Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Tipo de estudo
Intervalo de ano de publicação
1.
Cell Biochem Biophys ; 81(1): 151-160, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36251137

RESUMO

ATP-binding cassette transporter A-I (ABCA1) is an ubiquitously expressed protein whose main function is the transmembrane transport of cholesterol and phospholipids. Synthesis of ABCA1 protein in liver is necessary for high-density lipoprotein (HDL) formation in mammals. Thus, the mechanism of ABCA1 gene expression regulation in hepatocytes are of critical importance. Recently, we have found the insulin-dependent downregulation of other key player in the HDL formation-apolipoprotein A-I gene (J. Cell. Biochem., 2017, 118:382-396). Nothing is known about the role of insulin in the regulation of ABCA1 gene. Here we show for the first time that insulin decreases the mRNA and protein levels of ABCA1 in human hepatoma cell line HepG2. PI3K, p38, MEK1/2, JNK and mTORC1 signaling pathways are involved in the insulin-mediated downregulation of human ABCA1 gene. Transcription factors LXRα, LXRß, FOXO1 and NF-κB are important contributors to this process, while FOXA2 does not regulate ABCA1 gene expression. Insulin causes the decrease in FOXO1, LXRα and LXRß binding to ABCA1 promoter, which is likely the cause of the decrease in the gene expression. Interestingly, the murine ABCA1 gene seems to be not regulated by insulin in hepatocytes (in vitro and in vivo). We suggest that the reason for this discrepancy is the difference in the 5'-regulatory regions of human and murine ABCA1 genes.


Assuntos
Transportador 1 de Cassete de Ligação de ATP , Insulina , Receptores X do Fígado , Receptores Nucleares Órfãos , Animais , Humanos , Camundongos , Transportador 1 de Cassete de Ligação de ATP/genética , Carcinoma Hepatocelular , Linhagem Celular , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Insulina/metabolismo , Neoplasias Hepáticas , Receptores X do Fígado/genética , Receptores X do Fígado/metabolismo , Mamíferos/metabolismo , Receptores Nucleares Órfãos/genética , Receptores Nucleares Órfãos/metabolismo
3.
Biochemistry (Mosc) ; 87(11): 1252-1259, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36509728

RESUMO

Adiponectin is an adipose tissue hormone, participating in energy metabolism and involved in atherogenesis. Previously, it was found that adiponectin increases expression of the APOA1 (apolipoprotein A-1) gene in hepatocytes, but the mechanisms of this effect remained unexplored. Our aim was to investigate the role of adiponectin receptors AdipoR1/R2, AMP-activated protein kinase (AMPK), nuclear peroxisome proliferator-activated receptor alpha (PPARα) and liver X receptors (LXRs) in mediating the action of adiponectin on hepatic APOA1 expression in human hepatoma HepG2 cells. The level of APOA1 expression was determined by RT-qPCR and ELISA. We showed that the siRNA-mediated knockdown of genes coding for AdipoR1, AdipoR2, AMPK, PPARα, and LXRα and ß prevented adiponectin-induced APOA1 expression in HepG2 cells and demonstrated that interaction of PPARα and LXRs with the APOA1 gene hepatic enhancer is important for the adiponectin-dependent APOA1 transcription. The results of this study point out to the involvement of both types of adiponectin receptors, AMPK, PPARα, and LXRs in the adiponectin-dependent upregulation of the APOA1 expression.


Assuntos
Adiponectina , PPAR alfa , Humanos , PPAR alfa/genética , PPAR alfa/metabolismo , Adiponectina/genética , Adiponectina/metabolismo , Receptores X do Fígado/genética , Receptores de Adiponectina/genética , Receptores de Adiponectina/metabolismo , Células Hep G2 , Apolipoproteína A-I/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Expressão Gênica
4.
Eur J Cell Biol ; 97(3): 204-215, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29550264

RESUMO

C3 is an acute phase protein, and thus its plasma concentration increases quickly and drastically during the onset of inflammation. Insulin plays a complex role in inflammation. Elevated level of plasma C3 was shown to correlate with heightened fasting insulin levels and insulin resistance and appears to be a risk factor for the cardiovascular disease and atherosclerosis. The main source of plasma C3 is liver. Nothing is known about effects of insulin on C3 gene expression and protein secretion by hepatocytes. In light of these data we asked if insulin is capable of regulating C3 production in hepatocytes. Here we show that insulin downregulates C3 gene expression in human hepatoma cells HepG2 through activation of PI3K, mTORC1, p38 and MEK1/2 signaling pathways. Transcription factors PPARα, PPARγ, HNF4α and NF-κB are important contributors to this process. Insulin activates PPARγ through PI3K/Akt/mTORC1 pathway, which results in PPARγ binding to DR4 and DR0 cis-acting elements within the C3 promoter and subsequent displacement of HNF4α and PPARα from these sites. As a result PPARα/NF-κB complex, which exists on C3 promoter, is broken down and C3 gene expression is downregulated. The data obtained can potentially be used to explain the molecular mechanism underlying the correlation between heightened level of plasma C3 and insulin resistance in humans.


Assuntos
Complemento C3/biossíntese , Regulação da Expressão Gênica/fisiologia , Hepatócitos/metabolismo , Insulina/metabolismo , PPAR gama/metabolismo , Animais , Complemento C3/genética , Regulação para Baixo , Células Hep G2 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Transdução de Sinais/fisiologia
5.
J Cell Biochem ; 118(2): 382-396, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27404023

RESUMO

Apolipoprotein A-I (ApoA-I) is a key component of high density lipoproteins which possess anti-atherosclerotic and anti-inflammatory properties. Insulin is a crucial mediator of the glucose and lipid metabolism that has been implicated in atherosclerotic and inflammatory processes. Important mediators of insulin signaling such as Liver X Receptors (LXRs) and Forkhead Box A2 (FOXA2) are known to regulate apoA-I expression in liver. Forkhead Box O1 (FOXO1) is a well-known target of insulin signaling and a key mediator of oxidative stress response. Low doses of insulin were shown to activate apoA-I expression in human hepatoma HepG2 cells. However, the detailed mechanisms for these processes are still unknown. We studied the possible involvement of FOXO1, FOXA2, LXRα, and LXRß transcription factors in the insulin-mediated regulation of apoA-I expression. Treatment of HepG2 cells with high doses of insulin (48 h, 100 nM) suppresses apoA-I gene expression. siRNAs against FOXO1, FOXA2, LXRß, or LXRα abrogated this effect. FOXO1 forms a complex with LXRß and insulin treatment impairs FOXO1/LXRß complex binding to hepatic enhancer and triggers its nuclear export. Insulin as well as LXR ligand TO901317 enhance the interaction between FOXA2, LXRα, and hepatic enhancer. These data suggest that high doses of insulin downregulate apoA-I gene expression in HepG2 cells through redistribution of FOXO1/LXRß complex, FOXA2, and LXRα on hepatic enhancer of apoA-I gene. J. Cell. Biochem. 118: 382-396, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Apolipoproteína A-I/biossíntese , Carcinoma Hepatocelular/metabolismo , Regulação para Baixo/efeitos dos fármacos , Proteína Forkhead Box O1/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Insulina/farmacologia , Neoplasias Hepáticas/metabolismo , Receptores X do Fígado/metabolismo , Proteínas de Neoplasias/metabolismo , Carcinoma Hepatocelular/patologia , Células Hep G2 , Humanos , Hidrocarbonetos Fluorados/farmacologia , Neoplasias Hepáticas/patologia , Sulfonamidas/farmacologia
6.
Cell Stress Chaperones ; 22(1): 123-134, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27896567

RESUMO

Reactive oxygen species damage various cell components including DNA, proteins, and lipids, and these impairments could be a reason for severe human diseases including atherosclerosis. Forkhead box O1 (FOXO1), an important metabolic transcription factor, upregulates antioxidant and proapoptotic genes during oxidative stress. Apolipoprotein A-I (ApoA-I) forms high density lipoprotein (HDL) particles that are responsible for cholesterol transfer from peripheral tissues to liver for removal in bile in vertebrates. The main sources for plasma ApoA-I in mammals are liver and jejunum. Hepatic apoA-I transcription depends on a multitude of metabolic transcription factors. We demonstrate that ApoA-I synthesis and secretion are decreased during H2O2-induced oxidative stress in human hepatoma cell line HepG2. Here, we first show that FOXO1 binds to site B of apoA-I hepatic enhancer and downregulates apoA-I gene activity in HepG2 cells. Moreover, FOXO1 and LXRα transcription factors participate in H2O2-triggered downregulation of apoA-I gene together with Src, JNK, p38, and AMPK kinase cascades. Mutations of sites B or C as well as the administration of siRNAs against FOXO1 or LXRα to HepG2 cells abolished the hydrogen peroxide-mediated suppression of apoA-I gene.


Assuntos
Proteína Forkhead Box O1/metabolismo , Peróxido de Hidrogênio/toxicidade , Receptores X do Fígado/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Regulação para Baixo/efeitos dos fármacos , Proteína Forkhead Box O1/antagonistas & inibidores , Proteína Forkhead Box O1/genética , Células Hep G2 , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Receptores X do Fígado/antagonistas & inibidores , Receptores X do Fígado/genética , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Regulação para Cima/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
7.
J Cell Biochem ; 117(9): 2010-22, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26813964

RESUMO

Apolipoprotein A-I (ApoA-I) is the main anti-atherogenic component of human high-density lipoproteins (HDL). ApoA-I gene expression is regulated by several nuclear receptors, which are the sensors for metabolic changes during development of cardiovascular diseases. Activation of nuclear receptor PPARγ has been shown to impact lipid metabolism as well as inflammation. Here, we have shown that synthetic PPARγ agonist GW1929 decreases both ApoA-I mRNA and protein levels in HepG2 cells and the effect of GW1929 on apoA-I gene transcription depends on PPARγ. PPARγ binds to the sites A and C within the hepatic enhancer of apoA-I gene and the negative regulation of apoA-I gene transcription by PPARγ appears to be realized via the site C (-134 to -119). Ligand activation of PPARγ leads to an increase of LXRß and a decrease of PPARα binding to the apoA-I gene hepatic enhancer in HepG2 cells. GW1929 abolishes the TNFα-mediated decrease of ApoA-I mRNA expression in both HepG2 and Caco-2 cells but does not block TNFα-mediated inhibition of ApoA-I protein secretion by HepG2 cells. These data demonstrate that complex of PPARγ with GW1929 is a negative regulator involved in the control of ApoA-I expression and secretion in human hepatocyte- and enterocyte-like cells. J. Cell. Biochem. 117: 2010-2022, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Apolipoproteína A-I/biossíntese , Regulação para Baixo , Elementos Facilitadores Genéticos , Enterócitos/metabolismo , Hepatócitos/metabolismo , PPAR gama/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Apolipoproteína A-I/genética , Benzofenonas/farmacologia , Células CACO-2 , Células Hep G2 , Humanos , Receptores X do Fígado/genética , Receptores X do Fígado/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo , PPAR gama/genética , Fator de Necrose Tumoral alfa/genética , Tirosina/análogos & derivados , Tirosina/farmacologia
8.
Gene ; 524(2): 187-92, 2013 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-23628799

RESUMO

Complement C3 is involved in various protective and regulatory mechanisms of immune system. Recently it was established that C3 expression is regulated by nuclear receptors. Hepatic nuclear factor 4α (HNF4α) is a nuclear receptor critical for hepatic development and metabolism. We have shown that HNF4α is a positive regulator of C3 gene expression, realizing its effects through binding to two HNF4-response elements within the C3 promoter in HepG2 cells. TNFα is a well established positive regulator of C3 expression in hepatocytes during acute phase of inflammation. TNFα decreases the amount of HNF4α protein in HepG2 cells through NF-κB and MEK1/2 pathways thereby leading to a decrease in HNF4α bound to the C3 promoter. TNFα and HNF4α act in a synergetic way resulting in the potent activation of C3 transcription. These results suggest a novel mechanism of C3 regulation during acute phase response in HepG2 cells and display the mechanism of interaction of TNFα-induced pathways and HNF4α in transcriptional regulation of C3 gene.


Assuntos
Complemento C3/genética , Regulação da Expressão Gênica , Fator 4 Nuclear de Hepatócito/genética , Fator de Necrose Tumoral alfa/imunologia , Complemento C3/imunologia , Complemento C3/metabolismo , Células Hep G2 , Fator 4 Nuclear de Hepatócito/imunologia , Fator 4 Nuclear de Hepatócito/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/imunologia , Hepatócitos/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , NF-kappa B/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Mapeamento de Interação de Proteínas , Ativação Transcricional , Fator de Necrose Tumoral alfa/farmacologia , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA