Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Toxicol Rep ; 2: 1213-1221, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-28962464

RESUMO

Doxorubicin (Dox) has been used for more than four decades to treat cancer, particularly solid tumours and haematological malignancies. However, the administration of this drug is a matter of concern in the clinical community, since Dox therapy is commonly associated with dose-dependent cardiotoxicity. Attempts at alleviating drug generated cardiac damage using naturally occurring compounds with radical scavenging property are a promising area of research. p-Coumaric acid (pCA) is one such compound which has significant antiradical scavenging effect. This study aims to investigate the effect of pre and co-administration of pCA on mitigating or preventing Dox induced cardiotoxicity in vitro using H9c2 cardiomyoblast cell lines. Addition of pCA and Dox were performed for both treatment and control sets on H9c2 cells. Sulphorhodamine B assay was used to study the cytotoxic effect of pCA and Dox. The effect of the drug on cell morphology, cell viability and nuclear damage was studied using AO/EB and DAPI staining. ROS production was studied using DCFH-DA staining. Mitochondrial membrane potential and intracellular calcium levels were assessed by rhodamine 123 and Fura 2AM staining. pCA showed strong ABTS cation radical scavenging activity and FRAP activity in a dose dependent manner. The results showed that Dox has significant cytotoxic effect in a dose dependent manner while pCA, even at higher concentrations did not display any significant cytotoxicity on H9c2 cells. Both pre treatment and co- administration of pCA reduced the drug induced toxic effects on cell morphology and enhanced the number of viable cells in comparison to the Dox treated cells as evident from the AO/EB and DAPI staining images. The Dox induced ROS production was found to be significantly reduced in pCA pre-treated and co-administered cells. Dox induced changes in mitochondrial membrane potential and intracellular calcium levels were remarkably improved following pre and co-treatment of H9c2 cells with pCA. These results clearly suggest that pre-treatment and co-administration of pCA is a promising therapeutic intervention in managing Dox mediated cardiotoxicity.

2.
J Sci Food Agric ; 92(9): 1903-8, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22231513

RESUMO

BACKGROUND: Coconut kernel protein (CKP) has been reported to contain significant amounts of L-arginine. Its potential effect on glucose homeostasis, possibly through the nitric oxide synthase (NO) pathway, was therefore investigated in alloxan-induced diabetic rats. Diabetes was induced by a single intraperitoneal dose of alloxan (150 mg kg⁻¹ body weight). Experimental rats were grouped as follows: Group I, normal control; Group II, diabetic control; Group III, diabetic + CKP; Group IV, diabetic + L-arginine; Group V, diabetic + L-arginine + L-N(G)-Nitroarginine methyl ester (L-NAME). Purified CKP isolated from dried coconut kernel and L-arginine was administered to rats along with a semi-synthetic diet for 45 days. L-NAME (0.5 mg kg⁻¹ body weight) was given to Group V animals. After the experimental period, serum glucose, insulin, activities of liver nitric oxide synthase and arginase, liver glycogen levels and histopathology of the pancreas were evaluated. RESULTS: Serum glucose, insulin and antioxidant enzyme activities and liver glycogen levels were found to be restored to basal levels in CKP-fed rats. Decreased arginase and increased nitric oxide synthase (NOS) activities were found in CKP- and arginine-fed rats. L-NAME treatment showed a partial effect on these parameters. Histopathology revealed that CKP and L-arginine feeding reduced the diabetes-related pancreatic damage in treated rats compared to the diabetic control. CONCLUSION: The results observed in this study indicate that the potential antidiabetic activity of CKP may be through an arginine-NO pathway leading to pancreatic beta cell regeneration.


Assuntos
Arginina/uso terapêutico , Cocos/química , Diabetes Mellitus Experimental/tratamento farmacológico , Dieta , Proteínas Alimentares/uso terapêutico , Óxido Nítrico Sintase/metabolismo , Fitoterapia , Animais , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Arginase/metabolismo , Arginina/farmacologia , Glicemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Proteínas Alimentares/farmacologia , Glicogênio/metabolismo , Insulina/sangue , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , NG-Nitroarginina Metil Éster/farmacologia , Pâncreas/efeitos dos fármacos , Pâncreas/patologia , Preparações de Plantas/farmacologia , Preparações de Plantas/uso terapêutico , Proteínas de Plantas/farmacologia , Proteínas de Plantas/uso terapêutico , Ratos , Ratos Sprague-Dawley , Sementes/química , Transdução de Sinais
3.
Cancer Lett ; 297(2): 231-43, 2010 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-20831981

RESUMO

A novel pyranoside mimetic compound, DMBO (2-(2,6-difluorophenyl)-5-(4-methoxyphenyl)-1-oxa-3-azaspiro[5.5]undecane), was designed and synthesized. The sugar mimicking behavior of DMBO was addressed by its ability to bind several growth factors/cytokines such as vascular endothelial growth factor (VEGF), heparin-binding epidermal growth factor-like growth factor (HB-EGF), and tumor necrosis factor (TNF)-α as demonstrated by the recently developed surface plasmon resonance assay. DMBO exhibited strong anti-proliferation activity in vitro against tumor cells including a highly metastatic murine osteosarcoma cell line LM8G7 that secretes VEGF as well as two human ovarian cell lines, OVSAHO and SKOV-3, which secrete TNF-α and HB-EGF respectively. Furthermore, DMBO inhibited the metastatic activity to the mouse liver of LM8G7 cells injected from a lateral tail vein, and affected the heparan-degrading activity of LM8G7 cells. Here, we report that DMBO acts as a human heparanase inhibitor in vitro possibly as a substrate mimetic. DMBO also inhibited the migration and invasion of LM8G7 cells and angiogenic events such as endothelial cell proliferation, migration and capillary tube-like formation in vitro. More prominently, the administration of DMBO with heparin resulted in synergistic anti-tumor effects in mouse modelofosteosarcoma. These preclinical data shows the potential anti-cancer effects of DMBO.


Assuntos
Benzoxazinas/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fatores de Crescimento do Endotélio Vascular/metabolismo , Inibidores da Angiogênese/farmacologia , Animais , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Feminino , Glucuronidase/antagonistas & inibidores , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Humanos , Neoplasias Hepáticas Experimentais/tratamento farmacológico , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas Experimentais/secundário , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Osteossarcoma/tratamento farmacológico , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Ligação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...