Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Cereb Cortex ; 30(3): 913-928, 2020 03 14.
Article in English | MEDLINE | ID: mdl-31298263

ABSTRACT

Neural progenitor proliferation, neuronal migration, areal organization, and pioneer axon wiring are critical events during early forebrain development, yet remain incompletely understood, especially in human. Here, we studied forebrain development in human embryos aged 5 to 8 postconceptional weeks (WPC5-8), stages that correspond to the neuroepithelium/early marginal zone (WPC5), telencephalic preplate (WPC6 & 7), and incipient cortical plate (WPC8). We show that early telencephalic neurons are formed at the neuroepithelial stage; the most precocious ones originate from local telencephalic neuroepithelium and possibly from the olfactory placode. At the preplate stage, forebrain organization is quite similar in human and mouse in terms of areal organization and of differentiation of Cajal-Retzius cells, pioneer neurons, and axons. Like in mice, axons from pioneer neurons in prethalamus, ventral telencephalon, and cortical preplate cross the diencephalon-telencephalon junction and the pallial-subpallial boundary, forming scaffolds that could guide thalamic and cortical axons at later stages. In accord with this model, at the early cortical plate stage, corticofugal axons run in ventral telencephalon in close contact with scaffold neurons, which express CELSR3 and FZD3, two molecules that regulates formation of similar scaffolds in mice.


Subject(s)
Axons/physiology , Neurons/physiology , Prosencephalon/embryology , Cell Adhesion Molecules, Neuronal/metabolism , Cells, Cultured , Extracellular Matrix Proteins/metabolism , Gestational Age , Gonadotropin-Releasing Hormone/metabolism , Humans , Nerve Tissue Proteins/metabolism , Neural Pathways/embryology , Neural Pathways/metabolism , Neurons/metabolism , Prosencephalon/metabolism , Reelin Protein , Serine Endopeptidases/metabolism
2.
Semin Cell Dev Biol ; 76: 1-2, 2018 04.
Article in English | MEDLINE | ID: mdl-28988957
3.
Development ; 144(22): 4061-4077, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29138289

ABSTRACT

The cerebral cortex covers the rostral part of the brain and, in higher mammals and particularly humans, plays a key role in cognition and consciousness. It is populated with neuronal cell bodies distributed in radially organized layers. Understanding the common and lineage-specific molecular mechanisms that orchestrate cortical development and evolution are key issues in neurobiology. During evolution, the cortex appeared in stem amniotes and evolved divergently in two main branches of the phylogenetic tree: the synapsids (which led to present day mammals) and the diapsids (reptiles and birds). Comparative studies in organisms that belong to those two branches have identified some common principles of cortical development and organization that are possibly inherited from stem amniotes and regulated by similar molecular mechanisms. These comparisons have also highlighted certain essential features of mammalian cortices that are absent or different in diapsids and that probably evolved after the synapsid-diapsid divergence. Chief among these is the size and multi-laminar organization of the mammalian cortex, and the propensity to increase its area by folding. Here, I review recent data on cortical neurogenesis, neuronal migration and cortical layer formation and folding in this evolutionary perspective, and highlight important unanswered questions for future investigation.


Subject(s)
Biological Evolution , Birds/embryology , Cerebral Cortex/embryology , Mammals/embryology , Reptiles/embryology , Animals , Humans , Neurogenesis , Reelin Protein
4.
Semin Cell Dev Biol ; 69: 102-110, 2017 09.
Article in English | MEDLINE | ID: mdl-28716607

ABSTRACT

Cadherin EGF LAG seven-pass G-type receptors 1, 2 and 3 (CELSR1-3) form a family of three atypical cadherins with multiple functions in epithelia and in the nervous system. During the past decade, evidence has accumulated for a key role of CELSR1 in epithelial planar cell polarity (PCP), and for CELSR2 and CELSR3 in ciliogenesis and neural development, especially neuron migration and axon guidance in the central, peripheral and enteric nervous systems. Phenotypes in mutant mice indicate that CELSR proteins work in concert with FZD3 and FZD6, but several questions remain. Apart from PCP signaling pathways implicating CELSR1 that begin to be unraveled, little is known about other signals generated by CELSR2 and CELSR3. A crucial question concerns the putative ligands that trigger signaling, in particular what is the role of WNT factors. Another critical issue is the identification of novel intracellular pathways and effectors that relay and transmit signals in receptive cells? Answers to those questions should further our understanding of the role of those important molecules not only in development but also in regeneration and disease.


Subject(s)
Cadherins/chemistry , Cadherins/metabolism , Animals , Axon Guidance , Body Patterning , Brain/physiology , Cadherins/genetics , Humans , Protein Domains
5.
Nat Commun ; 7: 13509, 2016 11 16.
Article in English | MEDLINE | ID: mdl-27848932

ABSTRACT

The diaphanous homologue Diaph3 (aka mDia2) is a major regulator of actin cytoskeleton. Loss of Diaph3 has been constantly associated with cytokinesis failure ascribed to impaired accumulation of actin in the cleavage furrow. Here we report that Diaph3 is required before cell fission, to ensure the accurate segregation of chromosomes. Inactivation of the Diaph3 gene causes a massive loss of cortical progenitor cells, with subsequent depletion of intermediate progenitors and neurons, and results in microcephaly. In embryonic brain extracts, Diaph3 co-immunoprecipitates with BubR1, a key regulator of the spindle assembly checkpoint (SAC). Diaph3-deficient cortical progenitors have decreased levels of BubR1 and fail to properly activate the SAC. Hence, they bypass mitotic arrest and embark on anaphase in spite of incorrect chromosome segregation, generating aneuploidy. Our data identify Diaph3 as a major guard of cortical progenitors, unravel novel functions of Diaphanous formins and add insights into the pathobiology of microcephaly.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Embryonic Stem Cells/physiology , M Phase Cell Cycle Checkpoints/physiology , Microtubule-Associated Proteins/metabolism , NADPH Dehydrogenase/metabolism , Neural Stem Cells/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Formins , Gene Expression Regulation/physiology , Gene Knockdown Techniques , HEK293 Cells , Humans , Mice , Mitosis/physiology , Mutation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
6.
Dev Biol ; 419(2): 298-310, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27612405

ABSTRACT

The dorsal surface of the mouse tongue is covered by ~7000 papillae, asymmetric epithelial protrusions that are precisely oriented to create a stereotyped macroscopic pattern. Within the context of this large-scale pattern, neighboring papillae exhibit a high degree of local order that minimizes the differences in their orientations. We show here that the orientations of lingual papillae are under the control of the core planar cell polarity (PCP) genes Vangl1, Vangl2, and Celsr1. Using K14-Cre and Nkx2.5-Cre to induce conditional knockout of Vangl1 and/or Vangl2 in the tongue epithelium, we observe more severe disruptions to local order among papillae with inactivation of larger numbers of Vangl genes, a greater role for Vangl2 than Vangl1, and a more severe phenotype with the Vangl2 Looptail (Lp) allele than the Vangl2 null allele, consistent with a dominant negative mode of action of the Vangl2Lp allele. Interestingly, Celsr1-/- tongues show disruption of both local and global order, with many papillae in the anterior tongue showing a reversed orientation. To quantify each of these phenotypes, we have developed and applied three procedures for sampling the orientations of papillae and assessing the degree of order on different spatial scales. The experiments reported here establish the dorsal surface of the mouse tongue as a favorable system for studying PCP control of epithelial patterning.


Subject(s)
Body Patterning/physiology , Carrier Proteins/physiology , Membrane Proteins/physiology , Mice/anatomy & histology , Nerve Tissue Proteins/physiology , Receptors, G-Protein-Coupled/physiology , Tongue/anatomy & histology , Alleles , Animals , Body Patterning/genetics , Carrier Proteins/genetics , Cell Polarity/physiology , Epithelial Cells/metabolism , Gene Deletion , Gene Dosage , Gene Knockout Techniques , Genetic Association Studies , Membrane Proteins/deficiency , Membrane Proteins/genetics , Models, Biological , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Phenotype , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Tongue/embryology
7.
Dev Biol ; 417(1): 40-9, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27395006

ABSTRACT

The caudal migration of facial branchiomotor (FBM) neurons from rhombomere (r) 4 to r6 in the hindbrain is an excellent model to study neuronal migration mechanisms. Although several Wnt/Planar Cell Polarity (PCP) components are required for FBM neuron migration, only Celsr1, an atypical cadherin, regulates the direction of migration in mice. In Celsr1 mutants, a subset of FBM neurons migrates rostrally instead of caudally. Interestingly, Celsr1 is not expressed in the migrating FBM neurons, but rather in the adjacent floor plate and adjoining ventricular zone. To evaluate the contribution of different expression domains to neuronal migration, we conditionally inactivated Celsr1 in specific cell types. Intriguingly, inactivation of Celsr1 in the ventricular zone of r3-r5, but not in the floor plate, leads to rostral migration of FBM neurons, greatly resembling the migration defect of Celsr1 mutants. Dye fill experiments indicate that the rostrally-migrated FBM neurons in Celsr1 mutants originate from the anterior margin of r4. These data suggest strongly that Celsr1 ensures that FBM neurons migrate caudally by suppressing molecular cues in the rostral hindbrain that can attract FBM neurons.


Subject(s)
Cell Movement/physiology , Facial Nerve/embryology , Neurogenesis/physiology , Receptors, G-Protein-Coupled/metabolism , Rhombencephalon/embryology , Animals , Facial Nerve/metabolism , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Motor Neurons/cytology , Receptors, G-Protein-Coupled/genetics
8.
Cereb Cortex ; 26(7): 3323-34, 2016 07.
Article in English | MEDLINE | ID: mdl-27170656

ABSTRACT

Celsr3 and Fzd3 regulate the development of reciprocal thalamocortical projections independently of their expression in cortical or thalamic neurons. To understand this cell non autonomous mechanism further, we tested whether Celsr3 and Fzd3 could act via Isl1-positive guidepost cells. Isl1-positive cells appear in the forebrain at embryonic day (E) 9.5-E10.5 and, from E12.5, they form 2 contingents in ventral telencephalon and prethalamus. In control mice, corticothalamic axons run in the ventral telencephalic corridor in close contact with Isl1-positive cells. When Celsr3 or Fzd3 is inactivated in Isl1-expressing cells, corticofugal fibers stall and loop in the ventral telencephalic corridor of high Isl1 expression, and thalamic axons fail to cross the diencephalon-telencephalon junction (DTJ). At E12.5, before thalamic and cortical axons emerge, pioneer projections from Isl1-positive cells cross the DTJ from both sides in control but not mutant embryos. These early projections appear to act like a bridge to guide later growing thalamic axons through the DTJ. Our data suggest that Celsr3 and Fzd3 orchestrate the formation of a scaffold of pioneer neurons and their axons. This scaffold extends from prethalamus to ventral telencephalon and subcortex, and steers reciprocal corticothalamic fibers.


Subject(s)
Axons/metabolism , Cadherins/metabolism , Cerebral Cortex/embryology , Frizzled Receptors/metabolism , Receptors, Cell Surface/metabolism , Thalamus/embryology , Animals , Cadherins/genetics , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Frizzled Receptors/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Mice, Transgenic , Neuronal Outgrowth/physiology , RNA, Messenger/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Receptors, Cell Surface/genetics , Thalamus/cytology , Thalamus/metabolism , Tissue Culture Techniques , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Invest Ophthalmol Vis Sci ; 57(6): 2788-96, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27214687

ABSTRACT

PURPOSE: Frizzled3 (Fzd3), a member of the core planar cell polarity (PCP) family in mammals, contributes to visual development by guiding axonal projections of some retinal ganglion cells. However, its other functions in the maturation of the visual system, especially the retina, remain elusive. The present study explores the role of Fzd3 in retinal development by focusing on rod bipolar cells (RBCs). METHODS: Frizzled3 was conditionally removed from the retina of Isl1-Cre;Fzd3f/- mice using the Cre-loxP system. Electroretinograms (ERGs) were performed to measure the light response of retinas. Frizzled3 expression was monitored by ß-galactosidase (ß-gal) staining and anti-ß-gal immunostaining. Immunofluorescence was used to examine cellular distribution and morphology during development, and electron microscopy was applied to visualize the dendritic invaginations of RBCs. RESULTS: Electroretinograms showed decreased b-wave amplitudes, and lower b- to a-wave ratios in Isl1-Cre;Fzd3f/- than in control (Isl1-Cre;Fzd3f/+) mice. In RBCs, where Fzd3 was expressed and inactivated, the planar organization, shape, and orientation of somas were disrupted. From P10, dendrites of these RBCs displayed reduced arborization with mistargeting. Furthermore, their dendritic invaginations into rod terminals were suppressed, and the density of rod ribbons in the OPL was reduced. CONCLUSIONS: Frizzled3 is required to shape the pattern of RBC somas and dendrites, and the structural and functional connectivity between rods and RBCs. Our results highlight novel functions for Fzd3 in regulating retinal development.


Subject(s)
Frizzled Receptors/genetics , Gene Expression Regulation, Developmental , Retinal Bipolar Cells/physiology , Retinal Ganglion Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Animals , Animals, Newborn , Cell Count , Disease Models, Animal , Electroretinography , Female , Frizzled Receptors/metabolism , Male , Mice , Mice, Mutant Strains , Microscopy, Electron , Retinal Ganglion Cells/ultrastructure , Retinal Rod Photoreceptor Cells/ultrastructure
10.
Nat Commun ; 7: 10936, 2016 Mar 04.
Article in English | MEDLINE | ID: mdl-26939553

ABSTRACT

Sequential generation of neurons and glial cells during development is critical for the wiring and function of the cerebral cortex. This process requires accurate coordination of neural progenitor cell (NPC) fate decisions, by NPC-autonomous mechanisms as well as by negative feedback from neurons. Here, we show that neurogenesis is protracted and gliogenesis decreased in mice with mutations of genes Celsr3 and Fzd3. This phenotype is not due to gene inactivation in progenitors, but rather in immature cortical neurons. Mutant neurons are unable to upregulate expression of Jag1 in response to cortical Wnt7, resulting in blunted activation of Notch signalling in NPC. Thus, Celsr3 and Fzd3 enable immature neurons to respond to Wnt7, upregulate Jag1 and thereby facilitate feedback signals that tune the timing of NPC fate decisions via Notch activation.


Subject(s)
Cadherins/metabolism , Frizzled Receptors/metabolism , Gene Expression Regulation/physiology , Proto-Oncogene Proteins/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Wnt Proteins/metabolism , Animals , Bromodeoxyuridine , Cadherins/genetics , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Female , Frizzled Receptors/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mutation , Neurogenesis/physiology , Pregnancy , Proto-Oncogene Proteins/genetics , Receptors, Cell Surface/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism , Serrate-Jagged Proteins , Staining and Labeling , Wnt Proteins/genetics
11.
Int J Biochem Cell Biol ; 64: 11-4, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25813877

ABSTRACT

The assembly of functional neuronal circuits depends on the correct wiring of axons and dendrites. To reach their targets, axons are guided by a variety of extracellular guidance cues, including Netrins, Ephrins, Semaphorins and Slits. Corresponding receptors in the growth cone, the dynamic structure at the tip of the growing axon, sense and integrate these positional signals, and activate downstream effectors to regulate cytoskeletal organization. In addition to the four canonical families of axon guidance cues mentioned above, some proteins that regulate planar cell polarity were recently found to be critical for axon guidance. The seven-transmembrane domain receptors Celsr3 and Fzd3, in particular, control the development of most longitudinal tracts in the central nervous system, and axon navigation in the peripheral, sympathetic and enteric nervous systems. Despite their unequivocally important role, however, underlying molecular mechanisms remain elusive. We do not know which extracellular ligands they recognize, whether they have co-receptors in the growth cone, and what their downstream effectors are. Here, we review some recent advances and discuss future trends in this emerging field.


Subject(s)
Cadherins/physiology , Frizzled Receptors/physiology , Growth Cones/physiology , Receptors, Cell Surface/physiology , Animals , Chemotaxis , Humans , Signal Transduction
12.
Development ; 141(23): 4558-68, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25406397

ABSTRACT

The oviduct is an important organ in reproduction where fertilization occurs, and through which the fertilized eggs are carried to the uterus in mammals. This organ is highly polarized, where the epithelium forms longitudinal folds along the ovary-uterus axis, and the epithelial multicilia beat towards the uterus to transport the ovulated ova. Here, we analyzed the postnatal development of mouse oviduct and report that multilevel polarities of the oviduct are regulated by a planar cell polarity (PCP) gene, Celsr1. In the epithelium, Celsr1 is concentrated in the specific cellular boundaries perpendicular to the ovary-uterus axis from postnatal day 2. We found a new feature of cellular polarity in the oviduct - the apical surface of epithelial cells is elongated along the ovary-uterus axis. In Celsr1-deficient mice, the ciliary motion is not orchestrated along the ovary-uterus axis and the transport ability of beating cilia is impaired. Epithelial cells show less elongation and randomized orientation, and epithelial folds show randomized directionality and ectopic branches in the mutant. Our mosaic analysis suggests that the geometry of epithelial cells is primarily regulated by Celsr1 and as a consequence the epithelial folds are aligned. Taken together, we reveal the characteristics of the multilevel polarity formation processes in the mouse oviduct epithelium and suggest a novel function of the PCP pathway for proper tissue morphogenesis.


Subject(s)
Cell Polarity/physiology , Epithelial Cells/physiology , Organogenesis/physiology , Oviducts/embryology , Receptors, G-Protein-Coupled/metabolism , Animals , Bromodeoxyuridine , Cell Polarity/genetics , Cell Shape/physiology , Cilia/physiology , DNA Primers/genetics , Female , Fluorescence , Immunohistochemistry , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Microspheres , Oviducts/ultrastructure , Reverse Transcriptase Polymerase Chain Reaction
13.
Ann N Y Acad Sci ; 1333: 43-64, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25424900

ABSTRACT

The class of adhesion G protein-coupled receptors (aGPCRs), with 33 human homologs, is the second largest family of GPCRs. In addition to a seven-transmembrane α-helix-a structural feature of all GPCRs-the class of aGPCRs is characterized by the presence of a large N-terminal extracellular region. In addition, all aGPCRs but one (GPR123) contain a GPCR autoproteolysis-inducing (GAIN) domain that mediates autoproteolytic cleavage at the GPCR autoproteolysis site motif to generate N- and a C-terminal fragments (NTF and CTF, respectively) during protein maturation. Subsequently, the NTF and CTF are associated noncovalently as a heterodimer at the plasma membrane. While the biological function of the GAIN domain-mediated autocleavage is not fully understood, mounting evidence suggests that the NTF and CTF possess distinct biological activities in addition to their function as a receptor unit. We discuss recent advances in understanding the biological functions, signaling mechanisms, and disease associations of the aGPCRs.


Subject(s)
Cell Adhesion , Receptors, G-Protein-Coupled/physiology , Animals , Developmental Disabilities/genetics , Humans , Mutation , Neoplasms/genetics , Signal Transduction , Synapses/physiology
14.
Nat Neurosci ; 17(9): 1171-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25108913

ABSTRACT

The cadherin Celsr3 regulates the directional growth and targeting of axons in the CNS, but whether it acts in collaboration with or in parallel to other guidance cues is unknown. Furthermore, the function of Celsr3 in the peripheral nervous system is still largely unexplored. Here we show that Celsr3 mediates pathfinding of motor axons innervating the hindlimb. In mice, Celsr3-deficient axons of the peroneal nerve segregate from those of the tibial nerve but fail to extend dorsally, and they stall near the branch point. Mutant axons respond to repulsive ephrinA-EphA forward signaling and glial cell-derived neurotrophic factor (GDNF). However, they are insensitive to attractive EphA-ephrinA reverse signaling. In transfected cells, Celsr3 immunoprecipitates with ephrinA2, ephrinA5, Ret, GDNF family receptor α1 (GFRα1) and Frizzled3 (Fzd3). The function of Celsr3 is Fzd3 dependent but Vangl2 independent. Our results provide evidence that the Celsr3-Fzd3 pathway interacts with EphA-ephrinA reverse signaling to guide motor axons in the hindlimb.


Subject(s)
Axons/physiology , Cadherins/genetics , Hindlimb/innervation , Motor Neurons/physiology , Peroneal Nerve/physiology , Receptors, Cell Surface/genetics , Tibial Nerve/physiology , Animals , Cadherins/metabolism , Cells, Cultured , Clubfoot/embryology , Clubfoot/genetics , Ephrin-A2/metabolism , Ephrin-A5/metabolism , Female , Frizzled Receptors/metabolism , Green Fluorescent Proteins/genetics , HEK293 Cells , Hindlimb/abnormalities , Humans , Male , Mice, Knockout , Motor Neurons/ultrastructure , Peroneal Nerve/cytology , Peroneal Nerve/embryology , Pregnancy , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Tibial Nerve/cytology , Tibial Nerve/embryology
15.
Proc Natl Acad Sci U S A ; 111(30): E3129-38, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25024228

ABSTRACT

In the nervous system, cilia dysfunction perturbs the circulation of the cerebrospinal fluid, thus affecting neurogenesis and brain homeostasis. A role for planar cell polarity (PCP) signaling in the orientation of cilia (rotational polarity) and ciliogenesis is established. However, whether and how PCP regulates cilia positioning in the apical domain (translational polarity) in radial progenitors and ependymal cells remain unclear. By analysis of a large panel of mutant mice, we show that two PCP signals are operating in ciliated cells. The first signal, controlled by cadherin, EGF-like, laminin G-like, seven-pass, G-type receptor (Celsr) 2, Celsr3, Frizzled3 (Fzd3) and Van Gogh like2 (Vangl2) organizes multicilia in individual cells (single-cell polarity), whereas the second signal, governed by Celsr1, Fzd3, and Vangl2, coordinates polarity between cells in both radial progenitors and ependymal cells (tissue polarity). Loss of either of these signals is associated with specific defects in the cytoskeleton. Our data reveal unreported functions of PCP and provide an integrated view of planar polarization of the brain ciliated cells.


Subject(s)
Cell Polarity/physiology , Cytoskeleton/metabolism , Ependyma/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Signal Transduction/physiology , Animals , Cilia/genetics , Cilia/metabolism , Cytoskeleton/genetics , Ependyma/cytology , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics
16.
Proc Natl Acad Sci U S A ; 111(29): E2996-3004, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-25002511

ABSTRACT

Celsr3 and Fzd3, members of "core planar cell polarity" (PCP) genes, were shown previously to control forebrain axon guidance and wiring by acting in axons and/or guidepost cells. Here, we show that Celsr2 acts redundantly with Celsr3, and that their combined mutation mimics that of Fzd3. The phenotypes generated upon inactivation of Fzd3 in different forebrain compartments are similar to those in conditional Celsr2-3 mutants, indicating that Fzd3 and Celsr2-3 act in the same population of cells. Inactivation of Celsr2-3 or Fzd3 in thalamus does not affect forebrain wiring, and joint inactivation in cortex and thalamus adds little to cortical inactivation alone in terms of thalamocortical projections. On the other hand, joint inactivation perturbs strongly the formation of the barrel field, which is unaffected upon single cortical or thalamic inactivation, indicating a role for interactions between thalamic axons and cortical neurons in cortical arealization. Unexpectedly, forebrain wiring is normal in mice defective in Vangl1 and Vangl2, showing that, contrary to epithelial PCP, axon guidance can be Vangl independent in some contexts. Our results suggest that Celsr2-3 and Fzd3 regulate axonal navigation in the forebrain by using mechanisms different from classical epithelial PCP, and require interacting partners other than Vangl1-2 that remain to be identified.


Subject(s)
Cadherins/metabolism , Carrier Proteins/metabolism , Frizzled Receptors/metabolism , Membrane Proteins/metabolism , Nerve Net/metabolism , Nerve Tissue Proteins/metabolism , Prosencephalon/metabolism , Receptors, Cell Surface/metabolism , Animals , Axons/metabolism , Cerebral Cortex/metabolism , Gene Silencing , Integrases/metabolism , Mice , Mutation/genetics , Phenotype , Thalamus/metabolism
17.
J Neurosci ; 33(49): 19071-85, 2013 Dec 04.
Article in English | MEDLINE | ID: mdl-24305805

ABSTRACT

How growth cones detect small concentration differences of guidance cues for correct steering remains a long-standing puzzle. Commissural axons engage planar cell polarity (PCP) signaling components to turn anteriorly in a Wnt gradient after midline crossing. We found here that Frizzled3, a Wnt receptor, undergoes endocytosis via filopodia tips. Wnt5a increases Frizzled3 endocytosis, which correlates with filopodia elongation. We discovered an unexpected antagonism between Dishevelleds, which may function as a signal amplification mechanism in filopodia where PCP signaling is activated: Dishevelled2 blocks Dishevelled1-induced Frizzled3 hyperphosphorylation and membrane accumulation. A key component of apical-basal polarity (A-BP) signaling, aPKC, also inhibits Dishevelled1-induced Frizzled3 hyperphosphorylation. Celsr3, another PCP component, is required in commissural neurons for anterior turning. Frizzled3 hyperphosphorylation is increased in Celsr3 mutant mice, where PCP signaling is impaired, suggesting Frizzled3 hyperphosphorylation does correlate with loss of PCP signaling in vivo. Furthermore, we found that the small GTPase, Arf6, which is required for Frizzled3 endocytosis, is essential for Wnt-promoted outgrowth, highlighting the importance of Frizzled3 recycling in PCP signaling in growth cone guidance. In a Wnt5a gradient, more Frizzled3 endocytosis and activation of atypical protein kinase C was observed on the side of growth cones facing higher Wnt5a concentration, suggesting that spatially controlled Frizzled3 endocytosis is part of the key mechanism for growth cone steering.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Endocytosis/physiology , Frizzled Receptors/physiology , Growth Cones/physiology , Phosphoproteins/physiology , Pseudopodia/physiology , Wnt Proteins/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Avidin/metabolism , Axons/physiology , Biotinylation , Cell Polarity/physiology , Cells, Cultured , Dishevelled Proteins , Endocytosis/genetics , Female , Frizzled Receptors/genetics , Glutathione Transferase/metabolism , Glycoside Hydrolases/metabolism , Immunohistochemistry , Immunoprecipitation , Male , Mice , Neurons/physiology , Phosphoproteins/genetics , Phosphoric Monoester Hydrolases/metabolism , Plasmids/genetics , RNA, Small Interfering/genetics , Rats , Signal Transduction/physiology , rac GTP-Binding Proteins/metabolism
18.
Nat Rev Neurosci ; 14(8): 525-35, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23839596

ABSTRACT

Planar cell polarity (PCP) is complementary to the intrinsic polarization of single cells and refers to the global coordination of cell behaviour in the plane of a tissue and, by extension, to the signalling pathways that control it. PCP is most evident in cell sheets, and research into PCP was for years confined to studies in Drosophila melanogaster. However, PCP has more recently emerged as an important phenomenon in vertebrates, in which it regulates various developmental processes and is associated with multiple disorders. In particular, core PCP genes are crucial for the development and function of the nervous system. They are involved in neural tube closure, ependymal polarity, neuronal migration, dendritic growth and axon guidance.


Subject(s)
Cell Polarity/genetics , Membrane Proteins/genetics , Nervous System/cytology , Nervous System/metabolism , Animals , Body Patterning/genetics , Cell Movement/genetics , Membrane Proteins/metabolism , Models, Biological , Nervous System/embryology , Nervous System/growth & development , Neurons/cytology , Neurons/physiology , Signal Transduction/genetics
19.
J Clin Invest ; 123(4): 1763-72, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23478408

ABSTRACT

A highly complex network of intrinsic enteric neurons is required for the digestive and homeostatic functions of the gut. Nevertheless, the genetic and molecular mechanisms that regulate their assembly into functional neuronal circuits are currently unknown. Here we report that the planar cell polarity (PCP) genes Celsr3 and Fzd3 are required during murine embryogenesis to specifically control the guidance and growth of enteric neuronal projections relative to the longitudinal and radial gut axes. Ablation of these genes disrupts the normal organization of nascent neuronal projections, leading to subtle changes of axonal tract configuration in the mature enteric nervous system (ENS), but profound abnormalities in gastrointestinal motility. Our data argue that PCP-dependent modules of connectivity established at early stages of enteric neurogenesis control gastrointestinal function in adult animals and provide the first evidence that developmental deficits in ENS wiring may contribute to the pathogenesis of idiopathic bowel disorders.


Subject(s)
Cadherins/genetics , Embryonic Development/genetics , Enteric Nervous System/embryology , Frizzled Receptors/genetics , Neurogenesis/genetics , Receptors, Cell Surface/genetics , Animals , Axons/physiology , Cadherins/metabolism , Cells, Cultured , Colon/physiology , Enteric Nervous System/metabolism , Enteric Nervous System/physiology , Frizzled Receptors/metabolism , Gastrointestinal Motility , Gastrointestinal Tract/embryology , Gastrointestinal Tract/innervation , Gastrointestinal Tract/physiology , Gene Expression Regulation, Developmental , Mice , Neurons/physiology , Receptors, Cell Surface/metabolism
20.
Prog Mol Biol Transl Sci ; 116: 193-214, 2013.
Article in English | MEDLINE | ID: mdl-23481196

ABSTRACT

Cadherin EGF LAG seven-pass G-type receptors 1, 2, and 3 (Celsr1-3) form a family of three atypical cadherins with multiple functions in epithelia and in the nervous system. During the past decade, evidence has accumulated for important and distinct roles of Celsr1-3 in planar cell polarity (PCP) during the development of the brain and some other organs. Although Celsr function in PCP is conserved from flies to mammals, other functions may be more distantly related, with Celsr working only with one or a subset of the classical core PCP partners. Here, we review the literature on Celsr, focusing on PCP and particularly on brain development.


Subject(s)
Brain/metabolism , Cadherins/classification , Cadherins/metabolism , Cell Polarity , Vertebrates/metabolism , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...