Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
PLoS One ; 11(2): e0147535, 2016.
Article in English | MEDLINE | ID: mdl-26859889

ABSTRACT

The heterotrimeric G protein subunit Gsα couples receptors to activate adenylyl cyclase and is required for the intracellular cAMP response and protein kinase A (PKA) activation. Gsα is ubiquitously expressed in many cell types; however, the role of Gsα in neural crest cells (NCCs) remains unclear. Here we report that NCCs-specific Gsα knockout mice die within hours after birth and exhibit dramatic craniofacial malformations, including hypoplastic maxilla and mandible, cleft palate and craniofacial skeleton defects. Histological and anatomical analysis reveal that the cleft palate in Gsα knockout mice is a secondary defect resulting from craniofacial skeleton deficiencies. In Gsα knockout mice, the morphologies of NCCs-derived cranial nerves are normal, but the development of dorsal root and sympathetic ganglia are impaired. Furthermore, loss of Gsα in NCCs does not affect cranial NCCs migration or cell proliferation, but significantly accelerate osteochondrogenic differentiation. Taken together, our study suggests that Gsα is required for neural crest cells-derived craniofacial development.


Subject(s)
Facial Bones/embryology , GTP-Binding Protein alpha Subunits, Gs/metabolism , Morphogenesis , Skull/embryology , Animals , Cell Differentiation , Cleft Palate/embryology , Cleft Palate/genetics , Facial Bones/abnormalities , GTP-Binding Protein alpha Subunits, Gs/deficiency , GTP-Binding Protein alpha Subunits, Gs/genetics , Ganglia, Sympathetic/embryology , Gene Knockout Techniques , Mice , Mutation , Neural Crest/cytology , Neural Crest/metabolism , Skull/abnormalities , Spinal Nerve Roots/embryology
2.
Congenit Anom (Kyoto) ; 53(3): 122-6, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23998265

ABSTRACT

The spinal nerve, which is composed of dorsal root ganglion (DRG) sensory axons and spinal motor axons, forms the dorsal ramus projecting to the dorsal musculature. By using the free-floating immunohistochemistry method, we closely examined the spatiotemporal pattern of the formation of the dorsal ramus and the relationship between its projection to the myotome/dorsal musculature and semaphorin 3A (Sema3A), which is an axonal guidance molecule. In embryonic day (E) 10.5-E11.5 wild-type mouse embryos, we clearly showed the existence of a waiting period for the dorsal ramus projection to the myotome. In contrast, in E10.5-E11.5 Sema3A-deficient embryos, the dorsal ramus fibers projected beyond the edge of the myotome without exhibiting the waiting period for projection. These results strongly suggest that the delayed innervation by dorsal ramus fibers may be caused by Sema3A-induced axon repulsion derived from the myotome. Next, by performing culture experiments, we confirmed that E12.5 mouse axons responded to Sema3A-induced repulsion. Together, our results imply that Sema3A may play a key role in the proper development of the dorsal ramus projection.


Subject(s)
Muscle, Skeletal/innervation , Semaphorin-3A/metabolism , Spinal Nerve Roots/metabolism , Animals , Axons/metabolism , Mice , Mice, Knockout , Semaphorin-3A/genetics , Spinal Nerve Roots/embryology
3.
J Comp Neurol ; 521(13): 2987-3002, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23504940

ABSTRACT

The avian cervical spinal cord includes motoneurons (MNs) that send their axons through the dorsal roots. They have been called dorsal motoneurons (dMNs) and assumed to correspond to MNs of the accessory nerve that innervate the cucullaris muscle (SAN-MNs). However, their target muscles have not been elucidated to date. The present study sought to determine the targets and the specific combination of transcription factors expressed by dMNs and SAN-MNs and to describe the detailed development of dMNs. Experiments with tracing techniques confirmed that axons of dMNs innervated the cucullaris muscle. Retrogradely labeled dMNs were distributed in the ventral horn of C3 and more caudal segments. In most cases, some dMNs were also observed in the C2 segment. It was also demonstrated that SAN-MNs existed in the ventral horn of the C1-2 segments and the adjacent caudal hindbrain. Both SAN-MNs and dMNs expressed Isl1 but did not express Isl2, MNR2, or Lhx3. Rather, these MNs expressed Phox2b, a marker for branchial motoneurons (brMNs), although the intensity of expression was weaker. Dorsal MNs and SAN-MNs were derived from the Nkx2.2-positive precursor domain and migrated dorsally. Dorsal MNs remain in the ventral domain of the neural tube, unlike brMNs in the brainstem. These results indicate that dMNs and SAN-MNs belong to a common MN population innervating the cucullaris muscle and also suggest that they are similar to brMNs of the brainstem, although there are differences in Phox2b expression and in the final location of each population. J. Comp. Neurol. 521: 2987-3002, 2013. © 2013 Wiley Periodicals, Inc.


Subject(s)
Motor Neurons/physiology , Muscle, Skeletal/physiology , Spinal Nerve Roots/cytology , Accessory Nerve/physiology , Animals , Biotin/analogs & derivatives , Biotin/metabolism , Chick Embryo , Dextrans/metabolism , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Muscle, Skeletal/embryology , Nerve Tissue Proteins/metabolism , Neural Pathways/physiology , Phenylurea Compounds/administration & dosage , Spinal Nerve Roots/embryology , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish Proteins
4.
Genesis ; 50(7): 552-60, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22368151

ABSTRACT

Prrxl1-CreER(T2) transgenic mice expressing tamoxifen-inducible Cre recombinase were generated by modifying a Prrxl1-containing BAC clone. Cre recombination activity was examined in Prrxl1-CreER(T2); Rosa26 reporter mice at various embryonic and postnatal stages. Pregnant mice were treated with a single dose of tamoxifen at embryonic day (E) 9.5 or E12.5, and X-gal staining was performed 2 days later. Strong X-gal staining was observed in the somatosensory ganglia (e.g., dorsal root and trigeminal ganglia) and the first central sites for processing somatosensory information (e.g., spinal dorsal horn and trigeminal nerve-associated nuclei). When tamoxifen was administered at postnatal day (P) 20 or in adulthood (P120), strong Cre recombination activity was present in the primary somatosensory ganglia, while weak Cre recombination activity was found in the spinal dorsal horn, mesencephalic trigeminal nucleus, principal sensory trigeminal nucleus, and spinal trigeminal nucleus. This mouse line provides a useful tool for exploring genes' functions in the somatosensory system in a time-controlled way.


Subject(s)
Afferent Pathways/physiology , Homeodomain Proteins/genetics , Mice, Transgenic , Nerve Tissue Proteins/genetics , Somatosensory Cortex/physiology , Spinal Nerve Roots/physiology , Transcription Factors/genetics , Trigeminal Ganglion/physiology , Afferent Pathways/embryology , Animals , Chromosomes, Artificial, Bacterial , Embryo, Mammalian , Female , Founder Effect , Gene Expression Regulation, Developmental/drug effects , Genes, Reporter , Integrases/genetics , Mice , Pregnancy , Promoter Regions, Genetic , Proteins/genetics , RNA, Untranslated , Recombination, Genetic/drug effects , Somatosensory Cortex/embryology , Spinal Nerve Roots/embryology , Tamoxifen/administration & dosage , Time Factors , Trigeminal Ganglion/embryology
5.
Adv Clin Exp Med ; 21(6): 735-42, 2012.
Article in English | MEDLINE | ID: mdl-23457131

ABSTRACT

BACKGROUND: The median nerve is an important nerve leaving the brachial plexus. Median nerve damages may result from tunnel syndromes or injuries. The nerve anatomical variants are of great clinical importance in hand surgery. OBJECTIVES: Clinical evaluation of median nerve divergence from brachial plexus morphological variability in foetal period. MATERIAL AND METHODS: The material consisted of 220 brachial plexus sections derived from 110 foetuses aged 14-32 weeks of foetal life (50 females and 60 males, in CRL: 80-233 mm). The survey incorporated the following methods: dissection, anthropological, image digital acquisition, Image J computer transformation system, GIMP programme and statistical methods. Typology assessment was based on 0/1 system. Sexual dimorphism and symmetry were examined. RESULTS: Median nerve left directly lateral cord in 5 cases. In 59 (26.81%) plexuses, anterior division of middle trunk co-created median nerves anomalies. The total of 9 types of anterior division of middle trunk as well as of median nerve were distinguished. Median nerve double root leaving lateral cord was observed in 10 (9.09%) cases, whereas triple lateral root was seen in one case. In 1/3 of the examined plexuses, median nerve roots combined to form the nerve beneath humeral bone head and even in ½ of the bone distal length (type II and III). Type II prevailed more often on the left side. CONCLUSIONS: Median nerve roots as well as the median nerve itself are characteristic for significant morphological variability. Nerve roots low junction into median nerve is clinically favourable as it can prevent nerve damage during injuries.


Subject(s)
Fetus/anatomy & histology , Fetus/embryology , Median Nerve/anatomy & histology , Median Nerve/embryology , Female , Humans , Male , Spinal Nerve Roots/anatomy & histology , Spinal Nerve Roots/embryology
6.
Glia ; 57(13): 1450-7, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19243017

ABSTRACT

Neural crest (NC) cells are a multipotent, highly migratory cell population that generates most of the components of the peripheral nervous system (PNS), including the glial Schwann cells (SC) and boundary cap (BC) cells. These latter cells are located at the interface between the central nervous system and PNS, at the exit/entry points of ventral motor/dorsal sensory axons and give rise to all SC in the nerve roots and to a subset of nociceptive neurons and satellite cells in the dorsal root ganglia. In the present study we have compared BC cells with two closely related cell types, NC and Schwann cell precursors (SCpr), by RNA profiling. This led to the definition of a set of 10 genes that show specific expression in BC cells and/or in their derivatives along the nerve roots. Analysis of the expression of these genes during mouse development revealed novel features, of those most important are: (i) dorsal and ventral nerve root BC cell derivatives express different sets of genes, suggesting that they have distinct properties; (ii) these cells undergo major modifications in their gene expression pattern between embryonic days 14.5 and 17.5, possibly linked to the SCpr-immature Schwann cell transition; (iii) nerve roots SC differ from more distal SC not only in their origins and locations, but also in their gene expression patterns. In conclusion, the identification of these novel makers opens the way for a detailed characterization of BC cells in both mouse and man.


Subject(s)
Gene Expression Regulation, Developmental , Neural Crest/embryology , Neural Crest/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/metabolism , Early Growth Response Protein 2/genetics , Early Growth Response Protein 2/metabolism , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , RNA/metabolism , Receptors, Atrial Natriuretic Factor/genetics , Receptors, Atrial Natriuretic Factor/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Schwann Cells/physiology , Spinal Nerve Roots/embryology , Spinal Nerve Roots/physiology , Stem Cells/physiology
7.
J Anat ; 213(2): 194-201, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18537848

ABSTRACT

Ventral roots in all mammalian species, including humans, contain significant numbers of unmyelinated axons, many of them afferents transmitting nociceptive signals from receptive fields in skin, viscera, muscles and joints. Observations in cats indicate that these afferents do not enter the spinal cord via the ventral root, but rather turn distally and enter the dorsal root. Some unmyelinated axons are postganglionic autonomic efferents that innervate blood vessels of the root and the pia mater. In the feline L7 segment, a substantial proportion of unmyelinated axons are not detectable until late in perinatal development. The mechanisms inducing this late ingrowth, and the recruitment of Schwann cells (indispensable, at this stage, for axonal survival and sustenance), are unknown. We have counted axons and Schwann cells in both ends of the L7 ventral root in young kittens and made the following observations. (1) The total number of axons detectable in the root increased throughout the range of investigated ages. (2) The number of myelinated axons was similar in the root's proximal and distal ends. The increased number of unmyelinated axons with age is thus due to increased numbers of small unmyelinated axons. (3) The number of separated large probably promyelin axons was about the same in the proximal and distal ends of the root. (4) Schwann cells appeared to undergo redistribution, from myelinated to unmyelinated axons. (5) During redistribution of Schwann cells they first appear as aberrant Schwann cells and then become endoneurial X-cells temporarily free of axonal contact. We hypothesize that unmyelinated axons invade the ventral root from its distal end, that this ingrowth is particularly intense during the first postnatal month and that disengaged Schwann cells, eliminated from myelinated motoneuron axons, provide the ingrowing axons with structural and trophic support.


Subject(s)
Cats/growth & development , Neurons, Afferent/physiology , Schwann Cells/physiology , Spinal Nerve Roots/growth & development , Aging/pathology , Animals , Animals, Newborn , Axons/physiology , Axons/ultrastructure , Cats/anatomy & histology , Cats/embryology , Cell Nucleus/ultrastructure , Microscopy, Electron , Nerve Fibers, Unmyelinated/physiology , Nerve Fibers, Unmyelinated/ultrastructure , Neurons, Afferent/ultrastructure , Schwann Cells/ultrastructure , Spinal Nerve Roots/embryology , Spinal Nerve Roots/ultrastructure
8.
J Comput Neurosci ; 25(1): 39-63, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18322788

ABSTRACT

Many developing neural systems exhibit spontaneous activity (O'Donovan, Curr Opin Neurobiol 9:94-104, 1999; Feller, Neuron 22:653-656, 1999) characterized by episodes of discharge (active phases) when many cells are firing, separated by silent phases during which few cells fire. Various models exhibit features of episodic behavior by means of recurrent excitation for supporting an episode and slow activity-dependent depression for terminating one. The basic mechanism has been analyzed using mean-field, firing-rate models. Firing-rate models are typically formulated ad hoc, not derived from a spiking network description, and the effects of substantial heterogeneity amongst the units are not usually considered. Here we develop an excitatory network of spiking neurons (N-cell model) with slow synaptic depression to model episodic rhythmogenesis. This N-cell model displays episodic behavior over a range of heterogeneity in bias currents. Important features of the episodic behavior include orderly recruitment of individual cells during silent phases and existence of a dynamical process whereby a small critical subpopulation of intermediate excitability conveys synaptic drive from active to silent cells. We also derive a general self-consistency equation for synaptic drive that includes cell heterogeneity explicitly. We use this mean-field description to expose the dynamical bistability that underlies episodic behavior in the heterogeneous network. In a systematic numerical study we find that the robustness of the episodic behavior improves with increasing heterogeneity. Furthermore, the heterogeneity of depression variables (imparted by the heterogeneity in cellular firing thresholds) plays an important role in this improvement: it renders the network episodic behavior more robust to variations in excitability than if depression is uniformized. We also investigate the effects of noise vs. heterogeneity on the robustness of episodic behavior, especially important for the developing nervous system. We demonstrate that noise-induced episodes are very fragile, whereas heterogeneity-produced episodic rhythm is robust.


Subject(s)
Action Potentials/physiology , Computer Simulation , Models, Neurological , Neural Networks, Computer , Neurons, Efferent/physiology , Animals , Chick Embryo , Nerve Net/physiology , Periodicity , Spinal Nerve Roots/cytology , Spinal Nerve Roots/embryology
9.
Neural Dev ; 2: 28, 2007 Dec 18.
Article in English | MEDLINE | ID: mdl-18088409

ABSTRACT

BACKGROUND: During spinal cord development, expression of chicken SEMAPHORIN6A (SEMA6A) is almost exclusively found in the boundary caps at the ventral motor axon exit point and at the dorsal root entry site. The boundary cap cells are derived from a population of late migrating neural crest cells. They form a transient structure at the transition zone between the peripheral nervous system (PNS) and the central nervous system (CNS). Ablation of the boundary cap resulted in emigration of motoneurons from the ventral spinal cord along the ventral roots. Based on its very restricted expression in boundary cap cells, we tested for a role of Sema6A as a gate keeper between the CNS and the PNS. RESULTS: Downregulation of Sema6A in boundary cap cells by in ovo RNA interference resulted in motoneurons streaming out of the spinal cord along the ventral roots, and in the failure of dorsal roots to form and segregate properly. PlexinAs interact with class 6 semaphorins and are expressed by both motoneurons and sensory neurons. Knockdown of PlexinA1 reproduced the phenotype seen after loss of Sema6A function both at the ventral motor exit point and at the dorsal root entry site of the lumbosacral spinal cord. Loss of either PlexinA4 or Sema6D function had an effect only at the dorsal root entry site but not at the ventral motor axon exit point. CONCLUSION: Sema6A acts as a gate keeper between the PNS and the CNS both ventrally and dorsally. It is required for the clustering of boundary cap cells at the PNS/CNS interface and, thus, prevents motoneurons from streaming out of the ventral spinal cord. At the dorsal root entry site it organizes the segregation of dorsal roots.


Subject(s)
Body Patterning/genetics , Central Nervous System/embryology , Neuroglia/metabolism , Peripheral Nervous System/embryology , Semaphorins/metabolism , Animals , COS Cells , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Differentiation/genetics , Cell Movement/genetics , Central Nervous System/cytology , Central Nervous System/metabolism , Chick Embryo , Chlorocebus aethiops , Down-Regulation/genetics , Motor Neurons/cytology , Motor Neurons/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Crest/cytology , Neural Crest/embryology , Neural Crest/metabolism , Neuroglia/cytology , Peripheral Nervous System/cytology , Peripheral Nervous System/metabolism , RNA Interference , Semaphorins/genetics , Spinal Cord/cytology , Spinal Cord/embryology , Spinal Cord/metabolism , Spinal Nerve Roots/cytology , Spinal Nerve Roots/embryology , Spinal Nerve Roots/metabolism
10.
Neuroreport ; 18(16): 1645-9, 2007 Oct 29.
Article in English | MEDLINE | ID: mdl-17921861

ABSTRACT

In the early stages, the dorsal root ganglion neurons extend their axons toward the dorsal spinal cord. We previously showed that surround repulsion by semaphorin 3A prevents sensory axons from straying from their paths. The finding, however, that sensory trajectories toward the dorsal spinal cord are almost normal in semaphorin 3A-deficient littermates raises the possibility that a chemoattraction-based mechanism also contributes to the formation of sensory axonal projections. By employing culture assays, we show that the dorsal spinal cord secretes chemoattractants for the dorsal root ganglion axons. Furthermore, we demonstrate that the activity of a dorsal spinal cord-derived cue is specific for early sensory axons. These results suggest that dorsal spinal cord-derived chemoattractants contribute to the formation of the initial trajectories of sensory axons.


Subject(s)
Axons/metabolism , Ganglia, Spinal/embryology , Nerve Growth Factors/metabolism , Neurons, Afferent/metabolism , Spinal Cord/embryology , Spinal Nerve Roots/embryology , Afferent Pathways/cytology , Afferent Pathways/embryology , Afferent Pathways/metabolism , Animals , Axons/ultrastructure , Biological Assay , Body Patterning/physiology , Cell Culture Techniques , Cell Differentiation/physiology , Cells, Cultured , Chemotactic Factors/metabolism , Chick Embryo , Coculture Techniques , Cues , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Growth Cones/metabolism , Growth Cones/ultrastructure , Neurons, Afferent/cytology , Receptors, Nerve Growth Factor/metabolism , Semaphorin-3A/deficiency , Semaphorin-3A/genetics , Spinal Cord/metabolism , Spinal Nerve Roots/cytology , Spinal Nerve Roots/metabolism
11.
Glia ; 55(1): 93-103, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17024657

ABSTRACT

Connexins are transmembrane proteins forming gap junction channels for direct intercellular and, for example in myelinating glia cells, intracellular communication. In mature myelin-forming Schwann cells, expression of multiple connexins, i.e. connexin (Cx) 43, Cx29, Cx32, and Cx46 (after nerve injury) has been detected. However, little is known about connexin protein expression during Schwann cell development. Here we use histochemical methods on wildtype and Cx29lacZ transgenic mice to investigate the developmental expression of connexins in the Schwann cell lineage. Our data demonstrate that in the mouse Cx43, Cx29, and Cx32 protein expression is activated in a developmental sequence that is clearly correlated with major developmental steps in the lineage. Only Cx43 was expressed from neural crest cells onwards. Cx29 protein expression was absent from neural crest cells but appeared as neural crest cells generated precursors (embryonic day 12) both in vivo and in vitro. This identifies Cx29 as a novel marker for cells of the defined Schwann cell lineage. The only exception to this were dorsal roots, where the expression of Cx29 was delayed four days relative to ventral roots and spinal nerves. Expression of Cx32 commenced postnatally, coinciding with the onset of myelination. Thus, the coordinated expression of connexin proteins in cells of the embryonic and postnatal Schwann cell lineage might point to a potential role in peripheral nerve development and maturation.


Subject(s)
Cell Differentiation/physiology , Connexins/metabolism , Nerve Tissue Proteins/metabolism , Neural Crest/metabolism , Peripheral Nervous System/embryology , Peripheral Nervous System/growth & development , Schwann Cells/metabolism , Stem Cells/metabolism , Animals , Animals, Newborn , Biomarkers , Cell Lineage/physiology , Cells, Cultured , Connexin 43 , Connexins/genetics , Female , Gap Junctions/metabolism , Gene Expression Regulation, Developmental/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Neural Crest/cytology , Peripheral Nervous System/cytology , Schwann Cells/cytology , Spinal Nerve Roots/cytology , Spinal Nerve Roots/embryology , Spinal Nerve Roots/metabolism , Stem Cells/cytology , Gap Junction beta-1 Protein
12.
Dev Dyn ; 235(8): 2071-82, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16691572

ABSTRACT

A wide range of physiological and behavioral processes, such as social, sexual, and maternal behaviors, learning and memory, and osmotic homeostasis are influenced by the neurohypophysial peptides oxytocin and vasopressin. Disruptions of these hormone systems have been linked to several neurobehavioral disorders, including autism, Prader-Willi syndrome, affective disorders, and obsessive-compulsive disorder. Studies in zebrafish promise to reveal the complex network of regulatory genes and signaling pathways that direct the development of oxytocin- and vasopressin-like neurons, and provide insight into factors involved in brain disorders associated with disruption of these systems. Isotocin, which is homologous to oxytocin, is expressed early, in a simple pattern in the developing zebrafish brain. Single-minded 1 (sim1), a member of the bHLH-PAS family of transcriptional regulatory genes, is required for terminal differentiation of mammalian oxytocin cells and is a master regulator of neurogenesis in Drosophila. Here we show that sim1 is expressed in the zebrafish forebrain and is required for isotocin cell development. The expression pattern of sim1 mRNA in the embryonic forebrain is dynamic and complex, and overlaps with isotocin expression in the preoptic area. We provide evidence that the role of sim1 in zebrafish neuroendocrine cell development is evolutionarily conserved with that of mammals.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Oxytocin/analogs & derivatives , Repressor Proteins/metabolism , Transcription, Genetic/genetics , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/classification , Basic Helix-Loop-Helix Transcription Factors/genetics , Body Patterning , Diencephalon/embryology , Diencephalon/metabolism , Gene Expression Regulation, Developmental , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Oxytocin/metabolism , Phylogeny , Prosencephalon/embryology , Prosencephalon/metabolism , RNA, Messenger/genetics , Repressor Proteins/classification , Repressor Proteins/genetics , Spinal Nerve Roots/embryology , Spinal Nerve Roots/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish/genetics , Zebrafish Proteins/classification , Zebrafish Proteins/genetics
13.
J Neurophysiol ; 93(5): 2922-33, 2005 May.
Article in English | MEDLINE | ID: mdl-15574794

ABSTRACT

Locally projecting inhibitory interneurons play a crucial role in the patterning and timing of network activity. However, because of their relative inaccessibility, little is known about their development or incorporation into circuits. In this study, we characterized the functional onset, neurotransmitters, rostrocaudal spread, and funicular distribution of one such spinal interneuronal circuit during development. The R-interneuron is the avian homologue of the mammalian Renshaw cell. Both cell types receive input from motoneuron recurrent collaterals and make direct connections back onto motoneurons. By stimulating motoneurons projecting in a given ventral root and recording the response in adjacent ventral roots, we demonstrate that the R-interneuron circuit becomes functional between embryonic day 6 (E6) and E7. This ventral root response is observed at E11 and at E14 until it can no longer be detected at E16. Using bath-applied neurotransmitter receptor antagonists, we were able to demonstrate that the circuit is predominately nicotinic and GABAergic from E7.5 to E15. We also found a glutamatergic component to the pathway throughout this developmental period. The R-interneuron projects three or more segments both rostrally and caudally through the ventrolateral funiculus. The distribution of this circuit may become more locally focused between E7.5 and E15.


Subject(s)
Interneurons/physiology , Nerve Net/embryology , Neural Inhibition/physiology , Spinal Cord/cytology , 2-Amino-5-phosphonovalerate/pharmacology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Age Factors , Analysis of Variance , Animals , Bicuculline/pharmacology , Chick Embryo , Drug Interactions , Electric Stimulation/methods , Embryo, Mammalian , Embryo, Nonmammalian , Excitatory Amino Acid Antagonists/pharmacology , GABA Antagonists/pharmacology , Glycine Agents/pharmacology , In Vitro Techniques , Interneurons/classification , Interneurons/drug effects , Interneurons/radiation effects , Mecamylamine/pharmacology , Nerve Net/cytology , Neural Networks, Computer , Nicotinic Antagonists/pharmacology , Spinal Cord/embryology , Spinal Nerve Roots/drug effects , Spinal Nerve Roots/embryology , Spinal Nerve Roots/physiology , Strychnine/pharmacology
14.
Neuron ; 37(3): 403-15, 2003 Feb 06.
Article in English | MEDLINE | ID: mdl-12575949

ABSTRACT

Spinal motor neurons must extend their axons into the periphery through motor exit points (MEPs), but their cell bodies remain within spinal motor columns. It is not known how this partitioning is established in development. We show here that motor neuron somata are confined to the CNS by interactions with a neural crest subpopulation, boundary cap (BC) cells that prefigure the sites of spinal MEPs. Elimination of BC cells by surgical or targeted genetic ablation does not perturb motor axon outgrowth but results in motor neuron somata migrating out of the spinal cord by translocating along their axons. Heterologous neural crest grafts in crest-ablated embryos stop motor neuron emigration. Thus, before the formation of a mature transitional zone at the MEP, BC cells maintain a cell-tight boundary that allows motor axons to cross but blocks neuron migration.


Subject(s)
Motor Neurons/physiology , Neural Crest/cytology , Neural Crest/embryology , Spinal Cord/cytology , Spinal Cord/embryology , Transcription Factors , Animals , Axons/physiology , Cell Movement/physiology , Chick Embryo , DNA-Binding Proteins/genetics , Denervation , Mice , Mice, Mutant Strains , Microsurgery , Motor Neurons/ultrastructure , Neural Crest/transplantation , PAX3 Transcription Factor , Paired Box Transcription Factors , Quail , Spinal Nerve Roots/cytology , Spinal Nerve Roots/embryology
15.
J Neurosci ; 22(11): 4509-21, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12040058

ABSTRACT

The present study uses the embryonic chick to examine in vivo the mechanisms and regulation of Schwann cell programmed cell death (PCD) in spinal and cranial peripheral nerves. Schwann cells are highly dependent on the presence of axons for survival because the in ovo administration of NMDA, which excitotoxically eliminates motoneurons and their axons by necrosis, results in a significant increase in apoptotic Schwann cell death. Additionally, pharmacological and surgical manipulation of axon numbers also affects the relative amounts of Schwann cell PCD. Schwann cells undergoing both normal and induced PCD display an apoptotic-like cell death, using a caspase-dependent pathway. Furthermore, axon elimination results in upregulation of the p75 and platelet-derived growth factor receptors in mature Schwann cells within the degenerating ventral root. During early development, Schwann cells are also dependent on axon-derived mitogens; the loss of axons results in a decrease in Schwann cell proliferation. Axon removal during late embryonic stages, however, elicits an increase in proliferation, as is expected from these more differentiated Schwann cells. In rodents, Schwann cell survival is regulated by glial growth factor (GGF), a member of the neuregulin family of growth factors. GGF administration to chick embryos selectively rescued Schwann cells during both normal PCD and after the loss of axons, whereas other trophic factors tested had no effect on Schwann cell survival. In conclusion, avian Schwann cells exhibit many similarities to mammalian Schwann cells in terms of their dependence on axon-derived signals during early and later stages of development.


Subject(s)
Apoptosis/physiology , Axons/physiology , Neuregulin-1/metabolism , Schwann Cells/cytology , Animals , Axons/ultrastructure , Caspase Inhibitors , Cell Division/physiology , Chick Embryo , Cysteine Proteinase Inhibitors/pharmacology , N-Methylaspartate/pharmacology , Neuregulins/metabolism , Oculomotor Nerve/cytology , Oculomotor Nerve/drug effects , Oculomotor Nerve/embryology , Peripheral Nerves/cytology , Peripheral Nerves/drug effects , Peripheral Nerves/embryology , Receptor, Nerve Growth Factor/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Schwann Cells/drug effects , Schwann Cells/ultrastructure , Signal Transduction , Spinal Nerve Roots/cytology , Spinal Nerve Roots/drug effects , Spinal Nerve Roots/embryology , Up-Regulation/physiology
16.
Eur J Neurosci ; 14(5): 809-20, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11576185

ABSTRACT

Using a multiple-site optical recording technique with a voltage-sensitive dye, we found that widely spreading depolarization waves were evoked by dorsal root stimulation in embryonic chick spinal cords. Spatiotemporal maps of the depolarization waves showed that the signals were mainly distributed in the ventral half of the slice, with the highest activity in the ventrolateral area. The propagation velocity of the waves was estimated to be in the order of mm/s. Depolarization waves were evoked in the ventral root-cut preparation, but not in the dorsal root-cut preparation, suggesting that the wave was triggered by synaptic inputs from the primary afferents, and that activation of the motoneurons was not essential for wave generation. In intact spinal cord-brain preparations, the depolarization wave propagated rostrally and caudally for a distance of several spinal segments in normal Ringer's solution. In a Mg(2+)-free solution, the amplitude and extent of the signals were markedly enhanced, and the depolarization wave triggered in the cervical spinal cord propagated to the brainstem and the cerebellum. The depolarization wave demonstrated here had many similarities with the vagus nerve-evoked depolarization wave reported previously. The results suggest that functional cell-to-cell communication systems mediated by the depolarization wave are widely generated in the embryonic central nervous system, and could play a role in large-scale coactivation of the neurons in the spinal cord and brain.


Subject(s)
Brain Stem/physiology , Cerebellum/physiology , Rhodanine/analogs & derivatives , Spinal Cord/physiology , Animals , Brain Mapping , Brain Stem/cytology , Brain Stem/embryology , Cell Communication/physiology , Cerebellum/cytology , Cerebellum/embryology , Chick Embryo , Coloring Agents , Electric Stimulation , Motor Neurons/physiology , Neural Pathways , Spinal Cord/cytology , Spinal Cord/embryology , Spinal Nerve Roots/cytology , Spinal Nerve Roots/embryology , Spinal Nerve Roots/physiology , Spinal Nerves/cytology , Spinal Nerves/embryology , Spinal Nerves/physiology , Thiazolidines , Vagus Nerve/cytology , Vagus Nerve/embryology , Vagus Nerve/physiology
17.
Eur. j. anat ; 5(1): 1-9, mayo 2001. ilus, tab
Article in En | IBECS | ID: ibc-15536

ABSTRACT

Neuronal maturation in the central nervous system, as well as in some cells deriving from neural crest, is accompanied by a swich in the expression of cytoskeletal intermediate filament proteins. Whether this occurs in humans and the exact timing of this change in human dorsal root and sympathetic ganglia are matters still open to debate. The present study was designed to analyze these issues in human embryos (estimated gestational age -e.g.a.- ranging between 6 and 12 weeks), as well as the possible co-expression of more than one intermediate filament protein in both embryos and adults. A panel of commercially available antibodies against vimentin, glial fibrillary acidic protein and neurofilament proteins was used. Glial fibrillary acidic protein was consistently absent in both developing and adult dorsal root or sympathetic ganglia. Conversely, embryonic neurons, satellite glial cells and Schwann cells displayed vimentin immunoreactivity. The number of vimentin immunoreactive neurons decreased progressively, and it was absent from neurons by 12 weeks e.g.a., while it persisted in satellite glial and Schwann cells. By adulthood, the pattern of distribution was identical. The occurrence of neurofilament proteins in peripheral neurons was a regular feature from early developmental stages to adulthood, and a time-dependent increase in the percentage of neurons containing phosphorylated neurofilaments was observed. The present results demonstrate that developing human dorsal root and sympathetic ganglion neurons co-express vimentin and neurofilaments for a short time, but that the intermediate filaments for mature neurons are neurofilaments. Our findings also show that co-expression or a switch in the expression of intermediate filament proteins do not occur in satellite glial cells or Schwann cells, which normally contain vimentin and not glial fibrillary acidic protein (AU)


La maduración neuronal en poblaciones neuronales discretas del sistema central nervioso, así como en algunas células que derivan de la cresta neural es acompañada de un cambio en la expresión de las proteínas citoesqueléticas de filamentos intermedios. El que esto ocurre en los seres humanos y la cronología exacta de este cambio en los ganglios de la raíz dorsal y simpáticos no se ha dilucidado todavía. El presente estudio fue diseñado para analizar estos problemas en embriones humanos (edad gestacional estimada –e.g.e- en el intervalo entre 6 y 12 semanas), así como la posible co-expresión de más de una proteína de filamentos intermedios, tanto en embriones como en adultos. Se utilizó un panel de anticuerpos comerciales frente a la vimentina, a la proteína fibrilar glial ácida, y a las proteínas de los neurofilamentos. La proteína fibrilar glial ácida estaba consistentemente ausente de los ganglios de la raíz dorsal y simpáticos, tanto en el adulto como en embriones en desarrollo. Por el contrario, las neuronas embrionarias, las células gliales satélites y las células de Schwann exhibían inmunoreactividad frente a la vimentina. El número de neuronas con inmunoreactividad para la vimentina descendió progresivamente y este compuesto no se detectaba en las neuronas a una e.g.e. de 12 semanas, mientras que persistía en las células Schwann. Llegada la edad adulta, el patrón de distribución era idéntico. La presencia de proteínas de neurofilamentos en las neuronas periféricas se apreció como una característica consistente a partir de las fases tempranas del desarrollo hasta la edad adulta, observándose un descenso dependiente del tiempo en el porcentaje de neuronas que contenían neurofilamentos fosforilados. Los resultados aquí obtenidos demuestran que las neuronas de los ganglios de la raíz dorsal y simpáticos humanos en desarrollo co-expresan la vimentina y los neurofilamentos durante un periodo corto de tiempo, pero los filamentos intermedios de las neuronas maduras son neurofilamentos. Nuestros hallazgos también muestran que la co-expresión o un cambio en la expresión de las proteínas de los filamentos intermedios no ocurren en las células gliales ni en las de Schwann, las cuales normalmente contienen vimentina y no la proteína fibrilar glial ácida (AU)


Subject(s)
Adult , Humans , Intermediate Filament Proteins/analysis , Ganglia, Sympathetic/chemistry , Ganglia, Sympathetic/embryology , Spinal Nerve Roots/chemistry , Spinal Nerve Roots/embryology , Neurons/chemistry , Vimentin/analysis
18.
Dev Biol ; 238(1): 47-63, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11783993

ABSTRACT

CNS glia have integral roles in directing axon migration of both vertebrates and insects. In contrast, very little is known about the roles of PNS glia in axonal pathfinding. In vertebrates and Drosophila, anatomical evidence shows that peripheral glia prefigure the transition zones through which axons migrate into and out of the CNS. Therefore, peripheral glia could guide axons at the transition zone. We used the Drosophila model system to test this hypothesis by ablating peripheral glia early in embryonic neurodevelopment via targeted overexpression of cell death genes grim and ced-3. The effects of peripheral glial loss on sensory and motor neuron development were analyzed. Motor axons initially exit the CNS in abnormal patterns in the absence of peripheral glia. However, they must use other cues within the periphery to find their correct target muscles since early pathfinding errors are largely overcome. When peripheral glia are lost, sensory axons show disrupted migration as they travel centrally. This is not a result of motor neuron defects, as determined by motor/sensory double-labeling experiments. We conclude that peripheral glia prefigure the CNS/PNS transition zone and guide axons as they traverse this region.


Subject(s)
Axons/metabolism , Central Nervous System/physiology , Drosophila Proteins , Neuroglia/physiology , Neurons/physiology , Peripheral Nervous System/physiology , Spinal Cord/physiology , Animals , Caenorhabditis elegans Proteins , Caspases/genetics , Caspases/metabolism , Cell Movement , Cell Nucleus/metabolism , Central Nervous System/embryology , Drosophila , Immunohistochemistry , Microscopy, Confocal , Models, Biological , Mutation , Neuroglia/metabolism , Neurons/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Peripheral Nervous System/embryology , Spinal Cord/embryology , Spinal Nerve Roots/embryology , Spinal Nerve Roots/physiology , Time Factors , beta-Galactosidase/metabolism
19.
Glia ; 32(1): 71-83, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10975912

ABSTRACT

Some years ago we showed that in the adult rat and mouse neuropeptide Y (NPY) is expressed by olfactory ensheathing cells, a special type of glial cell involved in guiding of the continuously renewing olfactory axons. In the present study, using immunohistochemistry combined with confocal laser scanning microscopy, and mRNA in situ hybridization, we have analyzed whether NPY is also expressed in other axon-related glial cell systems during prenatal development. NPY was found to be expressed in the dorsal and ventral rootlets along the spinal cord from E13 and onward and in the rootlets of the cranial nerves and in the sensory ganglia from E14 and onward. In some cases, NPY-immunoreactivity (IR) was also found along peripheral nerves. NPY-IR was expressed in a dot-like fashion, similar to the NPY expression observed in olfactory ensheathing cells. At E18 the NPY-immunoreactive dots had disappeared from almost all ganglia and rootlets, and only in the most central part of the rootlets some weak dot-like NPY-IR was observed. At E20 it had disappeared completely from all rootlets and nerves, except the olfactory nerve. Most of the dot-like NPY-IR did not co-localize with the neuronal marker PGP 9.5. Based on its spatiotemporal expression, it is concluded that NPY is expressed by Schwann cell precursors. NPY expressed by Schwann cell precursors might have a role in axonal growth or axonal guidance, or both.


Subject(s)
Neuropeptide Y/metabolism , Peripheral Nervous System/embryology , Peripheral Nervous System/metabolism , Schwann Cells/metabolism , Stem Cells/metabolism , Age Factors , Animals , Cranial Nerves/cytology , Cranial Nerves/embryology , Cranial Nerves/metabolism , Female , Fetus , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Ganglia, Spinal/metabolism , Peripheral Nervous System/cytology , Pregnancy , Rats , Rats, Sprague-Dawley , Schwann Cells/cytology , Spinal Nerve Roots/cytology , Spinal Nerve Roots/embryology , Spinal Nerve Roots/metabolism , Stem Cells/cytology , Thiolester Hydrolases/metabolism , Ubiquitin Thiolesterase
20.
J Cell Biol ; 148(5): 1035-46, 2000 Mar 06.
Article in English | MEDLINE | ID: mdl-10704452

ABSTRACT

Neuregulin-1 provides an important axonally derived signal for the survival and growth of developing Schwann cells, which is transmitted by the ErbB2/ErbB3 receptor tyrosine kinases. Null mutations of the neuregulin-1, erbB2, or erbB3 mouse genes cause severe deficits in early Schwann cell development. Here, we employ Cre-loxP technology to introduce erbB2 mutations late in Schwann cell development, using a Krox20-cre allele. Cre-mediated erbB2 ablation occurs perinatally in peripheral nerves, but already at E11 within spinal roots. The mutant mice exhibit a widespread peripheral neuropathy characterized by abnormally thin myelin sheaths, containing fewer myelin wraps. In addition, in spinal roots the Schwann cell precursor pool is not correctly established. Thus, the Neuregulin signaling system functions during multiple stages of Schwann cell development and is essential for correct myelination. The thickness of the myelin sheath is determined by the axon diameter, and we suggest that trophic signals provided by the nerve determine the number of times a Schwann cell wraps an axon.


Subject(s)
Genes, erbB-2/genetics , Myelin Sheath/metabolism , Peripheral Nervous System Diseases/genetics , Schwann Cells/metabolism , Stem Cells/metabolism , Viral Proteins , Animals , Axons/ultrastructure , Cell Count , DNA-Binding Proteins/genetics , Early Growth Response Protein 2 , Gene Targeting , Integrases/genetics , Mice , Mice, Neurologic Mutants , Mutagenesis , Myelin Sheath/genetics , Myelin Sheath/ultrastructure , Neuregulin-1/metabolism , Peripheral Nervous System Diseases/etiology , Recombination, Genetic , Schwann Cells/cytology , Schwann Cells/ultrastructure , Sciatic Nerve/growth & development , Sciatic Nerve/pathology , Sciatic Nerve/ultrastructure , Signal Transduction/genetics , Spinal Nerve Roots/embryology , Spinal Nerve Roots/pathology , Stem Cells/cytology , Stem Cells/ultrastructure , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...