Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biosci Trends ; 14(2): 144-150, 2020 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-32295993

RESUMO

The ST3GAL4 gene encodes the enzyme Galß1-4GlcNAc α2,3 sialyltransferase (ST3Gal IV). This enzyme participates in the synthesis of the sialyl Lewis x antigen. In different cancer types altered expression of this antigen has been reported. The transcriptional regulation of this gene is very complex, different mRNA variants (V1-V10) have been reported and are originated by the activity of different promoters and alternative splicing. Only the promoter that gives rise to the V3 variant has not been previously reported. The objective of this work was to identify and characterize the V3 promoter of the ST3GAL4 gene. For this, the putative V3 promoter of the ST3GAL4 gene was delimited by in silico analysis. The complete promoter and smaller versions were cloned in a reporter plasmid. The constructs were transfected in the HaCaT cells and the promoter activity was evaluated by luciferase reporter assays. The cloned region showed promoter activity, and the basal activity was not dependent on TATA boxes. However, the GC boxes, an initiator element (Inr) and downstream promoter element (DPE) could contribute to basal activity. The promoter contains several binding sites for the nuclear factor of activated T-cells (NFAT) that could participate in inducible activity during the immune response. The minimal promoter corresponds to a fragment of approximately 300 bp, located in the position -347 b to -40 b. The characterization of the V3 promoter of the ST3GAL4 gene completes the study of the four promoters of this gene, this contributes to the understanding of its complex transcription regulation.


Assuntos
Regiões Promotoras Genéticas , Sialiltransferases/genética , Transcrição Gênica , Processamento Alternativo , Sítios de Ligação , Linhagem Celular , Clonagem Molecular , Biologia Computacional , Humanos , Isoenzimas/genética , Fatores de Transcrição NFATC/metabolismo
2.
Infect Agent Cancer ; 13: 25, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30038662

RESUMO

BACKGROUND: Aberrant glycosylation is a characteristic of tumour cells. The expression of certain glycan structures has been associated with poor prognosis. In cervical carcinoma, changes in the expression levels of some glycogenes have been associated with lymph invasion. Human papillomavirus (HPV) infection is one of the most important factors underlying the development of cervical cancer. The HPV oncoproteins E6 and E7 have been implicated in cervical carcinogenesis and can modify the host gene expression profile. The roles of these oncoproteins in glycosylation changes have not been previously reported. METHODS: To determine the effect of the E6 and E7 oncoproteins on glycogene expression we partially silenced the E6 and E7 oncogenes in HeLa cells, we performed a microarray expression assay to identify altered glycogenes and quantified the mRNA levels of glycogenes by RT-qPCR. A protein-protein interaction network was constructed to identify potentially altered glycosylation pathways. RESULTS: The microarray analysis showed 9 glycogenes that were upregulated and 7 glycogenes that were downregulated in HeLa shE6/E7 cells. Some of these genes participate in glycosylation related to Notch proteins and O-glycans antigens. CONCLUSIONS: Our results support that E6 and E7 oncoproteins could modify glycogene expression the products of which participate in the synthesis of structures implicated in proliferation, adhesion and apoptosis.

3.
Stem Cells Int ; 2012: 980941, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22619684

RESUMO

Progress in understanding neurodegenerative cell biology in Parkinson's disease (PD) has been hampered by a lack of predictive and relevant cellular models. In addition, the lack of an adequate in vitro human neuron cell-based model has been an obstacle for the uncover of new drugs for treating PD. The ability to generate induced pluripotent stem cells (iPSCs) from PD patients and a refined capacity to differentiate these iPSCs into DA neurons, the relevant disease cell type, promises a new paradigm in drug development that positions human disease pathophysiology at the core of preclinical drug discovery. Disease models derived from iPSC that manifest cellular disease phenotypes have been established for several monogenic diseases, but iPSC can likewise be used for phenotype-based drug screens in complex diseases for which the underlying genetic mechanism is unknown. Here, we highlight recent advances as well as limitations in the use of iPSC technology for modelling PD "in a dish" and for testing compounds against human disease phenotypes in vitro. We discuss how iPSCs are being exploited to illuminate disease pathophysiology, identify novel drug targets, and enhance the probability of clinical success of new drugs.

4.
J Cell Biol ; 194(3): 489-503, 2011 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-21807879

RESUMO

Coordination between functionally related adjacent tissues is essential during development. For example, formation of trunk neural crest cells (NCCs) is highly influenced by the adjacent mesoderm, but the molecular mechanism involved is not well understood. As part of this mechanism, fibroblast growth factor (FGF) and retinoic acid (RA) mesodermal gradients control the onset of neurogenesis in the extending neural tube. In this paper, using gain- and loss-of-function experiments, we show that caudal FGF signaling prevents premature specification of NCCs and, consequently, premature epithelial-mesenchymal transition (EMT) to allow cell emigration. In contrast, rostrally generated RA promotes EMT of NCCs at somitic levels. Furthermore, we show that FGF and RA signaling control EMT in part through the modulation of elements of the bone morphogenetic protein and Wnt signaling pathways. These data establish a clear role for opposition of FGF and RA signaling in control of the timing of NCC EMT and emigration and, consequently, coordination of the development of the central and peripheral nervous system during vertebrate trunk elongation.


Assuntos
Transição Epitelial-Mesenquimal , Fatores de Crescimento de Fibroblastos/metabolismo , Crista Neural/citologia , Tretinoína/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Morfogenéticas Ósseas/fisiologia , Ciclo Celular , Movimento Celular , Sistema Nervoso Central/embriologia , Embrião de Galinha , Eletroporação , Transição Epitelial-Mesenquimal/genética , Regulação da Expressão Gênica no Desenvolvimento , Sistema de Sinalização das MAP Quinases , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Crista Neural/metabolismo , Crista Neural/fisiologia , Sistema Nervoso Periférico/embriologia , Reação em Cadeia da Polimerase , Transdução de Sinais , Fatores de Transcrição/biossíntese , Proteínas Wnt/metabolismo
5.
EMBO Rep ; 11(6): 466-72, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20448664

RESUMO

Genes of the SOX family of high-mobility group transcription factors are essential during nervous system development. In this study, we show that SOX5 is expressed by neural progenitors in the chick spinal cord and is turned off as differentiation proceeds. The overexpression of SOX5 in neural progenitors causes premature cell cycle exit and prevents terminal differentiation. Conversely, knocking down SOX5 protein extends the proliferative period of neural progenitors and causes marked cell death in a dorsal interneuron (dI3) population. Furthermore, SOX5 reduces WNT-beta-catenin signalling, thereby triggering the expression of the negative regulator of the pathway axin2. We propose that SOX5 regulates the timing of cell cycle exit by opposing WNT-beta-catenin activity on cell cycle progression.


Assuntos
Ciclo Celular , Neurônios/citologia , Fatores de Transcrição SOXD/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Ciclo Celular/genética , Diferenciação Celular/genética , Embrião de Galinha , Regulação para Baixo/genética , Regulação da Expressão Gênica no Desenvolvimento , Interneurônios/citologia , Interneurônios/metabolismo , Fatores de Transcrição SOXD/genética , Medula Espinal/citologia , Medula Espinal/embriologia , Medula Espinal/metabolismo , Células-Tronco/citologia , Fatores de Tempo , beta Catenina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...