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.
J Cell Biochem ; 118(4): 754-763, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27618583

RESUMO

Increasing apolipoproteinA-I (apoA-I) production may be anti-atherogenic. Thus, there is a need to identify regulatory factors involved. Transcription of apoA-I involves peroxisome-proliferator-activated-receptor-alpha (PPARα) activation, but endoplasmic reticulum (ER) -stress and inflammation also influence apoA-I production. To unravel why PPARα agonist GW7647 increased apoA-I production compared to PPARα agonist fenofibric acid (FeAc) in human hepatocellular carcinoma (HepG2) and colorectal adenocarcinoma (CaCo-2) cells, gene expression profiles were compared. Microarray analyses suggested CCAAT/enhancer-binding-protein-beta (C/EBP-ß) involvement in the FeAc condition. Therefore, C/EBP-ß silencing and isoform-specific overexpression experiments were performed under ER-stressed, inflammatory and non-inflammatory conditions. mRNA expression of C/EBP-ß, ATF3, NF-IL3 and GDF15 were upregulated by FeAc compared to GW7647 in both cell lines, while DDIT3 and DDIT4 mRNA were only upregulated in HepG2 cells. This ER-stress related signature was associated with decreased apoA-I secretion. After ER-stress induction by thapsigargin or FeAc addition, intracellular apoA-I concentrations decreased, while ER-stress marker expression (CHOP, XBP1s, C/EBP-ß) increased. Cytokine addition increased intracellular C/EBP-ß levels and lowered apoA-I concentrations. Although a C/EBP binding place is present in the apoA-I promoter, C/EBP-ß silencing or isoform-specific overexpression did not affect apoA-I production in inflammatory, non-inflammatory and ER-stressed conditions. Therefore, C/EBP-ß is not a target to influence hepatic apoA-I production. J. Cell. Biochem. 118: 754-763, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Apolipoproteína A-I/biossíntese , Butiratos/farmacologia , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Fenofibrato/análogos & derivados , PPAR alfa/agonistas , Compostos de Fenilureia/farmacologia , Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Proteína beta Intensificadora de Ligação a CCAAT/antagonistas & inibidores , Proteína beta Intensificadora de Ligação a CCAAT/genética , Células CACO-2 , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fenofibrato/farmacologia , Perfilação da Expressão Gênica , Inativação Gênica , Células Hep G2 , Humanos , Inflamação/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Tapsigargina/farmacologia
2.
Eur J Gastroenterol Hepatol ; 21(6): 642-9, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19445040

RESUMO

BACKGROUND: Increasing HDL cholesterol concentrations by stimulating de-novo apolipoprotein A-I (apoA-I) production in the liver and/or in the small intestine is a potential strategy to reduce coronary heart disease risk. Although there is quite some knowledge concerning regulatory effects in the liver, less is known concerning potential agents that could elevate de-novo apoA-I production in the small intestine. METHODS: Therefore, we compared side-by-side effects of various peroxisome proliferator-activated receptor (PPAR)alpha, PPARgamma, retinoid-X-receptor alpha, and farnesoid-X-receptor agonists on de-novo apoA-I production in differentiated CaCo-2 and HepG2 cells. RESULTS: For PPARa agonists, we showed that GW7647 elevated apoA-I concentrations in the medium of both cell models, whereas WY14643 elevated only de-novo apoA-I concentrations in differentiated CaCo-2 cells. Unexpectedly, fenofibric acid lowered apoA-I medium concentrations in both cell lines, which could not be explained by a lack of PPAR transactivation or a lack of retinoid-X-receptor a activation. For farnesoid-X-receptor agonists, chenodeoxycholic acid strongly reduced apoA-I concentrations both in differentiated CaCo-2 and HepG2 cells, whereas GW4064 and taurocholate only lowered apoA-I in CaCo-2 cells (GW4064) or in HepG2 cells (taurocholate). However, overall effects of all individual components on apoA-I production in differentiated CaCo-2 and HepG2 cells were highly correlated (r = 0.68; P = 0.037; N=9). CONCLUSION: We conclude that differentiated CaCo-2 cells are suitable models to study de-novo small intestinal apoA-I production in vitro enabling the possibility to screen for potential bioactive dietary components. This cell model may also determine small-intestinal-specific effects, as some discrepancy was found between both cell models.


Assuntos
Apolipoproteína A-I/biossíntese , Intestino Delgado/metabolismo , Modelos Biológicos , Anticolesterolemiantes/farmacologia , Butiratos/farmacologia , Células CACO-2 , Diferenciação Celular , Avaliação Pré-Clínica de Medicamentos , Células Hep G2 , Humanos , Intestino Delgado/efeitos dos fármacos , Isoxazóis/farmacologia , Proliferadores de Peroxissomos/farmacologia , Compostos de Fenilureia/farmacologia , Pirimidinas/farmacologia , Receptores Citoplasmáticos e Nucleares/agonistas
3.
J Lipid Res ; 49(4): 790-6, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18162663

RESUMO

Policosanol is a mixture of long-chain primary aliphatic saturated alcohols. Previous studies in humans and animals have shown that these compounds improved lipoprotein profiles. However, more-recent placebo-controlled studies could not confirm these promising effects. Octacosanol (C28), the main component of sugarcane-derived policosanol, is assumed to be the bioactive component. This has, however, never been tested in an in vivo study that compared individual policosanol components side by side. Here we present that neither the individual policosanol components (C24, C26, C28, or C30) nor the natural policosanol mixture (all 30 mg/100 g diet) lowered serum cholesterol concentrations in LDL receptor knock-out (LDLr(+/-)) mice. Moreover, there was no effect on gene expression profiles of LDLr, ABCA1, HMG-CoA synthase 1, and apolipoprotein A-I (apoA-I) in hepatic and small intestinal tissue of female LDLr(+/-) mice after the 7 week intervention period. Finally, none of the individual policosanols or their respective long-chain fatty acids or aldehydes affected de novo apoA-I protein production in vitro in HepG2 and CaCo-2 cells. Therefore, we conclude that the evaluated individual policosanols, as well as the natural policosanol mixture, have no potential for reducing coronary heart disease risk through effects on serum lipoprotein concentrations.


Assuntos
Colesterol/metabolismo , Álcoois Graxos/química , Álcoois Graxos/farmacologia , Receptores de LDL/deficiência , Receptores de LDL/metabolismo , Animais , Apolipoproteína A-I/biossíntese , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Emulsões/química , Emulsões/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Heterozigoto , Humanos , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de LDL/genética , Soluções
4.
Nutr Metab Cardiovasc Dis ; 17(8): 616-28, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17703927

RESUMO

Dyslipidemia leading to coronary heart diseases (CHD) enables venues to prevent or treat CHD by other strategies than only lowering serum LDL cholesterol (LDL-C) concentrations, which is currently the most frequently targeted change. Unlike LDL-C, elevated high-density lipoprotein cholesterol (HDL-C) concentrations may protect against the development of CHD as demonstrated in numerous large-scale epidemiological studies. In this review we describe that besides elevating serum HDL-C concentrations by increasing alpha-HDL particles, approaches to elevate HDL-C concentrations by increasing pre-beta HDL particle concentrations seems more attractive. Besides infusion of apoA-I(Milano), using apoA-I mimetics, or delipidation of alpha-HDL particles, elevating de novo apoA-I production may be a suitable target to functionally increase pre-beta HDL particle concentrations. Therefore, a detailed description of the molecular pathways underlying apoA-I synthesis and secretion, completed with an overview of known effects of pharmacological and nutritional compounds on apoA-I synthesis will be presented. This knowledge may ultimately be applied in developing dietary intervention strategies to elevate apoA-I production and serum HDL-C concentrations and consequently lower CHD risk.


Assuntos
Anticolesterolemiantes/uso terapêutico , Apolipoproteína A-I/biossíntese , Doença das Coronárias/prevenção & controle , Hipercolesterolemia/tratamento farmacológico , Hipolipemiantes/uso terapêutico , Apolipoproteína A-I/sangue , HDL-Colesterol/sangue , LDL-Colesterol/sangue , Doença das Coronárias/sangue , Humanos , Hipercolesterolemia/complicações , Fatores de Risco , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...