Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
2.
Neurosci Lett ; 716: 134621, 2020 01 18.
Article in English | MEDLINE | ID: mdl-31726180

ABSTRACT

Amyotrophic Lateral Sclerosis (ALS) is a devastating and fatal neurodegenerative disease affecting approximately 30,000 individuals in the United States. The average age of onset is 55 years and progression of the disease is rapid with most patients dying of respiratory failure within 3-5 years. Currently available therapeutics have modest effects on patient survival, underscoring the immediate need for more effective medicines. Recent technological advances in next generation sequencing have led to a substantial uptick in the discovery of genes linked to ALS. Since 90 % of ALS cases are sporadic, risk genes identified in familial cases provide invaluable insights into the molecular pathogenesis of the disease. Most notably, TDP-43-expressing neuronal inclusions and C9orf72 mutations have emerged as the key pathological and genetic hallmarks, respectively, of ALS. In this review, we will discuss recent advances in modifiers of TDP-43 toxicity, with an emphasis on Ataxin-2, one of the most well-characterized TDP-43 modifiers. An understanding of Ataxin-2 function and related biological pathways could provide a framework for the discovery of other novel modifiers of TDP-43. We will also describe the pathogenic mechanisms underlying C9orf72 toxicity and how these impact the disease process. Finally, we will explore emerging therapeutic strategies for dampening TDP-43 and C9orf72 toxicity and, ultimately, slowing or halting the progression of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , C9orf72 Protein , DNA-Binding Proteins , Animals , Humans
4.
Curr Biol ; 26(17): 2379-87, 2016 09 12.
Article in English | MEDLINE | ID: mdl-27451901

ABSTRACT

Sensory dendrite arbors are patterned through cell-autonomously and non-cell-autonomously functioning factors [1-3]. Yet, only a few non-cell-autonomously acting proteins have been identified, including semaphorins [4, 5], brain-derived neurotrophic factors (BDNFs) [6], UNC-6/Netrin [7], and the conserved MNR-1/Menorin-SAX-7/L1CAM cell adhesion complex [8, 9]. This complex acts from the skin to pattern the stereotypic dendritic arbors of PVD and FLP somatosensory neurons in Caenorhabditis elegans through the leucine-rich transmembrane receptor DMA-1/LRR-TM expressed on PVD neurons [8, 9]. Here we describe a role for the diffusible C. elegans protein LECT-2, which is homologous to vertebrate leukocyte cell-derived chemotaxin 2 (LECT2)/Chondromodulin II. LECT2/Chondromodulin II has been implicated in a variety of pathological conditions [10-13], but the developmental functions of LECT2 have remained elusive. We find that LECT-2/Chondromodulin II is required for development of PVD and FLP dendritic arbors and can act as a diffusible cue from a distance to shape dendritic arbors. Expressed in body-wall muscles, LECT-2 decorates neuronal processes and hypodermal cells in a pattern similar to the cell adhesion molecule SAX-7/L1CAM. LECT-2 functions genetically downstream of the MNR-1/Menorin-SAX-7/L1CAM adhesion complex and upstream of the DMA-1 receptor. LECT-2 localization is dependent on SAX-7/L1CAM, but not on MNR-1/Menorin or DMA-1/LRR-TM, suggesting that LECT-2 functions as part of the skin-derived MNR-1/Menorin-SAX-7/L1CAM adhesion complex. Collectively, our findings suggest that LECT-2/Chondromodulin II acts as a muscle-derived, diffusible cofactor together with a skin-derived cell adhesion complex to orchestrate the molecular interactions of three tissues during patterning of somatosensory dendrites.


Subject(s)
Body Patterning , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/physiology , Chemotactic Factors/genetics , Amino Acid Sequence , Animals , Base Sequence , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/metabolism , Chemotactic Factors/chemistry , Chemotactic Factors/metabolism , Cues , Dendrites/physiology , Larva/genetics , Larva/growth & development , Larva/physiology , Muscles/metabolism , Sequence Alignment , Skin/metabolism
5.
Genes Dev ; 29(24): 2617-32, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26680304

ABSTRACT

Commissural axon guidance depends on a myriad of cues expressed by intermediate targets. Secreted semaphorins signal through neuropilin-2/plexin-A1 receptor complexes on post-crossing commissural axons to mediate floor plate repulsion in the mouse spinal cord. Here, we show that neuropilin-2/plexin-A1 are also coexpressed on commissural axons prior to midline crossing and can mediate precrossing semaphorin-induced repulsion in vitro. How premature semaphorin-induced repulsion of precrossing axons is suppressed in vivo is not known. We discovered that a novel source of floor plate-derived, but not axon-derived, neuropilin-2 is required for precrossing axon pathfinding. Floor plate-specific deletion of neuropilin-2 significantly reduces the presence of precrossing axons in the ventral spinal cord, which can be rescued by inhibiting plexin-A1 signaling in vivo. Our results show that floor plate-derived neuropilin-2 is developmentally regulated, functioning as a molecular sink to sequester semaphorins, preventing premature repulsion of precrossing axons prior to subsequent down-regulation, and allowing for semaphorin-mediated repulsion of post-crossing axons.


Subject(s)
Axons/physiology , Commissural Interneurons/physiology , Neuropilin-2/metabolism , Semaphorins/metabolism , Animals , Cells, Cultured , Commissural Interneurons/cytology , Embryo, Mammalian , Gene Deletion , Gene Expression Regulation, Developmental , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuropilin-2/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signal Transduction
6.
Cell ; 155(2): 308-20, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24120132

ABSTRACT

Sensory dendrites depend on cues from their environment to pattern their growth and direct them toward their correct target tissues. Yet, little is known about dendrite-substrate interactions during dendrite morphogenesis. Here, we describe MNR-1/menorin, which is part of the conserved Fam151 family of proteins and is expressed in the skin to control the elaboration of "menorah"-like dendrites of mechanosensory neurons in Caenorhabditis elegans. We provide biochemical and genetic evidence that MNR-1 acts as a contact-dependent or short-range cue in concert with the neural cell adhesion molecule SAX-7/L1CAM in the skin and through the neuronal leucine-rich repeat transmembrane receptor DMA-1 on sensory dendrites. Our data describe an unknown pathway that provides spatial information from the skin substrate to pattern sensory dendrite development nonautonomously.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Dendrites/metabolism , Membrane Proteins/metabolism , Neurons/metabolism , Amino Acid Sequence , Animals , Caenorhabditis elegans Proteins/genetics , Cloning, Molecular , Gene Knockdown Techniques , Membrane Proteins/genetics , Molecular Sequence Data , Sequence Alignment
7.
Neural Dev ; 8: 15, 2013 Jul 31.
Article in English | MEDLINE | ID: mdl-23902858

ABSTRACT

BACKGROUND: Spinal commissural axons represent a model system for deciphering the molecular logic that regulates the guidance of midline-crossing axons in the developing central nervous system (CNS). Whether the same or specific sets of guidance signals control the navigation of molecularly distinct subtypes of these axons remains an open and largely unexplored question. Although it is well established that post-crossing commissural axons alter their responsiveness to midline-associated guidance cues, our understanding of the repulsive mechanisms that drive the post-crossing segments of these axons away from the midline and whether the underlying guidance systems operate in a commissural axon subtype-specific manner, remains fragmentary at best. RESULTS: Here, we utilize axonally targeted transgenic reporter mice to visualize genetically distinct dorsal interneuron (dI)1 and dI4 commissural axons and show that the repulsive class 3 semaphorin (Sema3) guidance receptor Neuropilin 2 (Npn2), is selectively expressed on the dI1 population and is required for the guidance of post-crossing dI1, but not dI4, axons. Consistent with these observations, the midline-associated Npn2 ligands, Sema3F and Sema3B, promote the collapse of dI1, but not dI4, axon-associated growth cones in vitro. We also identify, for the first time, a discrete GABAergic population of ventral commissural neurons/axons in the embryonic mouse spinal cord that expresses Npn2, and show that Npn2 is required for the proper guidance of their post-crossing axons. CONCLUSIONS: Together, our findings indicate that Npn2 is selectively expressed in distinct populations of commissural neurons in both the dorsal and ventral spinal cord, and suggest that Sema3-Npn2 signaling regulates the guidance of post-crossing commissural axons in a population-specific manner.


Subject(s)
Axons/metabolism , Neuropilin-2/metabolism , Spinal Cord/metabolism , Animals , Gene Expression Regulation, Developmental/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuropilin-2/genetics , Spinal Cord/cytology , Spinal Cord/embryology
8.
J Neurosci ; 32(44): 15377-87, 2012 Oct 31.
Article in English | MEDLINE | ID: mdl-23115176

ABSTRACT

The axons of spinal projection neurons transmit sensory information to the brain by ascending within highly organized longitudinal tracts. However, the molecular mechanisms that control the sorting of these axons within the spinal cord and their directed growth to poorly defined targets are not understood. Here, we show that an interplay between Robo and the cell adhesion molecule, N-cadherin, sorts spinal commissural axons into appropriate longitudinal tracts within the spinal cord, and thereby facilitates their brain targeting. Specifically, we show that d1 and d2 spinal commissural axons join the lateral funiculus within the spinal cord and target the cerebellum in chick embryos, and that these axons contribute to the spinocerebellar projection in transgenic reporter mice. Disabling Robo signaling or overexpressing N-cadherin on these axons prevents the formation of the lateral funiculus and the spinocerebellar tract, and simultaneously perturbing Robo and N-cadherin function rescues both phenotypes in chick embryos. Consistent with these observations, disabling Robo function in conditional N-cadherin knock-out mice results in a wild-type-like lateral funiculus. Together, these findings suggest that spinal projection axons must be sorted into distinct longitudinal tracts within the spinal cord proper to project to their brain targets.


Subject(s)
Axons/physiology , Cadherins/physiology , Nerve Tissue Proteins/physiology , Receptors, Immunologic/physiology , Spinal Cord/physiology , Spinocerebellar Tracts/growth & development , Spinocerebellar Tracts/physiology , Animals , Cadherins/genetics , Cell Adhesion , Cerebellum/cytology , Cerebellum/growth & development , Cerebellum/physiology , Chick Embryo , Electroporation , Functional Laterality/physiology , Mice , Mice, Knockout , Mutation/genetics , Mutation/physiology , Phenotype , Plasmids/genetics , Receptors, Dopamine D1/physiology , Receptors, Dopamine D2/physiology , Rhombencephalon/physiology , Spinal Cord/cytology , Roundabout Proteins
9.
Front Mol Neurosci ; 5: 59, 2012.
Article in English | MEDLINE | ID: mdl-22586366

ABSTRACT

The directed and stereotypical growth of axons to their synaptic targets is a crucial phase of neural circuit formation. Many axons in the developing vertebrate and invertebrate central nervous systems (CNSs), including those that remain on their own (ipsilateral), and those that cross over to the opposite (commissural), side of the midline project over long distances along the anterior-posterior (A-P) body axis within precisely positioned longitudinally oriented tracts to facilitate the transmission of information between CNS regions. Despite the widespread distribution and functional importance of these longitudinal tracts, the mechanisms that regulate their formation and projection to poorly characterized synaptic targets remain largely unknown. Nevertheless, recent studies carried out in a variety of invertebrate and vertebrate model systems have begun to elucidate the molecular logic that controls longitudinal axon guidance.

10.
Development ; 139(8): 1435-46, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22399681

ABSTRACT

Mammalian motor circuits control voluntary movements by transmitting signals from the central nervous system (CNS) to muscle targets. To form these circuits, motor neurons (MNs) must extend their axons out of the CNS. Although exit from the CNS is an indispensable phase of motor axon pathfinding, the underlying molecular mechanisms remain obscure. Here, we present the first identification of a genetic pathway that regulates motor axon exit from the vertebrate spinal cord, utilizing spinal accessory motor neurons (SACMNs) as a model system. SACMNs are a homogeneous population of spinal MNs with axons that leave the CNS through a discrete lateral exit point (LEP) and can be visualized by the expression of the cell surface protein BEN. We show that the homeodomain transcription factor Nkx2.9 is selectively required for SACMN axon exit and identify the Robo2 guidance receptor as a likely downstream effector of Nkx2.9; loss of Nkx2.9 leads to a reduction in Robo2 mRNA and protein within SACMNs and SACMN axons fail to exit the spinal cord in Robo2-deficient mice. Consistent with short-range interactions between Robo2 and Slit ligands regulating SACMN axon exit, Robo2-expressing SACMN axons normally navigate through LEP-associated Slits as they emerge from the spinal cord, and fail to exit in Slit-deficient mice. Our studies support the view that Nkx2.9 controls SACMN axon exit from the mammalian spinal cord by regulating Robo-Slit signaling.


Subject(s)
Axons/metabolism , Homeodomain Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Motor Neurons/physiology , Nerve Tissue Proteins/metabolism , Receptors, Immunologic/metabolism , Spinal Cord/physiology , Transcription Factors/metabolism , Animals , Gene Expression Regulation, Developmental , Ligands , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Signal Transduction , Spinal Cord/embryology , Spinal Cord/metabolism , Roundabout Proteins
11.
Development ; 138(3): 507-18, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21205795

ABSTRACT

The establishment of cell type-specific dendritic arborization patterns is a key phase in the assembly of neuronal circuitry that facilitates the integration and processing of synaptic and sensory input. Although studies in Drosophila and vertebrate systems have identified a variety of factors that regulate dendrite branch formation, the molecular mechanisms that control this process remain poorly defined. Here, we introduce the use of the Caenorhabditis elegans PVD neurons, a pair of putative nociceptors that elaborate complex dendritic arbors, as a tractable model for conducting high-throughput RNAi screens aimed at identifying key regulators of dendritic branch formation. By carrying out two separate RNAi screens, a small-scale candidate-based screen and a large-scale screen of the ~3000 genes on chromosome IV, we retrieved 11 genes that either promote or suppress the formation of PVD-associated dendrites. We present a detailed functional characterization of one of the genes, bicd-1, which encodes a microtubule-associated protein previously shown to modulate the transport of mRNAs and organelles in a variety of organisms. Specifically, we describe a novel role for bicd-1 in regulating dendrite branch formation and show that bicd-1 is likely to be expressed, and primarily required, in PVD neurons to control dendritic branching. We also present evidence that bicd-1 operates in a conserved pathway with dhc-1 and unc-116, components of the dynein minus-end-directed and kinesin-1 plus-end-directed microtubule-based motor complexes, respectively, and interacts genetically with the repulsive guidance receptor unc-5.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Dendrites/metabolism , Drosophila Proteins/metabolism , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cytoplasmic Dyneins/genetics , Cytoplasmic Dyneins/metabolism , Drosophila Proteins/genetics , Kinesins/genetics , Kinesins/metabolism , Protein Binding , RNA Interference
12.
Int J Mol Sci ; 12(12): 8539-61, 2011.
Article in English | MEDLINE | ID: mdl-22272090

ABSTRACT

Living organisms heavily rely on the function of motor circuits for their survival and for adapting to ever-changing environments. Unique among central nervous system (CNS) neurons, motor neurons (MNs) project their axons out of the CNS. Once in the periphery, motor axons navigate along highly stereotyped trajectories, often at considerable distances from their cell bodies, to innervate appropriate muscle targets. A key decision made by pathfinding motor axons is whether to exit the CNS through dorsal or ventral motor exit points (MEPs). In contrast to the major advances made in understanding the mechanisms that regulate the specification of MN subtypes and the innervation of limb muscles, remarkably little is known about how MN axons project out of the CNS. Nevertheless, a limited number of studies, mainly in Drosophila, have identified transcription factors, and in some cases candidate downstream effector molecules, that are required for motor axons to exit the spinal cord. Notably, specialized neural crest cell derivatives, referred to as Boundary Cap (BC) cells, pre-figure and demarcate MEPs in vertebrates. Surprisingly, however, BC cells are not required for MN axon exit, but rather restrict MN cell bodies from ectopically migrating along their axons out of the CNS. Here, we describe the small set of studies that have addressed motor axon exit in Drosophila and vertebrates, and discuss our fragmentary knowledge of the mechanisms, which guide motor axons out of the CNS.


Subject(s)
Axons/metabolism , Motor Neurons/metabolism , Neurogenesis , Spinal Cord/embryology , Animals , Axons/physiology , Gene Expression Regulation, Developmental , Motor Neurons/cytology , Motor Neurons/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Spinal Cord/cytology , Spinal Cord/metabolism
13.
Cell Adh Migr ; 3(3): 300-4, 2009.
Article in English | MEDLINE | ID: mdl-19556886

ABSTRACT

In the developing nervous system, pathfinding axons navigate through a series of intermediate targets in order to form synaptic connections. Vertebrate spinal commissural axons extend toward and across the floor plate (FP), a key intermediate target located at the ventral midline (VM). Subsequently, post-crossing commissural axons grow either alongside or significant distances away from the floor plate (FP), but never re-cross the VM. Consistent with this behavior, post-crossing commissural axons lose responsiveness to the FP-associated chemoattractants, Netrin-1 and SHH, and gain responsiveness to Slits, which are potent midline repellents, in vitro. In addition, the results of several in vivo studies suggest that the upregulation of Slit-binding repulsive Robo receptors, Robo1/2, alters the responsiveness of decussated commissural axons to midline guidance cues. Nevertheless, in vertebrates, it is unclear whether Robo1/2 are the sole or major repellent receptors responsible for driving these commissural axons away from the VM and preventing their re-entry into the FP. We recently re-visited these issues in the chick spinal cord by assessing the consequences of manipulating Robo expression on commissural axons in ovo. Our findings suggest that, at least in chick embryos, the upregulation of repulsive Robos on post-crossing axons alters the responsiveness of these axons to midline repellents and facilitates their expulsion from, but is not likely to have a significant role in preventing their re-entry into the VM.


Subject(s)
Axons/metabolism , Nerve Tissue Proteins/metabolism , Spinal Cord/metabolism , Animals , Chick Embryo , Mice , Models, Biological , Spinal Cord/embryology
14.
J Neurosci ; 28(35): 8698-708, 2008 Aug 27.
Article in English | MEDLINE | ID: mdl-18753371

ABSTRACT

In vertebrate embryos, most spinal commissural axons cross the ventral midline (VM) and project either alongside or significant distances away from the floor plate (FP). The upregulation of repulsive Robo1/2 receptors on postcrossing commissural axons, in mammals, presumably allows these axons to respond to the midline-associated repellents, Slit1-3, facilitating their expulsion from, and prohibiting their reentry into, the FP. Compelling data suggest that Robo3 represses Robo1/2 function on precrossing axons and that Robo1/2 inhibit attractive guidance receptors on postcrossing axons, thereby ensuring that decussated axons are selectively responsive to midline Slits. However, whether Robo1/2 expel decussated commissural axons from the VM and/or prevent their reentry into the FP has not been explicitly established in vivo. Furthermore, some commissural axons do not require Robo1/2 to elaborate appropriate contralateral projections in the mouse spinal cord. Here, we use unilateral in ovo electroporation together with Atoh1 and Neurog1 enhancer elements to visualize, and assess the consequences of manipulating Robo expression on, dl1 and dl2 chick commissural axons. In response to misexpressing a cytoplasmic truncation of Robo1 and/or Robo2, which should block all Robo-ligand interactions, postcrossing commissural axons extend alongside, but do not project away from or reenter the FP. In contrast, misexpression of full-length Robo2 prevents many commissural axons from crossing the VM. Together, these findings support key and selective in vivo roles for Robo receptors in presumably altering the responsiveness of decussated commissural axons and facilitating their expulsion from the VM within the chick spinal cord.


Subject(s)
Axons/physiology , Gene Expression Regulation, Developmental/genetics , Nerve Tissue Proteins/metabolism , Receptors, Immunologic/metabolism , Spinal Cord/cytology , Spinal Cord/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chick Embryo , Electroporation/methods , Functional Laterality , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/genetics , Receptors, Immunologic/genetics , Roundabout Proteins
15.
Mol Cell Neurosci ; 36(4): 462-71, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17884558

ABSTRACT

In vertebrates, spinal commissural axons project along a transverse path toward and across the floor plate (FP). Post-crossing commissural axons alter their responsiveness to FP-associated guidance cues and turn to project longitudinally in a fasciculated manner prior to extending away from the midline. The upregulation of the neural cell adhesion molecule L1 on crossed commissural axon segments has been proposed to facilitate pathfinding on the contralateral side of the FP. To explore this possibility in vivo, we used Math1 regulatory sequences to target L1 to commissural axons before they cross the ventral midline. L1 mis-expression did not alter the distribution of commissural axon-associated markers or the ventral extension of commissural axons toward the midline. However, commissural axons often stalled or inappropriately projected into the longitudinal plane at the ipsilateral FP margin. These observations suggest that L1-mediated pathfinding decisions are normally delayed until axons have crossed the ventral midline (VM).


Subject(s)
Cell Differentiation/physiology , Growth Cones/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neural Pathways/embryology , Neural Pathways/metabolism , Spinal Cord/embryology , Spinal Cord/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cues , Functional Laterality/physiology , Gene Expression Regulation, Developmental/genetics , Gene Targeting/methods , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Growth Cones/ultrastructure , Mice , Mice, Transgenic , Neural Cell Adhesion Molecule L1/genetics , Neural Pathways/cytology , Spinal Cord/cytology , Up-Regulation/physiology , tau Proteins/genetics , tau Proteins/metabolism
16.
J Neurosci ; 26(35): 8909-14, 2006 Aug 30.
Article in English | MEDLINE | ID: mdl-16943546

ABSTRACT

EphB receptors and their ephrin-B ligands are required for midline guidance decisions at several rostrocaudal levels of the developing CNS. In the embryonic vertebrate spinal cord, ephrin-B3 is localized to the floor plate (FP) at the ventral midline (VM), ephrin-B1 and ephrin-B2 are expressed in the dorsal spinal cord, and decussated EphB receptor-bearing commissural axons navigate between these ventral and dorsal ephrin-B domains. Despite these compelling expression patterns, the in vivo role(s) for EphB and ephrin-B proteins in regulating the guidance of spinal commissural axons has not been established. Here, we use DiI (1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate) labeling to assess the pathfinding of commissural axons in the spinal cords of ephrin-B and EphB mutant mouse embryos. In mice lacking ephrin-B3 or multiple EphB receptors, a significant number of axons followed aberrant trajectories in the immediate vicinity of the VM. Furthermore, forked transverse commissural (FTC) axons, a unique class of commissural axons that continues to project in the transverse plane on the contralateral side of the FP, were present at a markedly higher frequency in ephrin-B3 and EphB mutants, compared with wild-type embryos. Neither the midline guidance errors nor excessive numbers of FTC axons were observed in the spinal cords of ephrin-B3(lacz) mice that express a truncated form of ephrin-B3, which is capable of forward but not reverse signaling. In contrast to the midline guidance defects observed in EphB and ephrin-B3 mutant embryos, wild-type-like contralateral projections were observed in mice lacking ephrin-B1 and/or ephrin-B2.


Subject(s)
Axons/physiology , Ephrin-B3/physiology , Receptors, Eph Family/physiology , Spinal Cord/embryology , Animals , Carbocyanines , Embryo, Mammalian/physiology , Ephrin-B3/genetics , Fluorescent Dyes , Mice , Mice, Knockout , Neural Pathways/embryology , Receptors, Eph Family/genetics , Synaptic Transmission
17.
Dev Biol ; 296(2): 499-513, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16854408

ABSTRACT

In vertebrate embryos, commissural axons extend toward and across the floor plate (FP), an intermediate target at the ventral midline (VM) of the spinal cord. After decussating, many commissural axons turn into the longitudinal plane and elaborate diverse projections. FP contact is thought to alter the responsiveness of these axons so that they can exit the FP and adopt new trajectories. However, a requirement for the FP in shaping contralateral commissural projections has not been established in higher vertebrates. Here we further analyze to what extent FP contact is necessary for the elaboration of decussated commissural projections both in cultured, FP-excised spinal cord preparations and in gli2-deficient mice, which lack a FP. In FP-lacking spinal cords, we observe a large number of appropriately projecting contralateral commissural projections in vivo and in vitro. Surprisingly, even though gli2 mutants lack a FP, slit1-3 mRNA and their receptors (Robo1/2) are expressed in a wild-type-like manner. In addition, blocking Robo-Slit interactions in FP-lacking spinal cord explants prevents commissural axons from leaving the VM and turning longitudinally. Thus, compared to FP contact, Slit-Robo interactions are more critical for driving commissural axons out of the VM and facilitating the elaboration of a subset of contralateral commissural projections.


Subject(s)
Spinal Cord/embryology , Animals , Axons/metabolism , Axons/physiology , Embryo Culture Techniques , Female , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/physiology , Mice , Mice, Knockout , Neural Pathways/physiology , Spinal Cord/cytology , Spinal Cord/metabolism , Zinc Finger Protein GLI1
18.
Nat Neurosci ; 9(8): 996-8, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16829956

ABSTRACT

In addition to their role as chemorepellent netrin-1 receptors, UNC5 proteins may mediate cell death because they induce apoptosis in cultured cells. To test this in vivo, we generated Unc5a (formerly Unc5h1) knockout mice and found that this deletion decreased apoptosis and increased the number of neurons in the spinal cord. In contrast, loss of netrin-1 (Ntn1) did not affect the amount of apoptosis, suggesting that NTN1 is not required for neuronal apoptosis in vivo.


Subject(s)
Apoptosis/physiology , Nerve Growth Factors/metabolism , Neurons/metabolism , Receptors, Cell Surface/metabolism , Spinal Cord/cytology , Spinal Cord/embryology , Tumor Suppressor Proteins/metabolism , Animals , Mice , Mice, Knockout , Nerve Growth Factors/genetics , Netrin Receptors , Netrin-1 , Neurons/pathology , Receptors, Cell Surface/genetics , Spinal Cord/abnormalities , Spinal Cord/metabolism , Tumor Suppressor Proteins/genetics
19.
J Comp Neurol ; 497(5): 734-50, 2006 Aug 10.
Article in English | MEDLINE | ID: mdl-16786562

ABSTRACT

Contact-dependent interactions between EphB receptors and ephrin-B ligands mediate a variety of cell-cell communication events in the developing and mature central nervous system (CNS). These predominantly repulsive interactions occur at the interface between what are considered to be mutually exclusive EphB and ephrin-B expression domains. We previously used receptor and ligand affinity probes to show that ephrin-B ligands are expressed in the floor plate and within a dorsal region of the embryonic mouse spinal cord, while EphB receptors are present on decussated segments of commissural axons that navigate between these ephrin-B domains. Here we present the generation and characterization of two new monoclonal antibodies, mAb EfB1-3, which recognizes EphB1, EphB2, and EphB3, and mAb efrnB1, which is specific for ephrin-B1. We use these reagents and polyclonal antibodies specific for EphB1, EphB2, EphB3, or ephrin-B1 to describe the spatiotemporal expression patterns of EphB receptors and ephrin-B1 in the vertebrate spinal cord. Consistent with affinity probe binding, we show that EphB1, EphB2, and EphB3 are each preferentially expressed on decussated segments of commissural axons in vivo and in vitro, and that ephrin-B1 is expressed in a dorsal domain of the spinal cord that includes the roof plate. In contrast to affinity probe binding profiles, we show here that EphB1, EphB2, and EphB3 are present on the ventral commissure, and that EphB1 and EphB3 are expressed on axons that compose the dorsal funiculus. In addition, we unexpectedly find that mesenchymal cells, which surround the spinal cord and dorsal root ganglion, express ephrin-B1.


Subject(s)
Axons/metabolism , Ephrin-B1/metabolism , Receptors, Eph Family/metabolism , Spinal Cord/metabolism , Animals , Chick Embryo , Cricetinae , Ephrin-B3/metabolism , Female , Immunohistochemistry , Mice , Mice, Inbred BALB C , Rats , Rats, Sprague-Dawley , Receptor, EphB2/metabolism , Spinal Cord/cytology , Spinal Cord/embryology , Tissue Distribution , Zebrafish
20.
J Neurosci ; 25(44): 10119-30, 2005 Nov 02.
Article in English | MEDLINE | ID: mdl-16267219

ABSTRACT

Within the developing vertebrate spinal cord, motor neuron subtypes are distinguished by the settling positions of their cell bodies, patterns of gene expression, and the paths their axons follow to exit the CNS. The inclusive set of cues required to guide a given motor axon subtype from cell body to target has yet to be identified, in any species. This is attributable, in part, to the unavailability of markers that demarcate the complete trajectory followed by a specific class of spinal motor axons. Most spinal motor neurons extend axons out of the CNS through ventral exit points. In contrast, spinal accessory motor neurons (SACMNs) project dorsally directed axons through lateral exit points (LEPs), and these axons assemble into the spinal accessory nerve (SAN). Here we show that an antibody against BEN/ALCAM/SC1/DM-GRASP/MuSC selectively labels mouse SACMNs and can be used to trace the pathfinding of SACMN axons. We use this marker, together with a battery of transcription factor-deficient or guidance cue/receptor-deficient mice to identify molecules required for distinct stages of SACMN development. Specifically, we find that Gli2 is required for the initial extension of axons from SACMN cell bodies, and that netrin-1 and its receptor Dcc are required for the proper dorsal migration of these cells and the dorsally directed extension of SACMN axons toward the LEPs. Furthermore, in the absence of the transcription factor Nkx2.9, SACMN axons fail to exit the CNS. Together, these findings suggest molecular mechanisms that are likely to regulate key steps in SACMN development.


Subject(s)
Accessory Nerve/embryology , Accessory Nerve/metabolism , Axons/metabolism , Motor Neurons/metabolism , Accessory Nerve/cytology , Animals , Cell Differentiation/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Motor Neurons/cytology , Muscle Proteins/biosynthesis , Nuclear Proteins/biosynthesis , Pregnancy , Spinal Cord/embryology , Spinal Cord/metabolism , Trans-Activators/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...