1
|
Roome RB, Yadav A, Flores L, Puarr A, Nardini D, Richardson A, Waclaw RR, Arkell RM, Menon V, Johnson JE, Levine AJ. Ontogeny of the spinal cord dorsal horn. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643370. [PMID: 40161840 PMCID: PMC11952496 DOI: 10.1101/2025.03.14.643370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The dorsal horn of the mammalian spinal cord is an exquisite example of form serving function. It is comprised of diverse neuronal populations stacked into laminae, each of which receives different circuit connections and plays specialized roles in behavior. An outstanding question is how this organization emerges during development from an apparently homogeneous pool of neural progenitors. Here, we found that dorsal neurons are diversified by time, with families of related cell types born as temporal cohorts, and by a spatial-molecular gradient that specifies the full array of individual cell types. Excitatory dorsal neurons then settle in a chronotopic arrangement that transforms their progressive birthdates into anatomical order. This establishes the dorsal horn laminae, as these neurons are also required for spatial organization of inhibitory neurons and sensory axons. This work reveals essential ontogenetic principles that shape dorsal progenitors into the diverse cell types and architecture that subserve sensorimotor behavior.
Collapse
Affiliation(s)
- Robert Brian Roome
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Archana Yadav
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Lydia Flores
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA
| | - Amrit Puarr
- Maternal-foetal Precision Health Laboratory, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Diana Nardini
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Alexander Richardson
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Ronald R. Waclaw
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ruth M. Arkell
- Maternal-foetal Precision Health Laboratory, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Vilas Menon
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jane E. Johnson
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Ariel J. Levine
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
- Lead contact
| |
Collapse
|
2
|
Murcia-Belmonte V, Chauvin G, Coca Y, Escalante A, Klein R, Herrera E. EphA4 Mediates EphrinB1-Dependent Adhesion in Retinal Ganglion Cells. J Neurosci 2025; 45:e0043242024. [PMID: 39622649 PMCID: PMC11756631 DOI: 10.1523/jneurosci.0043-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 10/04/2024] [Accepted: 11/11/2024] [Indexed: 01/24/2025] Open
Abstract
Eph/ephrin signaling is crucial for organizing retinotopic maps in vertebrates. Unlike other EphAs, which are expressed in the embryonic ventral retina, EphA4 is found in the retinal ganglion cell (RGC) layer at perinatal stages, and its role in mammalian visual system development remains unclear. Using classic in vitro stripe assays, we demonstrate that, while RGC axons are repelled by ephrinB2, they grow on ephrinB1 stripes through EphA4-mediated adhesion. In vivo, retinal axons from EphA4-deficient mice from either sex show impaired arborization in the medial, but not lateral, regions of the superior colliculus that express ephrinB1. Gain-of-function experiments further reveal that ephrinB1-mediated adhesion depends on EphA4 tyrosine kinase activity but it is independent of its sterile alpha motif. Together, our findings suggest that EphA4/ephrinB1 forward signaling likely facilitates adhesion between retinal axon terminals and cells in the medial colliculus, contributing to the establishment of proper connectivity within the visual system.
Collapse
Affiliation(s)
- Verónica Murcia-Belmonte
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), Campus San Juan, Alicante 03550, Spain
| | - Géraud Chauvin
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), Campus San Juan, Alicante 03550, Spain
| | - Yaiza Coca
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), Campus San Juan, Alicante 03550, Spain
| | - Augusto Escalante
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), Campus San Juan, Alicante 03550, Spain
| | - Rüdiger Klein
- Department 'Molecules - Signals - Development', Max Planck Institute for Biological Intelligence, Martinsried 82152, Germany
| | - Eloísa Herrera
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), Campus San Juan, Alicante 03550, Spain
| |
Collapse
|
3
|
Barnada SM, Giner de Gracia A, Morenilla-Palao C, López-Cascales MT, Scopa C, Waltrich FJ, Mikkers HMM, Cicardi ME, Karlin J, Trotti D, Peterson KA, Brugmann SA, Santen GWE, McMahon SB, Herrera E, Trizzino M. ARID1A-BAF coordinates ZIC2 genomic occupancy for epithelial-to-mesenchymal transition in cranial neural crest specification. Am J Hum Genet 2024; 111:2232-2252. [PMID: 39226899 PMCID: PMC11480806 DOI: 10.1016/j.ajhg.2024.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 09/05/2024] Open
Abstract
The BAF chromatin remodeler regulates lineage commitment including cranial neural crest cell (CNCC) specification. Variants in BAF subunits cause Coffin-Siris syndrome (CSS), a congenital disorder characterized by coarse craniofacial features and intellectual disability. Approximately 50% of individuals with CSS harbor variants in one of the mutually exclusive BAF subunits, ARID1A/ARID1B. While Arid1a deletion in mouse neural crest causes severe craniofacial phenotypes, little is known about the role of ARID1A in CNCC specification. Using CSS-patient-derived ARID1A+/- induced pluripotent stem cells to model CNCC specification, we discovered that ARID1A-haploinsufficiency impairs epithelial-to-mesenchymal transition (EMT), a process necessary for CNCC delamination and migration from the neural tube. Furthermore, wild-type ARID1A-BAF regulates enhancers associated with EMT genes. ARID1A-BAF binding at these enhancers is impaired in heterozygotes while binding at promoters is unaffected. At the sequence level, these EMT enhancers contain binding motifs for ZIC2, and ZIC2 binding at these sites is ARID1A-dependent. When excluded from EMT enhancers, ZIC2 relocates to neuronal enhancers, triggering aberrant neuronal gene activation. In mice, deletion of Zic2 impairs NCC delamination, while ZIC2 overexpression in chick embryos at post-migratory neural crest stages elicits ectopic delamination from the neural tube. These findings reveal an essential ARID1A-ZIC2 axis essential for EMT and CNCC delamination.
Collapse
Affiliation(s)
- Samantha M Barnada
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Aida Giner de Gracia
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas- Universidad Miguel Hernández, CSIC-UMH). Campus San Juan, Avd. Ramón y Cajal s/n, 03550 San Juan de Alicante, Spain
| | - Cruz Morenilla-Palao
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas- Universidad Miguel Hernández, CSIC-UMH). Campus San Juan, Avd. Ramón y Cajal s/n, 03550 San Juan de Alicante, Spain
| | - Maria Teresa López-Cascales
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas- Universidad Miguel Hernández, CSIC-UMH). Campus San Juan, Avd. Ramón y Cajal s/n, 03550 San Juan de Alicante, Spain
| | - Chiara Scopa
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA, USA
| | - Francis J Waltrich
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Harald M M Mikkers
- Department of Cell & Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Maria Elena Cicardi
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jonathan Karlin
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Davide Trotti
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Samantha A Brugmann
- Division of Developmental Biology, Department of Pediatrics at Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Gijs W E Santen
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Steven B McMahon
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Eloísa Herrera
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas- Universidad Miguel Hernández, CSIC-UMH). Campus San Juan, Avd. Ramón y Cajal s/n, 03550 San Juan de Alicante, Spain.
| | - Marco Trizzino
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA; Department of Life Sciences, Imperial College London, London, UK.
| |
Collapse
|
4
|
Minto MS, Sotelo-Fonseca JE, Ramesh V, West AE. Genome binding properties of Zic transcription factors underlie their changing functions during neuronal maturation. BMC Biol 2024; 22:189. [PMID: 39218853 PMCID: PMC11367862 DOI: 10.1186/s12915-024-01989-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The Zic family of transcription factors (TFs) promote both proliferation and maturation of cerebellar granule neurons (CGNs), raising the question of how a single, constitutively expressed TF family can support distinct developmental processes. Here we use an integrative experimental and bioinformatic approach to discover the regulatory relationship between Zic TF binding and changing programs of gene transcription during postnatal CGN differentiation. RESULTS We first established a bioinformatic pipeline to integrate Zic ChIP-seq data from the developing mouse cerebellum with other genomic datasets from the same tissue. In newborn CGNs, Zic TF binding predominates at active enhancers that are co-bound by developmentally regulated TFs including Atoh1, whereas in mature CGNs, Zic TF binding consolidates toward promoters where it co-localizes with activity-regulated TFs. We then performed CUT&RUN-seq in differentiating CGNs to define both the time course of developmental shifts in Zic TF binding and their relationship to gene expression. Mapping Zic TF binding sites to genes using chromatin looping, we identified the set of Zic target genes that have altered expression in RNA-seq from Zic1 or Zic2 knockdown CGNs. CONCLUSIONS Our data show that Zic TFs are required for both induction and repression of distinct, developmentally regulated target genes through a mechanism that is largely independent of changes in Zic TF binding. We suggest that the differential collaboration of Zic TFs with other TF families underlies the shift in their biological functions across CGN development.
Collapse
Affiliation(s)
- Melyssa S Minto
- Program in Computational Biology and Bioinformatics, Duke University, Durham, NC, 27710, USA
- Omics, Epidemiology and Analytics Program, RTI International, Research Triangle Park, NC, 27709, USA
| | | | - Vijyendra Ramesh
- Department of Neurobiology, Duke University, Durham, NC, 27710, USA
| | - Anne E West
- Department of Neurobiology, Duke University, Durham, NC, 27710, USA.
| |
Collapse
|
5
|
Herrera E, Chédotal A, Mason C. Development of the Binocular Circuit. Annu Rev Neurosci 2024; 47:303-322. [PMID: 38635868 DOI: 10.1146/annurev-neuro-111020-093230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
Seeing in three dimensions is a major property of the visual system in mammals. The circuit underlying this property begins in the retina, from which retinal ganglion cells (RGCs) extend to the same or opposite side of the brain. RGC axons decussate to form the optic chiasm, then grow to targets in the thalamus and midbrain, where they synapse with neurons that project to the visual cortex. Here we review the cellular and molecular mechanisms of RGC axonal growth cone guidance across or away from the midline via receptors to cues in the midline environment. We present new views on the specification of ipsi- and contralateral RGC subpopulations and factors implementing their organization in the optic tract and termination in subregions of their targets. Lastly, we describe the functional and behavioral aspects of binocular vision, focusing on the mouse, and discuss recent discoveries in the evolution of the binocular circuit.
Collapse
Affiliation(s)
- Eloísa Herrera
- Instituto de Neurociencias (CSIC-UMH), Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Alicante, Spain;
| | - Alain Chédotal
- Université Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, Lyon, France
- Institut de Pathologie, Groupe Hospitalier Est, Hospices Civils de Lyon, Lyon, France
- Institut de la Vision, INSERM, Sorbonne Université, Paris, France;
| | - Carol Mason
- Departments of Pathology and Cell Biology, Neuroscience, and Ophthalmology, Zuckerman Institute, Columbia University, New York, NY, USA;
| |
Collapse
|
6
|
Malchow J, Eberlein J, Li W, Hogan BM, Okuda KS, Helker CSM. Neural progenitor-derived Apelin controls tip cell behavior and vascular patterning. SCIENCE ADVANCES 2024; 10:eadk1174. [PMID: 38968355 PMCID: PMC11225789 DOI: 10.1126/sciadv.adk1174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 05/31/2024] [Indexed: 07/07/2024]
Abstract
During angiogenesis, vascular tip cells guide nascent vascular sprouts to form a vascular network. Apelin, an agonist of the G protein-coupled receptor Aplnr, is enriched in vascular tip cells, and it is hypothesized that vascular-derived Apelin regulates sprouting angiogenesis. We identify an apelin-expressing neural progenitor cell population in the dorsal neural tube. Vascular tip cells exhibit directed elongation and migration toward and along the apelin-expressing neural progenitor cells. Notably, restoration of neural but not vascular apelin expression in apelin mutants remedies the angiogenic defects of mutants. By functional analyses, we show the requirement of Apelin signaling for tip cell behaviors, like filopodia formation and cell elongation. Through genetic interaction studies and analysis of transgenic activity reporters, we identify Apelin signaling as a modulator of phosphoinositide 3-kinase and extracellular signal-regulated kinase signaling in tip cells in vivo. Our results suggest a previously unidentified neurovascular cross-talk mediated by Apelin signaling that is important for tip cell function during sprouting angiogenesis.
Collapse
Affiliation(s)
- Julian Malchow
- Faculty of Biology, Cell Signaling and Dynamics, Philipps-University of Marburg, Marburg, Germany
| | - Jean Eberlein
- Faculty of Biology, Cell Signaling and Dynamics, Philipps-University of Marburg, Marburg, Germany
| | - Wei Li
- Faculty of Biology, Cell Signaling and Dynamics, Philipps-University of Marburg, Marburg, Germany
| | - Benjamin M. Hogan
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria 3000, Australia
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Kazuhide S. Okuda
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria 3000, Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, Victoria, Australia
- Centre for Cardiovascular Biology and Disease Research, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Christian S. M. Helker
- Faculty of Biology, Cell Signaling and Dynamics, Philipps-University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, Marburg, Germany
| |
Collapse
|
7
|
Ventriglia S, Kalcheim C. From neural tube to spinal cord: The dynamic journey of the dorsal neuroepithelium. Dev Biol 2024; 511:26-38. [PMID: 38580174 DOI: 10.1016/j.ydbio.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/21/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024]
Abstract
In a developing embryo, formation of tissues and organs is remarkably precise in both time and space. Through cell-cell interactions, neighboring progenitors coordinate their activities, sequentially generating distinct types of cells. At present, we only have limited knowledge, rather than a systematic understanding, of the underlying logic and mechanisms responsible for cell fate transitions. The formation of the dorsal aspect of the spinal cord is an outstanding model to tackle these dynamics, as it first generates the peripheral nervous system and is later responsible for transmitting sensory information from the periphery to the brain and for coordinating local reflexes. This is reflected first by the ontogeny of neural crest cells, progenitors of the peripheral nervous system, followed by formation of the definitive roof plate of the central nervous system and specification of adjacent interneurons, then a transformation of roof plate into dorsal radial glia and ependyma lining the forming central canal. How do these peripheral and central neural branches segregate from common progenitors? How are dorsal radial glia established concomitant with transformation of the neural tube lumen into a central canal? How do the dorsal radial glia influence neighboring cells? This is only a partial list of questions whose clarification requires the implementation of experimental paradigms in which precise control of timing is crucial. Here, we outline some available answers and still open issues, while highlighting the contributions of avian models and their potential to address mechanisms of neural patterning and function.
Collapse
Affiliation(s)
- Susanna Ventriglia
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, 9112102, P.O.Box 12272, Israel.
| | - Chaya Kalcheim
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, 9112102, P.O.Box 12272, Israel.
| |
Collapse
|
8
|
Safronov BV, Szucs P. Novel aspects of signal processing in lamina I. Neuropharmacology 2024; 247:109858. [PMID: 38286189 DOI: 10.1016/j.neuropharm.2024.109858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/12/2024] [Accepted: 01/25/2024] [Indexed: 01/31/2024]
Abstract
The most superficial layer of the spinal dorsal horn, lamina I, is a key element of the nociceptive processing system. It contains different types of projection neurons (PNs) and local-circuit neurons (LCNs) whose functional roles in the signal processing are poorly understood. This article reviews recent progress in elucidating novel anatomical features and physiological properties of lamina I PNs and LCNs revealed by whole-cell recordings in ex vivo spinal cord. This article is part of the Special Issue on "Ukrainian Neuroscience".
Collapse
Affiliation(s)
- Boris V Safronov
- Neuronal Networks Group, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
| | - Peter Szucs
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; HUN-REN-DE Neuroscience Research Group, Debrecen, Hungary
| |
Collapse
|
9
|
Minto M, Sotelo-Fonseca JE, Ramesh V, West AE. Genome binding properties of Zic transcription factors underlie their changing functions during neuronal maturation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.04.574185. [PMID: 38260638 PMCID: PMC10802290 DOI: 10.1101/2024.01.04.574185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Background The Zic family of transcription factors (TFs) promote both proliferation and maturation of cerebellar granule neurons (CGNs), raising the question of how a single, constitutively expressed TF family can support distinct developmental processes. Here we use an integrative experimental and bioinformatic approach to discover the regulatory relationship between Zic TF binding and changing programs of gene transcription during CGN differentiation. Results We first established a bioinformatic pipeline to integrate Zic ChIP-seq data from the developing mouse cerebellum with other genomic datasets from the same tissue. In newborn CGNs, Zic TF binding predominates at active enhancers that are co-bound by developmentally-regulated TFs including Atoh1, whereas in mature CGNs, Zic TF binding consolidates toward promoters where it co-localizes with activity-regulated TFs. We then performed CUT&RUN-seq in differentiating CGNs to define both the time course of developmental shifts in Zic TF binding and their relationship to gene expression. Mapping Zic TF binding sites to genes using chromatin looping, we identified the set of Zic target genes that have altered expression in RNA-seq from Zic1 or Zic2 knockdown CGNs. Conclusion Our data show that Zic TFs are required for both induction and repression of distinct, developmentally regulated target genes through a mechanism that is largely independent of changes in Zic TF binding. We suggest that the differential collaboration of Zic TFs with other TF families underlies the shift in their biological functions across CGN development.
Collapse
Affiliation(s)
- Melyssa Minto
- Duke University, Program in Computational Biology and Bioinformatics, Durham, NC 27710
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, NC 27709
| | | | | | - Anne E. West
- Duke University, Department of Neurobiology, Durham, NC 27710
| |
Collapse
|
10
|
Fries M, Brown TW, Jolicoeur C, Boulan B, Boudreau-Pinsonneault C, Javed A, Abram P, Cayouette M. Pou3f1 orchestrates a gene regulatory network controlling contralateral retinogeniculate projections. Cell Rep 2023; 42:112985. [PMID: 37590135 DOI: 10.1016/j.celrep.2023.112985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 05/26/2023] [Accepted: 08/01/2023] [Indexed: 08/19/2023] Open
Abstract
The balance of contralateral and ipsilateral retinogeniculate projections is critical for binocular vision, but the transcriptional programs regulating this process remain ill defined. Here we show that the Pou class homeobox protein POU3F1 is expressed in nascent mouse contralateral retinal ganglion cells (cRGCs) but not ipsilateral RGCs (iRGCs). Upon Pou3f1 inactivation, the proportion of cRGCs is reduced in favor of iRGCs, leading to abnormal projection ratios at the optic chiasm. Conversely, misexpression of Pou3f1 in progenitors increases the production of cRGCs. Using CUT&RUN and RNA sequencing in gain- and loss-of-function assays, we demonstrate that POU3F1 regulates expression of several key members of the cRGC gene regulatory network. Finally, we report that POU3F1 is sufficient to induce RGC-like cell production, even in late-stage retinal progenitors of Atoh7 knockout mice. This work uncovers POU3F1 as a regulator of the cRGC transcriptional program, opening possibilities for optic nerve regenerative therapies.
Collapse
Affiliation(s)
- Michel Fries
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; Molecular Biology Program, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Thomas W Brown
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 1A1, Canada
| | - Christine Jolicoeur
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada
| | - Benoit Boulan
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada
| | - Camille Boudreau-Pinsonneault
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 1A1, Canada
| | - Awais Javed
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; Molecular Biology Program, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Pénélope Abram
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada
| | - Michel Cayouette
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; Molecular Biology Program, Université de Montréal, Montreal, QC H3C 3J7, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 1A1, Canada; Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada.
| |
Collapse
|
11
|
Zhai J, Xu Y, Wan H, Yan R, Guo J, Skory R, Yan L, Wu X, Sun F, Chen G, Zhao W, Yu K, Li W, Guo F, Plachta N, Wang H. Neurulation of the cynomolgus monkey embryo achieved from 3D blastocyst culture. Cell 2023; 186:2078-2091.e18. [PMID: 37172562 DOI: 10.1016/j.cell.2023.04.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 12/15/2022] [Accepted: 04/12/2023] [Indexed: 05/15/2023]
Abstract
Neural tube (NT) defects arise from abnormal neurulation and result in the most common birth defects worldwide. Yet, mechanisms of primate neurulation remain largely unknown due to prohibitions on human embryo research and limitations of available model systems. Here, we establish a three-dimensional (3D) prolonged in vitro culture (pIVC) system supporting cynomolgus monkey embryo development from 7 to 25 days post-fertilization. Through single-cell multi-omics analyses, we demonstrate that pIVC embryos form three germ layers, including primordial germ cells, and establish proper DNA methylation and chromatin accessibility through advanced gastrulation stages. In addition, pIVC embryo immunofluorescence confirms neural crest formation, NT closure, and neural progenitor regionalization. Finally, we demonstrate that the transcriptional profiles and morphogenetics of pIVC embryos resemble key features of similarly staged in vivo cynomolgus and human embryos. This work therefore describes a system to study non-human primate embryogenesis through advanced gastrulation and early neurulation.
Collapse
Affiliation(s)
- Jinglei Zhai
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yanhong Xu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Haifeng Wan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Rui Yan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Jing Guo
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Robin Skory
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Long Yan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Xulun Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Fengyuan Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Gang Chen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Wentao Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Kunyuan Yu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China.
| | - Fan Guo
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China.
| | - Nicolas Plachta
- Department of Cell and Developmental Biology, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Hongmei Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China.
| |
Collapse
|
12
|
Klein R, Wilkinson D, Herrera E. Editorial - Friedrich Bonhoeffer (1932-2021). Neuroscience 2023; 508:1-2. [PMID: 36427670 DOI: 10.1016/j.neuroscience.2022.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Rüdiger Klein
- Max-Planck Institute for Biological Intelligence, Am Klopferspitz 18, 82152 Martinsried, Germany
| | | | - Eloisa Herrera
- Instituto de Neurociencias (CSIC-UMH), Av. Ramón y Cajal s/n, San Juan de Alicante, Alicante, Spain.
| |
Collapse
|
13
|
Fernández‐Nogales M, López‐Cascales MT, Murcia‐Belmonte V, Escalante A, Fernández‐Albert J, Muñoz‐Viana R, Barco A, Herrera E. Multiomic Analysis of Neurons with Divergent Projection Patterns Identifies Novel Regulators of Axon Pathfinding. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200615. [PMID: 35988153 PMCID: PMC9561852 DOI: 10.1002/advs.202200615] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/30/2022] [Indexed: 06/15/2023]
Abstract
Axon pathfinding is a key step in neural circuits formation. However, the transcriptional mechanisms regulating its progression remain poorly understood. The binary decision of crossing or avoiding the midline taken by some neuronal axons during development represents a robust model to investigate the mechanisms that control the selection of axonal trajectories. Here, to identify novel regulators of axon guidance, this work compares the transcriptome and chromatin occupancy profiles of two neuronal subpopulations, ipsilateral (iRGC) and contralateral retinal ganglion cells (cRGC), with similar functions but divergent axon trajectories. These analyses retrieved a number of genes encoding for proteins not previously implicated in axon pathfinding. In vivo functional experiments confirm the implication of some of these candidates in axonal navigation. Among the candidate genes, γ-synuclein is identified as essential for inducing midline crossing. Footprint and luciferase assays demonstrate that this small-sized protein is regulated by the transcription factor (TF) Pou4f1 in cRGCs. It is also shown that Lhx2/9 are specifically expressed in iRGCs and control a program that partially overlaps with that regulated by Zic2, previously described as essential for iRGC specification. Overall, the analyses identify dozens of new molecules potentially involved in axon guidance and reveal the regulatory logic behind the selection of axonal trajectories.
Collapse
Affiliation(s)
- Marta Fernández‐Nogales
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| | - Maria Teresa López‐Cascales
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| | - Verónica Murcia‐Belmonte
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| | - Augusto Escalante
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| | - Jordi Fernández‐Albert
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| | - Rafael Muñoz‐Viana
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| | - Angel Barco
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| | - Eloísa Herrera
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| |
Collapse
|
14
|
Herrera E, Escalante A. Transcriptional Control of Axon Guidance at Midline Structures. Front Cell Dev Biol 2022; 10:840005. [PMID: 35265625 PMCID: PMC8900194 DOI: 10.3389/fcell.2022.840005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
The development of the nervous system is a time-ordered and multi-stepped process that includes neurogenesis and neuronal specification, axonal navigation, and circuits assembly. During axonal navigation, the growth cone, a dynamic structure located at the tip of the axon, senses environmental signals that guide axons towards their final targets. The expression of a specific repertoire of receptors on the cell surface of the growth cone together with the activation of a set of intracellular transducing molecules, outlines the response of each axon to specific guidance cues. This collection of axon guidance molecules is defined by the transcriptome of the cell which, in turn, depends on transcriptional and epigenetic regulators that modify the structure and DNA accessibility to determine what genes will be expressed to elicit specific axonal behaviors. Studies focused on understanding how axons navigate intermediate targets, such as the floor plate of vertebrates or the mammalian optic chiasm, have largely contributed to our knowledge of how neurons wire together during development. In fact, investigations on axon navigation at these midline structures led to the identification of many of the currently known families of proteins that act as guidance cues and their corresponding receptors. Although the transcription factors and the regulatory mechanisms that control the expression of these molecules are not well understood, important advances have been made in recent years in this regard. Here we provide an updated overview on the current knowledge about the transcriptional control of axon guidance and the selection of trajectories at midline structures.
Collapse
|
15
|
Hirsch D, Kohl A, Wang Y, Sela-Donenfeld D. Axonal Projection Patterns of the Dorsal Interneuron Populations in the Embryonic Hindbrain. Front Neuroanat 2022; 15:793161. [PMID: 35002640 PMCID: PMC8738170 DOI: 10.3389/fnana.2021.793161] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Unraveling the inner workings of neural circuits entails understanding the cellular origin and axonal pathfinding of various neuronal groups during development. In the embryonic hindbrain, different subtypes of dorsal interneurons (dINs) evolve along the dorsal-ventral (DV) axis of rhombomeres and are imperative for the assembly of central brainstem circuits. dINs are divided into two classes, class A and class B, each containing four neuronal subgroups (dA1-4 and dB1-4) that are born in well-defined DV positions. While all interneurons belonging to class A express the transcription factor Olig3 and become excitatory, all class B interneurons express the transcription factor Lbx1 but are diverse in their excitatory or inhibitory fate. Moreover, within every class, each interneuron subtype displays its own specification genes and axonal projection patterns which are required to govern the stage-by-stage assembly of their connectivity toward their target sites. Remarkably, despite the similar genetic landmark of each dINs subgroup along the anterior-posterior (AP) axis of the hindbrain, genetic fate maps of some dA/dB neuronal subtypes uncovered their contribution to different nuclei centers in relation to their rhombomeric origin. Thus, DV and AP positional information has to be orchestrated in each dA/dB subpopulation to form distinct neuronal circuits in the hindbrain. Over the span of several decades, different axonal routes have been well-documented to dynamically emerge and grow throughout the hindbrain DV and AP positions. Yet, the genetic link between these distinct axonal bundles and their neuronal origin is not fully clear. In this study, we reviewed the available data regarding the association between the specification of early-born dorsal interneuron subpopulations in the hindbrain and their axonal circuitry development and fate, as well as the present existing knowledge on molecular effectors underlying the process of axonal growth.
Collapse
Affiliation(s)
- Dana Hirsch
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel.,Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Ayelet Kohl
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Yuan Wang
- Department of Biomedical Sciences, Program in Neuroscience, College of Medicine, Florida State University, Tallahassee, FL, United States
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
16
|
Sousa E, Flames N. Transcriptional regulation of neuronal identity. Eur J Neurosci 2021; 55:645-660. [PMID: 34862697 PMCID: PMC9306894 DOI: 10.1111/ejn.15551] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 11/29/2022]
Abstract
Neuronal diversity is an intrinsic feature of the nervous system. Transcription factors (TFs) are key regulators in the establishment of different neuronal identities; how are the actions of different TFs coordinated to orchestrate this diversity? Are there common features shared among the different neuron types of an organism or even among different animal groups? In this review, we provide a brief overview on common traits emerging on the transcriptional regulation of neuron type diversification with a special focus on the comparison between mouse and Caenorhabditis elegans model systems. In the first part, we describe general concepts on neuronal identity and transcriptional regulation of gene expression. In the second part of the review, TFs are classified in different categories according to their key roles at specific steps along the protracted process of neuronal specification and differentiation. The same TF categories can be identified both in mammals and nematodes. Importantly, TFs are very pleiotropic: Depending on the neuron type or the time in development, the same TF can fulfil functions belonging to different categories. Finally, we describe the key role of transcriptional repression at all steps controlling neuronal diversity and propose that acquisition of neuronal identities could be considered a metastable process.
Collapse
Affiliation(s)
- Erick Sousa
- Developmental Neurobiology Unit, Instituto de Biomedicina de Valencia IBV-CSIC, Valencia, Spain
| | - Nuria Flames
- Developmental Neurobiology Unit, Instituto de Biomedicina de Valencia IBV-CSIC, Valencia, Spain
| |
Collapse
|
17
|
Sánchez-Huertas C, Herrera E. With the Permission of Microtubules: An Updated Overview on Microtubule Function During Axon Pathfinding. Front Mol Neurosci 2021; 14:759404. [PMID: 34924953 PMCID: PMC8675249 DOI: 10.3389/fnmol.2021.759404] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/01/2021] [Indexed: 01/27/2023] Open
Abstract
During the establishment of neural circuitry axons often need to cover long distances to reach remote targets. The stereotyped navigation of these axons defines the connectivity between brain regions and cellular subtypes. This chemotrophic guidance process mostly relies on the spatio-temporal expression patterns of extracellular proteins and the selective expression of their receptors in projection neurons. Axon guidance is stimulated by guidance proteins and implemented by neuronal traction forces at the growth cones, which engage local cytoskeleton regulators and cell adhesion proteins. Different layers of guidance signaling regulation, such as the cleavage and processing of receptors, the expression of co-receptors and a wide variety of intracellular cascades downstream of receptors activation, have been progressively unveiled. Also, in the last decades, the regulation of microtubule (MT) assembly, stability and interactions with the submembranous actin network in the growth cone have emerged as crucial effector mechanisms in axon pathfinding. In this review, we will delve into the intracellular signaling cascades downstream of guidance receptors that converge on the MT cytoskeleton of the growing axon. In particular, we will focus on the microtubule-associated proteins (MAPs) network responsible of MT dynamics in the axon and growth cone. Complementarily, we will discuss new evidences that connect defects in MT scaffold proteins, MAPs or MT-based motors and axon misrouting during brain development.
Collapse
Affiliation(s)
- Carlos Sánchez-Huertas
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Alicante, Spain
| | | |
Collapse
|
18
|
Cárdenas A, Borrell V. A protocol for in ovo electroporation of chicken and snake embryos to study forebrain development. STAR Protoc 2021; 2:100692. [PMID: 34382018 PMCID: PMC8339381 DOI: 10.1016/j.xpro.2021.100692] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
In vivo electroporation has become a key technique to study genetic mechanisms of brain development. However, electroporation of the embryonic pallium in oviparous species, interesting for evolutionary studies but distinct from in utero electroporation, is quite infrequent. Here, we detail the in ovo electroporation of the developing pallium in chick and snake embryos. This protocol allows gene manipulation through introducing exogenous DNA into brain progenitor cells and can be adapted to any type of gene manipulation of the embryonic telencephalon. For complete information on the use and execution of this protocol, please refer to Cárdenas et al. (2018). In ovo electroporation of dorsal telencephalon in chick and snake embryos Maximal viability of the embryos makes the protocol highly efficient The simplicity of the procedure makes it accessible to non-expert researchers Adaptable to any type of gene manipulation of the embryonic telencephalon
Collapse
Affiliation(s)
- Adrián Cárdenas
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, 03550 Alacant, Spain
| | - Víctor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, 03550 Alacant, Spain
| |
Collapse
|
19
|
Wurmser M, Muppavarapu M, Tait CM, Laumonnerie C, González-Castrillón LM, Wilson SI. Robo2 Receptor Gates the Anatomical Divergence of Neurons Derived From a Common Precursor Origin. Front Cell Dev Biol 2021; 9:668175. [PMID: 34249921 PMCID: PMC8263054 DOI: 10.3389/fcell.2021.668175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/03/2021] [Indexed: 12/25/2022] Open
Abstract
Sensory information relayed to the brain is dependent on complex, yet precise spatial organization of neurons. This anatomical complexity is generated during development from a surprisingly small number of neural stem cell domains. This raises the question of how neurons derived from a common precursor domain respond uniquely to their environment to elaborate correct spatial organization and connectivity. We addressed this question by exploiting genetically labeled mouse embryonic dorsal interneuron 1 (dI1) neurons that are derived from a common precursor domain and give rise to spinal projection neurons with distinct organization of cell bodies with axons projecting either commissurally (dI1c) or ipsilaterally (dI1i). In this study, we examined how the guidance receptor, Robo2, which is a canonical Robo receptor, influenced dI1 guidance during embryonic development. Robo2 was enriched in embryonic dI1i neurons, and loss of Robo2 resulted in misguidance of dI1i axons, whereas dI1c axons remained unperturbed within the mantle zone and ventral commissure. Further, Robo2 profoundly influenced dI1 cell body migration, a feature that was partly dependent on Slit2 signaling. These data suggest that dI1 neurons are dependent on Robo2 for their organization. This work integrated with the field support of a model whereby canonical Robo2 vs. non-canonical Robo3 receptor expression facilitates projection neurons derived from a common precursor domain to read out the tissue environment uniquely giving rise to correct anatomical organization.
Collapse
Affiliation(s)
- Maud Wurmser
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | | | | | | | | | - Sara Ivy Wilson
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| |
Collapse
|
20
|
Morenilla-Palao C, López-Cascales MT, López-Atalaya JP, Baeza D, Calvo-Díaz L, Barco A, Herrera E. A Zic2-regulated switch in a noncanonical Wnt/βcatenin pathway is essential for the formation of bilateral circuits. SCIENCE ADVANCES 2020; 6:6/46/eaaz8797. [PMID: 33188033 PMCID: PMC7673756 DOI: 10.1126/sciadv.aaz8797] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 09/30/2020] [Indexed: 05/06/2023]
Abstract
The Wnt pathway is involved in a wide array of biological processes during development and is deregulated in many pathological scenarios. In neurons, Wnt proteins promote both axon extension and repulsion, but the molecular mechanisms underlying these opposing axonal responses are unknown. Here, we show that Wnt5a is expressed at the optic chiasm midline and promotes the crossing of retinal axons by triggering an alternative Wnt pathway that depends on the accumulation of βcatenin but does not activate the canonical pathway. In ipsilateral neurons, the transcription factor Zic2 switches this alternative Wnt pathway by regulating the expression of a set of Wnt receptors and intracellular proteins. In combination with this alternative Wnt pathway, the asymmetric activation of EphB1 receptors at the midline phosphorylates βcatenin and elicits a repulsive response. This alternative Wnt pathway and its Zic2-triggered switch may operate in other contexts that require a two-way response to Wnt ligands.
Collapse
Affiliation(s)
- Cruz Morenilla-Palao
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Campus San Juan, Av. Ramón y Cajal s/n, Alicante 03550, Spain
| | - María Teresa López-Cascales
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Campus San Juan, Av. Ramón y Cajal s/n, Alicante 03550, Spain
| | - José P López-Atalaya
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Campus San Juan, Av. Ramón y Cajal s/n, Alicante 03550, Spain
| | - Diana Baeza
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Campus San Juan, Av. Ramón y Cajal s/n, Alicante 03550, Spain
| | - Luís Calvo-Díaz
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Campus San Juan, Av. Ramón y Cajal s/n, Alicante 03550, Spain
| | - Angel Barco
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Campus San Juan, Av. Ramón y Cajal s/n, Alicante 03550, Spain
| | - Eloísa Herrera
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Campus San Juan, Av. Ramón y Cajal s/n, Alicante 03550, Spain.
| |
Collapse
|
21
|
Wei-Hua W, Ning Z, Qian C, Dao-Wen J. ZIC2 promotes cancer stem cell traits via up-regulating OCT4 expression in lung adenocarcinoma cells. J Cancer 2020; 11:6070-6080. [PMID: 32922547 PMCID: PMC7477430 DOI: 10.7150/jca.44367] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023] Open
Abstract
Background: Accumulating evidence has revealed the importance of cancer stem cells (CSCs) in self-renewal and chemoresistance. Previous studies reported high expression of ZIC2 was closely associated with tumorigenesis and CSC traits. However, the role of ZIC2 as a crucial factor for regulating CSC properties in lung adenocarcinoma (LAC) remains elusive. Methods: RT-PCR and WB assay were employed to assess ZIC2 expression in 20 LAC tumor tissues and the matched non-cancerous tissues. The role of ZIC2 in LAC CSC were analyzed by evaluation of CSC-related markers expression and spheroid formation in vitro. Cisplatin and paclitaxel resistance capacities were evaluated by CCK8 assay, colony formation assay, and flow cytometry analysis. Subcutaneous NOD/SCID mice models were generated to assess in vivo CSC features. Results: High expression of ZIC2 was found in LAC tumor tissues and indicated a poor overall survival in LAC patients. ZIC2 upregulated an array of CSCs-related genes, including EpCAM, OCT4, SOX2, NANOG, C-Myc and Bmi-1. Knockdown of ZIC2 inhibited sphere-forming capacity and decreased cisplatin and paclitaxel resistance. However, overexpression of ZIC2 achieved opposite effects. Mechanically, ZIC2 acts upstream of OCT4 to promote its expression, resulting in enhancement of CSC traits in LAC. Conclusion: Our results demonstrated that ZIC2 was crucial for promoting CSC traits in LAC cells, and served as a potential biomarker for predicting prognosis. The ZIC2-OCT4 network will facilitate the evaluation of the potential therapeutic efficacy of chemotherapy and predict patient sensitivity to treatment.
Collapse
Affiliation(s)
- Wang Wei-Hua
- Department of thoracic surgery, Minhang Hospital, Fudan University, Shanghai 201100, China
| | - Zhou Ning
- Department of thoracic surgery, Minhang Hospital, Fudan University, Shanghai 201100, China
| | - Chen Qian
- Department of general surgery, Minhang Hospital, Fudan University, Shanghai 201100, China
| | - Jiang Dao-Wen
- Department of thoracic surgery, Minhang Hospital, Fudan University, Shanghai 201100, China
| |
Collapse
|
22
|
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
|
23
|
Roig-Puiggros S, Vigouroux RJ, Beckman D, Bocai NI, Chiou B, Davimes J, Gomez G, Grassi S, Hoque A, Karikari TK, Kiffer F, Lopez M, Lunghi G, Mazengenya P, Meier S, Olguín-Albuerne M, Oliveira MM, Paraíso-Luna J, Pradhan J, Radiske A, Ramos-Hryb AB, Ribeiro MC, Schellino R, Selles MC, Singh S, Theotokis P, Chédotal A. Construction and reconstruction of brain circuits: normal and pathological axon guidance. J Neurochem 2019; 153:10-32. [PMID: 31630412 DOI: 10.1111/jnc.14900] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/14/2019] [Accepted: 10/17/2019] [Indexed: 02/06/2023]
Abstract
Perception of our environment entirely depends on the close interaction between the central and peripheral nervous system. In order to communicate each other, both systems must develop in parallel and in coordination. During development, axonal projections from the CNS as well as the PNS must extend over large distances to reach their appropriate target cells. To do so, they read and follow a series of axon guidance molecules. Interestingly, while these molecules play critical roles in guiding developing axons, they have also been shown to be critical in other major neurodevelopmental processes, such as the migration of cortical progenitors. Currently, a major hurdle for brain repair after injury or neurodegeneration is the absence of axonal regeneration in the mammalian CNS. By contrasts, PNS axons can regenerate. Many hypotheses have been put forward to explain this paradox but recent studies suggest that hacking neurodevelopmental mechanisms may be the key to promote CNS regeneration. Here we provide a seminar report written by trainees attending the second Flagship school held in Alpbach, Austria in September 2018 organized by the International Society for Neurochemistry (ISN) together with the Journal of Neurochemistry (JCN). This advanced school has brought together leaders in the fields of neurodevelopment and regeneration in order to discuss major keystones and future challenges in these respective fields.
Collapse
Affiliation(s)
| | - Robin J Vigouroux
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Danielle Beckman
- California National Primate Research Center, UC Davis, Davis, California, USA
| | - Nadia I Bocai
- Laboratory of Amyloidosis and Neurodegeneration, Fundación Instituto Leloir, Buenos Aires, Argentina.,Instituto de Investigaciones Bioquímicas de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Brian Chiou
- Department of Pediatrics, University of California - San Francisco, San Francisco, California, USA
| | - Joshua Davimes
- Faculty of Health Sciences School of Anatomical Sciences, University of the Witwatersrand, Parktown Johannesburg, South Africa
| | - Gimena Gomez
- Laboratorio de Parkinson Experimental, Instituto de Investigaciones Farmacológicas (ININFA-CONICET-UBA), Ciudad Autónoma de Buenos Aires, Argentina
| | - Sara Grassi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Ashfaqul Hoque
- Metabolic Signalling Laboratory, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,School of Life Sciences, University of Warwick, Coventry, UK.,Midlands Integrative Biosciences Training Partnership, University of Warwick, Coventry, UK
| | - Frederico Kiffer
- Division of Radiation Health, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Mary Lopez
- Institute for Stroke and Dementia Research, LMU Munich, Munich, Germany
| | - Giulia Lunghi
- Department of Medical Biotechnology and Translational Medicin, University of Milano, Segrate, Italy
| | - Pedzisai Mazengenya
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sonja Meier
- Queensland Brain Institute, The University of Queensland, St Lucia, Queensland, Australia
| | - Mauricio Olguín-Albuerne
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Mauricio M Oliveira
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juan Paraíso-Luna
- Ramón y Cajal Institute of Health Research (IRYCIS), Department of Biochemistry and Molecular Biology and University Research Institute in Neurochemistry (IUIN), Complutense University, Madrid, Spain.,Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jonu Pradhan
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Andressa Radiske
- Memory Research Laboratory, Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Ana Belén Ramos-Hryb
- Instituto de Biología y Medicina Experimental (IBYME)-CONICET, Buenos Aires, Argentina.,Grupo de Neurociencia de Sistemas, Instituto de Fisiología y Biofísica (IFIBIO) Bernardo Houssay, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Mayara C Ribeiro
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, New York, USA
| | - Roberta Schellino
- Neuroscience Department "Rita Levi-Montalcini" and Neuroscience Institute Cavalieri Ottolenghi, University of Torino, Torino, Italy
| | - Maria Clara Selles
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Shripriya Singh
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Paschalis Theotokis
- Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Thessaloniki, Macedonia, Greece
| | - Alain Chédotal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| |
Collapse
|
24
|
Identification of Spinal Neurons Contributing to the Dorsal Column Projection Mediating Fine Touch and Corrective Motor Movements. Neuron 2019; 104:749-764.e6. [PMID: 31586516 DOI: 10.1016/j.neuron.2019.08.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 07/22/2019] [Accepted: 08/16/2019] [Indexed: 12/22/2022]
Abstract
Tactile stimuli are integrated and processed by neuronal circuits in the deep dorsal horn of the spinal cord. Several spinal interneuron populations have been implicated in tactile information processing. However, dorsal horn projection neurons that contribute to the postsynaptic dorsal column (PSDC) pathway transmitting tactile information to the brain are poorly characterized. Here, we show that spinal neurons marked by the expression of Zic2creER mediate light touch sensitivity and textural discrimination. A subset of Zic2creER neurons are PSDC neurons that project to brainstem dorsal column nuclei, and chemogenetic activation of Zic2 PSDC neurons increases sensitivity to light touch stimuli. Zic2 neurons receive direct input from the cortex and brainstem motor nuclei and are required for corrective motor movements. These results suggest that Zic2 neurons integrate sensory input from cutaneous afferents with descending signals from the brain to promote corrective movements and transmit processed touch information back to the brain. VIDEO ABSTRACT.
Collapse
|
25
|
Comer JD, Alvarez S, Butler SJ, Kaltschmidt JA. Commissural axon guidance in the developing spinal cord: from Cajal to the present day. Neural Dev 2019; 14:9. [PMID: 31514748 PMCID: PMC6739980 DOI: 10.1186/s13064-019-0133-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 08/23/2019] [Indexed: 12/11/2022] Open
Abstract
During neuronal development, the formation of neural circuits requires developing axons to traverse a diverse cellular and molecular environment to establish synaptic contacts with the appropriate postsynaptic partners. Essential to this process is the ability of developing axons to navigate guidance molecules presented by specialized populations of cells. These cells partition the distance traveled by growing axons into shorter intervals by serving as intermediate targets, orchestrating the arrival and departure of axons by providing attractive and repulsive guidance cues. The floor plate in the central nervous system (CNS) is a critical intermediate target during neuronal development, required for the extension of commissural axons across the ventral midline. In this review, we begin by giving a historical overview of the ventral commissure and the evolutionary purpose of decussation. We then review the axon guidance studies that have revealed a diverse assortment of midline guidance cues, as well as genetic and molecular regulatory mechanisms required for coordinating the commissural axon response to these cues. Finally, we examine the contribution of dysfunctional axon guidance to neurological diseases.
Collapse
Affiliation(s)
- J 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
| | - S Alvarez
- Department of Neurobiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.,Molecular Biology Interdepartmental Graduate Program, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - S J Butler
- Department of Neurobiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - J A Kaltschmidt
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
26
|
Friocourt F, Kozulin P, Belle M, Suárez R, Di‐Poï N, Richards LJ, Giacobini P, Chédotal A. Shared and differential features of Robo3 expression pattern in amniotes. J Comp Neurol 2019; 527:2009-2029. [DOI: 10.1002/cne.24648] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 12/19/2018] [Accepted: 01/21/2019] [Indexed: 12/11/2022]
Affiliation(s)
| | - Peter Kozulin
- The Queensland Brain Institute The University of Queensland Brisbane Queensland Australia
| | - Morgane Belle
- Sorbonne Université, INSERM, CNRS Institut de la Vision Paris France
| | - Rodrigo Suárez
- The Queensland Brain Institute The University of Queensland Brisbane Queensland Australia
| | - Nicolas Di‐Poï
- Research Program in Developmental Biology, Institute of Biotechnology University of Helsinki Helsinki Finland
| | - Linda J. Richards
- The Queensland Brain Institute The University of Queensland Brisbane Queensland Australia
- The School of Biomedical Sciences The University of Queensland Brisbane Queensland Australia
| | - Paolo Giacobini
- University of Lille, UMR‐S 1172, Centre de Recherche Jean‐Pierre AUBERT Lille France
- Laboratory of Development and Plasticity of the Neuroendocrine Brain INSERM, UMR‐S 1172 Lille France
- FHU 1,000 Days for Health School of Medicine Lille France
| | - Alain Chédotal
- Sorbonne Université, INSERM, CNRS Institut de la Vision Paris France
| |
Collapse
|
27
|
Herrera E, Agudo-Barriuso M, Murcia-Belmonte V. Cranial Pair II: The Optic Nerves. Anat Rec (Hoboken) 2018; 302:428-445. [DOI: 10.1002/ar.23922] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/19/2017] [Accepted: 05/14/2017] [Indexed: 11/12/2022]
Affiliation(s)
- Eloísa Herrera
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH); Av. Santiago Ramón y Cajal, s/n., 03550 Sant Joan d'Alacant Alicante Spain
| | - Marta Agudo-Barriuso
- Departamento de Oftalmología, Facultad de Medicina; Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIB-Arrixaca); Murcia Spain
| | - Verónica Murcia-Belmonte
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH); Av. Santiago Ramón y Cajal, s/n., 03550 Sant Joan d'Alacant Alicante Spain
| |
Collapse
|
28
|
Cárdenas A, Villalba A, de Juan Romero C, Picó E, Kyrousi C, Tzika AC, Tessier-Lavigne M, Ma L, Drukker M, Cappello S, Borrell V. Evolution of Cortical Neurogenesis in Amniotes Controlled by Robo Signaling Levels. Cell 2018; 174:590-606.e21. [PMID: 29961574 PMCID: PMC6063992 DOI: 10.1016/j.cell.2018.06.007] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 02/24/2018] [Accepted: 06/01/2018] [Indexed: 11/29/2022]
Abstract
Cerebral cortex size differs dramatically between reptiles, birds, and mammals, owing to developmental differences in neuron production. In mammals, signaling pathways regulating neurogenesis have been identified, but genetic differences behind their evolution across amniotes remain unknown. We show that direct neurogenesis from radial glia cells, with limited neuron production, dominates the avian, reptilian, and mammalian paleocortex, whereas in the evolutionarily recent mammalian neocortex, most neurogenesis is indirect via basal progenitors. Gain- and loss-of-function experiments in mouse, chick, and snake embryos and in human cerebral organoids demonstrate that high Slit/Robo and low Dll1 signaling, via Jag1 and Jag2, are necessary and sufficient to drive direct neurogenesis. Attenuating Robo signaling and enhancing Dll1 in snakes and birds recapitulates the formation of basal progenitors and promotes indirect neurogenesis. Our study identifies modulation in activity levels of conserved signaling pathways as a primary mechanism driving the expansion and increased complexity of the mammalian neocortex during amniote evolution.
Collapse
Affiliation(s)
- Adrián Cárdenas
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, 03550 Alacant, Spain
| | - Ana Villalba
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, 03550 Alacant, Spain
| | - Camino de Juan Romero
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, 03550 Alacant, Spain
| | - Esther Picó
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, 03550 Alacant, Spain
| | - Christina Kyrousi
- Developmental Neurobiology, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Athanasia C Tzika
- Department Genetics and Evolution, University of Geneva, 1205 Geneva, Switzerland; SIB Swiss Institute of Bioinformatics, 1211 Geneva, Switzerland
| | | | - Le Ma
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sydney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Micha Drukker
- Institute of Stem Cell Research and the Induced Pluripotent Stem Cell Core Facility, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | - Silvia Cappello
- Developmental Neurobiology, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Víctor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, 03550 Alacant, Spain.
| |
Collapse
|
29
|
Kridsada K, Niu J, Haldipur P, Wang Z, Ding L, Li JJ, Lindgren AG, Herrera E, Thomas GM, Chizhikov VV, Millen KJ, Luo W. Roof Plate-Derived Radial Glial-like Cells Support Developmental Growth of Rapidly Adapting Mechanoreceptor Ascending Axons. Cell Rep 2018; 23:2928-2941. [PMID: 29874580 PMCID: PMC6174691 DOI: 10.1016/j.celrep.2018.05.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 03/02/2018] [Accepted: 05/03/2018] [Indexed: 11/28/2022] Open
Abstract
Spinal cord longitudinal axons comprise some of the longest axons in our body. However, mechanisms that drive this extra long-distance axonal growth are largely unclear. We found that ascending axons of rapidly adapting (RA) mechanoreceptors closely abut a previously undescribed population of roof plate-derived radial glial-like cells (RGLCs) in the spinal cord dorsal column, which form a network of processes enriched with growth-promoting factors. In dreher mutant mice that lack RGLCs, the lengths of ascending RA mechanoreceptor axon branches are specifically reduced, whereas their descending and collateral branches, and other dorsal column and sensory pathways, are largely unaffected. Because the number and intrinsic growth ability of RA mechanoreceptors are normal in dreher mice, our data suggest that RGLCs provide critical non-cell autonomous growth support for the ascending axons of RA mechanoreceptors. Together, our work identifies a developmental mechanism specifically required for long-range spinal cord longitudinal axons.
Collapse
Affiliation(s)
- Kim Kridsada
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jingwen Niu
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA; Shriners Hospital's Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Lewis Katz School of Medicine, Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Parthiv Haldipur
- Seattle Children's Hospital Research Institute, Center for Integrative Brain Research, Seattle, WA 98105, USA
| | - Zhiping Wang
- Department of Biostatistics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Long Ding
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jian J Li
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Eloisa Herrera
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), Campus San Juan, Av. Ramón y Cajal s/n, Alicante 03550, Spain
| | - Gareth M Thomas
- Shriners Hospital's Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Lewis Katz School of Medicine, Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA
| | - Victor V Chizhikov
- Department of Anatomy and Neurobiology, Health Science Center, University of Tennessee, Memphis, TN 38163, USA
| | - Kathleen J Millen
- Seattle Children's Hospital Research Institute, Center for Integrative Brain Research, Seattle, WA 98105, USA.
| | - Wenqin Luo
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
30
|
Kou CTJ, Kandpal RP. Differential Expression Patterns of Eph Receptors and Ephrin Ligands in Human Cancers. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7390104. [PMID: 29682554 PMCID: PMC5851329 DOI: 10.1155/2018/7390104] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 01/11/2018] [Accepted: 01/22/2018] [Indexed: 12/20/2022]
Abstract
Eph receptors constitute the largest family of receptor tyrosine kinases, which are activated by ephrin ligands that either are anchored to the membrane or contain a transmembrane domain. These molecules play important roles in the development of multicellular organisms, and the physiological functions of these receptor-ligand pairs have been extensively documented in axon guidance, neuronal development, vascular patterning, and inflammation during tissue injury. The recognition that aberrant regulation and expression of these molecules lead to alterations in proliferative, migratory, and invasive potential of a variety of human cancers has made them potential targets for cancer therapeutics. We present here the involvement of Eph receptors and ephrin ligands in lung carcinoma, breast carcinoma, prostate carcinoma, colorectal carcinoma, glioblastoma, and medulloblastoma. The aberrations in their abundances are described in the context of multiple signaling pathways, and differential expression is suggested as the mechanism underlying tumorigenesis.
Collapse
Affiliation(s)
- Chung-Ting Jimmy Kou
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Raj P. Kandpal
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
31
|
Abstract
The formation of the nervous system is a multistep process that yields a mature brain. Failure in any of the steps of this process may cause brain malfunction. In the early stages of embryonic development, neural progenitors quickly proliferate and then, at a specific moment, differentiate into neurons or glia. Once they become postmitotic neurons, they migrate to their final destinations and begin to extend their axons to connect with other neurons, sometimes located in quite distant regions, to establish different neural circuits. During the last decade, it has become evident that Zic genes, in addition to playing important roles in early development (e.g., gastrulation and neural tube closure), are involved in different processes of late brain development, such as neuronal migration, axon guidance, and refinement of axon terminals. ZIC proteins are therefore essential for the proper wiring and connectivity of the brain. In this chapter, we review our current knowledge of the role of Zic genes in the late stages of neural circuit formation.
Collapse
|
32
|
Ruiz-Reig N, Studer M. Rostro-Caudal and Caudo-Rostral Migrations in the Telencephalon: Going Forward or Backward? Front Neurosci 2017; 11:692. [PMID: 29311773 PMCID: PMC5742585 DOI: 10.3389/fnins.2017.00692] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/23/2017] [Indexed: 11/13/2022] Open
Abstract
The generation and differentiation of an appropriate number of neurons, as well as its distribution in different parts of the brain, is crucial for the proper establishment, maintenance and plasticity of neural circuitries. Newborn neurons travel along the brain in a process known as neuronal migration, to finalize their correct position in the nervous system. Defects in neuronal migration produce abnormalities in the brain that can generate neurodevelopmental pathologies, such as autism, schizophrenia and intellectual disability. In this review, we present an overview of the developmental origin of the different telencephalic subdivisions and a description of migratory pathways taken by distinct neural populations traveling long distances before reaching their target position in the brain. In addition, we discuss some of the molecules implicated in the guidance of these migratory paths and transcription factors that contribute to the correct migration and integration of these neurons.
Collapse
|
33
|
Guidance of retinal axons in mammals. Semin Cell Dev Biol 2017; 85:48-59. [PMID: 29174916 DOI: 10.1016/j.semcdb.2017.11.027] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 11/17/2017] [Accepted: 11/20/2017] [Indexed: 11/21/2022]
Abstract
In order to navigate through the surrounding environment many mammals, including humans, primarily rely on vision. The eye, composed of the choroid, sclera, retinal pigmented epithelium, cornea, lens, iris and retina, is the structure that receives the light and converts it into electrical impulses. The retina contains six major types of neurons involving in receiving and modifying visual information and passing it onto higher visual processing centres in the brain. Visual information is relayed to the brain via the axons of retinal ganglion cells (RGCs), a projection known as the optic pathway. The proper formation of this pathway during development is essential for normal vision in the adult individual. Along this pathway there are several points where visual axons face 'choices' in their direction of growth. Understanding how these choices are made has advanced significantly our knowledge of axon guidance mechanisms. Thus, the development of the visual pathway has served as an extremely useful model to reveal general principles of axon pathfinding throughout the nervous system. However, due to its particularities, some cellular and molecular mechanisms are specific for the visual circuit. Here we review both general and specific mechanisms involved in the guidance of mammalian RGC axons when they are traveling from the retina to the brain to establish precise and stereotyped connections that will sustain vision.
Collapse
|
34
|
Marcucci F, Murcia-Belmonte V, Wang Q, Coca Y, Ferreiro-Galve S, Kuwajima T, Khalid S, Ross ME, Mason C, Herrera E. The Ciliary Margin Zone of the Mammalian Retina Generates Retinal Ganglion Cells. Cell Rep 2017; 17:3153-3164. [PMID: 28009286 DOI: 10.1016/j.celrep.2016.11.016] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/23/2016] [Accepted: 11/01/2016] [Indexed: 10/20/2022] Open
Abstract
The retina of lower vertebrates grows continuously by integrating new neurons generated from progenitors in the ciliary margin zone (CMZ). Whether the mammalian CMZ provides the neural retina with retinal cells is controversial. Live imaging of embryonic retina expressing eGFP in the CMZ shows that cells migrate laterally from the CMZ to the neural retina where differentiated retinal ganglion cells (RGCs) reside. Because Cyclin D2, a cell-cycle regulator, is enriched in ventral CMZ, we analyzed Cyclin D2-/- mice to test whether the CMZ is a source of retinal cells. Neurogenesis is diminished in Cyclin D2 mutants, leading to a reduction of RGCs in the ventral retina. In line with these findings, in the albino retina, the decreased production of ipsilateral RGCs is correlated with fewer Cyclin D2+ cells. Together, these results implicate the mammalian CMZ as a neurogenic site that produces RGCs and whose proper generation depends on Cyclin D2 activity.
Collapse
Affiliation(s)
- Florencia Marcucci
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Veronica Murcia-Belmonte
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández), 03550 Sant Joan d'Alacant, Spain
| | - Qing Wang
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Yaiza Coca
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández), 03550 Sant Joan d'Alacant, Spain
| | - Susana Ferreiro-Galve
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández), 03550 Sant Joan d'Alacant, Spain
| | - Takaaki Kuwajima
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Sania Khalid
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - M Elizabeth Ross
- Center for Neurogenetics, Feil Family Brain & Mind Research Institute, Weill Cornell Medical College, New York, NY 10021, USA
| | - Carol Mason
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| | - Eloisa Herrera
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández), 03550 Sant Joan d'Alacant, Spain.
| |
Collapse
|
35
|
Zic-Proteins Are Repressors of Dopaminergic Forebrain Fate in Mice and C. elegans. J Neurosci 2017; 37:10611-10623. [PMID: 28972122 DOI: 10.1523/jneurosci.3888-16.2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 09/07/2017] [Accepted: 09/15/2017] [Indexed: 11/21/2022] Open
Abstract
In the postnatal forebrain regionalized neural stem cells along the ventricular walls produce olfactory bulb (OB) interneurons with varying neurotransmitter phenotypes and positions. To understand the molecular basis of this region-specific variability we analyzed gene expression in the postnatal dorsal and lateral lineages in mice of both sexes from stem cells to neurons. We show that both lineages maintain transcription factor signatures of their embryonic site of origin, the pallium and subpallium. However, additional factors, including Zic1 and Zic2, are postnatally expressed in the dorsal stem cell compartment and maintained in the lineage that generates calretinin-positive GABAergic neurons for the OB. Functionally, we show that Zic1 and Zic2 induce the generation of calretinin-positive neurons while suppressing dopaminergic fate in the postnatal dorsal lineage. We investigated the evolutionary conservation of the dopaminergic repressor function of Zic proteins and show that it is already present in C. elegansSIGNIFICANCE STATEMENT The vertebrate brain generates thousands of different neuron types. In this work we investigate the molecular mechanisms underlying this variability. Using a genomics approach we identify the transcription factor signatures of defined neural stem cells and neuron populations. Based thereon we show that two related transcription factors, Zic1 and Zic2, are essential to control the balance between two defined neuron types in the postnatal brain. We show that this mechanism is conserved in evolutionary very distant species.
Collapse
|
36
|
Katori S, Noguchi-Katori Y, Itohara S, Iwasato T. Spinal RacGAP α-Chimaerin Is Required to Establish the Midline Barrier for Proper Corticospinal Axon Guidance. J Neurosci 2017; 37:7682-7699. [PMID: 28747385 PMCID: PMC6596649 DOI: 10.1523/jneurosci.3123-16.2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 05/11/2017] [Accepted: 06/27/2017] [Indexed: 12/17/2022] Open
Abstract
In the developing CNS, the midline barrier, which comprises guidance molecule-expressing midline glial somata and processes, plays a pivotal role in midline axon guidance. Accumulating evidence has revealed the molecular mechanisms by which the midline barrier ensures proper midline guidance for axons. In contrast, the mechanisms for establishing the midline barrier remain obscure. Here, we report that Rac-specific GTPase-activating protein (RacGAP) α-chimaerin is required for both axonal repulsion at and establishment of the midline barrier in the spinal cord. We generated cortex-specific and spinal-cord-specific α-chimaerin gene (Chn1) knock-out mice (Cx-Chn1KO and Sp-Chn1KO mice, respectively) and found that both showed aberrant corticospinal tract (CST) axon midline crossing in the spinal cord. Strikingly, Sp-Chn1KO mice had breaks (holes) in the ephrinB3(+) spinal midline barrier and EphA4(+) CST axons aberrantly crossed the midline through these holes. During normal embryonic development, EphA4(+) spinal cells are located in juxta-midline areas but are excluded from the midline. In contrast, in Chn1KO embryos, several EphA4(+) cells were aberrantly relocated into the midline and the midline barrier was broken around these cells. Similarly, the spinal cord midline of Epha4KO mice was invaded by juxta-midline EphA4 cells (i.e., Epha4 promoter-active cells) during the embryonic stage and holes were formed in the midline barrier. Juxta-midline EphA4 cells in the spinal cord expressed α-chimaerin. We propose that spinal α-chimaerin aids in establishing an intact spinal midline barrier by mediating juxta-midline EphA4(+) cell repulsion, thus preventing these cells from breaking into the ephrinB3(+) midline barrier.SIGNIFICANCE STATEMENT The midline barrier plays a critical role in midline axon guidance, which is fundamental to the formation of neural circuits that are responsible for proper left-right coordination of the body. Studies have revealed some of the mechanisms underlying how the midline barrier navigates axons. In contrast, the establishment of the midline barrier during embryonic development remains unclear. In this study, we determined that α-chimaerin is required for the formation of an intact midline barrier. Spinal-cord-specific α-chimaerin knock-out mice had spinal midline barriers with numerous breaks (holes), through which corticospinal axons aberrantly crossed the midline. We propose that α-chimaerin protects the midline barrier by mediating cell-repulsive signaling in juxta-midline cells, which prevents these cells from invading the midline.
Collapse
Affiliation(s)
- Shota Katori
- Division of Neurogenetics, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan
| | - Yukiko Noguchi-Katori
- Division of Neurogenetics, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan
| | - Shigeyoshi Itohara
- Laboratory for Behavioral Genetics, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan, and
| | - Takuji Iwasato
- Division of Neurogenetics, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan,
- Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, Shizuoka 411-8540, Japan
| |
Collapse
|
37
|
Sedykh I, Yoon B, Roberson L, Moskvin O, Dewey CN, Grinblat Y. Zebrafish zic2 controls formation of periocular neural crest and choroid fissure morphogenesis. Dev Biol 2017; 429:92-104. [PMID: 28689736 DOI: 10.1016/j.ydbio.2017.07.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 05/30/2017] [Accepted: 07/06/2017] [Indexed: 12/31/2022]
Abstract
The vertebrate retina develops in close proximity to the forebrain and neural crest-derived cartilages of the face and jaw. Coloboma, a congenital eye malformation, is associated with aberrant forebrain development (holoprosencephaly) and with craniofacial defects (frontonasal dysplasia) in humans, suggesting a critical role for cross-lineage interactions during retinal morphogenesis. ZIC2, a zinc-finger transcription factor, is linked to human holoprosencephaly. We have previously used morpholino assays to show zebrafish zic2 functions in the developing forebrain, retina and craniofacial cartilage. We now report that zebrafish with genetic lesions in zebrafish zic2 orthologs, zic2a and zic2b, develop with retinal coloboma and craniofacial anomalies. We demonstrate a requirement for zic2 in restricting pax2a expression and show evidence that zic2 function limits Hh signaling. RNA-seq transcriptome analysis identified an early requirement for zic2 in periocular neural crest as an activator of alx1, a transcription factor with essential roles in craniofacial and ocular morphogenesis in human and zebrafish. Collectively, these data establish zic2 mutant zebrafish as a powerful new genetic model for in-depth dissection of cell interactions and genetic controls during craniofacial complex development.
Collapse
Affiliation(s)
- Irina Sedykh
- Department of Integrative Biology, University of Wisconsin, Madison, WI 53706, USA; Department of Neuroscience, University of Wisconsin, Madison, WI 53706, USA
| | - Baul Yoon
- Department of Integrative Biology, University of Wisconsin, Madison, WI 53706, USA; Department of Neuroscience, University of Wisconsin, Madison, WI 53706, USA; Genetics Ph. D. Training Program, University of Wisconsin, Madison, WI 53706, USA
| | - Laura Roberson
- Department of Integrative Biology, University of Wisconsin, Madison, WI 53706, USA; Department of Neuroscience, University of Wisconsin, Madison, WI 53706, USA
| | - Oleg Moskvin
- Primate Research Center, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Colin N Dewey
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Yevgenya Grinblat
- Department of Integrative Biology, University of Wisconsin, Madison, WI 53706, USA; Department of Neuroscience, University of Wisconsin, Madison, WI 53706, USA; McPherson Eye Research Institute, University of Wisconsin, Madison, WI, 53706, USA.
| |
Collapse
|
38
|
Kim JY, Shaker MR, Lee JH, Lee B, Kim H, Sun W. Identification of molecular markers distinguishing adult neural stem cells in the subventricular and subcallosal zones. Anim Cells Syst (Seoul) 2017; 21:152-159. [PMID: 30460064 PMCID: PMC6138335 DOI: 10.1080/19768354.2017.1324522] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 04/14/2017] [Accepted: 04/17/2017] [Indexed: 12/15/2022] Open
Abstract
Neural stem cells (NSCs) in the adult subventricular zone (SVZ) are regionally specified and have distinct molecular gene expression signatures. Recently, we identified the subcallosal zone (SCZ) as a novel brain region where adult NSCs maintain and spontaneously produce neuroblasts. In an attempt to isolate genes specifically expressed in the SCZ or SVZ, microarray analyses of their differentially expressing transcripts were done. The comparison between neurospheres generated from SVZ and SCZ revealed differential expression >1.5-fold in two groups in only 83 genes, representing <0.03% of the genes examined, suggesting that these two populations are largely similar. The differential expression patterns SCZ and SVZ genes were confirmed by RT-PCR and Western blots. The selective expressions of two genes (CRBP1, HMGA1) in SVZ-NSCs were further confirmed by immunohistochemistry. These molecular markers could be useful for further molecular and cellular characterization of NSCs.
Collapse
Affiliation(s)
- Joo Yeon Kim
- Department of Anatomy, Korea University College of Medicine, Seoul, South Korea
| | - Mohammed R. Shaker
- Department of Anatomy, Korea University College of Medicine, Seoul, South Korea
| | - Ju-Hyun Lee
- Department of Anatomy, Korea University College of Medicine, Seoul, South Korea
| | - Boram Lee
- Department of Anatomy, Korea University College of Medicine, Seoul, South Korea
| | - Hyun Kim
- Department of Anatomy, Korea University College of Medicine, Seoul, South Korea
| | - Woong Sun
- Department of Anatomy, Korea University College of Medicine, Seoul, South Korea
| |
Collapse
|
39
|
Friocourt F, Chédotal A. The Robo3 receptor, a key player in the development, evolution, and function of commissural systems. Dev Neurobiol 2017; 77:876-890. [DOI: 10.1002/dneu.22478] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 12/04/2016] [Accepted: 12/06/2016] [Indexed: 12/15/2022]
Affiliation(s)
- François Friocourt
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision; 17 Rue Moreau Paris 75012 France
| | - Alain Chédotal
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision; 17 Rue Moreau Paris 75012 France
| |
Collapse
|
40
|
Murcia-Belmonte V, Expósito G, Herrera E. Time-Lapse Imaging and Cell Tracking of Migrating Cells in Slices and Flattened Telencephalic Vesicles. ACTA ACUST UNITED AC 2017; 79:3.31.1-3.31.12. [PMID: 28398641 DOI: 10.1002/cpns.24] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neuronal migration is a vital process needed for subsequent assembly and function of neural circuitry during embryonic development. The vast majority of neural progenitors are generated far from their final destination and need to migrate considerable distances to reach their specific cortical layer. Innovations in cell culture techniques and fluorescence microscopy now facilitate the direct visualization of cell movements during cortical development. Here, a detailed protocol to record and analyze a particular type of early migrating neurons, the Cajal Retzius Cells, during the development of the telencephalic vesicles in mammals is described. This method applied to other reporter mouse lines or to electroporated mouse embryos can be also used to analyze the migration of different types of moving neurons during cortical development. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Verónica Murcia-Belmonte
- Instituto de Neurociencias de Alicante, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH, Alicante, Spain
| | - Giovanna Expósito
- Instituto de Neurociencias de Alicante, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH, Alicante, Spain
| | - Eloísa Herrera
- Instituto de Neurociencias de Alicante, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH, Alicante, Spain
| |
Collapse
|
41
|
Friocourt F, Lafont AG, Kress C, Pain B, Manceau M, Dufour S, Chédotal A. Recurrent DCC gene losses during bird evolution. Sci Rep 2017; 7:37569. [PMID: 28240285 PMCID: PMC5327424 DOI: 10.1038/srep37569] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 10/31/2016] [Indexed: 01/07/2023] Open
Abstract
During development, midline crossing by axons brings into play highly conserved families of receptors and ligands. The interaction between the secreted ligand Netrin-1 and its receptor Deleted in Colorectal Carcinoma (DCC) is thought to control midline attraction of crossing axons. Here, we studied the evolution of this ligand/receptor couple in birds taking advantage of a wealth of newly sequenced genomes. From phylogeny and synteny analyses we can infer that the DCC gene has been conserved in most extant bird species, while two independent events have led to its loss in two avian groups, passeriformes and galliformes. These convergent accidental gene loss events are likely related to chromosome Z rearrangement. We show, using whole-mount immunostaining and 3Disco clearing, that in the nervous system of all birds that have a DCC gene, DCC protein expression pattern is similar to other vertebrates. Surprisingly, we show that the early developmental pattern of commissural tracts is comparable in all birds, whether or not they have a DCC receptor. Interestingly, only 4 of the 5 genes encoding secreted netrins, the DCC ligands in vertebrates, were found in birds, but Netrin-5 was absent. Together, these results support a remarkable plasticity of commissural axon guidance mechanisms in birds.
Collapse
Affiliation(s)
- François Friocourt
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision, 17 Rue Moreau, 75012 Paris, France
| | - Anne-Gaelle Lafont
- Muséum National d’Histoire Naturelle, Sorbonne Universités, Research Unit BOREA, Biology of Aquatic Organisms and Ecosystems, CNRS 7208, IRD207, UPMC, UCN, Paris, France
| | - Clémence Kress
- Université Lyon 1, INSERM, INRA, Stem Cell and Brain Research Institute, U1208, USC1361, 69500 Bron, France
| | - Bertrand Pain
- Université Lyon 1, INSERM, INRA, Stem Cell and Brain Research Institute, U1208, USC1361, 69500 Bron, France
| | - Marie Manceau
- Center for Interdisciplinary Research in Biology, CNRS UMR 7241, Collège de France, 75005 Paris, France
| | - Sylvie Dufour
- Muséum National d’Histoire Naturelle, Sorbonne Universités, Research Unit BOREA, Biology of Aquatic Organisms and Ecosystems, CNRS 7208, IRD207, UPMC, UCN, Paris, France
| | - Alain Chédotal
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision, 17 Rue Moreau, 75012 Paris, France
| |
Collapse
|
42
|
Serradj N, Agger SF, Hollis ER. Corticospinal circuit plasticity in motor rehabilitation from spinal cord injury. Neurosci Lett 2016; 652:94-104. [PMID: 27939980 DOI: 10.1016/j.neulet.2016.12.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 11/29/2016] [Accepted: 12/02/2016] [Indexed: 11/18/2022]
Abstract
Restoring corticospinal function after spinal cord injury is a significant challenge as the corticospinal tract elicits no substantive, spontaneous regeneration, and its interruption leaves a permanent deficit. The corticospinal circuit serves multiple motor and sensory functions within the mammalian nervous system as the direct link between isocortex and spinal cord. Maturation of the corticospinal circuit involves the refinement of projections within the spinal cord and a subsequent refinement of motor maps within the cortex. The plasticity of these cortical motor maps mirrors the acquisition of skilled motor learning, and both the maps and motor skills are disrupted following injury to the corticospinal tract. The motor cortex exhibits the capacity to incorporate changes in corticospinal projections induced by both spontaneous and therapeutic-mediated plasticity of corticospinal axons through appropriate rehabilitation. An understanding of the mechanisms of corticospinal plasticity in motor learning will undoubtedly help inform strategies to improve motor rehabilitation after spinal cord injury.
Collapse
Affiliation(s)
- Najet Serradj
- Burke Medical Research Institute, White Plains, New York, NY 10605, United States
| | - Sydney F Agger
- Burke Medical Research Institute, White Plains, New York, NY 10605, United States
| | - Edmund R Hollis
- Burke Medical Research Institute, White Plains, New York, NY 10605, United States; Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10065, United States.
| |
Collapse
|
43
|
Abstract
…once the development was ended, the founts of growth and regeneration of the axons and dendrites dried up irrevocably. Santiago Ramón y Cajal Cajal's neurotropic theory postulates that the complexity of the nervous system arises from the collaboration of neurotropic signals from neuronal and non-neuronal cells and that once development has ended, a paucity of neurotropic signals means that the pathways of the central nervous system are "fixed, ended, immutable". While the capacity for regeneration and plasticity of the central nervous system may not be quite as paltry as Cajal proposed, regeneration is severely limited in scope as there is no spontaneous regeneration of long-distance projections in mammals and therefore limited opportunity for functional recovery following spinal cord injury. It is not a far stretch from Cajal to hypothesize that reappropriation of the neurotropic programs of development may be an appropriate strategy for reconstitution of injured circuits. It has become clear, however, that a significant number of the molecular cues governing circuit development become re-active after injury and many assume roles that paradoxically obstruct the functional re-wiring of severed neural connections. Therefore, the problem to address is how individual neural circuits respond to specific molecular cues following injury, and what strategies will be necessary for instigating functional repair or remodeling of the injured spinal cord.
Collapse
Affiliation(s)
- Edmund R Hollis
- Burke Medical Research Institute, White Plains, NY, USA.
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
44
|
Satoh D, Pudenz C, Arber S. Context-Dependent Gait Choice Elicited by EphA4 Mutation in Lbx1 Spinal Interneurons. Neuron 2016; 89:1046-58. [DOI: 10.1016/j.neuron.2016.01.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 12/18/2015] [Accepted: 01/12/2016] [Indexed: 12/23/2022]
|
45
|
Mechanisms of ephrin-Eph signalling in development, physiology and disease. Nat Rev Mol Cell Biol 2016; 17:240-56. [PMID: 26790531 DOI: 10.1038/nrm.2015.16] [Citation(s) in RCA: 461] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Eph receptor Tyr kinases and their membrane-tethered ligands, the ephrins, elicit short-distance cell-cell signalling and thus regulate many developmental processes at the interface between pattern formation and morphogenesis, including cell sorting and positioning, and the formation of segmented structures and ordered neural maps. Their roles extend into adulthood, when ephrin-Eph signalling regulates neuronal plasticity, homeostatic events and disease processes. Recently, new insights have been gained into the mechanisms of ephrin-Eph signalling in different cell types, and into the physiological importance of ephrin-Eph in different organs and in disease, raising questions for future research directions.
Collapse
|
46
|
Abstract
UNLABELLED Human mutations in ZIC2 have been identified in patients with holoprosencephaly and schizophrenia. Similarly, Zic2 mutant mice exhibit holoprosencephaly in homozygosis and behavioral and morphological schizophrenic phenotypes associated with forebrain defects in heterozygosis. Despite the devastating effects of mutations in Zic2, the cellular and molecular mechanisms that provoke Zic2-deficiency phenotypes are yet unclear. Here, we report a novel role for this transcription factor in the migration of three different types of forebrain neurons: the Cajal-Retzius cells that populate the surface of the telencephalic vesicles, an amygdaloid group of cells originated in the caudal pole of the telencephalic pallium, and a cell population that travels from the prethalamic neuroepithelium to the ventral lateral geniculate nucleus. Our results also suggest that the receptor EphB1, previously identified as a Zic2 target, may mediate, at least partially, Zic2-dependent migratory events. According to these results, we propose that deficiencies in cell motility and guidance contribute to most of the forebrain pathologies associated with Zic2 mutations. SIGNIFICANCE STATEMENT Although the phenotype of Zic2 mutant individuals was reported more than 10 years ago, until now, the main function of this transcription factor during early development has not been precisely defined. Here, we reveal a previously unknown role for Zic2 in the migration of forebrain neurons such as Cajal-Retzius cells, interneurons moving to the ventral lateral geniculate nucleus, and neocortical cells going to the amygdala. We believe that the role of this transcription factor in certain populations of migratory cells contributes to defects in cortical layering and hypocellularity in the ventral LGN and amygdala and will contribute to our understanding of the devastating phenotypes associated with Zic2 mutations in both humans and mice.
Collapse
|
47
|
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
|
48
|
Neurons in the lateral part of the lumbar spinal cord show distinct novel axon trajectories and are excited by short propriospinal ascending inputs. Brain Struct Funct 2015; 221:2343-60. [PMID: 25912439 DOI: 10.1007/s00429-015-1046-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 04/15/2015] [Indexed: 10/23/2022]
Abstract
The role of spinal dorsal horn propriospinal connections in nociceptive processing is not yet established. Recently described, rostrocaudally oriented axon collaterals of lamina I projection and local-circuit neurons (PNs and LCNs) running in the dorsolateral funiculus (DLF) may serve as the anatomical substrate for intersegmental processing. Putative targets of these axons include lateral dendrites of superficial dorsal horn neurons, including PNs, and also neurons in the lateral spinal nucleus (LSN) that are thought to be important integrator units receiving, among others, visceral sensory information. Here we used an intact spinal cord preparation to study intersegmental connections within the lateral part of the superficial dorsal horn. We detected brief monosynaptic and prolonged polysynaptic excitation of lamina I and LSN neurons when stimulating individual dorsal horn neurons located caudally, even in neighboring spinal cord segments. These connections, however, were infrequent. We also revealed that some projection neurons outside the dorsal grey matter and in the LSN have distinct, previously undescribed course of their projection axon. Our findings indicate that axon collaterals of lamina I PNs and LCNs in the DLF rarely form functional connections with other lamina I and LSN neurons and that the majority of their targets are on other elements of the dorsal horn. The unique axon trajectories of neurons in the dorsolateral aspect of the spinal cord, including the LSN do not fit our present understanding of midline axon guidance and suggest that their function and development differ from the neurons inside lamina I. These findings emphasize the importance of understanding the connectivity matrix of the superficial dorsal horn in order to decipher spinal sensory information processing.
Collapse
|
49
|
Zic2 is an enhancer-binding factor required for embryonic stem cell specification. Mol Cell 2015; 57:685-694. [PMID: 25699711 PMCID: PMC5009765 DOI: 10.1016/j.molcel.2015.01.007] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 11/09/2014] [Accepted: 01/05/2015] [Indexed: 12/03/2022]
Abstract
The Zinc finger protein of the cerebellum 2 (Zic2) is one of the vertebrate homologs of the Drosophila pair-rule gene odd-paired (opa). Our molecular and biochemical studies demonstrate that Zic2 preferentially binds to transcriptional enhancers and is required for the regulation of gene expression in embryonic stem cells. Detailed genome-wide and molecular studies reveal that Zic2 can function with Mbd3/NuRD in regulating the chromatin state and transcriptional output of genes linked to differentiation. Zic2 is required for proper differentiation of ES cells, similar to what has been previously reported for Mbd3/NuRD. Our study identifies Zic2 as a key factor in the execution of transcriptional fine-tuning with Mbd3/NuRD in ES cells through interactions with enhancers. Our study also points to the role of the Zic family of proteins as enhancer-specific binding factors functioning in development.
Collapse
|
50
|
Soares CA, Mason CA. Transient ipsilateral retinal ganglion cell projections to the brain: Extent, targeting, and disappearance. Dev Neurobiol 2015; 75:1385-401. [PMID: 25788284 DOI: 10.1002/dneu.22291] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/13/2015] [Accepted: 03/15/2015] [Indexed: 12/27/2022]
Abstract
During development of the mammalian eye, the first retinal ganglion cells (RGCs) that extend to the brain are located in the dorsocentral (DC) retina. These RGCs extend to either ipsilateral or contralateral targets, but the ipsilateral projections do not survive into postnatal periods. The function and means of disappearance of the transient ipsilateral projection are not known. We have followed the course of this transient early ipsilateral cohort of RGCs, paying attention to how far they extend, whether they enter targets and if so, which ones, and the time course of their disappearance. The DC ipsilateral RGC axons were traced using DiI labeling at E13.5 and E15.5 to compare the proportion of ipsi- versus contralateral projections during the first period of growth. In utero electroporation of E12.5 retina with GFP constructs was used to label axons that could be visualized at succeeding time points into postnatal ages. Our results show that the earliest ipsilateral axons grow along the cellular border of the brain, and are segregated from the laterally positioned contralateral axons from the same retinal origin. In agreement with previous reports, although many early RGCs extend ipsilaterally, after E16 their number rapidly declines. Nonetheless, some ipsilateral axons from the DC retina enter the superior colliculus and arborize minimally, but very few enter the dorsal lateral geniculate nucleus and those that do extend only short branches. While the mechanism of selective axonal disappearance remains elusive, these data give further insight into establishment of the visual pathways.
Collapse
Affiliation(s)
- Célia A Soares
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York.,Life and Health Science Research Institute, School of Health Sciences, University of Minho, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Carol A Mason
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York.,Department of Neuroscience, College of Physicians and Surgeons, Columbia University, New York.,Department of Ophthalmology, College of Physicians and Surgeons, Columbia University, New York
| |
Collapse
|