Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Drug Metab Dispos ; 50(5): 704-715, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34949673

RESUMO

Ritonavir (RTV), a pharmacoenhancer used in anti-HIV regimens, can induce liver damage. RTV is primarily metabolized by cytochrome P450 3A4 (CYP3A4) in the liver. HNF4A antisense RNA 1 (HNF4A-AS1) and HNF1A antisense RNA 1 (HNF1A-AS1) are long noncoding RNAs that regulate the expression of pregnane X receptor (PXR) and CYP3A4. This study investigated the role and underlying mechanisms of HNF4A-AS1 and HNF1A-AS1 in RTV-induced hepatotoxicity. HNF4A-AS1 and HNF1A-AS1 were knocked down by small hairpin RNAs in Huh7 and HepG2 cells. Lactate dehydrogenase and reactive oxygen species assays were performed to assess RTV-induced hepatotoxicity. Chromatin immunoprecipitation quantitative real-time polymerase chain reaction was used to detect PXR enrichment and histone modifications in the CYP3A4 promoter. HNF4A-AS1 knockdown increased PXR and CYP3A4 expression and exacerbated RTV-induced cytotoxicity, whereas HNF1A-AS1 knockdown generated the opposite phenotype. Mechanistically, enrichment of PXR and trimethylation of histone 3 lysine 4 (H3K4me3) in the CYP3A4 promoter was increased, and trimethylation of histone 3 lysine 27 (H3K27me3) was decreased after HNF4A-AS1 knockdown. However, PXR and H3K4me3 enrichment decreased after HNF1A-AS1 knockdown. Alterations in RTV-induced hepatotoxicity caused by decreasing HNF4A-AS1 or HNF1A-AS1 were reversed by knockdown or overexpression of PXR. Increased susceptibility to RTV-induced liver injury caused by the PXR activator rifampicin was attenuated by HNF4A-AS1 overexpression or HNF1A-AS1 knockdown. Taken together, these results revealed that HNF4A-AS1 and HNF1A-AS1 modulated RTV-induced hepatotoxicity by regulating CYP3A4 expression, primarily by affecting the binding of PXR and histone modification status in the CYP3A4 promoter. SIGNIFICANCE STATEMENT: HNF4A-AS1 and HNF1A-AS1, transcribed separately from neighboring antisense genes of the human transcription factor genes HNF4A and HNF1A, were identified as long noncoding RNAs that can affect RTV-induced hepatotoxicity and susceptibility to RTV-induced hepatotoxicity caused by rifampicin exposure, mainly by affecting the expression of CY3A4 via alterations in PXR enrichment and histone modification status in the CYP3A4 promoter. This discovery provides directions for further research on the mechanisms of RTV-induced liver injury.


Assuntos
Carcinoma Hepatocelular , Doença Hepática Crônica Induzida por Substâncias e Drogas , Neoplasias Hepáticas , RNA Longo não Codificante , Receptores de Esteroides , Carcinoma Hepatocelular/genética , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Fator 1 Nuclear de Hepatócito/metabolismo , Fator 4 Nuclear de Hepatócito/genética , Fator 4 Nuclear de Hepatócito/metabolismo , Histonas/metabolismo , Humanos , Neoplasias Hepáticas/genética , Lisina , RNA Antissenso/genética , RNA Longo não Codificante/genética , Receptores de Esteroides/metabolismo , Rifampina/toxicidade , Ritonavir/toxicidade
2.
Sci Rep ; 10(1): 9999, 2020 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-32561878

RESUMO

Brief refeeding times (~60 min) enhanced hepatic Angptl8 expression in fasted mice. We cloned the mouse Angptl8 promoter region to characterise this rapid refeeding-induced increase in hepatic Angptl8 expression. Deletion of the -309/-60 promoter region significantly attenuated basal promoter activity in hepatocytes. A computational motif search revealed a potential binding motif for hepatocyte nuclear factor 1α/1ß (HNF-1α/ß) at -84/-68 bp of the promoter. Mutation of the HNF-1 binding site significantly decreased the promoter activity in hepatocytes, and the promoter carrying the mutated HNF-1 site was not transactivated by co-transfection of HNF-1 in a non-hepatic cell line. Silencing Hnf-1 in hepatoma cells and mouse primary hepatocytes reduced Angptl8 protein levels. Electrophoretic mobility-shift assays confirmed direct binding of Hnf-1 to its Angptl8 promoter binding motif. Hnf-1α expression levels increased after short-term refeeding, paralleling the enhanced in vivo expression of the Angptl8 protein. Chromatin immunoprecipitation (ChIP) confirmed the recruitment of endogenous Hnf-1 to the Angptl8 promoter region. Insulin-treated primary hepatocytes showed increased expression of Angptl8 protein, but knockdown of Hnf-1 completely abolished this enhancement. HNF-1 appears to play essential roles in the rapid refeeding-induced increases in Angptl8 expression. HNF-1α may therefore represent a primary medical target for ANGPTL8-related metabolic abnormalities. The study revealed the transcriptional regulation of the mouse hepatic Angptl8 gene by HNF-1.


Assuntos
Proteínas Semelhantes a Angiopoietina/genética , Regulação da Expressão Gênica , Fator 1 Nuclear de Hepatócito/genética , Fígado/metabolismo , Transcrição Gênica/genética , Proteína 8 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina/metabolismo , Animais , Linhagem Celular , Imunoprecipitação da Cromatina , Fator 1 Nuclear de Hepatócito/metabolismo , Hepatócitos/metabolismo , Camundongos , Regiões Promotoras Genéticas
3.
J Appl Toxicol ; 39(12): 1640-1650, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31385618

RESUMO

Acetaminophen (APAP) is an antipyretic and analgesic, which is commonly associated with drug-induced hepatic injury. C2-ceramide plays a key role in mediating cell life activities, and oltipraz was extensively studied as a cancer chemopreventive agent. Glutathione S-transferase A1 (GSTA1) acts as a vital liver detoxification enzyme. Hepatocyte nuclear factor 1 (HNF-1) regulates various cellular signaling pathways. In this study, we investigated the effects of C2-ceramide and oltipraz on APAP-induced hepatocyte injury and the changes of HNF-1 and GSTA1. Results showed that C2-ceramide (6 µmol/L) exacerbated APAP-induced hepatocyte injury and caused a significant decrease (P < .01) in HNF-1 and GSTA1 expressions. Meanwhile, GSTA1 content in supernatant was significantly increased (P < .01). In contrast, oltipraz (8 µmol/L) reduced the injury and significantly elevated (P < .01) HNF-1 and GSTA1 expressions while GSTA1 content in supernatant was significantly decreased (P < .01). In conclusion, these findings revealed that C2-ceramide inhibited HNF-1 and GSTA1 expression and exacerbated hepatocyte injury, while oltipraz treatment results in the reduction of hepatocyte injury, and promoted HNF-1 and GSTA1 expression. Additionally, the changes in HNF-1 and GSTA1 were related to APAP-induced hepatocyte injury. These results were useful to investigate the mechanism of an antipyretic and analgesic drug combination.


Assuntos
Acetaminofen/toxicidade , Analgésicos não Narcóticos/toxicidade , Glutationa Transferase/metabolismo , Fator 1 Nuclear de Hepatócito/metabolismo , Hepatócitos/efeitos dos fármacos , Pirazinas/farmacologia , Esfingosina/análogos & derivados , Antioxidantes/metabolismo , Técnicas de Cultura de Células , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Interações Medicamentosas , Expressão Gênica/efeitos dos fármacos , Glutationa Transferase/genética , Células Hep G2 , Fator 1 Nuclear de Hepatócito/genética , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Esfingosina/farmacologia , Tionas , Tiofenos
4.
Andrology ; 7(5): 741-747, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31050198

RESUMO

BACKGROUND: The epithelial lining of the human epididymis is critical for sperm maturation. This process requires distinct specialized functions in the head, body, and tail of the duct. These region-specific properties are maintained by distinct gene expression profiles which are governed by transcription factor networks, non-coding RNAs, and other factors. MATERIALS AND METHODS: We used genome-wide protocols including DNase-seq, RNA-seq and ChIP-seq to characterize open (active) chromatin, the transcriptome and occupancy of specific transcription factors (TFs) respectively, in caput, corpus, and cauda segments of adult human epididymis tissue and primary human epididymis epithelial (HEE) cell cultures derived from them. RNA-seq following TF depletion or activation, combined with gene ontology analysis also determined TF targets. RESULTS: Among regional differentially expressed transcripts were epithelial-selective transcription factors (TFs), microRNAs, and antiviral response genes. Caput-enriched TFs included hepatocyte nuclear factor 1 (HNF1) and the androgen receptor (AR), both of which were also predicted to occupy cis-regulatory elements identified as open chromatin in HEE cells. HNF1 targets were identified genome-wide using ChIP-seq, in HEE cells. Next, siRNA-mediated depletion of HNF1 revealed a pivotal role for this TF in coordinating epithelial water and solute transport in caput epithelium. The importance of AR in HEE cells was shown by AR ChIP-seq, and by RNA-seq after synthetic androgen (R1881) treatment. AR has a distinct transcriptional program in the HEE cells and likely recruits different co-factors (RUNX1 and CEBPß) in comparison to those used in prostate epithelium. DISCUSSION AND CONCLUSION: Our data identify many transcription factors that regulate the development and differentiation of HEE cells. Moreover, a comparison between immature and adult HEE cells showed key TFs in the transition to fully differentiated function of this epithelium. These data may help identify new targets to treat male infertility and have the potential to open new avenues for male contraception.


Assuntos
Biologia Computacional/métodos , Epididimo/metabolismo , Regulação da Expressão Gênica/genética , Redes Reguladoras de Genes/genética , Perfilação da Expressão Gênica , Fator 1 Nuclear de Hepatócito/metabolismo , Humanos , Masculino , Maturação do Esperma/genética , Transcriptoma/genética
5.
Cardiovasc Res ; 115(11): 1646-1658, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-30715251

RESUMO

AIMS: We previously reported that sodium-dependent glucose cotransporter 1 (SGLT1) is highly expressed in cardiomyocytes and is further up-regulated in ischaemia. This study aimed to determine the mechanisms by which SGLT1 contributes to ischaemia/reperfusion (I/R) injury. METHODS AND RESULTS: Mice with cardiomyocyte-specific knockdown of SGLT1 (TGSGLT1-DOWN) and wild-type controls were studied. In vivo, the left anterior descending coronary artery was ligated for 30 min and reperfused for 48 h. Ex vivo, isolated perfused hearts were exposed to 20 min no-flow and up to 2 h reperfusion. In vitro, HL-1 cells and isolated adult murine ventricular cardiomyocytes were exposed to 1 h hypoxia and 24 h reoxygenation (H/R). We found that TGSGLT1-DOWN hearts were protected from I/R injury in vivo and ex vivo, with decreased infarct size, necrosis, dysfunction, and oxidative stress. 5'-AMP-activated protein kinase (AMPK) activation increased SGLT1 expression, which was abolished by extracellular signal-related kinase (ERK) inhibition. Co-immunoprecipitation studies showed that ERK, but not AMPK, interacts directly with SGLT1. AMPK activation increased binding of the hepatocyte nuclear factor 1 and specificity protein 1 transcription factors to the SGLT1 gene, and HuR to SGLT1 mRNA. In cells, up-regulation of SGLT1 during H/R was abrogated by AMPK inhibition. Co-immunoprecipitation studies showed that SGLT1 interacts with epidermal growth factor receptor (EGFR), and EGFR interacts with protein kinase C (PKC). SGLT1 overexpression activated PKC and NADPH oxidase 2 (Nox2), which was attenuated by PKC inhibition, EGFR inhibition, and/or disruption of the interaction between EGFR and SGLT1. CONCLUSION: During ischaemia, AMPK up-regulates SGLT1 through ERK, and SGLT1 interacts with EGFR, which in turn increases PKC and Nox2 activity and oxidative stress. SGLT1 may represent a novel therapeutic target for mitigating I/R injury.


Assuntos
Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Transportador 1 de Glucose-Sódio/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Proteína Semelhante a ELAV 1/metabolismo , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Fator 1 Nuclear de Hepatócito/metabolismo , Masculino , Camundongos Knockout , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miócitos Cardíacos/patologia , NADPH Oxidase 2/metabolismo , NADPH Oxidases/metabolismo , Necrose , Estresse Oxidativo , Proteína Quinase C/metabolismo , Transdução de Sinais , Transportador 1 de Glucose-Sódio/deficiência , Transportador 1 de Glucose-Sódio/genética
6.
Biochem Biophys Res Commun ; 495(2): 1758-1765, 2018 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-29233692

RESUMO

The chipmunk hibernation-related proteins (HPs) HP-20 and HP-27 are components of a 140-kDa complex that dramatically decreases in the blood during hibernation. The HP-20 and HP-27 genes are expressed specifically in the liver and are downregulated in hibernating chipmunks. Hibernation-associated physiological changes are assumed to be under genetic control. Therefore, to elucidate the molecular mechanisms of hibernation, here we examined the mechanisms behind the altered HP-20 and HP-27 gene expression in nonhibernating versus hibernating chipmunks. Chromatin immunoprecipitation (ChIP) analyses revealed that histone H3 on the HP-20 and HP-27 gene promoters was highly acetylated at lysine (K) 9 and K14 and highly trimethylated at K4 in the liver of nonhibernating chipmunks, while these active histone modifications were nearly absent in hibernating chipmunks. Furthermore, histone acetyltransferases and a histone methyltransferase were associated with the HP-20 and HP-27 gene promoters primarily in nonhibernating chipmunks. Consistent with a previous finding that HNF-1 and USF can activate HP-20 and HP-27 gene transcription by binding to the proximal promoter region, ChIP-quantitative PCR (qPCR) analyses revealed that significantly less HNF-1 and USF were bound to these gene promoters in hibernating than in nonhibernating chipmunks. These findings collectively indicated that the hibernation-associated HP-20 and HP-27 gene expression is epigenetically regulated at the transcriptional level by the binding of HNF-1 and USF to their proximal promoters, and that histone modification has a key role in hibernation-associated transcriptional regulation.


Assuntos
Proteínas Sanguíneas/genética , Proteínas Sanguíneas/fisiologia , Hibernação/genética , Hibernação/fisiologia , Sciuridae/genética , Sciuridae/fisiologia , Animais , Sequência de Bases , Epigênese Genética , Expressão Gênica , Fator 1 Nuclear de Hepatócito/metabolismo , Histonas/metabolismo , Masculino , Regiões Promotoras Genéticas , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica , Fatores Estimuladores Upstream/metabolismo
7.
Int J Mol Med ; 39(3): 749-756, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28204827

RESUMO

Proprotein convertase subtilisin/kexin type 9 (PCSK9) impedes low­density lipoprotein (LDL) receptor (LDLR)-mediated LDL-cholesterol uptake and has hence emerged as a critical regulator of serum cholesterol levels and a new therapeutic target for the treatment of hypercholesterolemia. Statins have been shown to elevate circulating PCSK9 levels by stimulating PCSK9 gene transcription, which reduces the clinical efficacy of statin in LDL­cholesterol reduction. The transcription of PCSK9 is partially controlled by the hepatocyte nuclear factor 1 (HNF1) binding site embedded in the proximal region of its promoter. In this study, we utilized adenoviral shRNA delivery vectors to generate liver-specific knockdown of HNF1α (Ad­shHNF1α) or HNF1ß (Ad­shHNF1ß) in hamsters to examine the impact of reduced hepatic expression of HNF1 transcription factors on statin­induced elevation of PCSK9 expression and serum cholesterol levels. We showed that the administration of rosuvastatin (RSV) to normolipidemic hamsters significantly augmented hepatic PCSK9 expression and serum PCSK9 levels. In addition, RSV treatment increased hepatic HNF1α protein levels without a clear effect on HNF1α mRNA expression. Injection of Ad-shHNF1α or Ad­shHNF1ß into hamsters both blunted RSV­induced elevation of PCSK9 serum concentration and hepatic mRNA and protein levels, which led to significant increases in liver LDLR protein abundance. Furthermore, hepatic depletion of HNF1 factors lowered circulating total cholesterol and non­high density lipoprotein cholesterol levels in RSV­treated hamsters. Our study demonstrates that both HNF1α and HNF1ß are positive regulators of hepatic PCSK9 transcription in hamster species and that transient, liver-specific knockdown of either HNF1α or HNF1ß could antagonize the RSV­induced elevation of serum PCSK9 and reduce circulating cholesterol levels.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Fator 1 Nuclear de Hepatócito/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Pró-Proteína Convertase 9/genética , Rosuvastatina Cálcica/farmacologia , Adenoviridae/genética , Animais , Sequência de Bases , Colesterol/sangue , Clonagem Molecular , Cricetinae , Técnicas de Silenciamento de Genes , Inativação Gênica , Vetores Genéticos/genética , Fator 1 Nuclear de Hepatócito/genética , Fator 1-alfa Nuclear de Hepatócito/genética , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Fator 1-beta Nuclear de Hepatócito/genética , Fator 1-beta Nuclear de Hepatócito/metabolismo , Fígado/metabolismo , Masculino , Pró-Proteína Convertase 9/sangue , Pró-Proteína Convertase 9/metabolismo , RNA Interferente Pequeno/genética , Receptores de LDL/metabolismo , Transdução de Sinais , Proteínas de Ligação a Elemento Regulador de Esterol/metabolismo , Transdução Genética
8.
Br Poult Sci ; 58(1): 19-25, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27844479

RESUMO

1. Albumin (ALB) is a serum protein most highly expressed in liver and regarded as an effective indicator for liver pathologies. The objectives of this study were to determine the expression of duck ALB gene (duALB) in various non-hepatic tissues and identify the potential cis-regulatory elements in the promoter. 2. A model was established to assess duALB promoter activity in different cell lines by construction of a duALB promoter-driven GFP (Green Fluorescent Protein)-expressing vector, which exhibited high expression activity in liver-derived cells and lower expression in other cells. Through the firefly luciferase reporter gene driven by a series of constructs carrying progressive deletions, the core transcriptional regulatory region within the duALB promoter was identified. Mutations in candidate-binding sites were made by site-directed mutagenesis. 3. The core transcriptional regulatory region was located in the -190/-51 bp region. This region contains three potential transcription factor-binding sites, one each for hepatocyte nuclear factor (HNF-3ß) (-158/-149), CCAAT/Enhancer-binding protein element (C/EBPα) (-119/-107) and nuclear factor-1 (HNF-1) (-67/-57). Site-directed mutagenesis of HNF-1 and C/EBPα-binding sites resulted in a significant reduction in duALB promoter activity. Two potential cis-regulatory elements (C/EBPα and HNF-1) were responsible for its transcriptional activity in liver-derived cells. 4. These findings contribute to the further understanding of the fundamental mechanisms of ALB gene regulation and the use of tissue-specific gene promoters to regulate tissue-specific expression of exogenous genes in vivo.


Assuntos
Patos/genética , Expressão Gênica/genética , Regiões Promotoras Genéticas/genética , Albumina Sérica/genética , Animais , Sítios de Ligação/genética , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , DNA/metabolismo , Regulação da Expressão Gênica/genética , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Fator 1 Nuclear de Hepatócito/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Fígado/metabolismo , Mutagênese Sítio-Dirigida
9.
Mol Cell Endocrinol ; 425: 94-102, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26808453

RESUMO

The luminal environment of the epididymis participates in sperm maturation and impacts male fertility. It is dependent on the coordinated expression of many genes encoding proteins with a role in epithelial transport. We identified cis-regulatory elements for critical genes in epididymis function, by mapping open chromatin genome-wide in human epididymis epithelial (HEE) cells. Bioinformatic predictions of transcription factors binding to the regulatory elements suggested an important role for hepatocyte nuclear factor 1 (HNF1) in the transcriptional program of these cells. Chromatin immunoprecipitation and deep sequencing (ChIP-seq) revealed HNF1 target genes in HEE cells. In parallel, the contribution of HNF1 to the transcriptome of HEE cells was determined by RNA-seq, following siRNA-mediated depletion of both HNF1α and HNF1ß transcription factors. Repression of these factors caused differential expression of 1892 transcripts (902 were downregulated and 990 upregulated) in comparison to non-targeting siRNAs. Differentially expressed genes with HNF1 ChIP-seq peaks within 20 kb were subject to gene ontology process enrichment analysis. Among the most significant processes associated with down-regulated genes were epithelial transport of water, phosphate and bicarbonate, all critical processes in epididymis epithelial function. Measurements of intracellular pH (pHi) confirmed a role for HNF1 in regulating the epididymis luminal environment.


Assuntos
Epididimo/metabolismo , Redes Reguladoras de Genes , Fator 1 Nuclear de Hepatócito/genética , Fator 1 Nuclear de Hepatócito/metabolismo , Elementos Reguladores de Transcrição , Células Cultivadas , Imunoprecipitação da Cromatina , Biologia Computacional/métodos , Epididimo/citologia , Regulação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Transcrição Gênica
10.
J Pharmacol Exp Ther ; 355(3): 429-41, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26427720

RESUMO

Cytosolic sulfotransferase 1C2 (SULT1C2) is expressed in the kidney, stomach, and liver of rats; however, the mechanisms regulating expression of this enzyme are not known. We evaluated transcriptional regulation of SULT1C2 by mevalonate (MVA)-derived intermediates in primary cultured rat hepatocytes using several cholesterol synthesis inhibitors. Blocking production of mevalonate with the 3-hydroxy-3-methylglutaryl-CoA reductase inhibitor pravastatin (30 µM), reduced SULT1C2 mRNA content by ∼40% whereas the squalene synthase inhibitor squalestatin (SQ1, 0.1 µM), which causes accumulation of nonsterol isoprenoids, increased mRNA content by 4-fold. Treatment with MVA (10 mM) strongly induced SULT1C2 mRNA by 12-fold, and this effect was blocked by inhibiting squalene epoxidase but not by more distal cholesterol inhibitors, indicating the effects of MVA are mediated by postsqualene metabolites. Using rapid amplification of cDNA ends (RACE), we characterized the 5' end of SULT1C2 mRNA and used this information to generate constructs for promoter analysis. SQ1 and MVA increased reporter activity by ∼1.6- and 3-fold, respectively, from a construct beginning 49 base pairs (bp) upstream from the longest 5'-RACE product (-3140:-49). Sequence deletions from this construct revealed a hepatocyte nuclear factor 1 (HNF1) element (-2558), and mutation of this element reduced basal (75%) and MVA-induced (30%) reporter activity and attenuated promoter activation following overexpression of HNF1α or 1ß. However, the effects of SQ1 were localized to a more proximal promoter region (-281:-49). Collectively, our findings demonstrate that cholesterol biosynthetic intermediates influence SULT1C2 expression in rat primary hepatocytes. Further, HNF1 appears to play an important role in mediating basal and MVA-induced SULT1C2 transcription.


Assuntos
Colesterol/biossíntese , Regulação Enzimológica da Expressão Gênica/fisiologia , Hepatócitos/enzimologia , Sulfotransferases/biossíntese , Sulfotransferases/genética , Animais , Anticolesterolemiantes/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Fator 1 Nuclear de Hepatócito/genética , Fator 1 Nuclear de Hepatócito/metabolismo , Hepatócitos/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Masculino , Ácido Mevalônico/farmacologia , Cultura Primária de Células , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Esqualeno Mono-Oxigenase/antagonistas & inibidores , Sulfotransferases/efeitos dos fármacos , Transfecção , Ácidos Tricarboxílicos/farmacologia
11.
Drug Metab Pharmacokinet ; 30(2): 188-97, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25989892

RESUMO

Constitutive androstane receptor (CAR) is one of the principal regulators of hepatic cytochrome P450s (CYPs) 3A (CYP3A). cDNA-mediated expression of a mature rat CAR (rCAR) into rat hepatoma cells induced CYP3A1 and CYP2B mRNAs. Aberrant rCAR failed in these inductions. Three important human CYP3A4 regulatory elements (REs), proximal ER6 (proER6), xenobiotic responsive enhancer module (XREM) and constitutive liver enhancer module (CLEM), support constitutive and inducible expression of CYP3As mediated by CAR and pregnane X receptor (PXR). NHR-scan software predicted proER6, XREM and CLEM at -255 b, -8 kb and -11.5 kb, respectively of CYP3A4, but neither XREM nor CLEM was predicted in rat CYP3A. A luciferase reporter construct carrying a 5'-flanking sequence of CYP3A1 (-31,739 to -31,585 from its transcription initiation site) revealed important for the rCAR-dependent transactivation of CYP3A1. This region includes two putative binding motifs of nuclear receptors (DR4 and DR2), a putative hepatocyte nuclear factor-1 binding motif (HNF1), nuclear factor-kappa B binding motif (NFκB), activator protein 1 binding motif (AP-1), and ecotropic viral integration site 1 binding motif (Evi1). We hereby conclude DR4 and/or DR2 motifs being primarily responsible and HNF1 being synergistically functioning elements for the rCAR-mediated transcription of CYP3A1.


Assuntos
Região 5'-Flanqueadora , Citocromo P-450 CYP3A/genética , Hepatócitos/enzimologia , Receptores Citoplasmáticos e Nucleares/genética , Elementos de Resposta , Animais , Sítios de Ligação , Carcinoma Hepatocelular/enzimologia , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Receptor Constitutivo de Androstano , Citocromo P-450 CYP3A/metabolismo , Dexametasona/farmacologia , Regulação Enzimológica da Expressão Gênica , Genes Reporter , Fator 1 Nuclear de Hepatócito/metabolismo , Hepatócitos/efeitos dos fármacos , Neoplasias Hepáticas/enzimologia , Neoplasias Hepáticas/genética , Masculino , Ligação Proteica , RNA Mensageiro/metabolismo , Ratos Endogâmicos F344 , Receptores Citoplasmáticos e Nucleares/metabolismo , Transcrição Gênica , Ativação Transcricional , Transfecção , Ácidos Tricarboxílicos/farmacologia
12.
Int J Cancer ; 135(3): 585-97, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24382740

RESUMO

Targeted approaches have revealed frequent epigenetic alterations in ovarian cancer, but the scope and relation of these changes to histologic subtype of disease is unclear. Genome-wide methylation and expression data for 14 clear cell carcinoma (CCC), 32 non-CCC and four corresponding normal cell lines were generated to determine how methylation profiles differ between cells of different histological derivations of ovarian cancer. Consensus clustering showed that CCC is epigenetically distinct. Inverse relationships between expression and methylation in CCC were identified, suggesting functional regulation by methylation, and included 22 hypomethylated (UM) genes and 276 hypermethylated (HM) genes. Categorical and pathway analyses indicated that the CCC-specific UM genes were involved in response to stress and many contain hepatocyte nuclear factor (HNF) 1-binding sites, while the CCC-specific HM genes included members of the estrogen receptor alpha (ERalpha) network and genes involved in tumor development. We independently validated the methylation status of 17 of these pathway-specific genes, and confirmed increased expression of HNF1 network genes and repression of ERalpha pathway genes in CCC cell lines and primary cancer tissues relative to non-CCC specimens. Treatment of three CCC cell lines with the demethylating agent Decitabine significantly induced expression for all five genes analyzed. Coordinate changes in pathway expression were confirmed using two primary ovarian cancer datasets (p < 0.0001 for both). Our results suggest that methylation regulates specific pathways and biological functions in CCC, with hypomethylation influencing the characteristic biology of the disease while hypermethylation contributes to the carcinogenic process.


Assuntos
Adenocarcinoma de Células Claras/genética , Metilação de DNA , Epigenômica , Neoplasias Ovarianas/genética , Adenocarcinoma de Células Claras/patologia , Feminino , Fator 1 Nuclear de Hepatócito/genética , Fator 1 Nuclear de Hepatócito/metabolismo , Humanos , Estadiamento de Neoplasias , Neoplasias Ovarianas/patologia , Reação em Cadeia da Polimerase em Tempo Real , Células Tumorais Cultivadas
13.
Meat Sci ; 94(4): 474-9, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23628452

RESUMO

The aim of this study was to investigate the association and expression of HNF1A gene as a candidate gene for meat and carcass quality traits in pigs. Statistical analysis revealed that the g.8260 A>G polymorphism significantly associated with pH 24(H), meat percentage and muscle area in the F2 Duroc × Pietrain (DuPi, n=313) and with pH 24(L), fat area and backfat thickness in the Pietrain (Pi, n=110) population. HNF1A mRNA and protein expressions were higher (p<0.05) in animals with the low post-mortem muscle pH 24(L). The promoter methylation profiling suggested that methylation was not involved on HNF1A expression regulation (p>0.05) in animal with divergent muscle pH. In conclusion, polymorphism in porcine HNF1A gene could be used as a candidate marker to improve the meat and carcass quality traits, with the consideration of breed-specific effect.


Assuntos
Tecido Adiposo/metabolismo , Cruzamento , Expressão Gênica , Fator 1 Nuclear de Hepatócito/genética , Carne/análise , Músculo Esquelético/metabolismo , Polimorfismo de Nucleotídeo Único , Animais , Dieta , Gorduras na Dieta/análise , Regulação da Expressão Gênica , Estudos de Associação Genética , Fator 1 Nuclear de Hepatócito/metabolismo , Concentração de Íons de Hidrogênio , Carne/normas , Metilação , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Sus scrofa
14.
Hum Mutat ; 32(10): 1153-60, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21681857

RESUMO

N-acetylglutamate synthase (NAGS) catalyzes the conversion of glutamate and acetyl-CoA to NAG, the essential allosteric activator of carbamyl phosphate synthetase I, the first urea cycle enzyme in mammals. A 17-year-old female with recurrent hyperammonemia attacks, the cause of which remained undiagnosed for 8 years in spite of multiple molecular and biochemical investigations, showed markedly enhanced ureagenesis (measured by isotope incorporation) in response to N-carbamylglutamate (NCG). This led to sequencing of the regulatory regions of the NAGS gene and identification of a deleterious single-base substitution in the upstream enhancer. The homozygous mutation (c.-3064C>A), affecting a highly conserved nucleotide within the hepatic nuclear factor 1 (HNF-1) binding site, was not found in single nucleotide polymorphism databases and in a screen of 1,086 alleles from a diverse population. Functional assays demonstrated that this mutation decreases transcription and binding of HNF-1 to the NAGS gene, while a consensus HNF-1 binding sequence enhances binding to HNF-1 and increases transcription. Oral daily NCG therapy restored ureagenesis in this patient, normalizing her biochemical markers, and allowing discontinuation of alternate pathway therapy and normalization of her diet with no recurrence of hyperammonemia. Inc.


Assuntos
Aminoácido N-Acetiltransferase/genética , Elementos Facilitadores Genéticos , Glutamatos/uso terapêutico , Deleção de Sequência , Distúrbios Congênitos do Ciclo da Ureia/tratamento farmacológico , Distúrbios Congênitos do Ciclo da Ureia/genética , Adolescente , Alelos , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Criança , Feminino , Frequência do Gene , Glutamatos/metabolismo , Células Hep G2 , Fator 1 Nuclear de Hepatócito/metabolismo , Humanos , Motivos de Nucleotídeos , Polimorfismo de Nucleotídeo Único , Alinhamento de Sequência , Resultado do Tratamento , Distúrbios Congênitos do Ciclo da Ureia/metabolismo
15.
J Biol Chem ; 286(19): 17259-69, 2011 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-21454713

RESUMO

HDL is a negative risk factor for atherosclerosis because of its multiple atheroprotective functions. Inflammation converts HDL particles from anti-atherogenic to pro-atherogenic, and this transformation is associated with changes in HDL structure and composition. Apolipoprotein M (apoM) has been recently shown to play a role in the maturation of HDL in plasma and to protect from atherosclerosis. ApoM gene is expressed primarily in the liver and kidney and is down-regulated by pro-inflammatory signals. We now show that the human apoM promoter harbors a dual specificity regulatory element in the proximal region that binds hepatocyte nuclear factor 1 (HNF-1) and members of the AP-1 family of pro-inflammatory transcription factors (c-Jun and JunB). Overexpression of c-Jun or JunB repressed both the basal and the HNF-1-mediated transactivation of the human apoM promoter. Treatment of HepG2 cells with potent inflammation-inducing phorbol esters or overexpression of PKCα was associated with a marked inhibition of apoM gene expression in a c-Jun/JunB-dependent manner. We provide evidence for a novel mechanism of inflammation-induced transcriptional repression that is based on the competition between HNF-1 and Jun proteins for binding to the same regulatory region. A similar mechanism accounts for the down-regulation of the liver-specific apolipoprotein A-II gene by Jun factors. Our studies provide novel insights on the mechanisms that control the expression of liver-specific apolipoprotein genes during inflammation and could affect the maturation and the functionality of HDL particles.


Assuntos
Apolipoproteínas/metabolismo , Regulação da Expressão Gênica , Fator 1 Nuclear de Hepatócito/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Apolipoproteínas M , Linhagem Celular , HDL-Colesterol/metabolismo , Humanos , Inflamação , Lipocalinas , Modelos Biológicos , Mutação , Ésteres de Forbol/metabolismo , Regiões Promotoras Genéticas , Fator de Transcrição AP-1/metabolismo , Ativação Transcricional
16.
J Biol Chem ; 286(8): 6049-60, 2011 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-21163946

RESUMO

Resveratrol, a polyphenol compound, is known for its effects on energy homeostasis. With properties of energy sensors mediating effects of calorie restriction, sirtuins are targets of resveratrol. The mammalian sirtuin homolog SIRT1 is a protein deacetylase playing a role in glucose metabolism and islet function. Here, we investigated the effects of resveratrol and possible link with SIRT1 in ß-cells. Insulinoma INS-1E cells and human islets were cultured with resveratrol before analyzing their physiological responses. Treatment of INS-1E cells for 24 h with 25 µM resveratrol resulted in marked potentiation of glucose-stimulated insulin secretion. This effect was associated with elevated glycolytic flux, resulting in increased glucose oxidation, ATP generation, and mitochondrial oxygen consumption. Such changes correlated with up-regulation of key genes for ß-cell function, i.e. Glut2, glucokinase, Pdx-1, Hnf-1α, and Tfam. In human islets, chronic resveratrol treatment similarly increased both the glucose secretory response and expression of the same set of genes, eventually restoring the glucose response in islets obtained from one type 2 diabetic donor. Overexpression of Sirt1 in INS-1E cells potentiated resveratrol effects on insulin secretion. Conversely, inhibition of SIRT1 achieved either by expression of an inactive mutant or by using the EX-527 inhibitor, both abolished resveratrol effects on glucose responses. Treatment of INS-1E cells with EX-527 also prevented resveratrol-induced up-regulation of Glut2, glucokinase, Pdx-1, and Tfam. Resveratrol markedly enhanced the glucose response of INS-1E cells and human islets, even after removal of the compound from the medium. These effects were mediated by and fully dependent on active SIRT1, defining a new role for SIRT1 in the regulation of insulin secretion.


Assuntos
Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Sirtuína 1/metabolismo , Animais , Carbazóis/farmacologia , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Inibidores Enzimáticos/farmacologia , Glucoquinase/genética , Glucoquinase/metabolismo , Glucose/farmacologia , Transportador de Glucose Tipo 2/genética , Transportador de Glucose Tipo 2/metabolismo , Fator 1 Nuclear de Hepatócito/genética , Fator 1 Nuclear de Hepatócito/metabolismo , Proteínas de Grupo de Alta Mobilidade/genética , Proteínas de Grupo de Alta Mobilidade/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Insulina/genética , Secreção de Insulina , Células Secretoras de Insulina/citologia , Mitocôndrias/genética , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Consumo de Oxigênio/fisiologia , Ratos , Resveratrol , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/genética , Estilbenos/farmacologia , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
17.
Exp Biol Med (Maywood) ; 235(1): 32-9, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20404016

RESUMO

The incidence of hepatocellular carcinoma is rising due to alcohol drinking, hepatitis C viral infection and metabolic syndrome. Differential expression of CYP2E1 may play a pleiotropic role in the multistep process of liver carcinogenesis. Considerable attention has focused on the antitumor effect of trichostatin A (TSA) as well as CYP2E1 expression-induced apoptosis of cancer cells. However, very few studies have examined the mechanisms by which TSA has an antitumor effect and its association to CYP2E1 expression. The current study examined the action of TSA on CYP2E1 expression and the role of CYP2E1 in inducing apoptosis of HepG2 cells. Our data showed that TSA selectively induced CYP2E1 in four studied human hepatocellular carcinoma (HCC) cell lines (Huh7, PLC/PRF/5, Hep3B and HepG2), but not in normal primary human hepatocytes. TSA-mediated up-regulation of CYP2E1 expression was associated with histone H3 acetylation and the recruitment of HNF-1 and HNF-3beta to the CYP2E1 promoter in HepG2 cells. siRNA-mediated knockdown experiments showed that TSA-induced caspase-3 cleavage was decreased due to reduced expression of CYP2E1 in HepG2 cells. Moreover, down-regulation of CYP2E1 was accompanied by decreased production of mitochondrial reactive oxygen species. These results suggest that histone modification is involved in CYP2E1 gene expression and that CYP2E1-dependent mitochondrial oxidative stress plays a role in TSA-induced apoptosis.


Assuntos
Apoptose/fisiologia , Carcinoma Hepatocelular/metabolismo , Citocromo P-450 CYP2E1/genética , Histonas/metabolismo , Neoplasias Hepáticas/metabolismo , Acetilação , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Sequência de Bases , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Caspase 3/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Citocromo P-450 CYP2E1/biossíntese , Inibidores do Citocromo P-450 CYP2E1 , Primers do DNA/genética , Indução Enzimática/efeitos dos fármacos , Expressão Gênica , Fator 1 Nuclear de Hepatócito/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Histonas/química , Humanos , Ácidos Hidroxâmicos/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Estresse Oxidativo , RNA Interferente Pequeno/genética
18.
Mol Nutr Food Res ; 54(10): 1445-51, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20425755

RESUMO

Maltase and glucoamylase are derived from the same mRNA and are responsible for digestion of starch in the small intestine. Their jejunal activities in rodents are induced by a high-starch/low-fat (HS)-diet. However, it is unknown whether jejunal expression of the maltase-glucoamylase (Mgam) gene is enhanced by the HS-diet. In this study, we found that jejunal Mgam mRNA was increased by a HS-diet in mice. We showed that the HS-diet increased acetylation of histones, bindings of a coactivator, Creb binding protein (CREBBP), and the transcriptional factors caudal type homeobox 2 (CDX2) and HNF1 homeobox (HNF1) in the promoter/enhancer and transcriptional regions of Mgam gene. This suggests that the increase in the jejunal activity of maltase and glucoamylase caused by a HS-diet in mice is regulated at the mRNA level through histone acetylation and binding of CREBBP, CDX2 and HNF1 in the promoter/enhancer and transcriptional regions of Mgam gene.


Assuntos
Dieta , Indução Enzimática , Jejuno/enzimologia , Amido/administração & dosagem , alfa-Glucosidases/biossíntese , Acetilação , Animais , Fator de Transcrição CDX2 , Proteína de Ligação a CREB/metabolismo , Proteínas de Ligação a DNA/metabolismo , Dieta/efeitos adversos , Dieta com Restrição de Gorduras , Fator 1 Nuclear de Hepatócito/metabolismo , Histonas/metabolismo , Proteínas de Homeodomínio/metabolismo , Jejuno/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fases de Leitura Aberta , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Amido/efeitos adversos , Fatores de Transcrição/metabolismo , alfa-Glucosidases/genética
19.
BMC Mol Biol ; 11: 9, 2010 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-20096102

RESUMO

BACKGROUND: UDP-glucuronosyltransferase 1A1 (UGT1A1) is a pivotal enzyme involved in metabolism of SN-38, the active metabolite of irinotecan commonly used to treat metastatic colorectal cancer. We previously demonstrated aberrant methylation of specific CpG dinucleotides in UGT1A1-negative cells, and revealed that methylation state of the UGT1A1 5'-flanking sequence is negatively correlated with gene transcription. Interestingly, one of these CpG dinucleotides (CpG -4) is found close to a HNF1 response element (HRE), known to be involved in activation of UGT1A1 gene expression, and within an upstream stimulating factor (USF) binding site. RESULTS: Gel retardation assays revealed that methylation of CpG-4 directly affect the interaction of USF1/2 with its cognate sequence without altering the binding for HNF1-alpha. Luciferase assays sustained a role for USF1/2 and HNF1-alpha in UGT1A1 regulation in colon cancer cells. Based on the differential expression profiles of HNF1A gene in colon cell lines, we also assessed whether methylation affects its expression. In agreement with the presence of CpG islands in the HNF1A promoter, treatments of UGT1A1-negative HCT116 colon cancer cells with a DNA methyltransferase inhibitor restore HNF1A gene expression, as observed for UGT1A1. CONCLUSIONS: This study reveals that basal UGT1A1 expression in colon cells is positively regulated by HNF1-alpha and USF, and negatively regulated by DNA methylation. Besides, DNA methylation of HNF1A could also play an important role in regulating additional cellular drug metabolism and transporter pathways. This process may contribute to determine local inactivation of drugs such as the anticancer agent SN-38 by glucuronidation and define tumoral response.


Assuntos
Neoplasias do Colo/genética , Regulação Neoplásica da Expressão Gênica , Glucuronosiltransferase/genética , Fator 1 Nuclear de Hepatócito/metabolismo , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Sequência de Bases , Neoplasias do Colo/metabolismo , Ilhas de CpG , Citidina Trifosfato/análogos & derivados , Citidina Trifosfato/farmacologia , Metilação de DNA , Ensaio de Desvio de Mobilidade Eletroforética , Glucuronosiltransferase/metabolismo , Células HCT116 , Células HT29 , Fator 1 Nuclear de Hepatócito/genética , Humanos , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Regulação para Cima
20.
Arterioscler Thromb Vasc Biol ; 30(3): 568-74, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20007910

RESUMO

OBJECTIVE: Our aim was to elucidate mechanisms involved in the acquisition of lipid transport properties during enterocyte differentiation. METHODS AND RESULTS: We show that lipid mobilization via apolipoprotein B lipoproteins is dependent on the expression of microsomal triglyceride transfer protein (MTP) during differentiation of Caco-2 cells into enterocyte-like cells. Mechanistic studies showed that binding of the nuclear receptor family 2 group F member 1 (NR2F1) to the DR1 element in the MTTP promoter suppresses MTTP expression in undifferentiated cells. During cellular differentiation, NR2F1 expression and its binding to MTTP promoter decline and MTP induction ensues. Moreover, undifferentiated cells express inositol-requiring enzyme 1beta (IRE1beta), a protein that posttranscriptionally degrades MTP mRNA, and its expression substantially decreases during differentiation, contributing to MTP induction. Immunohistochemical studies revealed a significant negative relationship between the expressions of MTP and NR2F1/IRE1beta in undifferentiated and differentiated Caco-2 cells, as well as in crypt-villus and jejunum-colon axes of mouse intestine. CONCLUSIONS: We propose that transcriptional and posttranscriptional mechanisms involving NR2F1 and IRE1beta ensure low MTP expression in undifferentiated intestinal cells and avoid apolipoprotein B lipoprotein biosynthesis.


Assuntos
Fator I de Transcrição COUP/metabolismo , Proteínas de Transporte/metabolismo , Diferenciação Celular , Endorribonucleases/metabolismo , Mucosa Intestinal/metabolismo , Lipoproteínas/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Apolipoproteínas B/metabolismo , Sequência de Bases , Células CACO-2 , Colo/citologia , Colo/metabolismo , Enterócitos/citologia , Enterócitos/metabolismo , Fator 1 Nuclear de Hepatócito/genética , Fator 1 Nuclear de Hepatócito/metabolismo , Fator 4 Nuclear de Hepatócito/genética , Fator 4 Nuclear de Hepatócito/metabolismo , Humanos , Jejuno/citologia , Jejuno/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Dados de Sequência Molecular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA