Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 42(10): 113195, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37816355

RESUMO

Fatty acids have long been considered essential to brain development; however, the involvement of their synthesis in nervous system formation is unclear. We generate mice with knockout of GPSN2, an enzyme for synthesis of very-long-chain fatty acids (VLCFAs) and investigate the effects. Both GPSN2-/- and GPSN2+/- mice show abnormal neuronal networks as a result of impaired neuronal polarity determination. Lipidomics of GPSN2-/- embryos reveal that ceramide synthesis is specifically inhibited depending on FA length; namely, VLCFA-containing ceramide is reduced. We demonstrate that lipid rafts are highly enriched in growth cones and that GPSN2+/- neurons lose gangliosides in their membranes. Application of C24:0 ceramide, but not C16:0 ceramide or C24:0 phosphatidylcholine, to GPSN2+/- neurons rescues both neuronal polarity determination and lipid-raft density in the growth cone. Taken together, our results indicate that VLCFA synthesis contributes to physiological neuronal development in brain network formation, in particular neuronal polarity determination through the formation of lipid rafts.


Assuntos
Ceramidas , Esfingolipídeos , Animais , Camundongos , Ácidos Graxos , Microdomínios da Membrana , Neurônios
3.
Neurochem Res ; 47(9): 2668-2682, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35347634

RESUMO

Mammalian axon growth has mechanistic similarities with axon regeneration. The growth cone is an important structure that is involved in both processes, and GAP-43 (growth associated protein-43 kDa) is believed to be the classical molecular marker. Previously, we used growth cone phosphoproteomics to demonstrate that S96 and T172 of GAP-43 in rodents are highly phosphorylated sites that are phosphorylated by c-jun N-terminal protein kinase (JNK). We also revealed that phosphorylated (p)S96 and pT172 antibodies recognize growing axons in the developing brain and regenerating axons in adult peripheral nerves. In rodents, S142 is another putative JNK-dependent phosphorylation site that is modified at a lower frequency than S96 and T172. Here, we characterized this site using a pS142-specific antibody. We confirmed that pS142 was detected by co-expressing mouse GAP-43 and JNK1. pS142 antibody labeled growth cones and growing axons in developing mouse neurons. pS142 was sustained until at least nine weeks after birth in mouse brains. The pS142 antibody could detect regenerating axons following sciatic nerve injury in adult mice. Comparison of amino acid sequences indicated that rodent S142 corresponds to human S151, which is predicted to be a substrate of the MAPK family, which includes JNK. Thus, we confirmed that the pS142 antibody recognized human phospho-GAP-43 using activated JNK1, and also that its immunostaining pattern in neurons differentiated from human induced pluripotent cells was similar to those observed in mice. These results indicate that the S142 residue is phosphorylated by JNK1 and that the pS142 antibody is a new candidate molecular marker for axonal growth in both rodents and human.


Assuntos
Axônios , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Regeneração Nervosa , Animais , Axônios/metabolismo , Proteína GAP-43/metabolismo , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Mamíferos/metabolismo , Camundongos , Regeneração Nervosa/fisiologia , Fosforilação , Serina/metabolismo
4.
Front Cell Dev Biol ; 9: 660349, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34235144

RESUMO

During brain development, axon outgrowth and its subsequent pathfinding are reliant on a highly motile growth cone located at the tip of the axon. Actin polymerization that is regulated by actin-depolymerizing factors homology (ADF-H) domain-containing family drives the formation of lamellipodia and filopodia at the leading edge of growth cones for axon guidance. However, the precise localization and function of ADF-H domain-containing proteins involved in axon extension and retraction remain unclear. We have previously shown that transcripts and proteins of coactosin-like protein 1 (COTL1), an ADF-H domain-containing protein, are observed in neurites and axons in chick embryos. Coactosin overexpression analysis revealed that this protein was localized to axonal growth cones and involved in axon extension in the midbrain. We further examined the specific distribution of coactosin and cofilin within the growth cone using superresolution microscopy, structured illumination microscopy, which overcomes the optical diffraction limitation and is suitable to the analysis of cellular dynamic movements. We found that coactosin was tightly associated with F-actin bundles at the growth cones and that coactosin overexpression promoted the expansion of lamellipodia and extension of growth cones. Coactosin knockdown in oculomotor neurons resulted in an increase in the levels of the inactive, phosphorylated form of cofilin and dysregulation of actin polymerization and axonal elongation, which suggests that coactosin promoted axonal growth in a cofilin-dependent manner. Indeed, the application of a dominant-negative form of LIMK1, a downstream effector of GTPases, reversed the effect of coactosin knockdown on axonal growth by enhancing cofilin activity. Combined, our results indicate that coactosin functions promote the assembly of protrusive actin filament arrays at the leading edge for growth cone motility.

5.
Mol Brain ; 14(1): 66, 2021 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-33832520

RESUMO

GAP-43 is a vertebrate neuron-specific protein and that is strongly related to axon growth and regeneration; thus, this protein has been utilized as a classical molecular marker of these events and growth cones. Although GAP-43 was biochemically characterized more than a quarter century ago, how this protein is related to these events is still not clear. Recently, we identified many phosphorylation sites in the growth cone membrane proteins of rodent brains. Two phosphorylation sites of GAP-43, S96 and T172, were found within the top 10 hit sites among all proteins. S96 has already been characterized (Kawasaki et al., 2018), and here, phosphorylation of T172 was characterized. In vitro (cultured neurons) and in vivo, an antibody specific to phosphorylated T172 (pT172 antibody) specifically recognized cultured growth cones and growing axons in developing mouse neurons, respectively. Immunoblotting showed that pT172 antigens were more rapidly downregulated throughout development than those of pS96 antibody. From the primary structure, this phosphorylation site was predicted to be conserved in a wide range of animals including primates. In the developing marmoset brainstem and in differentiated neurons derived from human induced pluripotent stem cells, immunoreactivity with pT172 antibody revealed patterns similar to those in mice. pT172 antibody also labeled regenerating axons following sciatic nerve injury. Taken together, the T172 residue is widely conserved in a wide range of mammals including primates, and pT172 is a new candidate molecular marker for growing axons.


Assuntos
Axônios/metabolismo , Biomarcadores/metabolismo , Proteína GAP-43/metabolismo , Mamíferos/metabolismo , Fosfotreonina/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos/metabolismo , Encéfalo/embriologia , Callithrix , Células Cultivadas , Furões , Proteína GAP-43/química , Cones de Crescimento/metabolismo , Células HEK293 , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos Endogâmicos C57BL , Regeneração Nervosa , Fosforilação , Primatas , Nervo Isquiático/lesões
6.
Front Mol Neurosci ; 13: 150, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32922262

RESUMO

Neuronal polarization and growth are developmental processes that occur during neuronal cell differentiation. The molecular signaling mechanisms involved in these events in in vivo mammalian brain remain unclear. Also, cellular events of the neuronal polarization process within a given neuron are thought to be constituted of many independent intracellular signal transduction pathways (the "tug-of-war" model). However, in vivo results suggest that such pathways should be cooperative with one another among a given group of neurons in a region of the brain. Lipid rafts, specific membrane domains with low fluidity, are candidates for the hotspots of such intracellular signaling. Among the signals reported to be involved in polarization, a number are thought to be present or translocated to the lipid rafts in response to extracellular signals. As part of our analysis, we discuss how such novel molecular mechanisms are combined for effective regulation of neuronal polarization and growth, focusing on the significance of the lipid rafts, including results based on recently introduced methods.

7.
J Biochem ; 168(3): 295-303, 2020 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-32289170

RESUMO

Actin-microtubule crosstalk is implicated in the formation of cellular protrusions, but the mechanism remains unclear. In this study, we examined the regulation of cell protrusion involving a ubiquitously expressed microtubule-associated protein (MAP) 4, and its superfamily proteins, neuronal MAP2 and tau. Fluorescence microscopy revealed that these MAPs bound to F-actin and microtubules simultaneously, and formed F-actin/microtubule hybrid bundles. The hybrid bundle-forming activity was in the order of MAP2 > MAP4 ≫ tau. Interestingly, the microtubule assembly-promoting activity of MAP4 and MAP2, but not of tau, was upregulated by their interaction with F-actin. When MAP4 was overexpressed in NG108-15 cells, the number of cell processes and maximum process length of each cell increased significantly by 28% and 30%, respectively. Super-resolution microscopy revealed that 95% of microtubules in cell processes colocalized with F-actin, and MAP4 was always found in their vicinity. These results suggest that microtubule elongation along F-actin induced by MAP4 contributes to the formation of cellular protrusions. Since MAP4, MAP2 and tau had different crosstalk activity between F-actin and microtubules, it is likely that the functional differentiation of these MAPs is a driving force for neural evolution, causing significant changes in cell morphology.


Assuntos
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Extensões da Superfície Celular/metabolismo , Glioma/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Neuroblastoma/metabolismo , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Escherichia coli/genética , Escherichia coli/metabolismo , Glioma/patologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Microscopia Confocal , Proteínas Associadas aos Microtúbulos/genética , Neuritos/metabolismo , Neuroblastoma/patologia , Plasmídeos/genética , Plasmídeos/metabolismo , Ligação Proteica , Ratos , Transfecção , Proteínas tau/metabolismo
8.
J Biochem ; 168(1): 23-32, 2020 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-32044995

RESUMO

Lemur kinase 1 (LMTK1) is a membrane-bound Ser/Thr kinase that is expressed in neurons. There are two splicing variants of LMTK1 with different membrane binding modes, viz., cytosolic LMTK1A that binds to membranes through palmitoylation at the N-terminal cysteines and LMTK1B, an integral membrane protein with transmembrane sequences. We recently reported that LMTK1A regulates axon outgrowth and spine formation in neurons. However, data about LMTK1B are scarce. We analysed the expression and cellular localization of LMTK1B along with its role in axon and spine formation. We found that both LMTK1B and LMTK1A were expressed equally in the cerebral cortex and cerebellum of the mouse brain. Similar to LMTK1A, the wild type of LMTK1B was localized to Rab11-positive pericentrosomal compartment. The kinase negative (kn) mutant of LMTK1B was found to be associated with an increase in the tubular form of endoplasmic reticulum (ER), which was not the case with LMTK1A kn. Furthermore, unlike LMTK1A kn, LMTK1B kn did not stimulate the axon outgrowth and spine formation. These results suggest that while LMTK1A and LMTK1B share a common function in recycling endosomal trafficking at the pericentrosomal compartment, LMTK1B has an additional unique function in vesicle transport in the ER region.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Axônios/fisiologia , Encéfalo/crescimento & desenvolvimento , Crescimento Neuronal/fisiologia , Proteínas Tirosina Quinases/metabolismo , Frações Subcelulares/metabolismo , Animais , Encéfalo/metabolismo , Células Cultivadas , Cricetinae , Retículo Endoplasmático/metabolismo , Endossomos/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Isoformas de Proteínas
9.
J Neurosci ; 38(44): 9459-9467, 2018 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-30381437

RESUMO

Superresolution microscopy (SM) techniques are among the revolutionary methods for molecular and cellular observations in the 21st century. SM techniques overcome optical limitations, and several new observations using SM lead us to expect these techniques to have a large impact on neuroscience in the near future. Several types of SM have been developed, including structured illumination microscopy (SIM), stimulated emission depletion microscopy (STED), and photoactivated localization microscopy (PALM)/stochastic optical reconstruction microscopy (STORM), each with special features. In this Minisymposium, experts in these different types of SM discuss the new structural and functional information about specific important molecules in neuroscience that has been gained with SM. Using these techniques, we have revealed novel mechanisms of endocytosis in nerve growth, fusion pore dynamics, and described quantitative new properties of excitatory and inhibitory synapses. Additional powerful techniques, including single molecule-guided Bayesian localization SM (SIMBA) and expansion microscopy (ExM), alone or combined with super-resolution observation, are also introduced in this session.


Assuntos
Encéfalo/citologia , Microscopia Eletrônica de Transmissão/métodos , Rede Nervosa/citologia , Neurociências/métodos , Imagem Óptica/métodos , Animais , Encéfalo/ultraestrutura , Humanos , Microscopia Eletrônica de Transmissão/tendências , Microscopia de Fluorescência/métodos , Microscopia de Fluorescência/tendências , Rede Nervosa/ultraestrutura , Neurociências/tendências , Imagem Óptica/tendências
10.
iScience ; 4: 190-203, 2018 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-30240740

RESUMO

Neuronal growth cones are essential for nerve growth and regeneration, as well as for the formation and rearrangement of the neural network. To elucidate phosphorylation-dependent signaling pathways and establish useful molecular markers for axon growth and regeneration, we performed a phosphoproteomics study of mammalian growth cones, which identified >30,000 phosphopeptides of ∼1,200 proteins. The phosphorylation sites were highly proline directed and primarily MAPK dependent, owing to the activation of JNK, suggesting that proteins that undergo proline-directed phosphorylation mediate nerve growth in the mammalian brain. Bioinformatics analysis revealed that phosphoproteins were enriched in microtubules and the cortical cytoskeleton. The most frequently phosphorylated site was S96 of GAP-43 (growth-associated protein 43-kDa), a vertebrate-specific protein involved in axon growth. This previously uncharacterized phosphorylation site was JNK dependent. S96 phosphorylation was specifically detected in growing and regenerating axons as the most frequent target of JNK signaling; thus it represents a promising new molecular marker for mammalian axonal growth and regeneration.

11.
Neurochem Int ; 119: 71-76, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-28962923

RESUMO

Growth cones, which are the highly motile tips of extending neuronal processes in developing neurons, have many vesicles. These vesicles are likely essential for the membrane expansion that is required for nerve growth, and probably coordinate with rearrangement of the cytoskeletons. Such mechanisms are poorly understood from molecular and cell biological aspects. Recently, we used superresolution microscopic approaches and described new mechanisms that are involved in the interaction between the vesicles and F-actin in the leading edge of the peripheral domain. Vesicles mainly accumulate in the central domain of growth cones. However, the dynamics of vesicles in each domain, for example, clathrin dependency, are totally distinct from each other. Here, we discuss the diversity of the dynamics of vesicular and related proteins that play different roles in nerve growth.


Assuntos
Citoesqueleto de Actina/metabolismo , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Cones de Crescimento/metabolismo , Animais , Humanos , Neurogênese/fisiologia , Neurônios/metabolismo
12.
J Neurosci ; 37(15): 4046-4064, 2017 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-28275160

RESUMO

Lipid raft domains, where sphingolipids and cholesterol are enriched, concentrate signaling molecules. To examine how signaling protein complexes are clustered in rafts, we focused on the functions of glycoprotein M6a (GPM6a), which is expressed at a high concentration in developing mouse neurons. Using imaging of lipid rafts, we found that GPM6a congregated in rafts in a GPM6a palmitoylation-dependent manner, thereby contributing to lipid raft clustering. In addition, we found that signaling proteins downstream of GPM6a, such as Rufy3, Rap2, and Tiam2/STEF, accumulated in lipid rafts in a GPM6a-dependent manner and were essential for laminin-dependent polarity during neurite formation in neuronal development. In utero RNAi targeting of GPM6a resulted in abnormally polarized neurons with multiple neurites. These results demonstrate that GPM6a induces the clustering of lipid rafts, which supports the raft aggregation of its associated downstream molecules for acceleration of neuronal polarity determination. Therefore, GPM6a acts as a signal transducer that responds to extracellular signals.SIGNIFICANCE STATEMENT Lipid raft domains, where sphingolipids and cholesterol are enriched, concentrate signaling molecules. We focused on glycoprotein M6a (GPM6a), which is expressed at a high concentration in developing neurons. Using imaging of lipid rafts, we found that GPM6a congregated in rafts in a palmitoylation-dependent manner, thereby contributing to lipid raft clustering. In addition, we found that signaling proteins downstream of GPM6a accumulated in lipid rafts in a GPM6a-dependent manner and were essential for laminin-dependent polarity during neurite formation. In utero RNAi targeting of GPM6a resulted in abnormally polarized neurons with multiple neurites. These results demonstrate that GPM6a induces the clustering of lipid rafts, which supports the raft aggregation of its associated downstream molecules for acceleration of polarity determination. Therefore, GPM6a acts as a signal transducer that responds to extracellular signals.


Assuntos
Líquido Extracelular/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Microdomínios da Membrana/genética , Microdomínios da Membrana/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Transdução de Sinais/fisiologia , Sequência de Aminoácidos , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Análise por Conglomerados , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Gravidez
13.
Cell Rep ; 18(9): 2203-2216, 2017 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-28249165

RESUMO

The growth cone is an essential structure for nerve growth. Although its membrane and cytoskeleton are likely to interact coordinately during nerve growth, the mechanisms are unknown due to their close proximity. Here, we used superresolution microscopy to simultaneously observe vesicles and F-actin in growth cones. We identified a novel vesicular generation mechanism that is independent of clathrin and dependent on endophilin-3- and dynamin-1 and that occurs proximal to the leading edge simultaneously with fascin-1-dependent F-actin bundling. In contrast to conventional clathrin-dependent endocytosis, which occurs distal from the leading edge at the basal surfaces of growth cones, this mechanism was distinctly observed at the apical surface using 3D imaging and was involved in mediating axon growth. Reduced endophilin or fascin inhibited this endocytic mechanism. These results suggest that, at the leading edge, vesicles are coordinately generated and transported with actin bundling during nerve growth.


Assuntos
Actinas/metabolismo , Neurogênese/fisiologia , Neurônios/metabolismo , Neurônios/fisiologia , Vesículas Transportadoras/metabolismo , Animais , Axônios/metabolismo , Axônios/fisiologia , Proteínas de Transporte/metabolismo , Células Cultivadas , Clatrina/metabolismo , Citoesqueleto/metabolismo , Dinamina I/metabolismo , Endocitose/fisiologia , Cones de Crescimento/metabolismo , Cones de Crescimento/fisiologia , Camundongos , Proteínas dos Microfilamentos/metabolismo , Microscopia/métodos , Proteínas do Tecido Nervoso/metabolismo
14.
Neurosci Res ; 70(1): 85-90, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21238513

RESUMO

The growth cone plays crucial roles in neural wiring, synapse formation, and axonal regeneration. Continuous rearrangement of cytoskeletal elements and targeting of transported vesicles to the plasma membrane are essential to growth cone motility; however, the proteins directly involved in these processes and their specific functions are not well established. We recently identified 17 proteins as functional marker proteins of the mammalian growth cone and as neuronal growth-associated proteins in rat cortical neurons (nGAPs; Nozumi et al., 2009). To determine whether these 17 proteins are growth cone markers in other neuronal cell types, we examined their expression and function in PC12D cells. We found that all 17 nGAPs were highly concentrated in the growth cones of PC12D cells, and that knockdown of all of them by RNAi reduced or inhibited neurite outgrowth, indicating that all of the 17 nGAPs may be general growth cone markers. Among them, eight proteins were shown to regulate the amount of F-actin in PC12D growth cones. Two of these nGAP that are cytoskeletal proteins, Cap1 and Sept2, increased the mean growth cone area and the mean neurite length by regulating the amount of F-actin; Sept2 also induced filopodial growth. Taken together, our data suggested that some of the nGAPs were generalized markers of the growth cone in multiple neuronal cell types and some of them, such as Cap1 and Sept2, regulated growth cone morphology through rearrangement of F-actin and thereby controlled neurite outgrowth.


Assuntos
Proteínas do Citoesqueleto/biossíntese , Proteínas do Citoesqueleto/fisiologia , Fatores de Crescimento Neural/biossíntese , Fatores de Crescimento Neural/fisiologia , Septinas/biossíntese , Septinas/fisiologia , Actinas/metabolismo , Animais , Diferenciação Celular/genética , Proteínas do Citoesqueleto/genética , Regulação para Baixo/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fatores de Crescimento Neural/genética , Neuritos/fisiologia , Células PC12 , Interferência de RNA/fisiologia , Ratos , Septinas/genética
15.
J Theor Biol ; 266(4): 712-22, 2010 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-20688081

RESUMO

Neuronal growth cones migrate directionally under the control of axon guidance molecules, thereby forming synapses in the developing brain. The signal transduction system by which a growth cone detects surrounding guidance molecules, analyzes the detected signals, and then determines the overall behavior remains undetermined. In this study, we describe a novel stochastic model of this behavior that utilizes multiple sensors on filopodia to respond to guidance molecules. Overall growth cone behavior is determined by using only the concentration gradients of guidance molecules in the immediate vicinity of each sensor. The detected signal at each sensor, which is treated as a vector quantity, is sent to the growth cone center and then integrated to determine axonal growth in the next step by means of a simple vector operation. We compared the results of computer simulations of axonal growth with observations of actual axonal growth from co-culture experiments using olfactory bulb and septum. The probabilistic distributions of axonal growth generated by the computer simulation were consistent with those obtained from the culture experiments, indicating that our model accurately simulates growth cone behavior. We believe that this model will be useful for elucidating the as yet unknown mechanisms responsible for axonal growth in vivo.


Assuntos
Axônios/metabolismo , Cones de Crescimento/metabolismo , Modelos Biológicos , Transdução de Sinais , Algoritmos , Animais , Células Cultivadas , Simulação por Computador , Imageamento Tridimensional , Ratos , Ratos Sprague-Dawley , Processos Estocásticos
16.
Proc Natl Acad Sci U S A ; 106(40): 17211-6, 2009 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-19805073

RESUMO

Identification of proteins in the mammalian growth cone has the potential to advance our understanding of this critical regulator of neuronal growth and formation of neural circuit; however, to date, only one growth cone marker protein, GAP-43, has been reported. Here, we successfully used a proteomic approach to identify 945 proteins present in developing rat forebrain growth cones, including highly abundant, membrane-associated and actin-associated proteins. Almost 100 of the proteins appear to be highly enriched in the growth cone, as determined by quantitative immunostaining, and for 17 proteins, the results of RNAi suggest a role in axon growth. Most of the proteins we identified have not previously been implicated in axon growth and thus their identification presents a significant step forward, providing marker proteins and candidate neuronal growth-associated proteins.


Assuntos
Biomarcadores/análise , Cones de Crescimento/metabolismo , Proteoma/análise , Proteômica/métodos , Proteínas Adaptadoras de Transdução de Sinal/análise , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Animais Recém-Nascidos , Cateninas/análise , Cateninas/genética , Células Cultivadas , Cromatografia Líquida , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Immunoblotting , Imuno-Histoquímica/métodos , Espectrometria de Massas , Microscopia de Fluorescência , Proteínas Associadas aos Microtúbulos/análise , Proteínas Associadas aos Microtúbulos/genética , Fatores de Crescimento Neural/análise , Fatores de Crescimento Neural/genética , Proteínas do Tecido Nervoso/análise , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Neurônios/metabolismo , Prosencéfalo/embriologia , Prosencéfalo/crescimento & desenvolvimento , Prosencéfalo/metabolismo , Proteoma/genética , Interferência de RNA , Ratos , Ratos Transgênicos , Fatores de Tempo
17.
Neurosci Res ; 61(2): 219-24, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18423674

RESUMO

RNA interference (RNAi) is the simplest way of examining gene function by inhibiting expression. However, due to the low rate of introducing short interfering RNA (siRNA) into neurons, it is difficult to discriminate into which neurons that have been successfully introduced. Here, we used neurons from transgenic rats expressing enhanced green fluorescent protein (EGFP), and we simultaneously applied small interfering RNAs (siRNAs) against EGFP and a targeted gene to the EGFP-expressing neurons. EGFP fluorescence and immunoreactivity of the protein were then assessed by immunofluorescence microscopy. Quantitative analysis of the immunofluorescence confirmed that loss of EGFP closely correlates with loss of the target protein. These results indicate that this method can be used in a wider range of the neuroscientific research, especially in genome-wide studies.


Assuntos
Marcação de Genes/métodos , Proteínas de Fluorescência Verde/genética , Neurônios/fisiologia , Interferência de RNA , Animais , Animais Geneticamente Modificados , Córtex Cerebral/fisiologia , Microscopia de Fluorescência , RNA Interferente Pequeno , Ratos
18.
J Cell Sci ; 116(Pt 12): 2577-83, 2003 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-12734400

RESUMO

The insulin receptor tyrosine kinase substrate p53 (IRSp53) links Rac and WAVE2 and has been implicated in lamellipodia protrusion. Recently, however, IRSp53 has been reported to bind to both Cdc42 and Mena to induce filopodia. To shed independent light on IRSp53 function we determined the localisations and dynamics of IRSp53 and WAVE2 in B16 melanoma cells. In cells spread well on a laminin substrate, IRSp53 was localised by antibody labelling at the tips of both lamellipodia and filopodia. The same localisation was observed in living cells with IRSp53 tagged with enhanced green florescence protein (EGFP-IRSp53), but only during protrusion. From the transfection of deletion mutants the N-terminal region of IRSp53, which binds active Rac, was shown to be responsible for its localisation. Although IRSp53 has been reported to regulate filopodia formation with Mena, EGFP-IRSp53 showed the same localisation in MVD7 Ena/VASP (vasodilator stimulated phosphoprotein) family deficient cells. WAVE2 tagged with DsRed1 colocalised with EGFP-IRSp53 at the tips of protruding lamellipodia and filopodia and, in double-transfected cells, the IRSp53 signal in filopodia decreased before that of WAVE2 during retraction. These results suggest an alternative modulatory role for IRSp53 in the extension of both filopodia and lamellipodia, through WAVE2.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas do Citoesqueleto , Proteínas dos Microfilamentos/metabolismo , Pseudópodes/metabolismo , Animais , Moléculas de Adesão Celular/deficiência , Moléculas de Adesão Celular/genética , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proteínas de Fluorescência Verde , Proteínas Luminescentes , Camundongos , Microscopia de Vídeo , Mutação/genética , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Estrutura Terciária de Proteína/genética , Transporte Proteico/fisiologia , Pseudópodes/ultraestrutura , Proteínas Recombinantes de Fusão/metabolismo , Família de Proteínas da Síndrome de Wiskott-Aldrich
19.
J Cell Sci ; 116(Pt 2): 239-46, 2003 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-12482910

RESUMO

The formation and extension of filopodia in response to an extracellular stimulus by guidance cues determine the path of growth cone advance. Actin-filament bundling and actin polymerization at the tips supply the driving force behind the formation and elongation. We tried to clarify how signals in response to extracellular cues are transformed to induce filopodial generation and extension. Observations on the formation process of filopodia at growth cones in the neuroblastoma cell line NG108 showed that WAVE (WASP (Wiskott-Aldrich syndrome protein)-family verprolin homologous protein) isoforms played crucial and distinct roles in this process. WAVE1 was continuously distributed along the leading edge only and was not found in the filopodia. WAVE2 and WAVE3 discretely localized at the initiation sites of microspikes on the leading edge and also concentrated at the tips of protruding filopodia. We further found that WAVE isoforms localized at the filopodial tips through SHD (SCAR homology domain), next to its leucine zipper-like motif. Furthermore, time-lapse observations of filopodial formation in living cells showed that WAVE2 and WAVE3 were continuously expressed at the tips of filopodia during elongation. These results indicate that WAVE2 or WAVE3 may guide the actin bundles into the filopodia and promote actin assembly at the tips.


Assuntos
Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Cones de Crescimento/metabolismo , Proteínas dos Microfilamentos/metabolismo , Sistema Nervoso/embriologia , Pseudópodes/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Compartimento Celular/fisiologia , Imunofluorescência , Modelos Biológicos , Sistema Nervoso/citologia , Sistema Nervoso/metabolismo , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína/fisiologia , Pseudópodes/ultraestrutura , Família de Proteínas da Síndrome de Wiskott-Aldrich
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...