Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Proteomics ; 269: 104720, 2022 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-36089189

RESUMO

Loss of fragile X retardation protein (FMRP) leads to fragile X syndrome (FXS), a common cause of inherited intellectual disability. Protein lysine acetylation (K-ac), a reversible post-translational modification of proteins, is associated with the regulation of brain development and neuropathies. However, a comprehensive hippocampal K-ac protein profile in response to FMRP deficiency has not been reported until now. Using LC-MS/MS to analyze the enriched K-ac peptides, this study identified 1629 K-ac hits across 717 proteins in the mouse hippocampus, and these proteins were enriched in several metabolic processes. Of them, 51 K-ac hits across 45 proteins were significantly changed upon loss of FMRP. These altered K-ac proteins were enriched in energy metabolic processes including carboxylic acid metabolism process, aerobic respiration and citrate cycle, linking with several neurological disorders such as lactic acidosis, Lewy body disease, Leigh disease and encephalopathies. In the mouse hippocampus and the hippocampal HT-22 cells, FMRP deficiency could induce altered K-ac modification of several key enzymes, decrease in ATP and increase in lactate. Thus, this study identified a global hippocampal lysine acetylome and an altered K-ac protein profile upon loss of FMRP linked to abnormal energy metabolism, implicating in the pathogenesis of FXS. SIGNIFICANCE: Fragile X syndrome (FXS) is a common inherited neurodevelopment disorder characterized by intellectual disability and an increased risk for autism spectrum disorder. FXS is resulted from silencing of the FMR1 gene, which induces loss of its encoding protein FMRP. Molecular and metabolic changes of Fmr1-null animal models of FXS have been identified to potentially contribute to the pathogenesis of FXS. Here, we used a TMT-labeled quantitative proteomic analysis of the peptides enriched by anti-K-ac antibodies and identified a global K-ac protein profile in the mouse hippocampus with a total of 1629 K-ac peptides on 717 proteins. Of them, 51 K-ac peptides regarding 45 proteins altered in response to loss of FMRP, which were enriched in energy metabolic processes and were implicated in several neurological disorders. Thus this study for the first time provides a global hippocampal lysine acetylome upon FMRP deficiency linked to abnormal metabolic pathways, which may contribute to pathogenic mechanism of FXS.


Assuntos
Transtorno do Espectro Autista , Síndrome do Cromossomo X Frágil , Deficiência Intelectual , Trifosfato de Adenosina/metabolismo , Animais , Ácidos Carboxílicos , Cromatografia Líquida , Citratos , Modelos Animais de Doenças , Metabolismo Energético , Proteína do X Frágil da Deficiência Intelectual/genética , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/genética , Síndrome do Cromossomo X Frágil/metabolismo , Síndrome do Cromossomo X Frágil/patologia , Hipocampo/metabolismo , Lactatos , Lisina/metabolismo , Camundongos , Camundongos Knockout , Proteômica , Espectrometria de Massas em Tandem
2.
Cell Mol Neurobiol ; 42(3): 777-790, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33058074

RESUMO

Arachidonic acid (AA), a polyunsaturated fatty acid, is involved in the modulation of neuronal excitability in the brain. Arachidonate lipoxygenase 3 (ALOXE3), a critical enzyme in the AA metabolic pathway, catalyzes the derivate of AA into hepoxilins. However, the expression pattern of ALOXE3 and its role in the brain has not been described until now. Here we showed that the levels of Aloxe3 mRNA and protein kept increasing since birth and reached the highest level at postnatal day 30 in the mouse hippocampus and temporal cortex. Histomorphological analyses indicated that ALOXE3 was enriched in adult hippocampus, somatosensory cortex and striatum. The distribution was restricted to the neurites of function-specific subregions, such as mossy fibre connecting hilus and CA3 neurons, termini of Schaffer collateral projections, and the layers III and IV of somatosensory cortex. The spatiotemporal expression pattern of ALOXE3 suggests its potential role in the modulation of neural excitability and seizure susceptibility. In fact, decreased expression of ALOXE3 and elevated concentration of AA in the hippocampus was found after status epilepticus (SE) induced by pilocarpine. Local overexpression of ALOXE3 via adeno-associated virus gene transfer restored the elevated AA level induced by SE, alleviated seizure severities by increasing the latencies to myclonic switch, clonic convulsions and tonic hindlimb extensions, and decreased the mortality rate in the pilocarpine-induced SE model. These results suggest that the expression of ALOXE3 is a crucial regulator of AA metabolism in brain, and potentially acts as a regulator of neural excitability, thereby controlling brain development and seizure susceptibility.


Assuntos
Convulsões , Estado Epiléptico , Animais , Encéfalo/metabolismo , Hipocampo/metabolismo , Camundongos , Pilocarpina , Convulsões/induzido quimicamente , Convulsões/genética , Convulsões/metabolismo , Estado Epiléptico/induzido quimicamente
3.
J Proteomics ; 214: 103633, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31911195

RESUMO

Nicotine, a major addictive component in tobacco, plays an important role in the changes of body weight upon smoking and its cessation. Here we showed that nicotine-treated mice exhibited weight loss and nicotine withdrawal led to weight gain. Using TMT-based proteomic analysis, we obtained the different hypothalamic protein profiles in response to nicotine and its withdrawal. A total of ~5000 proteins were identified from the hypothalamus with 50 altered proteins upon 28-day nicotine treatment and 28 altered proteins upon 15-day nicotine withdrawal. Of the altered proteins, CASP3, LCMT2, GRIN2D, CCNT2, FADS3 and MRPS18B were inversely changed in response to nicotine and withdrawal, coincidence with the change of body weight. Of them, CASP3, LCMT2, GRIN2D and CCNT2 were found to be associated with several GO terms and KEGG pathways linking with cell apoptosis, neurotransmission and metabolism. Further Western blot and RT-qPCR analyses confirmed that the levels of the 4 proteins CASP3, LCMT2, GRIN2D and CCNT2, instead of their mRNA transcripts, altered in response to nicotine and withdrawal. Thus this study provides nicotine- and withdrawal-induced hypothalamic protein profiles and suggests potential roles of these altered proteins in the change of body weight. SIGNIFICANCE: Cigarette smoking is one of important factors harming human health. Most smokers tend to have lower body weights and smoking cessation often lead to overweight or obesity, which is an important reason for smokers to insist on smoking. It is known that nicotine, a critical component in tobacco, is associated with the alteration in body weight by affecting hypothalamic function. Through TMT-based proteomic analysis, this study identified differential hypothalamic protein profiles in response to nicotine treatment and its withdrawal, and 4 nicotine- and withdrawal-induced contrary proteins CASP3, LCMT2, GRIN2D and CCNT2 are involved in several enriched GO terms and KEGG pathways, which are associated with cell apoptosis, neurotransmission and metabolism. Our study may provide novel targets for further investigation of the molecular mechanisms of nicotine- and withdrawal-induced alteration in body weight.


Assuntos
Nicotina , Proteoma , Animais , Peso Corporal , Hipotálamo , Camundongos , Nicotina/efeitos adversos , Proteômica
4.
Neuroscience ; 404: 282-296, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30742966

RESUMO

Fragile X mental retardation protein (FMRP), a key determinant of normal brain development and neuronal plasticity, plays critical roles in nucleocytoplasmic shuttling of mRNAs. However, the factors involved in FMRP nuclear localization remain to be determined. Using cross-species sequence comparison, we show that an aspartate in position 132 (D132), located within the conserved nuclear localization signal (NLS) of FMRP, appears in human and other mammals, while glutamate 132 (E132) appears in rodents and birds. Human FMRP-D132E alters the secondary structure of the protein and reduces its nuclear localization, while the reciprocal substitution in mouse FMRP-E132D promotes its nuclear localization. Human FMRP could interact with poly(A)-binding protein 1 (PABP1) which is impeded by the D132E mutation. Reversely, mouse FMRP could not interact with PABP1, but the E132D mutation leads to the FMRP-PABP1 interaction. We further show that overexpression of human FMRP-D132E mutant promotes the formation of cytoplasmic aggregates of PABP1 in human cells, but not of mouse FMRP-E132D in mouse cells. PABP1 knockdown reduces the nuclear localization of human FMRP, but not mouse FMRP. Furthermore, RNase A treatment decreases the PABP1 levels in the anti-V5-immunoprecipitates using the V5-hFMRP-transfected cells, suggesting an interaction between human FMRP and PABP1 in an RNA-dependent fashion. Thus, our data suggest that the FMRP protein with the human-used D132 accommodates a novel protein-RNA-protein interaction which may implicate a connection between FMRP residue transition and neural evolution.


