Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proteomics ; 18(10): e1700064, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29645342

RESUMO

The number of small proteins (SPs) encoded in the Escherichia coli genome is unknown, as current bioinformatics and biochemical techniques make short gene and small protein identification challenging. One method of small protein identification involves adding an epitope tag to the 3' end of a short open reading frame (sORF) on the chromosome, with synthesis confirmed by immunoblot assays. In this study, this strategy was used to identify new E. coli small proteins, tagging 80 sORFs in the E. coli genome, and assayed for protein synthesis. The selected sORFs represent diverse sequence characteristics, including degrees of sORF conservation, predicted transmembrane domains, sORF direction with respect to flanking genes, ribosome binding site (RBS) prediction, and ribosome profiling results. Of 80 sORFs, 36 resulted in encoded synthesized proteins-a 45% success rate. Modeling of detected versus non-detected small proteins analysis showed predictions based on RBS prediction, transcription data, and ribosome profiling had statistically-significant correlation with protein synthesis; however, there was no correlation between current sORF annotation and protein synthesis. These results suggest substantial numbers of small proteins remain undiscovered in E. coli, and existing bioinformatics techniques must continue to improve to facilitate identification.


Assuntos
Biologia Computacional/métodos , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Anotação de Sequência Molecular , Fases de Leitura Aberta , Biossíntese de Proteínas , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Genoma Bacteriano , Ribossomos
2.
PLoS Med ; 5(6): e117, 2008 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-18593214

RESUMO

BACKGROUND: Prenatal exposure of the developing brain to cocaine causes morphological and behavioral abnormalities. Recent studies indicate that cocaine-induced proliferation inhibition and/or apoptosis in neural progenitor cells may play a pivotal role in causing these abnormalities. To understand the molecular mechanism through which cocaine inhibits cell proliferation in neural progenitors, we sought to identify the molecules that are responsible for mediating the effect of cocaine on cell cycle regulation. METHODS AND FINDINGS: Microarray analysis followed by quantitative real-time reverse transcription PCR was used to screen cocaine-responsive and cell cycle-related genes in a neural progenitor cell line where cocaine exposure caused a robust anti-proliferative effect by interfering with the G1-to-S transition. Cyclin A2, among genes related to the G1-to-S cell cycle transition, was most strongly down-regulated by cocaine. Down-regulation of cyclin A was also found in cocaine-treated human primary neural and A2B5+ progenitor cells, as well as in rat fetal brains exposed to cocaine in utero. Reversing cyclin A down-regulation by gene transfer counteracted the proliferation inhibition caused by cocaine. Further, we found that cocaine-induced accumulation of reactive oxygen species, which involves N-oxidation of cocaine via cytochrome P450, promotes cyclin A down-regulation by causing an endoplasmic reticulum (ER) stress response, as indicated by increased phosphorylation of eIF2alpha and expression of ATF4. In the developing rat brain, the P450 inhibitor cimetidine counteracted cocaine-induced inhibition of neural progenitor cell proliferation as well as down-regulation of cyclin A. CONCLUSIONS: Our results demonstrate that down-regulation of cyclin A underlies cocaine-induced proliferation inhibition in neural progenitors. The down-regulation of cyclin A is initiated by N-oxidative metabolism of cocaine and consequent ER stress. Inhibition of cocaine N-oxidative metabolism by P450 inhibitors may provide a preventive strategy for counteracting the adverse effects of cocaine on fetal brain development.


Assuntos
Cocaína/farmacologia , Neurônios/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Animais , Ciclo Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular , Células Cultivadas , Cimetidina/farmacologia , Ciclina A/genética , Ciclina A/metabolismo , Ciclina A2 , Regulação para Baixo , Feminino , Humanos , Fosforilação , Gravidez , Ratos , Ratos Sprague-Dawley , Transfecção
3.
Mol Cell Biol ; 23(13): 4649-62, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12808104

RESUMO

Recent studies have revealed a positive correlation between astrocyte apoptosis and rapid disease progression in persons with neurodegenerative diseases. Glycogen synthase kinase 3beta (GSK-3beta) is a molecular regulator of cell fate in the central nervous system and a target of the phosphatidylinositol 3-kinase (PI-3K) pathway. We have therefore examined the role of the PI-3K pathway, and of GSK-3beta, in regulating astrocyte survival. Our studies indicate that inhibition of PI-3K leads to apoptosis in primary cortical astrocytes. Furthermore, overexpression of a constitutively active GSK-3beta mutant (S9A) is sufficient to cause astrocyte apoptosis, whereas an enzymatically inactive GSK-3beta mutant (K85M) has no effect. In light of reports on the interplay between GSK-3beta and nuclear factor kappaB (NF-kappaB), and because of the antiapoptotic activity of NF-kappaB, we examined the effect of GSK-3beta overexpression on NF-kappaB activation. These experiments revealed strong inhibition of NF-kappaB activation in astrocytes upon overexpression of the S9A, but not the K85M, mutant of GSK-3beta. This was accompanied by stabilization of the NF-kappaB-inhibitory protein, IkappaBalpha and down-regulation of IkappaB kinase (IKK) activity. These findings therefore implicate GSK-3beta as a regulator of NF-kappaB activation in astrocytes and suggest that the pro-apoptotic effects of GSK-3beta may be mediated at least in part through the inhibition of NF-kappaB pathway.


Assuntos
Apoptose , Astrócitos/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Astrócitos/citologia , Células Cultivadas , Ciclo-Oxigenase 2 , DNA/metabolismo , DNA Complementar/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Glutationa Transferase/metabolismo , Glicogênio Sintase Quinase 3 beta , Quinase I-kappa B , Immunoblotting , Marcação In Situ das Extremidades Cortadas , Isoenzimas/metabolismo , Luciferases/metabolismo , Mutação , NF-kappa B/antagonistas & inibidores , Fosfatidilinositol 3-Quinases/metabolismo , Testes de Precipitina , Prostaglandina-Endoperóxido Sintases/metabolismo , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Transcrição Gênica
4.
Brain Res ; 1112(1): 1-15, 2006 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-16901471

RESUMO

AF5 neural cells derived from fetal rat mesencephalic tissue were immortalized with a truncated SV40 LT vector lacking the p53-inactivating domain to maintain long-term cultures with a p53-responsive phenotype. This study examined p53 function in producing programmed cell death in propagating AF5 neural cells after exposure to hydrogen peroxide (H2O2) and the kinase inhibitor staurosporine (STSP). Concentration-dependent exposure of AF5 cells to 0-800 mM H2O2 and STSP at 0-1000 nM revealed increasing cytotoxicity from MTS cell viability assays. Apoptosis occurred at 400 mM H2O2 as evidenced by subG1 DNA and Annexin V flow cytometry analyses and cellular immunofluorescence staining with propidium iodide, anti-Annexin V and DAPI. DNA fragmentation, caspase-3/7 activity and cytochrome c release into cytosol also confirmed H2O2-mediated apoptotic events. p53 protein levels were increased over 24 h by H2O2 in a coordinated fashion with mdm2 expression. p53 activation by H2O2 was evidenced by elevated Ser15 phosphorylation, increased luciferase p53 reporter activity and upregulation of the downstream p53 targets p21(waf1) and apoptotic proteins, bax, Noxa and PUMA. STSP exposure produced apoptosis demonstrated by DNA fragmentation, caspase-3/7 activity, cytochrome c release and over 24 h was accompanied by sustained increase in p53 and Ser15 phosphorylation, rise in p21(waf1) and bax and a transient increase in p53 reporter activity but without Annexin V binding. These findings demonstrate that AF5 cells undergo apoptosis in response to H2O2-mediated oxidative stress and signal pathway disruption by STSP that therefore would be useful in studies related to p53-dependent neuronal cell death and neurodegeneration.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Peróxido de Hidrogênio/farmacologia , Neurônios/efeitos dos fármacos , Oxidantes/farmacologia , Estaurosporina/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Análise de Variância , Animais , Anexina A5/metabolismo , Western Blotting/métodos , Caspases/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Citometria de Fluxo/métodos , Imunofluorescência/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Marcação In Situ das Extremidades Cortadas/métodos , Proteínas do Tecido Nervoso/metabolismo , Ratos , Fatores de Tempo
5.
Brain Res ; 1010(1-2): 55-61, 2004 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-15126117

RESUMO

We have previously demonstrated that pretreatment with bone morphogenetic protein 7 (BMP7), a trophic factor in the TGFbeta superfamily, reduces ischemia-induced brain infarction induced by middle cerebral artery ligation in rats. Since the mitogen-activated protein kinase (MAPK) pathway is involved in many TGFbeta-mediated responses, we examined the interaction of BMP7 and MAPK in primary cultures obtained from the cerebral cortex of E16-17 rat embryos. Lactate dehydrogenase (LDH) in the media was used as an index of cell death. BMP7 did not alter LDH levels at low concentration (1.25 nM), but exhibited increased cellular toxicity at higher concentration (>12.5 nM). BMP7 at the low concentration significantly attenuated H2O2-induced increases in LDH activity and decreases in neuronal density. Pharmacological interactions were used to examine if MAPK was involved in this response. BMP7-induced protection was antagonized by the p42,44 MAPK kinase inhibitors PD98059 and U0125. The p38 MAPK antagonist SB203580, and their inactive analog SB202474, also attenuated BMP7-induced protection, suggesting that the interaction with p38 MAPK is nonspecific. Previous studies have indicated that SB202474 has inhibitory effects on other protein kinases. We found that the protein kinase C inhibitor chelerythrine antagonized BMP7-induced protection against H2O2. Western blot analysis indicated that BMP7 increased phosphorylation of p42,44 MAPK and PKC. Taken together, our data suggest that BMP7 is neuroprotective at low concentrations in primary cortical cell culture. The protective effects of BMP7 may involve the activation of p42,44 MAPK and PKC.


Assuntos
Proteínas Morfogenéticas Ósseas/farmacologia , Córtex Cerebral/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Proteína Quinase C/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , Animais , Proteína Morfogenética Óssea 7 , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/enzimologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Peróxido de Hidrogênio/antagonistas & inibidores , Peróxido de Hidrogênio/toxicidade , L-Lactato Desidrogenase/efeitos dos fármacos , L-Lactato Desidrogenase/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína Quinase 1 Ativada por Mitógeno/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Oxidantes/antagonistas & inibidores , Oxidantes/toxicidade , Fosforilação/efeitos dos fármacos , Proteínas de Plantas , Proteína Quinase C/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Quinases p38 Ativadas por Mitógeno
6.
J Addict Med ; 3(4): 218-26, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20948987

RESUMO

OBJECTIVES: Prenatal exposure to cocaine causes cytoarchitectural alterations in the developing neocortex. Previously, we reported that cocaine inhibits neural progenitor cell proliferation through oxidative endoplasmic reticulum stress and consequent down-regulation of cyclin A, whereas cyclin A expression was increased in astrocytes. In the present study, cell type-specific responses to cocaine were further explored. METHODS: Gene expression profiles were examined in five types of cells obtained from the human fetal cerebral cortex at 20 weeks gestation. Cells were treated with 100 µM cocaine in vitro for 24 hr, followed by gene expression analysis using a human neural/stem cell/drug abuse-focused cDNA array, with verification by quantitative real-time RT-PCR. RESULTS: Cocaine influenced transcription of distinct categories of genes in a cell type-specific manner. Cocaine down-regulated cytoskeleton-related genes including ezrin, γ2 actin, α3d tubulin and α8 tubulin in neural and/or A2B5+ progenitor cells. In contrast, cocaine modulated immune and cell death-related genes in microglia and astrocytes. In microglia, cocaine up-regulated the immunoregulatory and pro-apoptotic genes IL-1ß and BAX. In astrocytes, cocaine down-regulated the immune response gene glucocorticoid receptor and up-regulated the anti-apoptotic genes 14-3-3 ε and HVEM. Therefore, cell types comprising the developing neocortex show differential responses to cocaine. CONCLUSIONS: These data suggest that cocaine causes cytoskeletal abnormalities leading to disturbances in neural differentiation and migration in progenitor cells, while altering immune and apoptotic responses in glia. Understanding the mechanisms of cocaine's effects on human CNS cells may help in the development of therapeutic strategies to prevent or ameliorate cocaine-induced impairments in fetal brain development.

7.
Cell Tissue Res ; 324(1): 1-8, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16408195

RESUMO

We have previously described an immortal rat central-nervous-system progenitor cell line, AF5, which is able to exit the cell cycle and assume a differentiated state with neuronal properties. The phenotypic specification of differentiated AF5 cells, however, is not known. In the present study, when induced to differentiate by serum starvation in Neurobasal medium, AF5 cells down-regulate glial fibrillary acidic protein and up-regulate expression of beta-III-tubulin, medium-molecular-weight neurofilament protein, and neuronal growth-associated protein 43. Expression of the gamma-aminobutyric acid (GABA) lineage marker, glutamic acid decarboxylase 67 (GAD67), increases during differentiation, suggesting that AF5 cells adopt a GABAergic lineage. Time-course analysis of the GABAergic neuron specification transcription factor, Pitx2, by reverse transcription/polymerase chain reaction, has shown an increase in the Pitx2 transcript 48 h after initiation of differentiation. In differentiated AF5 cells, expression of the Pitx2 target gene products GAD65 and GABA transporter-1 increases. Cellular GABA levels in differentiated AF5 cells increase by about 26-fold, and GABA release into the medium is 150-fold higher compared with that of undifferentiated cells. Therefore, AF5 cells can be induced to differentiate to a neuronal phenotype with a GABAergic lineage.


Assuntos
Diferenciação Celular , Linhagem da Célula , Neurônios/citologia , Células-Tronco/citologia , Ácido gama-Aminobutírico/metabolismo , Animais , Células Cultivadas , Meios de Cultura/farmacologia , Regulação da Expressão Gênica , Glutamato Descarboxilase/metabolismo , Proteínas de Homeodomínio/metabolismo , Isoenzimas/metabolismo , Ratos , Fatores de Transcrição/metabolismo , Ácido gama-Aminobutírico/biossíntese , Proteína Homeobox PITX2
8.
Exp Neurol ; 191 Suppl 1: S45-59, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15629761

RESUMO

Immortalized central nervous system (CNS) cell lines are useful as in vitro models for innumerable purposes such as elucidating biochemical pathways, studies of effects of drugs, and ultimately, such cells may also be useful for neural transplantation. The SV40 large T (LT) oncoprotein, commonly used for immortalization, interacts with several cell cycle regulatory factors, including binding and inactivating p53 and retinoblastoma family cell-cycle regulators. In an attempt to define the minimal requirements of SV40 T antigen for immortalizing cells of CNS origin, we constructed T155c, encoding the N-terminal 155 amino acids of LT. The p53 binding region is known to reside in the C-terminal region of LT. An additional series of mutants was produced to further narrow the molecular targets for immortalization, and plasmid vectors were constructed for each. In a p53 temperature sensitive cell line model, T64-7B, expression of T155c and all constructs having mutations outside of the first 82 amino acids were capable of overriding cell-cycle block at the non-permissive growth temperature. Several cell lines were produced from fetal rat mesencephalic and cerebral cortical cultures using the T155c construct. The E107K construct contained a mutation in the Rb binding region, but was nonetheless capable of overcoming cell cycle block in T64-7B cell and immortalizing primary cultured cells. Cells immortalized with T155c were often highly dependent on the presence of bFGF for growth. Telomerase activity, telomere length, growth rates, and integrity of the p53 gene in cells immortalized with T155c did not change over 100 population doublings in culture, indicating that cells immortalized with T155c were generally stable during long periods of continuous culture.


Assuntos
Antígenos Virais de Tumores/genética , Transformação Celular Viral/genética , Córtex Cerebral/citologia , Mesencéfalo/citologia , Fragmentos de Peptídeos/genética , Vírus 40 dos Símios/genética , Animais , Ciclo Celular/genética , Divisão Celular/efeitos dos fármacos , Linhagem Celular Transformada , Células Cultivadas , Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Células Clonais , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Mesencéfalo/embriologia , Mesencéfalo/metabolismo , Mutagênese Sítio-Dirigida , Fragmentos de Peptídeos/biossíntese , Ratos , Ratos Sprague-Dawley , Telomerase/metabolismo , Telômero/química , Telômero/metabolismo , Temperatura , Transfecção , Proteína Supressora de Tumor p53/genética
9.
Mol Pharmacol ; 63(6): 1238-47, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12761333

RESUMO

The expression of inducible nitric-oxide synthase (iNOS) in vascular smooth muscle cells leads to prolonged vasorelaxation in vivo and contributes to the profound vasodilation induced by bacterial lipopolysaccharide (LPS) in septic shock. This induction of iNOS depends, in large part, on activation of nuclear factor (NF)-kappaB. Hypertonicity regulates the activity of NF-kappaB in different cell lines; as such, we propose that it should also regulate the expression of iNOS. Thus, the goal of this study was to determine whether hypertonicity regulates iNOS expression and function in smooth muscle cells and to elucidate the mechanism(s) underlying this process. Treatment of hamster ductus deferens (DDT1MF-2) cells and porcine aortic smooth muscle cells with either mannitol (50 mM) or NaCl (50 mM) reduced LPS-stimulated iNOS expression and nitric oxide release. Both of these agents also reduced the activation of NF-kappaB induced by LPS, tumor necrosis factor-alpha and interleukin-1beta in smooth muscle cells. This inhibitory action was caused by suppression of IkappaB-alpha phosphorylation, a prerequisite for ubiquitination and degradation of this protein, and showed additivity with N-benzoyloxycarbonyl (Z)-Leu-Leu-leucinal (MG-132), an inhibitor of proteasomal degradation of IkappaB-alpha. Furthermore, exposure to mannitol inhibited the activity of IkappaB kinase, an enzyme involved in phosphorylation of IkappaB-alpha. Mannitol was unable to affect the induction of iNOS produced by overexpression of RelA in DDT1MF-2 cells, suggesting that this agent does not have additional downstream inhibitory actions on this activated NF-kappaB subunit. Taken together, these data suggest that these hypertonic solutions may prove useful as anti-inflammatory agents, especially against conditions associated with increased NF-kappaB activity.


Assuntos
Lipopolissacarídeos/farmacologia , Manitol/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , NF-kappa B/metabolismo , Óxido Nítrico Sintase/biossíntese , Animais , Cricetinae , Diuréticos Osmóticos/farmacologia , Interações Medicamentosas , Expressão Gênica/efeitos dos fármacos , Quinase I-kappa B , Músculo Liso Vascular/enzimologia , NF-kappa B/antagonistas & inibidores , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Proteínas Serina-Treonina Quinases/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/efeitos dos fármacos , Suínos
10.
Stem Cells ; 21(6): 647-53, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14595124

RESUMO

Three promoters, cellular polypeptide chain elongation factor 1 alpha (EF1), cytomegalovirus (CMV), and Rous sarcoma virus (RSV) were examined for stable transgene expression in mouse embryonic stem (ES) cells and their progeny during dopaminergic neural differentiation. In undifferentiated ES cells the EF1 promoter was highly effective, while CMV had moderate activity. After 3 months in culture, expression of humanized renilla green fluorescent protein (hrGFP) was unchanged for the EF1 promoter and decreased for CMV. At the nestin-positive stage of differentiation, hrGFP and nestin were colocalized in about 20% of cells for EF1, in contrast to 80% of cells for the CMV promoter. In tyrosine hydroxylase (TH)-positive neurons neither the EF1 nor CMV promoter were effective. The RSV promoter was inactive in undifferentiated, nestin-positive, and TH-positive cells. Thus, EF1 and CMV are effective promoters for transgene expression in undifferentiated ES cells and nestin-positive neural precursors.


Assuntos
Diferenciação Celular , Dopamina/metabolismo , Embrião de Mamíferos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Pluripotentes/metabolismo , Regiões Promotoras Genéticas/fisiologia , Animais , Vírus do Sarcoma Aviário/genética , Vírus do Sarcoma Aviário/metabolismo , Linhagem Celular , Citomegalovirus/genética , Citomegalovirus/metabolismo , Embrião de Mamíferos/metabolismo , Vetores Genéticos , Proteínas de Fluorescência Verde , Humanos , Proteínas de Filamentos Intermediários/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Nestina , Neurônios/citologia , Neurônios/metabolismo , Fator 1 de Elongação de Peptídeos/genética , Fator 1 de Elongação de Peptídeos/metabolismo , Células-Tronco Pluripotentes/citologia , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA