Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Proc Natl Acad Sci U S A ; 114(22): E4462-E4471, 2017 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-28500272

RESUMO

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.


Assuntos
Transtorno Bipolar , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Lítio/farmacologia , Modelos Biológicos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Animais , Transtorno Bipolar/genética , Transtorno Bipolar/metabolismo , Transtorno Bipolar/fisiopatologia , Química Encefálica , Cálcio/metabolismo , Células Cultivadas , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/química , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Proteômica
2.
Biophys J ; 117(10): 1963-1980, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31668749

RESUMO

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.


Assuntos
Cálcio/metabolismo , Simulação por Computador , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Espinhas Dendríticas/metabolismo , Transdução de Sinais , Adenilil Ciclases/metabolismo , Animais , Sinalização do Cálcio , Calmodulina/metabolismo , Ativação Enzimática , Feminino , Cinética , Masculino , Modelos Biológicos , Modelos Moleculares , Neurônios/metabolismo , Diester Fosfórico Hidrolases/metabolismo , Fosforilação , Ratos Sprague-Dawley
3.
Mol Cell Neurosci ; 68: 36-45, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25827095

RESUMO

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.


Assuntos
Dinamina III/metabolismo , Dinamina I/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Terminações Pré-Sinápticas/metabolismo , Animais , Células Cultivadas , Dinamina I/genética , Dinamina III/genética , Embrião de Mamíferos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/citologia , Imunossupressores/farmacologia , Neurônios/efeitos dos fármacos , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/ultraestrutura , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , Quinoxalinas/farmacologia , Ratos , Ratos Wistar , Bloqueadores dos Canais de Sódio/farmacologia , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Sinapses/ultraestrutura , Sinaptossomos/metabolismo , Tacrolimo/análogos & derivados , Tacrolimo/farmacologia , Tetrodotoxina/farmacologia , Fatores de Tempo
4.
Mol Biol Cell ; 35(2): ar23, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38088877

RESUMO

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.


Assuntos
Actinas , Proteínas de Membrana , Actinas/metabolismo , Proteínas de Membrana/metabolismo , Espinhas Dendríticas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína Quinase C/metabolismo , Fosforilação
5.
J Biol Chem ; 287(18): 14984-93, 2012 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-22403409

RESUMO

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.


Assuntos
Doença de Alzheimer/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteína Fosfatase 2/metabolismo , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/patologia , Motivos de Aminoácidos , Animais , Bovinos , Linhagem Celular , Neurônios/patologia , Peptídeos/farmacologia , Ligação Proteica/efeitos dos fármacos , Isoformas de Proteínas/metabolismo , Transdução de Sinais/efeitos dos fármacos
6.
Proc Natl Acad Sci U S A ; 106(11): 4507-12, 2009 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-19240213

RESUMO

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.


Assuntos
Córtex Cerebral/citologia , Espinhas Dendríticas/patologia , Proteínas do Tecido Nervoso/fisiologia , Receptor ErbB-2/fisiologia , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antipsicóticos/farmacologia , Sistema Nervoso Central , Clozapina/farmacologia , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Neuregulina-1 , Proteínas Oncogênicas v-erbB/deficiência , Proteínas Oncogênicas v-erbB/fisiologia , Receptores de Glutamato
7.
Front Mol Neurosci ; 15: 1099554, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36710926

RESUMO

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.

8.
Nat Commun ; 13(1): 6037, 2022 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-36229429

RESUMO

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.


Assuntos
Actinas , N-Metilaspartato , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animais , Espinhas Dendríticas/metabolismo , Forminas , Isquemia/metabolismo , Camundongos , N-Metilaspartato/metabolismo , Neurônios/metabolismo , Receptores de Glutamato/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Água/metabolismo
9.
Mol Biol Cell ; 33(7): ar64, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35352947

RESUMO

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.


Assuntos
Actinas , Cones de Crescimento , Actinas/metabolismo , Animais , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Cones de Crescimento/metabolismo , Camundongos , Microtúbulos/metabolismo , Fatores de Crescimento Neural/metabolismo , Neurônios/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo
10.
BMC Neurosci ; 12: 100, 2011 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-21989414

RESUMO

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.


Assuntos
Citometria por Imagem/métodos , Neuritos/ultraestrutura , Neurogênese/fisiologia , Validação de Programas de Computador , Software/normas , Algoritmos , Animais , Técnicas de Cultura de Células/métodos , Linhagem Celular , Biologia Computacional/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Disseminação de Informação/métodos , Camundongos , Fatores de Crescimento Neural/fisiologia , Neuritos/fisiologia
11.
Front Physiol ; 12: 657074, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34220531

RESUMO

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.

12.
Neuron ; 52(5): 746-8, 2006 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-17145496

RESUMO

Dendritic spines form and grow during hippocampal long-term potentiation (LTP). In this issue of Neuron, a new study by Park et al. uses both serial reconstruction electron microscopy and time-lapse imaging to show that plasma membrane for such spine expansion is trafficked from recycling endosomes that reside locally at the spines themselves.


Assuntos
Espinhas Dendríticas/fisiologia , Hipocampo/citologia , Plasticidade Neuronal/fisiologia , Actinas/metabolismo , Actinas/fisiologia , Animais , Membrana Celular/fisiologia , Membrana Celular/ultraestrutura , Endossomos/fisiologia , Endossomos/ultraestrutura , Hipocampo/fisiologia , Humanos , Potenciação de Longa Duração/fisiologia , Microscopia Eletrônica , Sinapses/fisiologia
13.
Curr Biol ; 17(15): R611-4, 2007 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-17686437

RESUMO

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.


Assuntos
Neuritos/metabolismo , Animais , Proteínas do Domínio Duplacortina , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Neuropeptídeos/metabolismo , Fosforilação
14.
Neuron ; 48(1): 77-90, 2005 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-16202710

RESUMO

Spine morphology is regulated by intracellular signals, like PKC, that affect cytoskeletal and membrane dynamics. We investigated the role of MARCKS (myristoylated, alanine-rich C-kinase substrate) in dendrites of 3-week-old hippocampal cultures. MARCKS associates with membranes via the combined action of myristoylation and a polybasic effector domain, which binds phospholipids and/or F-actin, unless phosphorylated by PKC. Knockdown of endogenous MARCKS using RNAi reduced spine density and size. PKC activation induced similar effects, which were prevented by expression of a nonphosphorylatable mutant. Moreover, expression of pseudophosphorylated MARCKS was, by itself, sufficient to induce spine loss and shrinkage, accompanied by reduced F-actin content. Nonphosphorylatable MARCKS caused spine elongation and increased the mobility of spine actin clusters. Surprisingly, it also decreased spine density via a novel mechanism of spine fusion, an effect that required the myristoylation sequence. Thus, MARCKS is a key factor in the maintenance of dendritic spines and contributes to PKC-dependent morphological plasticity.


Assuntos
Espinhas Dendríticas/fisiologia , Hipocampo/citologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas de Membrana/fisiologia , Neurônios/citologia , Proteína Quinase C/metabolismo , Actinas/metabolismo , Análise de Variância , Animais , Células Cultivadas , Espinhas Dendríticas/efeitos dos fármacos , Diagnóstico por Imagem , Embrião de Mamíferos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Fluorimunoensaio/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Proteínas de Fluorescência Verde/biossíntese , Imuno-Histoquímica/métodos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Modelos Biológicos , Mutagênese/efeitos dos fármacos , Mutagênese/fisiologia , Substrato Quinase C Rico em Alanina Miristoilada , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp/métodos , Terminações Pré-Sinápticas/metabolismo , RNA Interferente Pequeno/farmacologia , Ratos , Sinapses/efeitos dos fármacos , Sinapses/fisiologia , Sinaptofisina/metabolismo , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Tempo , Transfecção/métodos
15.
Trends Cell Biol ; 12(10): 455-7, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12441241

RESUMO

A Keystone symposium held in Taos, New Mexico, 21-26 March 2002 provided the setting for a pioneering gathering of cell biologists and neuroscientists. Under the guidance of organizers Tom Pollard, James Sabry and Carla Schatz, the meeting, entitled 'Cellular Motility and Signaling in the Wiring and Plasticity of Nervous Systems', brought together two groups of researchers that ordinarily rarely connect at scientific conferences. The goal of this new collegial interchange was to fuse expertise on cytoskeletal dynamics with emerging ideas in neuronal development.


Assuntos
Citoesqueleto/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Transdução de Sinais/fisiologia
16.
J Cell Biol ; 157(7): 1187-96, 2002 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-12082079

RESUMO

MAP2 and tau exhibit microtubule-stabilizing activities that are implicated in the development and maintenance of neuronal axons and dendrites. The proteins share a homologous COOH-terminal domain, composed of three or four microtubule binding repeats separated by inter-repeats (IRs). To investigate how MAP2 and tau stabilize microtubules, we calculated 3D maps of microtubules fully decorated with MAP2c or tau using cryo-EM and helical image analysis. Comparing these maps with an undecorated microtubule map revealed additional densities along protofilament ridges on the microtubule exterior, indicating that MAP2c and tau form an ordered structure when they bind microtubules. Localization of undecagold attached to the second IR of MAP2c showed that IRs also lie along the ridges, not between protofilaments. The densities attributable to the microtubule-associated proteins lie in close proximity to helices 11 and 12 and the COOH terminus of tubulin. Our data further suggest that the evolutionarily maintained differences observed in the repeat domain may be important for the specific targeting of different repeats to either alpha or beta tubulin. These results provide strong evidence suggesting that MAP2c and tau stabilize microtubules by binding along individual protofilaments, possibly by bridging the tubulin interfaces.


Assuntos
Citoesqueleto/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas tau/metabolismo , Animais , Sítios de Ligação , Bovinos , Humanos , Imageamento Tridimensional , Camundongos , Proteínas Associadas aos Microtúbulos/química , Microtúbulos/química , Microtúbulos/ultraestrutura , Modelos Biológicos , Modelos Moleculares , Mutação , Ligação Proteica , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Tubulina (Proteína)/química , Tubulina (Proteína)/isolamento & purificação , Proteínas tau/química
17.
J Vis Exp ; (150)2019 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-31524872

RESUMO

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.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Neuritos/metabolismo , Neurogênese , Sinapses/metabolismo , Sinalização do Cálcio , Linhagem Celular , Sobrevivência Celular , Células Cultivadas , Dendritos/metabolismo , Eletrodos , Humanos , Neurogênese/fisiologia , Células-Tronco Pluripotentes/citologia , Proteólise , Reprodutibilidade dos Testes
18.
Neuron ; 98(2): 235-237, 2018 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-29673473

RESUMO

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.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animais , Dendritos , Neurogênese , Irmãos
19.
Curr Biol ; 14(5): 363-71, 2004 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-15028210

RESUMO

BACKGROUND: MAP2 and tau are abundant microtubule-associated proteins (MAPs) in neurons. The development of neuronal dendrites and axons requires a dynamic interaction between microtubules and actin filaments. MAPs represent good candidates to mediate such interactions. Although MAP2c and tau have similar, well-characterized microtubule binding activities, their actin interaction is poorly understood. RESULTS: Here, we show by using a cosedimentation assay that MAP2c binds F-actin. Upon actin binding, MAP2c organizes F-actin into closely packed actin bundles. Moreover, we show by using a deletion approach that MAP2c's microtubule binding domain (MTBD) is both necessary and sufficient for both F-actin binding and bundling activities. Surprisingly, even though the MAP2 and tau MTBDs share high sequence homology and possess similar microtubule binding activities, tau is unable to bind or bundle F-actin. Furthermore, experiments with chimeric proteins demonstrate that the actin binding activity fully correlates with the ability to promote neurite initiation in neuroblastoma cells. CONCLUSIONS: These results provide the first demonstration that the MAP2c and tau MTBD domains exhibit distinct properties, diverging in actin binding and neurite initiation activities. These results implicate a novel actin function for MAP2c in neuronal morphogenesis and furthermore suggest that actin interactions could contribute to functional differences between MAP2 and tau in neurons.


Assuntos
Actinas/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Neurônios/metabolismo , Proteínas tau/metabolismo , Animais , Sítios de Ligação/fisiologia , Células Cultivadas , Centrifugação , Primers do DNA , Eletroforese em Gel de Poliacrilamida , Imuno-Histoquímica , Microscopia Eletrônica , Microtúbulos/ultraestrutura , Morfogênese/fisiologia , Neurônios/fisiologia , Proteínas Recombinantes de Fusão/metabolismo
20.
J Neurosci ; 23(29): 9479-90, 2003 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-14573527

RESUMO

During neurite initiation, cells surrounded by a flattened, actin-rich lamellipodium transform to produce thin, microtubule-filled neurite shafts tipped by actin-rich growth cones, but little is known about this transformation. Our detailed time-lapse analyses of cultured hippocampal neurons, a widely used model system for neuronal development, revealed that neurites emerge from segmented lamellipodia, which then gradually extend from the cell body to become nascent growth cones. This suggests that actin- and microtubule-rich structures are reorganized in a coordinated manner. We hypothesized that proteins such as microtubule-associated protein 2 (MAP2), which can interact with both cytoskeletal components, might be critically involved in neurite initiation. Live-cell video and fluorescence microscopy in Neuro-2a cells showed that expression of MAP2c triggers neurite formation via rapid accumulation and bundling of stable, MAP2c-bound microtubules, concurrent with a gradual transformation of lamellipodia into nascent growth cones. The microtubule-stabilizing agent Taxol did not mimic this effect, suggesting that the ability of MAP2c to stabilize microtubules is not sufficient for neurite initiation. However, combination of Taxol treatment with actin disruption induced robust process formation, suggesting that inhibitory effects of F-actin need to be overcome as well. Neurite initiation by MAP2c required its microtubule-binding domain and was enhanced by its binding domain for cAMP-dependent protein kinase (PKA). MAP2c mutants defective in both PKA and microtubule binding acted as dominant negative inhibitors of neurite initiation in neuroblastoma cells and primary hippocampal neurons. Together, these data suggest that MAP2c bears functions that both stabilize microtubules and directly or indirectly alter actin organization during neurite initiation.


Assuntos
Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Neuritos/fisiologia , Neuritos/ultraestrutura , Pseudópodes/ultraestrutura , Actinas/genética , Actinas/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Genes Dominantes , Hipocampo/citologia , Humanos , Camundongos , Microscopia de Vídeo , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/farmacologia , Microtúbulos/metabolismo , Neuritos/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/ultraestrutura , Pseudópodes/metabolismo , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA