphB receptor expression levels are low. Overexpression of EphB2 at E2 resulted in misrouted axons that deflected away from transfected midline cells. This effect was observed when midline cells were transfected but not when NM cells alone were transfected, suggesting that EphB2 acts non-cell autonomously and through reverse signaling. These data suggest an inhibitory role for midline Eph receptors, in which low levels permit axon growth and subsequently high levels prohibit growth after axons have crossed the midline.
Collapse
Affiliation(s)
- Karina S Cramer
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA.
| | | | | |
Collapse
|
25
|
Ogawa Y, Takebayashi H, Takahashi M, Osumi N, Iwasaki Y, Ikenaka K. Gliogenic radial glial cells show heterogeneity in the developing mouse spinal cord. Dev Neurosci 2006; 27:364-77. [PMID: 16280634 DOI: 10.1159/000088452] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2005] [Accepted: 04/12/2005] [Indexed: 11/19/2022] Open
Abstract
The central nervous system of the mammalian embryo is organized according to the expression of region-specific transcription factors along the anteroposterior and/or the dorsoventral axis. For example, the dorsal ventricular zone (VZ) of the embryonic spinal cord expresses Pax3 and Pax7, the ventral VZ expresses Pax6, and the more ventral VZ expresses Nkx2.2. Properties of neuronal precursors located in the VZ are determined by the characteristic expression patterns of these transcription factors, leading to the generation of distinct classes of neurons. Recent studies demonstrated that radial glial cells produce neurons in addition to glia during central nervous system development. Thus, neuronal precursor diversity may be dependent upon the diversity of radial glial cells. To investigate this hypothesis, we analyzed the expression of radial glial cell markers and transcription factors in the mouse embryonic spinal cord. We show that radial glial cells indeed express domain-specific transcription factor. Moreover, they varied in expression of the astrocyte-specific glutamate transporter. The region where the astrocyte-specific glutamate transporter is strongly expressed in the ventral radial glial cells is closely related to the Pax6-expressing domain, and the weakly expressing region corresponding to the Nkx2.2-expressing domain. Furthermore, dorsal radial fibers expressed ephrin-B1. Thus, different types of radial glial cells exist in different domains defined by the transcription factor expression at E12.5. We also show that this diversity continues to the gliogenic stage of radial glial cells. This raises the idea that astrocytes generated from different domains along the dorsoventral axis in the mouse spinal cord have distinct characteristics.
Collapse
Affiliation(s)
- Yasuhiro Ogawa
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Japan
| | | | | | | | | | | |
Collapse
|
26
|
Abstract
The map of the retina onto the optic tectum is a highly conserved feature of the vertebrate visual system; the mechanism by which this mapping is accomplished during development is a long-standing problem of neurobiology. The early suggestion by Roger Sperry that the map is formed through interactions between retinal ganglion cell axons and target cells within the tectum has gained significant experimental support and widespread acceptance. Nonetheless, reports in a variety of species indicate that some aspects of retinotopic order exist within the optic tract, leading to the suggestion that this "preordering" of retinal axons may play a role in the formation of the mature tectal map. A satisfactory account of pretarget order must provide the mechanism by which such axon order develops. Insofar as this mechanism must ultimately be determined genetically, the mouse suggests itself as the natural species in which to pursue these studies. Quantitative and repeatable methods are required to assess the contribution of candidate genes in mouse models. For these reasons, we have undertaken a quantitative study of the degree of retinotopic order within the optic tract and nerve of wild-type mice both before and after the development of the retinotectal map. Our methods are based on tract tracing using lipophilic dyes, and our results indicate that there is a reestablishment of dorsoventral but not nasotemporal retinal order when the axons pass through the chiasm and that this order is maintained throughout the subsequent tract. Furthermore, this dorsoventral retinotopic order is well established by the day after birth, long before the final target zone is discernible within the tectum. We conclude that pretarget sorting of axons according to origin along the dorsoventral axis of the retina is both spatially and chronologically appropriate to contribute to the formation of the retinotectal map, and we suggest that these methods be used to search for the molecular basis of such order by using available mouse genetic models.
Collapse
Affiliation(s)
- Daniel T Plas
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | |
Collapse
|
27
|
Lane S, McDermott K, Dockery P, Fraher J. The developing cervical spinal ventral commissure of the rat: a highly controlled axon-glial system. ACTA ACUST UNITED AC 2005; 33:489-501. [PMID: 15906157 DOI: 10.1007/s11068-004-0512-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2004] [Revised: 09/13/2004] [Accepted: 09/13/2004] [Indexed: 10/25/2022]
Abstract
The floor plate of the neural tube is of major importance in determining axonal behaviour, such that, having crossed, decussating axons do not cross back again. The ventral commissure (VC) of the spinal cord forms immediately ventral to the floor plate shortly after neural tube closure. It is the principal location in which decussating axons cross the midline. It is probably also of major importance in neural tube development, but has received relatively little attention. This study analyses the growth and development of the rat VC and also axon-glial relationships within it throughout the crucial prenatal period of extensive transmedian axon growth, when key biochemical interactions between the two tissues are taking place. The morphometric, stereological and immunohistochemical methods used show that the axonal and glial populations remain in a finely balanced equilibrium throughout a period of almost a hundred-fold growth of both elements. At all stages axons are highly segregated into small bundles of constant size by glial processes, to which they are closely apposed. Thus, glial-axon contact is remarkably precocious, uniquely intimate and persists throughout VC development. This suggests that the relationship between the two tissues is highly controlled through interactions between them. The VC is likely to be the physical basis of a second set of glial-axonal interactions, namely, those which are well known to influence axon crossing behaviour. In mediating these, the extensive axon-glial contact is an ideal arrangement for molecular transfer between them, and is probably the substrate for altering axon responsiveness and ensuring reliable transmedian decussation. The VC is therefore a segregating matrix temporally and spatially specialised for a range of key developmental axon-glial interactions.
Collapse
Affiliation(s)
- Sile Lane
- Department of Anatomy/Neuroscience, BioSciences Institute, National University of Ireland Cork, Cork, Ireland
| | | | | | | |
Collapse
|
28
|
Calò L, Spillantini M, Nicoletti F, Allen ND. Nurr1 co-localizes with EphB1 receptors in the developing ventral midbrain, and its expression is enhanced by the EphB1 ligand, ephrinB2. J Neurochem 2005; 92:235-45. [PMID: 15663472 DOI: 10.1111/j.1471-4159.2004.02853.x] [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] [Indexed: 11/30/2022]
Abstract
Both ephrins and the transcription factor, Nurr1, are critically involved in CNS development and, particularly, in the ontogenesis of the nigro-striatal system. Here we examined whether the ephrin receptor, EphB1, and Nurr1 share a similar expression pattern in the embryonic brain and whether expression of Nurr1 is under the control of EphB1 activation. The transcripts of EphB1 receptor and Nurr1 showed a similar pattern of expression in the ventral midbrain of mice at early stages of embryonic development (E11.5 and E12.5). At later stages (E15.5), only Nurr1 mRNA could still be detected in significant amounts in the A9-A10 regions of the ventral midbrain, whereas the two transcripts still showed a similar pattern of expression in discrete regions of the hindbrain. To examine whether activation of EphB1 receptor could induce the expression of Nurr1 in the ventral midbrain, we applied the EphB1 ligand, ephrinB2, to explants of embryonic mouse ventral midbrain. Low concentrations of clustered ephrinB2 (0.25 microg/mL) enhanced Nurr1 mRNA and protein levels, whereas higher concentrations were inactive. We conclude that activation of EphB1 receptors by appropriate concentrations of its ligand ephrinB2 might contribute to the acquisition of a dopaminergic fate in developing midbrain ventral neurones.
Collapse
Affiliation(s)
- Laura Calò
- Laboratory of Cognitive and Developmental Neurobiology, The Babraham Institute, Babraham, Cambridge, UK.
| | | | | | | |
Collapse
|
29
|
Wang Y, Ying GX, Liu X, Wang WY, Dong JH, Ni ZM, Zhou CF. Induction of ephrin-B1 and EphB receptors during denervation-induced plasticity in the adult mouse hippocampus. Eur J Neurosci 2005; 21:2336-46. [PMID: 15932593 DOI: 10.1111/j.1460-9568.2005.04093.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Abstract It has been widely demonstrated that Eph receptors and their ephrin ligands play multiple pivotal roles in the development of the nervous system. However, less is known about their roles in the adult brain. Here we reported the expression of ephrin-B1 and its cognate EphB receptors in the adult mouse hippocampus at 3, 7, 15, 30 and 60 days after transections of the entorhinal afferents. In situ hybridization and immunohistochemistry showed the time-dependent up-regulation of ephrin-B1 in the denervated areas of the hippocampus, which initiated at 3 days postlesion (dpl), reached maximal levels at 7-15 dpl, remained slightly elevated at 30 dpl and recovered to normal levels by 60 dpl. Double labeling of ephrin-B1 and glial fibrillary acidic protein revealed that ephrin-B1-expressing cells in the denervated areas were reactive astrocytes. Furthermore, a ligand-binding assay using ephrin-B1/Fc chimera protein also displayed the up-regulation of EphB receptors in the denervated areas of the hippocampus in a similar manner to that of ephrin-B1. Within the first week postlesion, the EphB receptors were expressed by reactive astrocytes. After 7 dpl, however, EphB receptors were expressed not only by reactive astrocytes but also first by sprouting axons and later by regrowing dendrites. These results suggest that the ephrin-B1/EphB system may participate in the lesion-induced plasticity processes in the adult mouse hippocampus.
Collapse
Affiliation(s)
- Yan Wang
- Key Laboratory of Neurobiology, Shanghai Institute of Physiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, PR China
| | | | | | | | | | | | | |
Collapse
|
30
|
Siddiqui SA, Cramer KS. Differential expression of Eph receptors and ephrins in the cochlear ganglion and eighth cranial nerve of the chick embryo. J Comp Neurol 2005; 482:309-19. [PMID: 15669077 DOI: 10.1002/cne.20396] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The cochleovestibular ganglion of the chick differentiates at early embryonic stages as VIIIth nerve axons enter the brainstem. The tonotopic organization of the auditory portion of the VIIIth nerve can be discerned at the time axons initially reach their brainstem targets. The mechanisms underlying this early organization are not known. Eph receptor tyrosine kinases and their ligands, the ephrins, have a demonstrated role in guiding axons to topographically appropriate locations in other areas of the nervous system. In order to begin to test whether Eph proteins have a similar role in the auditory system, we investigated the tonotopic expression of several Eph receptors and ephrins in the VIIIth nerve during embryonic ages corresponding to the initial innervation of the auditory brainstem. Expression patterns of EphA4, EphB2, EphB5, ephrin-A2, and ephrin-B1 were evaluated immunohistochemically at embryonic days 4 through 10. Protein expression was observed in the cochlear ganglion and VIIIth nerve axons at these ages. EphB5, ephrin-A2, and ephrin-B1 were expressed throughout the nerve. EphA4 and EphB2 had complementary expression patterns within the nerve, with EphA4 expression higher in the dorsolateral part of the nerve and EphB2 expression higher in the ventromedial part of the nerve. These regions may correspond to auditory and vestibular components, respectively. Moreover, EphA4 expression was higher toward the low-frequency region of both the centrally and peripherally projecting branches of cochlear ganglion cells. Regional variation of Eph protein expression may influence the target selection and topography of developing VIIIth nerve projections.
Collapse
Affiliation(s)
- Shazia A Siddiqui
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, California 92697, USA
| | | |
Collapse
|
31
|
Martínez A, Soriano E. Functions of ephrin/Eph interactions in the development of the nervous system: emphasis on the hippocampal system. ACTA ACUST UNITED AC 2005; 49:211-26. [PMID: 16111551 DOI: 10.1016/j.brainresrev.2005.02.001] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2004] [Revised: 02/01/2005] [Accepted: 02/04/2005] [Indexed: 12/20/2022]
Abstract
Ephrins and their Eph receptors are membrane-anchored proteins that have key roles in the development of the Central Nervous System. The main characteristics of ephrin/Eph interactions are that their effect is mediated by cell-to-cell contacts and that they can propagate bidirectional signals downstream of the ligand-receptor complex. These characteristics make ephrins and Eph receptors critical cues in the regulation of migrating cells or axons, and in the establishment of tissue patterns and topographic maps in distinct regions of the developing brain. In addition, ephrins and Eph receptors regulate synapse formation and plasticity. These roles would be promoted by complementary gradual expression of receptors and ligands in the neurons involved. Although, historically, ephrins and Eph receptors have been considered as repulsion signals through barriers or gradients, new evidence indicates that they may be both inhibitory and permissive/active cues depending on expression levels. The expression of distinct ligands and receptors in the developing and mature hippocampus suggests that these proteins are involved in distinct processes during the development and maturation of the hippocampal region. In fact, recent studies have shown that ephrin/Eph signaling participates in the formation of the layer-specific patterns of hippocampal afferents, in synaptogenesis and in plasticity. Therefore, ephrin/Eph interactions should be considered a crucial system in the development and maturation of the brain regions, including the hippocampus.
Collapse
Affiliation(s)
- Albert Martínez
- Neuronal Development and Regeneration Group (S1-A1), Department of Cell Biology, University of Barcelona/Barcelona Science Park, Josep Samitier 1-5, Barcelona E-08028, Spain.
| | | |
Collapse
|
32
|
Abstract
The human brain assembles an incredible network of over a billion neurons. Understanding how these connections form during development in order for the brain to function properly is a fundamental question in biology. Much of this wiring takes place during embryonic development. Neurons are generated in the ventricular zone, migrate out, and begin to differentiate. However, neurons are often born in locations some distance from the target cells with which they will ultimately form connections. To form connections, neurons project long axons tipped with a specialized sensing device called a growth cone. The growing axons interact directly with molecules within the environment through which they grow. In order to find their targets, axonal growth cones use guidance molecules that can either attract or repel them. Understanding what these guidance cues are, where they are expressed, and how the growth cone is able to transduce their signal in a directionally specific manner is essential to understanding how the functional brain is constructed. In this chapter, we review what is known about the mechanisms involved in axonal guidance. We discuss how the growth cone is able to sense and respond to its environment and how it is guided by pioneering cells and axons. As examples, we discuss current models for the development of the spinal cord, the cerebral cortex, and the visual and olfactory systems.
Collapse
Affiliation(s)
- Céline Plachez
- Department of Anatomy and Neurobiology, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| | | |
Collapse
|
33
|
Kadison SR, Kaprielian Z. Diversity of contralateral commissural projections in the embryonic rodent spinal cord. J Comp Neurol 2004; 472:411-22. [PMID: 15065116 DOI: 10.1002/cne.20086] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In vertebrate embryos, the axons of spinal commissural neurons grow toward and across the floor plate, a specialized structure located at the ventral midline. Although the initial segment of this trajectory has been intensively studied, relatively little is known about commissural axon pathfinding on the contralateral side of the floor plate in higher vertebrates. We recently demonstrated that many embryonic mouse and chick spinal commissural axons follow a complex trajectory once they cross the ventral midline. Here we use focal applications of 1,1'-dioctadecyl-3,3,3',3' tetramethylindocarbocyanine perchlorate (DiI) to identify four different contralateral commissural trajectories, two of which have not previously been described in the embryonic rodent spinal cord. Intermediate longitudinal commissural (ILC) axons travel away from the floor plate along an arcuate trajectory into intermediate regions of the spinal cord. In contrast, medial longitudinal commissural (MLC) axons grow alongside the floor plate, projecting primarily in the rostral direction. Bifurcating longitudinal commissural (BLC) axons branch into rostrally and caudally directed projections. Forked transverse commissural (FTC) axons either execute two orthogonal turns before crossing the floor plate or extend directly across the floor plate. We also show a variation in the relative frequencies of individual contralateral commissural projections along the dorsoventral and anteroposterior axes of the spinal cord. In addition, using a novel culture system, we demonstrate that commissural axons elaborate ILC-, MLC-, BLC-, and FTC-like trajectories in vitro. These results provide a basis for examining the mechanisms that regulate commissural axon pathfinding on the contralateral side of the floor plate in the embryonic rodent spinal cord.
Collapse
|
34
|
Long H, Sabatier C, Ma L, Plump A, Yuan W, Ornitz DM, Tamada A, Murakami F, Goodman CS, Tessier-Lavigne M. Conserved roles for Slit and Robo proteins in midline commissural axon guidance. Neuron 2004; 42:213-23. [PMID: 15091338 DOI: 10.1016/s0896-6273(04)00179-5] [Citation(s) in RCA: 356] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2003] [Revised: 02/24/2004] [Accepted: 03/12/2004] [Indexed: 11/30/2022]
Abstract
In Drosophila, Slit at the midline activates Robo receptors on commissural axons, thereby repelling them out of the midline into distinct longitudinal tracts on the contralateral side of the central nervous system. In the vertebrate spinal cord, Robo1 and Robo2 are expressed by commissural neurons, whereas all three Slit homologs are expressed at the ventral midline. Previous analysis of Slit1;Slit2 double mutant spinal cords failed to reveal a defect in commissural axon guidance. We report here that when all six Slit alleles are removed, many commissural axons fail to leave the midline, while others recross it. In addition, Robo1 and Robo2 single mutants show guidance defects that reveal a role for these two receptors in guiding commissural axons to different positions within the ventral and lateral funiculi. These results demonstrate a key role for Slit/Robo signaling in midline commissural axon guidance in vertebrates.
Collapse
Affiliation(s)
- Hua Long
- Howard Hughes Medical Institute, Department of Biological Sciences, Stanford University, Stanford, CA 94305, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
The mouse optic chiasm is a model for axon guidance at the midline and for analyzing how binocular vision is patterned. Recent work has identified several molecular players that influence the binary decision that retinal ganglion cells make at the optic chiasm, to either cross or avoid the midline. An ephrin-B localized to the midline, together with an EphB receptor and a zinc-finger transcription factor expressed exclusively in the ventrotemporal retina where ipsilaterally projecting retinal ganglion cells are located, comprise a molecular program for the uncrossed pathway. In addition, the mechanisms for axon divergence in the optic chiasm are discussed in the context of other popular models for midline axon guidance.
Collapse
Affiliation(s)
- Scott E Williams
- Department of Pathology, Center for Neurobiology and Behavior, Columbia University, College of Physicians and Surgeons, 630 W. 168(th) Street, New York, NY 10032, USA
| | | | | |
Collapse
|
36
|
Abstract
Cell adhesion molecules of the immunoglobulin superfamily (IgSF CAMs) were discovered 25 years ago based on their role in cell-cell adhesion. Ever since, they have played a major role in developmental neuroscience research. The elucidation of IgSF CAM structure and function has been tightly linked to the establishment of new areas of research. Over the years, our view of the role of the IgSF CAMs has changed. First, they were thought to provide "specific glue" segregating subtypes of cells in the nervous system. Soon it became clear that IgSF CAMs can do much more. The focus shifted from simple adhesion to CAM-associated signaling that was shown to be involved in the promotion of axon growth and the regulation of cell migration. From there it was a small step to axon guidance, a field that has been given a lot of attention during the last decade. More recently, the involvement of IgSF CAMs in synapse formation and maturation has been discovered, although this last step in the formation of neural circuits was thought to be the domain of other families of cell adhesion molecules, such as the neuroligins, the neurexins, and the cadherins. Certainly, the most striking discovery in the context of IgSF CAMs has been the diversity of signaling mechanisms that are associated with them. The versatility of signals and their complexity make IgSF CAMs a perfect tool for brain development.
Collapse
Affiliation(s)
- E T Stoeckli
- Institute of Zoology, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland,
| |
Collapse
|
37
|
Mann F, Miranda E, Weinl C, Harmer E, Holt CE. B-type Eph receptors and ephrins induce growth cone collapse through distinct intracellular pathways. JOURNAL OF NEUROBIOLOGY 2003; 57:323-36. [PMID: 14608666 PMCID: PMC3683941 DOI: 10.1002/neu.10303] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Forward and reverse signaling mediated by EphB tyrosine kinase receptors and their transmembrane ephrin-B ligands play important roles in axon pathfinding, yet little is known about the intracellular pathways involved. Here we have used growth cones from the ventral (EphB receptor-bearing) and dorsal (ephrin-B-bearing) embryonic Xenopus retina to investigate the signaling mechanisms in both forward and reverse directions. We report that unclustered, but not clustered, EphB2 ectodomains trigger fast (5-10 min) transient collapse responses in growth cones. This collapse response is mediated by low levels of intracellular cyclic GMP and requires proteasome function. In contrast, clustered, but not unclustered, ephrin-B1 ectodomains cause slow (30-60 min) growth cone collapse that depends on high cGMP levels and is insensitive to inhibition of the proteasomal pathway. Upon receptor-ligand binding, endocytosis occurs in the reverse direction (EphB2-Fc into dorsal retinal growth cones), but not the forward direction, and is also sensitive to proteasomal inhibition. Endocytosis is functionally important because blocking of EphB2 internalization inhibits growth cone collapse. Our data reveal that distinct signaling mechanisms exist for B-type Eph/ephrin-mediated growth cone guidance and suggest that endocytosis provides a fast mechanism for switching off signaling in the reverse direction.
