Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
BMC Bioinformatics ; 25(1): 143, 2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38566033

RESUMO

BACKGROUND: Liquid-liquid phase separation (LLPS) by biomolecules plays a central role in various biological phenomena and has garnered significant attention. The behavior of LLPS is strongly influenced by the characteristics of RNAs and environmental factors such as pH and temperature, as well as the properties of proteins. Recently, several databases recording LLPS-related biomolecules have been established, and prediction models of LLPS-related phenomena have been explored using these databases. However, a prediction model that concurrently considers proteins, RNAs, and experimental conditions has not been developed due to the limited information available from individual experiments in public databases. RESULTS: To address this challenge, we have constructed a new dataset, RNAPSEC, which serves each experiment as a data point. This dataset was accomplished by manually collecting data from public literature. Utilizing RNAPSEC, we developed two prediction models that consider a protein, RNA, and experimental conditions. The first model can predict the LLPS behavior of a protein and RNA under given experimental conditions. The second model can predict the required conditions for a given protein and RNA to undergo LLPS. CONCLUSIONS: RNAPSEC and these prediction models are expected to accelerate our understanding of the roles of proteins, RNAs, and environmental factors in LLPS.


Assuntos
Proteínas Intrinsicamente Desordenadas , RNA , RNA/genética , Proteínas Intrinsicamente Desordenadas/química
2.
Neurosci Lett ; 810: 137317, 2023 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-37286070

RESUMO

Fragile X syndrome (FXS) is a developmental disorder characterized by intellectual disability and autistic-like behaviors. These symptoms are supposed to result from dysregulated translation in pre- and postsynapses, resulting in aberrant synaptic plasticity. Although most drug development research on FXS has focused on aberrant postsynaptic functions by excess translation in postsynapses, the effect of drug candidates on FXS in presynaptic release is largely unclear. In this report, we developed a novel assay system using neuron ball culture with beads to induce presynapse formation, allowing for the analysis of presynaptic phenotypes, including presynaptic release. Metformin, which is shown to rescue core phenotypes in FXS mouse model by normalizing dysregulated translation, ameliorated the exaggerated presynaptic release of neurons of FXS model mouse using this assay system. Furthermore, metformin suppressed the excess accumulation of the active zone protein Munc18-1, which is supposed to be locally translated in presynapses. These results suggest that metformin rescues both postsynaptic and presynaptic phenotypes by inhibiting excess translation in FXS neurons.


Assuntos
Síndrome do Cromossomo X Frágil , Animais , Camundongos , Modelos Animais de Doenças , Proteína do X Frágil da Deficiência Intelectual/genética , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/tratamento farmacológico , Síndrome do Cromossomo X Frágil/genética , Síndrome do Cromossomo X Frágil/metabolismo , Camundongos Knockout , Neurônios/metabolismo , Sinapses/fisiologia
3.
Mol Brain ; 13(1): 167, 2020 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-33323119

RESUMO

Corticogenesis is one of the most critical and complicated processes during embryonic brain development. Any slight impairment in corticogenesis could cause neurodevelopmental disorders such as Fragile X syndrome (FXS), of which symptoms contain intellectual disability (ID) and autism spectrum disorder (ASD). Fragile X mental retardation protein (FMRP), an RNA-binding protein responsible for FXS, shows strong expression in neural stem/precursor cells (NPCs) during corticogenesis, although its function during brain development remains largely unknown. In this study, we attempted to identify the FMRP target mRNAs in the cortical primordium using RNA immunoprecipitation sequencing analysis in the mouse embryonic brain. We identified 865 candidate genes as targets of FMRP involving 126 and 118 genes overlapped with ID and ASD-associated genes, respectively. These overlapped genes were enriched with those related to chromatin/chromosome organization and histone modifications, suggesting the involvement of FMRP in epigenetic regulation. We further identified a common set of 17 FMRP "core" target genes involved in neurogenesis/FXS/ID/ASD, containing factors associated with Ras/mitogen-activated protein kinase, Wnt/ß-catenin, and mammalian target of rapamycin (mTOR) pathways. We indeed showed overactivation of mTOR signaling via an increase in mTOR phosphorylation in the Fmr1 knockout (Fmr1 KO) neocortex. Our results provide further insight into the critical roles of FMRP in the developing brain, where dysfunction of FMRP may influence the regulation of its mRNA targets affecting signaling pathways and epigenetic modifications.


Assuntos
Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Organogênese , Serina-Treonina Quinases TOR/metabolismo , Via de Sinalização Wnt , Proteínas ras/metabolismo , Animais , Transtorno do Espectro Autista/genética , Embrião de Mamíferos/metabolismo , Proteína do X Frágil da Deficiência Intelectual/genética , Regulação da Expressão Gênica no Desenvolvimento , Deficiência Intelectual/genética , Masculino , Camundongos Endogâmicos C57BL , Neurogênese/genética , Organogênese/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
4.
Biomolecules ; 10(5)2020 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-32344905

RESUMO

During neural development, growth cones, very motile compartments of tips of axons, lead axonal extension to the correct targets. Subsequently, presynapses, another axonal compartment with vigorous trafficking of synaptic vesicles, emerge to form functional synapses with postsynapses. In response to extracellular stimuli, the immediate supply of proteins by local translation within these two axonal compartments far from cell bodies confers high motility of growth cones and active vesicle trafficking in presynapses. Although local translation in growth cones and presynapses occurs at a very low level compared with cell bodies and even dendrites, recent progress in omics and visualization techniques with subcellular fractionation of these compartments has revealed the actual situation of local translation within these two axonal compartments. Here, the increasing evidence for local protein synthesis in growth cones and presynapses for axonal and synaptic functions has been reviewed. Furthermore, the mechanisms regulating local translation in these two compartments and pathophysiological conditions caused by dysregulated local translation are highlighted.


Assuntos
Cones de Crescimento/metabolismo , Terminações Pré-Sinápticas/metabolismo , Biossíntese de Proteínas , Animais , Cones de Crescimento/fisiologia , Humanos , Terminações Pré-Sinápticas/fisiologia , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo
5.
Artigo em Inglês | MEDLINE | ID: mdl-32184710

RESUMO

Fragile X mental retardation protein (FMRP) is an RNA-binding protein that regulates local translation in dendrites and spines for synaptic plasticity. In axons, FMRP is implicated in axonal extension and axon guidance. We previously demonstrated the involvement of FMRP in growth cone collapse via a translation-dependent response to Semaphorin-3A (Sema3A), a repulsive axon guidance factor. In the case of attractive axon guidance factors, RNA-binding proteins such as zipcode binding protein 1 (ZBP1) accumulate towards the stimulated side of growth cones for local translation. However, it remains unclear how Sema3A effects FMRP localization in growth cones. Here, we show that levels of FMRP in growth cones of hippocampal neurons decreased after Sema3A stimulation. This decrease in FMRP was suppressed by the ubiquitin-activating enzyme E1 enzyme inhibitor PYR-41 and proteasome inhibitor MG132, suggesting that the ubiquitin-proteasome pathway is involved in Sema3A-induced FMRP degradation in growth cones. Moreover, the E1 enzyme or proteasome inhibitor suppressed Sema3A-induced increases in microtubule-associated protein 1B (MAP1B) in growth cones, suggesting that the ubiquitin-proteasome pathway promotes local translation of MAP1B, whose translation is mediated by FMRP. These inhibitors also blocked the Sema3A-induced growth cone collapse. Collectively, our results suggest that Sema3A promotes degradation of FMRP in growth cones through the ubiquitin-proteasome pathway, leading to growth cone collapse via local translation of MAP1B. These findings reveal a new mechanism of axon guidance regulation: degradation of the translational suppressor FMRP via the ubiquitin-proteasome pathway.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/metabolismo , Cones de Crescimento/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Semaforina-3A/metabolismo , Transdução de Sinais/fisiologia , Ubiquitina/metabolismo , Animais , Células Cultivadas , Proteína do X Frágil da Deficiência Intelectual/análise , Cones de Crescimento/química , Hipocampo/química , Hipocampo/metabolismo , Camundongos , Complexo de Endopeptidases do Proteassoma/análise , Semaforina-3A/análise , Ubiquitina/análise
6.
BMJ Case Rep ; 12(8)2019 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-31451453

RESUMO

This report discusses a case of superior mesenteric artery (SMA) syndrome in a previously healthy 15-year-old boy with no weight loss or other common risk factors. The patient presented to the emergency department with acute bilious vomiting and epigastric pain after acute consumption of a meal and excessive quantities of water. The patient was diagnosed with SMA syndrome based on the findings of contrasted CT of the abdomen. In early puberty, boys have a significant increase in lean body mass and a concomitant loss of adipose tissues. These pubertal changes lead to a narrowing of the aortomesenteric space. The acute consumption of food and water caused a transient obstruction at the already-narrowed space, which resulted in the manifestation of SMA syndrome. This case demonstrates that pubertal growth spurt is a risk factor for SMA syndrome, and acute excessive ingestion can trigger SMA syndrome among those in puberty.


Assuntos
Bulimia/complicações , Puberdade/fisiologia , Síndrome da Artéria Mesentérica Superior , Tomografia Computadorizada por Raios X/métodos , Dor Abdominal/diagnóstico , Dor Abdominal/etiologia , Adolescente , Bulimia/fisiopatologia , Diagnóstico Diferencial , Humanos , Masculino , Intensificação de Imagem Radiográfica/métodos , Radiografia Abdominal/métodos , Síndrome da Artéria Mesentérica Superior/diagnóstico , Síndrome da Artéria Mesentérica Superior/etiologia , Síndrome da Artéria Mesentérica Superior/fisiopatologia , Síndrome da Artéria Mesentérica Superior/prevenção & controle , Vômito/diagnóstico , Vômito/etiologia
7.
J Vis Exp ; (150)2019 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-31424445

RESUMO

During neuronal development, synapse formation is an important step to establish neural circuits. To form synapses, synaptic proteins must be supplied in appropriate order by transport from cell bodies and/or local translation in immature synapses. However, it is not fully understood how synaptic proteins accumulate in synapses in proper order. Here, we present a novel method to analyze presynaptic formation by using the combination of neuron ball culture with beads to induce presynapse formation. Neuron balls that is neuronal cell aggregates provide axonal sheets far from cell bodies and dendrites, so that weak fluorescent signals of presynapses can be detected by avoiding overwhelming signals of cell bodies. As beads to trigger presynapse formation, we use beads conjugated with leucine-rich repeat transmembrane neuronal 2 (LRRTM2), an excitatory presynaptic organizer. Using this method, we demonstrated that vesicular glutamate transporter 1 (vGlut1), a synaptic vesicle protein, accumulated in presynapses faster than Munc18-1, an active zone protein. Munc18-1 accumulated translation-dependently in presynapse even after removing cell bodies. This finding indicates the Munc18-1 accumulation by local translation in axons, not transport from cell bodies. In conclusion, this method is suitable to analyze accumulation of synaptic proteins in presynapses and source of synaptic proteins. As neuron ball culture is simple and it is not necessary to use special apparatus, this method could be applicable to other experimental platforms.


Assuntos
Neurônios/fisiologia , Sinapses/fisiologia , Animais , Axônios/metabolismo , Células Cultivadas , Camundongos , Proteínas Munc18/metabolismo , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo
8.
Neurosci Res ; 146: 36-47, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30240639

RESUMO

Fragile X mental retardation protein (FMRP), a causative gene (FMR1) product of Fragile X syndrome (FXS), is an RNA-binding protein to regulate local protein synthesis in dendrites for postsynaptic functions. However, involvement of FMRP in local protein synthesis in axons for presynaptic functions remains unclear. Here we investigated role of FMRP in local translation of the active zone protein Munc18-1 during presynapse formation. We found that leucine-rich repeat transmembrane neuronal 2 (LRRTM2)-conjugated beads, which promotes synchronized presynapse formation, induced simultaneous accumulation of FMRP and Munc18-1 in presynapses of axons of mouse cortical neurons in neuronal cell aggregate culture. The LRRTM2-induced accumulation of Munc18-1 in presynapses was observed in axons protein-synthesis-dependently, even physically separated from cell bodies. The accumulation of Munc18-1 was enhanced in Fmr1-knockout (KO) axons as compared to wild type (WT), suggesting FMRP-regulated suppression for local translation of Munc18-1 in axons during presynapse formation. Using naturally formed synapses of dissociated culture, structured illumination microscope revealed that accumulation of Munc18-1 puncta in Fmr1-KO neurons increased significantly at 19 days in vitro, as compared to WT. Our findings lead the possibility that excessive accumulation of Munc18-1 in presynapses at early stage of synaptic development in Fmr1-KO neurons may have a critical role in impaired presynaptic functions in FXS.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/fisiologia , Proteínas de Membrana/metabolismo , Proteínas Munc18/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/fisiologia , Sinapses/metabolismo , Animais , Axônios/metabolismo , Córtex Cerebral , Dendritos/metabolismo , Síndrome do Cromossomo X Frágil , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
9.
Cell Adh Migr ; 10(6): 627-640, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27392015

RESUMO

Numerous cell adhesion molecules, extracellular matrix proteins and axon guidance molecules participate in neuronal network formation through local effects at axo-dendritic, axo-axonic or dendro-dendritic contact sites. In contrast, neurotrophins and their receptors play crucial roles in neural wiring by sending retrograde signals to remote cell bodies. Semaphorin 3A (Sema3A), a prototype of secreted type 3 semaphorins, is implicated in axon repulsion, dendritic branching and synapse formation via binding protein neuropilin-1 (NRP1) and the signal transducing protein PlexinAs (PlexAs) complex. This review focuses on Sema3A retrograde signaling that regulates dendritic localization of AMPA-type glutamate receptor GluA2 and dendritic patterning. This signaling is elicited by activation of NRP1 in growth cones and is propagated to cell bodies by dynein-dependent retrograde axonal transport of PlexAs. It also requires interaction between PlexAs and a high-affinity receptor for nerve growth factor, toropomyosin receptor kinase A. We propose a control mechanism by which retrograde Sema3A signaling regulates the glutamate receptor localization through trafficking of cis-interacting PlexAs with GluA2 along dendrites; this remote signaling may be an alternative mechanism to local adhesive contacts for neural network formation.


Assuntos
Dendritos/metabolismo , Espaço Intracelular/metabolismo , Semaforina-3A/metabolismo , Transdução de Sinais , Animais , Axônios/metabolismo , Humanos , Sinapses/metabolismo
10.
Nat Commun ; 5: 3424, 2014 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-24599038

RESUMO

The dendritic targeting of neurotransmitter receptors is vital for dendritic development and function. However, how such localization is established remains unclear. Here we show that semaphorin 3A (Sema3A) signalling at the axonal growth cone is propagated towards the cell body by retrograde axonal transport and drives AMPA receptor GluA2 to the distal dendrites, which regulates dendritic development. Sema3A enhances glutamate receptor interacting protein 1-dependent localization of GluA2 in dendrites, which is blocked by knockdown of cytoplasmic dynein heavy chain. PlexinA (PlexA), a receptor component for Sema3A, interacts with GluA2 at the immunoglobulin-like Plexin-transcription-factor domain (PlexA-IPT) in somatodendritic regions. Overexpression of PlexA-IPT suppresses dendritic localization of GluA2 and induces aproximal bifurcation phenotype in the apical dendrites of CA1 hippocampal neurons. Thus, we propose a control mechanism by which retrograde Sema3A signalling regulates the glutamate receptor localization through trafficking of cis-interacting PlexA with GluA2 along dendrites.


Assuntos
Dendritos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores de AMPA/metabolismo , Receptores de Superfície Celular/metabolismo , Semaforina-3A/metabolismo , Transdução de Sinais , Animais , Axônios/metabolismo , Western Blotting , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/embriologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Células Cultivadas , Feminino , Transferência Ressonante de Energia de Fluorescência , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Microscopia Confocal , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Neurônios/metabolismo , Interferência de RNA , Ratos , Ratos Wistar , Receptores de AMPA/genética , Receptores de Superfície Celular/genética , Semaforina-3A/genética , Imagem com Lapso de Tempo/métodos
11.
Dev Neurobiol ; 74(3): 397-406, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23897634

RESUMO

There is increasing evidence that localized mRNAs in axons and growth cones play an important role in axon extension and pathfinding via local translation. A few studies have revealed the presence of microRNAs (miRNAs) in axons, which may control local protein synthesis during axon development. However, so far, there has been no attempt to screen for axon-enriched miRNAs and to validate their possible localization to growth cones of developing axons from neurons of the central nervous system. In this study, the localization of miRNAs in axons and growth cones in cortical neurons was examined using a "neuron ball" culture method that is suitable to prepare axonal miRNAs with high yield and purity. Axonal miRNAs prepared from the neuron ball cultures of mouse cortical neurons were analyzed by quantitative real-time RT-PCR. Among 375 miRNAs that were analyzed, 105 miRNAs were detected in axons, and six miRNAs were significantly enriched in axonal fractions when compared with cell body fractions. Fluorescence in situ hybridization revealed that two axon-enriched miRNAs, miR-181a-1* and miR-532, localized as distinct granules in distal axons and growth cones. The association of these miRNAs with the RNA-induced silencing complex further supported their function to regulate mRNA levels or translation in the brain. These results suggest a mechanism to localize specific miRNAs to distal axons and growth cones, where they could be involved in local mRNA regulation. These findings provide new insight into the presence of axonal miRNAs and motivate further analysis of their function in local protein synthesis underlying axon guidance.


Assuntos
Axônios/metabolismo , Córtex Cerebral/metabolismo , Cones de Crescimento/metabolismo , Hipocampo/metabolismo , MicroRNAs/metabolismo , Neurônios/metabolismo , Animais , Técnicas de Cultura de Células , Células Cultivadas , Grânulos Citoplasmáticos/metabolismo , Imunoprecipitação , Hibridização in Situ Fluorescente , Camundongos , Complexo de Inativação Induzido por RNA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Dev Neurobiol ; 73(3): 230-46, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23008267

RESUMO

Nerve growth cones contain mRNA and its translational machinery and thereby synthesize protein locally. The regulatory mechanisms in the growth cone, however, remain largely unknown. We previously found that the calcium entry-induced increase of phosphorylation of eukaryotic elongation factor-2 (eEF2), a key component of mRNA translation, within growth cones showed growth arrest of neurites. Because dephosphorylated eEF2 and phosphorylated eEF2 are known to promote and inhibit mRNA translation, respectively, the data led to the hypothesis that eEF2-mediating mRNA translation may regulate neurite outgrowth. Here, we validated the hypothesis by using a chromophore-assisted light inactivation (CALI) technique to examine the roles of localized eEF2 and eEF2 kinase (EF2K), a specific calcium calmodulin-dependent enzyme for eEF2 phosphorylation, in advancing growth cones of cultured chick dorsal root ganglion (DRG) neurons. The phosphorylated eEF2 was weakly distributed in advancing growth cones, whereas eEF2 phosphorylation was increased by extracellular adenosine triphosphate (ATP)-evoked calcium transient through P2 purinoceptors in growth cones and resulted in growth arrest of neurites. The increase of eEF2 phosphorylation within growth cones by inhibition of protein phosphatase 2A known to dephosphorylate eEF2 also showed growth arrest of neurites. CALI of eEF2 within growth cones resulted in retardation of neurite outgrowth, whereas CALI of EF2K enhanced neurite outgrowth temporally. Moreover, CALI of EF2K abolished the ATP-induced retardation of neurite outgrowth. These findings suggest that an eEF2 phosphorylation state localized to the growth cone regulates neurite outgrowth.


Assuntos
Fator de Iniciação 2 em Eucariotos/metabolismo , Cones de Crescimento/metabolismo , Neuritos/metabolismo , Neurogênese/fisiologia , Animais , Western Blotting , Embrião de Galinha , Quinase do Fator 2 de Elongação/metabolismo , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Fosforilação
13.
J Neurosci ; 32(35): 11905-18, 2012 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-22933777

RESUMO

Semaphorin3A (Sema3A) exerts a wide variety of biological functions by regulating reorganization of actin and tubulin cytoskeletal proteins through signaling pathways including sequential phosphorylation of collapsin response mediator protein 1 (CRMP1) and CRMP2 by cyclin-dependent kinase-5 and glycogen synthase kinase-3ß (GSK3ß). To delineate how GSK3ß mediates Sema3A signaling, we here determined the substrates of GSK3ß involved. Introduction of either GSK3ß mutants, GSK3ß-R96A, L128A, or K85M into chick dorsal root ganglion (DRG) neurons suppressed Sema3A-induced growth cone collapse, thereby suggesting that unprimed as well as primed substrates are involved in Sema3A signaling. Axin-1, a key player in Wnt signaling, is an unprimed substrate of GSK3ß. The phosphorylation of Axin-1 by GSK3ß accelerates the association of Axin-1 with ß-catenin. Immunocytochemical studies revealed that Sema3A induced an increase in the intensity levels of ß-catenin in the DRG growth cones. Axin-1 siRNA knockdown suppressed Sema3A-induced growth cone collapse. The reintroduction of RNAi-resistant Axin-1 (rAxin-1)-wt rescued the responsiveness to Sema3A, while that of nonphosphorylated mutants, rAxin S322A/S326A/S330A and T485A/S490A/S497A, did not. Sema3A also enhanced the colocalization of GSK3ß, Axin-1, and ß-catenin in the growth cones. The increase of ß-catenin in the growth cones was suppressed by the siRNA knockdown of Axin-1. Furthermore, either Axin-1 or ß-catenin RNAi knockdown suppressed the internalization of Sema3A. These results suggest that Sema3A induces the formation of GSK3ß/Axin-1/ß-catenin complex, which regulates signaling cascade of Sema3A via an endocytotic mechanism. This finding should provide clue for understanding of mechanisms of a wide variety of biological functions of Sema3A.


Assuntos
Proteína Axina/fisiologia , Quinase 3 da Glicogênio Sintase/fisiologia , Semaforina-3A/fisiologia , Transdução de Sinais/fisiologia , beta Catenina/fisiologia , Animais , Proteína Axina/genética , Células Cultivadas , Embrião de Galinha , Endocitose/genética , Feminino , Gânglios Espinais/metabolismo , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Humanos , Masculino , Camundongos , Mutagênese Sítio-Dirigida , Técnicas de Cultura de Órgãos , Fosforilação/genética , Ratos , Semaforina-3A/genética , Transdução de Sinais/genética , Especificidade por Substrato/genética , beta Catenina/genética
14.
Expert Opin Ther Targets ; 16(9): 933-44, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22834859

RESUMO

INTRODUCTION: The semaphorins were initially described as axon guidance molecules that play important roles in the development of nervous system. Recent studies suggest that semaphorins and their receptors also exert such diverse functions as immune response, control of vascular endothelial cell motility and invasion of many types of cancer cells. AREAS COVERED: The available results concerning application of class 3 semaphorins and their inhibitors for the treatment in animal disease models. EXPERT OPINION: Because semaphorins are now recognized as key players in immune, cardiovascular, bone metabolism and neurological system, semaphorins and their receptors are most promising therapeutic targets for various disease states. As semaphorins exert their diverse or even opposing activities in vivo, more elaborate studies on pathophysiology and signal transduction mechanisms of semaphorins are required.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Semaforinas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Animais , Humanos , Modelos Moleculares , Conformação Proteica , Semaforinas/química , Semaforinas/genética
15.
Mol Brain ; 4: 40, 2011 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-22051374

RESUMO

BACKGROUND: The small non-coding microRNAs play an important role in development by regulating protein translation, but their involvement in axon guidance is unknown. Here, we investigated the role of microRNA-134 (miR-134) in chemotropic guidance of nerve growth cones. RESULTS: We found that miR-134 is highly expressed in the neural tube of Xenopus embryos. Fluorescent in situ hybridization also showed that miR-134 is enriched in the growth cones of Xenopus spinal neurons in culture. Importantly, overexpression of miR-134 mimics or antisense inhibitors blocked protein synthesis (PS)-dependent attractive responses of Xenopus growth cones to a gradient of brain-derived neurotrophic factor (BDNF). However, miR-134 mimics or inhibitors had no effect on PS-independent bidirectional responses of Xenopus growth cones to bone morphogenic protein 7 (BMP7). Our data further showed that Xenopus LIM kinase 1 (Xlimk1) mRNA is a potential target of miR-134 regulation. CONCLUSIONS: These findings demonstrate a role for miR-134 in translation-dependent guidance of nerve growth cones. Different guidance cues may act through distinct signaling pathways to elicit PS-dependent and -independent mechanisms to steer growth cones in response to a wide array of spatiotemporal cues during development.


Assuntos
Quimiotaxia , Cones de Crescimento/metabolismo , MicroRNAs/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Quimiotaxia/efeitos dos fármacos , Imunofluorescência , Regulação da Expressão Gênica/efeitos dos fármacos , Cones de Crescimento/efeitos dos fármacos , Cones de Crescimento/enzimologia , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Modelos Biológicos , Oligonucleotídeos Antissenso/farmacologia , Fosforilação/efeitos dos fármacos , Xenopus , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo
16.
J Neurosci ; 31(5): 1773-9, 2011 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-21289187

RESUMO

Collapsin response mediator protein 5 (CRMP5) is one of the CRMP members that expresses abundantly in the developing brain. To examine the in vivo function of CRMP5, we generated crmp5-deficient (crmp5(-/-)) mice. Anti-calbindin immunofluorescence studies of crmp5(-/-) mice revealed aberrant dendrite morphology; specifically, a decrease in the size of soma and diameter of primary dendrite of the cerebellar Purkinje cells at postnatal day 21 (P21) and P28, but not at P14. Coincidentally, CRMP5 is detected in Purkinje cells at P21 and P28 from crmp5(+/-) mice. In cerebellar slices of crmp5(-/-) mice, the induction of long-term depression of excitatory synaptic transmission between parallel fibers and Purkinje cells was deficient. Given that brain-derived neurotrophic factor (BDNF) plays major roles in dendritic development, we tried to elucidate the possible roles of CRMP5 in BDNF signaling. The effect of BDNF to induce dendritic branching was markedly attenuated in cultured crmp5(-/-) neurons. Furthermore, CRMP5 was tyrosine phosphorylated when coexpressed with neurotrophic tyrosine kinase receptor type 2 (TrkB), a receptor for BDNF, in HEK293T cells. These findings suggest that CRMP5 is involved in the development, maintenance and synaptic plasticity of Purkinje cells.


Assuntos
Amidoidrolases/metabolismo , Dendritos/metabolismo , Depressão Sináptica de Longo Prazo/fisiologia , Plasticidade Neuronal/fisiologia , Células de Purkinje/metabolismo , Receptor trkB/metabolismo , Transmissão Sináptica/fisiologia , Amidoidrolases/deficiência , Amidoidrolases/genética , Animais , Animais Recém-Nascidos , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Células Cultivadas , Cerebelo/metabolismo , Dendritos/efeitos dos fármacos , Células HEK293 , Humanos , Hidrolases , Imuno-Histoquímica , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Associadas aos Microtúbulos , Plasticidade Neuronal/efeitos dos fármacos , Reação em Cadeia da Polimerase , Células de Purkinje/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos
17.
J Neurosci ; 30(28): 9349-58, 2010 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-20631164

RESUMO

The localization of specific mRNAs and their local translation in growth cones of developing axons has been shown to play an important mechanism to regulate growth cone turning responses to attractive or repulsive cues. However, the mechanism whereby local translation and growth cone turning may be controlled by specific mRNA-binding proteins is unknown. Here we demonstrate that brain-derived neurotrophic factor (BDNF) signals the Src-dependent phosphorylation of the beta-actin mRNA zipcode binding protein 1 (ZBP1), which is necessary for beta-actin synthesis and growth cone turning. We raised a phospho-specific ZBP1 antibody to Tyr396, which is a Src phosphorylation site, and immunofluorescence revealed BDNF-induced phosphorylation of ZBP1 within growth cones. The BDNF-induced increase in fluorescent signal of a green fluorescent protein translation reporter with the 3' untranslated region of beta-actin was attenuated with the Src family kinase-specific inhibitor PP2 [4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine]. Furthermore, a nonphosphorylatable mutant, ZBP1 Y396F, suppressed the BDNF-induced and protein synthesis-dependent increase in beta-actin localization in growth cones. Last, the ZBP1 Y396F mutant blocked BDNF-induced attractive growth cone turning. These results indicate that phosphorylation of ZBP1 at Tyr396 within growth cones has a critical role to regulate local protein synthesis and growth cone turning. Our findings provide new insight into how the regulated phosphorylation of mRNA-binding proteins influences local translation underlying growth cone motility and axon guidance.


Assuntos
Actinas/biossíntese , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Cones de Crescimento/metabolismo , Neurônios/metabolismo , Proteínas de Ligação a RNA/metabolismo , Actinas/genética , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Imunofluorescência , Cones de Crescimento/efeitos dos fármacos , Processamento de Imagem Assistida por Computador , Hibridização in Situ Fluorescente , Neurônios/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Biossíntese de Proteínas/efeitos dos fármacos , Biossíntese de Proteínas/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Xenopus
18.
Artigo em Inglês | MEDLINE | ID: mdl-19826618

RESUMO

Fragile X syndrome, the most frequent form of familial mental retardation, is caused by mutation of the Fmr1 gene. Fmr1 encodes the fragile X mental retardation protein (FMRP), an mRNA binding protein regulating local, postsynaptic mRNA translation along dendrites necessary for long-term synaptic plasticity. However, recent studies on FMRP localization in axons and growth cones suggest a possible function in the regulation of local protein synthesis needed for axon guidance. Here, we have demonstrated that FMRP is involved in axonal and growth cone responses induced by the axon guidance factor, Semaphorin-3A (Sema3A). In cultured hippocampal neurons from wild type mice, Sema3A-induced growth cone collapse was protein synthesis-dependent. In contrast, Sema3A-induced growth cone collapse was attenuated in Fmr1 knock-out (KO) neurons and insensitive to protein synthesis inhibitors, suggesting that FMRP is involved in protein synthesis-dependent growth cone collapse. Sema3A increased phosphorylation of eukaryotic initiation factor 4E (eIF4E), an indicator of local translation, in distal axons and growth cones of wild type, but not Fmr1 KO neurons. Furthermore, Sema3A rapidly induced a protein synthesis-dependent increase in levels of microtubule associated protein 1B (MAP1B) in distal axons of wild type neurons, but this response was attenuated in Fmr1 KO neurons. These results suggest a possible role of FMRP to regulate local translation and axonal protein localization in response to Sema3A. This study reveals a new link between FMRP and semaphorin signaling in vitro, and raises the possibility that FMRP may have a critical role in semaphorin signaling in axon guidance during brain development.

19.
J Biol Chem ; 284(40): 27393-401, 2009 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-19652227

RESUMO

Collapsin response mediator protein 2 (CRMP2) is an intracellular protein that mediates signaling of Semaphorin3A (Sema3A), a repulsive axon guidance molecule. Fyn, a Src-type tyrosine kinase, is involved in the Sema3A signaling. However, the relationship between CRMP2 and Fyn in this signaling pathway is still unknown. In our research, we demonstrated that Fyn phosphorylated CRMP2 at Tyr(32) residues in HEK293T cells. Immunohistochemical analysis using a phospho-specific antibody at Tyr(32) of CRMP showed that Tyr(32)-phosphorylated CRMP was abundant in the nervous system, including dorsal root ganglion neurons, the molecular and Purkinje cell layer of adult cerebellum, and hippocampal fimbria. Overexpression of a nonphosphorylated mutant (Tyr(32) to Phe(32)) of CRMP2 in dorsal root ganglion neurons interfered with Sema3A-induced growth cone collapse response. These results suggest that Fyn-dependent phosphorylation of CRMP2 at Tyr(32) is involved in Sema3A signaling.


Assuntos
Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Fosfotirosina/metabolismo , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Semaforina-3A/metabolismo , Transdução de Sinais , Sequência de Aminoácidos , Animais , Anticorpos/imunologia , Especificidade de Anticorpos , Linhagem Celular , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Dados de Sequência Molecular , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/imunologia , Fosforilação , Ratos
20.
Nat Methods ; 6(5): 347-9, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19349979
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA