Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
EMBO J ; 40(14): e105712, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34057742

RESUMO

During development, neural progenitors are in proliferative and immature states; however, the molecular machinery that cooperatively controls both states remains elusive. Here, we report that cyclin D1 (CCND1) directly regulates both proliferative and immature states of cerebellar granule cell progenitors (GCPs). CCND1 not only accelerates cell cycle but also upregulates ATOH1 protein, an essential transcription factor that maintains GCPs in an immature state. In cooperation with CDK4, CCND1 directly phosphorylates S309 of ATOH1, which inhibits additional phosphorylation at S328 and consequently prevents S328 phosphorylation-dependent ATOH1 degradation. Additionally, PROX1 downregulates Ccnd1 expression by histone deacetylation of Ccnd1 promoter in GCPs, leading to cell cycle exit and differentiation. Moreover, WNT signaling upregulates PROX1 expression in GCPs. These findings suggest that WNT-PROX1-CCND1-ATOH1 signaling cascade cooperatively controls proliferative and immature states of GCPs. We revealed that the expression and phosphorylation levels of these molecules dynamically change during cerebellar development, which are suggested to determine appropriate differentiation rates from GCPs to GCs at distinct developmental stages. This study contributes to understanding the regulatory mechanism of GCPs as well as neural progenitors.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Cerebelo/crescimento & desenvolvimento , Cerebelo/metabolismo , Ciclina D1/metabolismo , Grânulos Citoplasmáticos/metabolismo , Fosforilação/fisiologia , Células-Tronco/metabolismo , Animais , Ciclo Celular/genética , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Proteínas Hedgehog/metabolismo , Camundongos , Neurogênese/fisiologia , Transdução de Sinais/fisiologia , Fatores de Transcrição
2.
J Neurosci ; 43(22): 3989-4004, 2023 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-37117011

RESUMO

The presentation of nicotinic acetylcholine receptors (nAChRs) on synaptic membranes is crucial for generating cholinergic circuits, some of which are associated with memory function and neurodegenerative disorders. Although the physiology and structure of nAChR, a cation channel comprising five subunits, have been extensively studied, little is known about how the receptor levels in interneuronal synapses are determined and which nAChR subunits participate in the regulatory process in cooperation with synaptic cleft matrices and intracellular proteins. By a genetic screen of Drosophila, we identified mutations in the nAChR subunit Dα5 gene as suppressors that restored the mutant phenotypes of hig, which encodes a secretory matrix protein localized to cholinergic synaptic clefts in the brain. Only the loss of function of Dα5 among the 10 nAChR subunits suppressed hig mutant phenotypes in both male and female flies. Dα5 behaved as a lethal factor when Hig was defective; loss of Dα5 in hig mutants rescued lethality, upregulating Dα6 synaptic levels. By contrast, levels of Dα5, Dα6, and Dα7 subunits were all reduced in hig mutants. These three subunits have distinct properties for interaction with Hig or trafficking, as confirmed by chimeric subunit experiments. Notably, the chimeric Dα5 protein, which has the extracellular sequences that display no positive interaction with Hig, exhibited abnormal distribution and lethality even in the presence of Hig. We propose that the sequestering subunit Dα5 functions by reducing synaptic levels of nAChR through internalization, and this process is blocked by Hig, which tethers Dα5 to the synaptic cleft matrix.SIGNIFICANCE STATEMENT Because the cholinergic synapse is one of the major synapses that generate various brain functions, numerous studies have sought to reveal the physiology and structure of the nicotinic acetylcholine receptor (nAChR). However, little is known about how synaptic levels of nAChR are controlled and which nAChR subunits participate in the regulatory process in cooperation with synaptic cleft matrices. By a genetic screen of Drosophila, we identified mutations in the nAChR subunit Dα5 gene as suppressors that restored the mutant phenotypes of hig, which encodes a secretory matrix protein localized to cholinergic synaptic clefts. Our data indicate that Dα5 functions in reducing synaptic levels of nAChR, and this process is blocked by Hig, which tethers Dα5 to the synaptic cleft matrix.


Assuntos
Proteínas de Drosophila , Receptores Nicotínicos , Animais , Feminino , Masculino , Colinérgicos , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Receptores Nicotínicos/metabolismo , Transmissão Sináptica
4.
Hum Mol Genet ; 23(5): 1345-64, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24179173

RESUMO

DNA damage repair is implicated in neurodegenerative diseases; however, the relative contributions of various DNA repair systems to the pathology of these diseases have not been investigated systematically. In this study, we performed a systematic in vivo screen of all available Drosophila melanogaster homolog DNA repair genes, and we tested the effect of their overexpression on lifespan and developmental viability in Spinocerebellar Ataxia Type 1 (SCA1) Drosophila models expressing human mutant Ataxin-1 (Atxn1). We identified genes previously unknown to be involved in CAG-/polyQ-related pathogenesis that function in multiple DNA damage repair systems. Beyond the significance of each repair system, systems biology analyses unraveled the core networks connecting positive genes in the gene screen that could contribute to SCA1 pathology. In particular, RpA1, which had the largest effect on lifespan in the SCA1 fly model, was located at the hub position linked to such core repair systems, including homologous recombination (HR). We revealed that Atxn1 actually interacted with RpA1 and its essential partners BRCA1/2. Furthermore, mutant but not normal Atxn1 impaired the dynamics of RpA1 in the nucleus after DNA damage. Uptake of BrdU by Purkinje cells was observed in mutant Atxn1 knockin mice, suggesting their abnormal entry to the S-phase. In addition, chemical and genetic inhibitions of Chk1 elongated lifespan and recovered eye degeneration. Collectively, we elucidated core networks for DNA damage repair in SCA1 that might include the aberrant usage of HR.


Assuntos
Dano ao DNA , Reparo do DNA , Drosophila/genética , Redes Reguladoras de Genes , Ataxias Espinocerebelares/genética , Animais , Animais Geneticamente Modificados , Ataxina-1 , Ataxinas , Ciclo Celular/genética , Quinase 1 do Ponto de Checagem , Modelos Animais de Doenças , Feminino , Vetores Genéticos/genética , Humanos , Longevidade/genética , Masculino , Mutagênese Insercional , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Proteínas Quinases/metabolismo , Células de Purkinje/metabolismo , Transdução de Sinais , Ataxias Espinocerebelares/metabolismo , Ataxias Espinocerebelares/mortalidade , Biologia de Sistemas
5.
EMBO J ; 29(14): 2446-60, 2010 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-20531390

RESUMO

Non-cell-autonomous effect of mutant proteins expressed in glia has been implicated in several neurodegenerative disorders, whereas molecules mediating the toxicity are currently not known. We identified a novel molecule named multiple alpha-helix protein located at ER (Maxer) downregulated by mutant ataxin-1 (Atx1) in Bergmann glia. Maxer is an endoplasmic reticulum (ER) membrane protein interacting with CDK5RAP3. Maxer anchors CDK5RAP3 to the ER and inhibits its function of Cyclin D1 transcription repression in the nucleus. The loss of Maxer eventually induces cell accumulation at G1 phase. It was also shown that mutant Atx1 represses Maxer and inhibits proliferation of Bergmann glia in vitro. Consistently, Bergmann glia are reduced in the cerebellum of mutant Atx1 knockin mice before onset. Glutamate-aspartate transporter reduction in Bergmann glia by mutant Atx1 and vulnerability of Purkinje cell to glutamate are both strengthened by Maxer knockdown in Bergmann glia, whereas Maxer overexpression rescues them. Collectively, these results suggest that the reduction of Maxer mediates functional deficiency of Bergmann glia, and might contribute to the non-cell-autonomous pathology of SCA1.


Assuntos
Retículo Endoplasmático/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso , Neuroglia/metabolismo , Proteínas Nucleares , Sequência de Aminoácidos , Animais , Ataxina-1 , Ataxinas , Proteínas de Ciclo Celular , Proliferação de Células , Transportador 1 de Aminoácido Excitatório/genética , Transportador 1 de Aminoácido Excitatório/metabolismo , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neuroglia/citologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ratos , Ratos Wistar , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Distribuição Tecidual , Proteínas Supressoras de Tumor
6.
Dev Cell ; 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38834071

RESUMO

Normal cells coordinate proliferation and differentiation by precise tuning of gene expression based on the dynamic shifts of the epigenome throughout the developmental timeline. Although non-mutational epigenetic reprogramming is an emerging hallmark of cancer, the epigenomic shifts that occur during the transition from normal to malignant cells remain elusive. Here, we capture the epigenomic changes that occur during tumorigenesis in a prototypic embryonal brain tumor, medulloblastoma. By comparing the epigenomes of the different stages of transforming cells in mice, we identify nuclear factor I family of transcription factors, known to be cell fate determinants in development, as oncogenic regulators in the epigenomes of precancerous and cancerous cells. Furthermore, genetic and pharmacological inhibition of NFIB validated a crucial role of this transcription factor by disrupting the cancer epigenome in medulloblastoma. Thus, this study exemplifies how epigenomic changes contribute to tumorigenesis via non-mutational mechanisms involving developmental transcription factors.

7.
Nat Commun ; 15(1): 458, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38302444

RESUMO

In the central nervous system, astrocytes enable appropriate synapse function through glutamate clearance from the synaptic cleft; however, it remains unclear how astrocytic glutamate transporters function at peri-synaptic contact. Here, we report that Down syndrome cell adhesion molecule (DSCAM) in Purkinje cells controls synapse formation and function in the developing cerebellum. Dscam-mutant mice show defects in CF synapse translocation as is observed in loss of function mutations in the astrocytic glutamate transporter GLAST expressed in Bergmann glia. These mice show impaired glutamate clearance and the delocalization of GLAST away from the cleft of parallel fibre (PF) synapse. GLAST complexes with the extracellular domain of DSCAM. Riluzole, as an activator of GLAST-mediated uptake, rescues the proximal impairment in CF synapse formation in Purkinje cell-selective Dscam-deficient mice. DSCAM is required for motor learning, but not gross motor coordination. In conclusion, the intercellular association of synaptic and astrocyte proteins is important for synapse formation and function in neural transmission.


Assuntos
Neuroglia , Neurônios , Animais , Camundongos , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Cerebelo/metabolismo , Ácido Glutâmico/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Células de Purkinje/metabolismo , Sinapses/metabolismo
8.
J Neurosci ; 30(42): 14091-101, 2010 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-20962230

RESUMO

Polyglutamine tract-binding protein-1 (PQBP1) is involved in the transcription-splicing coupling, and its mutations cause a group of human mental retardation syndromes. We generated a fly model in which the Drosophila homolog of PQBP1 (dPQBP1) is repressed by insertion of piggyBac. In classical odor conditioning, learning acquisition was significantly impaired in homozygous piggyBac-inserted flies, whereas the following memory retention was completely normal. Mushroom bodies (MBs) and antennal lobes were morphologically normal in dPQBP1-mutant flies. Projection neurons (PNs) were not reduced in number and their fiber connections were not changed, whereas gene expressions including NMDA receptor subunit 1 (NR1) were decreased in PNs. Targeted double-stranded RNA-mediated silencing of dPQBP1 in PNs, but not in MBs, similarly disrupted learning acquisition. NR1 overexpression in PNs rescued the learning disturbance of dPQBP1 mutants. HDAC (histone deacetylase) inhibitors, SAHA (suberoylanilide hydroxamic acid) and PBA (phenylbutyrate), that upregulated NR1 partially rescued the learning disturbance. Collectively, these findings identify dPQBP1 as a novel gene regulating learning acquisition at PNs.


Assuntos
Aprendizagem da Esquiva/fisiologia , Condicionamento Operante/fisiologia , Drosophila/fisiologia , Neurônios/fisiologia , Oligopeptídeos/genética , Oligopeptídeos/fisiologia , Olfato/genética , Olfato/fisiologia , Animais , Northern Blotting , Dendritos/metabolismo , Dendritos/ultraestrutura , Inibidores de Histona Desacetilases/farmacologia , Imuno-Histoquímica , Cloreto de Lítio/farmacologia , Corpos Pedunculados/fisiologia , Mutação/fisiologia , Desempenho Psicomotor/fisiologia , Piridinas/farmacologia , RNA/biossíntese , RNA/genética , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/biossíntese , Receptores de N-Metil-D-Aspartato/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Chem Commun (Camb) ; 57(46): 5630-5633, 2021 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-34018507

RESUMO

We designed a new caging group that can be photoactivated only in the presence of a non-endogenous enzyme when exposed to 405 nm light. Because cells or tissues can be genetically tagged by an exogenously expressed enzyme, this novel method can serve as a strategy for adding targeting abilities to photocaged compounds.


Assuntos
Nucleotídeos Cíclicos/síntese química , Células HeLa , Humanos , Luz , Estrutura Molecular , Nucleotídeos Cíclicos/química , Nucleotídeos Cíclicos/genética , Processos Fotoquímicos , Células Tumorais Cultivadas
10.
Genes Genet Syst ; 95(6): 303-314, 2021 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-33583916

RESUMO

yata mutants of Drosophila melanogaster exhibit phenotypes including progressive brain shrinkage, developmental abnormalities and shortened lifespan, whereas in mammals, null mutations of the yata ortholog Scyl1 result in motor neuron degeneration. yata mutation also causes defects in the anterograde intracellular trafficking of a subset of proteins including APPL, which is the Drosophila ortholog of mammalian APP, a causative molecule in Alzheimer's disease. SCYL1 binds and regulates the function of coat protein complex I (COPI) in secretory vesicles. Here, we reveal a role for the Drosophila YATA protein in the proper localization of COPI. Immunohistochemical analyses performed using confocal microscopy and structured illumination microscopy showed that YATA colocalizes with COPI and GM130, a cis-Golgi marker. Analyses using transgenically expressed YATA with a modified N-terminal sequence revealed that the N-terminal portion of YATA is required for the proper subcellular localization of YATA. Analysis using transgenically expressed YATA proteins in which the C-terminal sequence was modified revealed a function for the C-terminal portion of YATA in the subcellular localization of COPI. Notably, when YATA was mislocalized, it also caused the mislocalization of COPI, indicating that YATA plays a role in directing COPI to the proper subcellular site. Moreover, when both YATA and COPI were mislocalized, the staining pattern of GM130 revealed Golgi with abnormal elongated shapes. Thus, our in vivo data indicate that YATA plays a role in the proper subcellular localization of COPI.


Assuntos
Complexo I de Proteína do Envoltório/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas Quinases/metabolismo , Animais , Sítios de Ligação , Complexo I de Proteína do Envoltório/química , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster , Complexo de Golgi/metabolismo , Ligação Proteica , Proteínas Quinases/química , Proteínas Quinases/genética , Sinais Direcionadores de Proteínas , Transporte Proteico , Vesículas Secretórias/metabolismo
11.
Life Sci Alliance ; 4(7)2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34130995

RESUMO

The early-stage pathologies of frontotemporal lobal degeneration (FTLD) remain largely unknown. In VCPT262A-KI mice carrying VCP gene mutation linked to FTLD, insufficient DNA damage repair in neural stem/progenitor cells (NSCs) activated DNA-PK and CDK1 that disabled MCM3 essential for the G1/S cell cycle transition. Abnormal neural exit produced neurons carrying over unrepaired DNA damage and induced early-stage transcriptional repression-induced atypical cell death (TRIAD) necrosis accompanied by the specific markers pSer46-MARCKS and YAP. In utero gene therapy expressing normal VCP or non-phosphorylated mutant MCM3 rescued DNA damage, neuronal necrosis, cognitive function, and TDP43 aggregation in adult neurons of VCPT262A-KI mice, whereas similar therapy in adulthood was less effective. The similar early-stage neuronal necrosis was detected in PGRNR504X-KI, CHMP2BQ165X-KI, and TDPN267S-KI mice, and blocked by embryonic treatment with AAV-non-phospho-MCM3. Moreover, YAP-dependent necrosis occurred in neurons of human FTLD patients, and consistently pSer46-MARCKS was increased in cerebrospinal fluid (CSF) and serum of these patients. Collectively, developmental stress followed by early-stage neuronal necrosis is a potential target for therapeutics and one of the earliest general biomarkers for FTLD.


Assuntos
Degeneração Lobar Frontotemporal/patologia , Células-Tronco Neurais/metabolismo , Proteína com Valosina/metabolismo , Animais , Ciclo Celular , Linhagem da Célula/genética , Células Cultivadas , Dano ao DNA/genética , Dano ao DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Degeneração Lobar Frontotemporal/líquido cefalorraquidiano , Degeneração Lobar Frontotemporal/genética , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Necrose/metabolismo , Necrose/patologia , Células-Tronco Neurais/patologia , Neurônios/metabolismo , Proteína com Valosina/genética
12.
Biol Open ; 9(1)2020 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-31862863

RESUMO

We previously identified the Drosophila yata mutant, which showed phenotypes including progressive vacuolization of the white-coloured compound eye, progressive shrinkage of the brain and a shortened lifespan. The yata gene was shown to be involved in controlling intracellular trafficking of the Amyloid precursor protein-like protein, which is an orthologue of Amyloid precursor protein, which is a causative molecule of Alzheimer's disease. In this study, we examined the phenotype of the compound eye of the yata mutant using electron microscopy and confocal microscopy. We found that abnormal cellular structures that seemed to originate from bleb-like structures and contained vesicles and organelles, such as multivesicular bodies and autophagosomes, were observed in aged white; yata mutants and aged white mutants. These structures were not observed in newly eclosed flies and the presence of the structures was suppressed in flies grown under constant dark conditions after eclosion. The structures were not observed in newly eclosed red-eyed yata mutants or wild-type flies, but were observed in very aged red-eyed wild-type flies. Thus, our data suggest that the observed structures are formed as a result of changes associated with exposure to light after eclosion in white mutants, white; yata mutants and aged flies.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Olho Composto de Artrópodes/metabolismo , Olho Composto de Artrópodes/ultraestrutura , Proteínas de Drosophila/genética , Drosophila/genética , Drosophila/ultraestrutura , Proteínas do Olho/genética , Mutação , Proteínas Quinases/genética , Fatores Etários , Animais , Estudos de Associação Genética , Longevidade/genética , Fenótipo
13.
iScience ; 23(6): 101183, 2020 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-32498016

RESUMO

Impairments in synapse development are thought to cause numerous psychiatric disorders. Autism susceptibility candidate 2 (AUTS2) gene has been associated with various psychiatric disorders, such as autism and intellectual disabilities. Although roles for AUTS2 in neuronal migration and neuritogenesis have been reported, its involvement in synapse regulation remains unclear. In this study, we found that excitatory synapses were specifically increased in the Auts2-deficient primary cultured neurons as well as Auts2 mutant forebrains. Electrophysiological recordings and immunostaining showed increases in excitatory synaptic inputs as well as c-fos expression in Auts2 mutant brains, suggesting that an altered balance of excitatory and inhibitory inputs enhances brain excitability. Auts2 mutant mice exhibited autistic-like behaviors including impairments in social interaction and altered vocal communication. Together, these findings suggest that AUTS2 regulates excitatory synapse number to coordinate E/I balance in the brain, whose impairment may underlie the pathology of psychiatric disorders in individuals with AUTS2 mutations.

14.
Neuron ; 47(2): 201-13, 2005 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-16039563

RESUMO

The molecular machinery governing glutamatergic-GABAergic neuronal subtype specification is unclear. Here we describe a cerebellar mutant, cerebelless, which lacks the entire cerebellar cortex in adults. The primary defect of the mutant brains was a specific inhibition of GABAergic neuron production from the cerebellar ventricular zone (VZ), resulting in secondary and complete loss of external germinal layer, pontine, and olivary nuclei during development. We identified the responsible gene, Ptf1a, whose expression was lost in the cerebellar VZ but was maintained in the pancreas in cerebelless. Lineage tracing revealed that two types of neural precursors exist in the cerebellar VZ: Ptf1a-expressing and -nonexpressing precursors, which generate GABAergic and glutamatergic neurons, respectively. Introduction of Ptf1a into glutamatergic neuron precursors in the dorsal telencephalon generated GABAergic neurons with representative morphological and migratory features. Our results suggest that Ptf1a is involved in driving neural precursors to differentiate into GABAergic neurons in the cerebellum.


Assuntos
Cerebelo/citologia , Cerebelo/metabolismo , Sequências Hélice-Alça-Hélice/fisiologia , Neurônios/fisiologia , Peptidilprolil Isomerase/fisiologia , Ácido gama-Aminobutírico/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Bromodesoxiuridina/metabolismo , Calbindina 2 , Calbindinas , Contagem de Células/métodos , Morte Celular/fisiologia , Diferenciação Celular/fisiologia , Tamanho Celular , Cerebelo/anormalidades , Cerebelo/embriologia , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Fluorescência Verde , Imuno-Histoquímica/métodos , Hibridização in Situ Fluorescente/métodos , Marcação In Situ das Extremidades Cortadas/métodos , Técnicas In Vitro , Camundongos , Camundongos Mutantes , Modelos Neurológicos , Peptidilprolil Isomerase de Interação com NIMA , Neurônios/classificação , Fenótipo , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Proteína G de Ligação ao Cálcio S100/metabolismo , beta-Galactosidase/metabolismo
15.
PLoS One ; 13(9): e0204048, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30226901

RESUMO

APP (amyloid precursor protein), the causative molecule of Alzheimer's disease, is synthesized in neuronal cell bodies and subsequently transported to synapses. We previously showed that the yata gene is required for the synaptic transport of the APP orthologue in Drosophila melanogaster. In this study, we examined the effect of a reduction in yata expression in the Drosophila Alzheimer's disease model, in which expression of human mutant APP was induced. The synaptic localization of APP and other synaptic proteins was differentially inhibited by yata knockdown and null mutation. Expression of APP resulted in abnormal synaptic morphology and the premature death of animals. These phenotypes were partially but significantly rescued by yata knockdown, whereas yata knockdown itself caused no abnormality. Moreover, we observed that synaptic transmission accuracy was impaired in our model, and this phenotype was improved by yata knockdown. Thus, our data suggested that the phenotypes caused by APP can be partially prevented by inhibition of the synaptic localization of a subset of synaptic proteins including APP.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Drosophila melanogaster/metabolismo , Sinapses/metabolismo , Doença de Alzheimer/prevenção & controle , Animais , Modelos Animais de Doenças , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Feminino , Técnicas de Silenciamento de Genes , Masculino , Proteínas do Tecido Nervoso/metabolismo , Proteínas Quinases/genética
16.
J Neurosci ; 25(17): 4406-19, 2005 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-15858067

RESUMO

Rho-family GTPases play key roles in regulating cytoskeletal reorganization, contributing to many aspects of nervous system development. Their activities are known to be regulated by guanine nucleotide exchange factors (GEFs), in response to various extracellular cues. P-Rex1, a GEF for Rac, has been mainly investigated in neutrophils, in which this molecule contributes to reactive oxygen species formation. However, its role in the nervous system is essentially unknown. Here we describe the expression profile and a physiological function of P-Rex1 in nervous system development. In situ hybridization revealed that P-Rex1 is dynamically expressed in a variety of cells in the developing mouse brain, including some cortical and DRG neurons. In migrating neurons in the intermediate zone, P-Rex1 protein was found to localize in the leading process and adjacent cytoplasmic region. When transfected in pheochromocytoma PC12 cells, P-Rex1 can be activated by NGF, causing an increase in GTP-bound Rac1 and cell motility. Deletion analyses suggested roles for distinct domains of this molecule. Experiments using a P-Rex1 mutant lacking the Dbl-homology domain, a dominant-negative-like form, and small interfering RNA showed that endogenous P-Rex1 was involved in cell migration of PC12 cells and primary cultured neurons from the embryonic day 14 cerebral cortices, induced by extracellular stimuli (NGF, BDNF, and epidermal growth factor). Furthermore, in utero electroporation of the mutant protein into the embryonic cerebral cortex perturbed radial neuronal migration. These findings suggest that P-Rex1, which is expressed in a variety of cell types, is activated by extracellular cues such as neurotrophins and contributes to neuronal migration in the developing nervous system.


Assuntos
Movimento Celular/fisiologia , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Fatores de Crescimento Neural/metabolismo , Neurônios/fisiologia , Transdução de Sinais/fisiologia , Actinas/metabolismo , Animais , Northern Blotting/métodos , Encéfalo/anatomia & histologia , Encéfalo/embriologia , Encéfalo/metabolismo , Células Cultivadas , Embrião de Mamíferos , Imunofluorescência/métodos , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Fluorescência Verde/metabolismo , Humanos , Hibridização In Situ/métodos , Camundongos , Camundongos Endogâmicos ICR , Mutagênese/fisiologia , Fator de Crescimento Neural/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Ratos , Receptor trkB/metabolismo , Fatores de Tempo , Transfecção/métodos , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
17.
Biochim Open ; 3: 1-7, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29450125

RESUMO

ß-Galactosidase encoded by the Escherichia coli lacZ gene, is widely used as a reporter molecule in molecular biology in a wide variety of animals. ß-Galactosidase retains its enzymatic activity in cells or tissues even after fixation and can degrade X-Gal, a frequently used colormetric substrate, producing a blue color. Therefore, it can be used for the activity staining of fixed tissues. However, the enzymatic activity of the ß-galactosidase that is ectopically expressed in the non-fixed tissues of animals has not been extensively studied. Here, we report the characterization of ß-galactosidase activity in Drosophila tissues with and without fixation in various experimental conditions comparing the activity of two evolutionarily orthologous ß-galactosidases derived from the E. coli lacZ and Drosophila melanogaster DmelGal genes. We performed quantitative analysis of the activity staining of larval imaginal discs and an in vitro assay using larval lysates. Our data showed that both E. coli and Drosophila ß-galactosidase can be used for cell-type-specific activity staining, but they have their own preferences in regard to conditions. E. coli ß-galactosidase showed a preference for neutral pH but not for acidic pH compared with Drosophila ß-galactosidase. Our data suggested that both E. coli and Drosophila ß-galactosidase show enzymatic activity in the physiological conditions of living animals when they are ectopically expressed in a desired specific spatial and temporal pattern. This may enable their future application to studies of chemical biology using model animals.

18.
Mech Dev ; 113(1): 65-8, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11900975

RESUMO

STEF (Sif- and Tiam1-like exchange factor), a guanine nucleotide exchange factor, was identified as a candidate molecule in regulation of neural development. The STEF gene product specifically activates Rac1, a member of the Rho-like small G proteins. Here we report the detailed examination of the expression profile of the stef gene in the mouse brain. In situ hybridization revealed that the stef gene was expressed in a stage- and region-specific manner in the mouse brain; it was expressed during certain developmental stages in the cerebral cortex, the olfactory bulb, the rostral migratory pathway (RMP) and the hippocampus. In the cerebral cortex, stef transcripts were detected in migrating cells in the intermediate zone as well as neurons in the cortical plate. While the expression in the cerebral cortex was reduced at adult stages, considerable expression was found to be maintained in other regions (RMP, olfactory bulb, hippocampal formation), which are the tissues where neurons continue to undergo morphological remodeling including cellular migration, neurite extension and synapse formation even in adults. Thus, stef gene expression appears to correspond to neuronal morphological changes.


Assuntos
Encéfalo/embriologia , Fatores de Troca do Nucleotídeo Guanina/biossíntese , Neurônios/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Córtex Cerebral/embriologia , Hipocampo/embriologia , Hipocampo/metabolismo , Hibridização In Situ , Camundongos , Camundongos Endogâmicos ICR , Bulbo Olfatório/embriologia , RNA Complementar/metabolismo , Fatores de Tempo , Proteínas rac1 de Ligação ao GTP/fisiologia
19.
Gene Expr Patterns ; 3(3): 375-81, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12799088

RESUMO

Dbl-family guanine nucleotide exchange factors (Dbl-GEFs) act as activators of Rho-like small G proteins such as Rac1, Cdc42 and RhoA. Recently, some GEFs have been suggested to play important roles in the development of the nervous system. Here, we report a comprehensive expression profile analysis of 20 Dbl-GEFs that have yet to be well investigated. Northern analyses of murine mRNAs from brains of E13, E17, P7 and adult mice revealed expression of 18 out of 20 GEFs in some or all stages. In addition, we found that three human GEFs were highly expressed in the brain. Examination of the spatial expression patterns of five GEFs in embryos or neonatal brain by in situ hybridization revealed distinct patterns for each GEF. Our study reveals the dynamic and coordinated expression profiles of the Dbl-GEFs and provides a basic framework for understanding the function of GEFs in neural development.


Assuntos
Encéfalo/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fatores de Troca do Nucleotídeo Guanina/genética , Sequência de Aminoácidos , Animais , Encéfalo/metabolismo , Perfilação da Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/biossíntese , Humanos , Hibridização In Situ , Camundongos , Dados de Sequência Molecular , Família Multigênica , Alinhamento de Sequência
20.
PLoS One ; 9(12): e116567, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25551764

RESUMO

The fruit fly, Drosophila melanogaster, is a commonly used model organism for neurodegenerative diseases. Its major advantages include a short lifespan and its susceptibility to manipulation using sophisticated genetic techniques. Here, we report the systematic comparison of fly models of two polyglutamine (polyQ) diseases. We induced expression of the normal and mutant forms of full-length Ataxin-1 and Huntingtin exon 1 in cholinergic, dopaminergic, and motor neurons, and glial cells using cell type-specific drivers. We systematically analyzed their effects based on multiple phenotypes: eclosion rate, lifespan, motor performance, and circadian rhythms of spontaneous activity. This systematic assay system enabled us to quantitatively evaluate and compare the functional disabilities of different genotypes. The results suggest different effects of Ataxin-1 and Huntingtin on specific types of neural cells during development and in adulthood. In addition, we confirmed the therapeutic effects of LiCl and butyrate using representative models. These results support the usefulness of this assay system for screening candidate chemical compounds that modify the pathologies of polyQ diseases.


Assuntos
Ataxina-1/fisiologia , Drosophila melanogaster/genética , Proteínas Associadas aos Microtúbulos/fisiologia , Animais , Animais Geneticamente Modificados/metabolismo , Antracenos/farmacologia , Ataxina-1/genética , Ataxina-1/metabolismo , Comportamento Animal , Butiratos/farmacologia , Proteínas de Drosophila , Drosophila melanogaster/metabolismo , Drosophila melanogaster/fisiologia , Regulação da Expressão Gênica , Proteína Huntingtina , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Cloreto de Lítio/farmacologia , Longevidade/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Modelos Animais , Atividade Motora/genética , Fármacos Neuroprotetores/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA