Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Development ; 145(3)2018 02 08.
Article in English | MEDLINE | ID: mdl-29439133

ABSTRACT

Genetic factors underlying the human limb abnormality congenital talipes equinovarus ('clubfoot') remain incompletely understood. The spontaneous autosomal recessive mouse 'peroneal muscular atrophy' mutant (PMA) is a faithful morphological model of human clubfoot. In PMA mice, the dorsal (peroneal) branches of the sciatic nerves are absent. In this study, the primary developmental defect was identified as a reduced growth of sciatic nerve lateral motor column (LMC) neurons leading to failure to project to dorsal (peroneal) lower limb muscle blocks. The pma mutation was mapped and a candidate gene encoding LIM-domain kinase 1 (Limk1) identified, which is upregulated in mutant lateral LMC motor neurons. Genetic and molecular analyses showed that the mutation acts in the EphA4-Limk1-Cfl1/cofilin-actin pathway to modulate growth cone extension/collapse. In the chicken, both experimental upregulation of Limk1 by electroporation and pharmacological inhibition of actin turnover led to defects in hindlimb spinal motor neuron growth and pathfinding, and mimicked the clubfoot phenotype. The data support a neuromuscular aetiology for clubfoot and provide a mechanistic framework to understand clubfoot in humans.


Subject(s)
Charcot-Marie-Tooth Disease/embryology , Clubfoot/embryology , Clubfoot/genetics , Lim Kinases/genetics , Mutation , Animals , Axons , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Chick Embryo , Chromosome Mapping , Clubfoot/pathology , Disease Models, Animal , Female , Hindlimb/abnormalities , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Motor Neurons/pathology , Muscle, Skeletal/abnormalities , Muscle, Skeletal/innervation , Peroneal Nerve/abnormalities , Phenotype , Pregnancy , Receptor, EphA4/deficiency , Receptor, EphA4/genetics , Sciatic Nerve/abnormalities , Up-Regulation
2.
J Sleep Res ; 26(5): 539-546, 2017 10.
Article in English | MEDLINE | ID: mdl-28488395

ABSTRACT

Slow waves occurring during non-rapid eye movement sleep have been associated with neurobehavioural performance and memory. In addition, the duration of previous wakefulness and sleep impacts characteristics of these slow waves. However, molecular mechanisms regulating the dynamics of slow-wave characteristics remain poorly understood. The EphA4 receptor regulates glutamatergic transmission and synaptic plasticity, which have both been linked to sleep slow waves. To investigate if EphA4 regulates slow-wave characteristics during non-rapid eye movement sleep, we compared individual parameters of slow waves between EphA4 knockout mice and wild-type littermates under baseline conditions and after a 6-h sleep deprivation. We observed that, compared with wild-type mice, knockout mice display a shorter duration of positive and negative phases of slow waves under baseline conditions and after sleep deprivation. However, the mutation did not change slow-wave density, amplitude and slope, and did not affect the sleep deprivation-dependent changes in slow-wave characteristics, suggesting that EphA4 is not involved in the response to elevated sleep pressure. Our present findings suggest a role for EphA4 in shaping cortical oscillations during sleep that is independent from sleep need.


Subject(s)
Receptor, EphA4/deficiency , Receptor, EphA4/genetics , Sleep/physiology , Animals , Electroencephalography , Male , Mice , Mice, Knockout , Receptor, EphA4/metabolism , Sleep/genetics , Sleep Deprivation/genetics , Sleep Deprivation/physiopathology , Time Factors , Wakefulness/genetics , Wakefulness/physiology
3.
Sleep ; 39(3): 613-24, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26612390

ABSTRACT

STUDY OBJECTIVES: Optimal sleep is ensured by the interaction of circadian and homeostatic processes. Although synaptic plasticity seems to contribute to both processes, the specific players involved are not well understood. The EphA4 tyrosine kinase receptor is a cell adhesion protein regulating synaptic plasticity. We investigated the role of EphA4 in sleep regulation using electrocorticography in mice lacking EphA4 and gene expression measurements. METHODS: EphA4 knockout (KO) mice, Clock(Δ19/Δ19) mutant mice and littermates, C57BL/6J and CD-1 mice, and Sprague-Dawley rats were studied under a 12 h light: 12 h dark cycle, under undisturbed conditions or 6 h sleep deprivation (SLD), and submitted to a 48 h electrophysiological recording and/or brain sampling at different time of day. RESULTS: EphA4 KO mice showed less rapid eye movement sleep (REMS), enhanced duration of individual bouts of wakefulness and nonrapid eye movement sleep (NREMS) during the light period, and a blunted daily rhythm of NREMS sigma activity. The NREMS delta activity response to SLD was unchanged in EphA4 KO mice. However, SLD increased EphA4 expression in the thalamic/hypothalamic region in C57BL/6J mice. We further show the presence of E-boxes in the promoter region of EphA4, a lower expression of EphA4 in Clock mutant mice, a rhythmic expression of EphA4 ligands in several brain areas, expression of EphA4 in the suprachiasmatic nuclei of the hypothalamus (SCN), and finally an unchanged number of cells expressing Vip, Grp and Avp in the SCN of EphA4 KO mice. CONCLUSIONS: Our results suggest that EphA4 is involved in circadian sleep regulation.


Subject(s)
Circadian Rhythm/physiology , Receptor, EphA4/metabolism , Sleep Deprivation/physiopathology , Sleep/physiology , Animals , CLOCK Proteins/genetics , Circadian Rhythm/genetics , Darkness , Electrocorticography , Electrophysiological Phenomena , Homeostasis , Light , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity , Promoter Regions, Genetic/genetics , Rats , Rats, Sprague-Dawley , Receptor, EphA4/biosynthesis , Receptor, EphA4/deficiency , Receptor, EphA4/genetics , Sleep/genetics , Sleep Deprivation/genetics , Sleep, REM/genetics , Sleep, REM/physiology , Suprachiasmatic Nucleus/metabolism , Thalamus/metabolism , Time Factors , Wakefulness/genetics , Wakefulness/physiology
4.
Development ; 142(1): 140-50, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25480914

ABSTRACT

The phenotype of excitatory cerebral cortex neurons is specified at the progenitor level, orchestrated by various intrinsic and extrinsic factors. Here, we provide evidence for a subcortical contribution to cortical progenitor regulation by thalamic axons via ephrin A5-EphA4 interactions. Ephrin A5 is expressed by thalamic axons and represents a high-affinity ligand for EphA4 receptors detected in cortical precursors. Recombinant ephrin A5-Fc protein, as well as ephrin A ligand-expressing, thalamic axons affect the output of cortical progenitor division in vitro. Ephrin A5-deficient mice show an altered division mode of radial glial cells (RGCs) accompanied by increased numbers of intermediate progenitor cells (IPCs) and an elevated neuronal production for the deep cortical layers at E13.5. In turn, at E16.5 the pool of IPCs is diminished, accompanied by reduced rates of generated neurons destined for the upper cortical layers. This correlates with extended infragranular layers at the expense of superficial cortical layers in adult ephrin A5-deficient and EphA4-deficient mice. We suggest that ephrin A5 ligands imported by invading thalamic axons interact with EphA4-expressing RGCs, thereby contributing to the fine-tuning of IPC generation and thus the proper neuronal output for cortical layers.


Subject(s)
Cerebral Cortex/cytology , Ephrin-A5/metabolism , Neurons, Afferent/cytology , Neurons, Afferent/metabolism , Receptor, EphA4/metabolism , Stem Cells/metabolism , Thalamus/cytology , Animals , Axons/metabolism , Cell Count , Cell Division , Embryo, Mammalian/cytology , Ependymoglial Cells/cytology , Ependymoglial Cells/metabolism , Ephrin-A5/deficiency , Ligands , Mice, Inbred C57BL , Neurogenesis , Receptor, EphA4/deficiency , Signal Transduction , Stem Cells/cytology , Thalamus/embryology , Thalamus/metabolism
5.
Birth Defects Res A Clin Mol Teratol ; 100(6): 507-11, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24753315

ABSTRACT

BACKGROUND: Interstitial 2q36 deletion is a rare event. Only two previously published cases of 2q36 deletions were characterized using array-CGH. This is the first case diagnosed prenatally. METHODS: We report on the prenatal diagnosis of a 2q36.1q36.3 interstitial deletion in a fetus with facial dysmorphism, spina bifida, and cleft palate. RESULTS: Array-CGH analysis revealed a 5.6 Mb interstitial deletion of the long arm of chromosome 2q36.1q36.3, including the PAX3 and EPHA4 genes. CONCLUSION: The present study reinforces the hypothesis that PAX3 haploinsufficiency may be associated with neural tube defects in humans and suggests that the EPHA4 gene might be implicated during palate development. This report also illustrates the added value of array-CGH to detect cryptic chromosomal imbalances in malformed fetuses and to improve genetic counseling prenatally.


Subject(s)
Abnormalities, Multiple/genetics , Chromosome Deletion , Chromosomes, Human, Pair 2 , Cleft Palate/genetics , Paired Box Transcription Factors/genetics , Receptor, EphA4/genetics , Spinal Dysraphism/genetics , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/pathology , Adult , Cleft Palate/diagnosis , Cleft Palate/pathology , Comparative Genomic Hybridization , Female , Fetus , Gene Expression Regulation, Developmental , Humans , Karyotyping , PAX3 Transcription Factor , Paired Box Transcription Factors/deficiency , Pregnancy , Prenatal Diagnosis , Receptor, EphA4/deficiency , Spinal Dysraphism/diagnosis , Spinal Dysraphism/pathology
6.
J Comp Neurol ; 520(8): 1702-20, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22121026

ABSTRACT

The guidance of axonal projections to ipsilateral and contralateral regions is essential for integration of bilateral sensory information and coordination of movement. In the development of olivocerebellar projections, newborn neurons of inferior olivary (IO) nuclei ventrally migrate from the hindbrain rhombic lip to the floor plate (FP). The cell bodies of IO neurons cannot cross the FP but their axons can, and thus IO neurons project their axons only to the contralateral cerebellar cortex. The molecular mechanisms determining the contralateral axonal projections of IO neurons, however, are obscure. The IO neurons and their axons express EphA4, whereas the FP expresses an EphA4 ligand, EphrinB3, from embryonic day 12.5. Therefore, we tested whether EphA4-deficient mice (EphA4(-/-) ) would show impairment in the development of olivocerebellar projections. We found that, in EphA4(-/-) embryos, some of the IO neurons projected their axons to the ipsilateral cerebellar cortex because the cell bodies of the IO neurons abnormally crossed the FP. Furthermore, even in adults, EphA4(-/-) cerebella were bilaterally innervated by unilateral IO subnuclei. These observations indicate that EphA4 is involved in the contralateral axonal projections of IO neurons by preventing their cell bodies from crossing the midline FP.


Subject(s)
Cell Movement/physiology , Functional Laterality/physiology , Neurogenesis/physiology , Neurons/cytology , Olivary Nucleus/embryology , Receptor, EphA4/metabolism , Animals , Axons/metabolism , Axons/ultrastructure , Body Patterning/physiology , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred ICR , Mice, Knockout , Neurons/metabolism , Olivary Nucleus/growth & development , Receptor, EphA4/deficiency
7.
J Neurosci ; 30(47): 16015-24, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-21106840

ABSTRACT

There is growing evidence that astrocytes play critical roles in neuron-glial interactions at the synapse. Astrocytes are believed to regulate presynaptic and postsynaptic structures and functions, in part, by the release of gliotransmitters such as glutamate, ATP, and d-serine; however, little is known of how neurons and astrocytes communicate to regulate these processes. Here, we investigated a family of transmembrane proteins called ephrinBs and Eph receptors that are expressed in the synapse and are known to regulate synaptic transmission and plasticity. In addition to their presence on CA1 hippocampal neurons, we determined that ephrins and Eph receptors are also expressed on hippocampal astrocytes. Stimulation of hippocampal astrocytes with soluble ephrinB3, known to be expressed on CA1 postsynaptic dendrites, enhanced d-serine synthesis and release in culture. Conversely, ephrinB3 had no effect on d-serine release from astrocytes deficient in EphB3 and EphA4, which are the primary receptors for ephrinB3. Eph receptors mediate this response through interactions with PICK1 (protein interacting with C-kinase) and by dephosphorylating protein kinase C α to activate the conversion of l-serine to d-serine by serine racemase. These findings are supported in vivo, where reduced d-serine levels and synaptic transmissions are observed in the absence of EphB3 and EphA4. These data support a role for ephrins and Eph receptors in regulating astrocyte gliotransmitters, which may have important implications on synaptic transmission and plasticity.


Subject(s)
Astrocytes/metabolism , Ephrin-B3/physiology , Serine/biosynthesis , Serine/metabolism , Animals , Cells, Cultured , Ephrin-B3/deficiency , Hippocampus/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Neuronal Plasticity/genetics , Protein Biosynthesis/genetics , Receptor, EphA4/biosynthesis , Receptor, EphA4/deficiency , Receptor, EphA4/physiology , Serine/analogs & derivatives , Stereoisomerism , Synaptic Transmission/genetics
8.
Nat Neurosci ; 12(10): 1285-92, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19734893

ABSTRACT

Astrocytes are critical participants in synapse development and function, but their role in synaptic plasticity is unclear. Eph receptors and their ephrin ligands have been suggested to regulate neuron-glia interactions, and EphA4-mediated ephrin reverse signaling is required for synaptic plasticity in the hippocampus. Here we show that long-term potentiation (LTP) at the CA3-CA1 synapse is modulated by EphA4 in the postsynaptic CA1 cell and by ephrin-A3, a ligand of EphA4 that is found in astrocytes. Lack of EphA4 increased the abundance of glial glutamate transporters, and ephrin-A3 modulated transporter currents in astrocytes. Pharmacological inhibition of glial glutamate transporters rescued the LTP defects in EphA4 (Epha4) and ephrin-A3 (Efna3) mutant mice. Transgenic overexpression of ephrin-A3 in astrocytes reduces glutamate transporter levels and produces focal dendritic swellings possibly caused by glutamate excitotoxicity. These results suggest that EphA4/ephrin-A3 signaling is a critical mechanism for astrocytes to regulate synaptic function and plasticity.


Subject(s)
Ephrin-A3/metabolism , Glutamic Acid/metabolism , Long-Term Potentiation/physiology , Neuroglia/physiology , Neurons/physiology , Receptor, EphA4/metabolism , Animals , Animals, Newborn , Aspartic Acid/analogs & derivatives , Aspartic Acid/pharmacology , Biophysics , Disease Models, Animal , Electric Stimulation/methods , Ephrin-A3/genetics , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Postsynaptic Potentials/genetics , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/genetics , Hippocampus/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques , Patch-Clamp Techniques/methods , Pentylenetetrazole , Receptor, EphA4/deficiency , Seizures/chemically induced , Seizures/genetics , Seizures/physiopathology , Signal Transduction/physiology , Synapses/physiology , Up-Regulation/genetics
9.
Development ; 136(14): 2467-76, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19542359

ABSTRACT

Eph receptors are widely expressed during cerebral cortical development, yet a role for Eph signaling in the generation of cells during corticogenesis has not been shown. Cortical progenitor cells selectively express one receptor, EphA4, and reducing EphA4 signaling in cultured progenitors suppressed proliferation, decreasing cell number. In vivo, EphA4(-/-) cortex had a reduced area, fewer cells and less cell division compared with control cortex. To understand the effects of EphA4 signaling in corticogenesis, EphA4-mediated signaling was selectively depressed or elevated in cortical progenitors in vivo. Compared with control cells, cells with reduced EphA4 signaling were rare and mitotically inactive. Conversely, overexpression of EphA4 maintained cells in their progenitor states at the expense of subsequent maturation, enlarging the progenitor pool. These results support a role for EphA4 in the autonomous promotion of cell proliferation during corticogenesis. Although most ephrins were undetectable in cortical progenitors, ephrin B1 was highly expressed. Our analyses demonstrate that EphA4 and ephrin B1 bind to each other, thereby initiating signaling. Furthermore, overexpression of ephrin B1 stimulated cell division of neighboring cells, supporting the hypothesis that ephrin B1-initiated forward signaling of EphA4 promotes cortical cell division.


Subject(s)
Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Receptor, EphA4/metabolism , Animals , Cell Communication , Cell Proliferation , Cells, Cultured , Cerebral Cortex/cytology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Ephrin-B1/genetics , Ephrin-B1/metabolism , Female , Gene Expression Regulation, Developmental , Ligands , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Neurological , Pregnancy , Receptor, EphA4/deficiency , Receptor, EphA4/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
10.
Development ; 135(19): 3281-90, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18755772

ABSTRACT

Brain structures, whether mature or developing, display a wide diversity of pattern and shape, such as layers, nuclei or segments. The striatum in the mammalian forebrain displays a unique mosaic organization (subdivided into two morphologically and functionally defined neuronal compartments: the matrix and the striosomes) that underlies important functional features of the basal ganglia. Matrix and striosome neurons are generated sequentially during embryonic development, and segregate from each other to form a mosaic of distinct compartments. However, the molecular mechanisms that underlie this time-dependent process of neuronal segregation remain largely unknown. Using a novel organotypic assay, we identified ephrin/Eph family members as guidance cues that regulate matrix/striosome compartmentalization. We found that EphA4 and its ephrin ligands displayed specific temporal patterns of expression and function that play a significant role in the spatial segregation of matrix and striosome neurons. Analysis of the striatal patterning in ephrin A5/EphA4 mutant mice further revealed the requirement of EphA4 signalling for the proper sorting of matrix and striosome neuronal populations in vivo. These data constitute the first identification of genes involved in striatal compartmentalization, and reveal a novel mechanism by which the temporal control of guidance cues enables neuronal segregation, and thereby the generation of complex cellular patterns in the brain.


Subject(s)
Body Patterning/physiology , Corpus Striatum/embryology , Corpus Striatum/metabolism , Ephrin-A5/metabolism , Receptor, EphA4/metabolism , Animals , Body Patterning/genetics , Cell Adhesion , Corpus Striatum/cytology , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Ephrin-A5/deficiency , Ephrin-A5/genetics , Female , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Models, Neurological , Pregnancy , Receptor, EphA4/deficiency , Receptor, EphA4/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
11.
J Neurochem ; 106(2): 682-95, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18410519

ABSTRACT

The ephrin receptors EphA4 and EphB2 have been implicated in synaptogenesis and long-term potentiation in the cerebral cortex and hippocampus, where they are generally viewed as post-synaptic receptors. To determine the precise distribution of EphA4 and EphB2 in mature brain synapses, we used subcellular fractionation and electron microscopy to examine the adult mouse forebrain/midbrain. EphA4 and EphB2 were both enriched in microsomes and synaptosomes. In synaptosomes, they were present in the membrane and the synaptic vesicle fractions. While EphA4 was tightly associated with PSD-95-enriched post-synaptic density fractions, EphB2 was easily extracted with detergents. In contrast, both receptors were found in the pre-synaptic active zone fraction. By electron microscopy, EphA4 was mainly detected in axon terminals, whereas EphB2 was more frequently detected in large dendritic shafts, in the hippocampus and cerebral cortex. However, in the ventrobasal thalamus, EphB2 was detected most frequently in axon terminals and thin dendritic shafts. The localization of EphA4 and EphB2 in multiple compartments of neurons and synaptic junctions suggests that they interact with several distinct scaffolding proteins and play diverse roles at synapses.


Subject(s)
Presynaptic Terminals/metabolism , Prosencephalon/ultrastructure , Receptor, EphA4/metabolism , Receptor, EphB2/metabolism , Synapses/metabolism , Animals , Disks Large Homolog 4 Protein , Guanylate Kinases , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Immunoelectron/methods , Neurons/metabolism , Neurons/ultrastructure , Presynaptic Terminals/ultrastructure , Receptor, EphA4/deficiency , Subcellular Fractions/metabolism , Subcellular Fractions/ultrastructure , Synapses/ultrastructure , Synaptosomes/metabolism , Synaptosomes/ultrastructure
12.
J Comp Neurol ; 504(5): 508-18, 2007 Oct 10.
Article in English | MEDLINE | ID: mdl-17702003

ABSTRACT

Axonal selection of ipsilateral and/or contralateral targets is essential for integrating bilateral sensory information and for coordinated movement. The molecular processes that determine ipsilateral and contralateral target choice are not fully understood. We examined this target selection in the developing auditory brainstem. Ventral cochlear nucleus (VCN) axons normally project to the medial nucleus of the trapezoid body (MNTB) only on the contralateral side. However, after unilateral removal of cochlear input in neonates, we found that axons from the unoperated VCN sprout and project to MNTB bilaterally. We found that EphA4 is expressed in the mouse auditory brainstem during development and during a sensitive period for ipsilateral sprouting, so we hypothesized that deletion of the Eph receptor EphA4 would impair target selection in these auditory pathways. Lipophilic dyes were used to evaluate quantitatively the brainstem projections in wild-type and EphA4-null mice. VCN-MNTB projections in EphA4-null mice were strictly contralateral, as in wild-type mice. However, after deafferentation, EphA4-null mice had a significant, threefold increase in the proportion of axons from the intact VCN that sprouted into ipsilateral MNTB compared with wild-type mice. Heterozygous mice had a twofold increase in these projections. These results demonstrate that EphA4 influences auditory brainstem circuitry selectively in response to deafferentation. Although this axon guidance molecule is not by itself necessary for appropriate target choice during normal development, it is a strong determinant of ipsilateral vs. contralateral target choice during deafferentation-induced plasticity.


Subject(s)
Auditory Pathways/physiopathology , Brain Stem/pathology , Denervation , Functional Laterality/physiology , Nerve Regeneration/genetics , Receptor, EphA4/deficiency , Amino Acids/metabolism , Animals , Animals, Newborn , Axons/physiology , Cochlea/injuries , Cochlea/surgery , Galactosides , Gene Expression Regulation, Developmental/physiology , Indoles , Mice , Mice, Knockout , Neuronal Plasticity/physiology , Time Factors
13.
J Immunol ; 177(2): 804-13, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16818734

ABSTRACT

In the present work, we have demonstrated in vivo an altered maturation of the thymic epithelium that results in defective T cell development which increases with age, in the thymus of Eph A4-deficient mice. The deficient thymi are hypocellular and show decreased proportions of double-positive (CD4+CD8+) cells which reach minimal numbers in 4-wk-old thymi. The EphA4 (-/-) phenotype correlates with an early block of T cell precursor differentiation that results in accumulation of CD44-CD25+ triple-negative cells and, sometimes, of CD44+CD25- triple-negative thymocytes as well as with increased numbers of apoptotic cells and an important reduction in the numbers of cycling thymocytes. Various approaches support a key role of the thymic epithelial cells in the observed phenotype. Thymic cytoarchitecture undergoes profound changes earlier than those found in the thymocyte maturation. Thymic cortex is extremely reduced and consists of densely packed thymic epithelial cells. Presumably the lack of forward Eph A4 signaling in the Eph A4 -/- epithelial cells affects their development and finally results in altered T cell development.


Subject(s)
Receptor, EphA4/deficiency , Receptor, EphA4/genetics , Thymus Gland/immunology , Thymus Gland/pathology , Animals , Apoptosis/genetics , Apoptosis/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Cycle/genetics , Cell Cycle/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Lymphopenia/genetics , Lymphopenia/immunology , Lymphopenia/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mice, SCID , Stromal Cells/immunology , Stromal Cells/metabolism , Stromal Cells/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Thymus Gland/metabolism
14.
J Comp Neurol ; 497(6): 864-75, 2006 Aug 20.
Article in English | MEDLINE | ID: mdl-16802330

ABSTRACT

Molecules involved in axon guidance have recently also been shown to play a role in blood vessel guidance. To examine whether axon guidance molecules, such as the EphA4 receptor tyrosine kinase, might also play a role in development of the central nervous system (CNS) vasculature and repair following CNS injury, we examined wild-type and EphA4 null mutant (-/-) mice. EphA4-/- mice exhibited an abnormal CNS vascular structure in both the cerebral cortex and the spinal cord, with disorganized branching and a 30% smaller diameter. During development, EphA4 was expressed on endothelial cells. This pattern of expression was not maintained in the adult. After spinal cord injury in wild-type mice, expression of EphA4 was markedly up-regulated on activated astrocytes, many of which were tightly associated with blood vessels. In EphA4-/- spinal cord following injury, astrocytes were not as tightly associated with blood vessels as the wild-type astrocytes. In uninjured EphA4-/- mice, the blood-brain barrier (BBB) appeared normal, but it showed prolonged leakage following spinal cord injury. These results support a role for EphA4 in CNS vascular formation and guidance during development and an additional role in BBB repair.


Subject(s)
Brain/blood supply , Brain/growth & development , Receptor, EphA4/physiology , Spinal Cord/blood supply , Spinal Cord/growth & development , Animals , Blood-Brain Barrier/growth & development , Central Nervous System/blood supply , Central Nervous System/growth & development , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, EphA4/deficiency
15.
J Neurosci ; 24(45): 10064-73, 2004 Nov 10.
Article in English | MEDLINE | ID: mdl-15537875

ABSTRACT

Spinal cord injury usually results in permanent paralysis because of lack of regrowth of damaged neurons. Here we demonstrate that adult mice lacking EphA4 (-/-), a molecule essential for correct guidance of spinal cord axons during development, exhibit axonal regeneration and functional recovery after spinal cord hemisection. Anterograde and retrograde tracing showed that axons from multiple pathways, including corticospinal and rubrospinal tracts, crossed the lesion site. EphA4-/- mice recovered stride length, the ability to walk on and climb a grid, and the ability to grasp with the affected hindpaw within 1-3 months of injury. EphA4 expression was upregulated on astrocytes at the lesion site in wild-type mice, whereas astrocytic gliosis and the glial scar were greatly reduced in lesioned EphA4-/- spinal cords. EphA4-/- astrocytes failed to respond to the inflammatory cytokines, interferon-gamma or leukemia inhibitory factor, in vitro. Neurons grown on wild-type astrocytes extended shorter neurites than on EphA4-/- astrocytes, but longer neurites when the astrocyte EphA4 was blocked by monomeric EphrinA5-Fc. Thus, EphA4 regulates two important features of spinal cord injury, axonal inhibition, and astrocytic gliosis.


Subject(s)
Astrocytes/pathology , Axons/physiology , Gliosis/genetics , Nerve Regeneration/physiology , Receptor, EphA4/physiology , Spinal Cord Injuries/physiopathology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Brain/pathology , Cell Division/drug effects , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Enzyme Activation , Immunoglobulin Fc Fragments/pharmacology , Interferon-gamma/pharmacology , Interleukin-6/pharmacology , Lameness, Animal/etiology , Lameness, Animal/physiopathology , Leukemia Inhibitory Factor , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurites/ultrastructure , Paraplegia/etiology , Paraplegia/physiopathology , Pyramidal Tracts/pathology , Receptor, EphA4/biosynthesis , Receptor, EphA4/deficiency , Receptor, EphA4/genetics , Recovery of Function , Red Nucleus/pathology , Spinal Cord Injuries/pathology , rho GTP-Binding Proteins/metabolism
16.
J Comp Neurol ; 458(1): 98-111, 2003 Mar 24.
Article in English | MEDLINE | ID: mdl-12577325

ABSTRACT

The correct assembly of the neural circuits that control movement requires the development of topographically organized pools of motoneurons within the spinal cord. The generation of a diverse array of motoneuron subtypes, which express differing transcription factors and cell-surface receptors, allows different motoneuron pools to be segregated to specific positions during development. In this investigation, we show that the Eph receptor tyrosine kinase, EphA4, appears to be important for the correct localization of a motoneuron pool to a specific position in the spinal cord. In the spinal cord of mice deficient in EphA4, the motoneuron pool that innervates the tibialis anterior muscle of the hindlimb is caudally displaced by approximately one vertebral segment. However, despite the abnormal position of the tibialis anterior motoneuron pool in the spinal cord of EphA4-deficient animals, the motoneurons of this pool still project to the tibialis anterior muscle of the hindlimb correctly. Additional analyses of other limb innervating motoneuron pools in the cervical and lumbar enlargements of the spinal cord of EphA4-deficient animals revealed them to be located in the appropriate segmental positions. Furthermore, we show that EphA4 does not appear to be important for spinal motoneuron survival as stereological quantification of the number of motoneurons present in the sciatic motoneuron pool of EphA4-deficient animals demonstrated these motoneurons to be present in the correct numbers. These observations suggest an important role for EphA4 in regulating the position of a specific motoneuron pool within the spinal cord.


Subject(s)
Motor Neurons/metabolism , Receptor, EphA4/metabolism , Spinal Cord/metabolism , Animals , Hindlimb/innervation , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal/innervation , Receptor, EphA4/deficiency , Spinal Cord/anatomy & histology , Transfection
17.
Nat Neurosci ; 6(2): 153-60, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12496762

ABSTRACT

Communication between glial cells and neurons is emerging as a critical parameter of synaptic function. However, the molecular mechanisms underlying the ability of glial cells to modify synaptic structure and physiology are poorly understood. Here we describe a repulsive interaction that regulates postsynaptic morphology through the EphA4 receptor tyrosine kinase and its ligand ephrin-A3. EphA4 is enriched on dendritic spines of pyramidal neurons in the adult mouse hippocampus, and ephrin-A3 is localized on astrocytic processes that envelop spines. Activation of EphA4 by ephrin-A3 was found to induce spine retraction, whereas inhibiting ephrin/EphA4 interactions distorted spine shape and organization in hippocampal slices. Furthermore, spine irregularities in pyramidal neurons from EphA4 knockout mice and in slices transfected with kinase-inactive EphA4 indicated that ephrin/EphA4 signaling is critical for spine morphology. Thus, our data support a model in which transient interactions between the ephrin-A3 ligand and the EphA4 receptor regulate the structure of excitatory synaptic connections through neuroglial cross-talk.


Subject(s)
Astrocytes/metabolism , Dendrites/metabolism , Ephrin-A3/metabolism , Hippocampus/metabolism , Receptor, EphA4/deficiency , Synapses/metabolism , Synaptic Transmission/genetics , Animals , Astrocytes/ultrastructure , Cell Communication/genetics , Cell Size/genetics , Dendrites/ultrastructure , Ephrin-A3/genetics , Ephrin-B1/metabolism , Ephrin-B1/pharmacology , Fluorescent Antibody Technique , Gene Expression Regulation/physiology , Hippocampus/abnormalities , Hippocampus/ultrastructure , Mice , Mice, Knockout , Neuronal Plasticity/genetics , Phosphotransferases/deficiency , Phosphotransferases/genetics , Pyramidal Cells/metabolism , Pyramidal Cells/ultrastructure , Receptor, EphA4/genetics , Receptors, Eph Family/drug effects , Receptors, Eph Family/metabolism , Recombinant Fusion Proteins , Signal Transduction/genetics , Synapses/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...