Assuntos
Núcleo Celular/metabolismo , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Proteína I de Ligação a Poli(A)/metabolismo , RNA/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Proteína do X Frágil da Deficiência Intelectual/química , Proteína do X Frágil da Deficiência Intelectual/genética , Células HEK293 , Humanos , Camundongos , Proteína I de Ligação a Poli(A)/química , Proteína I de Ligação a Poli(A)/genética , Ligação Proteica/fisiologia , Estrutura Secundária de Proteína , RNA/química , RNA/genética , Especificidade da Espécie
5.
J Proteomics ; 185: 39-50, 2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-29953961

RESUMO

Physical exercise has been reported to increase neurotrophin in brain tissues as hippocampus as well as increased neurotrophic level peripherally in blood plasma and might have an effect on/or affect molecular processes of energy metabolism (and homeostasis). In this study, using quantitative proteomic analysis, we obtained a plasma protein profile from the rat with long-term moderate exercise. A total of 752 proteins were identified in the plasma. Among them, 54 proteins were significant up-regulated and 47 proteins were down-regulated in the plasma of exercise group compared with the control group. Bioinformatic analyses showed that these altered proteins are widely involved in multiple biological processes, molecular functions and cellular components, which connect with 11 signaling pathways. Interestingly, 5 up-regulated proteins Rap1b, PTPN11, ARHGDIA, Cdc42 and YWHAE, confirmed by Western blots, are involved in the neurotrophin signaling pathway which shows the lowest P value among the identified pathways. Further analyses showed that the 5 neurotrophin-signaling-pathway-related proteins participate in two important protein-protein interaction networks associated to cell survival and apoptosis, axonal development, synapse formation and plasticity. This study provides an exercise-induced plasma protein profile, suggesting that long-term exercise enhances the proteins involved in neurotrophin signaling pathway which may contribute to health benefit. SIGNIFICANCE: Physical activity contributes to myriad benefits on body health across the lifespan. The changes in plasma proteins after chronic moderate exercise may be used as biomarkers for health and may also play important roles in increase of cardiovascular fitness, enhancement of immune competence, prevention of obesity, decrease of risk for neurological disorders, cancer, stroke, diabetes and other metabolic disorders. Using a TMT-based proteomic method, this study identified 101 altered proteins in the plasma of rats after long-term moderate treadmill running, which may provide novel biomarkers for further investigation of the underlying mechanism of physical exercise. We confirmed that exercise enhances 5 proteins of the neurotrophin signaling pathway that may contribute to health benefits.


Assuntos
Proteínas Sanguíneas/análise , Fatores de Crescimento Neural/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Condicionamento Físico Animal/métodos , Proteômica/métodos , Animais , Proteínas Sanguíneas/química , Proteínas Sanguíneas/metabolismo , Estudos de Casos e Controles , Cromatografia Líquida , Masculino , Neurogênese/fisiologia , Plasma/química , Plasma/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia , Espectrometria de Massas em Tandem , Fatores de Tempo
6.
Mol Neurobiol ; 54(4): 2831-2842, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27013471

RESUMO

Upregulation of sodium channel SCN3A expression in epileptic tissues is known to contribute to enhancing neuronal excitability and the development of epilepsy. Therefore, certain strategies to reduce SCN3A expression may be helpful for seizure control. Here, we reveal a novel role of valproate (VPA) in the epigenetic downregulation of Scn3a expression. We found that VPA, instead of carbamazepine (CBZ) and lamotrigine (LTG), could significantly downregulate Scn3a expression in mouse Neuro-2a cells. Luciferase assays and CpG methylation analyses showed that VPA induced the methylation at the -39C site in Scn3a promoter which decreased the promoter activity. We further showed that VPA downregulated the expression of methyl-CpG-binding domain protein 2 (MBD2) at the posttranscriptional level and knockdown of MBD2 increased Scn3a expression. In addition, we found that VPA induced the expression of fat mass and obesity-associated (FTO) protein and FTO knockdown abolished the repressive effects of VPA on MBD2 and Nav1.3 expressions. Furthermore, VPA, instead of other two anticonvulsant drugs, induced the expressions of Scn3a and Mbd2 and reduced Fto expression in the hippocampus of VPA-treated seizure mice. Taken together, this study suggests an epigenetic pathway for the VPA-induced downregulation of Scn3a expression, which provides a possible role of this pathway in the anticonvulsant action of VPA.


Assuntos
Anticonvulsivantes/farmacologia , Regulação para Baixo/genética , Epigênese Genética/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.3/genética , Ácido Valproico/farmacologia , Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Animais , Anticonvulsivantes/uso terapêutico , Linhagem Celular Tumoral , Ilhas de CpG/genética , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo/efeitos dos fármacos , Genes Reporter , Hipocampo/metabolismo , Masculino , Camundongos , Modelos Biológicos , Canal de Sódio Disparado por Voltagem NAV1.3/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Convulsões/tratamento farmacológico , Convulsões/genética , Transcrição Gênica/efeitos dos fármacos , Ácido Valproico/uso terapêutico
7.
Mol Neurobiol ; 54(4): 2585-2594, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-26993298

RESUMO

Fragile X mental retardation protein (FMRP), associated with fragile X syndrome, is known as an RNA-binding protein to regulate gene expression at post-transcriptional level in the brain. FMRP is also involved in microRNA (miRNA) biogenesis during the process of precursor miRNA (pre-miRNA) into mature miRNA. However, there is no description of the effect of FMRP on primary miRNA (pri-miRNA) processing. Here, we uncover a novel role of FMRP in pri-miRNA processing via controlling Drosha translation. We show that the expression of DROSHA protein, instead of its messenger RNA (mRNA) transcripts, is downregulated in both the hippocampus of Fmr1-knockout mice and the FMRP-knockdown Neuro-2a cells. Overexpression or knockdown FMRP does not alter Drosha mRNA stability. Immunoprecipitation and polysome analyses demonstrate that FMRP binds to the Drosha mRNA and enhances its translation. Additionally, we show that loss of FMRP in Fmr1-deficient mice results in the accumulation of three in six analyzed pri-miRNAs and the reduction of the corresponding pre-miRNAs and mature miRNAs. Thus, our data suggest that FMRP is involved in pri-miRNA processing via enhancing DROSHA expression that may play an important role in fragile X syndrome.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/metabolismo , MicroRNAs/genética , Biossíntese de Proteínas/genética , Processamento Pós-Transcricional do RNA/genética , Ribonuclease III/genética , Animais , Linhagem Celular Tumoral , Regulação para Baixo/genética , Técnicas de Silenciamento de Genes , Camundongos Knockout , MicroRNAs/metabolismo , Ligação Proteica/genética , Estabilidade de RNA/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ribonuclease III/metabolismo , Regulação para Cima/genética
8.
Neuropharmacology ; 113(Pt A): 480-489, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27816501

RESUMO

Abnormal expressions of sodium channel SCN1A and SCN3A genes alter neural excitability that are believed to contribute to the pathogenesis of epilepsy, a long-term risk of recurrent seizures. Ketogenic diet (KD), a high-fat and low-carbohydrate treatment for difficult-to-control (refractory) epilepsy in children, has been suggested to reverse gene expression patterns. Here, we reveal a novel role of GAPDH on the posttranscriptional regulation of mouse Scn1a and Scn3a expressions under seizure and KD conditions. We show that GAPDH binds to a conserved region in the 3' UTRs of human and mouse SCN1A and SCN3A genes, which decreases and increases genes' expressions by affecting mRNA stability through SCN1A 3' UTR and SCN3A 3' UTR, respectively. In seizure mice, the upregulation and phosphorylation of GAPDH enhance its binding to the 3' UTR, which lead to downregulation of Scn1a and upregulation of Scn3a. Furthermore, administration of KD generates ß-hydroxybutyric acid which rescues the abnormal expressions of Scn1a and Scn3a by weakening the GAPDH's binding to the element. Taken together, these data suggest that GAPDH-mediated expression regulation of sodium channel genes may be associated with epilepsy and the anticonvulsant action of KD.


Assuntos
Dieta Cetogênica , Gliceraldeído-3-Fosfato Desidrogenases/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Canal de Sódio Disparado por Voltagem NAV1.3/genética , Convulsões/dietoterapia , Convulsões/genética , Canais de Sódio/genética , Animais , Linhagem Celular Tumoral , Dieta Cetogênica/métodos , Células HEK293 , Humanos , Masculino , Camundongos , Canal de Sódio Disparado por Voltagem NAV1.1/biossíntese , Canal de Sódio Disparado por Voltagem NAV1.3/biossíntese , Ligação Proteica/fisiologia , Processamento Pós-Transcricional do RNA/fisiologia , Convulsões/metabolismo , Canais de Sódio/biossíntese
9.
Mol Neurobiol ; 51(3): 1053-63, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24906954

RESUMO

Fragile X syndrome (FXS), a common form of inherited mental retardation, is caused by a loss of expression of the fragile X mental retardation protein (FMRP). FMRP is involved in brain functions by interacting with mRNAs and microRNAs (miRNAs) that selectively control gene expression at translational level. However, little is known about the role of FMRP in regulating miRNA expression. Here, we found a development-dependant dynamic expression of Fmr1 gene (encoding FMRP) in mouse hippocampus with a small peak at postnatal day 7 (P7). MiRNA microarray analysis showed that the levels of 38 miRNAs showed a significant increase with about 15 ~ 250-folds and the levels of 26 miRNAs showed a significant decrease with only about 2 ~ 4-folds in the hippocampus of P7 Fmr1 knockout (KO) mice. The qRT-PCR assay showed that nine of the most increased miRNAs (>100-folds in microarrays) increased about 40 ~ 70-folds and their pre-miRNAs increased about 5 ~ 10-folds, but no significant difference in their pri-miRNA levels was observed, suggesting that the alterations of partial miRNAs are an indirect consequence of FMRP lacking. We further demonstrated that a set of protein-coding mRNAs, potentially targeted by the nine miRNAs, were down-regulated in the hippocampus of Fmr1 KO mice. Finally, luciferase assays demonstrated that miR-34b, miR-340, and miR-148a could down-regulate the reporter gene expression by interacting with the Met 3' UTR. Taken together, these findings suggest that the miRNA expression alterations resulted from the absence of FMRP might contribute to molecular pathology of FXS.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Síndrome do Cromossomo X Frágil/metabolismo , Perfilação da Expressão Gênica , MicroRNAs/biossíntese , MicroRNAs/genética , Animais , Linhagem Celular Tumoral , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Knockout
10.
Mol Neurobiol ; 50(2): 438-48, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24436055

RESUMO

Voltage-gated sodium channel α subunit type I (Nav1.1, encoded by SCN1A gene) plays a critical role in the initiation of action potential in the central nervous system. Downregulated expression of SCN1A is believed to be associated with epilepsy. Here, we found that the SCN1A promoter (P1c), located at the 5' untranslated exon 1c, drove the reporter gene expression in human NT2 cells, and a region between nt +53 and +62 downstream of the P1c promoter repressed the promoter activity. Further analyses showed that a scaffolding protein RACK1 (receptor for activated C kinase 1) was involved in binding to this silencer. Knockdown of RACK1 expression in NT2 cells deprived the repressive role of the silencer on the P1c promoter and increased SCN1A transcription, suggesting the potential involvement of RACK1 in negatively regulating SCN1A transcription via interaction with the silencer. Furthermore, we demonstrated that the binding of the protein complex including RACK1 to the SCN1A promoter motif was decreased in neuron-like differentiation of the NT2 cells induced by retinoic acid and resulted in the upregulation of SCN1A transcription. Taken together, this study reports a novel role of RACK1 in regulating SCN1A expression that participates in retinoic acid-induced neuronal differentiation of NT2 cells.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Mutação/genética , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Proteínas de Neoplasias/metabolismo , Receptores de Superfície Celular/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Expressão Gênica/genética , Humanos , Proteínas do Tecido Nervoso/metabolismo , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , Receptores de Quinase C Ativada , Transcrição Gênica , Tretinoína/farmacologia
11.
J Genet Genomics ; 38(4): 157-63, 2011 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-21530899

RESUMO

Transcription factors (TFs) play critical roles in the development of the nervous system, but the transcriptional regulatory mechanisms of these genes are poorly understood. Here we analyzed 5-kb of the 5' flanking genomic DNA sequences of 41 TF genes involved in neuronal development. The results showed that the TF genes tend to have higher GC contents in the proximal region and most of the TF genes have at least one proximal GC-rich (GC content > 60%) promoter with a CpG island. The promoter distribution analysis showed that the GC-poor promoters were sporadically distributed within the 5-kb flanking genomic sequence (FGS); however, more than half (37 of 70) of the GC-rich promoters were located in the proximal region between nucleotides -1 and -500. Luciferase assays showed that partial GC-rich promoters increased gene expression in SH-SY5Y cells and that CpG methylation repressed the promoter activity. This study suggests a potential general mechanism for regulation of TF expression.


Assuntos
Ilhas de CpG/genética , Sistema Nervoso/embriologia , Regiões Promotoras Genéticas/genética , Fatores de Transcrição/genética , Composição de Bases/genética , Sequência de Bases , Linhagem Celular Tumoral , Metilação de DNA/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Dados de Sequência Molecular
12.
J Mol Neurosci ; 44(2): 115-21, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21271300

RESUMO

Voltage-gated sodium channel α-subunit type III (Na(v)1.3) is mainly expressed in the central nervous system and is associated with neurological disorders. The expression of mouse Scn3a product (Na(v)1.3) mainly occurs in embryonic and early postnatal brain but not in adult brain. Here, we report for the first time the identification and characterization of the mouse Scn3a gene promoter region and regulation of the promoter activity by GC box and CpG methylation. Luciferase assay showed that the promoter region F1.2 (nt -1,049 to +157) had significantly higher activity in PC12 cells, comparing with that in SH-SY5Y cells and HEK293 cells. A stepwise 5' truncation of the promoter region found that the minimal functional promoter located within the region nt -168 to +157. Deletion of a GC box (nt -254 to -258) in the mouse Scn3a promoter decreased the promoter activity. CpG methylation of the F1.2 without the GC box completely repressed the promoter activity, suggesting that the GC box is a critical element in the CpG-methylated Scn3a promoter. These results suggest that the GC box and CpG methylation might play important roles in regulating mouse Scn3a gene expression.


Assuntos
Ilhas de CpG , Metilação de DNA , Fosfatos de Dinucleosídeos/metabolismo , Regiões Promotoras Genéticas/genética , Canais de Sódio/genética , Animais , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Humanos , Camundongos , Mutagênese Sítio-Dirigida , Canal de Sódio Disparado por Voltagem NAV1.3
13.
Mol Biol Rep ; 37(5): 2157-62, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19669666

RESUMO

Agrobacterium tumefaciens transfers DNA from its Ti plasmid to plant host cells. The genes located within the transferred DNA of Ti plasmid including the octopine synthase gene (OCS) are expressed in plant host cells. The 3'-flanking region of OCS gene, known as OCS terminator, is widely used as a transcriptional terminator of the transgenes in plant expression vectors. In this study, we found the reversed OCS terminator (3'-OCS-r) could drive expression of hygromycin phosphotransferase II gene (hpt II) and beta-glucuronidase gene in Escherichia coli, and expression of hpt II in A. tumefaciens. Furthermore, reverse transcription-polymerase chain reaction analysis revealed that an open reading frame (ORF12) that is located downstream to the 3'-OCS-r was transcribed in A. tumefaciens, which overlaps in reverse with the coding region of the OCS gene in octopine Ti plasmid.


Assuntos
Agrobacterium tumefaciens/enzimologia , Agrobacterium tumefaciens/genética , Aminoácido Oxirredutases/genética , Plasmídeos Indutores de Tumores em Plantas/genética , Células Procarióticas/metabolismo , Regiões Promotoras Genéticas/genética , Regiões Terminadoras Genéticas , Sequência de Bases , Escherichia coli/genética , Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Dados de Sequência Molecular , Fases de Leitura Aberta/genética , Recombinação Genética/genética , Sítio de Iniciação de Transcrição , Transcrição Gênica
14.
J Neurosci Res ; 86(15): 3375-81, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18655196

RESUMO

Voltage-gated sodium channels play critical roles in the excitability of the brain. A decreased level of Na(v)1.1 has been identified as the cause of severe myoclonic epilepsy in infancy. In the present study, we identified the transcription start site and three 5'-untranslated exons of SCN1A by using 5'-full RACE. The 2.5-kb region upstream of the transcription start site was targeted as a potential location of the promoter. The 2.5-kb genomic fragment (P(2.5), from +26 to -2,500) and the 2.7-kb fragment (P(2.7), P(2.5) combined with the 227-bp 5'-untranslated exons) were cloned to produce luciferase constructs. The P(2.5) and the P(2.7) drove luciferase gene expression in the human neuroblastoma cell line SH-SY5Y but not in the human embryonic kidney cell line HEK-293. The 5'-untranslated exons could greatly enhance gene expression in SH-SY5Y cells. The P(2.7) could be used as a functional unit to study the role of SCN1A noncoding sequences in gene expression. These findings will also help in exploring the possibility of promoter mutant-induced diseases and revealing the mechanism underlying the regulation of SCN1A expression in the normal brain.


Assuntos
Regiões 5' não Traduzidas/genética , Éxons/genética , Regulação da Expressão Gênica , Proteínas do Tecido Nervoso/genética , Regiões Promotoras Genéticas/genética , Canais de Sódio/genética , Sequência de Bases , Encéfalo/fisiologia , Linhagem Celular , Expressão Gênica , Humanos , Dados de Sequência Molecular , Canal de Sódio Disparado por Voltagem NAV1.1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA