Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
J Neurosci ; 34(17): 5895-908, 2014 Apr 23.
Article in English | MEDLINE | ID: mdl-24760849

ABSTRACT

During development extrinsic guidance cues modulate the peripheral actin network in growth cones to direct axons to their targets. We wanted to understand the role of the actin nucleator Arp2/3 in growth cone actin dynamics and guidance. Since growth cones migrate in association with diverse adhesive substrates during development, we probed the hypothesis that the functional significance of Arp2/3 is substrate dependent. We report that Arp2/3 inhibition led to a reduction in the number of filopodia and growth cone F-actin content on laminin and L1. However, we found substrate-dependent differences in growth cone motility, actin retrograde flow, and guidance after Arp2/3 inhibition, suggesting that its role, and perhaps that of other actin binding proteins, in growth cone motility is substrate dependent.


Subject(s)
Actin Cytoskeleton/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Actins/metabolism , Cell Movement/physiology , Growth Cones/metabolism , Animals , Axons/metabolism , Cells, Cultured , Chick Embryo , Laminin/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neurons/metabolism , Pseudopodia/metabolism
2.
J Neurochem ; 129(2): 221-34, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24164353

ABSTRACT

Motile growth cones lead growing axons through developing tissues to synaptic targets. These behaviors depend on the organization and dynamics of actin filaments that fill the growth cone leading margin [peripheral (P-) domain]. Actin filament organization in growth cones is regulated by actin-binding proteins that control all aspects of filament assembly, turnover, interactions with other filaments and cytoplasmic components, and participation in producing mechanical forces. Actin filament polymerization drives protrusion of sensory filopodia and lamellipodia, and actin filament connections to the plasma membrane link the filament network to adhesive contacts of filopodia and lamellipodia with other surfaces. These contacts stabilize protrusions and transduce mechanical forces generated by actomyosin activity into traction that pulls an elongating axon along the path toward its target. Adhesive ligands and extrinsic guidance cues bind growth cone receptors and trigger signaling activities involving Rho GTPases, kinases, phosphatases, cyclic nucleotides, and [Ca++] fluxes. These signals regulate actin-binding proteins to locally modulate actin polymerization, interactions, and force transduction to steer the growth cone leading margin toward the sources of attractive cues and away from repellent guidance cues.


Subject(s)
Actins/metabolism , Growth Cones/physiology , Actin Cytoskeleton/physiology , Animals , Cell Adhesion/physiology , Cell Movement/physiology , Humans , Microfilament Proteins/metabolism , Pseudopodia/physiology
3.
Cytoskeleton (Hoboken) ; 69(7): 496-505, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22328420

ABSTRACT

Axonal growth cones turn away from repulsive guidance cues. This may start with reduced protrusive motility in the region the growth cone leading margin that is closer to the source of repulsive cue. Using explants of E7 chick temporal retina, we examine the effects of two repulsive guidance cues, ephrin-A2 and slit3, on retinal ganglion cell growth cone protrusive activity, total F-actin, free F-actin barbed ends, and the activities (phosphorylation states) of actin regulatory proteins, ADF/cofilin and ezrin, radixin, moesin (ERM) proteins. Ephrin-A2 rapidly stops protrusive activity simultaneously with reducing F-actin, free barbed ends and the activities of ADF/cofilin and ERM proteins. Slit3 also stops protrusion and reduces the activities of ADF/cofilin and ERM proteins. We interpret these results as indicating that repulsive guidance cues inhibit actin polymerization and actin-membrane linkage to stop protrusive activity. Retrograde F-actin flow withdraws actin to the C-domain, where F-actin bundles interact with myosin II to generate contractile forces that can collapse and retract the growth cone. Our results suggest that common mechanisms are used by repulsive guidance cue to disable growth cone motility and remodel growing axon terminals.


Subject(s)
Actin Depolymerizing Factors/metabolism , DNA-Binding Proteins/metabolism , Ephrin-A2/pharmacology , Growth Cones/drug effects , Membrane Proteins/pharmacology , Retina/drug effects , Transcription Factors/metabolism , Actins/metabolism , Animals , Cells, Cultured , Chickens , Ephrin-A2/metabolism , Female , Growth Cones/metabolism , Humans , Male , Membrane Proteins/metabolism , Phosphorylation , Retina/embryology
4.
J Neurosci ; 32(1): 282-96, 2012 Jan 04.
Article in English | MEDLINE | ID: mdl-22219290

ABSTRACT

The development of a functioning neural network relies on responses of axonal growth cones to molecular guidance cues that are encountered en route to their target tissue. Nerve growth factor (NGF) and neurotrophin-3 serve as attractive cues for chick embryo sensory growth cones in vitro and in vivo, but little is known about the actin-binding proteins necessary to mediate this response. The evolutionarily conserved ezrin/radixin/moesin (ERM) family of proteins can tether actin filaments to the cell membrane when phosphorylated at a conserved threonine residue. Here we show that acute neurotrophin stimulation rapidly increases active phospho-ERM levels in chick sensory neuron growth cone filopodia, coincident with an increase in filopodial L1 and ß-integrin. Disrupting ERM function with a dominant-negative construct (DN-ERM) results in smaller and less motile growth cones with disorganized actin filaments. Previously, we found that NGF treatment increases actin-depolymerizing factor (ADF)/cofilin activity and growth cone F-actin (Marsick et al., 2010). Here, we show this F-actin increase, as well as attractive turning to NGF, is blocked when ERM function is disrupted despite normal activation of ADF/cofilin. We further show that DN-ERM expression disrupts leading edge localization of active ADF/cofilin and free F-actin barbed ends. Moreover, filopodial phospho-ERM levels are increased by incorporation of active ADF/cofilin and reduced by knockdown of L1CAM.Together, these data suggest that ERM proteins organize actin filaments in sensory neuron growth cones and are crucial for neurotrophin-induced remodeling of F-actin and redistribution of adhesion receptors.


Subject(s)
Actins/metabolism , Cytoskeletal Proteins/metabolism , Growth Cones/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Sensory Receptor Cells/metabolism , Animals , Chick Embryo , Chickens , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/genetics , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Growth Cones/drug effects , Growth Cones/ultrastructure , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Microfilament Proteins/antagonists & inhibitors , Microfilament Proteins/genetics , Primary Cell Culture , Sensory Receptor Cells/cytology , Sensory Receptor Cells/drug effects
5.
J Vis Exp ; (49)2011 Mar 17.
Article in English | MEDLINE | ID: mdl-21445046

ABSTRACT

The motile tips of growing axons are called growth cones. Growth cones lead navigating axons through developing tissues by interacting with locally expressed molecular guidance cues that bind growth cone receptors and regulate the dynamics and organization of the growth cone cytoskeleton. The main target of these navigational signals is the actin filament meshwork that fills the growth cone periphery and that drives growth cone motility through continual actin polymerization and dynamic remodeling. Positive or attractive guidance cues induce growth cone turning by stimulating actin filament (F-actin) polymerization in the region of the growth cone periphery that is nearer the source of the attractant cue. This actin polymerization drives local growth cone protrusion, adhesion of the leading margin and axonal elongation toward the attractant. Actin filament polymerization depends on the availability of sufficient actin monomer and on polymerization nuclei or actin filament barbed ends for the addition of monomer. Actin monomer is abundantly available in chick retinal and dorsal root ganglion (DRG) growth cones. Consequently, polymerization increases rapidly when free F-actin barbed ends become available for monomer addition. This occurs in chick DRG and retinal growth cones via the local activation of the F-actin severing protein actin depolymerizing factor (ADF/cofilin) in the growth cone region closer to an attractant. This heightened ADF/cofilin activity severs actin filaments to create new F-actin barbed ends for polymerization. The following method demonstrates this mechanism. Total content of F-actin is visualized by staining with fluorescent phalloidin. F-actin barbed ends are visualized by the incorporation of rhodamine-actin within growth cones that are permeabilized with the procedure described in the following, which is adapted from previous studies of other motile cells. When rhodamine-actin is added at a concentration above the critical concentration for actin monomer addition to barbed ends, rhodamine-actin assembles onto free barbed ends. If the attractive cue is presented in a gradient, such as being released from a micropipette positioned to one side of a growth cone, the incorporation of rhodamine-actin onto F-actin barbed ends will be greater in the growth cone side toward the micropipette. Growth cones are small and delicate cell structures. The procedures of permeabilization, rhodamine-actin incorporation, fixation and fluorescence visualization are all carefully done and can be conducted on the stage of an inverted microscope. These methods can be applied to studying local actin polymerization in migrating neurons, other primary tissue cells or cell lines.


Subject(s)
Actins/metabolism , Fluorescent Dyes/metabolism , Growth Cones/metabolism , Microscopy, Fluorescence/methods , Rhodamines/metabolism , Actins/chemistry , Animals , Cell Membrane Permeability , Chick Embryo , Fluorescent Dyes/chemistry , Ganglia, Spinal/chemistry , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Growth Cones/chemistry , Rhodamines/chemistry
6.
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
7.
Results Probl Cell Differ ; 48: 65-90, 2009.
Article in English | MEDLINE | ID: mdl-19582412

ABSTRACT

Actin filaments are thin polymers of the 42 kD protein actin. In mature axons a network of subaxolemmal actin filaments provide stability for membrane integrity and a substrate for short distance transport of cargos. In developing neurons dynamic regulation of actin polymerization and organization mediates axonal morphogenesis and axonal pathfinding to synaptic targets. Other changes in axonal shape, collateral branching, branch retraction, and axonal regeneration, also depend on actin filament dynamics. Actin filament organization is regulated by a diversity of actin-binding proteins (ABP). ABP are the focus of complex extrinsic and intrinsic signaling pathways, and many neurological pathologies and dysfunctions arise from defective regulation of ABP function.


Subject(s)
Actin Cytoskeleton/physiology , Actins/physiology , Axons , Cytoskeleton/physiology , Animals , Axons/pathology , Axons/ultrastructure , Biological Transport , Humans , Nerve Regeneration
8.
J Neurosci ; 29(3): 638-52, 2009 Jan 21.
Article in English | MEDLINE | ID: mdl-19158291

ABSTRACT

Recent evidence suggests that growth cone responses to guidance cues require local protein synthesis. Using chick neurons, we investigated whether protein synthesis is required for growth cones of several types to respond to guidance cues. First, we found that global inhibition of protein synthesis stops axonal elongation after 2 h. When protein synthesis inhibitors were added 15 min before adding guidance cues, we found no changes in the typical responses of retinal, sensory, and sympathetic growth cones. In the presence of cycloheximide or anisomycin, ephrin-A2, slit-3, and semaphorin3A still induced growth cone collapse and loss of actin filaments, nerve growth factor (NGF) and neurotrophin-3 still induced growth cone protrusion and increased filamentous actin, and sensory growth cones turned toward an NGF source. In compartmented chambers that separated perikarya from axons, axons grew for 24-48 h in the presence of cycloheximide and responded to negative and positive cues. Our results indicate that protein synthesis is not strictly required in the mechanisms for growth cone responses to many guidance cues. Differences between our results and other studies may exist because of different cellular metabolic levels in in vitro conditions and a difference in when axonal functions become dependent on local protein synthesis.


Subject(s)
Axons/metabolism , Cues , Growth Cones/physiology , Neurons/cytology , Actins/metabolism , Animals , Anisomycin/pharmacology , Axons/drug effects , Chick Embryo , Cycloheximide/pharmacology , Ephrin-A2/metabolism , Ganglia, Spinal/cytology , Green Fluorescent Proteins/genetics , Growth Cones/drug effects , Nerve Growth Factor/pharmacology , Neurons/drug effects , Neurons/metabolism , Organ Culture Techniques , Protein Synthesis Inhibitors/pharmacology , Retina/cytology , Semaphorin-3A/pharmacology , Statistics, Nonparametric , Time Factors , Transfection , rhoA GTP-Binding Protein/metabolism
9.
Dev Neurobiol ; 67(7): 976-86, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17506497

ABSTRACT

It is now well established that new proteins are synthesized in the distal segments of elongating axons, where they may play an essential role in some guidance decisions. It remains unclear, however, whether distal protein synthesis also plays an essential role in axon growth per se. Previous in vitro experiments have shown that blocking protein synthesis in distal axons has no effect on the rate of axonal advance. However, because these experiments were performed in vitro and over a relatively short time period, the role of distal protein synthesis over longer periods and in a native tissue environment remained untested. Here, we tested whether protein synthesis in distal axons plays an essential role in the elongation of descending axons in the embryonic spinal cord. We developed an in situ model of the brainstem-spinal projection of the embryonic chick, and developed a split-chamber method in which inhibitors of proteins synthesis could be applied independently to cell bodies in the brainstem or to distal axons in the spinal cord. When protein synthesis was blocked in distal axons, axon growth remained robust for 2 days, which is the length of the experiment. However, when protein synthesis was blocked only in the brainstem, axonal elongation in the spinal cord ceased within 6 h. These data showed that protein synthesis in the distal axon is not essential to continue the advance of axons. Rather, essential proteins are synthesized more proximally and then transported rapidly to the distal axon.


Subject(s)
Brain Stem/embryology , Cell Differentiation/physiology , Efferent Pathways/embryology , Growth Cones/metabolism , Nerve Tissue Proteins/biosynthesis , Spinal Cord/embryology , Animals , Axonal Transport/physiology , Brain Stem/cytology , Brain Stem/metabolism , Carbocyanines , Cell Compartmentation/physiology , Chick Embryo , Coculture Techniques , Efferent Pathways/cytology , Efferent Pathways/metabolism , Growth Cones/ultrastructure , Organ Culture Techniques , Protein Synthesis Inhibitors/pharmacology , Spinal Cord/cytology , Spinal Cord/metabolism , Time Factors
10.
J Neurobiol ; 66(14): 1564-83, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17058193

ABSTRACT

Embryonic birds and mammals are capable of axon regeneration after spinal cord injury, but this ability is lost during a discrete developmental transition. We recently showed that changes within maturing neurons, as opposed to changes solely in the spinal cord environment, significantly restrict axon regeneration during development. The developmental changes within neurons that limit axon regeneration remain unclear. One gap in knowledge is the identity of the adhesive receptors that embryonic neurons use to extend axons in the spinal cord. Here we test the roles of L1/NgCAM, beta1 integrin, and cadherins, using a coculture system in which embryonic chick brainstem neurons regenerate axons into an explant of embryonic spinal cord. By in vivo and in vitro methods, we found that brainstem neurons reduce axonal expression of L1 as they mature. Disrupting either L1 or beta1 integrin function individually in our coculture system partially inhibited growth of brainstem axons in spinal cords, while disrupting cadherin function alone had no effect. However, when all three adhesive receptors were blocked simultaneously, axon growth in the spinal cord was reduced by 90%. Using immunohistochemistry and in situ hybridization we show that during the period when neurons lose their regenerative capacity they reduce expression of mRNA for N-cadherin, and reduce axonal L1/NgCAM protein through a post-transcriptional mechanism. These data show that embryonic neurons use L1/NgCAM, beta1 integrin, and cadherin receptors for axon regeneration in the embryonic spinal cord, and raise the possibility that a reduced expression of these essential receptors may contribute to the low-regenerative capacity of older neurons.


Subject(s)
Axons/physiology , Cadherins/physiology , Integrin beta1/physiology , Neural Cell Adhesion Molecule L1/physiology , Regeneration/physiology , Spinal Cord/cytology , Age Factors , Amino Acids , Animals , Antibodies/pharmacology , Axons/drug effects , Brain Stem , Cadherins/immunology , Calcium/pharmacology , Chick Embryo , Dose-Response Relationship, Drug , Drug Interactions , Gene Expression Regulation, Developmental , Immunohistochemistry/methods , Integrin beta1/immunology , Neural Cell Adhesion Molecule L1/immunology , Neurons/cytology , Neurons/physiology , Organ Culture Techniques , Regeneration/drug effects , Spinal Cord/embryology , Transfection/methods
11.
J Neurobiol ; 66(4): 348-60, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16408302

ABSTRACT

Embryonic birds and mammals display a remarkable ability to regenerate axons after spinal injury, but then lose this ability during a discrete developmental transition. To explain this transition, previous research has emphasized the emergence of myelin and other inhibitory factors in the environment of the spinal cord. However, research in other CNS tracts suggests an important role for neuron-intrinsic limitations to axon regeneration. Here we re-examine this issue quantitatively in the hindbrain-spinal projection of the embryonic chick. Using heterochronic cocultures we show that maturation of the spinal cord environment causes a 55% reduction in axon regeneration, while maturation of hindbrain neurons causes a 90% reduction. We further show that young neurons transplanted in vivo into older spinal cord can regenerate axons into myelinated white matter, while older axons regenerate poorly and have reduced growth cone motility on a variety of growth-permissive ligands in vitro, including laminin, L1, and N-cadherin. Finally, we use video analysis of living growth cones to directly document an age-dependent decline in the motility of brainstem axons. These data show that developmental changes in both the spinal cord environment and in brainstem neurons can reduce regeneration, but that the effect of the environment is only partial, while changes in neurons by themselves cause a nearly complete reduction in regeneration. We conclude that maturational events within neurons are a primary cause for the failure of axon regeneration in the spinal cord.


Subject(s)
Aging/physiology , Brain Stem/embryology , Efferent Pathways/embryology , Growth Cones/physiology , Nerve Regeneration/physiology , Spinal Cord/embryology , Animals , Brain Stem/cytology , Brain Stem/physiology , Cadherins/metabolism , Cell Communication/physiology , Cell Differentiation/physiology , Chick Embryo , Coculture Techniques , Cues , Efferent Pathways/cytology , Efferent Pathways/physiology , Growth Cones/ultrastructure , Laminin/metabolism , Nerve Fibers, Myelinated/physiology , Nerve Fibers, Myelinated/ultrastructure , Neural Cell Adhesion Molecule L1/metabolism , Neuronal Plasticity/physiology , Organ Culture Techniques , Reticular Formation/cytology , Reticular Formation/embryology , Reticular Formation/physiology , Spinal Cord/cytology , Spinal Cord/physiology
12.
J Neurobiol ; 66(2): 103-14, 2006 Feb 05.
Article in English | MEDLINE | ID: mdl-16215999

ABSTRACT

Rho family GTPases have important roles in mediating the effects of guidance cues and growth factors on the motility of neuronal growth cones. We previously showed that the neurotrophin BDNF regulates filopodial dynamics on growth cones of retinal ganglion cell axons through activation of the actin regulatory proteins ADF and cofilin by inhibiting a RhoA-dependent pathway that phosphorylates (inactivates) ADF/cofilin. The GTPase Cdc42 has also been implicated in mediating the effects of positive guidance cues. In this article we investigated whether Cdc42 is involved in the effects of BDNF on filopodial dynamics. BDNF treatment increases Cdc42 activity in retinal neurons, and neuronal incorporation of constitutively active Cdc42 mimics the increases in filopodial number and length. Furthermore, constitutively active and dominant negative Cdc42 decreased and increased, respectively, the activity of RhoA in retinal growth cones, indicating crosstalk between these GTPases in retinal growth cones. Constitutively active Cdc42 mimicked the activation of ADF/cofilin that resulted from BDNF treatment, while dominant negative Cdc42 blocked the effects of BDNF on filopodia and ADF/cofilin. The inability of dominant negative Cdc42 to block ADF/cofilin activation and stimulation of filopodial dynamics by the ROCK inhibitor Y-27632 indicate interaction between Cdc42 and RhoA occurs upstream of ROCK. Our results demonstrate crosstalk occurs between GTPases in mediating the effects of BDNF on growth cone motility, and Cdc42 activity can promote actin dynamics via activation of ADF/cofilin.


Subject(s)
Actin Depolymerizing Factors/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Growth Cones/metabolism , Pseudopodia/metabolism , Retina/metabolism , cdc42 GTP-Binding Protein/metabolism , Animals , Cells, Cultured , Chick Embryo , Enzyme Activation/physiology , Fluorescent Antibody Technique , Image Processing, Computer-Assisted
13.
Nat Neurosci ; 9(1): 50-7, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16369480

ABSTRACT

Lis1 gene defects impair neuronal migration, causing the severe human brain malformation lissencephaly. Although much is known about its interactions with microtubules, microtubule-binding proteins such as CLIP-170, and with the dynein motor complex, the response of Lis1 to neuronal motility signals has not been elucidated. Lis1 deficiency is associated with deregulation of the Rho-family GTPases Cdc42, Rac1 and RhoA, and ensuing actin cytoskeletal defects, but the link between Lis1 and Rho GTPases remains unclear. We report here that calcium influx enhances neuronal motility through Lis1-dependent regulation of Rho GTPases. Lis1 promotes Cdc42 activation through interaction with the calcium sensitive GTPase scaffolding protein IQGAP1, maintaining the perimembrane localization of IQGAP1 and CLIP170 and thereby tethering microtubule ends to the cortical actin cytoskeleton. Lis1 thus is a key component of neuronal motility signal transduction that regulates the cytoskeleton by complexing with IQGAP1, active Cdc42 and CLIP-170 upon calcium influx.


Subject(s)
Calcium/physiology , Cell Movement/physiology , Microtubule-Associated Proteins/physiology , Neurons/physiology , cdc42 GTP-Binding Protein/physiology , ras GTPase-Activating Proteins/physiology , 1-Alkyl-2-acetylglycerophosphocholine Esterase , Actins/metabolism , Aniline Compounds , Animals , Blotting, Western , Calcium/metabolism , Cells, Cultured , Fluorescent Dyes , Hippocampus/cytology , Hippocampus/metabolism , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Knockout , Microscopy, Video , Plasmids/genetics , Transfection , Xanthenes , rho GTP-Binding Proteins/metabolism
14.
J Neurosci ; 25(2): 281-90, 2005 Jan 12.
Article in English | MEDLINE | ID: mdl-15647471

ABSTRACT

Modifier of cell adhesion (MOCA) is a member of the dedicator of cytokinesis 180 family of proteins and is highly expressed in CNS neurons. MOCA is associated with Alzheimer's disease tangles and regulates the accumulation of amyloid precursor protein and beta-amyloid. Here, we report that MOCA modulates cell-cell adhesion and morphology. MOCA increases the accumulation of adherens junction proteins, including N-cadherin and beta-catenin, whereas reducing endogenous MOCA expression lowers cell-cell aggregation and N-cadherin expression. MOCA colocalizes with N-cadherin and actin in areas of cell-cell and cell substratum contact and is expressed in neuronal processes. MOCA accumulates during neuronal differentiation, and its expression enhances NGF-induced neurite outgrowth and morphological complexity. We conclude that MOCA regulates N-cadherin-mediated cell-cell adhesion and neurite outgrowth.


Subject(s)
Cadherins/physiology , Cell Adhesion/physiology , Guanine Nucleotide Exchange Factors/physiology , Nerve Tissue Proteins/physiology , Neurites/ultrastructure , Animals , Cadherins/biosynthesis , Cell Aggregation/physiology , Cell Line , Cytoskeletal Proteins/biosynthesis , Humans , Molecular Sequence Data , PC12 Cells , RNA, Small Interfering , Rats , Trans-Activators/biosynthesis , Transfection , beta Catenin
15.
J Neurobiol ; 62(1): 134-47, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15452851

ABSTRACT

Growth cone navigation is guided by extrinsic environmental proteins, called guidance cues. Many in vitro studies have characterized growth cone turning up and down gradients of soluble guidance cues. Although previous studies have shown that axonal elongation rates can be regulated by gradients of surface-bound molecules, there are no convincing demonstrations of growth cones turning to migrate up a surface-bound gradient of an adhesive ligand or guidance cue. In order to test this mode of axonal guidance, we used a photo-immobilization technique to create grids and gradients of an adhesive laminin peptide on polystyrene culture dish surfaces. Chick embryo dorsal root ganglia (DRGs) were placed on peptide grid patterns containing surface-bound gradients of the IKVAV-containing peptide. DRG growth cones followed a path of surface-bound peptide to the middle of a perpendicularly oriented gradient with a 25% concentration difference across 30 microm. The majority of growth cones turned and migrated up the gradient, turning until they were oriented directly up the gradient. Growth cones slowed their migration when they encountered the gradient, but growth cone velocity returned to the previous rate after turning up or down the gradient. This resembles in vivo situations where growth cones slow at a choice point before changing the direction of axonal extension. Thus, these results support the hypothesis that mechanisms of axonal guidance include growth cone orientation by gradients of surface-bound adhesive molecules and guidance cues.


Subject(s)
Cell Differentiation/physiology , Chemotaxis/physiology , Ganglia, Spinal/embryology , Growth Cones/metabolism , Laminin/metabolism , Animals , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Communication/physiology , Cell Differentiation/drug effects , Chemotactic Factors/metabolism , Chemotactic Factors/pharmacology , Chemotaxis/drug effects , Chick Embryo , Cues , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Growth Cones/drug effects , Growth Cones/ultrastructure , Laminin/pharmacology , Lasers , Nerve Growth Factors/metabolism , Nerve Growth Factors/pharmacology , Neural Cell Adhesion Molecules/metabolism , Peptides/metabolism , Peptides/pharmacology , Photic Stimulation
16.
J Neurosci ; 24(47): 10741-9, 2004 Nov 24.
Article in English | MEDLINE | ID: mdl-15564592

ABSTRACT

The molecular mechanisms by which neurotrophins regulate growth cone motility are not well understood. This study investigated the signaling involved in transducing BDNF-induced increases of filopodial dynamics. Our results indicate that BDNF regulates filopodial length and number through a Rho kinase-dependent mechanism. Additionally, actin depolymerizing factor (ADF)/cofilin activity is necessary and sufficient to transduce the effects of BDNF. Our data indicate that activation of ADF/cofilin mimics the effects of BDNF on filopodial dynamics, whereas ADF/cofilin inactivity blocks the effects of BDNF. Furthermore, BDNF promotes the activation of ADF/cofilin by reducing the phosphorylation of ADF/cofilin. Although inhibition of myosin II also enhances filopodial length, our results indicate that BDNF signaling is independent of myosin II activity and that the two pathways result in additive effects on filopodial length. Thus, filopodial extension is regulated by at least two independent mechanisms. The BDNF-dependent pathway works via regulation of ADF/cofilin, independently of myosin II activity.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Growth Cones/physiology , Microfilament Proteins/physiology , Pseudopodia/physiology , Retina/ultrastructure , 14-3-3 Proteins/physiology , Actin Depolymerizing Factors , Animals , Brain-Derived Neurotrophic Factor/antagonists & inhibitors , Chick Embryo , Destrin , Growth Cones/ultrastructure , Heterocyclic Compounds, 4 or More Rings/pharmacology , Intracellular Signaling Peptides and Proteins , Microfilament Proteins/metabolism , Myosin Type II/antagonists & inhibitors , Myosin Type II/physiology , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/physiology , Signal Transduction/physiology , Tissue Culture Techniques , rho-Associated Kinases
17.
J Neurosci ; 24(18): 4363-72, 2004 May 05.
Article in English | MEDLINE | ID: mdl-15128850

ABSTRACT

The mechanisms by which neurotrophins regulate growth cone motility are unclear. We investigated the role of the p75 neurotrophin receptor (p75NTR) in mediating neurotrophin-induced increases in filopodial length. Our data demonstrate that neurotrophin binding to p75NTR is necessary and sufficient to regulate filopodial dynamics. Furthermore, retinal and dorsal root ganglion growth cones from p75 mutant mice are insensitive to neurotrophins but display enhanced filopodial lengths comparable with neurotrophin-treated wild-type growth cones. This suggests unoccupied p75NTR negatively regulates filopodia length. Furthermore, p75NTR regulates RhoA activity to mediate filopodial dynamics. Constitutively active RhoA blocks neurotrophin-induced increases in filopodial length, whereas inhibition of RhoA enhances filopodial lengths, similar to neurotrophin treatment. BDNF treatment of retinal neurons results in reduced RhoA activity. Furthermore, p75 mutant neurons display reduced levels of activated RhoA compared with wild-type counterparts, consistent with the enhanced filopodial lengths observed on mutant growth cones. These observations suggest that neurotrophins regulate filopodial dynamics by depressing the activation of RhoA that occurs through p75NTR signaling.


Subject(s)
Growth Cones/physiology , Pseudopodia/physiology , Receptors, Nerve Growth Factor/physiology , Signal Transduction/physiology , rhoA GTP-Binding Protein/metabolism , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Chick Embryo , Dose-Response Relationship, Drug , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Growth Cones/drug effects , Growth Cones/enzymology , Mice , Mice, Mutant Strains , Nerve Growth Factors/metabolism , Nerve Growth Factors/pharmacology , Neurons/drug effects , Neurons/metabolism , Neurons/ultrastructure , Pseudopodia/drug effects , Pseudopodia/enzymology , Receptor, Nerve Growth Factor , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Retina/cytology , Retina/embryology , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/genetics
18.
J Neurobiol ; 58(1): 92-102, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14598373

ABSTRACT

The motile behaviors of growth cones at the ends of elongating axons determine pathways of axonal connections in developing nervous systems. Growth cones express receptors for molecular guidance cues in the local environment, and receptor-guidance cue binding initiates cytoplasmic signaling that regulates the cytoskeleton to control growth cone advance, turning, and branching behaviors. The dynamic actin filaments of growth cones are frequently targets of this regulatory signaling. Rho GTPases are key mediators of signaling by guidance cues, although much remains to be learned about how growth cone responses are orchestrated by Rho GTPase signaling to change the dynamics of polymerization, transport, and disassembly of actin filaments. Binding of neurotrophins to Trk and p75 receptors on growth cones triggers changes in actin filament dynamics to regulate several aspects of growth cone behaviors. Activation of Trk receptors mediates local accumulation of actin filaments, while neurotrophin binding to p75 triggers local decrease in RhoA signaling that promotes lengthening of filopodia. Semaphorin IIIA and ephrin-A2 are guidance cues that trigger avoidance or repulsion of certain growth cones, and in vitro responses to these proteins include growth cone collapse. Dynamic changes in the activities of Rho GTPases appear to mediate responses to these cues, although it remains unclear what the changes are in actin filament distribution and dynamic reorganization that result in growth cone collapse. Growth cones in vivo simultaneously encounter positive and negative guidance cues, and thus, growth cone behaviors during axonal pathfinding reflect the complex integration of multiple signaling activities.


Subject(s)
Actin Cytoskeleton/physiology , Growth Cones/physiology , rho GTP-Binding Proteins/physiology , Animals
19.
J Neurosci ; 23(25): 8673-81, 2003 Sep 24.
Article in English | MEDLINE | ID: mdl-14507966

ABSTRACT

Lissencephaly is a severe brain malformation caused by impaired neuronal migration. Lis1, a causative gene, functions in an evolutionarily conserved nuclear translocation pathway regulating dynein motor and microtubule dynamics. Whereas microtubule contributions to neuronal motility are incompletely understood, the actin cytoskeleton is essential for crawling cell movement of all cell types investigated. Lis1 haploinsufficiency is shown here to also result in reduced filamentous actin at the leading edge of migrating neurons, associated with upregulation of RhoA and downregulation of Rac1 and Cdc42 activity. Disruption of RhoA function through pharmacological inhibition of its effector kinase, p160ROCK, restores normal Rac1 and Cdc42 activity and rescues the motility defect in Lis1+/- neurons. These data indicate a previously unrecognized role for Lis1 protein in neuronal motility by promoting actin polymerization through the regulation of Rho GTPase activity. This effect of Lis1 on GTPases does not appear to occur through direct Lis1 binding of Rho, but could involve Lis1 effects on Rho modulatory proteins or on microtubule dynamics.


Subject(s)
Cell Movement/genetics , Cytoskeleton/metabolism , Microtubule-Associated Proteins/deficiency , Neurons/physiology , rho GTP-Binding Proteins/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase , Actins/metabolism , Animals , Animals, Newborn , Cell Migration Inhibition , Cell Movement/drug effects , Cells, Cultured , Cytoskeleton/pathology , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/physiology , Gene Expression Regulation , Heterozygote , Intracellular Signaling Peptides and Proteins , Mice , Microtubule-Associated Proteins/genetics , Nervous System Malformations/genetics , Neurons/cytology , Neurons/drug effects , Phospholipases A/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/genetics , rho-Associated Kinases
20.
J Histochem Cytochem ; 51(4): 435-44, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12642622

ABSTRACT

Nerve growth factor (NGF) and semaphorin3A (Sema3A) are guidance cues found in pathways and targets of developing dorsal root ganglia (DRG) neurons. DRG growth cone motility is regulated by cytoplasmic signaling triggered by these molecules. We investigated interactions of NGF and Sema3A in modulating growth cone behaviors of axons extended from E7 chick embryo DRGs. Axons extending in collagen matrices were repelled by Sema3A released from transfected HEK293 cells. However, if an NGF-coated bead was placed adjacent to Sema3A-producing cells, axons converged at the NGF bead. Growth cones of DRGs raised in 10(-9) M NGF were more resistant to Sema3A-induced collapse than when DRGs were raised in 10(-11) M NGF. After overnight culture in 10(-11) M NGF, 1-hr treatment with 10(-9) M NGF also increased growth cone resistance to Sema3A. Pharmacological studies indicated that the activities of ROCK and PKG participate in the cytoskeletal alterations that lead to Sema3A-induced growth cone collapse, whereas PKA activity is required for NGF-mediated reduction of Sema3A-induced growth cone collapse. These results support the idea that growth cone responses to a guidance cue can be modulated by interactions involving coincident signaling by other guidance cues.


Subject(s)
Growth Cones/physiology , Nerve Growth Factor/physiology , Semaphorins/physiology , Signal Transduction/physiology , Animals , Cells, Cultured , Chick Embryo , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Ganglia, Spinal/metabolism , Ganglia, Spinal/ultrastructure , Humans , Intracellular Signaling Peptides and Proteins , Nerve Growth Factor/metabolism , Protein Serine-Threonine Kinases/metabolism , Semaphorins/metabolism , rho-Associated Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...