Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 185(21): 3931-3949.e26, 2022 10 13.
Article in English | MEDLINE | ID: mdl-36240740

ABSTRACT

Neural migration is a critical step during brain development that requires the interactions of cell-surface guidance receptors. Cancer cells often hijack these mechanisms to disseminate. Here, we reveal crystal structures of Uncoordinated-5 receptor D (Unc5D) in complex with morphogen receptor glypican-3 (GPC3), forming an octameric glycoprotein complex. In the complex, four Unc5D molecules pack into an antiparallel bundle, flanked by four GPC3 molecules. Central glycan-glycan interactions are formed by N-linked glycans emanating from GPC3 (N241 in human) and C-mannosylated tryptophans of the Unc5D thrombospondin-like domains. MD simulations, mass spectrometry and structure-based mutants validate the crystallographic data. Anti-GPC3 nanobodies enhance or weaken Unc5-GPC3 binding and, together with mutant proteins, show that Unc5/GPC3 guide migrating pyramidal neurons in the mouse cortex, and cancer cells in an embryonic xenograft neuroblastoma model. The results demonstrate a conserved structural mechanism of cell guidance, where finely balanced Unc5-GPC3 interactions regulate cell migration.


Subject(s)
Cell Movement , Glypicans/chemistry , Netrin Receptors/chemistry , Animals , Glypicans/metabolism , Humans , Mice , Mutant Proteins , Netrin Receptors/metabolism , Receptors, Cell Surface/metabolism , Single-Domain Antibodies , Thrombospondins
2.
BMC Biol ; 22(1): 131, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38831263

ABSTRACT

BACKGROUND: Fine characterization of gene expression patterns is crucial to understand many aspects of embryonic development. The chicken embryo is a well-established and valuable animal model for developmental biology. The period spanning from the third to sixth embryonic days (E3 to E6) is critical for many organ developments. Hybridization chain reaction RNA fluorescent in situ hybridization (HCR RNA-FISH) enables multiplex RNA detection in thick samples including embryos of various animal models. However, its use is limited by tissue opacity. RESULTS: We optimized HCR RNA-FISH protocol to efficiently label RNAs in whole mount chicken embryos from E3.5 to E5.5 and adapted it to ethyl cinnamate (ECi) tissue clearing. We show that light sheet imaging of HCR RNA-FISH after ECi clearing allows RNA expression analysis within embryonic tissues with good sensitivity and spatial resolution. Finally, whole mount immunofluorescence can be performed after HCR RNA-FISH enabling as exemplified to assay complex spatial relationships between axons and their environment or to monitor GFP electroporated neurons. CONCLUSIONS: We could extend the use of HCR RNA-FISH to older chick embryos by optimizing HCR RNA-FISH and combining it with tissue clearing and 3D imaging. The integration of immunostaining makes possible to combine gene expression with classical cell markers, to correlate expressions with morphological differentiation and to depict gene expressions in gain or loss of function contexts. Altogether, this combined procedure further extends the potential of HCR RNA-FISH technique for chicken embryology.


Subject(s)
In Situ Hybridization, Fluorescence , Animals , Chick Embryo , In Situ Hybridization, Fluorescence/methods , Fluorescent Antibody Technique/methods , Imaging, Three-Dimensional/methods , RNA/metabolism , RNA/genetics , Gene Expression Regulation, Developmental
4.
Semin Cell Dev Biol ; 85: 3-12, 2019 01.
Article in English | MEDLINE | ID: mdl-29277684

ABSTRACT

The navigation of commissural axons in the developing spinal cord has attracted multiple studies over the years. Many important concepts emerged from these studies which have enlighten the general mechanisms of axon guidance. The navigation of commissural axons is regulated by a series of cellular territories which provides the diverse guidance information necessary to ensure the successive steps of their pathfinding towards, across, and away from the ventral midline. In this review, we discuss how repulsive forces, by propelling, channelling, and confining commissural axon navigation, bring key contributions to the formation of this neuronal projection.


Subject(s)
Axon Guidance , Axons/metabolism , Spinal Cord/metabolism , Animals , Humans , Neurons/metabolism
5.
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
6.
Development ; 143(12): 2183-93, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27122172

ABSTRACT

In the hematopoietic system, Syk family tyrosine kinases are essential components of immunoreceptor ITAM-based signaling. While there is increasing data indicating the involvement of immunoreceptors in neural functions, the contribution of Syk kinases remains obscure. Previously, we identified phosphorylated forms of Syk kinases in specialized populations of migrating neurons or projecting axons. Moreover, we identified ephrin/Eph as guidance molecules utilizing the ITAM-bearing CD3zeta (Cd247) and associated Syk kinases for the growth cone collapse response induced in vitro Here, we show that in the developing spinal cord, Syk is phosphorylated in navigating commissural axons. By analyzing axon trajectories in open-book preparations of Syk(-/-); Zap70(-/-) mouse embryos, we show that Syk kinases are dispensable for attraction towards the midline but confer growth cone responsiveness to repulsive signals that expel commissural axons from the midline. Known to serve a repulsive function at the midline, ephrin B3/EphB2 are obvious candidates for driving the Syk-dependent repulsive response. Indeed, Syk kinases were found to be required for ephrin B3-induced growth cone collapse in cultured commissural neurons. In fragments of commissural neuron-enriched tissues, Syk is in a constitutively phosphorylated state and ephrin B3 decreased its level of phosphorylation. Direct pharmacological inhibition of Syk kinase activity was sufficient to induce growth cone collapse. In conclusion, Syk kinases act as a molecular switch of growth cone adhesive and repulsive responses.


Subject(s)
Axons/metabolism , Ephrin-B3/metabolism , Receptor, EphB2/metabolism , Signal Transduction , Spinal Cord/metabolism , Syk Kinase/metabolism , Animals , Embryo, Mammalian/metabolism , Endocytosis , Growth Cones/metabolism , Mice, Knockout , Phosphorylation
7.
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
8.
Int J Mol Sci ; 18(4)2017 Mar 24.
Article in English | MEDLINE | ID: mdl-28338624

ABSTRACT

Membrane microdomains or "lipid rafts" have emerged as essential functional modules of the cell, critical for the regulation of growth factor receptor-mediated responses. Herein we describe the dichotomy between caveolin-1 and caveolin-2, structural and regulatory components of microdomains, in modulating proliferation and differentiation. Caveolin-2 potentiates while caveolin-1 inhibits nerve growth factor (NGF) signaling and subsequent cell differentiation. Caveolin-2 does not appear to impair NGF receptor trafficking but elicits prolonged and stronger activation of MAPK (mitogen-activated protein kinase), Rsk2 (ribosomal protein S6 kinase 2), and CREB (cAMP response element binding protein). In contrast, caveolin-1 does not alter initiation of the NGF signaling pathway activation; rather, it acts, at least in part, by sequestering the cognate receptors, TrkA and p75NTR, at the plasma membrane, together with the phosphorylated form of the downstream effector Rsk2, which ultimately prevents CREB phosphorylation. The non-phosphorylatable caveolin-1 serine 80 mutant (S80V), no longer inhibits TrkA trafficking or subsequent CREB phosphorylation. MC192, a monoclonal antibody towards p75NTR that does not block NGF binding, prevents exit of both NGF receptors (TrkA and p75NTR) from lipid rafts. The results presented herein underline the role of caveolin and receptor signaling complex interplay in the context of neuronal development and tumorigenesis.


Subject(s)
Caveolin 1/metabolism , Cell Nucleus/metabolism , Membrane Microdomains/metabolism , Nerve Growth Factor/pharmacology , Signal Transduction/drug effects , Animals , Antibodies, Monoclonal/immunology , CREB-Binding Protein/metabolism , Caveolin 1/antagonists & inhibitors , Caveolin 1/genetics , Caveolin 2/antagonists & inhibitors , Caveolin 2/genetics , Caveolin 2/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Mice , Nerve Tissue Proteins , PC12 Cells , Phosphorylation/drug effects , Protein Binding , Protein Transport/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Rats , Receptor, Nerve Growth Factor/metabolism , Receptor, trkA/chemistry , Receptor, trkA/immunology , Receptor, trkA/metabolism , Receptors, Growth Factor , Receptors, Nerve Growth Factor/chemistry , Receptors, Nerve Growth Factor/immunology , Receptors, Nerve Growth Factor/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
9.
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
10.
Cereb Cortex ; 23(3): 604-14, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22368082

ABSTRACT

Previous work demonstrated that members of the semaphorin family, Sema3A and Sema3C, act as repulsive and attractive guidance signals, respectively, for cortical axons. During the development of corticofugal projections, these semaphorins are expressed in adjacent cortical zones, but there is a considerable overlap between Sema3A and Sema3C expression in the subventricular zone. We used different in vitro assays to examine the response of cortical axons exposed to defined mixtures of these opposing guidance cues. Results showed that even at very low concentrations, Sema3A overrides the effects of Sema3C. Moreover, experiments with function-blocking antibodies directed against neuropilin provided insights into how cortical axons integrate disparate guidance signals at the receptor level. These in vitro data suggest that the pathway of corticofugal axons is defined by an attractive cue in the intermediate zone, where Sema3C is expressed alone. To directly test this hypothesis in vivo, we performed axon-tracing experiments in Sema3C-deficient mice. Compared with wild-type animals, corticofugal axons take a more superficial route in Sema3C(-/-) mice, and the corticofugal pathway is more compacted. This phenotype is expected when an attractive cue for cortical axons, Sema3C, is eliminated and a repulsive cue, Sema3A, becomes predominant.


Subject(s)
Axons/metabolism , Cerebral Cortex/growth & development , Neurogenesis/physiology , Semaphorins/metabolism , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Coculture Techniques , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
Dev Cell ; 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39353435

ABSTRACT

Reconstructing functional neuronal circuits is one major challenge of central nervous system repair. Through activation of pro-growth signaling pathways, some neurons achieve long-distance axon regrowth. Yet, functional reconnection has hardly been obtained, as these regenerating axons fail to resume their initial trajectory and reinnervate their proper target. Axon guidance is considered to be active only during development. Here, using the mouse visual system, we show that axon guidance is still active in the adult brain in regenerative conditions. We highlight that regenerating retinal ganglion cell axons avoid one of their primary targets, the suprachiasmatic nucleus (SCN), due to Slit/Robo repulsive signaling. Together with promoting regeneration, silencing Slit/Robo in vivo enables regenerating axons to enter the SCN and form active synapses. The newly formed circuit is associated with neuronal activation and functional recovery. Our results provide evidence that axon guidance mechanisms are required to reconnect regenerating axons to specific brain nuclei.

12.
Nat Commun ; 15(1): 7065, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39152112

ABSTRACT

The sympathetic nervous system controls bodily functions including vascular tone, cardiac rhythm, and the "fight-or-flight response". Sympathetic chain ganglia develop in parallel with preganglionic motor nerves extending from the neural tube, raising the question of whether axon targeting contributes to sympathetic chain formation. Using nerve-selective genetic ablations and lineage tracing in mouse, we reveal that motor nerve-associated Schwann cell precursors (SCPs) contribute sympathetic neurons and satellite glia after the initial seeding of sympathetic ganglia by neural crest. Motor nerve ablation causes mispositioning of SCP-derived sympathoblasts as well as sympathetic chain hypoplasia and fragmentation. Sympathetic neurons in motor-ablated embryos project precociously and abnormally towards dorsal root ganglia, eventually resulting in fusion of sympathetic and sensory ganglia. Cell interaction analysis identifies semaphorins as potential motor nerve-derived signaling molecules regulating sympathoblast positioning and outgrowth. Overall, central innervation functions both as infrastructure and regulatory niche to ensure the integrity of peripheral ganglia morphogenesis.


Subject(s)
Ganglia, Sympathetic , Motor Neurons , Neural Crest , Schwann Cells , Sympathetic Nervous System , Animals , Sympathetic Nervous System/embryology , Mice , Motor Neurons/physiology , Schwann Cells/metabolism , Neural Crest/cytology , Neural Crest/metabolism , Ganglia, Sympathetic/cytology , Ganglia, Spinal , Semaphorins/metabolism , Semaphorins/genetics , Mice, Transgenic , Neuroglia/metabolism , Female
13.
Eur J Neurosci ; 37(10): 1573-83, 2013 May.
Article in English | MEDLINE | ID: mdl-23534961

ABSTRACT

Axonal projections in the CNS can be categorized as either crossed or uncrossed. Crossing and uncrossing of axons has been explained by attractive and repulsive molecules like Netrin-1 and Slits, which are secreted by midline structures. However, uncrossed projections can be established even in double knockout mice of slit1 and slit2 or of roundabout1 (robo1) and robo2, two receptors for Slits. Here, we found that a novel mechanism mediated by Neuropilin-2 (Nrp2) contributes to the formation of uncrossed projections of midbrain dopaminergic neurons (mDANs). Nrp2 transcriptional activities were detected in a subset of mDANs, and its protein was expressed in mDAN axons growing through the ipsilateral diencephalon. In nrp2(lac) (Z) (/lac) (Z) mice, mDAN axons aberrantly grew toward the ventral midline and even crossed it, suggesting that Nrp2 is necessary for the development of mDAN ipsilateral projections. We investigated the involvement of Semaphorin 3B (Sema3B) and Sema3F, two ligands of Nrp2, by analysing mDAN axon trajectories in single or double knockout mice. In both cases, mDAN axons still projected ipsilaterally, suggesting the involvement mechanisms independent of these Sema3s. Nrp2-deficient mDAN axons retained their responsiveness to Slit2, demonstrating that aberrant mDAN axons in nrp2(lac) (Z) (/lac) (Z) mice were not indirectly mediated by alterations in Slit/Robo signaling. Taken together, our results indicate that a novel mechanism mediated by Nrp2 contributes to the establishment of uncrossed projections by mDAN axons.


Subject(s)
Axons/metabolism , Dopaminergic Neurons/metabolism , Mesencephalon/metabolism , Neuropilin-2/genetics , Animals , Cells, Cultured , Dopaminergic Neurons/cytology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mesencephalon/cytology , Mesencephalon/embryology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuropilin-2/metabolism , Semaphorins/genetics , Semaphorins/metabolism , Transcription, Genetic
14.
EMBO Mol Med ; 15(3): e16629, 2023 03 08.
Article in English | MEDLINE | ID: mdl-36692026

ABSTRACT

Metastatic melanoma patients carrying a BRAFV600 mutation can be treated with a combination of BRAF and MEK inhibitors (BRAFi/MEKi), but innate and acquired resistance invariably occurs. Predicting patient response to targeted therapies is crucial to guide clinical decision. We describe here the development of a highly efficient patient-derived xenograft model adapted to patient melanoma biopsies, using the avian embryo as a host (AVI-PDXTM ). In this in vivo paradigm, we depict a fast and reproducible tumor engraftment of patient samples within the embryonic skin, preserving key molecular and phenotypic features. We show that sensitivity and resistance to BRAFi/MEKi can be reliably modeled in these AVI-PDXTM , as well as synergies with other drugs. We further provide proof-of-concept that the AVI-PDXTM models the diversity of responses of melanoma patients to BRAFi/MEKi, within days, hence positioning it as a valuable tool for the design of personalized medicine assays and for the evaluation of novel combination strategies.


Subject(s)
Melanoma , Proto-Oncogene Proteins B-raf , Animals , Humans , Proto-Oncogene Proteins B-raf/genetics , Melanoma/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Mitogen-Activated Protein Kinase Kinases , Mutation , Disease Models, Animal
15.
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
16.
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
17.
Cell Mol Life Sci ; 68(15): 2539-53, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21538161

ABSTRACT

Organisms with bilateral symmetry elaborate patterns of neuronal projections connecting both sides of the central nervous system at all levels of the neuraxis. During development, these so-called commissural projections navigate across the midline to innervate their contralateral targets. Commissural axon pathfinding has been extensively studied over the past years and turns out to be a highly complex process, implicating modulation of axon responsiveness to the various guidance cues that instruct axon trajectories towards, within and away from the midline. Understanding the molecular mechanisms allowing these switches of response to take place at the appropriate time and place is a major challenge for current research. Recent work characterized several instructive processes controlling the spatial and temporal fine-tuning of the guidance molecular machinery. These findings illustrate the molecular strategies by which commissural axons modulate their sensitivity to guidance cues during midline crossing and show that regulation at both transcriptional and post-transcriptional levels are crucial for commissural axon guidance.


Subject(s)
Axons/physiology , Central Nervous System/physiology , Synapses/physiology , Animals , Axonal Transport/genetics , Axonal Transport/physiology , Axons/metabolism , Cell Movement/genetics , Cell Movement/physiology , Central Nervous System/metabolism , Drosophila/genetics , Drosophila/physiology , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Humans , Membrane Proteins/genetics , Membrane Proteins/physiology , Models, Biological , Synapses/genetics , Synapses/metabolism
18.
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.

19.
Nat Commun ; 13(1): 2549, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35538114

ABSTRACT

Embryonic malignant transformation is concomitant to organogenesis, often affecting multipotent and migratory progenitors. While lineage relationships between malignant cells and their physiological counterparts are extensively investigated, the contribution of exogenous embryonic signals is not fully known. Neuroblastoma (NB) is a childhood malignancy of the peripheral nervous system arising from the embryonic trunk neural crest (NC) and characterized by heterogeneous and interconvertible tumor cell identities. Here, using experimental models mimicking the embryonic context coupled to proteomic and transcriptomic analyses, we show that signals released by embryonic sympathetic ganglia, including Olfactomedin-1, induce NB cells to shift from a noradrenergic to mesenchymal identity, and to activate a gene program promoting NB metastatic onset and dissemination. From this gene program, we extract a core signature specifically shared by metastatic cancers with NC origin. This reveals non-cell autonomous embryonic contributions regulating the plasticity of NB identities and setting pro-dissemination gene programs common to NC-derived cancers.


Subject(s)
Neural Crest , Neuroblastoma , Cell Differentiation/genetics , Child , Cues , Humans , Neuroblastoma/genetics , Neuroblastoma/pathology , Proteomics
20.
iScience ; 24(12): 103423, 2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34849474

ABSTRACT

Lack of preclinical patient-derived xenograft cancer models in which to conduct large-scale molecular studies seriously impairs the development of effective personalized therapies. We report here an in vivo concept consisting of implanting human tumor cells in targeted tissues of an avian embryo, delivering therapeutics, evaluating their efficacy by measuring tumors using light sheet confocal microscopy, and conducting large-scale RNA-seq analysis to characterize therapeutic-induced changes in gene expression. The model was established to recapitulate triple-negative breast cancer (TNBC) and validated using TNBC standards of care and an investigational therapeutic agent.

SELECTION OF CITATIONS
SEARCH DETAIL