Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 295(51): 17560-17572, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33453998

ABSTRACT

Hepatocyte nuclear factor-1ß (HNF-1ß) is a tissue-specific transcription factor that is required for normal kidney development and renal epithelial differentiation. Mutations of HNF-1ß produce congenital kidney abnormalities and inherited renal tubulopathies. Here, we show that ablation of HNF-1ß in mIMCD3 renal epithelial cells results in activation of ß-catenin and increased expression of lymphoid enhancer-binding factor 1 (LEF1), a downstream effector in the canonical Wnt signaling pathway. Increased expression and nuclear localization of LEF1 are also observed in cystic kidneys from Hnf1b mutant mice. Expression of dominant-negative mutant HNF-1ß in mIMCD3 cells produces hyperresponsiveness to exogenous Wnt ligands, which is inhibited by siRNA-mediated knockdown of Lef1. WT HNF-1ß binds to two evolutionarily conserved sites located 94 and 30 kb from the mouse Lef1 promoter. Ablation of HNF-1ß decreases H3K27 trimethylation repressive marks and increases ß-catenin occupancy at a site 4 kb upstream to Lef1. Mechanistically, WT HNF-1ß recruits the polycomb-repressive complex 2 that catalyzes H3K27 trimethylation. Deletion of the ß-catenin-binding domain of LEF1 in HNF-1ß-deficient cells abolishes the increase in Lef1 transcription and decreases the expression of downstream Wnt target genes. The canonical Wnt target gene, Axin2, is also a direct transcriptional target of HNF-1ß through binding to negative regulatory elements in the gene promoter. These findings demonstrate that HNF-1ß regulates canonical Wnt target genes through long-range effects on histone methylation at Wnt enhancers and reveal a new mode of active transcriptional repression by HNF-1ß.


Subject(s)
Hepatocyte Nuclear Factor 1-beta/metabolism , Lymphoid Enhancer-Binding Factor 1/metabolism , Wnt Signaling Pathway , Animals , Axin Protein/genetics , Axin Protein/metabolism , Binding Sites , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation , Hepatocyte Nuclear Factor 1-beta/deficiency , Hepatocyte Nuclear Factor 1-beta/genetics , Histones/metabolism , Kidney/cytology , Lymphoid Enhancer-Binding Factor 1/antagonists & inhibitors , Lymphoid Enhancer-Binding Factor 1/genetics , Methylation , Mice , Mice, Knockout , Mutagenesis , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/metabolism , Regulatory Elements, Transcriptional/genetics , Wnt3A Protein/metabolism , beta Catenin/metabolism
2.
Ital J Pediatr ; 45(1): 27, 2019 Feb 21.
Article in English | MEDLINE | ID: mdl-30791938

ABSTRACT

BACKGROUND: paucity of interlobular bile ducts is an important observation at liver biopsy in the diagnostic work-up of neonatal cholestasis. To date, other than in the Alagille syndrome, syndromic paucity of interlobular bile ducts has been documented in four cholestatic neonates with HFN1ß mutations. A syndromic phenotype, known as renal cysts and diabetes syndrome (RCAD), has been identified. This is usually characterized by a wide clinical spectrum, including renal cysts, maturity-onset diabetes of the young, exocrine pancreatic insufficiency, urogenital abnormalities and a not well established liver involvement. Herein we report a novel case of paucity of interlobular bile ducts due to an HFN1ß defect. CASE PRESENTATION: A 5-week-old boy was admitted to our department for cholestatic jaundice with increased gamma-glutamyl transpeptidase and an unremarkable clinical examination. He had been delivered by Caesarian section at 38 weeks' gestation from unrelated parents, with a birth weight of 2600 g (3rd percentile). Screening for cholestatic diseases, including Alagille syndrome, was negative except for a minor pulmonary artery stenosis at echocardiography and a doubt of a thoracic butterfly hemivertebra. The finding of hyperechogenic kidneys with multiple bilateral cortical cysts at ultrasound examination, associated with moderately impaired renal function with proteinuria, polyuria and metabolic acidosis, was suggestive of ciliopathy. A liver biopsy was performed revealing paucity of interlobular bile ducts, thus the diagnosis of Alagille syndrome was reconsidered. Although genetic tests for liver cholestatic diseases were performed with negative results for Alagille syndrome (JAG1 and NOTCH2), a de-novo missense mutation of HNF1ß gene was detected. At 18 months of age our patient has persistent cholestasis and his itching is not under satisfactory control. CONCLUSIONS: Alagille syndrome may not be the only syndrome determining paucity of interlobular bile ducts in neonates presenting with cholestasis and renal impairment, especially in small for gestational age newborns. We suggest that HNF1ß deficiency should also be ruled out, taking into consideration HNF1ß mutations, together with Alagille syndrome, in next generation sequencing strategies in neonates with cholestasis, renal impairment and/or paucity of interlobular bile ducts at liver biopsy.


Subject(s)
Alagille Syndrome/diagnosis , Cholestasis, Intrahepatic/etiology , Hepatocyte Nuclear Factor 1-beta/deficiency , Alagille Syndrome/complications , Humans , Infant , Male
3.
J Am Soc Nephrol ; 29(10): 2493-2509, 2018 10.
Article in English | MEDLINE | ID: mdl-30097458

ABSTRACT

BACKGROUND: Mutation of HNF1B, the gene encoding transcription factor HNF-1ß, is one cause of autosomal dominant tubulointerstitial kidney disease, a syndrome characterized by tubular cysts, renal fibrosis, and progressive decline in renal function. HNF-1ß has also been implicated in epithelial-mesenchymal transition (EMT) pathways, and sustained EMT is associated with tissue fibrosis. The mechanism whereby mutated HNF1B leads to tubulointerstitial fibrosis is not known. METHODS: To explore the mechanism of fibrosis, we created HNF-1ß-deficient mIMCD3 renal epithelial cells, used RNA-sequencing analysis to reveal differentially expressed genes in wild-type and HNF-1ß-deficient mIMCD3 cells, and performed cell lineage analysis in HNF-1ß mutant mice. RESULTS: The HNF-1ß-deficient cells exhibited properties characteristic of mesenchymal cells such as fibroblasts, including spindle-shaped morphology, loss of contact inhibition, and increased cell migration. These cells also showed upregulation of fibrosis and EMT pathways, including upregulation of Twist2, Snail1, Snail2, and Zeb2, which are key EMT transcription factors. Mechanistically, HNF-1ß directly represses Twist2, and ablation of Twist2 partially rescued the fibroblastic phenotype of HNF-1ß mutant cells. Kidneys from HNF-1ß mutant mice showed increased expression of Twist2 and its downstream target Snai2. Cell lineage analysis indicated that HNF-1ß mutant epithelial cells do not transdifferentiate into kidney myofibroblasts. Rather, HNF-1ß mutant epithelial cells secrete high levels of TGF-ß ligands that activate downstream Smad transcription factors in renal interstitial cells. CONCLUSIONS: Ablation of HNF-1ß in renal epithelial cells leads to the activation of a Twist2-dependent transcriptional network that induces EMT and aberrant TGF-ß signaling, resulting in renal fibrosis through a cell-nonautonomous mechanism.


Subject(s)
Gout/genetics , Gout/pathology , Hepatocyte Nuclear Factor 1-beta/genetics , Hyperuricemia/genetics , Hyperuricemia/pathology , Kidney Diseases/genetics , Kidney Diseases/pathology , Animals , Cell Line , Cell Lineage/genetics , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Female , Fibrosis , Genes, Dominant , Gout/metabolism , Hepatocyte Nuclear Factor 1-beta/deficiency , Hepatocyte Nuclear Factor 1-beta/metabolism , Humans , Hyperuricemia/metabolism , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Male , Mice , Mice, Transgenic , Mutation , Repressor Proteins/deficiency , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Twist-Related Protein 1/deficiency , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
4.
Am J Physiol Renal Physiol ; 315(1): F27-F35, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29561186

ABSTRACT

Mutations in hepatocyte nuclear factor 1ß (HNF1ß) cause autosomal dominant tubulointerstitial kidney disease (ADTKD-HNF1ß), and patients tend to develop renal cysts, maturity-onset diabetes of the young (MODY), and suffer from electrolyte disturbances, including hypomagnesemia, hypokalemia, and hypocalciuria. Previous HNF1ß research focused on the renal distal convoluted tubule (DCT) to elucidate the ADTKD-HNF1ß electrolyte phenotype, although 70% of Mg2+ is reabsorbed in the thick ascending limb of Henle's loop (TAL). An important regulator of Mg2+ reabsorption in the TAL is the calcium-sensing receptor (CaSR). This study used several methods to elucidate the role of HNF1ß in electrolyte reabsorption in the TAL. HNF1ß ChIP-seq data revealed a conserved HNF1ß binding site in the second intron of the CaSR gene. Luciferase-promoter assays displayed a 5.8-fold increase in CaSR expression when HNF1ß was present. Expression of the HNF1ß p.Lys156Glu mutant, which prevents DNA binding, abolished CaSR expression. Hnf1ß knockdown in an immortalized mouse kidney TAL cell line (MKTAL) reduced expression of the CaSR and Cldn14 (claudin 14) by 56% and 48%, respectively, while Cldn10b expression was upregulated 5.0-fold. These results were confirmed in a kidney-specific HNF1ß knockout mouse, which exhibited downregulation of the Casr by 81%. Cldn19 and Cldn10b expression levels were also decreased by 37% and 83%, respectively, whereas Cldn3 was upregulated by 4.6-fold. In conclusion, HNF1ß is a transcriptional activator of the CaSR. Consequently, patients with HNF1ß mutations may have reduced CaSR activity in the kidney, which could explain cyst progression and hyperabsorption of Ca2+ and Mg2+ in the TAL resulting in hypocalciuria.


Subject(s)
Hepatocyte Nuclear Factor 1-beta/metabolism , Loop of Henle/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Binding Sites , Calcium/metabolism , Claudins/genetics , Claudins/metabolism , Female , HEK293 Cells , Hepatocyte Nuclear Factor 1-beta/deficiency , Hepatocyte Nuclear Factor 1-beta/genetics , Humans , Magnesium/metabolism , Male , Mice, Knockout , Promoter Regions, Genetic , Protein Binding , Receptors, Calcium-Sensing/genetics , Receptors, Calcium-Sensing/metabolism , Receptors, G-Protein-Coupled/genetics , Renal Reabsorption , Transcription, Genetic , Transcriptional Activation
5.
J Biol Chem ; 290(41): 24793-805, 2015 Oct 09.
Article in English | MEDLINE | ID: mdl-26292219

ABSTRACT

The transcription factor hepatocyte nuclear factor-1ß (HNF-1ß) regulates tissue-specific gene expression in the kidney and other epithelial organs. Mutations of HNF-1ß produce kidney cysts, and previous studies have shown that HNF-1ß regulates the transcription of cystic disease genes, including Pkd2 and Pkhd1. Here, we combined chromatin immunoprecipitation and next-generation sequencing (ChIP-Seq) with microarray analysis to identify microRNAs (miRNAs) that are directly regulated by HNF-1ß in renal epithelial cells. These studies identified members of the epithelial-specific miR-200 family (miR-200b/200a/429) as novel transcriptional targets of HNF-1ß. HNF-1ß binds to two evolutionarily conserved sites located 28 kb upstream to miR-200b. Luciferase reporter assays showed that the HNF-1ß binding sites were located within a promoter that was active in renal epithelial cells. Mutations of the HNF-1ß binding sites abolished promoter activity. RT-PCR analysis revealed that a long noncoding RNA (lncRNA) is transcribed from the promoter and encodes the miR-200 cluster. Inhibition of the lncRNA with siRNAs decreased the levels of miR-200 but did not affect expression of the Ttll10 host gene. The expression of the lncRNA and miR-200 was decreased in kidneys from HNF-1ß knock-out mice and renal epithelial cells expressing dominant-negative mutant HNF-1ß. The expression of miR-200 targets, Zeb2 and Pkd1, was increased in HNF-1ß knock-out kidneys and in cells expressing mutant HNF-1ß. Overexpression of miR-200 decreased the expression of Zeb2 and Pkd1 in HNF-1ß mutant cells. These studies reveal a novel pathway whereby HNF-1ß directly contributes to the control of miRNAs that are involved in epithelial-mesenchymal transition and cystic kidney disease.


Subject(s)
Gene Expression Regulation , Hepatocyte Nuclear Factor 1-beta/metabolism , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Animals , Base Sequence , Epithelial Cells/metabolism , Gene Knockout Techniques , Genomics , HeLa Cells , Hepatocyte Nuclear Factor 1-beta/deficiency , Hepatocyte Nuclear Factor 1-beta/genetics , Homeodomain Proteins/genetics , Humans , Kidney/cytology , Mice , Mutation , Repressor Proteins/genetics , TRPP Cation Channels/genetics , Zinc Finger E-box Binding Homeobox 2
6.
J Lipid Res ; 56(4): 801-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25652089

ABSTRACT

The transcription factors hepatic nuclear factor (HNF)1α and HNF1ß can bind to the HNF1 site on the proprotein convertase subtilisin/kexin type 9 (PCSK9) promoter to activate transcription in HepG2 cells. However, it is unknown whether one or both HNF1 factors are obligatory for transactivating hepatic PCSK9 gene expression in vivo. We developed shRNA adenoviral constructs (Ad-shHNF1α and Ad-shHNF1ß) to examine the effects of knockdown of HNF1α or HNF1ß on PCSK9 expression and its consequent impact on LDL receptor (LDLR) protein levels in cultured hepatic cells and liver tissue. We demonstrated that infection with Ad-shHNF1α, but not Ad-shHNF1ß, markedly reduced PCSK9 mRNA expression in HepG2 cells with a concomitant increase in LDLR protein abundance. Injecting Ad-shHNF1α in mice fed a normal diet significantly (∼ 50%) reduced liver mRNA expression and serum concentration of PCSK9 with a concomitant increase (∼ 1.9-fold) in hepatic LDLR protein abundance. Furthermore, we observed a modest but significant reduction in circulating LDL cholesterol after knockdown of HNF1α in these normolipidemic mice. Consistent with the observation that knockdown of HNF1ß did not affect PCSK9 mRNA or protein expression in cultured hepatic cells, Ad-shHNF1ß infection in mice resulted in no change in the hepatic mRNA expression or serum content of PCSK9. Altogether, our study demonstrates that HNF1α, but not HNF1ß, is the primary positive regulator of PCSK9 transcription in mouse liver.


Subject(s)
Cholesterol, LDL/blood , Gene Knockdown Techniques , Hepatocyte Nuclear Factor 1-alpha/deficiency , Hepatocyte Nuclear Factor 1-alpha/genetics , Liver/metabolism , Proprotein Convertases/blood , Serine Endopeptidases/blood , Animals , Diet , Gene Expression Regulation, Enzymologic , HEK293 Cells , Hep G2 Cells , Hepatocyte Nuclear Factor 1-beta/deficiency , Hepatocyte Nuclear Factor 1-beta/genetics , Humans , Liver/cytology , Male , Mice , Organ Specificity , Proprotein Convertase 9 , RNA, Messenger/genetics , RNA, Messenger/metabolism
7.
Am J Med Genet A ; 164A(12): 3076-82, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25256560

ABSTRACT

Deletions of chromosome 17q12 [OMIM 614527] encompass a wide range of phenotypes, including renal cysts, diabetes mellitus, pancreatic structural abnormalities, genital tract anomalies, developmental delay, learning difficulties, and more recently, autism spectrum disorder and schizophrenia. To date, gastrointestinal malformations have not been fully characterized in this syndrome. In this case report, we describe a four-year-old girl with a 17q12 microdeletion who was born with duodenal atresia, bilateral renal cysts, left kidney dysplasia, a midline cystic structure at the conus medullaris, and dysmorphic features. Both the patient and her affected father were found to have a deletion of 17q12, which encompasses the HNF1B (hepatocyte nuclear factor beta). It is hypothesized that HNF1B may play a role in intestinal differentiation and development. Our clinical report further expands the pre-and post-natal presentation of this rare microdeletion syndrome.


Subject(s)
Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Chromosome Aberrations , Chromosomes, Human, Pair 17/genetics , Duodenal Obstruction/genetics , Hepatocyte Nuclear Factor 1-beta/deficiency , Phenotype , Female , Hepatocyte Nuclear Factor 1-beta/genetics , Humans , Intestinal Atresia , Microarray Analysis , Syndrome
8.
Nature ; 494(7435): 111-5, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23389544

ABSTRACT

Insulin resistance represents a hallmark during the development of type 2 diabetes mellitus and in the pathogenesis of obesity-associated disturbances of glucose and lipid metabolism. MicroRNA (miRNA)-dependent post-transcriptional gene silencing has been recognized recently to control gene expression in disease development and progression, including that of insulin-resistant type 2 diabetes. The deregulation of miRNAs miR-143 (ref. 4), miR-181 (ref. 5), and miR-103 and miR-107 (ref. 6) alters hepatic insulin sensitivity. Here we report that the expression of miR-802 is increased in the liver of two obese mouse models and obese human subjects. Inducible transgenic overexpression of miR-802 in mice causes impaired glucose tolerance and attenuates insulin sensitivity, whereas reduction of miR-802 expression improves glucose tolerance and insulin action. We identify Hnf1b (also known as Tcf2) as a target of miR-802-dependent silencing, and show that short hairpin RNA (shRNA)-mediated reduction of Hnf1b in liver causes glucose intolerance, impairs insulin signalling and promotes hepatic gluconeogenesis. In turn, hepatic overexpression of Hnf1b improves insulin sensitivity in Lepr(db/db) mice. Thus, this study defines a critical role for deregulated expression of miR-802 in the development of obesity-associated impairment of glucose metabolism through targeting of Hnf1b, and assigns Hnf1b an unexpected role in the control of hepatic insulin sensitivity.


Subject(s)
Gene Silencing , Glucose/metabolism , Hepatocyte Nuclear Factor 1-beta/deficiency , MicroRNAs/genetics , Obesity/genetics , Animals , Gene Expression Regulation , Gluconeogenesis , Glucose/biosynthesis , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Hepatocyte Nuclear Factor 1-beta/genetics , Hepatocyte Nuclear Factor 1-beta/metabolism , Humans , Insulin/metabolism , Insulin Resistance/genetics , Liver/metabolism , Mice , MicroRNAs/biosynthesis , Signal Transduction
9.
Hepatology ; 56(3): 1178-81, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22706971

ABSTRACT

Heterozygous deletion or mutation in hepatocyte nuclear factor 1 homeobox B/transcription factor 2 (HNF1B/TCF2) causes renal cyst and diabetes syndrome (OMIM #137920). Mice with homozygous liver-specific deletion of Hnf1ß revealed that a complete lack of this factor leads to ductopenia and bile duct dysplasia, in addition to mild hepatocyte defects. However, little is known about the hepatic consequences of deficient HNF1B function in humans. Three patients with heterozygous HNF1B deficiency were found to have normal bile duct formation on radiology and routine liver pathology. Electron microscopy revealed a paucity or absence of normal primary cilia. Therefore, heterozygous HNF1B deficiency is associated with ciliary anomalies in cholangiocytes, and this may cause cholestasis.


Subject(s)
Bile Ducts/cytology , Cilia , Epithelial Cells/pathology , Hepatocyte Nuclear Factor 1-beta/deficiency , Adult , Female , Humans , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...