Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Neuron ; 103(6): 1073-1085.e6, 2019 09 25.
Article in English | MEDLINE | ID: mdl-31400829

ABSTRACT

Injured axons fail to regenerate in the adult CNS, which contrasts with their vigorous growth during embryonic development. We explored the potential of re-initiating axon extension after injury by reactivating the molecular mechanisms that drive morphogenetic transformation of neurons during development. Genetic loss- and gain-of-function experiments followed by time-lapse microscopy, in vivo imaging, and whole-mount analysis show that axon regeneration is fueled by elevated actin turnover. Actin depolymerizing factor (ADF)/cofilin controls actin turnover to sustain axon regeneration after spinal cord injury through its actin-severing activity. This pinpoints ADF/cofilin as a key regulator of axon growth competence, irrespective of developmental stage. These findings reveal the central role of actin dynamics regulation in this process and elucidate a core mechanism underlying axon growth after CNS trauma. Thereby, neurons maintain the capacity to stimulate developmental programs during adult life, expanding their potential for plasticity. Thus, actin turnover is a key process for future regenerative interventions.


Subject(s)
Actins/metabolism , Axons/metabolism , Cofilin 1/genetics , Cofilin 2/genetics , Destrin/genetics , Growth Cones/pathology , Nerve Regeneration/genetics , Spinal Cord Injuries/genetics , Animals , Axons/pathology , Cofilin 1/metabolism , Cofilin 2/metabolism , Destrin/metabolism , Growth Cones/metabolism , Intravital Microscopy , Mice , Microscopy, Confocal , Neurons/metabolism , Neurons/pathology , Rats , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Time-Lapse Imaging
2.
PLoS Genet ; 11(3): e1005063, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25803043

ABSTRACT

Glycosphingolipids are key elements of cellular membranes, thereby, controlling a variety of cellular functions. Accumulation of the simple glycosphingolipid glucosylceramide results in life-threatening lipid storage-diseases or in male infertility. How glucosylceramide regulates cellular processes is ill defined. Here, we reveal that glucosylceramide accumulation in GBA2 knockout-mice alters cytoskeletal dynamics due to a more ordered lipid organization in the plasma membrane. In dermal fibroblasts, accumulation of glucosylceramide augments actin polymerization and promotes microtubules persistence, resulting in a higher number of filopodia and lamellipodia and longer microtubules. Similar cytoskeletal defects were observed in male germ and Sertoli cells from GBA2 knockout-mice. In particular, the organization of F-actin structures in the ectoplasmic specialization and microtubules in the sperm manchette is affected. Thus, glucosylceramide regulates cytoskeletal dynamics, providing mechanistic insights into how glucosylceramide controls signaling pathways not only during sperm development, but also in other cell types.


Subject(s)
Actins/metabolism , Cytoskeleton/genetics , Glucosylceramides/genetics , Lipid Metabolism/genetics , beta-Glucosidase/genetics , Actins/chemistry , Animals , Cell Membrane/metabolism , Cell Membrane/pathology , Cytoskeleton/metabolism , Cytoskeleton/pathology , Fibroblasts/metabolism , Glucosylceramides/chemistry , Glucosylceramides/metabolism , Humans , Male , Mice , Mice, Knockout , Microtubules/genetics , Microtubules/metabolism , Microtubules/pathology , Pseudopodia/genetics , Pseudopodia/metabolism , Pseudopodia/pathology , Sertoli Cells/metabolism , Sertoli Cells/pathology , beta-Glucosidase/metabolism
3.
Science ; 348(6232): 347-52, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25765066

ABSTRACT

After central nervous system (CNS) injury, inhibitory factors in the lesion scar and poor axon growth potential prevent axon regeneration. Microtubule stabilization reduces scarring and promotes axon growth. However, the cellular mechanisms of this dual effect remain unclear. Here, delayed systemic administration of a blood-brain barrier-permeable microtubule-stabilizing drug, epothilone B (epoB), decreased scarring after rodent spinal cord injury (SCI) by abrogating polarization and directed migration of scar-forming fibroblasts. Conversely, epothilone B reactivated neuronal polarization by inducing concerted microtubule polymerization into the axon tip, which propelled axon growth through an inhibitory environment. Together, these drug-elicited effects promoted axon regeneration and improved motor function after SCI. With recent clinical approval, epothilones hold promise for clinical use after CNS injury.


Subject(s)
Axons/drug effects , Cicatrix/prevention & control , Epothilones/administration & dosage , Nerve Regeneration/drug effects , Spinal Cord Injuries/drug therapy , Tubulin Modulators/administration & dosage , Animals , Axons/physiology , Cell Movement/drug effects , Cell Polarity/drug effects , Cicatrix/pathology , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , Meninges/drug effects , Meninges/pathology , Motor Activity/drug effects , Neurons/drug effects , Neurons/pathology , Rats , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology
4.
Bioarchitecture ; 3(4): 86-109, 2013.
Article in English | MEDLINE | ID: mdl-24002528

ABSTRACT

Neurons begin their life as simple spheres, but can ultimately assume an elaborate morphology with numerous, highly arborized dendrites, and long axons. This is achieved via an astounding developmental progression which is dependent upon regulated assembly and dynamics of the cellular cytoskeleton. As neurites emerge out of the soma, neurons break their spherical symmetry and begin to acquire the morphological features that define their structure and function. Neurons regulate their cytoskeleton to achieve changes in cell shape, velocity, and direction as they migrate, extend neurites, and polarize. Of particular importance, the organization and dynamics of actin and microtubules directs the migration and morphogenesis of neurons. This review focuses on the regulation of intrinsic properties of the actin and microtubule cytoskeletons and how specific cytoskeletal structures and dynamics are associated with the earliest phase of neuronal morphogenesis­neuritogenesis.


Subject(s)
Actins/metabolism , Cytoskeleton/metabolism , Neurites/metabolism , Neurons/metabolism , Animals , Humans , Morphogenesis , Neurons/cytology
5.
Neuron ; 76(6): 1091-107, 2012 Dec 20.
Article in English | MEDLINE | ID: mdl-23259946

ABSTRACT

Neurites are the characteristic structural element of neurons that will initiate brain connectivity and elaborate information. Early in development, neurons are spherical cells but this symmetry is broken through the initial formation of neurites. This fundamental step is thought to rely on actin and microtubule dynamics. However, it is unclear which aspects of the complex actin behavior control neuritogenesis and which molecular mechanisms are involved. Here, we demonstrate that augmented actin retrograde flow and protrusion dynamics facilitate neurite formation. Our data indicate that a single family of actin regulatory proteins, ADF/Cofilin, provides the required control of actin retrograde flow and dynamics to form neurites. In particular, the F-actin severing activity of ADF/Cofilin organizes space for the protrusion and bundling of microtubules, the backbone of neurites. Our data reveal how ADF/Cofilin organizes the cytoskeleton to drive actin retrograde flow and thus break the spherical shape of neurons.


Subject(s)
Actin Depolymerizing Factors/physiology , Actins/metabolism , Cell Shape/physiology , Cerebral Cortex/embryology , Destrin/physiology , Growth Cones/metabolism , Neurites/metabolism , Animals , Biological Transport , Cell Growth Processes/physiology , Cells, Cultured , Cerebral Cortex/cytology , Hippocampus/cytology , Hippocampus/embryology , In Vitro Techniques , Mice , Mice, Knockout , Microtubules/physiology , Neurogenesis/physiology
6.
Science ; 331(6019): 928-31, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21273450

ABSTRACT

Hypertrophic scarring and poor intrinsic axon growth capacity constitute major obstacles for spinal cord repair. These processes are tightly regulated by microtubule dynamics. Here, moderate microtubule stabilization decreased scar formation after spinal cord injury in rodents through various cellular mechanisms, including dampening of transforming growth factor-ß signaling. It prevented accumulation of chondroitin sulfate proteoglycans and rendered the lesion site permissive for axon regeneration of growth-competent sensory neurons. Microtubule stabilization also promoted growth of central nervous system axons of the Raphe-spinal tract and led to functional improvement. Thus, microtubule stabilization reduces fibrotic scarring and enhances the capacity of axons to grow.


Subject(s)
Axons/physiology , Cicatrix/prevention & control , Microtubules/metabolism , Paclitaxel/administration & dosage , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/physiopathology , Spinal Cord Regeneration , Animals , Cells, Cultured , Chondroitin Sulfate Proteoglycans/metabolism , Cicatrix/pathology , Female , Ganglia, Spinal/cytology , Kinesins/metabolism , Microtubules/drug effects , Paclitaxel/pharmacology , Protein Transport , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/physiology , Signal Transduction , Smad2 Protein/metabolism , Spinal Cord/cytology , Spinal Cord/drug effects , Spinal Cord Injuries/pathology , Transforming Growth Factor beta/metabolism
7.
Dev Neurobiol ; 70(8): 565-88, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20506164

ABSTRACT

Proper neural circuitry requires that growth cones, motile tips of extending axons, respond to molecular guidance cues expressed in the developing organism. However, it is unclear how guidance cues modify the cytoskeleton to guide growth cone pathfinding. Here, we show acute treatment with two attractive guidance cues, nerve growth factor (NGF) and netrin-1, for embryonic dorsal root ganglion and temporal retinal neurons, respectively, results in increased growth cone membrane protrusion, actin polymerization, and filamentous actin (F-actin). ADF/cofilin (AC) family proteins facilitate F-actin dynamics, and we found the inactive phosphorylated form of AC is decreased in NGF- or netrin-1-treated growth cones. Directly increasing AC activity mimics addition of NGF or netrin-1 to increase growth cone protrusion and F-actin levels. Extracellular gradients of NGF, netrin-1, and a cell-permeable AC elicit attractive growth cone turning and increased F-actin barbed ends, F-actin accumulation, and active AC in growth cone regions proximal to the gradient source. Reducing AC activity blunts turning responses to NGF and netrin. Our results suggest that gradients of NGF and netrin-1 locally activate AC to promote actin polymerization and subsequent growth cone turning toward the side containing higher AC activity.


Subject(s)
Actin Depolymerizing Factors/metabolism , Chemotaxis/physiology , Growth Cones/physiology , Nerve Growth Factor/metabolism , Nerve Growth Factors/metabolism , Neurons/physiology , Tumor Suppressor Proteins/metabolism , Actins/metabolism , Amphibian Proteins/metabolism , Animals , Avian Proteins/metabolism , Cell Membrane/physiology , Cell Movement/physiology , Cells, Cultured , Chick Embryo , Extracellular Space/metabolism , Ganglia, Spinal/embryology , Ganglia, Spinal/physiology , In Vitro Techniques , Netrin-1 , Phosphorylation , Protein Multimerization , Retinal Neurons/physiology , Spinal Cord/embryology , Spinal Cord/physiology , Xenopus laevis
8.
J Neurosci ; 30(20): 6930-43, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20484635

ABSTRACT

Neuronal migration and axon growth, key events during neuronal development, require distinct changes in the cytoskeleton. Although many molecular regulators of polarity have been identified and characterized, relatively little is known about their physiological role in this process. To study the physiological function of Rac1 in neuronal development, we have generated a conditional knock-out mouse, in which Rac1 is ablated in the whole brain. Rac1-deficient cerebellar granule neurons, which do not express other Rac isoforms, showed impaired neuronal migration and axon formation both in vivo and in vitro. In addition, Rac1 ablation disrupts lamellipodia formation in growth cones. The analysis of Rac1 effectors revealed the absence of the Wiskott-Aldrich syndrome protein (WASP) family verprolin-homologous protein (WAVE) complex from the plasma membrane of knock-out growth cones. Loss of WAVE function inhibited axon growth, whereas overexpression of a membrane-tethered WAVE mutant partially rescued axon growth in Rac1-knock-out neurons. In addition, pharmacological inhibition of the WAVE complex effector Arp2/3 also reduced axon growth. We propose that Rac1 recruits the WAVE complex to the plasma membrane to enable actin remodeling necessary for axon growth.


Subject(s)
Cell Movement/physiology , Neurons/physiology , Wiskott-Aldrich Syndrome Protein Family/metabolism , rac1 GTP-Binding Protein/metabolism , Angiopoietin-Like Protein 2 , Angiopoietin-like Proteins , Angiopoietins/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Axons/drug effects , Axons/metabolism , Bromodeoxyuridine/metabolism , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cells, Cultured , Cerebellum/cytology , Cerebellum/growth & development , Cofilin 1/metabolism , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Growth Cones/drug effects , Growth Cones/metabolism , Ki-67 Antigen/metabolism , Luminescent Proteins/genetics , Mice , Mice, Knockout , Mutation/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Organ Culture Techniques/methods , RNA Interference/physiology , RNA, Small Interfering/pharmacology , Transfection/methods , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/deficiency , rhoA GTP-Binding Protein/metabolism
10.
Dev Neurobiol ; 69(12): 761-79, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19513994

ABSTRACT

Axonogenesis involves a shift from uniform delivery of materials to all neurites to preferential delivery to the putative axon, supporting its more rapid extension. Waves, growth cone-like structures that propagate down the length of neurites, were shown previously to correlate with neurite growth in dissociated cultured hippocampal neurons. Waves are similar to growth cones in their structure, composition and dynamics. Here, we report that waves form in all undifferentiated neurites, but occur more frequently in the future axon during initial neuronal polarization. Moreover, wave frequency and their impact on neurite growth are altered in neurons treated with stimuli that enhance axonogenesis. Coincident with wave arrival, growth cones enlarge and undergo a marked increase in dynamics. Through their engorgement of filopodia along the neurite shaft, waves can induce de novo neurite branching. Actin in waves maintains much of its cohesiveness during transport whereas actin in nonwave regions of the neurite rapidly diffuses as measured by live cell imaging of photoactivated GFP-actin and photoconversion of Dendra-actin. Thus, waves represent an alternative axonal transport mechanism for actin. Waves also occur in neurons in organotypic hippocampal slices where they propagate along neurites in the dentate gyrus and the CA regions and induce branching. Taken together, our results indicate that waves are physiologically relevant and contribute to axon growth and branching via the transport of actin and by increasing growth cone dynamics.


Subject(s)
Axons/physiology , Growth Cones/physiology , Neurites/physiology , Neurons/cytology , Actins/metabolism , Animals , Axonal Transport/physiology , Axons/metabolism , Cell Movement/physiology , Cells, Cultured , Gene Transfer Techniques , Growth Cones/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/physiology , Image Processing, Computer-Assisted , Immunohistochemistry , Mice , Microscopy, Confocal , Neurites/metabolism , Neurons/metabolism , Neurons/physiology , Organ Culture Techniques , Rats
11.
J Alzheimers Dis ; 18(1): 35-50, 2009.
Article in English | MEDLINE | ID: mdl-19542631

ABSTRACT

Dissociated hippocampal neurons exposed to a variety of degenerative stimuli form neuritic cofilin-actin rods. Here we report on stimulus driven regional rod formation in organotypic hippocampal slices. Ultrastructural analysis of rods formed in slices demonstrates mitochondria and vesicles become entrapped within some rods. We developed a template for combining and mapping data from multiple slices, enabling statistical analysis for the identification of vulnerable sub-regions. Amyloid-beta (Abeta) induces rods predominantly in the dentate gyrus region, and Abeta-induced rods are reversible following washout. Rods that persist 24 h following transient (30 min) ATP-depletion are broadly distributed, whereas rods formed in response to excitotoxic glutamate localize within and nearby the pyramidal neurons. Time-lapse imaging of cofilin-GFP-expressing neurons within slices shows neuronal rod formation begins rapidly and peaks by 10 min of anoxia. In approximately 50% of responding neurons, Abeta-induced rod formation acts via cdc42, an upstream regulator of cofilin. These new observations support a role for cofilin-actin rods in stress-induced disruption of cargo transport and synaptic function within hippocampal neurons and suggest both cdc42-dependent and independent pathways modulate cofilin activity downstream from Abeta.


Subject(s)
Actins/ultrastructure , Amyloid beta-Peptides/toxicity , Brain Mapping/methods , Cofilin 1/ultrastructure , Hippocampus/ultrastructure , cdc42 GTP-Binding Protein/physiology , Actins/physiology , Animals , Chickens , Cofilin 1/physiology , Female , Hippocampus/drug effects , Hippocampus/physiology , Male , Mice , Mice, Knockout , Mice, Transgenic , Neurons/physiology , Neurons/ultrastructure , Organ Culture Techniques , Pregnancy , Rabbits , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Stress, Physiological , Synapses/metabolism , Synapses/ultrastructure
12.
Nat Rev Neurosci ; 9(2): 136-47, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18209731

ABSTRACT

Higher-order actin-based networks (actin superstructures) are important for growth-cone motility and guidance. Principles for generating, organizing and remodelling actin superstructures have emerged from recent findings in cell-free systems, non-neuronal cells and growth cones. This Review examines how actin superstructures are initiated de novo at the leading-edge membrane and how the spontaneous organization of actin superstructures is driven by ensembles of actin-binding proteins. How the regulation of actin-binding proteins can affect growth-cone turning and axonal regeneration is also discussed.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Differentiation/physiology , Central Nervous System/embryology , Central Nervous System/metabolism , Growth Cones/metabolism , Microfilament Proteins/metabolism , Actin Cytoskeleton/ultrastructure , Animals , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Movement/physiology , Central Nervous System/cytology , Growth Cones/ultrastructure , Humans , Neural Pathways/cytology , Neural Pathways/embryology , Neural Pathways/metabolism , Neuronal Plasticity/physiology
13.
J Neurosci ; 27(48): 13117-29, 2007 Nov 28.
Article in English | MEDLINE | ID: mdl-18045906

ABSTRACT

The establishment of polarity is an essential process in early neuronal development. Although a number of molecules controlling neuronal polarity have been identified, genetic evidence about their physiological roles in this process is mostly lacking. We analyzed the consequences of loss of Cdc42, a central regulator of polarity in multiple systems, on the polarization of mammalian neurons. Genetic ablation of Cdc42 in the brain led to multiple abnormalities, including striking defects in the formation of axonal tracts. Neurons from the Cdc42 null animals sprouted neurites but had a strongly suppressed ability to form axons both in vivo and in culture. This was accompanied by disrupted cytoskeletal organization, enlargement of the growth cones, and inhibition of filopodial dynamics. Axon formation in the knock-out neurons was rescued by manipulation of the actin cytoskeleton, indicating that the effects of Cdc42 ablation are exerted through modulation of actin dynamics. In addition, the knock-outs showed a specific increase in the phosphorylation (inactivation) of the Cdc42 effector cofilin. Furthermore, the active, nonphosphorylated form of cofilin was enriched in the axonal growth cones of wild-type, but not of mutant, neurons. Importantly, cofilin knockdown resulted in polarity defects quantitatively analogous to the ones seen after Cdc42 ablation. We conclude that Cdc42 is a key regulator of axon specification, and that cofilin is a physiological downstream effector of Cdc42 in this process.


Subject(s)
Actin Depolymerizing Factors/metabolism , Neurons/physiology , cdc42 GTP-Binding Protein/physiology , Actins/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/physiology , Axons/physiology , Cell Polarity/drug effects , Cell Polarity/physiology , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Embryo, Mammalian , Gene Expression Regulation/physiology , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Mice , Mice, Knockout , Microtubule-Associated Proteins , Phosphorylation , RNA Interference/physiology , cdc42 GTP-Binding Protein/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...