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








Base de dados
Intervalo de ano de publicação
1.
Cell Commun Signal ; 15(1): 50, 2017 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-29187201

RESUMO

BACKGROUND: The simplicity of Transforming Growth Factor ß (TGFß) signaling pathway, linear and non-amplified, hardly sustains its variety of responses. This is often justified by the complex regulation showed by Smad proteins, TGFß signaling intracellular transducers, object of post-translational modifications that modulate TGFß-dependent transcription. Protein acetylation is emerging as a compelling mechanism affecting the activities of significant transcription factors, including p53, FOXO or NF-kB. Smad proteins might be controlled by this mechanism, implying that accessory factors capable of altering Smads-transcriptional complexes acetylation status and hence regulate TGFß responses remain to be identified. Understanding this interaction may help in the assessment of TGFß signaling outcomes, extending from healthy physiology to pathological conditions and cancer. METHODS: A two-hybrid chimera interacting system allowed to identify Sirt1, a NAD+ dependent type III histone deacetylase, as a novel Smad2 interactor. Several well stablished cellular models were applied to characterize this interaction by means of co-immunoprecipitation of tagged proteins and immuno-fluorescence staining. The occurrence of the interaction at Smad2 driven transcriptomic complexes was studied by means of DNA-pull-down and chromatin immunoprecipitation (ChIP), while its effects were assessed by protein over-expression and siRNA applied into a TGFß-dependent reporter gene assay. RESULTS: The interaction was confirmed and observed to be enhanced upon Smad2 acetylation, a known feature of active and nuclear Smad2. However, Sirt1 did not play a major role in Smad2 deacetylation. Anti-Sirt1 ChIP showed increased recovery of promoter regions corresponding to Smad2-driven genes after TGFß-stimulation, while its occurrence at Smad2-dependent transcriptomic complexes on DNA was found to effectively modulate gene expression. CONCLUSIONS: Sirt1 presence on Smad2-driven TGFß-dependent regulatory elements was detected and found to increase after TGFß treatment. Moreover, Sirt1 overexpression resulted in a decrease of the activity of a Smad2-driven TGFß-dependent reporter gene, while Sirt1 interference increased its activity. This would confirm the relevance of the discovered Sirt1-Smad2 interaction for the regulation of TGFß-dependent gene transcription.


Assuntos
Sirtuína 1/metabolismo , Proteína Smad2/metabolismo , Transcrição Gênica , Fator de Crescimento Transformador beta/metabolismo , Acetilação , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Ligação Proteica , Domínios Proteicos , Transporte Proteico , Transdução de Sinais , Sirtuína 1/química , Proteína Smad2/química
3.
PLoS One ; 10(8): e0135324, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26284363

RESUMO

BACKGROUND: Post-traumatic large-surface or deep wounds often cannot progress to reepithelialisation because they become irresponsive in the inflammatory stage, so intervention is necessary to provide the final sealing epidermis. Previously we have shown that Amniotic Membrane (AM) induced a robust epithelialisation in deep traumatic wounds. METHODS AND FINDINGS: To better understand this phenomenon, we used keratinocytes to investigate the effect of AM on chronic wounds. Using keratinocytes, we saw that AM treatment is able to exert an attenuating effect upon Smad2 and Smad3 TGFß-induced phosphorylation while triggering the activation of several MAPK signalling pathways, including ERK and JNK1, 2. This also has a consequence for TGFß-induced regulation on cell cycle control key players CDK1A (p21) and CDK2B (p15). The study of a wider set of TGFß regulated genes showed that the effect of AM was not wide but very concrete for some genes. TGFß exerted a powerful cell cycle arrest; the presence of AM however prevented TGFß-induced cell cycle arrest. Moreover, AM induced a powerful cell migration response that correlates well with the expression of c-Jun protein at the border of the healing assay. Consistently, the treatment with AM of human chronic wounds induced a robust expression of c-Jun at the wound border. CONCLUSIONS: The effect of AM on the modulation of TGFß responses in keratinocytes that favours proliferation together with AM-induced keratinocyte migration is the perfect match that allows chronic wounds to move on from their non-healing state and progress into epithelialization. Our results may explain why the application of AM on chronic wounds is able to promote epithelialisation.


Assuntos
Âmnio/citologia , Proliferação de Células/efeitos dos fármacos , Queratinócitos/citologia , Fator de Crescimento Transformador beta/farmacologia , Cicatrização/fisiologia , Ferimentos Penetrantes/terapia , Âmnio/metabolismo , Animais , Ciclo Celular/efeitos dos fármacos , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Pulmão/citologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Sistema de Sinalização das MAP Quinases , Vison , Fosforilação , Proteínas Proto-Oncogênicas c-jun/metabolismo , Reepitelização , Proteína Smad2/metabolismo , Cicatrização/efeitos dos fármacos , Ferimentos Penetrantes/patologia
4.
PLoS One ; 7(7): e42271, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22860103

RESUMO

Wound healing is a biological process directed to the restoration of tissue that has suffered an injury. An important phase of wound healing is the generation of a basal epithelium able to wholly replace the epidermis of the wound. A broad range of products derived from fibroin and sericin from Bombyx mori silk are used to stimulate wound healing. However, so far the molecular mechanism underlying this phenomenon has not been elucidated. The aim of this work was to determine the molecular basis underlying wound healing properties of silk proteins using a cell model. For this purpose, we assayed fibroin and sericin in a wound healing scratch assay using MDA-MB-231 and Mv1Lu cells. Both proteins stimulated cell migration. Furthermore, treatment with sericin and fibroin involved key factors of the wound healing process such as upregulation of c-Jun and c-Jun protein phosphorylation. Moreover, fibroin and sericin stimulated the phosphorylation of ERK 1/2 and JNK 1/2 kinases. All these experiments were done in the presence of specific inhibitors for some of the cell signalling pathways referred above. The obtained results revealed that MEK, JNK and PI3K pathways are involved in fibroin and sericin stimulated cells migration. Inhibition of these three kinases prevented c-Jun upregulation and phosphorylation by fibroin or sericin. Fibroin and sericin were tested in the human keratinocyte cell line, HaCaT, with similar results. Altogether, our results showed that fibroin and sericin initiate cell migration by activating the MEK, JNK and PI3K signalling pathways ending in c-Jun activation.


Assuntos
Movimento Celular/fisiologia , Fibroínas/fisiologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Sericinas/fisiologia , Seda/química , Regulação para Cima/fisiologia , Animais , Bombyx , Linhagem Celular , Humanos , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA