Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
J Mol Cell Cardiol ; 138: 34-48, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31733201

RESUMO

Homocysteine (Hcy) is an independent risk factor for atherosclerosis, which is characterized by lipid accumulation in the atherosclerotic plaque. Increasing evidence supports that as the main receptor of high-density lipoprotein, scavenger receptor class B member 1 (SCARB1) is protective against atherosclerosis. However, the underlying mechanism regarding it in Hcy-mediated atherosclerosis remains unclear. Here, we found the remarkable inhibition of SCARB1 expression in atherosclerotic plaque and Hcy-treated foam cells, whereas overexpression of SCARB1 can suppress lipid accumulation in foam cells following Hcy treatment. Analysis of SCARB1 promoter showed that no significant change of methylation level was observed both in vivo and in vitro under Hcy treatment. Moreover, it was found that the negative regulation of DNMT3b on SCARB1 was due to the decreased recruitment of SP1 to SCARB1 promoter. Thus, we concluded that inhibition of SCARB1 expression induced by DNMT3b at least partly accelerated Hcy-mediated atherosclerosis through promoting lipid accumulation in foam cells, which was attributed to the decreased binding of SP1 to SCARB1 promoter. In our point, these findings will provide novel insight into an epigenetic mechanism for atherosclerosis.


Assuntos
Aterosclerose/metabolismo , Aterosclerose/patologia , Antígenos CD36/metabolismo , DNA (Citosina-5-)-Metiltransferases/metabolismo , Homocisteína/efeitos adversos , Transdução de Sinais , Fator de Transcrição Sp1/metabolismo , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/metabolismo , Aterosclerose/complicações , Metilação de DNA/genética , Dieta , Progressão da Doença , Regulação para Baixo/genética , Células Espumosas/metabolismo , Células HEK293 , Humanos , Hiper-Homocisteinemia/complicações , Hiper-Homocisteinemia/patologia , Masculino , Metionina , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Placa Aterosclerótica/complicações , Placa Aterosclerótica/patologia , Regiões Promotoras Genéticas , Ligação Proteica , Células THP-1 , DNA Metiltransferase 3B
2.
Cell Death Dis ; 9(2): 169, 2018 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-29415998

RESUMO

Elevated homocysteine (Hcy) levels have been reported to be involved in liver injury, and autophagy plays an important role in normal hepatic physiology and pathophysiology, but the mechanism underlying Hcy regulated autophagy is currently unknown. In this study, CBS+/- mice were fed with regular diet for 12 weeks to establish a hyperhomocysteinemia (HHcy) model and HL-7702 cells were treated with Hcy, we found that Hcy increases autophagy and aggravates liver injury by downregulation of cystic fibrosis transmembrane conductance regulator (CFTR) expression in vivo and in vitro. Overexpression of CFTR inhibited the formation of autophagosomes and the expression of autophagy-related proteins BECN1, LC3-II/I and Atg12, while the expression of p62 increased in Hcy-treated hepatocytes and CBS+/- mice injected with lentivirus expressing CFTR. Further study showed that CFTR expression is regulated by the interaction of DNA methyltransferase 1 (DNMT1) and enhancer of zeste homolog 2 (EZH2), which, respectively, regulate DNA methylation and histone H3 lysine 27 trimethylation (H3K27me3). In conclusion, our study showed that Hcy activates autophagy by inhibition of CFTR expression via interaction between H3K27me3 and DNA methylation in the mouse liver. These findings provide new insight into the mechanism of Hcy-induced autophagy in liver injury.


Assuntos
Autofagia/efeitos dos fármacos , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Metilação de DNA/genética , Histonas/metabolismo , Homocisteína/farmacologia , Fígado/metabolismo , Lisina/metabolismo , Animais , Biomarcadores/metabolismo , Cistationina beta-Sintase/metabolismo , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Metilação de DNA/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Lentivirus/metabolismo , Fígado/efeitos dos fármacos , Fígado/ultraestrutura , Camundongos , Regiões Promotoras Genéticas/genética
3.
Mol Med Rep ; 16(6): 9181-9188, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29039510

RESUMO

Atherosclerosis (AS) is a progressive disease of multifactorial origin, which occurs in response to endothelial injury. Increased homocysteine (Hcy) is considered a major cause of endothelial dysfunction, oxidative stress and DNA methylation; however, the mechanisms remain to be fully elucidated. The aim of the present study was to investigate whether Hcy causes injury to endothelial cells (ECs) by the effect of lectin­like oxidized­low density lipoprotein receptor­1 (LOX­1) DNA methylation through toll­like receptor 4(TLR4)/nuclear factor (NF)­κB/DNA methyltransferase (DNMT)1. The ECs were treated with different concentrations of Hcy, and it was found that Hcy promoted the expression of TLR4, leading to EC injury. The effect of oxidative stress was analyzed by measuring superoxide dismutase, malondialdehyde and hydrogen peroxide in the ECs. In addition, the association between NF­κB and DNMT1 was examined by treatment of the ECs with pyrrolidine dithiocarbamate (PDTC). The results suggested that Hcy induced LOX­1 DNA hypomethyaltion to promote the expression levels of LOX­1. Taken together, Hcy injured the ECs through the effect of methylation and trans­sulfuration metabolism of LOX­1 through TLR4/NF­κB/DNMT1. Following injury to the ECs, lipids, particularly ox­LDL, accumulated in the sub­endothelial layer to promote the formation of AS.


Assuntos
DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Metilação de DNA , Células Endoteliais/metabolismo , Homocisteína/metabolismo , NF-kappa B/metabolismo , Estresse Oxidativo , Receptores Depuradores Classe E/genética , Receptor 4 Toll-Like/metabolismo , Biomarcadores , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Regulação da Expressão Gênica , Homocisteína/farmacologia , Humanos , Peróxido de Hidrogênio , Lipoproteínas LDL , Superóxido Dismutase/metabolismo , Receptor 4 Toll-Like/genética
4.
DNA Cell Biol ; 36(6): 462-474, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28472596

RESUMO

DNA methyltransferase 1 (DNMT1) and miRNAs are both important regulators of gene expression that have been implicated in the pathogenesis of atherosclerosis. This study was designed to elucidate the potential interaction between DNMT1 and miRNAs in the context of hyperhomocysteinemia (HHcy)-related atherosclerosis. In the aorta of ApoE-/- mice fed a high methionine diet, increased expression of miR-148a/152, with decreased DNMT1 mRNA and protein levels, was detected. Similar changes were observed in cultured foam cells stimulated with homocysteine. When miR-148a/152 was overexpressed using viral vectors, DNMT1 expression was suppressed, whereas the expression of adipose differentiation-related protein (ADRP) was enhanced, and the contents of total cholesterol (TC) and cholesteryl ester (CE) were increased in cultured foam cells. Conversely, downregulation of miR-148a/152 led to elevated DNMT1 expression, reduced ADRP expression, and lowered contents of TC and CE. The luciferase reporter assay verified that DNMT1 is a target gene for miR-148a/152 and overexpression of DNMT1 can partially reverse the miR-148a/152-induced lipid accumulation in foam cells. Meanwhile, we observed that DNMT1 overexpression enhanced DNA methylation and reduced miR-148a/152 expression. Our data showed reciprocal regulation between miR-148a/152 and DNMT1 in foam cells, which likely plays a critical role in HHcy-related atherosclerosis.


Assuntos
Aterosclerose/complicações , Aterosclerose/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Hiper-Homocisteinemia/complicações , MicroRNAs/genética , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/enzimologia , Aterosclerose/patologia , Linhagem Celular Tumoral , Colesterol/metabolismo , Ésteres do Colesterol/metabolismo , DNA (Citosina-5-)-Metiltransferase 1 , Metilação de DNA/efeitos dos fármacos , Células Espumosas/efeitos dos fármacos , Células Espumosas/metabolismo , Humanos , Metionina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas/genética
5.
J Cell Biochem ; 118(12): 4617-4627, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28485501

RESUMO

Hyperhomocysteinemia (HHcy) promotes atherogenesis by modification of histone acetylation patterns and regulation of miRNA expression while the underlying molecular mechanisms are not well known. In this study, we investigated the effects of homocysteine (Hcy) on the expression of histone deacetylase 1 (HDAC1) and tested our hypothesis that Hcy-induced atherosclerosis is mediated by increased HDAC1 expression, which is regulated by miR-34a. The expression of HDAC1 increased and acetylation of histone H3 at lysine 9 (H3K9ac) decreased in the aorta of ApoE-/- mice fed with high methionine diet, whereas miR-34a expression was inhibited. Over-expression of HDAC1 inhibited H3K9ac level and promoted the accumulation of total cholesterol, free cholesterol, and triglycerides in the foam cells. Furthermore, up-regulation of miR-34a reduced HDAC1 expression and inhibited the accumulation of total cholesterol (TC), free cholesterol (FC), and triglycerides (TG) in the foam cells. These data suggest that HDAC1-related H3K9ac plays a key role in Hcy-mediated lipid metabolism disorders, and that miR-34a may be a novel therapeutic target in Hcy-related atherosclerosis. J. Cell. Biochem. 118: 4617-4627, 2017. © 2017 Wiley Periodicals, Inc.


Assuntos
Aterosclerose/metabolismo , Colesterol/metabolismo , Células Espumosas/metabolismo , Histona Desacetilase 1/metabolismo , Homocisteína/metabolismo , MicroRNAs/metabolismo , Triglicerídeos/metabolismo , Acetilação , Animais , Apolipoproteínas E/deficiência , Aterosclerose/genética , Aterosclerose/patologia , Colesterol/genética , Células Espumosas/patologia , Histona Desacetilase 1/genética , Homocisteína/genética , Masculino , Camundongos , Camundongos Knockout , MicroRNAs/genética , Triglicerídeos/genética
6.
J Cell Biochem ; 118(9): 2921-2932, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28230279

RESUMO

Folate deficiency is a known risk factor for liver injury; however, the underlying mechanism remains unclear. In this study, we employed a high homocysteine-induced liver injury model of Apolipoprotein E-deficient (ApoE-/- ) mice fed high-methionine diet and found that high homocysteine induced endoplasmic reticulum (ER) stress and liver cell apoptosis by downregulation of cystic fibrosis transmembrane conductance regulator (CFTR) expression; observations that were attenuated with supplementation of dietary folate. The regulation on CFTR expression was mediated by CFTR promoter methylation and trimethylation of lysine 27 on histone H3 (H3K27me3). Mechanistically, folate inhibited homocysteine-induced CFTR promoter methylation and H3K27me3, which resulted in upregulation of CFTR expression, and reduced ER stress and liver cell apoptosis. Further study showed that folate inhibited the expression of DNA methyltransferase 1 and enhancer of zeste homolog 2, downregulated the cellular concentrations of S-adenosylmethionine (SAM) and S-adenosylhomocysteine (SAH) and upregulated the SAM/SAH ratio, leading to the inhibition of Hcy-induced DNA hypermethylation and H3K27me3 in CFTR promoter. In conclusion, our results provide insight into the protective role of folate in homocysteine-induced ER stress and liver cell apoptosis through the regulation of CFTR expression. J. Cell. Biochem. 118: 2921-2932, 2017. © 2017 Wiley Periodicals, Inc. HIGHLIGHTS: Folate protects hepatocytes of hyperhomocysteinemia mice from apoptosis. Folate alleviates Hcy-induced hepatocyte apoptosis. Folate inhibits Hcy-induced ER stress via upregulation of CFTR expression in hepatocytes. Folate inhibits Hcy-induced methylation of CFTR promotor and H3K27me3.


Assuntos
Apoptose/efeitos dos fármacos , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Metilação de DNA/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Ácido Fólico/farmacologia , Hepatócitos/metabolismo , Hiper-Homocisteinemia/metabolismo , Regiões Promotoras Genéticas , Animais , Apoptose/genética , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Metilação de DNA/genética , Estresse do Retículo Endoplasmático/genética , Hepatócitos/patologia , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/patologia , Camundongos , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA