Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Mol Biol Cell ; 35(2): ar23, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38088877

ABSTRACT

Myristoylated, alanine-rich C-kinase substrate (MARCKS) is an F-actin and phospholipid binding protein implicated in numerous cellular activities, including the regulation of morphology in neuronal dendrites and dendritic spines. MARCKS contains a lysine-rich effector domain that mediates its binding to plasma membrane phosphatidylinositol-4,5-biphosphate (PI(4,5)P2) in a manner controlled by PKC and calcium/calmodulin. In neurons, manipulations of MARCKS concentration and membrane targeting strongly affect the numbers, shapes, and F-actin properties of dendritic spines, but the mechanisms remain unclear. Here, we tested the hypothesis that the effects of MARCKS on dendritic spine morphology are due to its capacity to regulate the availability of plasma membrane PI(4,5)P2. We observed that the concentration of free PI(4,5)P2 on the dendritic plasma membrane was inversely proportional to the concentration of MARCKS. Endogenous PI(4,5)P2 levels were increased or decreased, respectively, by acutely overexpressing either phosphatidylinositol-4-phosphate 5-kinase (PIP5K) or inositol polyphosphate 5-phosphatase (5ptase). PIP5K, like MARCKS depletion, induced severe spine shrinkage; 5ptase, like constitutively membrane-bound MARCKS, induced aberrant spine elongation. These phenotypes involved changes in actin properties driven by the F-actin severing protein cofilin. Collectively, these findings support a model in which neuronal activity regulates actin-dependent spine morphology through antagonistic interactions of MARCKS and PI(4,5)P2.


Subject(s)
Actins , Membrane Proteins , Actins/metabolism , Membrane Proteins/metabolism , Dendritic Spines/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Kinase C/metabolism , Phosphorylation
2.
Nat Commun ; 13(1): 6037, 2022 10 13.
Article in English | MEDLINE | ID: mdl-36229429

ABSTRACT

During early ischemic brain injury, glutamate receptor hyperactivation mediates neuronal death via osmotic cell swelling. Here we show that ischemia and excess NMDA receptor activation cause actin to rapidly and extensively reorganize within the somatodendritic compartment. Normally, F-actin is concentrated within dendritic spines. However, <5 min after bath-applied NMDA, F-actin depolymerizes within spines and polymerizes into stable filaments within the dendrite shaft and soma. A similar actinification occurs after experimental ischemia in culture, and photothrombotic stroke in mouse. Following transient NMDA incubation, actinification spontaneously reverses. Na+, Cl-, water, and Ca2+ influx, and spine F-actin depolymerization are all necessary, but not individually sufficient, for actinification, but combined they induce activation of the F-actin polymerization factor inverted formin-2 (INF2). Silencing of INF2 renders neurons vulnerable to cell death and INF2 overexpression is protective. Ischemia-induced dendritic actin reorganization is therefore an intrinsic pro-survival response that protects neurons from death induced by cell edema.


Subject(s)
Actins , N-Methylaspartate , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Dendritic Spines/metabolism , Formins , Ischemia/metabolism , Mice , N-Methylaspartate/metabolism , Neurons/metabolism , Receptors, Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Water/metabolism
3.
Mol Biol Cell ; 33(7): ar64, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35352947

ABSTRACT

Neuron navigator 1 (Nav1) is a cytoskeleton-associated protein expressed during brain development that is necessary for proper neuritogenesis, but the underlying mechanisms are poorly understood. Here we show that Nav1 is present in elongating axon tracts during mouse brain embryogenesis. We found that depletion of Nav1 in cultured neurons disrupts growth cone morphology and neurotrophin-stimulated neuritogenesis. In addition to regulating both F-actin and microtubule properties, Nav1 promotes actin-rich membrane ruffles in the growth cone and promotes macropinocytosis at those membrane ruffles, including internalization of the TrkB receptor for the neurotrophin brain-derived neurotropic factor (BDNF). Growth cone macropinocytosis is important for downstream signaling, neurite targeting, and membrane recycling, implicating Nav1 in one or more of these processes. Depletion of Nav1 also induces transient membrane blebbing via disruption of signaling in the Rho GTPase signaling pathway, supporting the novel role of Nav1 in dynamic actin-based membrane regulation at the cell periphery. These data demonstrate that Nav1 works at the interface of microtubules, actin, and plasma membrane to organize the cell periphery and promote uptake of growth and guidance cues to facilitate neural morphogenesis during development.


Subject(s)
Actins , Growth Cones , Actins/metabolism , Animals , Cells, Cultured , Cytoskeletal Proteins/metabolism , Growth Cones/metabolism , Mice , Microtubules/metabolism , Nerve Growth Factors/metabolism , Neurons/metabolism , Receptors, Nerve Growth Factor/metabolism
4.
Front Mol Neurosci ; 15: 1099554, 2022.
Article in English | MEDLINE | ID: mdl-36710926

ABSTRACT

Neuron navigators (Navigators) are cytoskeletal-associated proteins important for neuron migration, neurite growth, and axon guidance, but they also function more widely in other tissues. Recent studies have revealed novel cellular functions of Navigators such as macropinocytosis, and have implicated Navigators in human disorders of axon growth. Navigators are present in most or all bilaterian animals: vertebrates have three Navigators (NAV1-3), Drosophila has one (Sickie), and Caenorhabditis elegans has one (Unc-53). Structurally, Navigators have conserved N- and C-terminal regions each containing specific domains. The N-terminal region contains a calponin homology (CH) domain and one or more SxIP motifs, thought to interact with the actin cytoskeleton and mediate localization to microtubule plus-end binding proteins, respectively. The C-terminal region contains two coiled-coil domains, followed by a AAA+ family nucleoside triphosphatase domain of unknown activity. The Navigators appear to have evolved by fusion of N- and C-terminal region homologs present in simpler organisms. Overall, Navigators participate in the cytoskeletal response to extracellular cues via microtubules and actin filaments, in conjunction with membrane trafficking. We propose that uptake of fluid-phase cues and nutrients and/or downregulation of cell surface receptors could represent general mechanisms that explain Navigator functions. Future studies developing new models, such as conditional knockout mice or human cerebral organoids may reveal new insights into Navigator function. Importantly, further biochemical studies are needed to define the activities of the Navigator AAA+ domain, and to study potential interactions among different Navigators and their binding partners.

5.
Front Physiol ; 12: 657074, 2021.
Article in English | MEDLINE | ID: mdl-34220531

ABSTRACT

Dendritic spines are small, bulbous protrusions along the dendrites of neurons and are sites of excitatory postsynaptic activity. The morphology of spines has been implicated in their function in synaptic plasticity and their shapes have been well-characterized, but the potential mechanics underlying their shape development and maintenance have not yet been fully understood. In this work, we explore the mechanical principles that could underlie specific shapes using a minimal biophysical model of membrane-actin interactions. Using this model, we first identify the possible force regimes that give rise to the classic spine shapes-stubby, filopodia, thin, and mushroom-shaped spines. We also use this model to investigate how the spine neck might be stabilized using periodic rings of actin or associated proteins. Finally, we use this model to predict that the cooperation between force generation and ring structures can regulate the energy landscape of spine shapes across a wide range of tensions. Thus, our study provides insights into how mechanical aspects of actin-mediated force generation and tension can play critical roles in spine shape maintenance.

6.
Biophys J ; 117(10): 1963-1980, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31668749

ABSTRACT

Dendritic spines are the primary excitatory postsynaptic sites that act as subcompartments of signaling. Ca2+ is often the first and most rapid signal in spines. Downstream of calcium, the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) pathway plays a critical role in the regulation of spine formation, morphological modifications, and ultimately, learning and memory. Although the dynamics of calcium are reasonably well-studied, calcium-induced cAMP/PKA dynamics, particularly with respect to frequency modulation, are not fully explored. In this study, we present a well-mixed model for the dynamics of calcium-induced cAMP/PKA dynamics in dendritic spines. The model is constrained using experimental observations in the literature. Further, we measured the calcium oscillation frequency in dendritic spines of cultured hippocampal CA1 neurons and used these dynamics as model inputs. Our model predicts that the various steps in this pathway act as frequency modulators for calcium, and the high frequency of calcium input is filtered by adenylyl cyclase 1 and phosphodiesterases in this pathway such that cAMP/PKA only responds to lower frequencies. This prediction has important implications for noise filtering and long-timescale signal transduction in dendritic spines. A companion manuscript presents a three-dimensional spatial model for the same pathway.


Subject(s)
Calcium/metabolism , Computer Simulation , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Dendritic Spines/metabolism , Signal Transduction , Adenylyl Cyclases/metabolism , Animals , Calcium Signaling , Calmodulin/metabolism , Enzyme Activation , Female , Kinetics , Male , Models, Biological , Models, Molecular , Neurons/metabolism , Phosphoric Diester Hydrolases/metabolism , Phosphorylation , Rats, Sprague-Dawley
7.
J Vis Exp ; (150)2019 08 28.
Article in English | MEDLINE | ID: mdl-31524872

ABSTRACT

Neurons differentiated in two-dimensional culture from human pluripotent stem-cell-derived neural progenitor cells (NPCs) represent a powerful model system to explore disease mechanisms and carry out high content screening (HCS) to interrogate compound libraries or identify gene mutation phenotypes. However, with human cells the transition from NPC to functional, mature neuron requires several weeks. Synapses typically start to form after 3 weeks of differentiation in monolayer culture, and several neuron-specific proteins, for example the later expressing pan-neuronal marker NeuN, or the layer 5/6 cerebral cortical neuron marker CTIP2, begin to express around 4-5 weeks post-differentiation. This lengthy differentiation time can be incompatible with optimal culture conditions used for small volume, multi-well HCS platforms. Among the many challenges are the need for well-adhered, uniformly distributed cells with minimal cell clustering, and culture procedures that foster long-term viability and functional synapse maturation. One approach is to differentiate neurons in a large volume format, then replate them at a later time point in HCS-compatible multi-wells. Some main challenges when using this replating approach concern reproducibility and cell viability, due to the stressful disruption of the dendritic and axonal network. Here we demonstrate a detailed and reliable procedure for enzymatically resuspending human induced pluripotent stem cell (hiPSC)-derived neurons after their differentiation for 4-8 weeks in a large-volume format, transferring them to 384-well microtiter plates, and culturing them for a further 1-3 weeks with excellent cell survival. This replating of human neurons not only allows the study of synapse assembly and maturation within two weeks from replating, but also enables studies of neurite regeneration and growth cone characteristics. We provide examples of scalable assays for neuritogenesis and synaptogenesis using a 384-well platform.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Neurites/metabolism , Neurogenesis , Synapses/metabolism , Calcium Signaling , Cell Line , Cell Survival , Cells, Cultured , Dendrites/metabolism , Electrodes , Humans , Neurogenesis/physiology , Pluripotent Stem Cells/cytology , Proteolysis , Reproducibility of Results
8.
Neuron ; 98(4): 864, 2018 05 16.
Article in English | MEDLINE | ID: mdl-29772205
9.
Neuron ; 98(2): 235-237, 2018 04 18.
Article in English | MEDLINE | ID: mdl-29673473

ABSTRACT

In this issue of Neuron, Liao and colleagues (2018) uncover a surprising way that the guidance molecule MIG14/Wntless operates in dendrite self-avoidance in sensorimotor neurons. Not only does MIG14/Wntless not require the soluble cue Wntless, but it can mediate direct cell-cell contact at the plasma membrane. During dendrite tip contact, MIG14/Wntless drives local bursts of WASP-dependent actin filament assembly that facilitate sister dendrite repulsion.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Dendrites , Neurogenesis , Siblings
10.
Proc Natl Acad Sci U S A ; 114(22): E4462-E4471, 2017 05 30.
Article in English | MEDLINE | ID: mdl-28500272

ABSTRACT

The molecular pathogenesis of bipolar disorder (BPD) is poorly understood. Using human-induced pluripotent stem cells (hiPSCs) to unravel such mechanisms in polygenic diseases is generally challenging. However, hiPSCs from BPD patients responsive to lithium offered unique opportunities to discern lithium's target and hence gain molecular insight into BPD. By profiling the proteomics of BDP-hiPSC-derived neurons, we found that lithium alters the phosphorylation state of collapsin response mediator protein-2 (CRMP2). Active nonphosphorylated CRMP2, which binds cytoskeleton, is present throughout the neuron; inactive phosphorylated CRMP2, which dissociates from cytoskeleton, exits dendritic spines. CRMP2 elimination yields aberrant dendritogenesis with diminished spine density and lost lithium responsiveness (LiR). The "set-point" for the ratio of pCRMP2:CRMP2 is elevated uniquely in hiPSC-derived neurons from LiR BPD patients, but not with other psychiatric (including lithium-nonresponsive BPD) and neurological disorders. Lithium (and other pathway modulators) lowers pCRMP2, increasing spine area and density. Human BPD brains show similarly elevated ratios and diminished spine densities; lithium therapy normalizes the ratios and spines. Consistent with such "spine-opathies," human LiR BPD neurons with abnormal ratios evince abnormally steep slopes for calcium flux; lithium normalizes both. Behaviorally, transgenic mice that reproduce lithium's postulated site-of-action in dephosphorylating CRMP2 emulate LiR in BPD. These data suggest that the "lithium response pathway" in BPD governs CRMP2's phosphorylation, which regulates cytoskeletal organization, particularly in spines, modulating neural networks. Aberrations in the posttranslational regulation of this developmentally critical molecule may underlie LiR BPD pathogenesis. Instructively, examining the proteomic profile in hiPSCs of a functional agent-even one whose mechanism-of-action is unknown-might reveal otherwise inscrutable intracellular pathogenic pathways.


Subject(s)
Bipolar Disorder , Induced Pluripotent Stem Cells/drug effects , Lithium/pharmacology , Models, Biological , Protein Processing, Post-Translational/drug effects , Animals , Bipolar Disorder/genetics , Bipolar Disorder/metabolism , Bipolar Disorder/physiopathology , Brain Chemistry , Calcium/metabolism , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/physiology , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Proteomics
11.
Mol Cell Neurosci ; 68: 36-45, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25827095

ABSTRACT

Neurons express three closely related dynamin genes. Dynamin 1 has long been implicated in the regulation of synaptic vesicle recycling in nerve terminals, and dynamins 2 and 3 were more recently shown also to contribute to synaptic vesicle recycling in specific and distinguishable ways. In cultured hippocampal neurons we found that chronic suppression of spontaneous network activity differentially regulated the targeting of endogenous dynamins 1 and 3 to nerve terminals, while dynamin 2 was unaffected. Specifically, when neural activity was chronically silenced for 1-2weeks by tetrodotoxin (TTX), the clustering of dynamin 1 at nerve terminals was reduced, while the clustering of dynamin 3 significantly increased. Moreover, dynamin 3 clustering was induced within hours by the sustained blockade of AMPA receptors, suggesting that AMPA receptors may function to prevent Dyn3 accumulation within nerve terminals. Clustering of dynamin 3 was induced by an antagonist of the calcium-dependent protein phosphatase calcineurin, but was not dependent upon intact actin filaments. TTX-induced clustering of Dyn3 occurred with a markedly slower time-course than the previously described clustering of synapsin 1. Potassium-induced depolarization rapidly de-clustered dynamin 3 from nerve terminals within minutes. These results, which have implications for homeostatic synapse restructuring, indicate that the three dynamins have evolved different regulatory mechanisms for trafficking to and from nerve terminals in response to changes in neural activity.


Subject(s)
Dynamin III/metabolism , Dynamin I/metabolism , Neurons/cytology , Neurons/metabolism , Presynaptic Terminals/metabolism , Animals , Cells, Cultured , Dynamin I/genetics , Dynamin III/genetics , Embryo, Mammalian , Excitatory Amino Acid Antagonists/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Immunosuppressive Agents/pharmacology , Neurons/drug effects , Presynaptic Terminals/drug effects , Presynaptic Terminals/ultrastructure , Protein Transport/drug effects , Protein Transport/genetics , Quinoxalines/pharmacology , Rats , Rats, Wistar , Sodium Channel Blockers/pharmacology , Synapses/drug effects , Synapses/metabolism , Synapses/ultrastructure , Synaptosomes/metabolism , Tacrolimus/analogs & derivatives , Tacrolimus/pharmacology , Tetrodotoxin/pharmacology , Time Factors
12.
Neuroreport ; 25(17): 1331-7, 2014 Dec 03.
Article in English | MEDLINE | ID: mdl-25304495

ABSTRACT

Increasing evidence suggests that cellular stress may underlie mood disorders such as bipolar disorder and major depression, particularly as lithium and its targets can protect against neuronal cell death. Here we describe N-methyl-D-aspartate (NMDA)-induced filamentous actin reorganization (NIFAR) as a useful in-vitro model for studying acute neurocellular stress and investigating the effects of mood stabilizers. Brief incubation of cultured neurons with NMDA (50 µM, 5 min) induces marked reorganization of F-actin within the somatodendritic domain of a majority of neurons. During NIFAR, F-actin is rapidly depleted from dendritic spines and aberrantly aggregates within the dendrite shaft. The widely used mood stabilizer lithium chloride prevented NIFAR in a time-dependent and dose-dependent manner, consistent with its known efficacy in treating bipolar disorder. Inhibitors of the lithium target glycogen synthase kinase 3 and its upstream activator phosphoinositide-3-kinase also prevented NIFAR. The antidepressant compounds imipramine and fluoxetine also attenuated NIFAR. These findings have potential relevance to neuropsychiatric diseases characterized by excessive glutamate receptor activity and synaptotoxicity. We propose that protection of the dendritic actin cytoskeleton may be a common mechanism shared by various mood stabilizers.


Subject(s)
Actins/metabolism , Antipsychotic Agents/pharmacology , Lithium Chloride/pharmacology , N-Methylaspartate/metabolism , Neurons/drug effects , Neurons/physiology , Animals , Antidepressive Agents/pharmacology , Cells, Cultured , Cytoskeleton/drug effects , Cytoskeleton/physiology , Dendrites/drug effects , Dendrites/physiology , Dose-Response Relationship, Drug , Fluoxetine/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Hippocampus/drug effects , Hippocampus/physiology , Imipramine/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Time Factors
13.
PLoS One ; 9(7): e102978, 2014.
Article in English | MEDLINE | ID: mdl-25068870

ABSTRACT

General anesthetics produce a reversible coma-like state through modulation of excitatory and inhibitory synaptic transmission. Recent evidence suggests that anesthetic exposure can also lead to sustained cognitive dysfunction. However, the subcellular effects of anesthetics on the structure of established synapses are not known. We investigated effects of the widely used volatile anesthetic isoflurane on the structural stability of hippocampal dendritic spines, a postsynaptic structure critical to excitatory synaptic transmission in learning and memory. Exposure to clinical concentrations of isoflurane induced rapid and non-uniform shrinkage and loss of dendritic spines in mature cultured rat hippocampal neurons. Spine shrinkage was associated with a reduction in spine F-actin concentration. Spine loss was prevented by either jasplakinolide or cytochalasin D, drugs that prevent F-actin disassembly. Isoflurane-induced spine shrinkage and loss were reversible upon isoflurane elimination. Thus, isoflurane destabilizes spine F-actin, resulting in changes to dendritic spine morphology and number. These findings support an actin-based mechanism for isoflurane-induced alterations of synaptic structure in the hippocampus. These reversible alterations in dendritic spine structure have important implications for acute anesthetic effects on excitatory synaptic transmission and synaptic stability in the hippocampus, a locus for anesthetic-induced amnesia, and have important implications for anesthetic effects on synaptic plasticity.


Subject(s)
Actins/metabolism , Anesthetics, Inhalation/pharmacology , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Isoflurane/pharmacology , Animals , Neuroglia/drug effects , Neuroglia/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Rats
14.
Mol Biol Cell ; 25(14): 2152-60, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24829386

ABSTRACT

Capping protein (CP) binds to barbed ends of growing actin filaments and inhibits elongation. CP is essential for actin-based motility in cell-free systems and in Dictyostelium. Even though CP is believed to be critical for creating the lamellipodial actin structure necessary for protrusion and migration, CP's role in mammalian cell migration has not been directly tested. Moreover, recent studies have suggested that structures besides lamellipodia, including lamella and filopodia, may have unappreciated roles in cell migration. CP has been postulated to be absent from filopodia, and thus its role in filopodial activity has remained unexplored. We report that silencing CP in both cultured mammalian B16F10 cells and in neurons of developing neocortex impaired cell migration. Moreover, we unexpectedly observed that low levels of CP were detectable in the majority of filopodia. CP depletion decreased filopodial length, altered filopodial shape, and reduced filopodial dynamics. Our results support an expansion of the potential roles that CP plays in cell motility by implicating CP in filopodia as well as in lamellipodia, both of which are important for locomotion in many types of migrating cells.


Subject(s)
CapZ Actin Capping Protein/physiology , Cell Movement , Pseudopodia/ultrastructure , Actins/metabolism , Animals , Cell Line, Tumor , Cell Shape , Gene Knockdown Techniques , Mice , Pseudopodia/metabolism
15.
PLoS One ; 9(4): e94787, 2014.
Article in English | MEDLINE | ID: mdl-24740405

ABSTRACT

A current model posits that cofilin-dependent actin severing negatively impacts dendritic spine volume. Studies suggested that increased cofilin activity underlies activity-dependent spine shrinkage, and that reduced cofilin activity induces activity-dependent spine growth. We suggest instead that both types of structural plasticity correlate with decreased cofilin activity. However, the mechanism of inhibition determines the outcome for spine morphology. RNAi in rat hippocampal cultures demonstrates that cofilin is essential for normal spine maintenance. Cofilin-F-actin binding and filament barbed-end production decrease during the early phase of activity-dependent spine shrinkage; cofilin concentration also decreases. Inhibition of the cathepsin B/L family of proteases prevents both cofilin loss and spine shrinkage. Conversely, during activity-dependent spine growth, LIM kinase stimulates cofilin phosphorylation, which activates phospholipase D-1 to promote actin polymerization. These results implicate novel molecular mechanisms and prompt a revision of the current model for how cofilin functions in activity-dependent structural plasticity.


Subject(s)
Cofilin 1/metabolism , Dendritic Spines/physiology , Down-Regulation , Neurons/physiology , Actins/metabolism , Animals , Cells, Cultured , Cofilin 1/genetics , Dendritic Spines/drug effects , Female , Hippocampus/cytology , Immunohistochemistry , Lim Kinases/metabolism , Long-Term Potentiation/physiology , Long-Term Synaptic Depression/physiology , Microscopy, Confocal , N-Methylaspartate/pharmacology , Neurons/drug effects , Neurons/metabolism , Phospholipase D/metabolism , Phosphorylation , Protein Binding , RNA Interference , Rats , Tacrolimus/pharmacology , Time-Lapse Imaging
16.
J Biol Chem ; 287(18): 14984-93, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22403409

ABSTRACT

The predominant brain microtubule-associated proteins MAP2 and tau play a critical role in microtubule cytoskeletal organization and function. We have previously reported that PP2A/Bα, a major protein phosphatase 2A (PP2A) holoenzyme, binds to and dephosphorylates tau, and regulates microtubule stability. Here, we provide evidence that MAP2 co-purifies with and is dephosphorylated by endogenous PP2A/Bα in bovine gray matter. It co-localizes with PP2A/Bα in immature and mature human neuronal cell bodies. PP2A co-immunoprecipitates with and directly interacts with MAP2. Using in vitro binding assays, we show that PP2A/Bα binds to MAP2c isoforms through a region encompassing the microtubule-binding domain and upstream proline-rich region. Tau and MAP2 compete for binding to and dephosphorylation by PP2A/Bα. Remarkably, the protein-tyrosine kinase Fyn, which binds to the proline-rich RTPPKSP motif conserved in both MAP2 and tau, inhibits the interaction of PP2A/Bα with either tau or MAP2c. The corresponding synthetic RTPPKSP peptide, but not the phosphorylated RpTPPKSP version, competes with Tau and MAP2c for binding to PP2A/Bα. Significantly, down-regulation of PP2A/Bα and deregulation of Fyn-Tau protein interactions have been linked to enhanced tau phosphorylation in Alzheimer disease. Together, our results suggest that PP2A/Bα is part of segregated MAP2 and tau signaling scaffolds that can coordinate the action of key kinases and phosphatases involved in modulating neuronal plasticity. Deregulation of these compartmentalized multifunctional protein complexes is likely to contribute to tau deregulation, microtubule disruption, and altered signaling in tauopathies.


Subject(s)
Alzheimer Disease/metabolism , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , tau Proteins/metabolism , Alzheimer Disease/pathology , Amino Acid Motifs , Animals , Cattle , Cell Line , Neurons/pathology , Peptides/pharmacology , Protein Binding/drug effects , Protein Isoforms/metabolism , Signal Transduction/drug effects
17.
BMC Neurosci ; 12: 100, 2011 Oct 11.
Article in English | MEDLINE | ID: mdl-21989414

ABSTRACT

BACKGROUND: To date, some of the most useful and physiologically relevant neuronal cell culture systems, such as high density co-cultures of astrocytes and primary hippocampal neurons, or differentiated stem cell-derived cultures, are characterized by high cell density and partially overlapping cellular structures. Efficient analytical strategies are required to enable rapid, reliable, quantitative analysis of neuronal morphology in these valuable model systems. RESULTS: Here we present the development and validation of a novel bioinformatics pipeline called NeuriteQuant. This tool enables fully automated morphological analysis of large-scale image data from neuronal cultures or brain sections that display a high degree of complexity and overlap of neuronal outgrowths. It also provides an efficient web-based tool to review and evaluate the analysis process. In addition to its built-in functionality, NeuriteQuant can be readily extended based on the rich toolset offered by ImageJ and its associated community of developers. As proof of concept we performed automated screens for modulators of neuronal development in cultures of primary neurons and neuronally differentiated P19 stem cells, which demonstrated specific dose-dependent effects on neuronal morphology. CONCLUSIONS: NeuriteQuant is a freely available open-source tool for the automated analysis and effective review of large-scale high-content screens. It is especially well suited to quantify the effect of experimental manipulations on physiologically relevant neuronal cultures or brain sections that display a high degree of complexity and overlap among neurites or other cellular structures.


Subject(s)
Image Cytometry/methods , Neurites/ultrastructure , Neurogenesis/physiology , Software Validation , Software/standards , Algorithms , Animals , Cell Culture Techniques/methods , Cell Line , Computational Biology/methods , Drug Evaluation, Preclinical/methods , Information Dissemination/methods , Mice , Nerve Growth Factors/physiology , Neurites/physiology
18.
Eur J Cell Biol ; 90(4): 291-300, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21146252

ABSTRACT

Regulation of the actin cytoskeleton is critical for neurite formation. Tropomodulins (Tmods) regulate polymerization at actin filament pointed ends. Previous experiments using a mouse model deficient for the neuron specific isoform Tmod2 suggested a role for Tmods in neuronal function by impacting processes underlying learning and memory. However, the role of Tmods in neuronal function on the cellular level remains unknown. Immunofluorescence localization of the neuronal isoforms Tmod1 and Tmod2 in cultured rat primary hippocampal neurons revealed that Tmod1 is enriched along the proximal part of F-actin bundles in lamellipodia of spreading cells and in growth cones of extending neurites, while Tmod2 appears largely cytoplasmic. Functional analysis of these Tmod isoforms in a mouse neuroblastoma N2a cell line showed that knockdown of Tmod2 resulted in a significant increase in the number of neurite-forming cells and in neurite length. While N2a cells compensated for Tmod2 knockdown by increasing Tmod1 levels, over-expression of exogenous Tmod1 had no effect on neurite outgrowth. Moreover, knockdown of Tmod1 increased the number of neurites formed per cell, without effect on the number of neurite-forming cells or neurite length. Taken together, these results indicate that Tmod1 and Tmod2 have mechanistically distinct inhibitory roles in neurite formation, likely mediated via different effects on F-actin dynamics and via differential localizations during early neuritogenesis.


Subject(s)
Neurites/physiology , Neurons/physiology , Tropomodulin/metabolism , Actin Cytoskeleton/metabolism , Actins/genetics , Animals , Cell Line, Tumor , Cells, Cultured , Cytoskeleton/genetics , Growth Cones/metabolism , Growth Cones/physiology , Hippocampus/physiology , Mice , Neurites/metabolism , Pseudopodia/metabolism , Pseudopodia/physiology , Rats , Tropomodulin/genetics
19.
Proc Natl Acad Sci U S A ; 106(11): 4507-12, 2009 Mar 17.
Article in English | MEDLINE | ID: mdl-19240213

ABSTRACT

Neuregulin-1 (NRG1) and its ErbB2/B4 receptors are encoded by candidate susceptibility genes for schizophrenia, yet the essential functions of NRG1 signaling in the CNS are still unclear. Using CRE/LOX technology, we have inactivated ErbB2/B4-mediated NRG1 signaling specifically in the CNS. In contrast to expectations, cell layers in the cerebral cortex, hippocampus, and cerebellum develop normally in the mutant mice. Instead, loss of ErbB2/B4 impairs dendritic spine maturation and perturbs interactions of postsynaptic scaffold proteins with glutamate receptors. Conversely, increased NRG1 levels promote spine maturation. ErbB2/B4-deficient mice show increased aggression and reduced prepulse inhibition. Treatment with the antipsychotic drug clozapine reverses the behavioral and spine defects. We conclude that ErbB2/B4-mediated NRG1 signaling modulates dendritic spine maturation, and that defects at glutamatergic synapses likely contribute to the behavioral abnormalities in ErbB2/B4-deficient mice.


Subject(s)
Cerebral Cortex/cytology , Dendritic Spines/pathology , Nerve Tissue Proteins/physiology , Receptor, ErbB-2/physiology , Signal Transduction , Adaptor Proteins, Signal Transducing , Animals , Antipsychotic Agents/pharmacology , Central Nervous System , Clozapine/pharmacology , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Neuregulin-1 , Oncogene Proteins v-erbB/deficiency , Oncogene Proteins v-erbB/physiology , Receptors, Glutamate
20.
Curr Biol ; 17(15): R611-4, 2007 Aug 07.
Article in English | MEDLINE | ID: mdl-17686437

ABSTRACT

A new study has shown that, near the tip of a growing axon, dephosphorylation of the microtubule-associated protein Doublecortin is controlled by protein phosphatase 1 and its regulator spinophilin. This results in spatially regulated microtubule bundling within the axon and more efficient axon outgrowth.


Subject(s)
Neurites/metabolism , Animals , Doublecortin Domain Proteins , Mice , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Neuropeptides/metabolism , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL
...