Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Biol Toxicol ; 37(1): 65-84, 2021 02.
Article in English | MEDLINE | ID: mdl-32623698

ABSTRACT

Bisphenol A (BPA) is an endocrine-disrupting chemical (EDC) associated with non-alcoholic fatty liver disease (NAFLD). The effects of gestational BPA exposure on hepatic lipid accumulation in offspring are not fully understood. Here, we investigate the sex-dependent effects of gestational BPA exposure on hepatic lipid and glucose metabolism in the offspring of mice to reveal the mechanisms underlying gestational BPA exposure-associated NAFLD. Pregnant mice were administered gavage with or without 1 µg kg-1 day-1 BPA at embryonic day 7.5 (E7.5)-E16.5. Hepatic glucose and lipid metabolism were evaluated in these models. Both male and female offspring mice exhibited hepatic fatty liver after BPA treatment. Lipid accumulation and dysfunction of glucose metabolism were observed in male offspring. We revealed abnormal expression of lipid regulators in the liver and that inhibition of peroxisome proliferator-activated receptor γ (PPARγ) repressed hepatic lipid accumulation induced by gestational BPA exposure. We also found a sex-dependent decrease of hepatocyte nuclear factor 1b (HNF1b) expression in male offspring. The transcriptional repression of PPARγ by HNF1b was confirmed in L02 cells. Downregulation of HNF1b, upregulation of PPARγ, and subsequent upregulation of hepatic lipid accumulation were essential for NAFLD development in male offspring gestationally exposed to BPA as well as BPA-exposed adult male mice. Dysregulation of the HNF1b/PPARγ pathway may be involved in gestational BPA exposure-induced NAFLD in male offspring. These data provide new insights into the mechanism of gestational BPA exposure-associated sex-dependent glucose and lipid metabolic dysfunction. Graphical abstract Schematic of the mechanism of gestational BPA exposure-induced glucose and lipid metabolic dysfunction.


Subject(s)
Benzhydryl Compounds/toxicity , Fatty Liver/chemically induced , Hepatocyte Nuclear Factor 1-beta/antagonists & inhibitors , PPAR gamma/metabolism , Phenols/toxicity , Prenatal Exposure Delayed Effects/pathology , Up-Regulation , Animals , Down-Regulation/drug effects , Estrogens/metabolism , Female , Gene Expression Regulation/drug effects , Glucose/metabolism , Hepatocyte Nuclear Factor 1-beta/metabolism , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Liver/drug effects , Liver/pathology , Liver/ultrastructure , Male , Mice, Inbred C57BL , Pregnancy , Transcription, Genetic/drug effects , Triglycerides/metabolism , Up-Regulation/drug effects
2.
J Am Soc Nephrol ; 28(11): 3205-3217, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28739648

ABSTRACT

AKI is a frequent condition that involves renal microcirculation impairment, infiltration of inflammatory cells with local production of proinflammatory cytokines, and subsequent epithelial disorders and mitochondrial dysfunction. Peroxisome proliferator-activated receptor γ coactivator 1-α (PPARGC1A), a coactivator of the transcription factor PPAR-γ that controls mitochondrial biogenesis and function, has a pivotal role in the early dysfunction of the proximal tubule and the subsequent renal repair. Here, we evaluated the potential role of hepatocyte nuclear factor-1ß (HNF-1ß) in regulating PPARGC1A expression in AKI. In mice, endotoxin injection to induce AKI also induced early and transient inflammation and PPARGC1A inhibition, which overlapped with downregulation of the HNF-1ß transcriptional network. In vitro, exposure of proximal tubule cells to the inflammatory cytokines IFN-γ and TNF-α led to inhibition of HNF-1ß transcriptional activity. Moreover, inhibition of HNF-1ß significantly reduced PPARGC1A expression and altered mitochondrial morphology and respiration in proximal tubule cells. Chromatin immunoprecipitation assays and PCR analysis confirmed HNF-1ß binding to the Ppargc1a promoter in mouse kidneys. We also demonstrated downregulation of renal PPARGC1A expression in a patient with an HNF1B germinal mutation. Thus, we propose that HNF-1ß links extracellular inflammatory signals to mitochondrial dysfunction during AKI partly via PPARGC1A signaling. Our findings further strengthen the view of HNF1B-related nephropathy as a mitochondrial disorder in adulthood.


Subject(s)
Acute Kidney Injury/metabolism , Hepatocyte Nuclear Factor 1-beta/physiology , Kidney Tubules, Proximal/metabolism , Mitochondria/metabolism , Acute Kidney Injury/etiology , Adult , Animals , Hepatocyte Nuclear Factor 1-beta/antagonists & inhibitors , Hepatocyte Nuclear Factor 1-beta/genetics , Humans , Mice, Inbred C57BL , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology
3.
Int J Gynecol Cancer ; 24(5): 838-43, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24804869

ABSTRACT

OBJECTIVE: Appropriate cell cycle checkpoints are essential for the maintenance of normal cells and chemosensitivity of cancer cells. Clear cell adenocarcinoma (CCA) of the ovary is highly resistant to chemotherapy. Hepatocyte nuclear factor-1ß (HNF-1ß) is known to be overexpressed in CCA, but its role and clinical significance is unclear. We investigated the role of HNF-1ß in regulation of the cell cycle in CCA. METHODS: To clarify the effects of HNF-1ß on cell cycle checkpoints, we compared the cell cycle distribution and the expression of key proteins involved in CCA cells in which HNF-1ß had been stably knocked down and in vector-control cell lines after treatment with bleomycin. HNF-1ß (+) cells were arrested in G2 phase because of DNA damage. RESULTS: HNF-1ß (-) cells died because of a checkpoint mechanism. G2 arrest of HNF-1ß (+) cells resulted from sustained CHK1 activation, a protein that plays a major role in the checkpoint mechanism. HNF-1ß (+) cells were treated with a CHK1 inhibitor after bleomycin treatment. Flow cytometric analysis of the cell cycle demonstrated that DNA damage-induced G2-arrested cells were released from the checkpoint and killed by a CHK1 inhibitor. CONCLUSIONS: The chemoresistance of CCA may be due to aberrant retention of the G2 checkpoint through overexpression of HNF-1ß. This is the first study demonstrating cell cycle regulation and chemosensitization by a CHK1 inhibitor in CCA.


Subject(s)
Apoptosis , Cell Cycle Checkpoints , DNA Damage , Drug Resistance, Neoplasm , G2 Phase , Hepatocyte Nuclear Factor 1-beta/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Protein Kinases/metabolism , Adenocarcinoma, Clear Cell/metabolism , Adenocarcinoma, Clear Cell/pathology , Blotting, Western , Cell Proliferation , Checkpoint Kinase 1 , Female , Flow Cytometry , Hepatocyte Nuclear Factor 1-beta/antagonists & inhibitors , Hepatocyte Nuclear Factor 1-beta/genetics , Humans , RNA, Small Interfering/genetics , Tumor Cells, Cultured
4.
Cell Tissue Res ; 357(1): 173-84, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24737489

ABSTRACT

In a previous study, the Notch pathway inhibited with N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (also called DAPT) was shown to promote the differentiation of fetal liver stem/progenitor cells (FLSPCs) into hepatocytes and to impair cholangiocyte differentiation. The precise mechanism for this, however, was not elucidated. Two mechanisms are possible: Notch inhibition might directly up-regulate hepatocyte differentiation via HGF (hepatocyte growth factor) and HNF (hepatocyte nuclear factor)-4α or might impair cholangiocyte differentiation thereby indirectly rendering hepatocyte differentiation as the dominant state. In this study, HGF and HNF expression was detected after the Notch pathway was inhibited. Although our initial investigation indicated that the inhibition of Notch induced hepatocyte differentiation with an efficiency similar to the induction via HGF, the results of this study demonstrate that Notch inhibition does not induce significant up-regulation of HGF or HNF-4α in FLSPCs. This suggests that Notch inhibition induces hepatocyte differentiation without the influence of HGF or HNF-4α. Moreover, significant down-regulation of HNF-1ß was observed, presumably dependent on an impairment of cholangiocyte differentiation. To confirm this presumption, HNF-1ß was blocked in FLSPCs and was followed by hepatocyte differentiation. The expression of markers of mature cholangiocyte was impaired and hepatocyte markers were elevated significantly. The data thus demonstrate that the inhibition of cholangiocyte differentiation spontaneously induces hepatocyte differentiation and further suggest that hepatocyte differentiation from FLSPCs occurs at the expense of the impairment of cholangiocyte differentiation, probably being enhanced partially via HNF-1ß down-regulation or Notch inhibition.


Subject(s)
Embryonic Stem Cells/cytology , Hepatocyte Nuclear Factor 1-beta/antagonists & inhibitors , Hepatocytes/cytology , Liver/cytology , Liver/embryology , Receptors, Notch/antagonists & inhibitors , Animals , Cell Culture Techniques , Cell Differentiation/physiology , Dipeptides/pharmacology , Rats , Rats, Inbred F344 , Signal Transduction , Transfection
5.
PLoS One ; 8(5): e63585, 2013.
Article in English | MEDLINE | ID: mdl-23704921

ABSTRACT

Epithelial repair following acute kidney injury (AKI) requires epithelial-mesenchyme-epithelial cycling associated with transient re-expression of genes normally expressed during kidney development as well as activation of growth factors and cytokine-induced signaling. In normal kidney, the Hnf-1ß transcription factor drives nephrogenesis, tubulogenesis and epithelial homeostasis through the regulation of epithelial planar cell polarity and expression of developmental or tubular segment-specific genes. In a mouse model of ischemic AKI induced by a 2-hours hemorrhagic shock, we show that expression of this factor is tightly regulated in the early phase of renal repair with a biphasic expression profile (early down-regulation followed by transient over-expression). These changes are associated to tubular epithelial differentiation as assessed by KSP-cadherin and megalin-cubilin endocytic complex expression analysis. In addition, early decrease in Hnf1b expression is associated with the transient over-expression of one of its main target genes, the suppressor of cytokine signaling Socs3, which has been shown essential for renal repair. In vitro, hypoxia induced early up-regulation of Hnf-1ß from 1 to 24 hours, independently of the hypoxia-inducible factor Hif-1α. When prolonged, hypoxia induced Hnf-1ß down-regulation while normoxia led to Hnf-1ß normalization. Last, Hnf-1ß down-regulation using RNA interference in HK-2 cells led to phenotype switch from an epithelial to a mesenchyme state. Taken together, we showed that Hnf-1ß may drive recovery from ischemic AKI by regulating both the expression of genes important for homeostasis control during organ repair and the state of epithelial cell differentiation.


Subject(s)
Epithelial Cells/metabolism , Epithelial Cells/pathology , Hepatocyte Nuclear Factor 1-beta/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney/metabolism , Kidney/pathology , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Animals , Biomarkers/metabolism , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cell Line , Disease Models, Animal , Endocytosis/drug effects , Epithelial Cells/drug effects , Female , Gene Expression Regulation/drug effects , Hepatocyte Nuclear Factor 1-beta/antagonists & inhibitors , Hepatocyte Nuclear Factor 1-beta/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kidney/drug effects , Kidney/physiopathology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/physiopathology , Mice , Mice, Inbred C57BL , Prolyl-Hydroxylase Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Shock, Hemorrhagic/complications , Shock, Hemorrhagic/pathology , Shock, Hemorrhagic/physiopathology , Wound Healing/drug effects
6.
Cancer Res ; 69(11): 4843-50, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19487294

ABSTRACT

We recently identified HSulf-1 as a down-regulated gene in ovarian carcinomas. Our previous analysis indicated that HSulf-1 inactivation in ovarian cancers is partly mediated by loss of heterozygosity and epigenetic silencing. Here, we show that variant hepatic nuclear factor 1 (vHNF1), encoded by transcription factor 2 gene (TCF2, HNF1beta), negatively regulates HSulf-1 expression in ovarian cancer. Immunoblot assay revealed that vHNF1 is highly expressed in HSulf-1-deficient OV207, SKOV3, and TOV-21G cell lines but not in HSulf-1-expressing OSE, OV167, and OV202 cells. By short hairpin RNA-mediated down-regulation of vHNF1 in TOV-21G cells and transient enhanced vHNF1 expression in OV202 cells, we showed that vHNF1 suppresses HSulf-1 expression in ovarian cancer cell lines. Reporter assay and chromatin immunoprecipitation experiments showed that vHNF1 is specifically recruited to HSulf-1 promoter at two different vHNF1-responsive elements in OV207 and TOV-21G cells. Additionally, down-regulation of vHNF1 expression in OV207 and TOV-21G cells increased cisplatin- or paclitaxel-mediated cytotoxicity as determined by both 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and clonogenic assays and this effect was reversed by down-regulation of HSulf-1. Moreover, nude mice bearing TOV-21G cell xenografts with stably down-regulated vHNF1 were more sensitive to cisplatin- or paclitaxel-induced cytotoxicity compared with xenografts of TOV-21G clonal lines with nontargeted control short hairpin RNA. Finally, immunohistochemical analysis of 501 ovarian tumors including 140 clear-cell tumors on tissue microarrays showed that vHNF1 inversely correlates to HSulf-1 expression. Collectively, these results indicate that vHNF1 acts as a repressor of HSulf-1 expression and might be a molecular target for ovarian cancer therapy.


Subject(s)
Carcinoma/genetics , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 1-beta/physiology , Ovarian Neoplasms/genetics , Sulfotransferases/genetics , Animals , Carcinoma/pathology , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Hepatocyte Nuclear Factor 1-beta/antagonists & inhibitors , Hepatocyte Nuclear Factor 1-beta/genetics , Hepatocyte Nuclear Factor 1-beta/metabolism , Humans , Mice , Mice, Nude , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/pathology , Protein Binding , RNA, Small Interfering/pharmacology , Response Elements , Sulfotransferases/physiology , Transfection , Xenograft Model Antitumor Assays
7.
Surgery ; 144(2): 168-73, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18656622

ABSTRACT

BACKGROUND: The sodium glucose cotransporter (SGLT1) is responsible for all active intestinal glucose uptake. Hepatocyte nuclear factors 1 alpha and beta (HNF 1 alpha and HNF 1 beta) activate the SGLT1 promoter, whereas GATA-binding protein 5 (GATA-5) and caudal-type homeobox protein 2 (CDX2) regulate transcription of other intestinal genes. We investigated SGLT1 regulation by these transcription factors using promoter studies and RNA interference. METHODS: Chinese hamster ovary (CHO) cells were transiently cotransfected with an SGLT1-luciferase promoter construct and combinations of expression vectors for HNF 1 alpha, HNF 1 beta, CDX2, and GATA-5. Caco-2 cells were stably transfected with knockdown vectors for either HNF 1 alpha or HNF 1 beta. mRNA levels of HNF 1 alpha, HNF 1 beta, and SGLT1 were determined using quantitative polymerase chain reaction (qPCR). RESULTS: HNF 1 alpha, GATA-5, and HNF 1 beta significantly activated the SGLT1 promoter (P < .05). Cotransfection of GATA-5 with HNF 1 alpha had an additive effect, whereas HNF 1 beta and CDX2 antagonized HNF 1 alpha and GATA-5. SGLT1 expression was significantly reduced in HNF 1 alpha or HNF 1 beta knockdowns (P < .001). HNF alpha knockdown significantly reduced HNF 1 beta expression and vice versa (P < .005). CONCLUSIONS: HNF 1 alpha and HNF 1 beta are important transcription factors for endogenous SGLT1 expression by cultured enterocytes. GATA-5 and CDX2 also regulate SGLT1 promoter activity and show cooperativity with the HNF1s. We, therefore, propose a multifactorial model for SGLT1 regulation, with interactions between HNF1, GATA-5, and CDX2 modulating intestinal glucose absorption.


Subject(s)
Gene Silencing , RNA Interference , Sodium-Glucose Transporter 1/metabolism , Transcription, Genetic , Animals , CDX2 Transcription Factor , CHO Cells , Caco-2 Cells , Cricetinae , Cricetulus , GATA5 Transcription Factor/antagonists & inhibitors , GATA5 Transcription Factor/pharmacology , Gene Expression Regulation , Hepatocyte Nuclear Factor 1-alpha/metabolism , Hepatocyte Nuclear Factor 1-alpha/pharmacology , Hepatocyte Nuclear Factor 1-beta/antagonists & inhibitors , Hepatocyte Nuclear Factor 1-beta/metabolism , Hepatocyte Nuclear Factor 1-beta/pharmacology , Homeodomain Proteins/pharmacology , Humans , Promoter Regions, Genetic/drug effects , Sodium-Glucose Transporter 1/genetics , Transfection
8.
J Biol Chem ; 282(51): 36837-44, 2007 Dec 21.
Article in English | MEDLINE | ID: mdl-17928287

ABSTRACT

Inducible nitric-oxide synthase (iNOS) plays a central role in the regulation of vascular function and response to injury. A central mediator controlling iNOS expression is transforming growth factor-beta (TGF-beta), which represses its expression through a mechanism that is poorly understood. We have identified a binding site in the iNOS promoter that interacts with the nuclear heterodimer TCF11/MafG using chromatin immunoprecipitation and mutation analyses. We demonstrate that binding at this site acts to repress the induction of iNOS gene expression by cytokines. We show that this repressor is induced by TGF-beta1 and by Smad6-short, which enhances TGF-beta signaling. In contrast, the up-regulation of TCF11/MafG binding could be suppressed by overexpression of the TGF-beta inhibitor Smad7, and a small interfering RNA to TCF11 blocked the suppression of iNOS by TGF-beta. The binding of TCF11/MafG to the iNOS promoter could be enhanced by phorbol 12-myristate 13-acetate and suppressed by the protein kinase C inhibitor staurosporine. Moreover, the induction of TCF11/MafG binding by TGF-beta and Smad6-short could be blocked by staurosporine, and the effect of TGF-beta was blocked by the selective protein kinase C inhibitor calphostin C. Consistent with the in vitro data, we found suppression of TCF11 coincident with iNOS up-regulation in a rat model of endotoxemia, and we observed a highly significant negative correlation between TCF11 and nitric oxide production. Furthermore, treatment with activated protein C, a serine protease effective in septic shock, blocked the down-regulation of TCF11 and suppressed endotoxin-induced iNOS. Overall, our results demonstrate a novel mechanism by which iNOS expression is regulated in the context of inflammatory activation.


Subject(s)
Gene Expression Regulation, Enzymologic , Hepatocyte Nuclear Factor 1-beta/metabolism , Nitric Oxide Synthase Type II/biosynthesis , Response Elements , Transforming Growth Factor beta1/metabolism , Animals , Carcinogens/pharmacology , Cells, Cultured , Dimerization , Disease Models, Animal , Endotoxemia/enzymology , Endotoxemia/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Hepatocyte Nuclear Factor 1-beta/antagonists & inhibitors , Hepatocyte Nuclear Factor 1-beta/genetics , Humans , Inflammation/enzymology , Inflammation/genetics , MafG Transcription Factor/genetics , MafG Transcription Factor/metabolism , Male , Mutation , Naphthalenes/pharmacology , Nitric Oxide Synthase Type II/genetics , Protein Kinase C/antagonists & inhibitors , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Repressor Proteins/genetics , Repressor Proteins/metabolism , Response Elements/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Smad6 Protein/genetics , Smad6 Protein/metabolism , Smad7 Protein/genetics , Smad7 Protein/metabolism , Staurosporine/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Transforming Growth Factor beta1/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...