Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Hum Mol Genet ; 22(22): 4502-15, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23804752

RESUMO

Lesch-Nyhan syndrome (LNS) is a neurodevelopmental disorder caused by mutations in the gene encoding the purine metabolic enzyme hypoxanthine-guanine phosphoribosyltransferase (HPRT). A series of motor, cognitive and neurobehavioral anomalies characterize this disease phenotype, which is still poorly understood. The clinical manifestations of this syndrome are believed to be the consequences of deficiencies in neurodevelopmental pathways that lead to disordered brain function. We have used microRNA array and gene ontology analysis to evaluate the gene expression of differentiating HPRT-deficient human neuron-like cell lines. We set out to identify dysregulated genes implicated in purine-based cellular functions. Our approach was based on the premise that HPRT deficiency affects preeminently the expression and the function of purine-based molecular complexes, such as guanine nucleotide exchange factors (GEFs) and small GTPases. We found that several microRNAs from the miR-17 family cluster and genes encoding GEF are dysregulated in HPRT deficiency. Most notably, our data show that the expression of the exchange protein activated by cAMP (EPAC) is blunted in HPRT-deficient human neuron-like cell lines and fibroblast cells from LNS patients, and is altered in the cortex, striatum and midbrain of HPRT knockout mouse. We also show a marked impairment in the activation of small GTPase RAP1 in the HPRT-deficient cells, as well as differences in cytoskeleton dynamics that lead to increased motility for HPRT-deficient neuron-like cell lines relative to control. We propose that the alterations in EPAC/RAP1 signaling and cell migration in HPRT deficiency are crucial for neuro-developmental events that may contribute to the neurological dysfunctions in LNS.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/metabolismo , Síndrome de Lesch-Nyhan/genética , MicroRNAs/genética , Proteínas rap1 de Ligação ao GTP/metabolismo , Animais , Linhagem Celular , Movimento Celular/fisiologia , Córtex Cerebral/metabolismo , Corpo Estriado/metabolismo , Citoesqueleto/metabolismo , Ontologia Genética , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Hipoxantina Fosforribosiltransferase/deficiência , Hipoxantina Fosforribosiltransferase/genética , Síndrome de Lesch-Nyhan/enzimologia , Masculino , Mesencéfalo/metabolismo , Camundongos , Camundongos Knockout , MicroRNAs/metabolismo , Família Multigênica , Análise de Sequência com Séries de Oligonucleotídeos , Transdução de Sinais , Proteínas rap1 de Ligação ao GTP/genética
2.
PLoS One ; 11(3): e0152692, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27031987

RESUMO

Cardiac glycosides (CGs), inhibitors of Na+/K+-ATPase (NKA), used clinically to treat heart failure, have garnered recent attention as potential anti-cancer and anti-viral agents. A high-throughput phenotypic screen designed to identify modulators of promyelocytic leukemia protein (PML) nuclear body (NB) formation revealed the CG gitoxigenin as a potent activator of PML. We demonstrate that multiple structurally distinct CGs activate the formation of PML NBs and induce PML protein SUMOylation in an NKA-dependent fashion. CG effects on PML occur at the post-transcriptional level, mechanistically distinct from previously described PML activators and are mediated through signaling events downstream of NKA. Curiously, genomic deletion of PML in human cancer cells failed to abrogate the cytotoxic effects of CGs and other apoptotic stimuli such as ceramide and arsenic trioxide that were previously shown to function through PML in mice. These findings suggest that alternative pathways can compensate for PML loss to mediate apoptosis in response to CGs and other apoptotic stimuli.


Assuntos
Glicosídeos Cardíacos/farmacologia , Proteínas Nucleares/metabolismo , Sumoilação/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Apoptose/efeitos dos fármacos , Glicosídeos Cardíacos/química , Chlorocebus aethiops , Deleção de Genes , Células HEK293 , Células HeLa , Humanos , Proteínas Nucleares/genética , Proteína da Leucemia Promielocítica , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , ATPase Trocadora de Sódio-Potássio/metabolismo , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Células Vero
3.
PLoS One ; 9(5): e96575, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24804781

RESUMO

Lesch-Nyhan Syndrome (LNS) is a neurodevelopmental disorder caused by mutations in the gene encoding the purine metabolic enzyme hypoxanthine-guanine phosphoribosyltransferase (HPRT). This syndrome is characterized by an array of severe neurological impairments that in part originate from striatal dysfunctions. However, the molecular and cellular mechanisms underlying these dysfunctions remain largely unidentified. In this report, we demonstrate that HPRT-deficiency causes dysregulated expression of key genes essential for striatal patterning, most notably the striatally-enriched transcription factor B-cell leukemia 11b (Bcl11b). The data also reveal that the down-regulated expression of Bcl11b in HPRT-deficient immortalized mouse striatal (STHdh) neural stem cells is accompanied by aberrant expression of some of its transcriptional partners and other striatally-enriched genes, including the gene encoding dopamine- and cAMP-regulated phosphoprotein 32, (DARPP-32). Furthermore, we demonstrate that components of the BDNF/TrkB signaling, a known activator of DARPP-32 striatal expression and effector of Bcl11b transcriptional activation are markedly increased in HPRT-deficient cells and in the striatum of HPRT knockout mouse. Consequently, the HPRT-deficient cells display superior protection against reactive oxygen species (ROS)-mediated cell death upon exposure to hydrogen peroxide. These findings suggest that the purine metabolic defect caused by HPRT-deficiency, while it may provide neuroprotection to striatal neurons, affects key genes and signaling pathways that may underlie the neuropathogenesis of LNS.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Corpo Estriado/patologia , Fosfoproteína 32 Regulada por cAMP e Dopamina/genética , Hipoxantina Fosforribosiltransferase/genética , Síndrome de Lesch-Nyhan/genética , Receptor trkB/genética , Transdução de Sinais/genética , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Morte Celular/genética , Diferenciação Celular/genética , Corpo Estriado/metabolismo , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Síndrome de Lesch-Nyhan/metabolismo , Síndrome de Lesch-Nyhan/patologia , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptor trkB/metabolismo
4.
PLoS One ; 8(5): e63333, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23691025

RESUMO

Lesch-Nyhan Disease (LND) is the result of mutations in the X-linked gene encoding the purine metabolic enzyme, hypoxanthine guanine phosphoribosyl transferase (HPRT). LND gives rise to severe neurological anomalies including mental retardation, dystonia, chorea, pyramidal signs and a compulsive and aggressive behavior to self injure. The neurological phenotype in LND has been shown to reflect aberrant dopaminergic signaling in the basal ganglia, however there are little data correlating the defect in purine metabolism to the neural-related abnormalities. In the present studies, we find that HPRT-deficient neuronal cell lines have reduced CREB (cAMP response element-binding protein) expression and intracellular cyclic AMP (cAMP), which correlates with attenuated CREB-dependent transcriptional activity and a reduced phosphorylation of protein kinase A (PKA) substrates such as synapsin (p-syn I). Of interest, we found increased expression of phosphodiesterase 10A (PDE10A) in HPRT-deficient cell lines and that the PDE10 inhibitor papaverine and PDE10A siRNA restored cAMP/PKA signaling. Furthermore, reconstitution of HPRT expression in mutant cells partly increased cAMP signaling synapsin phosphorylation. In conclusion, our data show that HPRT-deficiency alters cAMP/PKA signaling pathway, which is in part due to the increased of PDE10A expression and activity. These findings suggest a mechanistic insight into the possible causes of LND and highlight PDE10A as a possible therapeutic target for this intractable neurological disease.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Regulação Enzimológica da Expressão Gênica , Hipoxantina Fosforribosiltransferase/deficiência , Síndrome de Lesch-Nyhan/tratamento farmacológico , Diester Fosfórico Hidrolases/metabolismo , Transdução de Sinais , Linhagem Celular Tumoral , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Hipoxantina Fosforribosiltransferase/genética , Síndrome de Lesch-Nyhan/enzimologia , Síndrome de Lesch-Nyhan/patologia , MicroRNAs/genética , Terapia de Alvo Molecular , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sinapsinas/genética , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA