Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Exp Biol ; 222(Pt 19)2019 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-31488622

RESUMO

The Sap47 gene of Drosophila melanogaster encodes a highly abundant 47 kDa synaptic vesicle-associated protein. Sap47 null mutants show defects in synaptic plasticity and larval olfactory associative learning but the molecular function of Sap47 at the synapse is unknown. We demonstrate that Sap47 modulates the phosphorylation of another highly abundant conserved presynaptic protein, synapsin. Site-specific phosphorylation of Drosophila synapsin has repeatedly been shown to be important for behavioural plasticity but it was not known where these phospho-synapsin isoforms are localized in the brain. Here, we report the distribution of serine-6-phosphorylated synapsin in the adult brain and show that it is highly enriched in rings of synapses in the ellipsoid body and in large synapses near the lateral triangle. The effects of knockout of Sap47 or synapsin on olfactory associative learning/memory support the hypothesis that both proteins operate in the same molecular pathway. We therefore asked if this might also be true for other aspects of their function. We show that knockout of Sap47 but not synapsin reduces lifespan, whereas knockout of Sap47 and synapsin, either individually or together, affects climbing proficiency, as well as plasticity in circadian rhythms and sleep. Furthermore, electrophysiological assessment of synaptic properties at the larval neuromuscular junction (NMJ) reveals increased spontaneous synaptic vesicle fusion and reduced paired pulse facilitation in Sap47 and synapsin single and double mutants. Our results imply that Sap47 and synapsin cooperate non-uniformly in the control of synaptic properties in different behaviourally relevant neuronal networks of the fruitfly.


Assuntos
Comportamento Animal/fisiologia , Proteínas de Drosophila/genética , Drosophila melanogaster/fisiologia , Locomoção/genética , Longevidade/genética , Mutação/genética , Proteínas do Tecido Nervoso/genética , Plasticidade Neuronal/genética , Sinapsinas/metabolismo , Animais , Encéfalo/metabolismo , Ritmo Circadiano/fisiologia , Drosophila melanogaster/genética , Larva/metabolismo , Junção Neuromuscular/metabolismo , Fosforilação , Fosfosserina/metabolismo , Isoformas de Proteínas/metabolismo , Sinapsinas/genética
2.
Mov Disord ; 28(3): 392-5, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23401086

RESUMO

BACKGROUND: Emerging evidence has highlighted the pivotal role of the immune system in neurodegenerative diseases. This study investigated the impact of progressive neurodegeneration on the differentiation and development of hematopoietic stem cells in the peripheral blood of Parkinson's patients. METHODS: A colony-forming cell assay was established to study hematopoietic stem cells from venous blood of Parkinson's patients, and flow cytometry was used to analyze the expression of chemokine receptors on monocytes. RESULTS: We demonstrate that there is strong upregulation in the percentage of monocyte precursors in the peripheral blood of Parkinson's patients and asymptomatic high-risk individuals. We identify the receptor CCR2 as undergoing strong upregulation on the surface of classical monocytes in Parkinson's patients. CONCLUSIONS: The association between blood cell development and progressive cell death in the brain of Parkinson's patients should be further investigated as a potential dynamic biomarker and indicator of disease progression.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Monócitos/fisiologia , Doença de Parkinson/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Citometria de Fluxo , Células-Tronco Hematopoéticas/imunologia , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Masculino , Pessoa de Meia-Idade , Monócitos/imunologia , Doença de Parkinson/genética , Proteínas Serina-Treonina Quinases/genética , Receptores CCR2/metabolismo , Estatísticas não Paramétricas
3.
J Neurosci ; 31(9): 3508-18, 2011 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-21368063

RESUMO

The synapse-associated protein of 47 kDa (SAP47) is a member of a phylogenetically conserved gene family of hitherto unknown function. In Drosophila, SAP47 is encoded by a single gene (Sap47) and is expressed throughout all synaptic regions of the wild-type larval brain; specifically, electron microscopy reveals anti-SAP47 immunogold labeling within 30 nm of presynaptic vesicles. To analyze SAP47 function, we used the viable and fertile deletion mutant Sap47(156), which suffers from a 1.7 kb deletion in the regulatory region and the first exon. SAP47 cannot be detected by either immunoblotting or immunohistochemistry in Sap47(156) mutants. These mutants exhibit normal sensory detection of odorants and tastants as well as normal motor performance and basic neurotransmission at the neuromuscular junction. However, short-term plasticity at this synapse is distorted. Interestingly, Sap47(156) mutant larvae also show a 50% reduction in odorant-tastant associative learning ability; a similar associative impairment is observed in a second deletion allele (Sap47(201)) and upon reduction of SAP47 levels using RNA interference. In turn, transgenically restoring SAP47 in Sap47(156) mutant larvae rescues the defect in associative function. This report thus is the first to suggest a function for SAP47. It specifically argues that SAP47 is required for proper behavioral and synaptic plasticity in flies-and prompts the question whether its homologs are required for proper behavioral and synaptic plasticity in other species as well.


Assuntos
Proteínas de Drosophila/deficiência , Atividade Motora/fisiologia , Proteínas do Tecido Nervoso/deficiência , Plasticidade Neuronal/fisiologia , Sinapses/metabolismo , Animais , Animais Geneticamente Modificados , Proteínas de Drosophila/genética , Drosophila melanogaster , Técnicas de Silenciamento de Genes , Masculino , Proteínas do Tecido Nervoso/genética , Olfato/fisiologia , Sinapses/genética
4.
Hum Mol Genet ; 19(10): 1951-66, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20167579

RESUMO

Axonal transport and translation of beta-actin mRNA plays an important role for axonal growth and presynaptic differentiation in many neurons including hippocampal, cortical and spinal motor neurons. Several beta-actin mRNA-binding and transport proteins have been identified, including ZBP1, ZBP2 and hnRNP-R. hnRNP-R has been found as an interaction partner of the survival motor neuron protein that is deficient in spinal muscular atrophy. Little is known about the function of hnRNP-R in axonal beta-actin translocation. hnRNP-R and beta-actin mRNA are colocalized in axons. Recombinant hnRNP-R interacts directly with the 3'-UTR of beta-actin mRNA. We studied the role of hnRNP-R in motor neurons by knockdown in zebrafish embryos and isolated mouse motor neurons. Suppression of hnRNP-R in developing zebrafish embryos results in reduced axon growth in spinal motor neurons, without any alteration in motor neuron survival. ShRNA-mediated knockdown in isolated embryonic mouse motor neurons reduces beta-actin mRNA translocation to the axonal growth cone, which is paralleled by reduced axon elongation. Dendrite growth and neuronal survival were not affected by hnRNP-R depletion in these neurons. The loss of beta-actin mRNA in axonal growth cones of hnRNP-R-depleted motor neurons resembles that observed in Smn-deficient motor neurons, a model for the human disease spinal muscular atrophy. In particular, hnRNP-R-depleted motor neurons also exhibit defects in presynaptic clustering of voltage-gated calcium channels. Our data suggest that hnRNP-R-mediated axonal beta-actin mRNA translocation plays an essential physiological role for axon growth and presynaptic differentiation.


Assuntos
Actinas/metabolismo , Axônios/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Neurônios Motores/metabolismo , Transporte de RNA , Coluna Vertebral/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Regiões 3' não Traduzidas/genética , Actinas/genética , Animais , Axônios/patologia , Canais de Cálcio Tipo N/metabolismo , Separação Celular , Embrião não Mamífero/patologia , Técnicas de Silenciamento de Genes , Cones de Crescimento/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/genética , Camundongos , Neurônios Motores/patologia , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Peixe-Zebra/metabolismo
5.
Sci Rep ; 9(1): 4515, 2019 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-30872638

RESUMO

Mutations within Leucine-rich repeat kinase 2 (LRRK2) are associated with late-onset Parkinson's disease. The physiological function of LRRK2 and molecular mechanism underlying the pathogenic role of LRRK2 mutations remain uncertain. Here, we investigated the role of LRRK2 in intracellular signal transduction. We find that deficiency of Lrrk2 in rodents affects insulin-dependent translocation of glucose transporter type 4 (GLUT4). This deficit is restored during aging by prolonged insulin-dependent activation of protein kinase B (PKB, Akt) and Akt substrate of 160 kDa (AS160), and is compensated by elevated basal expression of GLUT4 on the cell surface. Furthermore, we find a crucial role of Rab10 phosphorylation by LRRK2 for efficient insulin signal transduction. Translating our findings into human cell lines, we find comparable molecular alterations in fibroblasts from Parkinson's patients with the known pathogenic G2019S LRRK2 mutation. Our results highlight the role of LRRK2 in insulin-dependent signalling with potential therapeutic implications.


Assuntos
Transportador de Glucose Tipo 4/metabolismo , Insulina/farmacologia , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Doença de Parkinson/patologia , Transdução de Sinais/efeitos dos fármacos , Animais , Sobrevivência Celular/efeitos dos fármacos , Fatores de Crescimento de Fibroblastos/farmacologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Camundongos , Crescimento Neuronal/efeitos dos fármacos , Doença de Parkinson/metabolismo , Fosforilação , Polimorfismo de Nucleotídeo Único , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Proteínas rab de Ligação ao GTP/metabolismo
6.
PLoS One ; 10(4): e0118947, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25830304

RESUMO

Mutations within the LRRK2 gene have been identified in Parkinson's disease (PD) patients and have been implicated in the dysfunction of several cellular pathways. Here, we explore how pathogenic mutations and the inhibition of LRRK2 kinase activity affect cytoskeleton dynamics in mouse and human cell systems. We generated and characterized a novel transgenic mouse model expressing physiological levels of human wild type and G2019S-mutant LRRK2. No neuronal loss or neurodegeneration was detected in midbrain dopamine neurons at the age of 12 months. Postnatal hippocampal neurons derived from transgenic mice showed no alterations in the seven parameters examined concerning neurite outgrowth sampled automatically on several hundred neurons using high content imaging. Treatment with the kinase inhibitor LRRK2-IN-1 resulted in no significant changes in the neurite outgrowth. In human fibroblasts we analyzed whether pathogenic LRRK2 mutations change cytoskeleton functions such as cell adhesion. To this end we compared the adhesion characteristics of human skin fibroblasts derived from six PD patients carrying one of three different pathogenic LRRK2 mutations and from four age-matched control individuals. The mutant LRRK2 variants as well as the inhibition of LRRK2 kinase activity did not reveal any significant cell adhesion differences in cultured fibroblasts. In summary, our results in both human and mouse cell systems suggest that neither the expression of wild type or mutant LRRK2, nor the inhibition of LRRK2 kinase activity affect neurite complexity and cellular adhesion.


Assuntos
Citoesqueleto/metabolismo , Dopamina/metabolismo , Mutação , Proteínas Serina-Treonina Quinases/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Citoesqueleto/efeitos dos fármacos , Feminino , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Expressão Gênica , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Doença de Parkinson/enzimologia , Doença de Parkinson/genética , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores
7.
BMC Neurosci ; 5: 16, 2004 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-15117418

RESUMO

BACKGROUND: Conserved proteins preferentially expressed in synaptic terminals of the nervous system are likely to play a significant role in brain function. We have previously identified and molecularly characterized the Sap47 gene which codes for a novel synapse associated protein of 47 kDa in Drosophila. Sequence comparison identifies homologous proteins in numerous species including C. elegans, fish, mouse and human. First hints as to the function of this novel protein family can be obtained by generating mutants for the Sap47 gene in Drosophila. RESULTS: Attempts to eliminate the Sap47 gene through targeted mutagenesis by homologous recombination were unsuccessful. However, several mutants were generated by transposon remobilization after an appropriate insertion line had become available from the Drosophila P-element screen of the Bellen/Hoskins/Rubin/Spradling labs. Characterization of various deletions in the Sap47 gene due to imprecise excision of the P-element identified three null mutants and three hypomorphic mutants. Null mutants are viable and fertile and show no gross structural or obvious behavioural deficits. For cell-specific over-expression and "rescue" of the knock-out flies a transgenic line was generated which expresses the most abundant transcript under the control of the yeast enhancer UAS. In addition, knock-down of the Sap47 gene was achieved by generating 31 transgenic lines expressing Sap47 RNAi constructs, again under UAS control. When driven by a ubiquitously expressed yeast transcription factor (GAL4), Sap47 gene suppression in several of these lines is highly efficient resulting in residual SAP47 protein concentrations in heads as low as 6% of wild type levels. CONCLUSION: The conserved synaptic protein SAP47 of Drosophila is not essential for basic synaptic function. The Sap47 gene region may be refractory to targeted mutagenesis by homologous recombination. RNAi using a construct linking genomic DNA to anti-sense cDNA in our hands is not more effective than using a cDNA-anti-sense cDNA construct. The tools developed in this study will now allow a detailed analysis of the molecular, cellular and systemic function of the SAP47 protein in Drosophila.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Fertilidade/genética , Viabilidade Fetal/genética , Proteínas do Tecido Nervoso/genética , Processamento Alternativo , Animais , Animais Geneticamente Modificados , Western Blotting , Sequência Conservada , Proteínas de Drosophila/biossíntese , Éxons , Deleção de Genes , Marcação de Genes/métodos , Humanos , Íntrons , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas do Tecido Nervoso/biossíntese , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , Interferência de RNA , Recombinação Genética , Deleção de Sequência , Homologia de Sequência de Aminoácidos , Transgenes
8.
J Parkinsons Dis ; 4(3): 483-98, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25000966

RESUMO

BACKGROUND: A major risk-factor for developing Parkinson's disease (PD) is genetic variability in leucine-rich repeat kinase 2 (LRRK2), most notably the p.G2019S mutation. Examination of the effects of this mutation is necessary to determine the etiology of PD and to guide therapeutic development. OBJECTIVE: Assess the behavioral consequences of LRRK2 p.G2019S overexpression in transgenic rats as they age and test the functional integrity of the nigro-striatal dopamine system. Conduct positron emission tomography (PET) neuroimaging to compare transgenic rats with previous data from human LRRK2 mutation carriers. METHODS: Rats overexpressing human LRRK2 p.G2019S were generated by BAC transgenesis and compared to non-transgenic (NT) littermates. Motor skill tests were performed at 3, 6 and 12 months-of-age. PET, performed at 12 months, assessed the density of dopamine and vesicular monoamine transporters (DAT and VMAT2, respectively) and measured dopamine synthesis, storage and availability. Brain tissue was assayed for D2, DAT, dopamine and cAMP-regulated phosphoprotein (DARPP32) and tyrosine hydroxylase (TH) expression by Western blot, and TH by immunohistochemistry. RESULTS: Transgenic rats had no abnormalities in measures of striatal dopamine function at 12 months. A behavioral phenotype was present, with LRRK2 p.G2019S rats performing significantly worse on the rotarod than non-transgenic littermates (26% reduction in average running duration at 6 months), but with normal performance in other motor tests. CONCLUSIONS: Neuroimaging using dopaminergic PET did not recapitulate prior studies in human LRRK2 mutation carriers. Consistently, LRRK2 p.G2019S rats do not develop overt neurodegeneration; however, they do exhibit behavioral abnormalities.


Assuntos
Modelos Animais de Doenças , Dopamina/metabolismo , Atividade Motora/genética , Neostriado/metabolismo , Doença de Parkinson/genética , Proteínas Serina-Treonina Quinases/genética , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Masculino , Neostriado/diagnóstico por imagem , Fosforilação , Tomografia por Emissão de Pósitrons , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Receptores de Dopamina D2/metabolismo , Teste de Desempenho do Rota-Rod , Tirosina 3-Mono-Oxigenase/metabolismo , Proteínas Vesiculares de Transporte de Monoamina/metabolismo
9.
Mech Dev ; 125(8): 700-11, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18524547

RESUMO

The synaptic growth of neurons during the development and adult life of an animal is a very dynamic and highly regulated process. During larval development in Drosophila new boutons and branches are added at the glutamatergic neuromuscular junction (NMJ) until a balance between neuronal activity and morphological structures is reached. Analysis of several Drosophila mutants suggest that bouton number and size might be regulated by separate signaling processes [Budnik, V., 1996. Synapse maturation and structural plasticity at Drosophila neuromuscular junctions. Curr. Opin. Neurobiol. 6, 858-867.]. Here we show a new role for Hangover as a negative regulator of bouton number at the NMJ. The hangover gene (hang) encodes a nuclear zinc finger protein. It has a function in neuronal plasticity mediating ethanol tolerance, a behavior that develops upon previous experience with ethanol. hang(AE10) mutants have more boutons and an extended synaptic span. Moreover, Hang expression in the motoneuron is required for the regulation of bouton number and the overall length of muscle innervation. However, the increase in bouton number does not correlate with a change in synaptic transmission, suggesting a mechanism independent from neuronal activity leads to the surplus of synaptic boutons. In contrast, we find that expression levels of the cell adhesion molecule Fasciclin II (FASII) are reduced in the hang mutant. This finding suggests that the increase in bouton number in hang mutants is caused by a reduction in FASII expression, thus, linking the regulation of nuclear gene expression with the addition of boutons at the NMJ regulated by cell adhesion molecules.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/fisiologia , Neurônios Motores/fisiologia , Junção Neuromuscular/fisiologia , Terminações Pré-Sinápticas/fisiologia , Animais , Animais Geneticamente Modificados , Moléculas de Adesão Celular Neuronais/biossíntese , Proteínas de Drosophila/biossíntese , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Larva , Neurônios Motores/metabolismo , Mutação , Junção Neuromuscular/crescimento & desenvolvimento , Plasticidade Neuronal/fisiologia , Transmissão Sináptica , Dedos de Zinco
10.
Neurodegener Dis ; 4(2-3): 261-9, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17596720

RESUMO

Bag1 acts as a cochaperone for Hsp70. However, it also binds to members of the RAF family and to Akt. In addition, bag1 and Hsp70 are part of a complex with glucocorticoid receptors and thus modulate glucocorticoid receptor-mediated transcriptional activation. In the developing nervous system, bag1 is expressed in at least two isoforms. The L-form (bag1L) contains a nuclear localization signal and thus can translocate to the nucleus. In contrast, the S-form (bag1S) is localized exclusively in the cytoplasm. Former studies have shown that B-RAF is essential for neurotrophin-mediated survival signaling in motoneurons and sensory neurons, and that bag1 plays a role in coordinating B-RAF and Akt function in this context. In the absence of B-RAF, embryonic motoneurons and sensory neurons are not able to survive, indicating that bag1 and B-RAF are essential mediators for neuronal survival in response to neurotrophic factors during development. However, the role of the complex containing bag1, Hsp70 and B-RAF in mediating neurite growth in response to neurotrophic factors remained unclear. We have therefore studied the effect of bag1 overexpression in rat phaeochromocytoma (PC12) cells. Upon NGF treatment, proliferating PC12 become postmitotic and grow out neuronal processes. Bag1S overexpression interferes with neurite extension in PC12 cells. In contrast, bag1L does not disturb neurite outgrowth. Interaction of bag1S with Hsp70 appears necessary for this effect. These data indicate that the cytosolic form of bag1 participates in neurotrophin-mediated neurite growth, and that interaction with Hsp70 plays a crucial role in this context.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Ligação a DNA/fisiologia , Proteínas de Choque Térmico HSP70/metabolismo , Neuritos/fisiologia , Fatores de Transcrição/fisiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/classificação , Expressão Gênica/fisiologia , Proteínas de Fluorescência Verde/biossíntese , Indóis , Fator de Crescimento Neural/farmacologia , Neuritos/efeitos dos fármacos , Proteínas de Neurofilamentos/metabolismo , Células PC12/citologia , Isoformas de Proteínas/fisiologia , Proteínas Proto-Oncogênicas B-raf/metabolismo , Ratos , Fatores de Transcrição/classificação , Transfecção/métodos
11.
Histochem Cell Biol ; 127(4): 439-48, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17102992

RESUMO

Primary neurons are a common tool for investigating gene function for survival and morphological and functional differentiation. Gene transfer techniques play an important role in this context. However, the efficacy of conventional gene transfer techniques, in particular for primary motoneurons is low so that it is not possible to distinguish whether the observed effects are representative for all neurons or only for the small subpopulation that expresses the transfected cDNA. In order to develop techniques that allow high gene transfer rates, we have optimized lentiviral-based gene transfer for cultured motoneurons by using a replication-defective viral vector system. These techniques result in transduction efficacies higher than 50%, as judged by EGFP expression under the control of SFFV or CMV promoters. Under the same conditions, survival and morphology of the cultured motoneurons was not altered, at least not when virus titers did not exceed a multiplicity of infection of 100. Under the same cell culture conditions, electroporation resulted in less than 5% transfected motoneurons and reduced survival. Therefore we consider this lentivirus-based gene transfer protocol as a suitable tool to study the effects of gene transfer on motoneuron survival, differentiation and function.


Assuntos
Lentivirus/genética , Neurônios Motores/metabolismo , Transfecção/métodos , Animais , Linhagem Celular , Células Cultivadas , Citomegalovirus/genética , Vírus Defeituosos/genética , Eletroporação/métodos , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Neurônios Motores/citologia , Regiões Promotoras Genéticas/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Vírus Formadores de Foco no Baço/genética , Fatores de Tempo
12.
Proc Natl Acad Sci U S A ; 103(20): 7871-6, 2006 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-16684879

RESUMO

Ciliary neurotrophic factor (Cntf) plays an essential role in postnatal maintenance of spinal motoneurons. Whereas the expression of this neurotrophic factor is low during embryonic development, it is highly up-regulated after birth in myelinating Schwann cells of rodents. To characterize the underlying transcriptional mechanisms, we have analyzed and compared the effects of various glial transcription factors. In contrast to Pit-1, Oct-1, Unc-86 homology region (POU) domain class 3, transcription factor 1 (Oct6/SCIP/Tst-1) and paired box gene 3 (Pax3), SRY-box-containing gene 10 (Sox10) induces Cntf expression in Schwann cells. Subsequent promoter analysis using luciferase reporter gene and EMSA identified the corresponding response elements within the Cntf promoter. Overexpression of Sox10 in primary sciatic nerve Schwann cells leads to a >100-fold up-regulation of Cntf protein, and suppression of Sox10 by RNA interference in the spontaneously immortalized Schwann cell line 32 reduces Cntf expression by >80%. Mice with heterozygous inactivation of the Sox10 gene show significantly reduced Cntf protein levels in sciatic nerves, indicating that Sox10 is necessary and sufficient for regulating Cntf expression in the peripheral nervous system.


Assuntos
Fator Neurotrófico Ciliar , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Proteínas de Grupo de Alta Mobilidade/metabolismo , Células de Schwann/fisiologia , Fatores de Transcrição/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Fator Neurotrófico Ciliar/genética , Fator Neurotrófico Ciliar/metabolismo , Proteínas de Ligação a DNA/genética , Genes Reporter , Proteínas de Grupo de Alta Mobilidade/genética , Camundongos , Regiões Promotoras Genéticas , Ratos , Fatores de Transcrição SOXE , Células de Schwann/citologia , Nervo Isquiático/citologia , Nervo Isquiático/metabolismo , Fatores de Transcrição/genética , Sítio de Iniciação de Transcrição
13.
Eur J Neurosci ; 20(3): 611-22, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15255973

RESUMO

Vertebrate synapsins are abundant synaptic vesicle phosphoproteins that have been proposed to fine-regulate neurotransmitter release by phosphorylation-dependent control of synaptic vesicle motility. However, the consequences of a total lack of all synapsin isoforms due to a knock-out of all three mouse synapsin genes have not yet been investigated. In Drosophila a single synapsin gene encodes several isoforms and is expressed in most synaptic terminals. Thus the targeted deletion of the synapsin gene of Drosophila eliminates the possibility of functional knock-out complementation by other isoforms. Unexpectedly, synapsin null mutant flies show no obvious defects in brain morphology, and no striking qualitative changes in behaviour are observed. Ultrastructural analysis of an identified 'model' synapse of the larval nerve muscle preparation revealed no difference between wild-type and mutant, and spontaneous or evoked excitatory junction potentials at this synapse were normal up to a stimulus frequency of 5 Hz. However, when several behavioural responses were analysed quantitatively, specific differences between mutant and wild-type flies are noted. Adult locomotor activity, optomotor responses at high pattern velocities, wing beat frequency, and visual pattern preference are modified. Synapsin mutant flies show faster habituation of an olfactory jump response, enhanced ethanol tolerance, and significant defects in learning and memory as measured using three different paradigms. Larval behavioural defects are described in a separate paper. We conclude that Drosophila synapsins play a significant role in nervous system function, which is subtle at the cellular level but manifests itself in complex behaviour.


Assuntos
Comportamento Animal/fisiologia , Drosophila/fisiologia , Sinapsinas/deficiência , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Potenciais de Ação/fisiologia , Animais , Animais Geneticamente Modificados , Comportamento Animal/efeitos dos fármacos , Western Blotting/métodos , Depressores do Sistema Nervoso Central/farmacologia , Clonagem Molecular/métodos , Condicionamento Operante/fisiologia , Análise Mutacional de DNA , Drosophila/genética , Estimulação Elétrica/métodos , Etanol/farmacologia , Potenciais Pós-Sinápticos Excitadores/genética , Imuno-Histoquímica/métodos , Técnicas de Imunoadsorção , Potenciais da Membrana/genética , Potenciais da Membrana/fisiologia , Microscopia Eletrônica , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Mutagênese/fisiologia , Junção Neuromuscular/genética , Junção Neuromuscular/metabolismo , Junção Neuromuscular/fisiologia , Desempenho Psicomotor/fisiologia , Comportamento Sexual/efeitos dos fármacos , Comportamento Sexual/fisiologia , Sinapses/metabolismo , Sinapses/ultraestrutura , Sinapsinas/genética , Sinapsinas/fisiologia , Vesículas Sinápticas/genética , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/ultraestrutura , Fatores de Tempo , Distribuição Tecidual , Percepção Visual/genética , Percepção Visual/fisiologia , Caminhada/fisiologia , Asas de Animais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA