Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Biochem Pharmacol ; 197: 114897, 2022 03.
Article in English | MEDLINE | ID: mdl-34968487

ABSTRACT

Acetaminophen (APAP) overdose is one of the leading causes of acute liver failure in the US and other developed countries, the molecular mechanisms of APAP-induced hepatotoxicity remain speculative. PIWI-interacting RNAs (piRNAs), a novel class of small non-coding RNAs, have been identified as epigenetic regulators of transposon silencing, mRNA deadenylation, and elimination. However, the functional role of piRNAs in APAP-induced liver injury remains unclear. In the current study, the piRNA profiles were constructed in HepaRG cells after APAP exposure, and the roles of piR-23210 in regulating nuclear receptors (NRs) expression, metabolizing enzymes expression, and consequently APAP-induced liver injury were systematically investigated. As a result, 57 upregulated piRNAs were identified after APAP exposure, indicating the stress-response characteristic of piRNA molecules. Subsequent in vitro and in vivo experiments proved that piR-23210 is a novel self-protective molecule that targets HNF1A and HNF4A transcripts by interacting with RNA binding protein Nucleolin (NCL), suppresses downstream CYPs (CYP2E1, CYP3A4, and CYP1A2) expression, and protects against APAP-induced liver injury. In conclusion, our findings provided new mechanistic clues revealing potential protective role of a piRNA against the hepatoxicity of APAP.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Hepatocyte Nuclear Factor 1-alpha/metabolism , Hepatocyte Nuclear Factor 4/metabolism , RNA, Small Interfering/metabolism , Animals , Chemical and Drug Induced Liver Injury/prevention & control , HEK293 Cells , Hep G2 Cells , Hepatocyte Nuclear Factor 1-alpha/antagonists & inhibitors , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Humans , Male , Mice , Mice, Inbred C57BL , RNA, Small Interfering/administration & dosage
2.
Cancer Res ; 81(23): 5849-5861, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34649947

ABSTRACT

Multiple noncoding natural antisense transcripts (ncNAT) are known to modulate key biological events such as cell growth or differentiation. However, the actual impact of ncNATs on cancer progression remains largely unknown. In this study, we identified a complete list of differentially expressed ncNATs in hepatocellular carcinoma. Among them, a previously undescribed ncNAT HNF4A-AS1L suppressed cancer cell growth by regulating its sense gene HNF4A, a well-known cancer driver, through a promoter-specific mechanism. HNF4A-AS1L selectively activated the HNF4A P1 promoter via HNF1A, which upregulated expression of tumor suppressor P1-driven isoforms, while having no effect on the oncogenic P2 promoter. RNA-seq data from 23 tissue and cancer types identified approximately 100 ncNATs whose expression correlated specifically with the activity of one promoter of their associated sense gene. Silencing of two of these ncNATs ENSG00000259357 and ENSG00000255031 (antisense to CERS2 and CHKA, respectively) altered the promoter usage of CERS2 and CHKA. Altogether, these results demonstrate that promoter-specific regulation is a mechanism used by ncNATs for context-specific control of alternative isoform expression of their counterpart sense genes. SIGNIFICANCE: This study characterizes a previously unexplored role of ncNATs in regulation of isoform expression of associated sense genes, highlighting a mechanism of alternative promoter usage in cancer.


Subject(s)
Carcinoma, Hepatocellular/pathology , Choline Kinase/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Liver Neoplasms/pathology , Membrane Proteins/metabolism , Promoter Regions, Genetic , RNA, Antisense/genetics , Sphingosine N-Acyltransferase/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Choline Kinase/antagonists & inhibitors , Choline Kinase/genetics , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Hepatocyte Nuclear Factor 4/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, SCID , Prognosis , Sphingosine N-Acyltransferase/antagonists & inhibitors , Sphingosine N-Acyltransferase/genetics , Tumor Cells, Cultured , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays
3.
J Hepatol ; 75(5): 1072-1082, 2021 11.
Article in English | MEDLINE | ID: mdl-34242702

ABSTRACT

BACKGROUND & AIMS: Our understanding of the interactions between HBV and its host cells is still quite limited. Spliceosome associated factor 1 (SART1) has recently been found to restrict HCV. Thus, we aimed to dissect its role in HBV infection. METHODS: SART1 was knocked down by RNA interference and over-expressed by lentiviral or adeno-associated virus (AAV) vectors in HBV-infected cell cultures and in vivo in HBV-infected mice. Luciferase reporter assays were used to determine viral or host factor promoter activities, and chromatin immunoprecipitation (ChIP) was used to investigate protein-DNA interactions. RESULTS: In HBV-infected cell cultures, downregulation of SART1 did not affect covalently closed circular HBV DNA but resulted in markedly enhanced HBV RNA, antigen expression and progeny virus production. On the other hand, HBV transcription and replication were significantly inhibited by overexpression of SART1. Similar results were observed in AAV-HBV-infected mice persistently replicating HBV. Inhibition of Janus kinases had no effect on SART1-mediated inhibition of HBV replication. HBV promoter assays revealed that SART1 reduced HBV core promoter activity. By screening known HBV transcription factors, we found that SART1 specifically suppressed the expression of hepatocyte nuclear factor 4α (HNF4α). Luciferase reporter and ChIP assays demonstrated a direct downregulation of HNF4α expression by association of SART1 with the HNF4α proximal P1 promoter element. CONCLUSIONS: We identify SART1 as a novel host factor suppressing HBV cccDNA transcription. Besides its effect on interferon-stimulated genes, SART1 exerts an anti-HBV activity by suppressing HNF4α expression, which is essential for transcription of HBV cccDNA. LAY SUMMARY: Hepatitis B virus (HBV) infects hepatocytes and persists in the form of covalently closed circular DNA (cccDNA), which remains a major obstacle to successful antiviral treatment. In this study, using various HBV models, we demonstrate that the protein SART1 restricts HBV cccDNA transcription by suppressing a key transcription factor, HNF4α.


Subject(s)
Antiviral Agents/metabolism , Gene Regulatory Networks/genetics , Hepatitis B/drug therapy , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Ribonucleoproteins, Small Nuclear/pharmacology , Antiviral Agents/immunology , Gene Expression Regulation/drug effects , Gene Regulatory Networks/drug effects , Hepatitis B/physiopathology , Hepatocyte Nuclear Factor 4/metabolism , Humans , Ribonucleoproteins, Small Nuclear/therapeutic use , Virus Replication/drug effects
4.
Cancer Gene Ther ; 28(3-4): 307-320, 2021 04.
Article in English | MEDLINE | ID: mdl-33199830

ABSTRACT

Gastric cancer (GC) is a common cancer and a leading cause of cancer-related deaths worldwide. Recent studies have supported the important role of long non-coding RNAs (lncRNAs) in GC progression. This study identified functional significance of X inactive specific transcript (XIST) in GC. The expression of XIST and EPHA1 in GC tissues and cells was measured. Then, dual luciferase reporter gene assay, RNA immunoprecipitation (RIP) assay and Chromatin Immunoprecipitation (ChIP) assay were performed to explore the interaction among XIST, EPHA1 and HNF4A. The effects of XIST on GG progression were evaluated by determining expression of proliferation- and invasion-related proteins (Ki67, PCNA, MMP-2, and MMP-9). Further, the functional role of XIST in GC with the involvement of NFκB pathway was also analyzed. Subsequently, the tumor growth in nude mice was evaluated. High expression of XIST and EPHA1 was observed in GC. XIST elevated EPHA1 expression by recruiting HNF4A. In addition, silencing of XIST inhibited GC progression in vitro and in vivo. Overexpressed XIST and EPHA1 yielded a reversed effect on cell proliferation and invasion. SN50 treatment (inhibitor of NFκB pathway) counteracted the promotive effect on GC cell proliferation and invasion mediated by XIST. The present study unveils that XIST increases the enrichment of HNF4A in the promoter region of EPHA1, thus promoting the deterioration of GC.


Subject(s)
Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Hepatocyte Nuclear Factor 4/metabolism , NF-kappa B/metabolism , RNA, Long Noncoding/genetics , Receptor, EphA1/metabolism , Stomach Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Disease Progression , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/biosynthesis , RNA, Long Noncoding/metabolism , Receptor, EphA1/genetics , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transcription, Genetic
5.
Mol Cell Endocrinol ; 513: 110867, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32422400

ABSTRACT

Hepatic nuclear factor 4 alpha (HNF4α) drives the expression of apolipoprotein B (ApoB), microsomal triglyceride transfer protein (MTP) and phospholipase A2 G12B (PLA2G12B), governing hepatic very-low-density lipoprotein (VLDL) production and secretion. Andrographolide (AP) is a major constituent isolated from Andrographis paniculata. We found that AP can disrupt the interaction between HNF4α and its coactivator peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α). Virtual docking and mutational analysis indicated that arginine 235 of HNF4α is essential for binding to AP. As a consequence of antagonizing the activity of HNF4α, AP suppresses the expression of ApoB, MTP and PLA2G12B and reduces the rate of hepatic VLDL secretion in vivo. AP additionally reduced gluconeogenesis via down-regulating the expression of HNF4α target genes phosphoenolpyruvate carboxykinase (Pepck) and glucose-6-phosphatase (G6pc). Collectively, our results suggest that AP affects liver function via modulating the transcriptional activity of HNF4α.


Subject(s)
Diterpenes/pharmacology , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Liver/drug effects , Animals , Cells, Cultured , Gluconeogenesis/drug effects , Gluconeogenesis/genetics , HEK293 Cells , Hep G2 Cells , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/antagonists & inhibitors , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Protein Binding/drug effects
6.
Pharmacol Res ; 157: 104840, 2020 07.
Article in English | MEDLINE | ID: mdl-32353589

ABSTRACT

Hepatocyte nuclear factor 4 alpha (HNF4α) regulates the expression of essential genes involved in very-low-density lipoprotein (VLDL) homeostasis and gluconeogenesis. 18ß-glycyrrhetinic acid (GA) is an active ingredient of Glycyrrhiza uralensis an herbal medicine used for treating liver aliments. In this study, we established that GA functions as a partial antagonist of HNF4α through HNF4α-driven reporter luciferase assay and co-immunoprecipitation experiments with co-activator PGC1α. By virtual docking and site-directed mutagenesis analysis, we confirmed that serine 190 and arginine 235 of HNF4α are both essential for GA to exert its antagonistic action on HNF4α. Importantly, GA suppressed the expression of HNF4α target genes such as apolipoprotein B (ApoB), microsomal triglyceride transfer protein (MTP) and phospholipase A2 G12B (PLA2G12B) modulating hepatic VLDL secretion in mice fed on a high fat diet. In addition, GA also suppressed gluconeogenesis and ameliorated glucose intolerance via down-regulating the expression of HNF4α target genes glucose-6-phosphatase (G6pc) and phosphoenolpyruvate carboxykinase (Pepck). Furthermore, GA significantly lowered blood glucose and improved insulin resistance in db/db mice. In all, we established that GA acts as a partial HNF4α antagonist modulating lipid and carbohydrate metabolism.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus/drug therapy , Glycyrrhetinic Acid/analogs & derivatives , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Hepatocytes/drug effects , Hypoglycemic Agents/pharmacology , Hypolipidemic Agents/pharmacology , Lipids/blood , Non-alcoholic Fatty Liver Disease/drug therapy , Animals , Blood Glucose/metabolism , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Disease Models, Animal , Gene Expression Regulation , Gluconeogenesis/drug effects , Glycyrrhetinic Acid/pharmacology , HEK293 Cells , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Insulin Resistance , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Signal Transduction
7.
Microbiol Immunol ; 64(5): 345-355, 2020 May.
Article in English | MEDLINE | ID: mdl-31981244

ABSTRACT

Chronic infection with hepatitis B virus (HBV) sometime induces lethal cirrhosis and hepatocellular carcinoma. Although nucleot(s)ide analogs are used as main treatment for HBV infection, the emergence of the drug-resistant viruses has become a problem. To discover novel antivirals with low side effects and low risk of emergence of resistant viruses, screening for anti-HBV compounds was performed with compound libraries of inhibitors targeting G-protein-coupled receptors (GPCRs). HepG2-hNTCP C4 cells infected with HBV were treated with various GPCR inhibitors and harvested at 14 day postinfection for quantification of core protein in the first screening or relaxed circular DNA in the second screening. Finally, we identified a cannabinoid receptor 1 inhibitor, rimonabant, as a candidate showing anti-HBV effect. In HepG2-hNTCP C4 cells, treatment with rimonabant suppressed HBV propagation at the viral RNA transcription step but had no effect on entry or covalently closed circular DNA level. The values of half maximal inhibitory concentration, half maximal effective concentration, and selectivity index of rimonabant in primary human hepatocyte (PHH) are 2.77 µm, 40.4 µm, and 14.6, respectively. Transcriptome analysis of rimonabant-treated primary hepatocytes by RNA sequencing revealed that the transcriptional activity of hepatocyte nuclear factor 4α (HNF4α), which is known to stimulate viral RNA synthesis, was depressed. By treatment of PHH with rimonabant, the expression level of HNF4α protein and the production of the messenger RNAs (mRNAs) of downstream factors promoted by HNF4α were reduced while the amount of HNF4α mRNA was not altered. These results suggest that treatment with rimonabant suppresses HBV propagation through the inhibition of HNF4α activity.


Subject(s)
Antiviral Agents/pharmacology , Hepatitis B virus/drug effects , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Rimonabant/pharmacology , Virus Replication/drug effects , Drug Discovery , Hep G2 Cells , Hepatitis B virus/physiology , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , RNA, Messenger/metabolism , RNA, Viral/metabolism
8.
Br J Cancer ; 120(5): 488-498, 2019 03.
Article in English | MEDLINE | ID: mdl-30792535

ABSTRACT

BACKGROUND: Gastric cancer (GC) is a highly heterogeneous disease with few "targeted" therapeutic options. Previously, we demonstrated involvement of the transcription factor HNF4α in human GC tumours, and the developmental signal mediator, WNT5A, as a prognostic GC biomarker. One previously developed HNF4α antagonist, BI6015, while not advancing beyond preclinical stages, remains useful for studying GC. METHODS: Here, we characterised the antineoplastic signalling activity of derivatives of BI6015, including transfer of the nitro group from the para position, relative to a methyl group on its benzene ring, to the ortho- and meta positions. We assessed binding efficacy, through surface plasmon resonance and docking studies, while biologic activity was assessed by antimitogenic efficacy against a panel of GC cell lines, and dysregulated transcriptomes, followed by pathway and subpathway analysis. RESULTS: The para derivative of BI6105 was found substantially more growth inhibitory, and effective, in downregulating numerous oncogenic signal pathways, including the embryonic cascade WNT. The ortho and meta derivatives, however, failed to downregulate WNT or other embryonic signalling pathways, unable to suppress GC growth. CONCLUSION: Straightforward strategies, employing bioinformatics analyses, to facilitate the effective design and development of "druggable" transcription factor inhibitors, are useful for targeting specific oncogenic signalling pathways, in GC and other cancers.


Subject(s)
Benzimidazoles/pharmacology , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Stomach Neoplasms/metabolism , Sulfonamides/pharmacology , Wnt Proteins/drug effects , Cell Line, Tumor , Drug Discovery , Humans , Molecular Docking Simulation , Signal Transduction , Substrate Specificity , Surface Plasmon Resonance , Wnt Proteins/metabolism
9.
J Lipid Res ; 60(4): 794-804, 2019 04.
Article in English | MEDLINE | ID: mdl-30709899

ABSTRACT

All-trans retinoic acid (atRA) is used to treat certain cancers and dermatologic diseases. A common adverse effect of atRA is hypercholesterolemia; cytochrome P450 (CYP) 7A repression is suggested as a driver. However, the underlying molecular mechanisms remain unclear. We investigated CYP7A1 expression in the presence of atRA in human hepatocytes and hepatic cell lines. In HepaRG cells, atRA increased cholesterol levels dose-dependently alongside dramatic decreases in CYP7A1 expression. Lentiviral-mediated CYP7A1 overexpression reversed atRA-induced cholesterol accumulation, suggesting that CYP7A1 repression mediated cholesterol accumulation. In CYP7A1 promoter reporter assays and gene-knockdown studies, altered binding of hepatocyte nuclear factor 4 α (HNF4α) to the proximal promoter was essential for atRA-mediated CYP7A1 repression. Pharmacologic inhibition of c-Jun N-terminal kinase (JNK) and ERK pathways attenuated atRA-mediated CYP7A1 repression and cholesterol accumulation. Overexpression of AP-1 (c-Jun/c-Fos), a downstream target of JNK and ERK, repressed CYP7A1 expression. In DNA pull-down and chromatin immunoprecipitation assays, AP-1 exhibited sequence-specific binding to the proximal CYP7A1 promoter region overlapping the HNF4α binding site, and atRA increased AP-1 but decreased HNF4α recruitment to the promoter. Collectively, these results indicate that atRA activates JNK and ERK pathways and the downstream target AP-1 represses HNF4α transactivation of the CYP7A1 promoter, potentially responsible for hypercholesterolemia.


Subject(s)
Cholesterol 7-alpha-Hydroxylase/metabolism , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Transcription Factor AP-1/metabolism , Tretinoin/pharmacology , Cells, Cultured , Cholesterol/analysis , Cholesterol/biosynthesis , Cholesterol 7-alpha-Hydroxylase/genetics , Dose-Response Relationship, Drug , Dyslipidemias , Hepatocyte Nuclear Factor 4/metabolism , Humans , Promoter Regions, Genetic/genetics
10.
J Gen Virol ; 99(5): 645-654, 2018 05.
Article in English | MEDLINE | ID: mdl-29561254

ABSTRACT

Hepatitis B virus (HBV) infection is a major health problem worldwide. Interleukin-35 (IL-35) is a definite immunosuppressive cytokine belonging to the IL-12 family. Nevertheless, the role of IL-35 in HBV replication remains elusive. In this study, we found that the level of HBV DNA replicative intermediates detected by qPCR and Southern blotting analysis was significantly increased by rhIL-35 in a dose-dependent manner. Moreover, HBV 3.5 kb mRNA levels were up-regulated by rhIL-35. The HBV core protein level as well as the HBsAg and HBeAg secretion levels were also increased by rhIL-35. Moreover, a mechanistic study demonstrated that IL-35 promoted HBV replication by enhancing the HBV core promoter activity. Importantly, hepatocyte nuclear factor 4α (HNF4α) was probably the target of IL-35. Mutation of the HNF4α-binding site on HBV core promoter or silencing HNF4α abolished the enhancement of HBV replication induced by IL-35. Finally, rhIL-35 was able to increase HBV replication in HBV transgenic mice. Taken together, our findings demonstrated that IL-35 has a novel role in HBV replication.


Subject(s)
Hepatitis B virus/drug effects , Hepatitis B virus/genetics , Hepatocyte Nuclear Factor 4/genetics , Interleukins/pharmacology , Virus Replication/drug effects , Animals , Gene Expression Regulation , Gene Silencing , Hep G2 Cells , Hepatitis B/immunology , Hepatitis B Surface Antigens/genetics , Hepatitis B e Antigens/genetics , Hepatitis B virus/physiology , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , RNA, Messenger , Recombinant Proteins/pharmacology , Signal Transduction , Up-Regulation
11.
Cell Physiol Biochem ; 41(6): 2419-2431, 2017.
Article in English | MEDLINE | ID: mdl-28467989

ABSTRACT

OBJECTIVE: Insulin resistance is a critical factor contributing to the pathogenesis of type 2 diabetes and other metabolic diseases. Recent studies have indicated that miR-338-3p plays an important role in cancer. Here, we investigated whether miR-338-3p mediates tumour necrosis factor-α (TNF-α)-induced hepatic insulin resistance. METHODS: The activation of the insulin signalling pathway and the level of glycogenesis were examined in the livers of the db/db and high fat diet (HFD)-fed mice and in HEP1-6 cells transfected with miR-338-3p mimic or inhibitor. Computational prediction of microRNA target, luciferase assay and Western blot were used to assess the miR-338-3p target. Chromatin immunoprecipitation (ChIP) assay was used to determine the transcriptional regulator of miR-338-3p. RESULTS: miR-338-3p was down-regulated in the livers of the db/db, HFD-fed and TNF-α-treated C57BL/6J mice, as well as in mouse HEP1-6 hepatocytes treated with TNF-α. Importantly the down-regulation of miR-338-3p induced insulin resistance, as indicated by impaired glucose tolerance and insulin tolerance. Further research showed that the down-regulated miR-338-3p resulted in the impaired AKT/ glycogen synthase kinase 3 beta (GSl·Gß) signalling pathway and glycogen synthesis. In contrast, hepatic over-expression of miR-338-3p rescued the TNF-α-induced insulin resistance. Moreover, protein phosphatase 4 regulator subunit 1 (PP4R1) was identified as a direct target of miR-338-3p that mediated hepatic insulin signalling by regulating protein phosphatase 4 (PP4). Finally we identified hepatic nuclear factor 4 alpha (HNF-4α) as the transcriptional regulator of miRNA-338-3p. CONCLUSIONS: Our studies provide novel insight into the critical role and molecular mechanism by which miR-338-3p is involved in TNF-α-induced hepatic insulin resistance. miR-338-3p might mediate TNF-α-induced hepatic insulin resistance by targeting PP4R1 to regulate PP4 expression.


Subject(s)
Gene Expression Regulation , Insulin Resistance , Liver/drug effects , MicroRNAs/metabolism , Phosphoprotein Phosphatases/genetics , Tumor Necrosis Factor-alpha/pharmacology , Animals , Base Sequence , Cell Line , Diet, High-Fat , Glycogen Synthase Kinase 3 beta/metabolism , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Insulin/pharmacology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Phosphoprotein Phosphatases/chemistry , Phosphoprotein Phosphatases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Signal Transduction
12.
Biosci Biotechnol Biochem ; 81(8): 1548-1552, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28475422

ABSTRACT

Hepatocyte nuclear factor 4α (HNF4α) is a nuclear receptor that regulates the expression of genes involved in a number of critical metabolic pathways. The modulation of HNF4α activity is thought to be a promising drug target pathway for hyperlipidemia. To identify compounds that reduce the activity of HNF4α, we conducted luciferase reporter assays using the promoter region of microsomal triglyceride transfer protein (MTP) gene, which contains an HNF4α-responsive element. Using this system, we show here that the flavonoid derivative 4'-nitro-6-hydroxyflavone (NOHF) suppresses MTP promoter activity. Treatment with NOHF caused a decrease in the expression of the HNF4α target gene. We also identified that NOHF triggers the AMP-activated protein kinase (AMPK) and accelerates the degradation of HNF4α protein. Knock-down of AMPK diminishes the effect of NOHF. These results indicate that NOHF is an AMPK activator and attenuates the transcriptional activity of HNF4α, at least in part, by accelerating HNF4α protein degradation.


Subject(s)
AMP-Activated Protein Kinases/genetics , Enzyme Inhibitors/pharmacology , Flavones/pharmacology , Flavonoids/pharmacology , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Hypolipidemic Agents/pharmacology , Nitro Compounds/pharmacology , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Enzyme Activation/drug effects , Gene Expression Regulation , Genes, Reporter , Hep G2 Cells , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Humans , Luciferases/genetics , Luciferases/metabolism , Promoter Regions, Genetic/drug effects , Proteolysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Response Elements , Signal Transduction
13.
Nucleic Acids Res ; 44(6): 2514-27, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26926107

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) is a complex multistep process in which phenotype switches are mediated by a network of transcription factors (TFs). Systematic characterization of all dynamic TFs controlling EMT state transitions, especially for the intermediate partial-EMT state, represents a highly relevant yet largely unexplored task. Here, we performed a computational analysis that integrated time-course EMT transcriptomic data with public cistromic data and identified three synergistic master TFs (ETS2, HNF4A and JUNB) that regulate the transition through the partial-EMT state. Overexpression of these regulators predicted a poor clinical outcome, and their elimination readily abolished TGF-ß-induced EMT. Importantly, these factors utilized a clique motif, physically interact and their cumulative binding generally characterized EMT-associated genes. Furthermore, analyses of H3K27ac ChIP-seq data revealed that ETS2, HNF4A and JUNB are associated with super-enhancers and the administration of BRD4 inhibitor readily abolished TGF-ß-induced EMT. These findings have implications for systematic discovery of master EMT regulators and super-enhancers as novel targets for controlling metastasis.


Subject(s)
Adenocarcinoma/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 4/genetics , Lung Neoplasms/genetics , Proto-Oncogene Protein c-ets-2/genetics , Transcription Factors/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Antineoplastic Agents/pharmacology , Azepines/pharmacology , Cell Cycle Proteins , Cell Line, Tumor , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Profiling , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Hepatocyte Nuclear Factor 4/metabolism , Histones/genetics , Histones/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phenotype , Proto-Oncogene Protein c-ets-2/antagonists & inhibitors , Proto-Oncogene Protein c-ets-2/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sequence Analysis, RNA , Signal Transduction , Smad3 Protein/genetics , Smad3 Protein/metabolism , Survival Analysis , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Transcriptome , Transforming Growth Factor beta/pharmacology , Triazoles/pharmacology
14.
Endocrinology ; 157(1): 207-19, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26496021

ABSTRACT

Exposure of humans to bisphenol A (BPA) is widespread and continuous. The effects of protracted exposure to BPA on the adult prostate have not been studied. We subjected Noble rats to 32 weeks of BPA (low or high dose) or 17ß-estradiol (E2) in conjunction with T replenishment. T treatment alone or untreated groups were used as controls. Circulating T levels were maintained within the physiological range in all treatment groups, whereas the levels of free BPA were elevated in the groups treated with T+low BPA (1.06 ± 0.05 ng/mL, P < .05) and T+high BPA (10.37 ± 0.43 ng/mL, P < .01) when compared with those in both controls (0.1 ± 0.05 ng/mL). Prostatic hyperplasia, low-grade prostatic intraepithelial neoplasia (PIN), and marked infiltration of CD4+ and CD8+ T cells into the PIN epithelium (P < .05) were observed in the lateral prostates (LPs) of T+low/high BPA-treated rats. In contrast, only hyperplasia and high-grade PIN, but no aberrant immune responses, were found in the T+E2-treated LPs. Genome-wide transcriptome analysis in LPs identified differential changes between T+BPA vs T+E2 treatment. Expression of multiple genes in the regulatory network controlled by hepatocyte nuclear factor 4α was perturbed by the T+BPA but not by the T+E2 exposure. Collectively these findings suggest that the adult rat prostate, under a physiologically relevant T environment, is susceptible to BPA-induced transcriptomic reprogramming, immune disruption, and aberrant growth dysregulation in a manner distinct from those caused by E2. They are more relevant to our recent report of higher urinary levels BPA found in patients with prostate cancer than those with benign disease.


Subject(s)
Benzhydryl Compounds/toxicity , Endocrine Disruptors/toxicity , Gene Expression Regulation, Neoplastic/drug effects , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Immunologic Surveillance/drug effects , Phenols/toxicity , Prostatic Intraepithelial Neoplasia/chemically induced , Prostatic Neoplasms/chemically induced , Animals , Benzhydryl Compounds/administration & dosage , Benzhydryl Compounds/blood , Dose-Response Relationship, Drug , Drug Implants , Endocrine Disruptors/administration & dosage , Endocrine Disruptors/blood , Estradiol/administration & dosage , Estradiol/pharmacology , Gene Expression Profiling , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Hormone Replacement Therapy , Lymphocyte Activation/drug effects , Male , Neoplasm Grading , Organ Size/drug effects , Phenols/administration & dosage , Phenols/blood , Prostate/drug effects , Prostate/immunology , Prostate/metabolism , Prostate/pathology , Prostatic Intraepithelial Neoplasia/immunology , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Rats, Inbred Strains , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Testosterone/administration & dosage , Testosterone/blood , Testosterone/pharmacology , Toxicokinetics
15.
Mol Pharmacol ; 88(4): 746-57, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26232425

ABSTRACT

Upon treatment with the pregnane X receptor (PXR) activator rifampicin (RIF), human hepatocellular carcinoma HepG2-derived ShP51 cells that stably express PXR showed epithelial-mesenchymal transition (EMT)-like morphological changes and migration. Our recent DNA microarrays have identified hepatocyte nuclear factor (HNF) 4α and insulin-like growth factor-binding protein (IGFBP) 1 mRNAs to be downregulated and upregulated, respectively, in RIF-treated ShP51 cells, and these regulations were confirmed by the subsequent real-time polymerase chain reaction and Western blot analyses. Using this cell system, we demonstrated here that the PXR-HNF4α-IGFBP1 pathway is an essential signal for PXR-induced morphological changes and migration. First, we characterized the molecular mechanism underlying the PXR-mediated repression of the HNF4α gene. Chromatin conformation capture and chromatin immunoprecipitation (ChIP) assays revealed that PXR activation by RIF disrupted enhancer-promoter communication and prompted deacetylation of histone H3 in the HNF4α P1 promoter. Cell-based reporter and ChIP assays showed that PXR targeted the distal enhancer of the HNF4α P1 promoter and stimulated dissociation of HNF3ß from the distal enhancer. Subsequently, small interfering RNA knockdown of HNF4α connected PXR-mediated gene regulation with the PXR-induced cellular responses, showing that the knockdown resulted in the upregulation of IGFBP1 and EMT-like morphological changes without RIF treatment. Moreover, recombinant IGFBP1 augmented migration, whereas an anti-IGFBP1 antibody attenuated both PXR-induced morphological changes and migration in ShP51 cells. PXR indirectly activated the IGFBP1 gene by repressing the HNF4α gene, thus enabling upregulation of IGFBP1 to change the morphology of ShP51 cells and cause migration. These results provide new insights into PXR-mediated cellular responses toward xenobiotics including therapeutics.


Subject(s)
Cell Movement/physiology , Cell Shape/physiology , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Hepatocyte Nuclear Factor 4/metabolism , Insulin-Like Growth Factor Binding Protein 1/biosynthesis , Receptors, Steroid/metabolism , Hep G2 Cells , Humans , Pregnane X Receptor
16.
Toxicology ; 333: 53-62, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-25868421

ABSTRACT

Perfluorooctanoic acid (PFOA) was shown to damage the liver of rodents and to impair embryonic development. At the molecular level, the hepatotoxic effects were attributed to the PFOA-mediated activation of peroxisome proliferator-activated receptor alpha (PPARα). In general, PPARα-dependent effects are less pronounced in humans than in rodents, and the hazard potential of PFOA for humans is controversially discussed. To analyse the effects of PFOA in human hepatocytes, a microarray analysis was conducted to screen for PFOA-mediated alterations in the transcriptome of human primary hepatocytes. A subsequent network analysis revealed that PFOA had an impact on several signalling pathways in addition to the well-known activation of PPARα. The microarray data confirmed earlier findings that PFOA: (i) affects the estrogen receptor ERα, (ii) activates the peroxisome proliferator-activated receptor gamma (PPARγ), and (iii) inhibits the function of the hepatocyte nuclear factor 4α (HNF4α) which is an essential factor for liver development and embryogenesis. Finally, as a novel finding, PFOA was shown to stimulate gene expression of the proto-oncogenes c-Jun and c-Fos. This was confirmed by using the HepG2 cell line as a model for human hepatocytes. PFOA stimulated cellular proliferation and the metabolic activity of the cells, and upregulated the expression of various cyclins which have a central function in the regulation of cell cycle control. Functional studies, however, indicated that PFOA had no impact on c-Jun and c-Fos phosphorylation and on AP-1-dependent gene transcription, thus demonstrating that PFOA-induced proliferation occurs largely independent of c-Jun and c-Fos.


Subject(s)
Caprylates/toxicity , Chemical and Drug Induced Liver Injury/etiology , Fluorocarbons/toxicity , Hepatocytes/drug effects , Signal Transduction/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Cyclins/genetics , Cyclins/metabolism , Dose-Response Relationship, Drug , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation , Gene Regulatory Networks , Hep G2 Cells , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Oligonucleotide Array Sequence Analysis , PPAR alpha/agonists , PPAR alpha/genetics , PPAR alpha/metabolism , PPAR gamma/agonists , PPAR gamma/genetics , PPAR gamma/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Time Factors , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcriptome
17.
Am J Physiol Renal Physiol ; 308(10): F1135-45, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25786776

ABSTRACT

The present study was carried out to investigate if hepatic nuclear factor (HNF)4α contributed to the high glucose-induced increase in stromal interacting molecule (STIM)1 protein abundance in glomerular mesangial cells (MCs). Western blot and immunofluorescence experiments showed HNF4α expression in MCs. Knockdown of HNF4α using a small interfering RNA approach significantly increased mRNA expression levels of both STIM1 and Orai1 and protein expression levels of STIM1 in cultured human MCs. Consistently, overexpression of HNF4α reduced expressed STIM1 protein expression in human embryonic kidney-293 cells. Furthermore, high glucose treatment did not significantly change the abundance of HNF4α protein in MCs but significantly attenuated HNF4α binding activity to the Stim1 promoter. Moreover, knockdown of HNF4α significantly augmented store-operated Ca(2+) entry, which is known to be gated by STIM1 and has recently been found to be antifibrotic in MCs. In agreement with those results, knockdown of HNF4α significantly attenuated the fibrotic response of high glucose. These results suggest that HNF4α negatively regulates STIM1 transcription in MCs. High glucose increases STIM1 expression levels by impairing HNF4α binding activity to the Stim1 promoter, which subsequently releases Stim1 transcription from HNF4α repression. Since the STIM1-gated store-operated Ca(2+) entry pathway in MCs has an antifibrotic effect, inhibition of HNF4α in MCs might be a potential therapeutic option for diabetic kidney disease.


Subject(s)
Glucose/pharmacology , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Hepatocyte Nuclear Factor 4/metabolism , Membrane Proteins/metabolism , Mesangial Cells/metabolism , Neoplasm Proteins/metabolism , Promoter Regions, Genetic/physiology , Up-Regulation/drug effects , Calcium/metabolism , Calcium Channels/metabolism , Cells, Cultured , Collagen Type IV/metabolism , Diabetic Nephropathies/metabolism , Dose-Response Relationship, Drug , Fibronectins/metabolism , HEK293 Cells , Hepatocyte Nuclear Factor 4/drug effects , Humans , In Vitro Techniques , Mesangial Cells/cytology , Mesangial Cells/drug effects , ORAI1 Protein , Protein Binding/drug effects , RNA, Small Interfering/pharmacology , Stromal Interaction Molecule 1 , Up-Regulation/physiology
18.
Cancer Discov ; 4(9): OF9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25185208
19.
Mol Cell Biol ; 34(17): 3291-304, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24980432

ABSTRACT

Transcriptional regulatory mechanisms likely contribute to the etiology of inflammatory bowel disease (IBD), as genetic variants associated with the disease are disproportionately found at regulatory elements. However, the transcription factors regulating colonic inflammation are unclear. To identify these transcription factors, we mapped epigenomic changes in the colonic epithelium upon inflammation. Epigenetic marks at transcriptional regulatory elements responded dynamically to inflammation and indicated a shift in epithelial transcriptional factor networks. Active enhancer chromatin structure at regulatory regions bound by the transcription factor hepatocyte nuclear factor 4α (HNF4A) was reduced during colitis. In agreement, upon an inflammatory stimulus, HNF4A was downregulated and showed a reduced ability to bind chromatin. Genetic variants that confer a predisposition to IBD map to HNF4A binding sites in the human colon cell line CaCo2, suggesting impaired HNF4A binding could underlie genetic susceptibility to IBD. Despite reduced HNF4A binding during inflammation, a temporal knockout model revealed HNF4A still actively protects against inflammatory phenotypes and promotes immune regulatory gene expression in the inflamed colonic epithelium. These findings highlight the potential for HNF4A agonists as IBD therapeutics.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , Colitis/genetics , Colitis/metabolism , Gene Regulatory Networks , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Animals , Binding Sites/genetics , Caco-2 Cells , Colitis/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Epigenesis, Genetic , Female , Gene Knockout Techniques , Genetic Predisposition to Disease , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Regulatory Elements, Transcriptional
20.
Nature ; 513(7516): 110-4, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25043045

ABSTRACT

Mutations in isocitrate dehydrogenase 1 (IDH1) and IDH2 are among the most common genetic alterations in intrahepatic cholangiocarcinoma (IHCC), a deadly liver cancer. Mutant IDH proteins in IHCC and other malignancies acquire an abnormal enzymatic activity allowing them to convert α-ketoglutarate (αKG) to 2-hydroxyglutarate (2HG), which inhibits the activity of multiple αKG-dependent dioxygenases, and results in alterations in cell differentiation, survival, and extracellular matrix maturation. However, the molecular pathways by which IDH mutations lead to tumour formation remain unclear. Here we show that mutant IDH blocks liver progenitor cells from undergoing hepatocyte differentiation through the production of 2HG and suppression of HNF-4α, a master regulator of hepatocyte identity and quiescence. Correspondingly, genetically engineered mouse models expressing mutant IDH in the adult liver show an aberrant response to hepatic injury, characterized by HNF-4α silencing, impaired hepatocyte differentiation, and markedly elevated levels of cell proliferation. Moreover, IDH and Kras mutations, genetic alterations that co-exist in a subset of human IHCCs, cooperate to drive the expansion of liver progenitor cells, development of premalignant biliary lesions, and progression to metastatic IHCC. These studies provide a functional link between IDH mutations, hepatic cell fate, and IHCC pathogenesis, and present a novel genetically engineered mouse model of IDH-driven malignancy.


Subject(s)
Bile Duct Neoplasms/pathology , Cell Differentiation/genetics , Cholangiocarcinoma/pathology , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Hepatocytes/pathology , Isocitrate Dehydrogenase/genetics , Mutant Proteins/metabolism , Animals , Bile Duct Neoplasms/enzymology , Bile Duct Neoplasms/genetics , Bile Ducts, Intrahepatic/enzymology , Bile Ducts, Intrahepatic/pathology , Cell Division/genetics , Cell Lineage/genetics , Cholangiocarcinoma/enzymology , Cholangiocarcinoma/genetics , Disease Models, Animal , Female , Glutarates/metabolism , Hepatocyte Nuclear Factor 4/biosynthesis , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/enzymology , Hepatocytes/metabolism , Humans , Isocitrate Dehydrogenase/metabolism , Male , Mice , Mice, Transgenic , Mutant Proteins/genetics , Mutation/genetics , Neoplasm Metastasis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras) , Stem Cells/pathology , ras Proteins/genetics , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...