Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.775
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 184(8): 2103-2120.e31, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33740419

ABSTRACT

During cell migration or differentiation, cell surface receptors are simultaneously exposed to different ligands. However, it is often unclear how these extracellular signals are integrated. Neogenin (NEO1) acts as an attractive guidance receptor when the Netrin-1 (NET1) ligand binds, but it mediates repulsion via repulsive guidance molecule (RGM) ligands. Here, we show that signal integration occurs through the formation of a ternary NEO1-NET1-RGM complex, which triggers reciprocal silencing of downstream signaling. Our NEO1-NET1-RGM structures reveal a "trimer-of-trimers" super-assembly, which exists in the cell membrane. Super-assembly formation results in inhibition of RGMA-NEO1-mediated growth cone collapse and RGMA- or NET1-NEO1-mediated neuron migration, by preventing formation of signaling-compatible RGM-NEO1 complexes and NET1-induced NEO1 ectodomain clustering. These results illustrate how simultaneous binding of ligands with opposing functions, to a single receptor, does not lead to competition for binding, but to formation of a super-complex that diminishes their functional outputs.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , GPI-Linked Proteins/metabolism , Nerve Tissue Proteins/metabolism , Oncogene Proteins/metabolism , Animals , Cell Adhesion Molecules, Neuronal/chemistry , Cell Movement , DCC Receptor/deficiency , DCC Receptor/genetics , GPI-Linked Proteins/chemistry , Growth Cones/physiology , Humans , Lateral Ventricles/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/chemistry , Neurons/cytology , Neurons/metabolism , Oncogene Proteins/chemistry , Oncogene Proteins/genetics , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction
2.
Annu Rev Cell Dev Biol ; 36: 61-83, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32603614

ABSTRACT

The brain is our most complex organ. During development, neurons extend axons, which may grow over long distances along well-defined pathways to connect to distant targets. Our current understanding of axon pathfinding is largely based on chemical signaling by attractive and repulsive guidance cues. These cues instruct motile growth cones, the leading tips of growing axons, where to turn and where to stop. However, it is not chemical signals that cause motion-motion is driven by forces. Yet our current understanding of the mechanical regulation of axon growth is very limited. In this review, I discuss the origin of the cellular forces controlling axon growth and pathfinding, and how mechanical signals encountered by growing axons may be integrated with chemical signals. This mechanochemical cross talk is an important but often overlooked aspect of cell motility that has major implications for many physiological and pathological processes involving neuronal growth.


Subject(s)
Axons/physiology , Chemistry , Animals , Axon Guidance/physiology , Biomechanical Phenomena , Growth Cones/physiology , Humans , Models, Biological
3.
Cell ; 162(1): 120-33, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26119341

ABSTRACT

Complicated neuronal circuits can be genetically encoded, but the underlying developmental algorithms remain largely unknown. Here, we describe a developmental algorithm for the specification of synaptic partner cells through axonal sorting in the Drosophila visual map. Our approach combines intravital imaging of growth cone dynamics in developing brains of intact pupae and data-driven computational modeling. These analyses suggest that three simple rules are sufficient to generate the seemingly complex neural superposition wiring of the fly visual map without an elaborate molecular matchmaking code. Our computational model explains robust and precise wiring in a crowded brain region despite extensive growth cone overlaps and provides a framework for matching molecular mechanisms with the rules they execute. Finally, ordered geometric axon terminal arrangements that are not required for neural superposition are a side product of the developmental algorithm, thus elucidating neural circuit connectivity that remained unexplained based on adult structure and function alone.


Subject(s)
Axons , Compound Eye, Arthropod/innervation , Computer Simulation , Drosophila/growth & development , Photoreceptor Cells, Invertebrate/physiology , Algorithms , Animals , Brain/cytology , Brain/physiology , Drosophila/cytology , Drosophila/physiology , Growth Cones
4.
Cell ; 162(5): 1140-54, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26317474

ABSTRACT

Axonal branching contributes substantially to neuronal circuit complexity. Studies in Drosophila have shown that loss of Dscam1 receptor diversity can fully block axon branching in mechanosensory neurons. Here we report that cell-autonomous loss of the receptor tyrosine phosphatase 69D (RPTP69D) and loss of midline-localized Slit inhibit formation of specific axon collaterals through modulation of Dscam1 activity. Genetic and biochemical data support a model in which direct binding of Slit to Dscam1 enhances the interaction of Dscam1 with RPTP69D, stimulating Dscam1 dephosphorylation. Single-growth-cone imaging reveals that Slit/RPTP69D are not required for general branch initiation but instead promote the extension of specific axon collaterals. Hence, although regulation of intrinsic Dscam1-Dscam1 isoform interactions is essential for formation of all mechanosensory-axon branches, the local ligand-induced alterations of Dscam1 phosphorylation in distinct growth-cone compartments enable the spatial specificity of axon collateral formation.


Subject(s)
Axons/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecules/metabolism , Receptor-Like Protein Tyrosine Phosphatases/metabolism , Animals , Cell Adhesion Molecules , Drosophila melanogaster/cytology , Growth Cones/metabolism
5.
Cell ; 158(2): 368-382, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25036633

ABSTRACT

Adenomatous polyposis coli (APC) is a microtubule plus-end scaffolding protein important in biology and disease. APC is implicated in RNA localization, although the mechanisms and functional significance remain unclear. We show APC is an RNA-binding protein and identify an RNA interactome by HITS-CLIP. Targets were highly enriched for APC-related functions, including microtubule organization, cell motility, cancer, and neurologic disease. Among the targets is ß2B-tubulin, known to be required in human neuron and axon migration. We show ß2B-tubulin is synthesized in axons and localizes preferentially to dynamic microtubules in the growth cone periphery. APC binds the ß2B-tubulin 3' UTR; experiments interfering with this interaction reduced ß2B-tubulin mRNA axonal localization and expression, depleted dynamic microtubules and the growth cone periphery, and impaired neuron migration. These results identify APC as a platform binding functionally related protein and RNA networks, and suggest a self-organizing model for the microtubule to localize synthesis of its own subunits.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Microtubules/metabolism , Neurogenesis , RNA-Binding Proteins/metabolism , Animals , Axons/metabolism , Base Sequence , Brain/cytology , Brain/metabolism , Cell Line , Cell Movement , Ganglia, Spinal/cytology , Genome-Wide Association Study , Growth Cones/metabolism , Mice , Molecular Sequence Data , Neurons/metabolism , Protein Interaction Maps , RNA, Messenger/metabolism , Rats , Sequence Alignment , Tubulin/metabolism
6.
Cell ; 154(2): 351-64, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23870124

ABSTRACT

Neuronal growth cones select synaptic partners through interactions with multiple cell surfaces in their environment. Many of these interactions are adhesive, yet it is unclear how growth cones integrate adhesive cues to direct their movements. Here, we examine the mechanisms that enable photoreceptors in the Drosophila visual system to choose synaptic partners. We demonstrate that the classical cadherin, N-cadherin, and an atypical cadherin, Flamingo, act redundantly to instruct the targeting choices made by every photoreceptor axon. These molecules gradually bias the spatial distribution of growth cone filopodia, polarizing each growth cone toward its future synaptic target before direct contact with the target occurs. We demonstrate that these molecules are localized to distinct subcellular domains and create a network of adhesive interactions distributed across many growth cones. Because this network comprises multiple redundant interactions, a complex wiring diagram can be constructed with extraordinary fidelity, suggesting a general principle.


Subject(s)
Cadherins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Growth Cones , Photoreceptor Cells, Invertebrate/metabolism , Synapses , Animals , Axons/metabolism , Growth Cones/metabolism , Retina/metabolism
7.
Cell ; 153(6): 1185-7, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23746834

ABSTRACT

The navigation of axons to their final destination can involve a sequence of steps that require different sets of guidance receptors. In this issue, Colak et al. show that regulated intra-axonal protein synthesis coupled to nonsense-mediated mRNA decay (NMD) controls a switch in Robo3.2 expression that is critical for navigation.


Subject(s)
Axons/metabolism , Embryo, Mammalian/metabolism , Growth Cones/metabolism , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Nonsense Mediated mRNA Decay , Spinal Cord/embryology , Animals , Receptors, Cell Surface
8.
Cell ; 153(6): 1252-65, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23746841

ABSTRACT

Growth cones enable axons to navigate toward their targets by responding to extracellular signaling molecules. Growth-cone responses are mediated in part by the local translation of axonal messenger RNAs (mRNAs). However, the mechanisms that regulate local translation are poorly understood. Here we show that Robo3.2, a receptor for the Slit family of guidance cues, is synthesized locally within axons of commissural neurons. Robo3.2 translation is induced by floor-plate-derived signals as axons cross the spinal cord midline. Robo3.2 is also a predicted target of the nonsense-mediated mRNA decay (NMD) pathway. We find that NMD regulates Robo3.2 synthesis by inducing the degradation of Robo3.2 transcripts in axons that encounter the floor plate. Commissural neurons deficient in NMD proteins exhibit aberrant axonal trajectories after crossing the midline, consistent with misregulation of Robo3.2 expression. These data show that local translation is regulated by mRNA stability and that NMD acts locally to influence axonal pathfinding.


Subject(s)
Axons/metabolism , Embryo, Mammalian/metabolism , Growth Cones/metabolism , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Nonsense Mediated mRNA Decay , Spinal Cord/embryology , Animals , Mice , Neurons/metabolism , Protein Biosynthesis , RNA Isoforms/metabolism , RNA Stability , Receptors, Cell Surface , Spinal Cord/metabolism
9.
Annu Rev Cell Dev Biol ; 30: 417-37, 2014.
Article in English | MEDLINE | ID: mdl-25150010

ABSTRACT

Precise connectivity in neuronal circuits is a prerequisite for proper brain function. The dauntingly complex environment encountered by axons and dendrites, even after navigation to their target area, prompts the question of how specificity of synaptic connections arises during development. We review developmental strategies and molecular mechanisms that are used by neurons to ensure their precise matching of pre- and postsynaptic elements. The emerging theme is that each circuit uses a combination of simple mechanisms to achieve its refined, often complex connectivity pattern. At increasing levels of resolution, from lamina choice to subcellular targeting, similar signaling concepts are reemployed to narrow the choice of potential matches. Temporal control over synapse development and synapse elimination further ensures the specificity of connections in the nervous system.


Subject(s)
Synapses/physiology , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/physiology , Cell Adhesion , Cell Adhesion Molecules, Neuronal/metabolism , Drosophila melanogaster/physiology , Eye Proteins/metabolism , Growth Cones/physiology , Humans , Membrane Proteins/metabolism , Neurons/physiology , Neurons/ultrastructure , Photoreceptor Cells, Invertebrate/physiology , Photoreceptor Cells, Invertebrate/ultrastructure , Retina/cytology , Synaptic Transmission , Time Factors
10.
Development ; 151(17)2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39157903

ABSTRACT

Ciliopathies are characterized by the absence or dysfunction of primary cilia. Despite the fact that cognitive impairments are a common feature of ciliopathies, how cilia dysfunction affects neuronal development has not been characterized in detail. Here, we show that primary cilium-mediated signaling is required cell-autonomously by neurons during neural circuit formation. In particular, a functional primary cilium is crucial during axonal pathfinding for the switch in responsiveness of axons at a choice point or intermediate target. Using different animal models and in vivo, ex vivo and in vitro experiments, we provide evidence for a crucial role of primary cilium-mediated signaling in long-range axon guidance. The primary cilium on the cell body of commissural neurons transduces long-range guidance signals sensed by growth cones navigating an intermediate target. In extension of our finding that Shh is required for the rostral turn of post-crossing commissural axons, we suggest a model implicating the primary cilium in Shh signaling upstream of a transcriptional change of axon guidance receptors, which in turn mediate the repulsive response to floorplate-derived Shh shown by post-crossing commissural axons.


Subject(s)
Axon Guidance , Axons , Cilia , Hedgehog Proteins , Signal Transduction , Cilia/metabolism , Animals , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Mice , Axons/metabolism , Growth Cones/metabolism , Neurons/metabolism
11.
Cell ; 149(7): 1549-64, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22726441

ABSTRACT

Secretory fibroblast growth factors (FGFs) and their receptors are known for their regulatory function in the early stages of neural development. FGF13, a nonsecretory protein of the FGF family, is expressed in cerebral cortical neurons during development and is a candidate gene for syndromal and nonspecific forms of X-chromosome-linked mental retardation (XLMR). However, its function during development remains unclear. We show that FGF13 acts intracellularly as a microtubule-stabilizing protein required for axon and leading process development and neuronal migration in the cerebral cortex. FGF13 is enriched in axonal growth cones and interacts directly with microtubules. Furthermore, FGF13 polymerizes tubulins and stabilizes microtubules. The loss of FGF13 impairs neuronal polarization and increases the branching of axons and leading processes. Genetic deletion of FGF13 in mice results in neuronal migration defects in both the neocortex and the hippocampus. FGF13-deficient mice also exhibit weakened learning and memory, which is correlated to XLMR patients' intellectual disability.


Subject(s)
Fibroblast Growth Factors/metabolism , Neurons/cytology , Neurons/metabolism , Amino Acid Sequence , Animals , Axons/metabolism , Cell Movement , Cell Polarity , Cerebral Cortex/metabolism , Disease Models, Animal , Female , Fibroblast Growth Factors/chemistry , Fibroblast Growth Factors/genetics , Growth Cones/metabolism , Hippocampus/cytology , Humans , Male , Mental Retardation, X-Linked/metabolism , Mice , Mice, Knockout , Microtubules/metabolism , Molecular Sequence Data , Polymerization , Tubulin/metabolism
12.
J Cell Sci ; 137(1)2024 01 01.
Article in English | MEDLINE | ID: mdl-38197773

ABSTRACT

Direct binding of netrin receptors with dynamic microtubules (MTs) in the neuronal growth cone plays an important role in netrin-mediated axon guidance. However, how netrin-1 (NTN1) regulates MT dynamics in axon turning remains a major unanswered question. Here, we show that the coupling of netrin-1 receptor DCC with tau (MAPT)-regulated MTs is involved in netrin-1-promoted axon attraction. Tau directly interacts with DCC and partially overlaps with DCC in the growth cone of primary neurons. Netrin-1 induces this interaction and the colocalization of DCC and tau in the growth cone. The netrin-1-induced interaction of tau with DCC relies on MT dynamics and TUBB3, a highly dynamic ß-tubulin isotype in developing neurons. Netrin-1 increased cosedimentation of DCC with tau and TUBB3 in MTs, and knockdown of either tau or TUBB3 mutually blocked this effect. Downregulation of endogenous tau levels by tau shRNAs inhibited netrin-1-induced axon outgrowth, branching and commissural axon attraction in vitro, and led to defects in spinal commissural axon projection in vivo. These findings suggest that tau is a key MT-associated protein coupling DCC with MT dynamics in netrin-1-promoted axon attraction.


Subject(s)
Axons , Growth Cones , Netrin-1 , Neurons , Microtubules
13.
J Cell Sci ; 137(14)2024 07 15.
Article in English | MEDLINE | ID: mdl-38910449

ABSTRACT

RhoA plays a crucial role in neuronal polarization, where its action restraining axon outgrowth has been thoroughly studied. We now report that RhoA has not only an inhibitory but also a stimulatory effect on axon development depending on when and where exerts its action and the downstream effectors involved. In cultured hippocampal neurons, FRET imaging revealed that RhoA activity selectively localized in growth cones of undifferentiated neurites, whereas in developing axons it displayed a biphasic pattern, being low in nascent axons and high in elongating ones. RhoA-Rho kinase (ROCK) signaling prevented axon initiation but had no effect on elongation, whereas formin inhibition reduced axon extension without significantly altering initial outgrowth. In addition, RhoA-mDia signaling promoted axon elongation by stimulating growth cone microtubule stability and assembly, as opposed to RhoA-ROCK signaling, which restrained growth cone microtubule assembly and protrusion.


Subject(s)
Axons , Growth Cones , Microtubules , Signal Transduction , rhoA GTP-Binding Protein , Microtubules/metabolism , Animals , rhoA GTP-Binding Protein/metabolism , Axons/metabolism , Growth Cones/metabolism , rho-Associated Kinases/metabolism , Hippocampus/metabolism , Hippocampus/cytology , Rats , Formins/metabolism , Cells, Cultured , Neurons/metabolism
14.
J Cell Sci ; 137(19)2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39257379

ABSTRACT

Association of tau (encoded by Mapt) with microtubules causes them to be labile, whereas association of MAP6 with microtubules causes them to be stable. As axons differentiate and grow, tau and MAP6 segregate from one another on individual microtubules, resulting in the formation of stable and labile domains. The functional significance of the yin-yang relationship between tau and MAP6 remains speculative, with one idea being that such a relationship assists in balancing morphological stability with plasticity. Here, using primary rodent neuronal cultures, we show that tau depletion has opposite effects compared to MAP6 depletion on the rate of neuronal development, the efficiency of growth cone turning, and the number of neuronal processes and axonal branches. Opposite effects to those seen with tau depletion were also observed on the rate of neuronal migration, in an in vivo assay, when MAP6 was depleted. When tau and MAP6 were depleted together from neuronal cultures, the morphological phenotypes negated one another. Although tau and MAP6 are multifunctional proteins, our results suggest that the observed effects on neuronal development are likely due to their opposite roles in regulating microtubule stability.


Subject(s)
Microtubule-Associated Proteins , Microtubules , Neurons , tau Proteins , tau Proteins/metabolism , Animals , Neurons/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Neurogenesis , Rats , Cells, Cultured , Axons/metabolism , Mice , Cell Movement , Growth Cones/metabolism
15.
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
16.
Development ; 150(7)2023 04 01.
Article in English | MEDLINE | ID: mdl-37014062

ABSTRACT

In the polarity/protrusion model of growth cone repulsion from UNC-6/netrin, UNC-6 first polarizes the growth cone of the VD motor neuron axon via the UNC-5 receptor, and then regulates protrusion asymmetrically across the growth cone based on this polarity. UNC-6 stimulates protrusion dorsally through the UNC-40/DCC receptor, and inhibits protrusion ventrally through UNC-5, resulting in net dorsal growth. Previous studies showed that UNC-5 inhibits growth cone protrusion via the flavin monooxygenases and potential destabilization of F-actin, and via UNC-33/CRMP and restriction of microtubule plus-end entry into the growth cone. We show that UNC-5 inhibits protrusion through a third mechanism involving TOM-1/tomosyn. A short isoform of TOM-1 inhibited protrusion downstream of UNC-5, and a long isoform had a pro-protrusive role. TOM-1/tomosyn inhibits formation of the SNARE complex. We show that UNC-64/syntaxin is required for growth cone protrusion, consistent with a role of TOM-1 in inhibiting vesicle fusion. Our results are consistent with a model whereby UNC-5 utilizes TOM-1 to inhibit vesicle fusion, resulting in inhibited growth cone protrusion, possibly by preventing the growth cone plasma membrane addition required for protrusion.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Caenorhabditis elegans/metabolism , Growth Cones/metabolism , Caenorhabditis elegans Proteins/metabolism , Axons/metabolism , Netrins/metabolism , Carrier Proteins/metabolism , Netrin Receptors/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Nerve Growth Factors/metabolism , Cell Adhesion Molecules/metabolism
17.
J Neurosci ; 44(27)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38692734

ABSTRACT

Aberrant condensation and localization of the RNA-binding protein (RBP) fused in sarcoma (FUS) occur in variants of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Changes in RBP function are commonly associated with changes in axonal cytoskeletal organization and branching in neurodevelopmental disorders. Here, we asked whether branching defects also occur in vivo in a model of FUS-associated disease. We use two reported Xenopus models of ALS/FTD (of either sex), the ALS-associated mutant FUS(P525L) and a mimic of hypomethylated FUS, FUS(16R). Both mutants strongly reduced axonal complexity in vivo. We also observed an axon looping defect for FUS(P525L) in the target area, which presumably arises due to errors in stop cue signaling. To assess whether the loss of axon complexity also had a cue-independent component, we assessed axonal cytoskeletal integrity in vitro. Using a novel combination of fluorescence and atomic force microscopy, we found that mutant FUS reduced actin density in the growth cone, altering its mechanical properties. Therefore, FUS mutants may induce defects during early axonal development.


Subject(s)
Amyotrophic Lateral Sclerosis , Axons , Frontotemporal Dementia , Mutation , RNA-Binding Protein FUS , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Axons/pathology , Axons/metabolism , Animals , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/metabolism , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Frontotemporal Dementia/metabolism , Female , Male , Xenopus laevis , Growth Cones/metabolism , Humans , Disease Models, Animal
18.
Semin Cell Dev Biol ; 140: 3-12, 2023 05 15.
Article in English | MEDLINE | ID: mdl-35817654

ABSTRACT

The axon is a sophisticated macromolecular machine composed of interrelated parts that transmit signals like spur gears transfer motion between parallel shafts. The growth cone is a fine sensor that integrates mechanical and chemical cues and transduces these signals through the generation of a traction force that pushes the tip and pulls the axon shaft forward. The axon shaft, in turn, senses this pulling force and transduces this signal in an orchestrated response, coordinating cytoskeleton remodeling and intercalated mass addition to sustain and support the advancing of the tip. Extensive research suggests that the direct application of active force is per se a powerful inducer of axon growth, potentially bypassing the contribution of the growth cone. This review provides a critical perspective on current knowledge of how the force is a messenger of axon growth and its mode of action for controlling navigation, including aspects that remain unclear. It also focuses on novel approaches and tools designed to mechanically manipulate axons, and discusses their implications in terms of potential novel therapies for re-wiring the nervous system.


Subject(s)
Axons , Growth Cones , Axons/physiology , Growth Cones/physiology , Actins , Neuronal Outgrowth
19.
Semin Cell Dev Biol ; 140: 63-71, 2023 05 15.
Article in English | MEDLINE | ID: mdl-35817655

ABSTRACT

Axon growth enables the rapid wiring of the central nervous system. Understanding this process is a prerequisite to retriggering it under pathological conditions, such as a spinal cord injury, to elicit axon regeneration. The last decades saw progress in understanding the mechanisms underlying axon growth. Most of these studies employed cultured neurons grown on flat surfaces. Only recently studies on axon growth were performed in 3D. In these studies, physiological environments exposed more complex and dynamic aspects of axon development. Here, we describe current views on axon growth and highlight gaps in our knowledge. We discuss how axons interact with the extracellular matrix during development and the role of the growth cone and its cytoskeleton within. Finally, we propose that the time is ripe to study axon growth in a more physiological setting. This will help us uncover the physiologically relevant mechanisms underlying axon growth, and how they can be reactivated to induce axon regeneration.


Subject(s)
Axons , Nerve Regeneration , Axons/physiology , Neurons , Central Nervous System , Neurogenesis/physiology , Growth Cones
20.
Semin Cell Dev Biol ; 140: 54-62, 2023 05 15.
Article in English | MEDLINE | ID: mdl-35927121

ABSTRACT

The concept of spatial confinement is the basis of cell positioning and guidance in in vitro studies. In vivo, it reflects many situations faced during embryonic development. In vitro, spatial confinement of neurons is achieved using different technological approaches: adhesive patterning, topographical structuring, microfluidics and the use of hydrogels. The notion of chemical or physical frontiers is particularly central to the behaviors of growth cones and neuronal processes under confinement. They encompass phenomena of cell spreading, boundary crossing, and path finding on surfaces with different adhesive properties. However, the most universal phenomenon related to confinement, regardless of how it is implemented, is the acceleration of neuronal growth. Overall, a bi-directional causal link emerges between the shape of the growth cone and neuronal elongation dynamics, both in vivo and in vitro. The sensing of adhesion discontinuities by filopodia and the subsequent spatial redistribution and size adaptation of these actin-rich filaments seem critical for the growth rate in conditions in which adhesive contacts and actin-associated clutching forces dominate. On the other hand, the involvement of microtubules, specifically demonstrated in 3D hydrogel environments and leading to ameboid-like locomotion, could be relevant in a wider range of growth situations. This review brings together a literature collected in distinct scientific fields such as development, mechanobiology and bioengineering that highlight the consequences of confinement and raise new questions at different cellular scales. Its ambition is to stimulate new research that could lead to a better understanding of what gives neurons their ability to establish and regulate their exceptional size.


Subject(s)
Actins , Neurons , Actins/metabolism , Neurons/metabolism , Growth Cones/metabolism , Neurites/metabolism , Microtubules/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL