Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
2.
Sci Adv ; 8(46): eabo4552, 2022 Nov 16.
Article in English | MEDLINE | ID: mdl-36399562

ABSTRACT

During corticogenesis, dynamic regulation of apical adhesion is fundamental to generate correct numbers and cell identities. While radial glial cells (RGCs) maintain basal and apical anchors, basal progenitors and neurons detach and settle at distal positions from the apical border. Whether diffusible signals delivered from the cerebrospinal fluid (CSF) contribute to the regulation of apical adhesion dynamics remains fully unknown. Secreted class 3 Semaphorins (Semas) trigger cell responses via Plexin-Neuropilin (Nrp) membrane receptor complexes. Here, we report that unconventional Sema3-Nrp preformed complexes are delivered by the CSF from sources including the choroid plexus to Plexin-expressing RGCs via their apical endfeet. Through analysis of mutant mouse models and various ex vivo assays mimicking ventricular delivery to RGCs, we found that two different complexes, Sema3B/Nrp2 and Sema3F/Nrp1, exert dual effects on apical endfeet dynamics, nuclei positioning, and RGC progeny. This reveals unexpected balance of CSF-delivered guidance molecules during cortical development.

3.
Neurogenetics ; 22(1): 43-51, 2021 03.
Article in English | MEDLINE | ID: mdl-33415589

ABSTRACT

Pathogenic variants in L1CAM, the gene encoding the L1 cell adhesion molecule, are responsible for a wide clinical spectrum including X-linked hydrocephalus with stenosis of the Sylvius aqueduct, MASA syndrome (mental retardation, aphasia, shuffling gait, adducted thumbs), and a form of spastic paraplegia (SPG1). A moderate phenotype with mild intellectual disability (ID) and X-linked partial corpus callosum agenesis (CCA) has only been related to L1CAM in one family. We report here a second family, including 5 patients with mild to moderate ID and partial CCA without signs usually associated with L1CAM pathogenic variations (such as hydrocephalus, pyramidal syndrome, thumb adductus, aphasia). We identified a previously unreported c.3226A > C transversion leading to a p.Thr1076Pro amino acid substitution in the fifth fibronectin type III domain (FnIII) of the protein which co-segregates with the phenotype within the family. We performed in vitro assays to assess the pathogenic status of this variation. First, the expression of the novel p.Thr1076Pro mutant in COS7 cells resulted in endoplasmic reticulum (ER) retention and reduced L1CAM cell surface expression, which is expected to affect both L1CAM-mediated cell-cell adhesion and neurite growth. Second, immunoblotting techniques showed that the immature form of the L1CAM protein was increased, indicating that this variation led to a lack of maturation of the protein. ID associated with CCA is not a common clinical presentation of L1CAM pathogenic variants. Genome-wide analyses will identify such variations and it is important to acknowledge this atypical phenotype.


Subject(s)
Agenesis of Corpus Callosum/genetics , Cerebral Aqueduct/abnormalities , Genetic Diseases, X-Linked/genetics , Hydrocephalus/genetics , Intellectual Disability/genetics , Mutation/genetics , Neural Cell Adhesion Molecule L1/genetics , Agenesis of Corpus Callosum/diagnosis , Female , Gene Deletion , Genome-Wide Association Study , Humans , Intellectual Disability/diagnosis , Pedigree , Young Adult
4.
Elife ; 92020 12 21.
Article in English | MEDLINE | ID: mdl-33345773

ABSTRACT

Spinal commissural axon navigation across the midline in the floor plate requires repulsive forces from local Slit repellents. The long-held view is that Slits push growth cones forward and prevent them from turning back once they became sensitized to these cues after midline crossing. We analyzed with fluorescent reporters Slits distribution and FP glia morphology. We observed clusters of Slit-N and Slit-C fragments decorating a complex architecture of glial basal process ramifications. We found that PC2 proprotein convertase activity contributes to this pattern of ligands. Next, we studied Slit-C acting via PlexinA1 receptor shared with another FP repellent, the Semaphorin3B, through generation of a mouse model baring PlexinA1Y1815F mutation abrogating SlitC but not Sema3B responsiveness, manipulations in the chicken embryo, and ex vivo live imaging. This revealed a guidance mechanism by which SlitC constantly limits growth cone exploration, imposing ordered and forward-directed progression through aligned corridors formed by FP basal ramifications.


Subject(s)
Commissural Interneurons/physiology , Spinal Cord/growth & development , Animals , Axons/physiology , Blotting, Western , Chick Embryo , Growth Cones/physiology , Mice , Microscopy, Fluorescence , Neural Tube/embryology , Neural Tube/growth & development , Spinal Cord/embryology
5.
Cell Rep ; 29(2): 347-362.e5, 2019 Oct 08.
Article in English | MEDLINE | ID: mdl-31597096

ABSTRACT

Accurate perception of guidance cues is crucial for cell and axon migration. During initial navigation in the spinal cord, commissural axons are kept insensitive to midline repellents. Upon midline crossing in the floor plate, they switch on responsiveness to Slit and Semaphorin repulsive signals and are thus propelled away and prevented from crossing back. Whether and how the different midline repellents control specific aspects of this navigation remain to be elucidated. We set up a paradigm for live-imaging and super-resolution analysis of PlexinA1, Neuropilin-2, and Robo1/2 receptor dynamics during commissural growth cone navigation in chick and mouse embryos. We uncovered a remarkable program of sensitization to midline cues achieved by unique spatiotemporal sequences of receptor allocation at the growth-cone surface that orchestrates receptor-specific growth-cone behavior changes. This reveals post-translational mechanisms whereby coincident guidance signals are temporally resolved to allow the generation of specific guidance responses.


Subject(s)
Axons/physiology , Nerve Tissue Proteins/metabolism , Semaphorins/metabolism , Animals , Cell Membrane/metabolism , Chick Embryo , Chickens , Embryo, Mammalian/metabolism , Growth Cones/metabolism , Mice , Nerve Tissue Proteins/chemistry , Protein Domains , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Receptors, Immunologic/metabolism , Recombinant Proteins/metabolism , Time Factors , Roundabout Proteins
6.
Dev Biol ; 442(2): 249-261, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30071216

ABSTRACT

The development of the sensory nervous system is the result of fine-tuned waves of neurogenesis and apoptosis which control the appropriate number of precursors and newly generated neurons and orient them toward a specific lineage. Neurotrophins and their tyrosine-kinase receptors (RTK) orchestrate this process. They have long been in the scope of the neurotrophic theory which established that a neuron is committed to die unless a trophic factor generated by its target provides it with a survival signal. The neural death has thus always been described as a "default" program, survival being the major player to control the number of cells. New insights have been brought by the gain of function studies which recently demonstrated that TrkC (NTRK3) is a "dependence receptor" able to actively trigger apoptosis in absence of its ligand NT-3. In order to address the role of TrkC pro-apoptotic activity in the control of sensory neurons number, we generated a TrkC gene-trap mutant mice. We found out that this new murine model recapitulates the sensory phenotype of TrkC constitutive mutants, with reduced DRG size and reduced number of DRG neurons. We engineered these mice strain with a lacZ reporter in order to follow the fate of neurons committed to a TrkC lineage and observed that they are specifically protected from NT-3 mediated apoptosis in NT-3/TrkC double knock-out embryos. Finally, using a chicken model we demonstrated that silencing NT-3 emanating from the ventral neural tube induced apoptosis in the DRG anlage. This apoptosis was inhibited by silencing TrkC. This work thus demonstrates that, during in vivo DRG development, TrkC behaves as a two-sided receptor transducing positive signals of neuronal survival in response to NT-3, but actively inducing neuronal cell death when unbound. This functional duality sets adequate number of neurons committed to a TrkC identity in the forming DRG.


Subject(s)
Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Receptor, trkC/metabolism , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism , Animals , Apoptosis/physiology , Cell Line , Cell Survival/physiology , Chick Embryo , Female , Ganglia, Spinal/embryology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism
7.
Cancer Cell ; 32(4): 427-443.e8, 2017 10 09.
Article in English | MEDLINE | ID: mdl-29017055

ABSTRACT

Neuroblastoma (NB) is a childhood cancer arising from sympatho-adrenal neural crest cells. Disseminated forms have high frequency of multiple tumoral foci whose etiology remains unknown; NB embryonic origin limits investigations in patients and current models. We developed an avian embryonic model driving human NB tumorigenesis in tissues homologous to patients. We found that aggressive NBs display a metastatic mode, secondary dissemination via peripheral nerves and aorta. Through tumor transcriptional profiling, we found that NB dissemination is induced by the shutdown of a pro-cohesion autocrine signal, SEMA3C, which constrains the tumoral mass. Lowering SEMA3C levels shifts the balance toward detachment, triggering NB cells to collectively evade the tumor. Together with patient cohort analysis, this identifies a microenvironment-driven pro-metastatic switch for NB.


Subject(s)
Neuroblastoma/secondary , Tumor Microenvironment , Adolescent , Adult , Animals , Cell Adhesion , Chick Embryo , Child , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasm Staging , Nerve Tissue Proteins/physiology , Neuroblastoma/etiology , Neuroblastoma/pathology , Receptors, Cell Surface/physiology , Semaphorins/genetics , Semaphorins/physiology
8.
Elife ; 62017 06 22.
Article in English | MEDLINE | ID: mdl-28639940

ABSTRACT

The diaphragm muscle is essential for breathing in mammals. Its asymmetric elevation during contraction correlates with morphological features suggestive of inherent left-right (L/R) asymmetry. Whether this asymmetry is due to L versus R differences in the muscle or in the phrenic nerve activity is unknown. Here, we have combined the analysis of genetically modified mouse models with transcriptomic analysis to show that both the diaphragm muscle and phrenic nerves have asymmetries, which can be established independently of each other during early embryogenesis in pathway instructed by Nodal, a morphogen that also conveys asymmetry in other organs. We further found that phrenic motoneurons receive an early L/R genetic imprint, with L versus R differences both in Slit/Robo signaling and MMP2 activity and in the contribution of both pathways to establish phrenic nerve asymmetry. Our study therefore demonstrates L-R imprinting of spinal motoneurons and describes how L/R modulation of axon guidance signaling helps to match neural circuit formation to organ asymmetry.


Subject(s)
Diaphragm/embryology , Diaphragm/innervation , Neural Pathways/embryology , Phrenic Nerve/embryology , Animals , Animals, Genetically Modified , Gene Expression Profiling , Mice , Motor Neurons/physiology , Nodal Protein/metabolism , Signal Transduction
9.
Nat Commun ; 6: 6366, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25721514

ABSTRACT

The spatial orientation of cell divisions is fundamental for tissue architecture and homeostasis. Here we analysed neuroepithelial progenitors in the developing mouse spinal cord to determine whether extracellular signals orient the mitotic spindle. We report that Semaphorin3B (Sema3B) released from the floor plate and the nascent choroid plexus in the cerebrospinal fluid (CSF) controls progenitor division orientation. Delivery of exogenous Sema3B to neural progenitors after neural tube opening in living embryos promotes planar orientation of their division. Preventing progenitor access to cues present in the CSF by genetically engineered canal obstruction affects the proportion of planar and oblique divisions. Sema3B knockout phenocopies the loss of progenitor access to the CSF. Sema3B binds to the apical surface of mitotic progenitors and exerts its effect via Neuropilin receptors, GSK3 activation and subsequent inhibition of the microtubule stabilizer CRMP2. Thus, extrinsic control mediated by the Semaphorin signalling orients progenitor divisions in neurogenic zones.


Subject(s)
Cell Division/physiology , Cell Polarity/physiology , Neuroepithelial Cells/physiology , Semaphorins/cerebrospinal fluid , Semaphorins/metabolism , Spinal Cord/embryology , Animals , Blotting, Western , Fluorescent Antibody Technique , HeLa Cells , Humans , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Neuropilins/metabolism , Spinal Cord/cytology , Statistics, Nonparametric
10.
Neuron ; 75(6): 1051-66, 2012 Sep 20.
Article in English | MEDLINE | ID: mdl-22998873

ABSTRACT

The Neurotrophic factor gdnf plays diverse developmental roles, supporting survival and also acting as a chemoattractant for axon and cell migration. We report that in the developing spinal cord, a focal source of gdnf is present in the floor plate (FP) where commissural axons cross the midline. Gdnf has no direct guidance properties but switches on the responsiveness of crossing commissural growth cones to the midline repellent Semaphorin3B by suppressing calpain-mediated processing of the Sema3B signaling coreceptor Plexin-A1. Analysis of single and double mutant mouse models indicates that although gdnf is the principal trigger of Sema3B midline repulsion, it acts with another FP cue, NrCAM. Finally, genetic and in vitro experiments provide evidence that this gdnf effect is RET independent and mediated by NCAM/GFRα1 signaling. This study identifies a regulator of midline crossing and reveals interplays between Semaphorin and gdnf signaling during axon guidance.


Subject(s)
Axons/physiology , Gene Expression Regulation, Developmental/physiology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Neural Cell Adhesion Molecules/metabolism , Neurons/cytology , Semaphorins/metabolism , Analysis of Variance , Animals , Axons/drug effects , Body Patterning/genetics , Calpain/metabolism , Cell Movement/drug effects , Cell Movement/genetics , Cells, Cultured , Embryo, Mammalian , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Glial Cell Line-Derived Neurotrophic Factor/deficiency , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Humans , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecules/genetics , Neurons/drug effects , Receptors, Cell Surface/metabolism , Semaphorins/genetics , Spinal Cord/cytology , Spinal Cord/embryology , Transfection
11.
Development ; 139(19): 3633-43, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22899844

ABSTRACT

The wiring of neuronal circuits requires complex mechanisms to guide axon subsets to their specific target with high precision. To overcome the limited number of guidance cues, modulation of axon responsiveness is crucial for specifying accurate trajectories. We report here a novel mechanism by which ligand/receptor co-expression in neurons modulates the integration of other guidance cues by the growth cone. Class 3 semaphorins (Sema3 semaphorins) are chemotropic guidance cues for various neuronal projections, among which are spinal motor axons navigating towards their peripheral target muscles. Intriguingly, Sema3 proteins are dynamically expressed, forming a code in motoneuron subpopulations, whereas their receptors, the neuropilins, are expressed in most of them. Targeted gain- and loss-of-function approaches in the chick neural tube were performed to enable selective manipulation of Sema3C expression in motoneurons. We show that motoneuronal Sema3C regulates the shared Sema3 neuropilin receptors Nrp1 and Nrp2 levels in opposite ways at the growth cone surface. This sets the respective responsiveness to exogenous Nrp1- and Nrp2-dependent Sema3A, Sema3F and Sema3C repellents. Moreover, in vivo analysis revealed a context where this modulation is essential. Motor axons innervating the forelimb muscles are exposed to combined expressions of semaphorins. We show first that the positioning of spinal nerves is highly stereotyped and second that it is compromised by alteration of motoneuronal Sema3C. Thus, the role of the motoneuronal Sema3 code could be to set population-specific axon sensitivity to limb-derived chemotropic Sema3 proteins, therefore specifying stereotyped motor nerve trajectories in their target field.


Subject(s)
Body Patterning/genetics , Chemotaxis/genetics , Extremities/embryology , Extremities/innervation , Motor Neurons/physiology , Semaphorins/physiology , Animals , Animals, Genetically Modified , Cells, Cultured , Chick Embryo , Gene Expression Regulation, Developmental , Growth Cones/metabolism , Growth Cones/physiology , HEK293 Cells , Humans , Motor Neurons/metabolism , Neurogenesis/genetics , Neurogenesis/physiology , Neuropilin-1/genetics , Neuropilin-1/metabolism , Neuropilin-1/physiology , Neuropilin-2/genetics , Neuropilin-2/metabolism , Neuropilin-2/physiology , Semaphorins/genetics , Semaphorins/metabolism , Spine/cytology , Spine/embryology , Spine/metabolism
12.
Neurogenetics ; 13(1): 49-59, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22222883

ABSTRACT

Mutations in the gene encoding the neural cell adhesion molecule L1CAM cause several neurological disorders collectively referred to as L1 syndrome. We report here a family case of X-linked hydrocephalus in which an obligate female carrier has two exonic L1CAM missense mutations in trans substituting amino acids in the first (p.W635C) or second (p.V768I) fibronectin-type III domains. We performed various biochemical and cell biological in vitro assays to evaluate the pathogenicity of these variants. Mutant L1-W635C protein accumulates in the endoplasmic reticulum (ER), is not transported into axons, and fails to promote L1CAM-mediated cell-cell adhesion as well as neurite growth. Immunoprecipitation experiments show that L1-W635C associates with the molecular ER chaperone calnexin and is modified by poly-ubiquitination. The mutant L1-V768I protein localizes at the cell surface, is not retained in the ER, and promotes neurite growth similar to wild-type L1CAM. However, the p.V768I mutation impairs L1CAM-mediated cell-cell adhesion albeit less severe than L1-W635C. These data indicate that p.W635C is a novel loss-of-function L1 syndrome mutation. The p.V768I mutation may represent a non-pathogenic variant or a variant associated with low penetrance. The poly-ubiquitination of L1-W635C and its association with the ER chaperone calnexin provide further insights into the molecular mechanisms underlying defective cell surface trafficking of L1CAM in L1 syndrome.


Subject(s)
Exons , Genetic Diseases, X-Linked/genetics , Genetic Variation , Hydrocephalus/genetics , Neural Cell Adhesion Molecule L1/genetics , Adult , Cell Line , Cerebral Aqueduct/abnormalities , Cerebral Aqueduct/metabolism , Cerebral Aqueduct/pathology , DNA Mutational Analysis , Female , Genetic Diseases, X-Linked/metabolism , Genetic Diseases, X-Linked/pathology , Humans , Hydrocephalus/metabolism , Hydrocephalus/pathology , Male , Middle Aged , Mutation , Neurons/cytology , Neurons/physiology , Pedigree
13.
Genes Dev ; 24(4): 396-410, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20159958

ABSTRACT

Commissural axon guidance requires complex modulations of growth cone sensitivity to midline-derived cues, but underlying mechanisms in vertebrates remain largely unknown. By using combinations of ex vivo and in vivo approaches, we uncovered a molecular pathway controlling the gain of response to a midline repellent, Semaphorin3B (Sema3B). First, we provide evidence that Semaphorin3B/Plexin-A1 signaling participates in the guidance of commissural projections at the vertebrate ventral midline. Second, we show that, at the precrossing stage, commissural neurons synthesize the Neuropilin-2 and Plexin-A1 Semaphorin3B receptor subunits, but Plexin-A1 expression is prevented by a calpain1-mediated processing, resulting in silencing commissural responsiveness. Third, we report that, during floor plate (FP) in-growth, calpain1 activity is suppressed by local signals, allowing Plexin-A1 accumulation in the growth cone and sensitization to Sema3B. Finally, we show that the FP cue NrCAM mediates the switch of Plexin-A1 processing underlying growth cone sensitization to Sema3B. This reveals pathway-dependent modulation of guidance receptor processing as a novel mechanism for regulating guidance decisions at intermediate targets.


Subject(s)
Axons/physiology , Neurons/cytology , Signal Transduction , Animals , Axons/metabolism , Calpain/metabolism , Cell Adhesion Molecules/metabolism , Chick Embryo , Embryo, Mammalian , Gene Expression Regulation, Developmental , Mice , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neuropilin-2/metabolism , Semaphorins/metabolism
14.
EMBO J ; 27(11): 1549-62, 2008 Jun 04.
Article in English | MEDLINE | ID: mdl-18464795

ABSTRACT

Axonal receptors for class 3 semaphorins (Sema3s) are heterocomplexes of neuropilins (Nrps) and Plexin-As signalling coreceptors. In the developing cerebral cortex, the Ig superfamily cell adhesion molecule L1 associates with Nrp1. Intriguingly, the genetic removal of L1 blocks axon responses of cortical neurons to Sema3A in vitro despite the expression of Plexin-As in the cortex, suggesting either that L1 substitutes for Plexin-As or that L1 and Plexin-A are both required and mediate distinct roles. We report that association of Nrp1 with L1 but not Plexin-As mediates the recruitment and activation of a Sema3A-induced focal adhesion kinase-mitogen-activated protein kinase cascade. This signalling downstream of L1 is needed for the disassembly of adherent points formed in growth cones and subsequently their collapse response to Sema3A. Plexin-As and L1 are coexpressed and present in common complexes in cortical neurons and both dominant-negative forms of Plexin-A and L1 impair their response to Sema3A. Consistently, Nrp1-expressing cortical projections are defective in mice lacking Plexin-A3, Plexin-A4 or L1. This reveals that specific signalling activities downstream of L1 and Plexin-As cooperate for mediating the axon guidance effects of Sema3A.


Subject(s)
Cerebral Cortex/growth & development , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Growth Cones/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neuropilin-1/metabolism , Semaphorin-3A/metabolism , Animals , Axons/metabolism , Cell Adhesion , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecule L1/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signal Transduction
15.
Development ; 134(24): 4491-501, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18039974

ABSTRACT

Class III semaphorins (SemaIIIs) are intercellular cues secreted by surrounding tissues to guide migrating cells and axons in the developing organism. This chemotropic activity is crucial for the formation of nerves and vasculature. Intriguingly, SemaIIIs are also synthesized by neurons during axon pathfinding, but their function as intrinsic cues remains unknown. We have explored the role of Sema3A expression in motoneurons during spinal nerve development. Loss- and gain-of-function in the neural tube of the chick embryo were undertaken to target Sema3A expression in motoneurons while preserving Sema3A sources localized in peripheral tissues, known to provide important repulsive information for delineating the routes of motor axons towards their ventral or dorsal targets. Strikingly, Sema3A overexpression induced defasciculation and exuberant growth of motor axon projections into these normally non-permissive territories. Moreover, knockdown studies showed that motoneuronal Sema3A is required for correct spinal nerve compaction and dorsal motor axon extension. Further analysis of Sema3A gain- and loss-of-function in ex vivo models revealed that Sema3A in motoneurons sets the level of sensitivity of their growth cones to exogenous Sema3A exposure. This regulation is associated with post-transcriptional and local control of the availability of the Sema3A receptor neuropilin 1 at the growth cone surface. Thus, by modulating the strength of Sema3A-mediated environmental repulsive constraints, Sema3A in motoneurons enables axons to extend more or less far away from these repulsive sources. Such interplay between intrinsic and extrinsic Sema3A may represent a fundamental mechanism in the accurate specification of axon pathways.


Subject(s)
Motor Neurons/metabolism , Neuropilin-1/metabolism , Semaphorin-3A/metabolism , Animals , Animals, Genetically Modified , Axons/drug effects , Axons/metabolism , Axons/ultrastructure , Base Sequence , Chick Embryo , DNA Primers/genetics , Gene Expression Regulation, Developmental , Models, Neurological , Motor Neurons/drug effects , Motor Neurons/ultrastructure , Neuropilin-1/genetics , RNA Interference , Semaphorin-3A/antagonists & inhibitors , Semaphorin-3A/genetics , Semaphorin-3A/pharmacology , Spinal Nerves/embryology , Spinal Nerves/metabolism
16.
Eur J Med Genet ; 50(3): 200-8, 2007.
Article in English | MEDLINE | ID: mdl-17383248

ABSTRACT

Mental retardation affects 2 to 3% of the population and is marked by significant etiological heterogeneity, including genetic and non genetic causes. FRAXA (FMR1) trinucleotide expansion is widely searched in routine screening, but found in only about 2% of the patients tested. Mutations of the MECP2 (methyl-CpG-binding protein) gene mainly cause Rett syndrome but were also shown to be involved in mental retardation. This study aimed to estimate the frequency of MECP2 gene mutations in a large group of mentally retarded patients without FRAXA expansion. Screening by heteroduplex analysis and SSCP followed by DNA sequencing of shifted bands were performed on 613 patients, including 442 males and 171 females. Eleven sequence variants were found, including nine polymorphisms. The two others may be pathogenetic. The first one, the double nucleotide substitution c.1162_1163delinsTA leading to a premature stop codon (p.Pro388X) was found in a female patient with random X-inactivation, presenting with borderline mental impairment without any features of Rett syndrome. The second one, the c.679C>G substitution, changing a glutamine to a glutamate in the transcriptional repression functional domain (p.Gln227Glu), was found in a female patient with a moderately biased X-chromosome inactivation profile and presenting with mild intellectual delay and minor psychotic features. The low mutation rate suggests that a large-scale routine screening for MECP2 in mentally retarded subjects is not cost-effective in clinical practice. Screening may be improved by a pre-selection based on clinical features that remain to be established.


Subject(s)
Intellectual Disability/genetics , Methyl-CpG-Binding Protein 2/genetics , Mutation , Amino Acid Substitution , Child , Codon, Nonsense/genetics , Cohort Studies , DNA/genetics , Female , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , France , Gene Frequency , Genetic Testing , Humans , Male , Point Mutation , Polymorphism, Single-Stranded Conformational
17.
Arch Ophthalmol ; 125(2): 213-6, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17296897

ABSTRACT

OBJECTIVE: To describe a new phenotype with an arginine-to-cysteine mutation at position 116 (Arg116Cys) in the CRYAA gene. METHODS: We investigated a 4-generation French family with autosomal dominant cataract and performed a genetic linkage analysis using microsatellite DNA markers encompassing 15 known cataract loci. Exons 1, 2, and 3 and flanking intronic sequences of the CRYAA gene were amplified and analyzed using direct sequencing. RESULTS: All of the affected individuals had nuclear cataract and iris coloboma. Genetic analysis revealed the previously described Arg116Cys mutation in the CRYAA gene in the heterozygous state in all of the affected members of the family but not in unaffected individuals. CONCLUSION: To our knowledge, this is the first case to date in which an Arg116Cys mutation in the CRYAA gene was associated with nuclear cataract and iris coloboma. CLINICAL RELEVANCE: This study indicates that an Arg116Cys mutation in the CRYAA gene could be associated with an unusual phenotype in affected individuals. In this family, the clinical observation of iris coloboma allows for the possibility of identifying individuals carrying the mutation. Iris coloboma is particularly important in terms of perinatal diagnosis because its detection in the newborn requires a careful and regular examination of the lens.


Subject(s)
Cataract/genetics , Coloboma/genetics , Crystallins/genetics , Iris/abnormalities , Microphthalmos/genetics , Mutation/genetics , Female , Genes, Dominant , Genetic Linkage , Humans , Male , Microsatellite Repeats , Pedigree , Phenotype
18.
Neuron ; 48(1): 63-75, 2005 Oct 06.
Article in English | MEDLINE | ID: mdl-16202709

ABSTRACT

Chemorepulsion by semaphorins plays a critical role during the development of neuronal projections. Although semaphorin-induced chemoattraction has been reported in vitro, the contribution of this activity to axon pathfinding is still unclear. Using genetic and culture models, we provide evidence that both attraction and repulsion by Sema3B, a secreted semaphorin, are critical for the positioning of a major brain commissural projection, the anterior commissure (AC). NrCAM, an immunoglobulin superfamily adhesion molecule of the L1 subfamily, associates with neuropilin-2 and is a component of a receptor complex for Sema3B and Sema3F. Finally, we show that activation of the FAK/Src signaling cascade distinguishes Sema3B-mediated attractive from repulsive axonal responses of neurons forming the AC, revealing a mechanism underlying the dual activity of this guidance cue.


Subject(s)
Neurons/metabolism , Olfactory Pathways , Semaphorins/physiology , Septal Nuclei/cytology , Adaptor Proteins, Signal Transducing/metabolism , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Axons/metabolism , Blotting, Northern/methods , Blotting, Western/methods , Cell Adhesion Molecules/metabolism , Cell Aggregation/drug effects , Cell Aggregation/physiology , Cells, Cultured , Chlorocebus aethiops , Cloning, Molecular/methods , Coculture Techniques/methods , Enzyme Inhibitors/pharmacology , Focal Adhesion Kinase 1/metabolism , Growth Cones/physiology , Immunohistochemistry/methods , Immunoprecipitation/methods , In Situ Hybridization/methods , Indoles/pharmacology , Mice , Mice, Knockout , Neuropilin-2/metabolism , Olfactory Pathways/growth & development , Olfactory Pathways/metabolism , Protein Binding/physiology , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction/methods , Semaphorins/deficiency , Septal Nuclei/growth & development , Septal Nuclei/metabolism , Signal Transduction/physiology , Sulfonamides/pharmacology , Transfection/methods , src-Family Kinases/physiology
19.
Eur J Hum Genet ; 12(7): 574-8, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15083168

ABSTRACT

Congenital microphthalmia is a developmental disorder characterized by shortened axial length of the eye. We have previously mapped the gene responsible for autosomal dominant colobomatous microphthalmia in a 5-generation family to chromosome 15q12-q15. Here, we set up a physical and transcript map of the 13.8 cM critical region, flanked by loci D15S1002 and D15S1040. Physical mapping and genetic linkage analysis using 20 novel polymorphic markers allowed the refinement of the disease locus to two intervals in close vicinity, namely a centromeric interval, bounded by microsatellite DNA markers m3-m17, and a telomeric interval, m76-m24, encompassing respectively 1.9 and 2.5 Mb. Moreover, we excluded three candidate genes, CKTSF1B1, KLF13 and CX36. Finally, although a phenomenon of anticipation was suggested by phenotypic and pedigree data, no abnormal expansion of three trinucleotide repeats mapping to the refine interval was found in affected individuals.


Subject(s)
Chromosomes, Human, Pair 15/genetics , Genes, Dominant/genetics , Microphthalmos/genetics , Cell Cycle Proteins/genetics , Computational Biology/methods , Connexins/genetics , Expressed Sequence Tags , Female , Genetic Linkage , Humans , Intercellular Signaling Peptides and Proteins/genetics , Kruppel-Like Transcription Factors , Male , Pedigree , Physical Chromosome Mapping , Polymorphism, Genetic , Repressor Proteins/genetics , Transcription, Genetic/genetics , Trinucleotide Repeats/genetics , Gap Junction delta-2 Protein
20.
Am J Med Genet A ; 122A(1): 37-41, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12949969

ABSTRACT

Isolated mental retardation is clinically and genetically heterogenous and may be inherited in an autosomal dominant, autosomal recessive, or X-linked manner. We report here a linkage analysis in a large family including 15 members, 6 of whom presenting X-linked non-syndromic mental retardation (MRX). Two-point linkage analysis using 23 polymorphic markers covering the entire X chromosome demonstrated significant linkage between the causative gene and DXS8055 with a maximum LOD score of 2.98 at theta = 0.00. Haplotype analysis indicated location for the disease gene in a 23.1 cM interval between DXS1106 and DXS8067. This MRX localization overlaps with 7 XLMR loci (MRX23, MRX27, MRX30, MRX35, MRX47, MRX53, and MRX63). This interval contains two genes associated with non-syndromic mental retardation (NSMR), namely the PAK3 gene, encoding a p21-activated kinase (MRX30 and MRX47) and the FACL4 gene encoding a fatty acyl-CoA ligase (MRX63). As skewed X-inactivation, an apparently constant feature in FACL4 carrier females was not observed in an obligate carrier belonging to the MRX family presented here, the PAK3 gene should be considered as the strongest candidate for this MRX locus.


Subject(s)
Chromosomes, Human, X , Mental Retardation, X-Linked/genetics , Chromosome Mapping , Female , Genetic Linkage , Humans , Male , Pedigree
SELECTION OF CITATIONS
SEARCH DETAIL
...