Collapse
Affiliation(s)
- Fanny Mann
- Department of Anatomy, University of Cambridge, Downing Street, Cambridge CB2 3DY, United Kingdom
| | | | | | | | | |
Collapse
|
38
|
Palmer A, Klein R. Multiple roles of ephrins in morphogenesis, neuronal networking, and brain function. Genes Dev 2003; 17:1429-50. [PMID: 12815065 DOI: 10.1101/gad.1093703] [Citation(s) in RCA: 220] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Amparo Palmer
- Max-Planck Institute of Neurobiology, Department of Molecular Neurobiology, D-82152 Martinsried, Germany.
| | | |
Collapse
|
39
|
Hahn AC, Emmons SW. The roles of an ephrin and a semaphorin in patterning cell-cell contacts in C. elegans sensory organ development. Dev Biol 2003; 256:379-88. [PMID: 12679110 DOI: 10.1016/s0012-1606(02)00129-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Ephrins and semaphorins regulate a wide variety of developmental processes, including axon guidance and cell migration. We have studied the roles of the ephrin EFN-4 and the semaphorin MAB-20 in patterning cell-cell contacts among the cells that give rise to the ray sensory organs of Caenorhabditis elegans. In wild-type, contacts at adherens junctions form only between cells belonging to the same ray. In efn-4 and mab-20 mutants, ectopic contacts form between cells belonging to different rays. Ectopic contacts also occur in mutants in regulatory genes that specify ray morphological identity. We used efn-4 and mab-20 reporters to investigate whether these ray identity genes function through activating expression of efn-4 or mab-20 in ray cells. mab-20 reporter expression in ray cells was unaffected by mutants in the Pax6 homolog mab-18 and the Hox genes egl-5 and mab-5, suggesting that these genes do not regulate mab-20 expression. We find that mab-18 is necessary for activating efn-4 reporter expression, but this activity alone is not sufficient to account for mab-18 function in controlling cell-cell contact formation. In egl-5 mutants, efn-4 reporter expression in certain ray cells was increased, inconsistent with a simple repulsion model for efn-4 action. The evidence indicates that ray identity genes primarily regulate ray morphogenesis by pathways other than through regulation of expression of semaphorin and ephrin.
Collapse
Affiliation(s)
- Andrew C Hahn
- Department of Molecular Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | |
Collapse
|
40
|
Beltran PJ, Bixby JL, Masters BA. Expression of PTPRO during mouse development suggests involvement in axonogenesis and differentiation of NT-3 and NGF-dependent neurons. J Comp Neurol 2003; 456:384-95. [PMID: 12532410 DOI: 10.1002/cne.10532] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Competition and cooperation between type II and type III receptor protein tyrosine phosphatases (RPTPs) regulate axon extension and pathfinding in Drosophila. The first step to investigate whether RPTPs influence axon growth in the more complex vertebrate nervous system is to identify which neurons express a particular RPTP. We studied the expression of mouse PTPRO, a type III RPTP with an extracellular region containing eight fibronectin type III domains, during embryogenesis and after birth. Mouse PTPRO mRNA is expressed exclusively in two cell types: neurons and kidney podocytes. Maximal expression in the brain was coincident with mid to late gestation and axonogenesis in the brain. We cloned two cDNAs, including a splice variant without sequence coding of 28 amino acids within the juxtamembrane domain that was found mostly in kidney. In situ hybridization detected mPTPRO mRNA in the cerebral cortex, olfactory bulb and nucleus, hippocampus, motor neurons, and the spinal cord midline. In addition, mPTPRO mRNA was found throughout dorsal root, cranial, and sympathetic ganglia and within kidney glomeruli. Mouse PTPRO mRNA was observed in neuron populations expressing TrkA, the high-affinity nerve growth factor receptor, or TrkC, the neurotrophin-3 receptor, and immunoreactive mPTPRO and TrkC colocalized in large dorsal root ganglia proprioceptive neurons. Our results suggest that mPTPRO is involved in the differentiation and axonogenesis of central and peripheral nervous system neurons, where it is in a position to modulate intracellular responses to neurotrophin-3 and/or nerve growth factor.
Collapse
Affiliation(s)
- Pedro J Beltran
- The Neuroscience Program and Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, Florida 33136, USA
| | | | | |
Collapse
|
41
|
Cavalcante LA, Garcia-Abreu J, Moura Neto V, Silva LC, Weissmüller G. Modulators of axonal growth and guidance at the brain midline with special reference to glial heparan sulfate proteoglycans. AN ACAD BRAS CIENC 2002; 74:691-716. [PMID: 12563418 DOI: 10.1590/s0001-37652002000400010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Bilaterally symmetric organisms need to exchange information between the left and right sides of their bodies to integrate sensory input and to coordinate motor control. Thus, an important choice point for developing axons is the Central Nervous System (CNS) midline. Crossing of this choice point is influenced by highly conserved, soluble or membrane-bound molecules such as the L1 subfamily, laminin, netrins, slits, semaphorins, Eph-receptors and ephrins, etc. Furthermore, there is much circumstantial evidence for a role of proteoglycans (PGs) or their glycosaminoglycan (GAG) moieties on axonal growth and guidance, most of which was derived from simplified models. A model of intermediate complexity is that of cocultures of young neurons and astroglial carpets (confluent cultures) obtained from medial and lateral sectors of the embryonic rodent midbrain soon after formation of its commissures. Neurite production in these cocultures reveals that, irrespective of the previous location of neurons in the midbrain, medial astrocytes exerted an inhibitory or non-permissive effect on neuritic growth that was correlated to a higher content of both heparan and chondroitin sulfates (HS and CS). Treatment with GAG lyases shows minor effects of CS and discloses a major inhibitory or non-permissive role for HS. The results are discussed in terms of available knowledge on the binding of HSPGs to interative proteins and underscore the importance of understanding glial polysaccharide arrays in addition to its protein complement for a better understanding of neuron-glial interactions.
Collapse
Affiliation(s)
- Leny A Cavalcante
- Instituto de Biofísica Carlos Chagas Filho, CCS, Universidade Federal do Rio de Janeiro, 21949-900, Rio de Janeiro, Brazil.
| | | | | | | | | |
Collapse
|
42
|
Cramer KS, Karam SD, Bothwell M, Cerretti DP, Pasquale EB, Rubel EW. Expression of EphB receptors and EphrinB ligands in the developing chick auditory brainstem. J Comp Neurol 2002; 452:51-64. [PMID: 12205709 DOI: 10.1002/cne.10399] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Nucleus magnocellularis (NM) in the avian auditory brainstem receives auditory input from nerve the VIIIth and projects bilaterally to nucleus laminaris (NL). This projection preserves binaural segregation in that ipsilateral NM projects to dorsal dendrites of NL and contralateral NM projects to ventral dendrites of NL. We have begun to examine the molecular signals that influence segregation of inputs onto discrete regions of NL cells. We previously showed that the Eph receptor, EphA4, is expressed selectively in the dorsal NL neuropil from embryonic day (E) 9 to E11, when NM axons grow into the NL neuropil. This asymmetric distribution suggests that EphA4 acts as a guidance molecule during binaural segregation. We report here on the developmental changes in the expression of two other Eph receptors, EphB2 and EphB5, and two ligands, ephrin-B1 and ephrin-B2, in the chick auditory brainstem. These proteins are expressed in the auditory nuclei during the maturation of the NM-NL projection. EphB2, EphB5, and ephrin-B1 are expressed in dorsal and ventral NL neuropil and at the midline of the brainstem at E10-E12. At this age, ephrin-B2, a ligand for EphB receptors and for EphA4, is expressed in NL cell bodies and NM-NL axons. The expression of these proteins diminishs in the posthatch ages examined. These results suggest that several members of the Eph family are involved in maturation of the nuclei and their projections. Moreover, ephrin-B2 in growing axons may interact with the asymmetrically expressed EphA4 during the establishment of binaural segregation.
Collapse
Affiliation(s)
- Karina S Cramer
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Washington, Seattle, Washington 98195, USA.
| | | | | | | | | | | |
Collapse
|
43
|
Butt SJB, Lebret JM, Kiehn O. Organization of left-right coordination in the mammalian locomotor network. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 2002; 40:107-17. [PMID: 12589910 DOI: 10.1016/s0165-0173(02)00194-7] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Neuronal circuits involved in left-right coordination are a fundamental feature of rhythmic locomotor movements. These circuits necessarily include commissural interneurons (CINs) that have axons crossing the midline of the spinal cord. The properties of CINs have been described in some detail in the spinal cords of a number of aquatic vertebrates including the Xenopus tadpole and the lamprey. However, their function in left-right coordination of limb movements in mammals is poorly understood. In this review we describe the present understanding of commissural pathways in the functioning of spinal cord central pattern generators (CPGs). The means by which reciprocal inhibition and integration of sensory information are maintained in swimming vertebrates is described, with similarities between the three basic populations of commissural interneurons highlighted. The subsequent section concentrates on recent evidence from mammalian limbed preparations and specifically the isolated spinal cord of the neonatal rat. Studies into the role of CPG elements during drug-induced locomotor-like activity have afforded a better understanding of the location of commissural pathways, such that it is now possible, using whole cell patch clamp, to record from anatomically defined CINs located in the rhythm-generating region of the lumbar segments. Initial results would suggest that the firing pattern of these neurons shows a greater diversity than that previously described in swimming central pattern generators. Spinal CINs play an important role in the generation of locomotor output. Increased knowledge as to their function in producing locomotion is likely to provide valuable insights into the spinal networks required for postural control and walking.
Collapse
Affiliation(s)
- Simon J B Butt
- Department of Neuroscience, Karolinska Institutet, Retzius Vag 8, S-171 77, Stockholm, Sweden
| | | | | |
Collapse
|
44
|
Runko E, Kaprielian Z. Expression of Vema in the developing mouse spinal cord and optic chiasm. J Comp Neurol 2002; 451:289-99. [PMID: 12210140 DOI: 10.1002/cne.10356] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
A critical phase of nervous system development is the formation of connections between axons and their synaptic targets. Intermediate targets play important roles in axon pathfinding by supplying growing axons with long- and short- range guidance cues at decision points along their trajectory. We recently identified Vema as a novel membrane-associated protein that is expressed at the ventral midline of the developing vertebrate central nervous system (CNS). We report that Vema is expressed in the floor plate, an intermediate target for pathfinding commissural axons located at the ventral midline of the developing mouse spinal cord. Interestingly, Vema expression overlaps with the position of an unique population of neurons situated at the midline of the ventral diencephalon and that function as intermediate targets for pathfinding retinal ganglion cell axons. The distribution of Vema in the developing spinal cord and optic chiasm resembles the expression patterns of a variety of molecules known to play important roles in axon guidance, including Robo2, Neuropilin2, and SSEA. The expression of Vema at two key choice points for pathfinding axons suggests an important role for this protein in regulating axon guidance at the midline of the developing mouse central nervous system.
Collapse
Affiliation(s)
- Erik Runko
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | |
Collapse
|
45
|
Takemoto M, Fukuda T, Sonoda R, Murakami F, Tanaka H, Yamamoto N. Ephrin-B3-EphA4 interactions regulate the growth of specific thalamocortical axon populations in vitro. Eur J Neurosci 2002; 16:1168-72. [PMID: 12383247 DOI: 10.1046/j.1460-9568.2002.02166.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The role was studied of ephrin-B3, a ligand of the Eph family of tyrosine kinase receptors, in the formation of cortical connectivity. In situ hybridization and immunohistochemistry showed that EphA4, a receptor of ephrin-B3, was expressed in the lateral thalamus (visual and somaotosensory thalamus) of the developing rat brain, but not in the medial thalamic nuclei which project to the limbic cortex. Correspondingly, ephrin-B3 was expressed strongly in the developing limbic cortex including amygdala, entorhinal cortex and hippocampus. To examine the action of ephrin-B3 on thalamic axons, either lateral or medial thalamic explants were cultured on membranes obtained from ephrin-B3-expressing COS cells. Axonal growth was inhibited for cells from the lateral thalamus but not from the medial thalamus. These results suggest that ephrin-B3 contributes to regional specificity by suppressing axonal growth of lateral thalamic neurons.
Collapse
Affiliation(s)
- Makoto Takemoto
- Neuroscience Laboratories, Graduate School of Frontier Biosciences, Osaka University, Toyonaka, Osaka 560-8531, Japan
| | | | | | | | | | | |
Collapse
|
46
|
Honda H, Mochizuki A. Formation and maintenance of distinctive cell patterns by coexpression of membrane-bound ligands and their receptors. Dev Dyn 2002; 223:180-92. [PMID: 11836783 DOI: 10.1002/dvdy.10042] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
We show that graded or checkerboard-like cell patterns, and segmental domains along a body axis, can be generated by cell behaviors involving differences in intercellular repulsion. A membrane-bound signal transduction system mediating contact-dependent cell interactions includes membrane-bound ligands (ephrins) and their receptors with tyrosine-kinase activity (Eph proteins). These molecules mediate both repulsive and attractive interactions under bilateral threshold control, i.e., cells expressing the receptors adhere to a surface bearing a critical density of ligand reciprocal to the density of receptor but are repelled by a surface with other densities of ligand (Honda [1998] J. Theor. Biol. 192:235-246). We extend this model. General membrane-bound ligands (not always ephrins) and their receptors are presumably coexpressed in a single cell under bilateral threshold control. Computer simulations of cell pattern formation showed that when coexpression of the ligand and receptor is reciprocal, the cells self-organize into a pattern of segmental domains or a graded cell arrangement along the body axis. The latter process interprets positional information in terms of protein molecules. When coexpression of the two species of molecules is not always reciprocal, the cells generate various patterns including checkerboard and kagome (star) patterns. The case of separate expression of ligands and receptors in different cells is also examined. The mechanism of differences in cell repulsion is compared with the differential cell adhesion hypothesis, which has been used to explain cell sorting.
Collapse
Affiliation(s)
- Hisao Honda
- Faculty of Health Science, Hyogo University, Kakogawa, Hyogo, Japan.
| | | |
Collapse
|
47
|
Schubert W, Kaprielian Z. Identification and characterization of a cell surface marker for embryonic rat spinal accessory motor neurons. J Comp Neurol 2001; 439:368-83. [PMID: 11596060 DOI: 10.1002/cne.1356] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The developing mammalian spinal cord contains distinct populations of motor neurons that can be distinguished by their cell body positions, by the expression of specific combinations of regulatory genes, and by the paths that their axons take to exit the central nervous system (CNS). Subclasses of spinal motor neurons are also thought to express specific cell surface proteins that function as receptors which control the guidance of their axons. We identified monoclonal antibody (mAb) SAC1 in a screen aimed at generating markers for specific subsets of neurons/axons in the developing rat spinal cord. During early embryogenesis, mAb SAC1 selectively labels a small subset of Isl1-positive motor neurons located exclusively within cervical segments of the spinal cord. Strikingly, these neurons extend mAb SAC1-positive axons along a dorsally directed trajectory toward the lateral exit points. Consistent with the finding that mAb SAC1 also labels spinal accessory nerves, these observations identify mAb SAC1 as a specific marker of spinal accessory motor neurons/axons. During later stages of embryogenesis, mAb SAC1 is transiently expressed on both dorsally and ventrally projecting spinal motor neurons/axons. Interestingly, mAb SAC1 also labels the notochord and floor plate during most stages of spinal cord development. The mAb SAC1 antigen is a 100-kD glycoprotein that is likely to be the rat homolog of SC1/BEN/DM-GRASP, a homophilic adhesion molecule that mediates axon outgrowth and fasciculation.
Collapse
Affiliation(s)
- W Schubert
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | |
Collapse
|
48
|
Lustig M, Erskine L, Mason CA, Grumet M, Sakurai T. Nr-CAM expression in the developing mouse nervous system: ventral midline structures, specific fiber tracts, and neuropilar regions. J Comp Neurol 2001; 434:13-28. [PMID: 11329126 DOI: 10.1002/cne.1161] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Nr-CAM is a member of the L1 subfamily of cell adhesion molecules (CAMs) that belong to the immunoglobulin superfamily. To explore the role of Nr-CAM in the developing nervous system, we prepared specific antibodies against both chick and mouse Nr-CAM using recombinant Fc fusion proteins of chick Nr-CAM and mouse Nr-CAM, respectively. First, we show the specificity of the new anti-chick Nr-CAM antibody compared with a previously employed antibody using the expression patterns of Nr-CAM in the chick spinal cord and floor plate and on commissural axons, where Nr-CAM has been implicated in axon guidance. Using the anti-mouse Nr-CAM antibody, we then studied the expression patterns of Nr-CAM in the developing mouse nervous system along with the patterns of two related CAMs, L1, which labels most growing axons, and TAG-1, which binds to Nr-CAM and has a more restricted distribution. Major sites that are positive for Nr-CAM are specialized glial formations in the ventral midline, including the floor plate in the spinal cord, the hindbrain and midbrain, the optic chiasm, and the median eminence in the forebrain. Similar to what is seen in the chick spinal cord, Nr-CAM is expressed on crossing fibers as they course through these areas. In addition, Nr-CAM is found in crossing fiber pathways, including the anterior commissure, corpus callosum, and posterior commissure, and in nondecussating pathways, such as the lateral olfactory tract and the habenulointerpeduncular tract. Nr-CAM, for the most part, is colocalized with TAG-1 in all of these systems. Based on in vitro studies indicating that the Nr-CAM-axonin-1/TAG-1 interaction is involved in peripheral axonal growth and guidance in the spinal cord [Lustig et al. (1999) Dev Biol 209:340-351; Fitzli et al. (2000) J Cell Biol 149:951-968], the expression patterns described herein implicate a role for this interaction in central nervous system axon growth and guidance, especially at points of decussation. Nr-CAM also is expressed in cortical regions, such as the olfactory bulb. In the hippocampus, however, TAG-1-positive areas are segregated from Nr-CAM-positive areas, suggesting that, in neuropilar regions, Nr-CAM interacts with molecules other than TAG-1.
Collapse
Affiliation(s)
- M Lustig
- Department of Pharmacology, New York University Medical Center, New York, New York 10016, USA
| | | | | | | | | |
Collapse
|
49
|
Abstract
A central feature of the developing nervous system is the midline region, which guides growing axons with both short- and long-range signals. New research shows that a trio of receptors, the Robos, are crucial in allowing axons to interpret these signals, ensuring correct route-finding within the emerging axon scaffold.
Collapse
Affiliation(s)
- S Guthrie
- MRC Centre for Developmental Neurobiology, 4th Floor, New Hunt's House, King's College, Guy's Campus, SE1 1UL, London, UK
| |
Collapse
|
50
|
Kullander K, Croll SD, Zimmer M, Pan L, McClain J, Hughes V, Zabski S, DeChiara TM, Klein R, Yancopoulos GD, Gale NW. Ephrin-B3 is the midline barrier that prevents corticospinal tract axons from recrossing, allowing for unilateral motor control. Genes Dev 2001; 15:877-88. [PMID: 11297511 PMCID: PMC312668 DOI: 10.1101/gad.868901] [Citation(s) in RCA: 193] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Growing axons follow highly stereotypical pathways, guided by a variety of attractive and repulsive cues, before establishing specific connections with distant targets. A particularly well-known example that illustrates the complexity of axonal migration pathways involves the axonal projections of motor neurons located in the motor cortex. These projections take a complex route during which they first cross the midline, then form the corticospinal tract, and ultimately connect with motor neurons in the contralateral side of the spinal cord. These obligatory contralateral connections account for why one side of the brain controls movement on the opposing side of the body. The netrins and slits provide well-known midline signals that regulate axonal crossings at the midline. Herein we report that a member of the ephrin family, ephrin-B3, also plays a key role at the midline to regulate axonal crossing. In particular, we show that ephrin-B3 acts as the midline barrier that prevents corticospinal tract projections from recrossing when they enter the spinal gray matter. We report that in ephrin-B3(-/-) mice, corticospinal tract projections freely recross in the spinal gray matter, such that the motor cortex on one side of the brain now provides bilateral input to the spinal cord. This neuroanatomical abnormality in ephrin-B3(-/-) mice correlates with loss of unilateral motor control, yielding mice that simultaneously move their right and left limbs and thus have a peculiar hopping gait quite unlike the alternate step gait displayed by normal mice. The corticospinal and walking defects in ephrin-B3(-/-) mice resemble those recently reported for mice lacking the EphA4 receptor, which binds ephrin-B3 as well as other ephrins, suggesting that the binding of EphA4-bearing axonal processes to ephrin-B3 at the midline provides the repulsive signal that prevents corticospinal tract projections from recrossing the midline in the developing spinal cord.
Collapse
Affiliation(s)
- K Kullander
- European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|