Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Cell Rep ; 42(5): 112493, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37149870

ABSTRACT

Axons crossing the CNS midline regulate their responsiveness to both attractive and repulsive cues. In this issue, Dailey-Krempel et al. find different modes of action for DCC isoforms and uncover evidence against the silencing model of axon guidance.


Subject(s)
Axons , Axons/physiology
2.
Dev Biol ; 476: 272-281, 2021 08.
Article in English | MEDLINE | ID: mdl-33905720

ABSTRACT

Muscle function is dependent on innervation by the correct motor nerves. Motor nerves are composed of motor axons which extend through peripheral tissues as a compact bundle, then diverge to create terminal nerve branches to specific muscle targets. As motor nerves approach their targets, they undergo a transition where the fasciculated nerve halts further growth then after a pause, the nerve later initiates branching to muscles. This transition point is potentially an intermediate target or guidepost to present specific cellular and molecular signals for navigation. Here we describe the navigation of the oculomotor nerve and its association with developing muscles in mouse embryos. We found that the oculomotor nerve initially grew to the eye three days prior to the appearance of any extraocular muscles. The oculomotor axons spread to form a plexus within a mass of cells, which included precursors of extraocular muscles and other orbital tissues and expressed the transcription factor Pitx2. The nerve growth paused in the plexus for more than two days, persisting during primary extraocular myogenesis, with a subsequent phase in which the nerve branched out to specific muscles. To test the functional significance of the nerve contact with Pitx2+ cells in the plexus, we used two strategies to genetically ablate Pitx2+ cells or muscle precursors early in nerve development. The first strategy used Myf5-Cre-mediated expression of diphtheria toxin A to ablate muscle precursors, leading to loss of extraocular muscles. The oculomotor axons navigated to the eye to form the main nerve, but subsequently largely failed to initiate terminal branches. The second strategy studied Pitx2 homozygous mutants, which have early apoptosis of Pitx2-expressing precursor cells, including precursors for extraocular muscles and other orbital tissues. Oculomotor nerve fibers also grew to the eye, but failed to stop to form the plexus, instead grew long ectopic projections. These results show that neither Pitx2 function nor Myf5-expressing cells are required for oculomotor nerve navigation to the eye. However, Pitx2 function is required for oculomotor axons to pause growth in the plexus, while Myf5-expressing cells are required for terminal branch initiation.


Subject(s)
Oculomotor Muscles/innervation , Oculomotor Nerve/embryology , Animals , Axons/metabolism , Female , Gene Expression/genetics , Gene Expression Regulation/genetics , Homeodomain Proteins/metabolism , Mice , Muscle Development , Myogenic Regulatory Factor 5/metabolism , Oculomotor Muscles/growth & development , Oculomotor Muscles/metabolism , Oculomotor Nerve/metabolism , Pregnancy , Transcription Factors/metabolism , Homeobox Protein PITX2
3.
Development ; 147(20)2020 10 29.
Article in English | MEDLINE | ID: mdl-32994163

ABSTRACT

Slit is a secreted protein that has a canonical function of repelling growing axons from the CNS midline. The full-length Slit (Slit-FL) is cleaved into Slit-N and Slit-C fragments, which have potentially distinct functions via different receptors. Here, we report that the BMP-1/Tolloid family metalloprotease Tolkin (Tok) is responsible for Slit proteolysis in vivo and in vitro. In Drosophilatok mutants lacking Slit cleavage, midline repulsion of axons occurs normally, confirming that Slit-FL is sufficient to repel axons. However, longitudinal axon guidance is highly disrupted in tok mutants and can be rescued by midline expression of Slit-N, suggesting that Slit is the primary substrate for Tok in the embryonic CNS. Transgenic restoration of Slit-N or Slit-C does not repel axons in Slit-null flies. Slit-FL and Slit-N are both biologically active cues with distinct axon guidance functions in vivo Slit signaling is used in diverse biological processes; therefore, differentiating between Slit-FL and Slit fragments will be essential for evaluating Slit function in broader contexts.


Subject(s)
Axons/metabolism , Bone Morphogenetic Protein 1/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Nerve Tissue Proteins/metabolism , Proteolysis , Amino Acid Sequence , Animals , Axon Guidance , Cell Membrane/metabolism , Drosophila Proteins/chemistry , Epistasis, Genetic , Extracellular Space/metabolism , Models, Biological , Mutation/genetics , Nerve Tissue Proteins/chemistry , Phenotype , Protein Binding
4.
RNA ; 26(10): 1414-1430, 2020 10.
Article in English | MEDLINE | ID: mdl-32522888

ABSTRACT

The majority of mouse and human genes are subject to alternative cleavage and polyadenylation (APA), which most often leads to the expression of two or more alternative length 3' untranslated region (3'-UTR) mRNA isoforms. In neural tissues, there is enhanced expression of APA isoforms with longer 3'-UTRs on a global scale, but the physiological relevance of these alternative 3'-UTR isoforms is poorly understood. Calmodulin 1 (Calm1) is a key integrator of calcium signaling that generates short (Calm1-S) and long (Calm1-L) 3'-UTR mRNA isoforms via APA. We found Calm1-L expression to be largely restricted to neural tissues in mice including the dorsal root ganglion (DRG) and hippocampus, whereas Calm1-S was more broadly expressed. smFISH revealed that both Calm1-S and Calm1-L were subcellularly localized to neural processes of primary hippocampal neurons. In contrast, cultured DRG showed restriction of Calm1-L to soma. To investigate the in vivo functions of Calm1-L, we implemented a CRISPR-Cas9 gene editing strategy to delete a small region encompassing the Calm1 distal poly(A) site. This eliminated Calm1-L expression while maintaining expression of Calm1-S Mice lacking Calm1-L (Calm1ΔL/ΔL ) exhibited disorganized DRG migration in embryos, and reduced experience-induced neuronal activation in the adult hippocampus. These data indicate that Calm1-L plays functional roles in the central and peripheral nervous systems.


Subject(s)
3' Untranslated Regions/genetics , CRISPR-Cas Systems/genetics , Calmodulin/genetics , Ganglia, Spinal/physiology , Hippocampus/physiology , Neurons/physiology , RNA Isoforms/genetics , RNA, Messenger/genetics , Animals , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Female , Gene Editing/methods , Mice , Mice, Inbred C57BL , Polyadenylation/genetics , Pregnancy
5.
Dev Biol ; 455(2): 449-457, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31356769

ABSTRACT

The developing spinal cord builds a boundary between the CNS and the periphery, in the form of a basement membrane. The spinal cord basement membrane is a barrier that retains CNS neuron cell bodies, while being selectively permeable to specific axon types. Spinal motor neuron cell bodies are located in the ventral neural tube next to the floor plate and project their axons out through the basement membrane to peripheral targets. However, little is known about how spinal motor neuron cell bodies are retained inside the ventral neural tube, while their axons can exit. In previous work, we found that disruption of Slit/Robo signals caused motor neuron emigration outside the spinal cord. In the current study, we investigate how Slit/Robo signals are necessary to keep spinal motor neurons within the neural tube. Our findings show that when Slit/Robo signals were removed from motor neurons, they migrated outside the spinal cord. Furthermore, this emigration was associated with abnormal basement membrane protein expression in the ventral spinal cord. Using Robo2 and Slit2 conditional mutants, we found that motor neuron-derived Slit/Robo signals were required to set up a normal basement membrane in the spinal cord. Together, our results suggest that motor neurons produce Slit signals that are required for the basement membrane assembly to retain motor neuron cell bodies within the spinal cord.


Subject(s)
Basement Membrane/physiology , Intercellular Signaling Peptides and Proteins/physiology , Motor Neurons/cytology , Nerve Tissue Proteins/physiology , Neural Tube/cytology , Receptors, Immunologic/physiology , Spinal Cord/embryology , Animals , Cell Movement , Dystroglycans/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mutation , Nerve Tissue Proteins/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/physiology , Receptors, Immunologic/genetics , Signal Transduction , Spinal Cord/cytology , Roundabout Proteins
6.
Neuroscience ; 402: 116-129, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30685539

ABSTRACT

The facial nerve is necessary for our ability to eat, speak, and make facial expressions. Both the axons and cell bodies of the facial nerve undergo a complex embryonic developmental pattern involving migration of the cell bodies caudally and tangentially through rhombomeres, and simultaneously the axons projecting to exit the hindbrain to form the facial nerve. Our goal in this study was to test the functions of the chemorepulsive receptors Robo1 and Robo2 in facial neuron migration and axon projection by analyzing genetically marked motor neurons in double-mutant mouse embryos through the migration time course, E10.0-E13.5. In Robo1/2 double mutants, axon projection and cell body migration errors were more severe than in single mutants. Most axons did not make it to their motor exit point, and instead projected into and longitudinally within the floor plate. Surprisingly, some facial neurons had multiple axons exiting and projecting into the floor plate. At the same time, a subset of mutant facial cell bodies failed to migrate caudally, and instead either streamed dorsally toward the exit point or shifted into the floor plate. We conclude that Robo1 and Robo2 have redundant functions to guide multiple aspects of the complex cell migration of the facial nucleus, as well as regulating axon trajectories and suppressing formation of ectopic axons.


Subject(s)
Axon Guidance , Axons/physiology , Cell Movement , Facial Nerve/embryology , Nerve Tissue Proteins/physiology , Receptors, Immunologic/physiology , Rhombencephalon/embryology , Animals , Mice, Transgenic , Motor Neurons/physiology , Roundabout Proteins
7.
Semin Cell Dev Biol ; 85: 78-83, 2019 01.
Article in English | MEDLINE | ID: mdl-29141180

ABSTRACT

Motor neurons differentiate from progenitor cells and cluster as motor nuclei, settling next to the floor plate in the brain stem and spinal cord. Although precise positioning of motor neurons is critical for their functional input and output, the molecular mechanisms that guide motor neurons to their proper positions remain poorly understood. Here, we review recent evidence of motor neuron positioning mechanisms, highlighting situations in which motor neuron cell bodies can migrate, and experiments that show that their migration is regulated by axon guidance cues. The view that emerges is that motor neurons are actively trapped or restricted in static positions, as the cells balance a push in the dorsal direction by repulsive Slit/Robo cues and a pull in the ventral direction by attractive Netrin-1/DCC cues. These new functions of guidance cues are necessary fine-tuning to set up patterns of motor neurons at their proper positions in the neural tube during embryogenesis.


Subject(s)
Axon Guidance , Cell Movement , Motor Neurons/cytology , Motor Neurons/metabolism , Neurogenesis/genetics
8.
Front Neuroanat ; 12: 96, 2018.
Article in English | MEDLINE | ID: mdl-30483071

ABSTRACT

Stereotypic cell migrations in the developing brain are fundamental for the proper patterning of brain regions and formation of neural networks. In this work, we uncovered in the developing rat, a population of neurons expressing tyrosine hydroxylase (TH) that migrates posteriorly from the alar plate of the midbrain, in neurophilic interaction with axons of the mesencephalic nucleus of the trigeminal nerve. A fraction of this population was also shown to traverse the mid-hindbrain boundary, reaching the vicinity of the locus coeruleus (LC) in rhombomere 1 (r1). This migratory population, however, does not have a noradrenergic (NA) phenotype and, in keeping with its midbrain origin, expresses Otx2 which is down regulated upon migration into the hindbrain. The interaction with the trigeminal mesencephalic axons is necessary for the arrangement and distribution of migratory cells as these aspects are dramatically altered in whole embryo cultures upon disruption of trigeminal axon projection by interfering with DCC function. Moreover, in mouse embryos in an equivalent developmental stage, we detected a cell population that also migrates caudally within the midbrain apposed to mesencephalic trigeminal axons but that does not express TH; a fraction of this population expresses calbindin instead. Overall, our work identified TH-expressing neurons from the rat midbrain alar plate that migrate tangentially over long distances within the midbrain and into the hindbrain by means of a close interaction with trigeminal mesencephalic axons. A different migratory population in this region and also in mouse embryos revealed diversity among the cells that follow this descending migratory pathway.

9.
J Dev Biol ; 6(4)2018 Oct 18.
Article in English | MEDLINE | ID: mdl-30340315

ABSTRACT

Navigating growth cones are exposed to multiple signals simultaneously and have to integrate competing cues into a coherent navigational response. Integration of guidance cues is traditionally thought to occur at the level of cytoskeletal dynamics. Drosophila studies indicate that cells exhibit a low level of continuous caspase protease activation, and that axon guidance cues can activate or suppress caspase activity. We base a model for axon guidance on these observations. By analogy with other systems in which caspase signaling has non-apoptotic functions, we propose that caspase signaling can either reinforce repulsion or negate attraction in response to external guidance cues by cleaving cytoskeletal proteins. Over the course of an entire trajectory, incorrectly navigating axons may pass the threshold for apoptosis and be eliminated, whereas axons making correct decisions will survive. These observations would also explain why neurotrophic factors can act as axon guidance cues and why axon guidance systems such as Slit/Robo signaling may act as tumor suppressors in cancer.

10.
Nat Cell Biol ; 20(10): 1159-1171, 2018 10.
Article in English | MEDLINE | ID: mdl-30250061

ABSTRACT

Actin-based protrusions are reinforced through positive feedback, but it is unclear what restricts their size, or limits positive signals when they retract or split. We identify an evolutionarily conserved regulator of actin-based protrusion: CYRI (CYFIP-related Rac interactor) also known as Fam49 (family of unknown function 49). CYRI binds activated Rac1 via a domain of unknown function (DUF1394) shared with CYFIP, defining DUF1394 as a Rac1-binding module. CYRI-depleted cells have broad lamellipodia enriched in Scar/WAVE, but reduced protrusion-retraction dynamics. Pseudopods induced by optogenetic Rac1 activation in CYRI-depleted cells are larger and longer lived. Conversely, CYRI overexpression suppresses recruitment of active Scar/WAVE to the cell edge, resulting in short-lived, unproductive protrusions. CYRI thus focuses protrusion signals and regulates pseudopod complexity by inhibiting Scar/WAVE-induced actin polymerization. It thus behaves like a 'local inhibitor' as predicted in widely accepted mathematical models, but not previously identified in cells. CYRI therefore regulates chemotaxis, cell migration and epithelial polarization by controlling the polarity and plasticity of protrusions.


Subject(s)
Cell Movement , Intracellular Signaling Peptides and Proteins/metabolism , Pseudopodia/metabolism , rac1 GTP-Binding Protein/metabolism , Actins/genetics , Actins/metabolism , Animals , COS Cells , Cell Line, Tumor , Chemotaxis/genetics , Chlorocebus aethiops , Dogs , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Madin Darby Canine Kidney Cells , Polymerization , Protein Binding , Pseudopodia/genetics , Signal Transduction/genetics , rac1 GTP-Binding Protein/genetics
11.
Dev Biol ; 432(1): 178-191, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28986144

ABSTRACT

In the spinal cord, motor axons project out the neural tube at specific exit points, then bundle together to project toward target muscles. The molecular signals that guide motor axons to and out of their exit points remain undefined. Since motor axons and their exit points are located near the floor plate, guidance signals produced by the floor plate and adjacent ventral tissues could influence motor axons as they project toward and out of exit points. The secreted Slit proteins are major floor plate repellents, and motor neurons express two Slit receptors, Robo1 and Robo2. Using mutant mouse embryos at early stages of motor axon exit, we found that motor exit points shifted ventrally in Robo1/2 or Slit1/2 double mutants. Along with the ventral shift, mutant axons had abnormal trajectories both within the neural tube toward the exit point, and after exit into the periphery. In contrast, the absence of the major ventral attractant, Netrin-1, or its receptor, DCC caused motor exit points to shift dorsally. Netrin-1 attraction on spinal motor axons was demonstrated by in vitro explant assays, showing that Netrin-1 increased outgrowth and attracted cultured spinal motor axons. The opposing effects of Slit/Robo and Netrin-1/DCC signals were tested genetically by combining Netrin-1 and Robo1/2 mutations. The location of exit points in the combined mutants was significantly recovered to their normal position compared to Netrin-1 or Robo1/2 mutants. Together, these results suggest that the proper position of motor exit points is determined by a "push-pull" mechanism, pulled ventrally by Netrin-1/DCC attraction and pushed dorsally by Slit/Robo repulsion.


Subject(s)
Axons/physiology , Glycoproteins/physiology , Motor Neurons/physiology , Nerve Tissue Proteins/physiology , Netrins/physiology , Spinal Cord/physiology , Animals , Axons/metabolism , Cell Movement/physiology , DCC Receptor/metabolism , Mice , Motor Neurons/cytology , Motor Neurons/metabolism , Nerve Tissue Proteins/metabolism , Netrins/metabolism , Neural Tube/cytology , Neural Tube/metabolism , Neural Tube/physiology , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Signal Transduction/genetics , Spinal Cord/cytology , Spinal Cord/metabolism , Tumor Suppressor Proteins/metabolism , Roundabout Proteins
12.
Sci Rep ; 6: 36491, 2016 11 07.
Article in English | MEDLINE | ID: mdl-27819291

ABSTRACT

LIM-homeodomain (HD) transcription factors form a multimeric complex and assign neuronal subtype identities, as demonstrated by the hexameric ISL1-LHX3 complex which gives rise to somatic motor (SM) neurons. However, the roles of combinatorial LIM code in motor neuron diversification and their subsequent differentiation is much less well understood. In the present study, we demonstrate that the ISL1 controls postmitotic cranial branchiomotor (BM) neurons including the positioning of the cell bodies and peripheral axon pathfinding. Unlike SM neurons, which transform into interneurons, BM neurons are normal in number and in marker expression in Isl1 mutant mice. Nevertheless, the movement of trigeminal and facial BM somata is stalled, and their peripheral axons are fewer or misrouted, with ectopic branches. Among genes whose expression level changes in previous ChIP-seq and microarray analyses in Isl1-deficient cell lines, we found that Slit2 transcript was almost absent from BM neurons of Isl1 mutants. Both ISL1-LHX3 and ISL1-LHX4 bound to the Slit2 enhancer and drove endogenous Slit2 expression in SM and BM neurons. Our findings suggest that combinations of ISL1 and LHX factors establish cell-type specificity and functional diversity in terms of motor neuron identities and/or axon development.


Subject(s)
Intercellular Signaling Peptides and Proteins/genetics , LIM-Homeodomain Proteins/genetics , Motor Neurons/physiology , Nerve Tissue Proteins/genetics , Neurogenesis/genetics , Transcription Factors/genetics , Animals , Axons/physiology , Cell Differentiation/genetics , Gene Expression Regulation, Developmental/genetics , Interneurons/physiology , Mice , Mice, Inbred C57BL , Transcription, Genetic/genetics , Trigeminal Motor Nucleus/physiology
13.
Neural Dev ; 11(1): 18, 2016 10 22.
Article in English | MEDLINE | ID: mdl-27770832

ABSTRACT

BACKGROUND: Oculomotor neurons develop initially like typical motor neurons, projecting axons out of the ventral midbrain to their ipsilateral targets, the extraocular muscles. However, in all vertebrates, after the oculomotor nerve (nIII) has reached the extraocular muscle primordia, the cell bodies that innervate the superior rectus migrate to join the contralateral nucleus. This motor neuron migration represents a unique strategy to form a contralateral motor projection. Whether migration is guided by diffusible cues remains unknown. METHODS: We examined the role of Slit chemorepellent signals in contralateral oculomotor migration by analyzing mutant mouse embryos. RESULTS: We found that the ventral midbrain expresses high levels of both Slit1 and 2, and that oculomotor neurons express the repellent Slit receptors Robo1 and Robo2. Therefore, Slit signals are in a position to influence the migration of oculomotor neurons. In Slit 1/2 or Robo1/2 double mutant embryos, motor neuron cell bodies migrated into the ventral midbrain on E10.5, three days prior to normal migration. These early migrating neurons had leading projections into and across the floor plate. In contrast to the double mutants, embryos which were mutant for single Slit or Robo genes did not have premature migration or outgrowth on E10.5, demonstrating a cooperative requirement of Slit1 and 2, as well as Robo1 and 2. To test how Slit/Robo midline repulsion is modulated, we found that the normal migration did not require the receptors Robo3 and CXCR4, or the chemoattractant, Netrin 1. The signal to initiate contralateral migration is likely autonomous to the midbrain because oculomotor neurons migrate in embryos that lack either nerve outgrowth or extraocular muscles, or in cultured midbrains that lacked peripheral tissue. CONCLUSION: Overall, our results demonstrate that a migratory subset of motor neurons respond to floor plate-derived Slit repulsion to properly control the timing of contralateral migration.


Subject(s)
Axon Guidance , Cell Movement , Intercellular Signaling Peptides and Proteins/physiology , Motor Neurons/physiology , Nerve Tissue Proteins/physiology , Oculomotor Nerve/growth & development , Receptors, Immunologic/physiology , Animals , Membrane Proteins/physiology , Mesencephalon/physiology , Mice , Nerve Growth Factors/physiology , Netrin-1 , Receptors, CXCR4/physiology , Receptors, Cell Surface , Signal Transduction , Tumor Suppressor Proteins/physiology , Roundabout Proteins
14.
Brain Struct Funct ; 221(1): 665-78, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25366972

ABSTRACT

The retroflex tract contains medial habenula efferents that target the hindbrain interpeduncular complex and surrounding areas. This tract displays a singular course. Initially, habenular axons extend ventralwards in front of the pretectum until they reach the basal plate. Next, they avoid crossing the local floor plate, sharply changing course caudalwards (the retroflexion alluded by the tract name) and navigate strictly antero-posteriorly across basal pretectum, midbrain and isthmus. Once they reach rhombomere 1, the habenular axons criss-cross the floor plate several times within the interpeduncular nuclear complex as they innervate it. Here we described the timing and details of growth phenomena as these axons navigate to their target. The first dorsoventral course apparently obeys Ntn1 attraction. We checked the role of local floor plate signaling in the decision to avoid the thalamic floor plate and bend caudalwards. Analyzing the altered floor and basal plates of Gli2 knockout mice, we found a contralateral projection of most habenular axons, plus ulterior bizarre navigation rostralwards. This crossing phenotype was due to a reduced expression of Slit repulsive cues, suggesting involvement of the floor-derived Robo-Slit system in the normal guidance of this tract. Using Slit and Robo mutant mice, open neural tube and co-culture assays, we determined that Robo1-Slit2 interaction is specifically required for impeding that medial habenular axons cross the thalamic floor plate. This pathfinding mechanism is essential to establish the functionally important habenulo-interpeduncular connection.


Subject(s)
Cell Movement , Habenula/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptors, Immunologic/metabolism , Thalamus/metabolism , Animals , Axons/metabolism , COS Cells , Chlorocebus aethiops , Coculture Techniques , Gene Expression Regulation, Developmental , Genotype , Gestational Age , Habenula/embryology , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Phenotype , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Signal Transduction , Thalamus/embryology , Tissue Culture Techniques , Transfection , Zinc Finger Protein Gli2 , Roundabout Proteins
15.
J Neurosci ; 35(33): 11707-18, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-26290247

ABSTRACT

Commissural axons grow along precise trajectories that are guided by several cues secreted from the ventral midline. After initial attraction to the floor plate using Netrin1 activation of its main attractive receptor, DCC (deleted in colorectal cancer), axons cross the ventral midline, and many turn to grow longitudinally on the contralateral side. After crossing the midline, axons are thought to lose their responsiveness to Netrin1 and become sensitive to midline Slit-Robo repulsion. We aimed to address the in vivo significance of Netrin1 in guiding post-crossing axon trajectories in mouse embryos. Surprisingly, in contrast to the spinal cord, Netrin1 and DCC mutants had abundant commissural axons crossing in the hindbrain. In Netrin1 and DCC mutants, many post-crossing axons made normal turns to grow longitudinally, but projected abnormally at angles away from the midline. In addition, exposure of cultured hindbrain explants to ectopic Netrin1 caused attractive deflection of post-crossing axons. Thus, Netrin1-DCC signaling is not required to attract pre-crossing axons toward the hindbrain floor plate, but is active in post-crossing guidance. Also in contrast with spinal cord, analysis of hindbrain post-crossing axons in Robo1/2 mutant embryos showed that Slit-Robo repulsive signaling was not required for post-crossing trajectories. Our findings show that Netrin1-DCC attractive signaling, but not Slit-Robo repulsive signaling, remains active in hindbrain post-crossing commissural axons to guide longitudinal trajectories, suggesting surprising regional diversity in commissural axon guidance mechanisms. SIGNIFICANCE STATEMENT: The left and right sides of the brainstem and spinal cord are connected primarily by axon fibers that grow across the ventral midline, and then away on the other side to their targets. Based on spinal cord, axons are initially attracted by diffusible attractive protein signals to approach and cross the midline, and then are thought to switch to repulsive cues to grow away on the opposite side. Our results in the hindbrain show that the major midline attractant, Netrin1, is not required for midline crossing. However, the post-crossing axons depend on Netrin1 attraction to set their proper trajectories on the other side. Overall, these findings suggest that commissural axons use distinct mechanisms to navigate in different CNS regions.


Subject(s)
Axons/physiology , Nerve Growth Factors/metabolism , Neurogenesis/physiology , Receptors, Cell Surface/metabolism , Rhombencephalon/cytology , Rhombencephalon/physiopathology , Tumor Suppressor Proteins/metabolism , Animals , Axons/ultrastructure , Cells, Cultured , DCC Receptor , Female , Gene Expression Regulation, Developmental/physiology , Male , Mice , Netrin-1
16.
Exp Neurol ; 269: 17-27, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25843547

ABSTRACT

Motor neurons send out axons to peripheral muscles while their cell bodies remain in the ventral spinal cord. The unique configuration of motor neurons spanning the border between the CNS and PNS has been explained by structural barriers such as boundary cap (BC) cells, basal lamina and radial glia. However, mechanisms in motor neurons that retain their position have not been addressed yet. Here we demonstrate that the Islet1 (Isl1) and Islet2 (Isl2) transcription factors, which are essential for acquisition of motor neuron identity, also contribute to restrict motor neurons within the neural tube. In mice that lack both Isl1 and Isl2, large numbers of motor neurons exited the neural tube, even prior to the appearance of BC cells at the ventral exit points. Transcriptional profiling of motor neurons derived from Isl1 null embryonic stem cells revealed that transcripts of major genes involved in repulsive mechanisms were misregulated. Particularly, expression of Neuropilin1 (Npr1) and Slit2 mRNA was diminished in Islet mutant mice, and these could be target genes of the Islet proteins. Consistent with this mechanism, Robo and Slit mutations in mice and knockdown of Npr1 and Slit2 in chick embryos caused motor neurons to migrate to the periphery. Together, our study suggests that Islet genes engage Robo-Slit and Neuropilin-Semaphorin signaling in motor neurons to retain motor somata within the CNS.


Subject(s)
Intercellular Signaling Peptides and Proteins/genetics , LIM-Homeodomain Proteins/genetics , Motor Neurons/metabolism , Nerve Tissue Proteins/genetics , Neural Tube/metabolism , Neuropilin-1/genetics , Semaphorins/metabolism , Transcription Factors/genetics , Animals , Axons/physiology , Cell Movement/physiology , Chick Embryo , Gene Expression Regulation, Developmental/genetics , LIM-Homeodomain Proteins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Neuropilin-1/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism
17.
Dev Biol ; 399(1): 68-79, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25530182

ABSTRACT

Motor neurons differentiate from a ventral column of progenitors and settle in static clusters, the motor nuclei, next to the floor plate. Within these cell clusters, motor neurons receive afferent input and project their axons out to muscle targets. The molecular mechanisms that position motor neurons in the neural tube remain poorly understood. The floor plate produces several types of guidance cues with well-known roles in attracting and repelling axons, including the Slit family of chemorepellents via their Robo receptors, and Netrin1 via its DCC attractive receptor. In the present study we found that Islet1(+) motor neuron cell bodies invaded the floor plate of Robo1/2 double mutant mouse embryos or Slit1/2/3 triple mutants. Misplaced neurons were born in their normal progenitor column, but then migrated tangentially into the ventral midline. Robo1 and 2 receptor expression in motor neurons was confirmed by reporter gene staining and anti-Robo antibody labeling. Mis-positioned motor neurons projected their axons longitudinally within the floor plate, and failed to reach their normal exit points. To test for potential counteracting ventral attractive signals, we examined Netrin-1 and DCC mutants, and found that motor neurons shifted dorsally in the hindbrain and spinal cord, suggesting that Netrin-1/DCC signaling normally attracts motor neurons closer to the floor plate. Our results show that motor neurons are actively migrating cells, and are normally trapped in a static position by Slit/Robo repulsion and Netrin-1/DCC attraction.


Subject(s)
Motor Neurons/metabolism , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Immunologic/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Axons/metabolism , Cell Body/metabolism , Cell Movement/genetics , Cell Movement/physiology , DCC Receptor , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Mice, Transgenic , Microscopy, Fluorescence , Mutation , Nerve Growth Factors/genetics , Nerve Tissue Proteins/genetics , Netrin-1 , Receptors, Cell Surface/genetics , Receptors, Immunologic/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Roundabout Proteins
18.
Neural Dev ; 9: 17, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-25056828

ABSTRACT

BACKGROUND: Longitudinal axons grow parallel to the embryonic midline to connect distant regions of the central nervous system. Previous studies suggested that repulsive midline signals guide pioneer longitudinal axons by blocking their entry into the floor plate; however, the role of midline attractants, and whether attractant signals may cooperate with repulsive signals, remains unclear. In this study we investigated the navigation of a set of pioneer longitudinal axons, the medial longitudinal fasciculus, in mouse embryos mutant for the Netrin/Deleted in Colorectal Cancer (DCC) attractants, and for Slit repellents, as well as the responses of explanted longitudinal axons in vitro. RESULTS: In mutants for Netrin1 chemoattractant or DCC receptor signaling, longitudinal axons shifted away from the ventral midline, suggesting that Netrin1/DCC signals act attractively to pull axons ventrally. Analysis of mutants in the three Slit genes, including Slit1/2/3 triple mutants, suggest that concurrent repulsive Slit/Robo signals push pioneer axons away from the ventral midline. Combinations of mutations between the Netrin and Slit guidance systems provided genetic evidence that the attractive and repulsive signals balance against each other. This balance is demonstrated in vitro using explant culture, finding that the cues can act directly on longitudinal axons. The explants also reveal an unexpected synergy of Netrin1 and Slit2 that promotes outgrowth. CONCLUSIONS: These results support a mechanism in which longitudinal trajectories are positioned by a push-pull balance between opposing Netrin and Slit signals. Our evidence suggests that longitudinal axons respond directly and simultaneously to both attractants and repellents, and that the combined signals constrain axons to grow longitudinally.


Subject(s)
Axons/physiology , Chemotaxis , Intracellular Signaling Peptides and Proteins/metabolism , Mesencephalon/embryology , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mesencephalon/metabolism , Mice , Mice, Transgenic , Nerve Growth Factors/genetics , Nerve Tissue Proteins/genetics , Netrin-1 , Neurons/cytology , Neurons/physiology , Tumor Suppressor Proteins/genetics
19.
Proc Natl Acad Sci U S A ; 111(28): E2851-7, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24982181

ABSTRACT

Ablation of a single miRNA gene rarely leads to a discernable developmental phenotype in mice, in some cases because of compensatory effects by other functionally related miRNAs. Here, we report that simultaneous inactivation of two functionally related miRNA clusters (miR-34b/c and miR-449) encoding five miRNAs (miR-34b, miR-34c, miR-449a, miR-449b, and miR-449c) led to sexually dimorphic, partial perinatal lethality, growth retardation, and infertility. These developmental defects correlated with the dysregulation of ∼ 240 target genes, which are mainly involved in three major cellular functions, including cell-fate control, brain development and microtubule dynamics. Our data demonstrate an essential role of a miRNA family in brain development, motile ciliogenesis, and spermatogenesis.


Subject(s)
Brain/embryology , Gene Expression Regulation, Developmental/physiology , MicroRNAs/metabolism , Multigene Family/physiology , Spermatogenesis/physiology , Animals , Cilia/genetics , Cilia/metabolism , Infertility, Male/genetics , Infertility, Male/metabolism , Male , Mice , Mice, Knockout , MicroRNAs/genetics
20.
Front Neuroanat ; 8: 43, 2014.
Article in English | MEDLINE | ID: mdl-24926237

ABSTRACT

The Nigrostriatal pathway (NSP) is formed by dopaminergic axons that project from the ventral midbrain to the dorsolateral striatum as part of the medial forebrain bundle. Previous studies have implicated chemotropic proteins in the formation of the NSP during development but little is known of the role of substrate-anchored signals in this process. We observed in mouse and rat embryos that midbrain dopaminergic axons ascend in close apposition to descending GAD65-positive axon bundles throughout their trajectory to the striatum. To test whether such interaction is important for dopaminergic axon pathfinding, we analyzed transgenic mouse embryos in which the GAD65 axon bundle was reduced by the conditional expression of the diphtheria toxin. In these embryos we observed dopaminergic misprojection into the hypothalamic region and abnormal projection in the striatum. In addition, analysis of Robo1/2 and Slit1/2 knockout embryos revealed that the previously described dopaminergic misprojection in these embryos is accompanied by severe alterations in the GAD65 axon scaffold. Additional studies with cultured dopaminergic neurons and whole embryos suggest that NCAM and Robo proteins are involved in the interaction of GAD65 and dopaminergic axons. These results indicate that the fasciculation between descending GAD65 axon bundles and ascending dopaminergic axons is required for the stereotypical NSP formation during brain development and that known guidance cues may determine this projection indirectly by instructing the pathfinding of the axons that are part of the GAD65 axon scaffold.

SELECTION OF CITATIONS
SEARCH DETAIL
...