Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Sci Signal ; 15(727): eabh2674, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35349303

ABSTRACT

Dual leucine-zipper kinase (DLK; a MAP3K) mediates neuronal responses to diverse injuries and insults through the c-Jun N-terminal kinase (JNK) family of mitogen-activated protein kinases (MAPKs). Here, we identified two ways through which DLK is coupled to the neural-specific isoform JNK3 to control prodegenerative signaling. JNK3 catalyzed positive feedback phosphorylation of DLK that further activated DLK and locked the DLK-JNK3 module in a highly active state. Neither homologous MAP3Ks nor a homologous MAPK could support this positive feedback loop. Unlike the related JNK1 isoform JNK2 and JNK3 promote prodegenerative axon-to-soma signaling and were endogenously palmitoylated. Moreover, palmitoylation targeted both DLK and JNK3 to the same axonal vesicles, and JNK3 palmitoylation was essential for axonal retrograde signaling in response to optic nerve crush injury in vivo. These findings provide previously unappreciated insights into DLK-JNK signaling relevant to neuropathological conditions and answer long-standing questions regarding the selective prodegenerative roles of JNK2 and JNK3.


Subject(s)
Axons , Lipoylation , Axons/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Neurons/metabolism , Signal Transduction
2.
Front Mol Neurosci ; 15: 726962, 2022.
Article in English | MEDLINE | ID: mdl-35264929

ABSTRACT

Axon branching is a fundamental aspect of neuronal morphogenesis, neuronal circuit formation, and response of the nervous system to injury. Sterile alpha and TIR motif containing 1 (SARM1) was initially identified as promoting Wallerian degeneration of axons. We now report a novel function of SARM1 in postnatal sensory neurons; the suppression of axon branching. Axon collateral branches develop from axonal filopodia precursors through the coordination of the actin and microtubule cytoskeleton. In vitro analysis revealed that cultured P0-2 dorsal root ganglion sensory neurons from a SARM1 knockout (KO) mouse exhibit increased numbers of collateral branches and axonal filopodia relative to wild-type neurons. In SARM1 KO mice, cutaneous sensory endings exhibit increased branching in the skin in vivo with normal density of innervation. Transient axonal actin patches serve as cytoskeletal platforms from which axonal filopodia emerge. Live imaging analysis of axonal actin dynamics showed that SARM1 KO neurons exhibit increased rates of axonal actin patch formation and increased probability that individual patches will give rise to a filopodium before dissipating. SARM1 KO axons contain elevated levels of drebrin and cortactin, two actin regulatory proteins that are positive regulators of actin patches, filopodia formation, and branching. Live imaging of microtubule plus tip dynamics revealed an increase in the rate of formation and velocity of polymerizing tips along the axons of SARM1 KO neurons. Stationary mitochondria define sites along the axon where branches may arise, and the axons of SARM1 KO sensory neurons exhibit an increase in stationary mitochondria. These data reveal SARM1 to be a negative regulator of axonal cytoskeletal dynamics and collateral branching.

3.
J Neurosci ; 41(31): 6637-6651, 2021 08 04.
Article in English | MEDLINE | ID: mdl-34252036

ABSTRACT

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


Subject(s)
Axon Guidance/physiology , Glycolysis/physiology , Growth Cones/metabolism , Sensory Receptor Cells/metabolism , Animals , Axons/physiology , Chick Embryo
4.
Elife ; 82019 12 02.
Article in English | MEDLINE | ID: mdl-31789589

ABSTRACT

Neurotrophins are growth factors that have a multitude of roles in the nervous system. We report that neurotrophins induce the fission of mitochondria along embryonic chick sensory axons driven by combined PI3K and Mek-Erk signaling. Following an initial burst of fission, a new steady state of neurotrophin-dependent mitochondria length is established. Mek-Erk controls the activity of the fission mediator Drp1 GTPase, while PI3K may contribute to the actin-dependent aspect of fission. Drp1-mediated fission is required for nerve growth factor (NGF)-induced collateral branching in vitro and expression of dominant negative Drp1 impairs the branching of axons in the developing spinal cord in vivo. Fission is also required for NGF-induced mitochondria-dependent intra-axonal translation of the actin regulatory protein cortactin, a previously determined component of NGF-induced branching. Collectively, these observations unveil a novel biological function of neurotrophins; the regulation of mitochondrial fission and steady state mitochondrial length and density in axons.

5.
Cell Rep ; 23(4): 1005-1019, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29694881

ABSTRACT

Mitochondria shape cytosolic calcium ([Ca2+]c) transients and utilize the mitochondrial Ca2+ ([Ca2+]m) in exchange for bioenergetics output. Conversely, dysregulated [Ca2+]c causes [Ca2+]m overload and induces permeability transition pore and cell death. Ablation of MCU-mediated Ca2+ uptake exhibited elevated [Ca2+]c and failed to prevent stress-induced cell death. The mechanisms for these effects remain elusive. Here, we report that mitochondria undergo a cytosolic Ca2+-induced shape change that is distinct from mitochondrial fission and swelling. [Ca2+]c elevation, but not MCU-mediated Ca2+ uptake, appears to be essential for the process we term mitochondrial shape transition (MiST). MiST is mediated by the mitochondrial protein Miro1 through its EF-hand domain 1 in multiple cell types. Moreover, Ca2+-dependent disruption of Miro1/KIF5B/tubulin complex is determined by Miro1 EF1 domain. Functionally, Miro1-dependent MiST is essential for autophagy/mitophagy that is attenuated in Miro1 EF1 mutants. Thus, Miro1 is a cytosolic Ca2+ sensor that decodes metazoan Ca2+ signals as MiST.


Subject(s)
Calcium/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics , Receptors, G-Protein-Coupled/metabolism , Stress, Physiological , rho GTP-Binding Proteins/metabolism , Animals , HeLa Cells , Humans , Mice , Mice, Mutant Strains , Mitochondria/genetics , Receptors, G-Protein-Coupled/genetics , rho GTP-Binding Proteins/genetics
6.
Dev Neurobiol ; 77(4): 454-473, 2017 04.
Article in English | MEDLINE | ID: mdl-27429169

ABSTRACT

Chondroitin sulfate proteoglycans (CSPGs) inhibit the formation of axon collateral branches. The regulation of the axonal cytoskeleton and mitochondria are important components of the mechanism of branching. Actin-dependent axonal plasticity, reflected in the dynamics of axonal actin patches and filopodia, is greatest along segments of the axon populated by mitochondria. It is reported that CSPGs partially depolarize the membrane potential of axonal mitochondria, which impairs the dynamics of the axonal actin cytoskeleton and decreases the formation and duration of axonal filopodia, the first steps in the mechanism of branching. The effects of CSPGs on actin cytoskeletal dynamics are specific to axon segments populated by mitochondria. In contrast, CSPGs do not affect the microtubule content of axons, or the localization of microtubules into axonal filopodia, a required step in the mechanism of branch formation. It is also reported that CSPGs decrease the mitochondria-dependent axonal translation of cortactin, an actin associated protein involved in branching. Finally, the inhibitory effects of CSPGs on axon branching, actin cytoskeletal dynamics and the axonal translation of cortactin are reversed by culturing neurons with acetyl-l-carnitine, which promotes mitochondrial respiration. Collectively these data indicate that CSPGs impair mitochondrial function in axons, an effect which contributes to the inhibition of axon branching. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 419-437, 2017.


Subject(s)
Actins/metabolism , Axons/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Cortactin/metabolism , Cytoskeleton/metabolism , Microtubules/metabolism , Mitochondria/metabolism , Pseudopodia/metabolism , Animals , Chick Embryo
7.
Dev Neurobiol ; 76(10): 1092-110, 2016 10.
Article in English | MEDLINE | ID: mdl-26731339

ABSTRACT

Drebrin is a cytoskeleton-associated protein which can interact with both actin filaments and the tips of microtubules. Its roles have been studied mostly in dendrites, and the functions of drebrin in axons are less well understood. In this study, we analyzed the role of drebrin, through shRNA-mediated depletion and overexpression, in the collateral branching of chicken embryonic sensory axons. We report that drebrin promotes the formation of axonal filopodia and collateral branches in vivo and in vitro. Live imaging of cytoskeletal dynamics revealed that drebrin promotes the formation of filopodia from precursor structures termed axonal actin patches. Endogenous drebrin localizes to actin patches and depletion studies indicate that drebrin contributes to the development of patches. In filopodia, endogenous drebrin localizes to the proximal portion of the filopodium. Drebrin was found to promote the stability of axonal filopodia and the entry of microtubule plus tips into axonal filopodia. The effects of drebrin on the stabilization of filopodia are independent of its effects on promoting microtubule targeting to filopodia. Inhibition of myosin II induces a redistribution of endogenous drebrin distally into filopodia, and further increases branching in drebrin overexpressing neurons. Finally, a 30 min treatment with the branch-inducing signal nerve growth factor increases the levels of axonal drebrin. This study determines the specific roles of drebrin in the regulation of the axonal cytoskeleton, and provides evidence that drebrin contributes to the coordination of the actin and microtubule cytoskeleton during the initial stages of axon branching. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 76: 1092-1110, 2016.


Subject(s)
Actins/metabolism , Axons/metabolism , Microtubules/metabolism , Neuropeptides/metabolism , Animals , Blotting, Western , Cells, Cultured , Chick Embryo , Electroporation , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Ganglia, Spinal/metabolism , Immunohistochemistry , Microscopy, Fluorescence , Nerve Growth Factor/metabolism , Pseudopodia/metabolism , Sensory Receptor Cells/metabolism
8.
Proc Natl Acad Sci U S A ; 113(3): 763-8, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26719418

ABSTRACT

Dual leucine-zipper kinase (DLK) is critical for axon-to-soma retrograde signaling following nerve injury. However, it is unknown how DLK, a predicted soluble kinase, conveys long-distance signals and why homologous kinases cannot compensate for loss of DLK. Here, we report that DLK, but not homologous kinases, is palmitoylated at a conserved site adjacent to its kinase domain. Using short-hairpin RNA knockdown/rescue, we find that palmitoylation is critical for DLK-dependent retrograde signaling in sensory axons. This functional importance is because of three novel cellular and molecular roles of palmitoylation, which targets DLK to trafficking vesicles, is required to assemble DLK signaling complexes and, unexpectedly, is essential for DLK's kinase activity. By simultaneously controlling DLK localization, interactions, and activity, palmitoylation ensures that only vesicle-bound DLK is active in neurons. These findings explain how DLK specifically mediates nerve injury responses and reveal a novel cellular mechanism that ensures the specificity of neuronal kinase signaling.


Subject(s)
Axons/metabolism , Axons/pathology , Caenorhabditis elegans Proteins/metabolism , Lipoylation , MAP Kinase Kinase Kinases/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/chemistry , Conserved Sequence , Evolution, Molecular , Fluorescent Dyes/metabolism , Gene Knockdown Techniques , HEK293 Cells , Humans , MAP Kinase Kinase Kinases/chemistry , Microfluidics , Models, Biological , Molecular Sequence Data , Mutation , Phosphorylation , Protein Binding , Protein Multimerization , Protein Transport , RNA, Small Interfering/metabolism , Rats , Sensory Receptor Cells/metabolism , Transfection , Transport Vesicles/metabolism
9.
Dev Neurobiol ; 75(12): 1441-61, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25846486

ABSTRACT

The localized debundling of the axonal microtubule array and the entry of microtubules into axonal filopodia are two defining features of collateral branching. We report that nerve growth factor (NGF), a branch-inducing signal, increases the frequency of microtubule debundling along the axon shaft of chicken embryonic sensory neurons. Sites of debundling correlate strongly with the localized targeting of microtubules into filopodia. Platinum replica electron microscopy suggests physical interactions between debundled microtubules and axonal actin filaments. However, as evidenced by depolymerization of actin filaments and inhibition of myosin II, actomyosin force generation does not promote debundling. In contrast, loss of actin filaments or inhibition of myosin II activity promotes debundling, indicating that axonal actomyosin forces suppress debundling. MAP1B is a microtubule associated protein that represses axon branching. Following treatment with NGF, microtubules penetrating filopodia during the early stages of branching exhibited lower levels of associated MAP1B. NGF increased and decreased the levels of MAP1B phosphorylated at a GSK-3ß site (pMAP1B) along the axon shaft and within axonal filopodia, respectively. The levels of MAP1B and pMAP1B were not altered at sites of debundling, relative to the rest of the axon. Unlike the previously determined effects of NGF on the axonal actin cytoskeleton, the effects of NGF on microtubule debundling were not affected by inhibition of protein synthesis. Collectively, these data indicate that NGF promotes localized axonal microtubule debundling, that actomyosin forces antagonize microtubule debundling, and that NGF regulates pMAP1B in axonal filopodia during the early stages of collateral branch formation.


Subject(s)
Avian Proteins/metabolism , Axons/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Nerve Growth Factor/metabolism , Pseudopodia/metabolism , Animals , Axons/ultrastructure , Chick Embryo , Fluorescent Antibody Technique , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Immunohistochemistry , Microscopy, Electron , Microtubules/ultrastructure , Oleanolic Acid/analogs & derivatives , Phosphorylation , Pseudopodia/ultrastructure , Saponins , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/ultrastructure , Transfection
10.
Cell Rep ; 5(6): 1564-75, 2013 Dec 26.
Article in English | MEDLINE | ID: mdl-24332852

ABSTRACT

The branching of axons is a fundamental aspect of nervous system development and neuroplasticity. We report that branching of sensory axons in the presence of nerve growth factor (NGF) occurs at sites populated by stalled mitochondria. Translational machinery targets to presumptive branching sites, followed by recruitment of mitochondria to these sites. The mitochondria promote branching through ATP generation and the determination of localized hot spots of active axonal mRNA translation, which contribute to actin-dependent aspects of branching. In contrast, mitochondria do not have a role in the regulation of the microtubule cytoskeleton during NGF-induced branching. Collectively, these observations indicate that sensory axons exhibit multiple potential sites of translation, defined by presence of translational machinery, but active translation occurs following the stalling and respiration of mitochondria at these potential sites of translation. This study reveals a local role for axonal mitochondria in the regulation of the actin cytoskeleton and axonal mRNA translation underlying branching.


Subject(s)
Axons/metabolism , Mitochondria/metabolism , Protein Biosynthesis , Sensory Receptor Cells/metabolism , Actin Cytoskeleton/metabolism , Adenosine Triphosphate/metabolism , Animals , Axons/physiology , Cell Growth Processes , Chickens , Microtubules/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sensory Receptor Cells/cytology , Sensory Receptor Cells/physiology
11.
J Neurosci ; 32(49): 17671-89, 2012 Dec 05.
Article in English | MEDLINE | ID: mdl-23223289

ABSTRACT

Nerve growth factor (NGF) induces collateral branching along sensory axons by promoting the formation of axonal filopodia dependent on the actin-nucleating Arp2/3 complex. This study shows that chicken embryonic sensory axons contain mRNAs for the actin-nucleating Arp2/3 complex activator WAVE1 and the complex stabilizer cortactin. NGF increases the axonal levels of WAVE1 and cortactin through localized protein synthesis even in axons isolated from the cell body. Inhibition of protein synthesis in severed axons impairs NGF-induced branching, the formation of axonal filopodia, and the initiation of Arp2/3-dependent axonal actin patches, which serve as precursors to the emergence of filopodia. Overexpression of WAVE1 or cortactin in axons not treated with NGF increased the rate of actin patch formation and the frequency of the emergence of filopodia from actin patches, respectively. Antisense inhibition of cortactin mRNA translation in isolated axons blocked NGF-induced filopodia. NGF also activated the Rac1 GTPase, which drives WAVE1 activity, in a protein synthesis-independent manner. Similarly, inhibition of protein synthesis did not impair the effects of NGF on the axonal microtubule cytoskeleton during branching. The effects of NGF on Rac1 activity and increases in axonal levels of WAVE1 and cortactin were both dependent on phosphoinositide 3-kinase (PI3K) signaling. Collectively, the data indicate that NGF promotes sensory axon branching through regulation of the actin cytoskeleton using both canonical signaling mechanisms and intra-axonal protein synthesis downstream of PI3K signaling. Finally, we present experimental evidence of axonal mRNA translation in sensory axons in the living embryonic spinal cord.


Subject(s)
Actin-Related Protein 2-3 Complex/metabolism , Axons/metabolism , Nerve Growth Factor/physiology , Pseudopodia/metabolism , Sensory Receptor Cells/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/physiology , Actin-Related Protein 2-3 Complex/genetics , Animals , Axons/drug effects , Axons/physiology , Cells, Cultured , Chick Embryo , Cortactin/metabolism , Growth Cones/drug effects , Growth Cones/metabolism , Microtubules/metabolism , Nerve Growth Factor/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Synthesis Inhibitors/pharmacology , Pseudopodia/drug effects , Sensory Receptor Cells/cytology , Sensory Receptor Cells/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Wiskott-Aldrich Syndrome Protein Family/metabolism , rac1 GTP-Binding Protein/biosynthesis
12.
J Neurosci Methods ; 199(2): 208-13, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21600922

ABSTRACT

The bacterial enzyme chondroitinase ABC (ChABC), which cleaves chondroitin sulfate glycosaminoglycan chains, can degrade inhibitory scar tissue formed following spinal cord injury, thereby promoting axonal growth and regeneration. However, delivering the active enzyme for prolonged periods presents practical limitations. To overcome these problems, we prepared a lentiviral vector (LV) encoding chondroitinase AC (Chase) together with the green fluorescent protein (GFP) reporter (Chase/LV) and demonstrated its expression and enzymatic activity in vitro and in vivo. Neural precursor cells infected with Chase/LV expressed the GFP reporter at levels that increased dramatically with time in culture. Enzymatic activity from the supernatant of the infected cells was demonstrated by dot blot assay using an antibody that recognizes the digested form of CSPG and was compared with the bacterial ChABC enzyme. Chick DRG cultures plated adjacent to the CSPG border and incubated with supernatant from Chase/LV-infected cells showed neurites growing into the CSPG area, a response similar to that after treatment with ChABC. In contrast, in control cultures, the neurites turned to avoid the inhibitory CSPG interface. Degradation of CSPG in these cultures was confirmed by specific CSPG antibodies. A single injection of Chase/LV into the spinal cord resulted in sustained secretion of the enzyme, whose activity was detected for 8 weeks by expression of GFP and evidence of the digested form of CSPG. This study demonstrates the efficacy of the Chase/LV vector and its potential as a therapeutic tool to reduce scar inhibition and promote axonal growth and repair following central nervous system injury.


Subject(s)
Chondroitin Lyases/genetics , Chondroitin Lyases/metabolism , Genetic Vectors/physiology , Lentivirus/genetics , Transduction, Genetic/methods , Animals , Axons/enzymology , Axons/physiology , Cells, Cultured , Chick Embryo , Female , Genetic Vectors/genetics , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Rats, Transgenic
13.
Dev Neurobiol ; 71(9): 747-58, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21557512

ABSTRACT

The emergence of axonal filopodia is the first step in the formation of axon collateral branches. In vitro, axonal filopodia emerge from precursor cytoskeletal structures termed actin patches. However, nothing is known about the cytoskeletal dynamics of the axon leading to the formation of filopodia in the relevant tissue environment. In this study we investigated the role of the actin nucleating Arp2/3 complex in the formation of sensory axon actin patches, filopodia, and branches. By combining in ovo chicken embryo electroporation mediated gene delivery with a novel acute ex vivo spinal cord preparation, we demonstrate that actin patches form along sensory axons and give rise to filopodia in situ. Inhibition of Arp2/3 complex function in vitro and in vivo decreases the number of axonal filopodia. In vitro, Arp2/3 complex subunits and upstream regulators localize to actin patches. Analysis of the organization of actin filaments in actin patches using platinum replica electron microscopy reveals that patches consist of networks of actin filaments, and filaments in axonal filopodia exhibit an organization consistent with the Arp2/3-based convergent elongation mechanism. Nerve growth factor (NGF) promotes formation of axonal filopodia and branches through phosphoinositide 3-kinase (PI3K). Inhibition of the Arp2/3 complex impairs NGF/PI3K-induced formation of axonal actin patches, filopodia, and the formation of collateral branches. Collectively, these data reveal that the Arp2/3 complex contributes to the formation of axon collateral branches through its involvement in the formation of actin patches leading to the emergence of axonal filopodia.


Subject(s)
Actin Cytoskeleton/metabolism , Actin-Related Protein 2/physiology , Actin-Related Protein 3/physiology , Axons/physiology , Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Growth Cones/physiology , Pseudopodia/metabolism , Actin Cytoskeleton/physiology , Actin-Related Protein 2/antagonists & inhibitors , Actin-Related Protein 3/antagonists & inhibitors , Animals , Chick Embryo , Chickens , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Primary Cell Culture , Pseudopodia/physiology , Sensory Receptor Cells/cytology , Sensory Receptor Cells/physiology
14.
Commun Integr Biol ; 4(1): 55-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21509179

ABSTRACT

The formation of axon collateral branches is a fundamental aspect of the development of neuronal circuits. Emergence of axonal filopodia from the axon is the first step in the formation of axon collateral branches and pre-synaptic structures. Using embryonic sensory axons as a model system, we have determined that axonal filopodia are formed from transient accumulations of F-actin within the axon, termed actin patches. We found that the branch-inducing factor NGF induces the formation of axonal actin patches and filopodia. NGF signaling, through PI3K, promotes the formation of localized axonal microdomains of PIP3 accumulation. The microdomains in turn drive formation of actin patches. Under basal conditions, only a subset of actin patches gives rise to filopodia, and many patches dissipate without forming a filopodium. Neither NGF nor direct activation of PI3K affects the probability that an actin patch will give rise to a filopodium. Thus, NGF increases formation of axonal filopodia through localized PI3K signaling that promotes the initiation of actin patch precursors to the formation of axonal filopodia. The promotion of actin patch formation by NGF may be mediated through a PI3K-TOR pathway driving intra-axonal protein synthesis. We propose the hypothesis that NGF signaling "turns up the volume" on the mechanism of filopodial formation by increasing axonal levels of the cytoskeletal proteins required for the orchestration of actin patch formation by PIP3 microdomains.

15.
J Neurosci ; 30(36): 12185-97, 2010 Sep 08.
Article in English | MEDLINE | ID: mdl-20826681

ABSTRACT

The initiation of axonal filopodia is the first step in the formation of collateral branches and synaptic structures. In sensory neurons, nerve growth factor (NGF) promotes the formation of axonal filopodia and branches. However, the signaling and cytoskeletal mechanisms of NGF-induced initiation of axonal filopodia are not clear. Axonal filopodia arise from precursor axonal cytoskeletal structures termed filamentous actin (F-actin) patches. Patches form spontaneously and are transient. Although filopodia emerge from patches, only a fraction of patches normally gives rise to filopodia. Using chicken sensory neurons and live imaging of enhanced yellow fluorescent protein (eYFP)-actin dynamics, we report that NGF promotes the formation of axonal filopodia by increasing the rate of F-actin patch formation but not the fraction of patches that give rise to filopodia. We also demonstrate that activation of the phosphatidylinositol 3-kinase (PI3K)-Akt pathway is sufficient and required for driving the formation of axonal F-actin patches, filopodia, and axon branches. Using the green fluorescent protein-plekstrin homology domain of Akt, which targets to PI3K-generated phosphatidylinositol-3,4,5-triphosphate (PIP(3)), we report localized microdomains of PIP(3) accumulation that form in synchrony with F-actin patches and that NGF promotes the formation of microdomains of PIP(3) and patches. Finally, we find that, in NGF, F-actin patches form in association with axonal mitochondria and oxidative phosphorylation is required for patch formation. This investigation demonstrates that surprisingly NGF promotes formation of axonal filopodia by increasing the formation of cytoskeletal filopodial precursors (patches) through localized microdomains of PI3K signaling but not the emergence of filopodia from patches.


Subject(s)
Axons/drug effects , Cytoskeleton/metabolism , Nerve Growth Factor/pharmacology , Neurons/cytology , Phosphatidylinositol 3-Kinases/metabolism , Pseudopodia/drug effects , Actins/genetics , Actins/metabolism , Animals , Antibodies/pharmacology , Axons/metabolism , Axons/ultrastructure , Cells, Cultured , Chick Embryo , Ganglia, Spinal/cytology , Gene Expression Regulation/drug effects , Integrin beta1/immunology , Luminescent Proteins/genetics , Microscopy, Confocal/methods , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/drug effects , Neurons/physiology , Peptides/pharmacology , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 4,5-Diphosphate/analogs & derivatives , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/physiology , Protein Transport/drug effects , Protein Transport/genetics , Pseudopodia/metabolism , Receptor, trkA/metabolism , Signal Transduction/drug effects , Time Factors , Transfection/methods
16.
Curr Biol ; 18(24): 1972-7, 2008 Dec 23.
Article in English | MEDLINE | ID: mdl-19084405

ABSTRACT

Inhibition of kinesin-5, a mitotic motor protein also expressed in neurons, causes axons to grow faster as a result of alterations in the forces on microtubules (MTs) in the axonal shaft. Here, we investigate whether kinesin-5 plays a role in growth-cone guidance. Growth-cone turning requires that MTs in the central (C-) domain enter the peripheral (P-) domain in the direction of the turn. We found that inhibition of kinesin-5 in cultured neurons prevents MTs from polarizing within growth cones and causes them to grow past cues that would normally cause them to turn. We found that kinesin-5 is enriched in the transition (T-) zone of the growth cone and that kinesin-5 is preferentially phosphorylated on the side opposite the invasion of MTs. Moreover, when a growth cone encounters a turning cue, phospho-kinesin-5 polarizes even before the growth cone turns. Additional studies indicate that kinesin-5 works in part by antagonizing cytoplasmic dynein and that these motor-driven forces function together with the dynamic properties of the MTs to determine whether MTs can enter the P-domain. We propose that kinesin-5 permits MTs to selectively invade one side of the growth cone by opposing their entry into the other side.


Subject(s)
Growth Cones/physiology , Kinesins/physiology , Animals , Cells, Cultured , Dyneins/physiology , Ganglia, Sympathetic/drug effects , Ganglia, Sympathetic/growth & development , Ganglia, Sympathetic/physiology , Ganglia, Sympathetic/ultrastructure , Growth Cones/drug effects , Growth Cones/ultrastructure , In Vitro Techniques , Kinesins/antagonists & inhibitors , Kinesins/genetics , Microtubules/drug effects , Microtubules/physiology , Movement/drug effects , Movement/physiology , Nerve Growth Factor/pharmacology , Phosphorylation , Pyrimidines/pharmacology , RNA, Small Interfering/genetics , Rats , Thiones/pharmacology
17.
Dev Neurobiol ; 67(10): 1305-20, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17638383

ABSTRACT

Axon extension involves the coordinated regulation of the neuronal cytoskeleton. Actin filaments drive protrusion of filopodia and lamellipodia while microtubules invade the growth cone, thereby providing structural support for the nascent axon. Furthermore, in order for axons to extend the growth cone must attach to the substratum. Previous work indicates that myosin II activity inhibits the advance of microtubules into the periphery of growth cones, and myosin II has also been implicated in mediating integrin-dependent cell attachment. However, it is not clear how the functions of myosin II in regulating substratum attachment and microtubule advance are integrated during axon extension. We report that inhibition of myosin II function decreases the rate of axon extension on laminin, but surprisingly promotes extension rate on polylysine. The differential effects of myosin II inhibition on axon extension rate are attributable to myosin II having the primary function of mediating substratum attachment on laminin, but not on polylysine. Conversely, on polylysine the primary function of myosin II is to inhibit microtubule advance into growth cones. Thus, the substratum determines the role of myosin II in axon extension by controlling the functions of myosin II that contribute to extension.


Subject(s)
Cell Differentiation/physiology , Central Nervous System/embryology , Central Nervous System/metabolism , Growth Cones/metabolism , Myosin Type II/metabolism , Animals , Cell Adhesion/physiology , Cell Communication/physiology , Cell Differentiation/drug effects , Chick Embryo , Extracellular Matrix/metabolism , Growth Cones/drug effects , Laminin/metabolism , Laminin/pharmacology , Microtubules/metabolism , Polylysine/metabolism , Polylysine/pharmacology , Time Factors
18.
J Parasitol ; 93(5): 1084-9, 2007 Oct.
Article in English | MEDLINE | ID: mdl-18163342

ABSTRACT

Parastrongyloides trichosuri is a nematode parasite of the Australian brush-tailed possums that can be propagated through many generations in vitro. This makes P. trichosuri uniquely suited for genetic investigations, including those involving transgenesis. However, an obstacle to its use as an experimental model has been the fact that its host is limited to Australia and New Zealand and that it cannot be exported because of its status as a protected species or agricultural pest, respectively. In previous studies, conventional laboratory animals such as rats, mice, rabbits, ferrets, and chickens have failed to support infections. In the present study, gerbils and short-tailed opossums proved similarly refractory to infection. In contrast, the sugar glider (Petaurus breviceps, family Petauridae) proved to be a good host for P. trichosuri. Patent infections resulted using as few as 6 infective larvae (L3i) and as many as 2,000 L3i. Large numbers of L3i (1,000-2,000) produced patent infections of much shorter duration than those seen when 100 L3i were initially given to the sugar glider. In one case, an infection initiated with 100 L3i was patent for over 1 yr. Parastrongyloides trichosuri is easily cryopreserved using a method developed for Strongyloides stercoralis. Thus, we have identified an experimental host for P. trichosuri that will make it possible to conduct research on this parasite in laboratories outside the endemic sites.


Subject(s)
Disease Models, Animal , Marsupialia/parasitology , Rhabditida Infections/parasitology , Rhabditida/pathogenicity , Animals , Cryopreservation , Feces/parasitology , Gerbillinae/parasitology , Male , Opossums/parasitology , Parasite Egg Count , Rhabditida/physiology
19.
Brain Res ; 965(1-2): 91-9, 2003 Mar 07.
Article in English | MEDLINE | ID: mdl-12591124

ABSTRACT

Rats use their vibrissae for a variety of exploratory tasks including location of objects and discrimination of texture. This study examines recovery in vibrissal function following a unilateral ischemic injury to the somatosensory cortex. Vibrissal function was examined in adult food-restricted rats performing on a two-texture discrimination device. Animals were trained and tested until the criteria of >80% correct choices was demonstrated on three consecutive days. Ischemic rats were constrained to use the affected whiskers by clipping the ipsilateral vibrissae. One group was tested after ischemia, a second group was trained before ischemia and then tested, and a third group was pre-trained and received whisker stimulation and tested post-ischemia. Nai;ve animals recovering from ischemia took longer to reach criteria than intact or unilateral trimmed control animals. Pre-trained animals with compression ischemia receiving whisker stimulation with sucrose water completed the task to criteria in the fewest number of trials. The results indicate that recovery of vibrissal function occurs following a unilateral ischemic injury. Histological analysis in animals without whisker stimulation indicates that the number of normal appearing cortical barrels following ischemia was inversely correlated to the number of trials needed to complete the behavioral task. This suggests that the natural recovery of the ability to discriminate textures is related to the degree of damage to the barrel cortex. The relationship between cortical barrels and behavioral recovery did not hold for the ischemic animals receiving whisker stimulation. This latter group demonstrated recovery despite marked anatomical lesions suggesting that the intervention influenced reorganization.


Subject(s)
Brain Ischemia/physiopathology , Cerebral Cortex/physiology , Exploratory Behavior/physiology , Vibrissae/physiology , Animals , Male , Physical Stimulation/methods , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...