Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Adicionar filtros








Intervalo de ano
1.
J. appl. oral sci ; 26: e20170004, 2018. tab, graf
Artigo em Inglês | LILACS, BBO | ID: biblio-893706

RESUMO

ABSTRACT Among the many graft materials that have been used for the treatment of bone defects in oral and maxillofacial regions is xenograft. To improve osteoconductive effects of xenografts, they have been combined with various biocompatible materials, such as hyaluronic acid and bone morphogenetic protein. Objective: To determine bone-healing capacity of high molecular weight hyaluronic acid (HA) combined with xenograft in rabbit calvarial bone defects. Material and methods: Ten adult male New Zealand rabbits (mean weight 3 kg) were included in the study. Three 6-mm-diameter bicortical cranial defects were created on calvarial bone of all rabbits. These defects were filled as follows: a) xenograft; b) HA+xenograft; c) autograft. One month after the first operation, rabbits were sacrificed. Specimens were evaluated histomorphometrically. Results: Considering multiple comparisons, differences regarding new bone were statistically significant between all groups (p<0.05). The volume of residual graft was significantly decreased in HA group compared to xenograft group (p=0.035). Marrow space, trabecular thickness (TbTh), trabecular width (TbWi), trabecular separation (TbSp), and number of node: number of terminus (NNd:NTm) in the autograft group were significantly better than xenograft and HA groups (p<0.05). However, regarding marrow space, TbTh, TbWi, TbSp, and NNd:NTm values, xenograft and HA groups showed similar results and the difference were not significant (p>0.05). Conclusion: These results support that high molecular weight hyaluronic acid could contribute to the healing of xenograft by improving the percentage of new bone formation and reducing the percentage of residual graft. However, HA did not significantly affect the quality of newly formed bone assessed by microarchitectural parameters.


Assuntos
Humanos , Animais , Masculino , Crânio/transplante , Cicatrização/efeitos dos fármacos , Regeneração Óssea/efeitos dos fármacos , Xenoenxertos/efeitos dos fármacos , Ácido Hialurônico/farmacologia , Coelhos , Crânio/efeitos dos fármacos , Materiais Biocompatíveis/farmacologia , Reprodutibilidade dos Testes , Transplante Ósseo/métodos , Resultado do Tratamento , Modelos Animais de Doenças , Autoenxertos/efeitos dos fármacos , Osso Esponjoso/efeitos dos fármacos , Ácido Hialurônico/química , Peso Molecular
2.
Braz. j. med. biol. res ; 47(12): 1021-1028, 12/2014. tab, graf
Artigo em Inglês | LILACS | ID: lil-727663

RESUMO

DNA hypomethylation may activate oncogene transcription, thus promoting carcinogenesis and tumor development. S-adenosylmethionine (SAM) is a methyl donor in numerous methylation reactions and acts as an inhibitor of intracellular demethylase activity, which results in hypermethylation of DNA. The main objectives of this study were to determine whether DNA hypomethylation correlated with vascular endothelial growth factor-C (VEGF-C) expression, and the effect of SAM on VEGF-C methylation and gastric cancer growth inhibition. VEGF-C expression was assayed by Western blotting and RT-qPCR in gastric cancer cells, and by immunohistochemistry in tumor xenografts. VEGF-C methylation was assayed by bisulfite DNA sequencing. The effect of SAM on cell apoptosis was assayed by flow cytometry analyses and its effect on cancer growth was assessed in nude mice. The VEGF-C promoters of MGC-803, BGC-823, and SGC-7901 gastric cancer cells, which normally express VEGF-C, were nearly unmethylated. After SAM treatment, the VEGF-C promoters in these cells were highly methylated and VEGF-C expression was downregulated. SAM also significantly inhibited tumor growth in vitro and in vivo. DNA methylation regulates expression of VEGF-C. SAM can effectively induce VEGF-C methylation, reduce the expression of VEGF-C, and inhibit tumor growth. SAM has potential as a drug therapy to silence oncogenes and block the progression of gastric cancer.


Assuntos
Animais , Humanos , Masculino , Antineoplásicos/farmacologia , Metilação de DNA/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , S-Adenosilmetionina/farmacologia , Neoplasias Gástricas/tratamento farmacológico , Fator C de Crescimento do Endotélio Vascular/metabolismo , Apoptose/efeitos dos fármacos , Western Blotting , Linhagem Celular Tumoral , Carcinogênese/efeitos dos fármacos , Metilação de DNA/genética , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/fisiologia , Xenoenxertos/efeitos dos fármacos , Imuno-Histoquímica , Camundongos Nus , Oncogenes/efeitos dos fármacos , Regiões Promotoras Genéticas/efeitos dos fármacos , Reação em Cadeia da Polimerase em Tempo Real , RNA Mensageiro/análise , Neoplasias Gástricas/metabolismo , Fator C de Crescimento do Endotélio Vascular/efeitos dos fármacos , Fator C de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA