Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
Add more filters











Publication year range
1.
Mil Med ; 189(Supplement_3): 63-66, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160822

ABSTRACT

INTRODUCTION: Peripheral nerve injury (PNI) occurs in approximately 3% of all trauma patients and can be challenging to treat, particularly when injury is severe such as with a long-segmental gap. Although peripheral nerves can regenerate after injury, functional recovery is often insufficient, leading to deficits in the quality of life of patients with PNI. Although nerve autografts are the gold standard of care, there are several disadvantages to their use, namely a lack of autologous nerve material for repair. This has led to the pursuit of alternative treatment methods such as axon guidance channels (AGCs). Second-generation AGCs have been shown to be able to deliver growth-enhancing substrates for nerve repair directly to the injury site. Although our laboratory has had success with second-generation AGCs filled with Schwann cells (SCs), SCs have their own set of issues clinically. Because of this, we have begun to utilize SC-derived exosomes as an alternative, as they have the appropriate protein markers, associate to axons in high concentrations, and are able to improve nerve regeneration. However, it is unknown how SC-derived exosomes may react within second-generation AGCs; thus, the aim of the present study was to assess the ability of SC-derived exosomes to be loaded into a second-generation AGC and how they would distribute within it. MATERIALS AND METHODS: A total of 4 dry second-generation AGCs were loaded with SC-derived exosomes that were derived from green fluorescent protein (GFP)-labeled SCs. They were subsequently frozen and sliced before imaging. RESULTS: Here, we present findings that SC-derived exosomes can be loaded into second-generation AGCs through our established loading method utilizing negative pressure and are able to survive and equally distribute along the length of the AGC. CONCLUSIONS: Although only 4 second-generation AGCs were utilized, these findings indicate a potential use for SC-derived exosomes within second-generation AGCs to treat severe PNI. Future research should focus on exploring this in greater detail and in different contexts to assess the ability of SC-derived exosomes to survive at the site of injury and treat PNI.


Subject(s)
Exosomes , Nerve Regeneration , Peripheral Nerve Injuries , Schwann Cells , Schwann Cells/physiology , Nerve Regeneration/physiology , Animals , Peripheral Nerve Injuries/therapy , Rats , Axon Guidance/physiology , Axons/physiology
2.
Elife ; 132024 Aug 19.
Article in English | MEDLINE | ID: mdl-39159057

ABSTRACT

The dorsal funiculus in the spinal cord relays somatosensory information to the brain. It is made of T-shaped bifurcation of dorsal root ganglion (DRG) sensory axons. Our previous study has shown that Slit signaling is required for proper guidance during bifurcation, but loss of Slit does not affect all DRG axons. Here, we examined the role of the extracellular molecule Netrin-1 (Ntn1). Using wholemount staining with tissue clearing, we showed that mice lacking Ntn1 had axons escaping from the dorsal funiculus at the time of bifurcation. Genetic labeling confirmed that these misprojecting axons come from DRG neurons. Single axon analysis showed that loss of Ntn1 did not affect bifurcation but rather altered turning angles. To distinguish their guidance functions, we examined mice with triple deletion of Ntn1, Slit1, and Slit2 and found a completely disorganized dorsal funiculus. Comparing mice with different genotypes using immunolabeling and single axon tracing revealed additive guidance errors, demonstrating the independent roles of Ntn1 and Slit. Moreover, the same defects were observed in embryos lacking their cognate receptors. These in vivo studies thus demonstrate the presence of multi-factorial guidance mechanisms that ensure proper formation of a common branched axonal structure during spinal cord development.


Subject(s)
Axon Guidance , Axons , Ganglia, Spinal , Nerve Tissue Proteins , Netrin-1 , Spinal Cord , Animals , Netrin-1/metabolism , Netrin-1/genetics , Mice , Spinal Cord/metabolism , Spinal Cord/embryology , Axons/metabolism , Axons/physiology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Axon Guidance/physiology , Ganglia, Spinal/metabolism , Ganglia, Spinal/embryology , Mice, Knockout , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics
3.
J Neurosci ; 44(30)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-38886055

ABSTRACT

During nervous system development, Sonic hedgehog (Shh) guides developing commissural axons toward the floor plate of the spinal cord. To guide axons, Shh binds to its receptor Boc and activates downstream effectors such as Smoothened (Smo) and Src family kinases (SFKs). SFK activation requires Smo activity and is also required for Shh-mediated axon guidance. Here we report that ß-arrestin1 and ß-arrestin2 (ß-arrestins) serve as scaffolding proteins that link Smo and SFKs in Shh-mediated axon guidance. We found that ß-arrestins are expressed in rat commissural neurons. We also found that Smo, ß-arrestins, and SFKs form a tripartite complex, with the complex formation dependent on ß-arrestins. ß-arrestin knockdown blocked the Shh-mediated increase in Src phosphorylation, demonstrating that ß-arrestins are required to activate Src kinase downstream of Shh. ß-arrestin knockdown also led to the loss of Shh-mediated attraction of rat commissural axons in axon turning assays. Expression of two different dominant-negative ß-arrestins, ß-arrestin1 V53D which blocks the internalization of Smo and ß-arrestin1 P91G-P121E which blocks its interaction with SFKs, also led to the loss of Shh-mediated attraction of commissural axons. In vivo, the expression of these dominant-negative ß-arrestins caused defects in commissural axon guidance in the spinal cord of chick embryos of mixed sexes. Thus we show that ß-arrestins are essential scaffolding proteins that connect Smo to SFKs and are required for Shh-mediated axon guidance.


Subject(s)
Axon Guidance , Hedgehog Proteins , beta-Arrestins , Animals , Hedgehog Proteins/metabolism , Rats , Axon Guidance/physiology , beta-Arrestins/metabolism , Arrestins/metabolism , Arrestins/genetics , Female , Axons/physiology , Axons/metabolism , Rats, Sprague-Dawley , Cells, Cultured , Smoothened Receptor/metabolism , Smoothened Receptor/genetics , src-Family Kinases/metabolism , Male , Spinal Cord/metabolism , Spinal Cord/embryology , Spinal Cord/cytology , Chick Embryo , Humans
4.
Ann Anat ; 254: 152247, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38458575

ABSTRACT

Neural guidance proteins participate in motor neuron migration, axonal projection, and muscle fiber innervation during development. One of the guidance proteins that participates in axonal pathfinding is Netrin-1. Despite the well-known role of Netrin-1 in embryogenesis of central nervous tissue, it is still unclear how the expression of this guidance protein contributes to primary innervation of the periphery, as well as reinnervation. This is especially true in the larynx where Netrin-1 is upregulated within the intrinsic laryngeal muscles after nerve injury and where blocking of Netrin-1 alters the pattern of reinnervation of the intrinsic laryngeal muscles. Despite this consistent finding, it is unknown how Netrin-1 expression contributes to guidance of the axons towards the larynx. Improved knowledge of Netrin-1's role in nerve regeneration and reinnervation post-injury in comparison to its role in primary innervation during embryological development, may provide insights in the search for therapeutics to treat nerve injury. This paper reviews the known functions of Netrin-1 during the formation of the central nervous system and during cranial nerve primary innervation. It also describes the role of Netrin-1 in the formation of the larynx and during recurrent laryngeal reinnervation following nerve injury in the adult.


Subject(s)
Larynx , Nerve Regeneration , Netrin-1 , Netrin-1/metabolism , Animals , Humans , Nerve Regeneration/physiology , Larynx/physiology , Nerve Growth Factors/metabolism , Nerve Growth Factors/physiology , Tumor Suppressor Proteins/metabolism , Axon Guidance/physiology
5.
Neuroscience ; 533: 36-52, 2023 11 21.
Article in English | MEDLINE | ID: mdl-37704063

ABSTRACT

Axon guidance molecules (AGM) are critical regulators of neural development and play a vital role in guiding axons to their target regions during spinal cord development. The correct wiring of neural circuits depends on these molecules' precise expression and function. Defects in axonal pathfinding, growth cone navigation, axonal branching, and synapse formation have far-reaching implications for neuronal circuit construction and function after CNS traumas, such as spinal cord injury (SCI), which affect the expression or activity of AGM. Ascending and descending paths in the spinal cord have been found to include many AGM, including Netrins, Slits, Semaphorins (Sema), Ephrins, and their receptors. In contrast to the repulsive signals like Slits and Semaphorins, which restrict axonal growth and guide axons away from unsuitable locations, Netrins are appealing guidance cues that encourage axonal growth and guidance. Defects in motor function and sensory processing can result from changes in the expression or activity of Ephrins or their receptors, which play an essential role in axonal guidance and synaptic plasticity in the spinal cord. Herein, we highlighted the expressions, functions, and mechanisms of AGM in ascending and descending spinal cord tracts, which can help us identify novel therapeutic targets to improve axonal regeneration and functional recovery after SCI.


Subject(s)
Semaphorins , Spinal Cord Injuries , Spinal Cord Regeneration , Humans , Axon Guidance/physiology , Axons/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord/metabolism , Ephrins/metabolism , Netrins/metabolism , Nerve Regeneration/physiology
6.
Development ; 150(19)2023 10 01.
Article in English | MEDLINE | ID: mdl-37747104

ABSTRACT

During neural circuit formation, axons navigate from one intermediate target to the next, until they reach their final target. At intermediate targets, axons switch from being attracted to being repelled by changing the guidance receptors on the growth cone surface. For smooth navigation of the intermediate target and the continuation of their journey, the switch in receptor expression has to be orchestrated in a precisely timed manner. As an alternative to changes in expression, receptor function could be regulated by phosphorylation of receptors or components of signaling pathways. We identified Cables1 as a linker between floor-plate exit of commissural axons, regulated by Slit/Robo signaling, and the rostral turn of post-crossing axons, regulated by Wnt/Frizzled signaling. Cables1 localizes ß-catenin, phosphorylated at tyrosine 489 by Abelson kinase, to the distal axon, which in turn is necessary for the correct navigation of post-crossing commissural axons in the developing chicken spinal cord.


Subject(s)
Axon Guidance , Axons , Axon Guidance/physiology , Axons/metabolism , Growth Cones , Spinal Cord/metabolism , Wnt Signaling Pathway , Animals , Chickens
7.
Neuroscience ; 508: 30-39, 2023 01 01.
Article in English | MEDLINE | ID: mdl-35940454

ABSTRACT

The molecular mechanisms of neural circuit formation have been of interest to Santiago Ramón y Cajal and thousands of neuroscientists sharing his passion for neural circuits ever since. Cajal was a brilliant observer and taught us about the connections and the morphology of neurons in the adult and developing nervous system. Clearly, we will not learn about molecular mechanisms by just looking at brain sections or cells in culture. Technically, we had to come a long way to today's possibilities that allow us to perturb target gene expression and watch the consequences of our manipulations on navigating axons in situ. In this review, we summarize landmark steps towards modern live-imaging approaches used to study the molecular basis of axon guidance.


Subject(s)
Axons , Neurons , Axons/metabolism , Neurons/physiology , Axon Guidance/physiology , Brain
8.
JCI Insight ; 7(16)2022 08 22.
Article in English | MEDLINE | ID: mdl-35993361

ABSTRACT

Metastatic pancreatic cancer (PDAC) has a poor clinical outcome with a 5-year survival rate below 3%. Recent transcriptome profiling of PDAC biopsies has identified 2 clinically distinct subtypes - the "basal-like" (BL) subtype with poor prognosis and therapy resistance compared with the less aggressive and drug-susceptible "classical" (CLA) subtype. However, the mechanistic events and environmental factors that promote the BL subtype identity are not very clear. Using preclinical models, patient-derived xenografts, and FACS-sorted PDAC patient biopsies, we report here that the axon guidance receptor, roundabout guidance receptor 3 (ROBO3), promotes the BL metastatic program via a potentially unique AXL/IL-6/phosphorylated STAT3 (p-STAT3) regulatory axis. RNA-Seq identified a ROBO3-mediated BL-specific gene program, while tyrosine kinase profiling revealed AXL as the key mediator of the p-STAT3 activation. CRISPR/dCas9-based ROBO3 silencing disrupted the AXL/p-STAT3 signaling axis, thereby halting metastasis and enhancing therapy sensitivity. Transcriptome analysis of resected patient tumors revealed that AXLhi neoplastic cells associated with the inflammatory stromal program. Combining AXL inhibitor and chemotherapy substantially restored a CLA phenotypic state and reduced disease aggressiveness. Thus, we conclude that a ROBO3-driven hierarchical network determines the inflammatory and prometastatic programs in a specific PDAC subtype.


Subject(s)
Axon Guidance , Pancreatic Neoplasms , Receptors, Cell Surface , Axon Guidance/genetics , Axon Guidance/physiology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prognosis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Axl Receptor Tyrosine Kinase
9.
Dev Biol ; 490: 50-52, 2022 10.
Article in English | MEDLINE | ID: mdl-35788000

ABSTRACT

The emergence of exquisitely organized axonal projections is one of the greatest wonders of nervous system development. In addition to growing along stereotyped directions, axons join one another as they extend. It is well known that axonal growth cones recognize cell surface guidance cues on axons and either grow along the axons or away from the axons. However, it is less well understood whether and how the growth cones communicate with each other and, if so, what do these interactions mean. Recent studies from our lab provided direct evidence that the growth cones do interact with each other during axon pathfinding. And this interaction is regulated by highly regulated protein-protein interactions among components of the planar cell polarity pathway. The disruption of these interactions lead to guidance defects and disorganization of axons. We propose that this local inter-growth cone PCP-like signaling mechanism reinforces and increases the sensitivity of the growth cone response to shallow Wnt gradients to turn in a precise and organized fashion.


Subject(s)
Axon Guidance , Growth Cones , Axon Guidance/physiology , Axons/metabolism , Cell Polarity , Communication , Growth Cones/metabolism , Spinal Cord/metabolism , Wnt Signaling Pathway
10.
Dev Dyn ; 251(3): 444-458, 2022 03.
Article in English | MEDLINE | ID: mdl-34374463

ABSTRACT

BACKGROUND: Proper guidance of neuronal axons to their targets is required to assemble neural circuits during the development of the nervous system. However, the mechanism by which the guidance of axonal growth cones is regulated by specific intermediaries activated by receptor signaling pathways to mediate cytoskeleton dynamics is unclear. Vav protein members have been proposed to mediate this process, prompting us to investigate their role in the limb selection of the axon trajectory of spinal lateral motor column (LMC) neurons. RESULTS: We found Vav2 and Vav3 expression in LMC neurons when motor axons grew into the limb. Vav2, but not Vav3, loss-of-function perturbed LMC pathfinding, while Vav2 gain-of-function exhibited the opposite effects, demonstrating that Vav2 plays an important role in motor axon growth. Vav2 knockdown also attenuated the redirectional phenotype of LMC axons induced by Dcc, but not EphA4, in vivo and lateral LMC neurite growth preference to Netrin-1 in vitro. This study showed that Vav2 knockdown and ectopic nonphosphorylable Vav2 mutant expression abolished the Src-induced stronger growth preference of lateral LMC neurites to Netrin-1, suggesting that Vav2 is downstream of Src in this context. CONCLUSIONS: Vav2 is essential for Netrin-1-regulated LMC motor axon pathfinding through Src interaction.


Subject(s)
Axon Guidance , Growth Cones , Netrin-1 , Proto-Oncogene Proteins c-vav , Animals , Axon Guidance/physiology , Axons/physiology , Growth Cones/physiology , Motor Neurons/physiology , Netrin-1/physiology , Proto-Oncogene Proteins c-vav/physiology
11.
Mol Cells ; 44(8): 549-556, 2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34385406

ABSTRACT

Decoding the molecular mechanisms underlying axon guidance is key to precise understanding of how complex neural circuits form during neural development. Although substantial progress has been made over the last three decades in identifying numerous axon guidance molecules and their functional roles, little is known about how these guidance molecules collaborate to steer growth cones to their correct targets. Recent studies in Drosophila point to the importance of the combinatorial action of guidance molecules, and further show that selective fasciculation and defasciculation at specific choice points serve as a fundamental strategy for motor axon guidance. Here, I discuss how attractive and repulsive guidance cues cooperate to ensure the recognition of specific choice points that are inextricably linked to selective fasciculation and defasciculation, and correct pathfinding decision-making.


Subject(s)
Axon Guidance/physiology , Drosophila melanogaster/physiology , Motor Neurons/physiology , Animals , Axon Fasciculation , Muscles/innervation , Neuromuscular Junction/physiology
12.
Int J Mol Sci ; 22(11)2021 Jun 06.
Article in English | MEDLINE | ID: mdl-34204060

ABSTRACT

The establishment of neuronal circuits requires neurons to develop and maintain appropriate connections with cellular partners in and out the central nervous system. These phenomena include elaboration of dendritic arborization and formation of synaptic contacts, initially made in excess. Subsequently, refinement occurs, and pruning takes places both at axonal and synaptic level, defining a homeostatic balance maintained throughout the lifespan. All these events require genetic regulations which happens cell-autonomously and are strongly influenced by environmental factors. This review aims to discuss the involvement of guidance cues from the Semaphorin family.


Subject(s)
Axon Guidance/physiology , Cues , Models, Neurological , Neurons/physiology , Semaphorins/metabolism , Animals , Humans , Neuronal Plasticity/physiology
13.
J Neurosci ; 41(31): 6637-6651, 2021 08 04.
Article in English | MEDLINE | ID: mdl-34252036

ABSTRACT

Understanding the bioenergetics of axon extension and maintenance has wide ranging implications for neurodevelopment and disease states. Glycolysis is a pathway consisting of 10 enzymes and separated into preparatory and payoff phases, the latter producing ATP. Using embryonic chicken sensory neurons, we report that glycolytic enzymes are found through the axon and the growth cone. Pharmacological inhibition of glycolysis in the presence of NGF impairs axon extension and growth cone dynamics within minutes without affecting axon maintenance. Experiments using microfluidic chambers show that the effect of inhibiting glycolysis on axon extension is local along distal axons and can be reversed by promoting mitochondrial respiration. Knockdown of GAPDH simplifies growth cone morphology and is rescued by shRNA-resistant GAPDH expression. Rescue of GAPDH using KillerRed fused to GAPDH followed by localized chromophore-assisted light inactivation of KillerRed-GAPDH in distal axons halts growth cone dynamics. Considering filament polymerization requires ATP, inhibition of glycolysis results in a paradoxical increase in axonal actin filament levels. The effect on actin filaments is because of enzymes before GAPDH, the first enzyme in the payoff phase. In the absence of NGF, inhibition of glycolysis along distal axons results in axon degeneration independent of cell death. These data indicate that the glycolytic pathway is operative in distal axons and contributes to the rate of axon extension and growth cone dynamics in the presence of NGF and that, in the absence of NGF, the axonal glycolytic pathway is required for axon maintenance.SIGNIFICANCE STATEMENT Elucidation of the sources of ATP required for axon extension and maintenance has implications for understanding the mechanism of neuronal development and diseases of the nervous system. While recent work has emphasized the importance of mitochondrial oxidative phosphorylation, the role of the glycolytic pathway in axon morphogenesis and maintenance remains minimally understood. The data reveal that the glycolytic pathway is required for normal sensory axon extension in the presence of NGF, while in the absence of NGF the glycolytic pathway is required for axon maintenance. The results have implications for the understanding of the bioenergetics of axon morphogenesis and plasticity and indicate that NGF has protective effects on sensory axon maintenance in hypoglycemic states.


Subject(s)
Axon Guidance/physiology , Glycolysis/physiology , Growth Cones/metabolism , Sensory Receptor Cells/metabolism , Animals , Axons/physiology , Chick Embryo
14.
Neuropharmacology ; 196: 108690, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34197891

ABSTRACT

Transcription-translation coupling leads to the production of proteins that are key for controlling essential neuronal processes that include neuronal development and changes in synaptic strength. Although these events have been a prevailing theme in neuroscience, the regulation of proteins via posttranslational signaling pathways are equally relevant for these neuronal processes. Ubiquitin is one type of posttranslational modification that covalently attaches to its targets/substrates. Ubiquitination of proteins play a key role in multiple signaling pathways, the predominant being removal of its substrates by a large molecular machine called the proteasome. Here, I review 40 years of progress on ubiquitination in the nervous system at glutamatergic synapses focusing on axon pathfinding, synapse formation, presynaptic release, dendritic spine formation, and regulation of postsynaptic glutamate receptors. Finally, I elucidate emerging themes in ubiquitin biology that may challenge our current understanding of ubiquitin signaling in the nervous system.


Subject(s)
Glutamic Acid/metabolism , Post-Synaptic Density/metabolism , Presynaptic Terminals/metabolism , Receptors, Glutamate/metabolism , Synapses/metabolism , Ubiquitin/metabolism , Ubiquitination/physiology , Animals , Axon Guidance/physiology , Dendritic Spines/metabolism , Dendritic Spines/physiology , Humans , Neurons/metabolism , Neurons/physiology , Post-Synaptic Density/physiology , Presynaptic Terminals/physiology , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/physiology , Protein Processing, Post-Translational/physiology , Receptors, Glutamate/physiology , Synapses/physiology
15.
J Am Soc Nephrol ; 32(9): 2331-2351, 2021 09.
Article in English | MEDLINE | ID: mdl-34140396

ABSTRACT

BACKGROUND: Mechanisms underlying the pro gression of diabetic kidney disease to ESKD are not fully understood. METHODS: We performed global microRNA (miRNA) analysis on plasma from two cohorts consisting of 375 individuals with type 1 and type 2 diabetes with late diabetic kidney disease, and targeted proteomics analysis on plasma from four cohorts consisting of 746 individuals with late and early diabetic kidney disease. We examined structural lesions in kidney biopsy specimens from the 105 individuals with early diabetic kidney disease. Human umbilical vein endothelial cells were used to assess the effects of miRNA mimics or inhibitors on regulation of candidate proteins. RESULTS: In the late diabetic kidney disease cohorts, we identified 17 circulating miRNAs, represented by four exemplars (miR-1287-5p, miR-197-5p, miR-339-5p, and miR-328-3p), that were strongly associated with 10-year risk of ESKD. These miRNAs targeted proteins in the axon guidance pathway. Circulating levels of six of these proteins-most notably, EFNA4 and EPHA2-were strongly associated with 10-year risk of ESKD in all cohorts. Furthermore, circulating levels of these proteins correlated with severity of structural lesions in kidney biopsy specimens. In contrast, expression levels of genes encoding these proteins had no apparent effects on the lesions. In in vitro experiments, mimics of miR-1287-5p and miR-197-5p and inhibitors of miR-339-5p and miR-328-3p upregulated concentrations of EPHA2 in either cell lysate, supernatant, or both. CONCLUSIONS: This study reveals novel mechanisms involved in progression to ESKD and points to the importance of systemic factors in the development of diabetic kidney disease. Some circulating miRNAs and axon guidance pathway proteins represent potential targets for new therapies to prevent and treat this condition.


Subject(s)
Axon Guidance/physiology , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 2/blood , Diabetic Nephropathies/etiology , Kidney Failure, Chronic/etiology , MicroRNAs/blood , Adult , Cohort Studies , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/blood , Female , Humans , Kidney Failure, Chronic/blood , Male , Middle Aged
16.
J Neurosci ; 41(31): 6617-6636, 2021 08 04.
Article in English | MEDLINE | ID: mdl-34131031

ABSTRACT

Axons navigate through the embryo to construct a functional nervous system. A missing part of the axon navigation puzzle is how a single axon traverses distinct anatomic choice points through its navigation. The dorsal root ganglia (DRG) neurons experience such choice points. First, they navigate to the dorsal root entry zone (DREZ), then halt navigation in the peripheral nervous system to invade the spinal cord, and then reinitiate navigation inside the CNS. Here, we used time-lapse super-resolution imaging in zebrafish DRG pioneer neurons to investigate how embryonic axons control their cytoskeleton to navigate to and invade at the correct anatomic position. We found that invadopodia components form in the growth cone even during filopodia-based navigation, but only stabilize when the axon is at the spinal cord entry location. Further, we show that intermediate levels of DCC and cAMP, as well as Rac1 activation, subsequently engage an axon invasion brake. Our results indicate that actin-based invadopodia components form in the growth cone and disruption of the invasion brake causes axon entry defects and results in failed behavioral responses, thereby demonstrating the importance of regulating distinct actin populations during navigational challenges.SIGNIFICANCE STATEMENT Correct spatiotemporal navigation of neuronal growth cones is dependent on extracellular navigational cues and growth cone dynamics. Here, we link dcc-mediated signaling to actin-based invadopodia and filopodia dynamics during pathfinding and entry into the spinal cord using an in vivo model of dorsal root ganglia (DRG) sensory axons. We reveal a molecularly-controlled brake on invadopodia stabilization until the sensory neuron growth cone is present at the dorsal root entry zone (DREZ), which is ultimately essential for growth cone entry into the spinal cord and behavioral response.


Subject(s)
Axon Guidance/physiology , DCC Receptor/metabolism , Signal Transduction/physiology , Zebrafish Proteins/metabolism , Animals , Ganglia, Spinal/embryology , Zebrafish
17.
J Neurosci ; 41(31): 6652-6672, 2021 08 04.
Article in English | MEDLINE | ID: mdl-34168008

ABSTRACT

A precise sequence of axon guidance events is required for the development of the ocular motor system. Three cranial nerves grow toward, and connect with, six extraocular muscles in a stereotyped pattern, to control eye movements. The signaling protein alpha2-chimaerin (α2-CHN) plays a pivotal role in the formation of the ocular motor system; mutations in CHN1, encoding α2-CHN, cause the human eye movement disorder Duane Retraction Syndrome (DRS). Our research has demonstrated that the manipulation of α2-chn signaling in the zebrafish embryo leads to ocular motor axon wiring defects, although the signaling cascades regulated by α2-chn remain poorly understood. Here, we demonstrate that several cytoskeletal regulatory proteins-collapsin response mediator protein 2 (CRMP2; encoded by the gene dpysl2), stathmin1, and stathmin 2-bind to α2-CHN. dpysl2, stathmin1, and especially stathmin2 are expressed by ocular motor neurons. We find that the manipulation of dpysl2 and of stathmins in zebrafish larvae leads to defects in both the axon wiring of the ocular motor system and the optokinetic reflex, impairing horizontal eye movements. Knockdowns of these molecules in zebrafish larvae of either sex caused axon guidance phenotypes that included defasciculation and ectopic branching; in some cases, these phenotypes were reminiscent of DRS. chn1 knock-down phenotypes were rescued by the overexpression of CRMP2 and STMN1, suggesting that these proteins act in the same signaling pathway. These findings suggest that CRMP2 and stathmins signal downstream of α2-CHN to orchestrate ocular motor axon guidance and to control eye movements.SIGNIFICANCE STATEMENT The precise control of eye movements is crucial for the life of vertebrate animals, including humans. In humans, this control depends on the arrangement of nerve wiring of the ocular motor system, composed of three nerves and six muscles, a system that is conserved across vertebrate phyla. Mutations in the protein alpha2-chimaerin have previously been shown to cause eye movement disorders (squint) and axon wiring defects in humans. Our recent work has unraveled how alpha2-chimaerin coordinates axon guidance of the ocular motor system in animal models. In this article, we demonstrate key roles for the proteins CRMP2 and stathmin 1/2 in the signaling pathway orchestrated by alpha2-chimaerin, potentially giving insight into the etiology of eye movement disorders in humans.


Subject(s)
Axon Guidance/physiology , Chimerin 1/metabolism , Motor Neurons/metabolism , Nerve Tissue Proteins/metabolism , Oculomotor Muscles/innervation , Stathmin/metabolism , Zebrafish Proteins/metabolism , Animals , Chimerin 1/genetics , Duane Retraction Syndrome/genetics , Eye Movements , Signal Transduction/physiology , Zebrafish
18.
Phys Rev Lett ; 126(11): 118101, 2021 Mar 19.
Article in English | MEDLINE | ID: mdl-33798338

ABSTRACT

During the development of the nervous system, neurons extend bundles of axons that grow and meet other neurons to form the neuronal network. Robust guidance mechanisms are needed for these bundles to migrate and reach their functional target. Directional information depends on external cues such as chemical or mechanical gradients. Unlike chemotaxis that has been extensively studied, the role and mechanism of durotaxis, the directed response to variations in substrate rigidity, remain unclear. We model bundle migration and guidance by rigidity gradients by using the theory of morphoelastic rods. We show that, at a rigidity interface, the motion of axon bundles follows a simple behavior analogous to optic ray theory and obeys Snell's law for refraction and reflection. We use this powerful analogy to demonstrate that axons can be guided by the equivalent of optical lenses and fibers created by regions of different stiffnesses.


Subject(s)
Axon Guidance/physiology , Models, Neurological , Nerve Net/growth & development , Animals , Axons/physiology , Biomechanical Phenomena , Nerve Net/physiology , Neurons/physiology , Xenopus
19.
Cereb Cortex ; 31(9): 4340-4356, 2021 07 29.
Article in English | MEDLINE | ID: mdl-33877363

ABSTRACT

Neocortical heterotopia consist of ectopic neuronal clusters that are frequently found in individuals with cognitive disability and epilepsy. However, their pathogenesis remains poorly understood due in part to a lack of tractable animal models. We have developed an inducible model of focal cortical heterotopia that enables their precise spatiotemporal control and high-resolution optical imaging in live mice. Here, we report that heterotopia are associated with striking patterns of circumferentially projecting axons and increased myelination around neuronal clusters. Despite their aberrant axonal patterns, in vivo calcium imaging revealed that heterotopic neurons remain functionally connected to other brain regions, highlighting their potential to influence global neural networks. These aberrant patterns only form when heterotopia are induced during a critical embryonic temporal window, but not in early postnatal development. Our model provides a new way to investigate heterotopia formation in vivo and reveals features suggesting the existence of developmentally modulated, neuron-derived axon guidance and myelination factors.


Subject(s)
Axon Guidance/physiology , Neocortex/cytology , Neocortex/physiology , Nerve Fibers, Myelinated/physiology , Neurons/physiology , Animals , Electroporation/methods , Female , Male , Mice , Neocortex/chemistry , Nerve Fibers, Myelinated/chemistry , Neurons/chemistry , Pregnancy
20.
J Neurosci ; 41(22): 4795-4808, 2021 06 02.
Article in English | MEDLINE | ID: mdl-33906900

ABSTRACT

Coordination of skilled movements and motor planning relies on the formation of regionally restricted brain circuits that connect cortex with subcortical areas during embryonic development. Layer 5 neurons that are distributed across most cortical areas innervate the pontine nuclei (basilar pons) by protrusion and extension of collateral branches interstitially along their corticospinal extending axons. Pons-derived chemotropic cues are known to attract extending axons, but molecules that regulate collateral extension to create regionally segregated targeting patterns have not been identified. Here, we discovered that EphA7 and EfnA5 are expressed in the cortex and the basilar pons in a region-specific and mutually exclusive manner, and that their repulsive activities are essential for segregating collateral extensions from corticospinal axonal tracts in mice. Specifically, EphA7 and EfnA5 forward and reverse inhibitory signals direct collateral extension such that EphA7-positive frontal and occipital cortical areas extend their axon collaterals into the EfnA5-negative rostral part of the basilar pons, whereas EfnA5-positive parietal cortical areas extend their collaterals into the EphA7-negative caudal part of the basilar pons. Together, our results provide a molecular basis that explains how the corticopontine projection connects multimodal cortical outputs to their subcortical targets.SIGNIFICANCE STATEMENT Our findings put forward a model in which region-to-region connections between cortex and subcortical areas are shaped by mutually exclusive molecules to ensure the fidelity of regionally restricted circuitry. This model is distinct from earlier work showing that neuronal circuits within individual cortical modalities form in a topographical manner controlled by a gradient of axon guidance molecules. The principle that a shared molecular program of mutually repulsive signaling instructs regional organization-both within each brain region and between connected brain regions-may well be applicable to other contexts in which information is sorted by converging and diverging neuronal circuits.


Subject(s)
Axon Guidance/physiology , Ephrin-A5/metabolism , Neocortex/embryology , Neural Pathways/embryology , Pons/embryology , Receptor, EphA7/metabolism , Animals , Female , Male , Mice , Mice, Inbred C57BL , Neocortex/metabolism , Neural Pathways/metabolism , Pons/pathology
SELECTION OF CITATIONS
SEARCH DETAIL