Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Food Chem Toxicol ; 163: 112938, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35314295

RESUMEN

A diet deficient in donors of methyl group, such as methionine, affects DNA methylation and hepatic lipid metabolism. Methionine also affects other epigenetic mechanisms, such as microRNAs. We investigated the effects of methionine-supplemented or methionine-deficient diets on the expression of chromatin-modifying genes, global DNA methylation, the expression and methylation of genes related to lipid metabolism, and the expression of microRNAs in mouse liver. Female Swiss albino mice were fed a control diet (0.3% methionine), a methionine-supplemented diet (2% methionine), and a methionine-deficient diet (0% methionine) for 10 weeks. The genes most affected by the methionine-supplemented diet were associated with histone and DNA methyltransferases activity, while the methionine-deficient diet mostly altered the expression of histone methyltransferases genes. Both diets altered the global DNA methylation and the expression and gene-specific methylation of the lipid metabolism gene Apoa5. Both diets altered the expression of several liver homeostasis-related microRNAs, including miR-190b-5p, miR-130b-3p, miR-376c-3p, miR-411-5p, miR-29c-3p, miR-295-3p, and miR-467d-5p, with the methionine-deficient diet causing a more substantial effect. The effects of improper amounts of methionine in the diet on liver pathologies may involve a cooperative action of chromatin-modifying genes, which results in an aberrant pattern of global and gene-specific methylation, and microRNAs responsible for liver homeostasis.


Asunto(s)
Metionina , MicroARNs , Animales , Cromatina/metabolismo , Metilación de ADN , Dieta , Epigénesis Genética , Femenino , Hígado , Ratones , MicroARNs/genética , MicroARNs/metabolismo
3.
Carcinogenesis ; 42(8): 1026-1036, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-33999989

RESUMEN

Hepatocellular carcinoma (HCC) presents with a high treatment resistance and poor prognosis. Early diagnosis and preventive approaches such as chemoprevention are essential for the HCC control. Therefore, we evaluated the chemopreventive effects of butyrate-containing structured lipids (STLs) administered during the promotion stage of hepatocarcinogenesis in rats submitted to the 'resistant hepatocyte' (RH) model. Administration of butyrate-containing STLs inhibited the incidence and mean number of visible hepatic nodules per rat and reduced the number and area of glutathione S-transferase placental form-positive (GST-P+) preneoplastic focal lesions in the livers. This was accompanied by the induction of apoptosis and an increased level of hepatic butyric acid. Treatment with butyrate-containing STLs resulted in increased histone H3 lysine 9 (H3K9) acetylation, reduction of total histone deacetylase (HDAC) activity, and lower levels of HDAC4 and HDAC6 proteins. The chemopreventive effect of butyrate-containing STLs was also associated with the increased nuclear compartmentalization of p53 protein and reduced expression of the Bcl-2 protein. In addition, rats treated with butyrate-containing STLs showed decreased DNA damage and telomerase activity in the livers. These results demonstrate that the suppressive activity of butyrate-containing STLs is associated with inhibition of elevated during hepatocarcinogenesis chromatin-modifying proteins HDAC4 and HDAC6, subcellular redistribution of the p53 protein, and decreased DNA damage and telomerase activity.


Asunto(s)
Butiratos/metabolismo , Daño del ADN , Gutatión-S-Transferasa pi/metabolismo , Histona Desacetilasa 6/metabolismo , Histona Desacetilasas/metabolismo , Lípidos/química , Neoplasias Hepáticas Experimentales/patología , Telomerasa/metabolismo , Animales , Carcinogénesis , Caspasa 3/metabolismo , Neoplasias Hepáticas Experimentales/enzimología , Neoplasias Hepáticas Experimentales/genética , Masculino , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Wistar , Fracciones Subcelulares/enzimología , Proteína p53 Supresora de Tumor/metabolismo , Ácido alfa-Linolénico/metabolismo
4.
Nutr Cancer ; 68(5): 719-33, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27266713

RESUMEN

Hepatocellular carcinoma (HCC) is an aggressive and life-threatening disease often diagnosed at intermediate or advanced stages, which substantially limits therapeutic approaches to its successful treatment. This indicates that the prevention of HCC may be the most promising strategy in reducing its incidence and mortality. Emerging evidence indicates that numerous nutrients and nonnutrient dietary bioactive components can reduce the occurrence and/or delay the development of HCC through modifications of deregulated epigenetic mechanisms. This review examines the existing knowledge on the epigenetic mechanism-based studies in in vitro and in vivo models of HCC on the chemopreventive potential of epigenetic food components, including dietary methyl-group donors, epigallocatechin-3-gallate, sodium butyrate, resveratrol, curcumin, and sulforaphane, on liver carcinogenesis. Future direction and potential challenges in the effective use of bioactive food constituents in the prevention of HCC are highlighted and discussed.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/prevención & control , Epigénesis Genética , Alimentos , Animales , Ácido Butírico/farmacología , Catequina/análogos & derivados , Catequina/farmacología , Línea Celular Tumoral , Curcumina/farmacología , Metilación de ADN/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Isotiocianatos/farmacología , Fitoquímicos/farmacología , Resveratrol , Estilbenos/farmacología , Sulfóxidos
5.
Oncotarget ; 7(17): 24339-47, 2016 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-27013579

RESUMEN

Hepatocellular carcinoma (HCC), an aggressive and the fastest growing life-threatening cancer worldwide, is often diagnosed at intermediate or advanced stages of the disease, which substantially limits therapeutic approaches for its successful treatment. This indicates that the prevention of hepatocarcinogenesis is probably the most promising approach to reduce both the HCC incidence and cancer-related mortality. In previous studies, we demonstrated a potent chemopreventive effect of tributyrin, a butyric acid prodrug, on experimental hepatocarcinogenesis. The cancer-inhibitory effect of tributyrin was linked to the suppression of sustained cell proliferation and induction of apoptotic cell death driven by an activation of the p53 apoptotic signaling pathway. The goal of the present study was to investigate the underlying molecular mechanisms linked to tributyrin-mediated p53 activation. Using in vivo and in vitro models of liver cancer, we demonstrate that an increase in the level of p53 protein in nuclei, a decrease in the level of cytoplasmic p53, and, consequently, an increase in the ratio of nuclear/cytoplasmic p53 in rat preneoplastic livers and in rat and human HCC cell lines caused by tributyrin or sodium butyrate treatments was associated with a marked increase in the level of nuclear chromosome region maintenance 1 (CRM1) protein. Mechanistically, the increase in the level of nuclear p53 protein was associated with a substantially reduced binding interaction between CRM1 and p53. The results demonstrate that the cancer-inhibitory activity of sodium butyrate and its derivatives on liver carcinogenesis may be attributed to retention of p53 and CRM1 proteins in the nucleus, an event that may trigger activation of p53-mediated apoptotic cell death in neoplastic cells.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Compartimento Celular/efectos de los fármacos , Carioferinas/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Receptores Citoplasmáticos y Nucleares/metabolismo , Triglicéridos/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Ácido Butírico/farmacología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Citoplasma/metabolismo , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Carioferinas/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Unión Proteica/efectos de los fármacos , Ratas Wistar , Receptores Citoplasmáticos y Nucleares/genética , Proteína p53 Supresora de Tumor/genética , Proteína Exportina 1
6.
Mol Nutr Food Res ; 60(2): 420-9, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26548572

RESUMEN

SCOPE: Emerging evidence indicates that the use of bioactive food components is a promising strategy to prevent the development of liver cancer. The goal of this study was to examine the chemopreventive effect of butyrate-containing structured lipids (STLs) produced by an enzymatic interesterification of tributyrin and flaxseed oil on rat hepatocarcinogenesis. METHODS AND RESULTS: Male Wistar rats were subjected to a classic "resistant hepatocyte" model of liver carcinogenesis and treated with STLs, tributyrin or flaxseed oil during the initial phases of hepatocarcinogenesis. Treatment with STLs and tributyrin strongly inhibited the development of preneoplastic liver lesions. The chemopreventive activity of tributyrin was associated with the induction of apoptosis and reduction of the expression of major activated hepatocarcinogenesis-related oncogenes. Treatment with STLs caused substantially greater inhibitory effects than tributyrin on oncogene expression. CONCLUSION: These results demonstrate that the tumor-suppressing activity of butyrate-containing STLs is associated with its ability to prevent and inhibit activation of major hepatocarcinogenesis-related oncogenes. Enrichment of histone H3K9me3 and H3K27me3 at the promoter of Myc and Ccnd1 genes may be related to the inhibitory effect on oncogene expression in the livers of STL-treated rats.


Asunto(s)
Anticarcinógenos/farmacología , Ácido Butírico/farmacología , Neoplasias Hepáticas Experimentales/prevención & control , Animales , Anticarcinógenos/química , Ácido Butírico/química , Regulación de la Expresión Génica/efectos de los fármacos , Histonas/genética , Histonas/metabolismo , Aceite de Linaza/química , Lípidos/química , Lípidos/farmacología , Neoplasias Hepáticas Experimentales/patología , Masculino , Oncogenes , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Lesiones Precancerosas/prevención & control , Ratas Wistar , Triglicéridos/química , Triglicéridos/farmacología
7.
Mol Nutr Food Res ; 58(7): 1502-12, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24827819

RESUMEN

SCOPE: A compromised nutritional status in methyl-group donors may provoke several molecular alterations triggering the development of nonalcoholic fatty liver disease (NAFLD) in humans and experimental animals. In this study, we investigated a role and the underlying molecular mechanisms of methionine metabolic pathway malfunctions in the pathogenesis of NAFLD. METHODS AND RESULTS: We fed female Swiss albino mice a control (methionine-adequate) diet and two experimental (methionine-deficient or methionine-supplemented) diets for 10 weeks, and the levels of one-carbon metabolites, expression of one-carbon and lipid metabolism genes in the livers were evaluated. We demonstrate that both experimental diets increased hepatic levels of S-adenosyl-l-homocysteine and homocysteine, altered expression of one-carbon and lipid metabolism genes, and caused lipid accumulation, especially in mice fed the methionine-deficient diet. Markers of oxidative and ER stress response were also elevated in the livers of mice fed either diet. CONCLUSION: Our findings indicate that both dietary methionine deficiency and methionine supplementation can induce molecular abnormalities in the liver associated with the development of NAFLD, including deregulation in lipid and one-carbon metabolic pathways, and induction of oxidative and ER stress. These pathophysiological events may ultimately lead to lipid accumulation in the livers, triggering the development of NAFLD.


Asunto(s)
Suplementos Dietéticos , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Metionina/administración & dosificación , Metionina/deficiencia , Animales , Femenino , Glutatión/sangre , Homocisteína/sangre , Hígado/metabolismo , Ratones , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Triglicéridos/sangre
8.
Int J Cancer ; 135(1): 7-18, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24302446

RESUMEN

The steady increase in the incidence and mortality of hepatocellular carcinoma (HCC) signifies a crucial need to understand better its pathogenesis to improve clinical management and prevention of the disease. The aim of this study was to investigate molecular mechanisms for the chemopreventive effects of folic acid and tributyrin alone or in combination on rat hepatocarcinogenesis. Male Wistar rats were subjected to a classic "resistant hepatocyte" model of liver carcinogenesis and treated with folic acid and tributyrin alone or in combination for 5 weeks during promotion stage. Treatment with folic acid and tributyrin alone or in combination strongly inhibited the development of glutathione-S-transferase placental form (GSTP)-positive foci. Microarray analysis showed significant changes in gene expression. A total of 498, 655 and 940 of differentially expressed genes, involved in cell cycle, p53-signaling, angiogenesis and Wnt pathways, was identified in the livers of rats treated with folic acid, tributyrin or folic acid and tributyrin. A detailed analysis of these differentially expressed genes revealed that treatments inhibited angiogenesis in the preneoplastic livers. This was evidenced by the fact that 30 out of 77 differentially expressed genes common to all three treatments are involved in the regulation of the angiogenesis pathway. The inhibition of angiogenesis was confirmed by reduced levels of CD34 protein. In conclusion, the tumor-suppressing activity of folic acid and tributyrin is associated with inhibition of angiogenesis at early stages of rat liver carcinogenesis. Importantly, the combination of folic acid and tributyrin has stronger chemopreventive effect than each of the compounds alone.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Ácido Fólico/administración & dosificación , Neoplasias Hepáticas/tratamiento farmacológico , Triglicéridos/administración & dosificación , Animales , Antígenos CD34/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Carcinogénesis/inducido químicamente , Carcinogénesis/efectos de los fármacos , Carcinogénesis/patología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Dietilnitrosamina/toxicidad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Gutatión-S-Transferasa pi/metabolismo , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Masculino , Proteínas de Neoplasias/biosíntesis , Neovascularización Patológica/tratamiento farmacológico , Ratas , Transcriptoma/genética
9.
Carcinogenesis ; 34(8): 1900-6, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23568954

RESUMEN

The reversibility of non-genotoxic phenotypic alterations has been explored in order to develop novel preventive and therapeutic approaches for cancer control. Previously, it has been demonstrated that histone deacetylase (HDAC) inhibitor tributyrin, a butyric acid prodrug, to have chemopreventive effects on rat hepatocarcinogenesis. The goal of this study was to determine molecular mechanisms associated with the chemopreventive activity of tributyrin. Male Wistar rats were allocated randomly to untreated control group and two experimental groups. Rats in the experimental group 1 were treated with maltodextrin (3g/kg body wt), and rats in experimental group 2 were treated with tributyrin (2g/kg body wt) daily for 8 weeks. Two weeks after treatment initiation, rats from experimental groups were subjected to a 'resistant hepatocyte' model of hepatocarcinogenesis. Treatment with tributyrin resulted in lower HDAC activity and Hdac3 and Hdac4 gene expression, and an increase of histone H3 lysine 9 and 18 and histone H4 lysine 16 acetylation as compared with the experimental group 1. In addition to the increase in histone acetylation, tributyrin caused an increase in the acetylation of the nuclear p53 protein. These changes were accompanied by a normalization of the p53-signaling network, particularly by the upregulation of pro-apoptotic genes, and a consequent increase of apoptosis and autophagy in the livers of tributyrin-treated rats. These results indicate that the chemopreventive activity of tributyrin may be related to an increase of histone and p53 acetylation, which could lead to the induction of the p53 apoptotic pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Ácido Butírico/farmacología , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/prevención & control , Profármacos/farmacología , Triglicéridos/farmacología , Proteína p53 Supresora de Tumor/genética , Acetilación , Animales , Apoptosis/genética , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Histonas/genética , Histonas/metabolismo , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/patología , Lisina/genética , Lisina/metabolismo , Masculino , Lesiones Precancerosas/tratamiento farmacológico , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Distribución Aleatoria , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba/efectos de los fármacos
10.
Nutr Res ; 27(1): 1365-1317, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18167510

RESUMEN

DNA methylation is an epigenetic feature that is associated with X chromosome inactivation, genomic imprinting, transcriptional silencing of genes and genomic stability. Folate provides a labile source of methyl groups which may be used for cellular methylation reactions including DNA methylation. The methylenetetrahydrofolate reductase (MTHFR) 677C-->T variant is an important determinant of folate nutriture and may influence DNA methylation. This study sought to assess the influence of the MTHFR C677T genotype on global leukocyte DNA methylation in young (18-45y) Mexican American women (n=43; 14 CC, 12 CT and 17 TT). Subjects consumed a folate restricted diet (135 mug DFE/d) for 7 wk followed by folate treatment with 400 or 800 mug DFE/d for 7 wk. Global leukocyte DNA methylation was assessed via the cytosine extension assay at week 0, week 7 (after folate restriction) and week 14 (after folate treatment). No main effects of MTHFR C677T genotype or folate intake were detected at any time point during the study. However, at the end of folate treatment (wk 14), DNA methylation was lower (P<0.05) in women with the MTHFR 677TT genotype relative to the CT or CC genotype. Because it is unlikely that folate treatment would result in methyl group loss, we suggest that there was a delay in DNA methylation response to folate intake. Overall, these data suggest that the MTHFR 677TT genotype and folate interact to lower global leukocyte DNA methylation patterns in young Mexican American women.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA