Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
eNeuro ; 11(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38937107

ABSTRACT

γ-Aminobutyric acid (GABA) is the principal inhibitory neurotransmitter in the adult brain which mediates its rapid effects on neuronal excitability via ionotropic GABAA receptors. GABA levels in the brain are critically dependent upon GABA-aminotransferase (GABA-AT) which promotes its degradation. Vigabatrin, a low-affinity GABA-AT inhibitor, exhibits anticonvulsant efficacy, but its use is limited due to cumulative ocular toxicity. OV329 is a rationally designed, next-generation GABA-AT inhibitor with enhanced potency. We demonstrate that sustained exposure to OV329 in mice reduces GABA-AT activity and subsequently elevates GABA levels in the brain. Parallel increases in the efficacy of GABAergic inhibition were evident, together with elevations in electroencephalographic delta power. Consistent with this, OV329 exposure reduced the severity of status epilepticus and the development of benzodiazepine refractory seizures. Thus, OV329 may be of utility in treating seizure disorders and associated pathologies that result from neuronal hyperexcitability.


Subject(s)
4-Aminobutyrate Transaminase , Anticonvulsants , Benzodiazepines , Seizures , gamma-Aminobutyric Acid , Animals , Anticonvulsants/pharmacology , Anticonvulsants/administration & dosage , Male , Benzodiazepines/pharmacology , 4-Aminobutyrate Transaminase/antagonists & inhibitors , 4-Aminobutyrate Transaminase/metabolism , Seizures/drug therapy , Seizures/metabolism , gamma-Aminobutyric Acid/metabolism , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Brain/drug effects , Brain/metabolism , Neural Inhibition/drug effects , Neural Inhibition/physiology , Mice , Electroencephalography , Disease Models, Animal , Status Epilepticus/drug therapy , Status Epilepticus/chemically induced , Status Epilepticus/metabolism , Female
2.
Mol Aspects Med ; 91: 101148, 2023 06.
Article in English | MEDLINE | ID: mdl-36257857

ABSTRACT

Advances in genome sequencing have greatly facilitated the identification of genomic variants underlying rare neurodevelopmental and neurodegenerative disorders. Understanding the fundamental causes of rare monogenic disorders has made gene therapy a possible treatment approach for these conditions. RNA interference (RNAi) technologies such as small interfering RNA (siRNA), microRNA (miRNA), and short hairpin RNA (shRNA), and other oligonucleotide-based modalities such as antisense oligonucleotides (ASOs) are being developed as potential therapeutic approaches for manipulating expression of the genes that cause a variety of neurological diseases. Here, we offer a brief review of the mechanism of action of these RNAi approaches; provide deeper discussion of the advantages, challenges, and specific considerations related to the development of RNAi therapeutics for neurological disease; and highlight examples of rare neurological diseases for which RNAi therapeutics hold great promise.


Subject(s)
MicroRNAs , Humans , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/therapeutic use , Genetic Therapy
3.
Neurochem Res ; 44(6): 1460-1474, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30569280

ABSTRACT

Remyelination promoting human IgMs effectively increase the number of myelinated axons in animal models of multiple sclerosis. Hence, they ultimately stimulate myelin production by oligodendrocytes (OLs); however, their exact mechanism of action remains to be elucidated, and in particular, it remains unclear whether they are directly targeting OLs, or their action is mediated by effects on other cell types. We assessed the effect of remyelination promoting antibody rHIgM22 on the proliferative response and on the ceramide/sphingosine 1-phosphate rheostat in mixed glial cell cultures (MGCs). rHIgM22 treatment caused a time-dependent increase in PDGFαR protein in MGCs. Forty-eight hours of treatment with rHIgM22 induced a dose-dependent proliferative response (evaluated as total cell number and as EdU(+) cell number) in MGCs. When the proliferation response of MGCs to rHIgM22 was analyzed as a function of the cell types, the most significant proliferative response was associated with GLAST(+) cells, i.e., astrocytes. In many cell types, the balance between different sphingolipid mediators (the "sphingolipid rheostat"), in particular ceramide and sphingosine 1-phosphate, is critical in determining the cell fate. rHIgM22 treatment in MGCs induced a moderate but significant inhibition of total acidic sphingomyelinase activity (measured in vitro on cell lysates), the main enzyme responsible for the stimulus-mediated production of ceramide, when treatment was performed in serum containing medium, but no significant differences were observed when antibody treatment was performed in the absence of serum. Moreover, rHIgM22 treatment, either in the presence or in absence of serum, had no effects on ceramide levels. On the other hand, rHIgM22 treatment for 24 h induced increased production and release of sphingosine 1-phosphate in the extracellular milieu of MGC. Release of sphingosine 1-phosphate upon rHIgM22 treatment was strongly reduced by a selective inhibitor of PDGFαR. Increased sphingosine 1-phosphate production does not seem to be mediated by regulation of the biosynthetic enzymes, sphingosine kinase 1 and 2, since protein levels of these enzymes and phosphorylation of sphingosine kinase 1 were unchanged upon rHIgM22 treatment. Instead, we observed a significant reduction in the levels of sphingosine 1-phosphate lyase 1, one of the key catabolic enzymes. Remarkably, rHIgM22 treatment under the same experimental conditions did not induce changes in the production and/or release of sphingosine 1-phosphate in pure astrocyte cultures. Taken together, these data suggest that rHIgM22 indirectly influences the proliferation of astrocytes in MGCs, by affecting the ceramide/sphingosine 1-phosphate balance. The specific cell population directly targeted by rHIgM22 remains to be identified, however our study unveils another aspect of the complexity of rHIgM22-induced remyelinating effect.


Subject(s)
Astrocytes/metabolism , Cell Proliferation/physiology , Immunoglobulin M/immunology , Myelin Sheath/metabolism , Remyelination/drug effects , Sphingolipids/metabolism , Animals , Ceramides/metabolism , Humans , Lysophospholipids/metabolism , Rats, Sprague-Dawley , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Recombinant Proteins/immunology , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Up-Regulation/drug effects
4.
J Neurotrauma ; 33(24): 2202-2216, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27125815

ABSTRACT

A porcine model of spinal cord injury (SCI) was used to evaluate the neuroprotective effects of magnesium chloride (MgCl2) within a polyethylene glycol (PEG) formulation, called "AC105" (Acorda Therapeutics Inc., Ardsley, NY). Specifically, we tested the hypothesis that AC105 would lead to greater tissue sparing at the injury site and improved behavioral outcome when delivered in a clinically realistic time window post-injury. Four hours after contusion/compression injury, Yucatan minipigs were randomized to receive a 30-min intravenous infusion of AC105, magnesium sulfate (MgSO4), or saline. Animals received 4 additional infusions of the same dose at 6-h intervals. Behavioral recovery was tested for 12 weeks using two-dimensional (2D) kinematics during weight-supported treadmill walking and the Porcine Injury Behavior Scale (PTIBS), a 10-point locomotion scale. Spinal cords were evaluated ex vivo by diffusion-weighted magnetic resonance imaging (MRI) and subjected to histological analysis. Treatment with AC105 or MgSO4 did not result in improvements in locomotor recovery on the PTIBS or in 2D kinematics on weight-supported treadmill walking. Diffusion weighted imaging (DWI) showed severe loss of tissue integrity at the impact site, with decreased fractional anisotropy and increased mean diffusivity; this was not improved with AC105 or MgSO4 treatment. Histological analysis revealed no significant increase in gray or white matter sparing with AC105 or MgSO4 treatment. Finally, AC105 did not result in higher Mg2+ levels in CSF than with the use of standard MgSO4. In summary, when testing AC105 in a porcine model of SCI, we were unable to reproduce the promising therapeutic benefits observed previously in less-severe rodent models of SCI.


Subject(s)
Disease Models, Animal , Magnesium Chloride/administration & dosage , Polyethylene Glycols/administration & dosage , Spinal Cord Injuries/diagnostic imaging , Spinal Cord Injuries/prevention & control , Acute Disease , Animals , Drug Compounding , Drug Evaluation, Preclinical/methods , Female , Locomotion/drug effects , Locomotion/physiology , Magnesium Chloride/chemistry , Polyethylene Glycols/chemistry , Random Allocation , Recovery of Function/drug effects , Recovery of Function/physiology , Spinal Cord Injuries/physiopathology , Swine , Swine, Miniature , Thoracic Vertebrae
5.
BMC Neurosci ; 9: 112, 2008 Nov 13.
Article in English | MEDLINE | ID: mdl-19014551

ABSTRACT

BACKGROUND: The Kv2.1 delayed-rectifier K+ channel regulates membrane excitability in hippocampal neurons where it targets to dynamic cell surface clusters on the soma and proximal dendrites. In the past, Kv2.1 has been assumed to be absent from the axon initial segment. RESULTS: Transfected and endogenous Kv2.1 is now demonstrated to preferentially accumulate within the axon initial segment (AIS) over other neurite processes; 87% of 14 DIV hippocampal neurons show endogenous channel concentrated at the AIS relative to the soma and proximal dendrites. In contrast to the localization observed in pyramidal cells, GAD positive inhibitory neurons within the hippocampal cultures did not show AIS targeting. Photoactivable-GFP-Kv2.1-containing clusters at the AIS were stable, moving <1 microm/hr with no channel turnover. Photobleach studies indicated individual channels within the cluster perimeter were highly mobile (FRAP tau=10.4+/-4.8 sec), supporting our model that Kv2.1 clusters are formed by the retention of mobile channels behind a diffusion-limiting perimeter. Demonstrating that the AIS targeting is not a tissue culture artifact, Kv2.1 was found in axon initial segments within both the adult rat hippocampal CA1, CA2, and CA3 layers and cortex. CONCLUSION: In summary, Kv2.1 is associated with the axon initial segment both in vitro and in vivo where it may modulate action potential frequency and back propagation. Since transfected Kv2.1 initially localizes to the AIS before appearing on the soma, it is likely multiple mechanisms regulate Kv2.1 trafficking to the cell surface.


Subject(s)
Axons/metabolism , Cerebral Cortex/metabolism , Hippocampus/metabolism , Neurons/metabolism , Shab Potassium Channels/metabolism , Animals , Cells, Cultured , Cerebral Cortex/cytology , Fluorescence Recovery After Photobleaching , Hippocampus/cytology , Neurites/metabolism , Neurons/ultrastructure , Protein Transport , Pyramidal Cells/metabolism , Pyramidal Cells/ultrastructure , Rats , Shab Potassium Channels/genetics , Transfection
6.
Exp Neurol ; 214(1): 78-86, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18722369

ABSTRACT

Statins elicit numerous favorable effects on central nervous system (CNS) injury, including inhibition of the rhoA/ROCK pathway. In the present study, we show that statins decrease acute astrocyte activation in CNS injury, and decrease chondroitin sulfate proteoglycan (CSPG) levels in astrocyte cultures as well as CNS injury. CSPG levels decreased by up to 45% in simvastatin-treated astrocyte cultures compared to control cultures. In simvastatin-treated animals, CSPG levels declined by 60% 8 days after brain stab injury, and by 62-64% 4 weeks after spinal cord injury (SCI). Glial fibrillary acid protein (GFAP) levels decreased in brain stab at 8 days after surgery/intervention, suggesting that statins produce a decrease in astrocyte activation. Attenuation of astrocyte activation may contribute to the decline in CSPG levels. However, there are likely other contributing factors, since GFAP levels were not a contributing factor in the decline of CSPG levels in astrocyte cultures. Robust locomotor improvements were not observed with any treatment. The numerous beneficial effects of statins on CNS injury render them an attractive candidate in the treatment of CNS injury.


Subject(s)
Astrocytes/drug effects , Brain Injuries/metabolism , Cerebral Cortex/drug effects , Chondroitin Sulfate Proteoglycans/metabolism , Simvastatin/pharmacology , Spinal Cord Injuries/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Blotting, Western , Cells, Cultured , Cerebral Cortex/metabolism , Glial Fibrillary Acidic Protein/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Immunohistochemistry , Male , Motor Activity/drug effects , Nerve Regeneration/drug effects , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Reverse Transcriptase Polymerase Chain Reaction
7.
J Neurotrauma ; 24(2): 411-20, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17376003

ABSTRACT

Glial scar represents a physical and molecular barrier to axonal regeneration and has become an important target for regeneration research in chronic spinal cord injury. Although many methods have been proven useful for the prevention of scar formation in an acute injury model, to date no effective method has been described to remove an existing glial scar in a chronic injury. The chronic lesion possesses an irregular shaped scar that lines the entire perimeter of the cavity. In the present study, we used rose bengal, a molecule commonly used for biological staining, injected into the cavity at the injury site of Long-Evans rat spinal cord (5 weeks after 25-mm contusion injury). Visible light was used to illuminate the injury site. Histological observation illustrates that at least partial glial scar tissue is ablated by rose bengal/illumination. The lack of glial fibrillary acidic protein (GFAP) immunoreactivity at the glial scar coupled with the reduction of GFAP density surrounding spared tissue suggests that this photochemical scar ablation preferentially kills astrocytes at the scar tissue but also reacts, to a lesser degree, in the spared tissue. There is an observed reduction of Basso, Beattie, and Bresnahan (BBB) scale scores after scar ablation, but it is not statistically significant from stabilized behavioral scoring prior to the scar ablation treatment. Our findings indicate that the rose bengal/illumination is feasible for ablation of the glial scar which surrounds an irregular lesion cavity in shape. The scar ablation might provide a permissive environment for the regenerating axons when enriched by cellular or drug therapy.


Subject(s)
Cicatrix/drug therapy , Fluorescent Dyes/therapeutic use , Neuroglia/physiology , Photochemotherapy/methods , Rose Bengal/therapeutic use , Spinal Cord Injuries/complications , Animals , Chronic Disease , Cicatrix/etiology , Cicatrix/metabolism , Female , Glial Fibrillary Acidic Protein/metabolism , Rats , Rats, Long-Evans , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Thoracic Vertebrae
8.
EMBO J ; 24(3): 473-86, 2005 Feb 09.
Article in English | MEDLINE | ID: mdl-15660133

ABSTRACT

Slingshot (SSH) phosphatases and LIM kinases (LIMK) regulate actin dynamics via a reversible phosphorylation (inactivation) of serine 3 in actin-depolymerizing factor (ADF) and cofilin. Here we demonstrate that a multi-protein complex consisting of SSH-1L, LIMK1, actin, and the scaffolding protein, 14-3-3zeta, is involved, along with the kinase, PAK4, in the regulation of ADF/cofilin activity. Endogenous LIMK1 and SSH-1L interact in vitro and co-localize in vivo, and this interaction results in dephosphorylation and downregulation of LIMK1 activity. We also show that the phosphatase activity of purified SSH-1L is F-actin dependent and is negatively regulated via phosphorylation by PAK4. 14-3-3zeta binds to phosphorylated slingshot, decreases the amount of slingshot that co-sediments with F-actin, but does not alter slingshot activity. Here we define a novel ADF/cofilin phosphoregulatory complex and suggest a new mechanism for the regulation of ADF/cofilin activity in mediating changes to the actin cytoskeleton.


Subject(s)
Microfilament Proteins/chemistry , Microfilament Proteins/metabolism , Phosphoprotein Phosphatases/chemistry , Phosphoprotein Phosphatases/metabolism , Protein Kinases/chemistry , Protein Kinases/metabolism , 14-3-3 Proteins/chemistry , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , Actin Depolymerizing Factors , Actins/metabolism , Animals , Binding Sites , Cell Line , Chickens , Destrin , Humans , In Vitro Techniques , Lim Kinases , Microfilament Proteins/genetics , Models, Biological , Multiprotein Complexes , Neurons/metabolism , Phosphoprotein Phosphatases/genetics , Protein Kinases/genetics , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , p21-Activated Kinases
9.
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
10.
Cell Microbiol ; 6(5): 459-71, 2004 May.
Article in English | MEDLINE | ID: mdl-15056216

ABSTRACT

Entry of Salmonella into mammalian cells is strictly dependent on the reorganization of actin cytoskeleton induced by a panel of Salmonella type III secreted proteins. Although several factors have been identified to be responsible for inducing the actin polymerization and stability, little is known about how the actin depolymerization contributes to Salmonella-induced actin rearrangements. We report here that activity cycles of host actin depolymerizing factor (ADF and cofilin) are modulated by Salmonella during bacterial entry. Efficient Salmonella internalization involves an initial dephosphorylation of ADF and cofilin followed by phosphorylation, suggesting that ADF and cofilin activities are increased briefly. Expression of a kinase dead form of an ADF/cofilin kinase (LIM kinase 1) or a catalytically inactive ADF/cofilin phosphatase (Slingshot), but not constitutively active LIM kinase 1 or wild-type Slingshot, resulted in decreased invasion. These data suggest that ADF/cofilin activities play a key role in the actin polymerization/depolymerization process induced by Salmonella. The activation of ADF/cofilin is brief and has to be reversed to facilitate efficient bacterial entry. Surprisingly, co-expression of constitutive active ADF and cofilin prevented efficient Salmonella entry, whereas expression of either one alone had no effect. We propose that ADF and cofilin actin-dynamizing activities and their activity cycling via phosphorylation are required for efficient Salmonella internalization.


Subject(s)
Endocytosis/physiology , Microfilament Proteins/metabolism , Salmonella Infections/metabolism , Salmonella/metabolism , Actin Depolymerizing Factors , Actins/metabolism , Animals , Bacterial Proteins/metabolism , Cell Surface Extensions/metabolism , Cytoskeleton/metabolism , Destrin , HeLa Cells , Humans , Lim Kinases , Microfilament Proteins/genetics , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Kinases/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
11.
Exp Cell Res ; 294(2): 392-405, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15023529

ABSTRACT

The expression of endogenous LIM kinase 1 (LIMK1) protein was investigated in embryonic and adult mice using a rat monoclonal antibody (mAb), which recognizes specifically the PDZ domain of LIMK1 and not LIMK2. Immunoblotting analysis revealed widespread expression of LIMK1 existing as a 70-kDa protein in tissues and in cell lines, with a higher mass form (approximately 75 kDa) present in some tissues and cell lines. Smaller isoforms of approximately 50 kDa were also occasionally evident. Immunofluorescence analysis demonstrated LIMK1 subcellular localization at focal adhesions in fibroblasts as revealed by co-staining with actin, paxillin and vinculin in addition to perinuclear (Golgi) and occasional nuclear localization. Furthermore, an association between LIMK1 and paxillin but not vinculin was identified by co-immunoprecipitation analysis. LIMK1 is enriched in both axonal and dendritic growth cones of E18 rat hippocampal pyramidal neurons where it is found in punctae that extend far out into filopodia, as well as in a perinuclear region identified as Golgi. In situ, we identify LIMK1 protein expression in all embryonic and adult tissues examined, albeit at different levels and in different cell populations. The rat monoclonal LIMK1 antibody recognizes proteins of similar size in cell and tissue extracts from numerous species. Thus, LIMK1 is a widely expressed protein that exists as several isoforms.


Subject(s)
Actins/metabolism , Cell Differentiation/physiology , DNA-Binding Proteins/metabolism , Embryo, Mammalian/enzymology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Antibodies, Monoclonal , COS Cells , Chick Embryo , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , DNA-Binding Proteins/genetics , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Focal Adhesions/metabolism , Golgi Apparatus/metabolism , Humans , Lim Kinases , Mice , Mice, Inbred C57BL , Molecular Weight , NIH 3T3 Cells , Neurites/metabolism , Organ Specificity , Paxillin , Phosphoproteins/metabolism , Protein Isoforms/metabolism , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary/physiology , Pseudopodia/metabolism , Rats
12.
Anat Sci Int ; 79(4): 173-83, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15633455

ABSTRACT

The regulation of filopodial dynamics by neurotrophins and other guidance cues plays an integral role in growth cone pathfinding. Filopodia are F-actin-based structures that explore the local environment, generate forces and play a role in growth cone translocation. Here, we review recent research showing that the actin-depolymerizing factor (ADF)/cofilin family of proteins mediates changes in the length and number of growth cone filopodia in response to brain-derived neurotrophic factor (BDNF). Although inhibition of myosin contractility also causes filopodial elongation, the elongation in response to BDNF does not occur through a myosin-dependent pathway. Active ADF/cofilin increases the rate of cycling between the monomer and polymer pools and is critical for the BDNF-induced changes. Thus, we discuss potential mechanisms by which ADF/cofilin may affect filopodial initiation and length change via its effects on F-actin dynamics in light of past research on actin and myosin function in growth cones.


Subject(s)
Growth Cones/metabolism , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Actin Depolymerizing Factors , Animals , Cell Line, Tumor , Destrin , Growth Cones/ultrastructure , Humans , Mice , Microfilament Proteins/ultrastructure , Nerve Tissue Proteins/ultrastructure
13.
J Neurobiol ; 58(1): 103-17, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14598374

ABSTRACT

Actin and microtubules are major cytoskeletal elements of most cells including neurons. In order for a cell to move and change shape, its cytoskeleton must undergo rearrangements that involve breaking down and reforming filaments. Many recent reviews have focused on the signaling pathways emanating from receptors that ultimately affect axon growth and growth cone steering. This particular review will address changes in the actin cytoskeleton modulated by the family of actin dynamizing proteins known as actin depolymerizing factor (ADF)/cofilin or AC proteins. Though much is known about inactivation of AC proteins through phosphorylation at ser3 by LIM or TES kinases, new mechanisms of regulation of AC have recently emerged. A novel phosphatase, slingshot (SSH), and the 14-3-3 family of regulatory proteins have also been found to affect AC activity. The potential role of AC proteins in modulating the actin organizational changes that accompany neurite initiation, axonogenesis, growth cone guidance, and dendritic spine formation will be discussed.


Subject(s)
Actins/metabolism , Cytoskeleton/physiology , Microfilament Proteins/metabolism , Neurons/physiology , Actin Depolymerizing Factors , Animals , Destrin
14.
Neuron ; 35(1): 3-5, 2002 Jul 03.
Article in English | MEDLINE | ID: mdl-12123600

ABSTRACT

LIM kinase 1 regulates actin filament dynamics through inhibition of ADF/cofilins. Surprisingly, nervous system development in LIM kinase 1 knockout mice is grossly normal, but the animals have deficits in spatial learning, alterations in LTP, and abnormalities in hippocampal dendritic spine structure. The findings are consistent with a role for LIMK-1 in synapse formation and function.


Subject(s)
Actin Cytoskeleton/metabolism , DNA-Binding Proteins/deficiency , Dendrites/metabolism , Hippocampus/abnormalities , Protein Serine-Threonine Kinases/deficiency , Synapses/metabolism , Animals , DNA-Binding Proteins/genetics , Dendrites/pathology , Hippocampus/pathology , Hippocampus/physiopathology , Learning Disabilities/genetics , Learning Disabilities/metabolism , Learning Disabilities/pathology , Lim Kinases , Long-Term Potentiation/physiology , Mice , Protein Kinases , Protein Serine-Threonine Kinases/genetics , Synapses/pathology , Synaptic Transmission/physiology
SELECTION OF CITATIONS
SEARCH DETAIL