Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 186
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Development ; 148(9)2021 05 01.
Article in English | MEDLINE | ID: mdl-33929509

ABSTRACT

The vertebrate retina is generated by retinal progenitor cells (RPCs), which produce >100 cell types. Although some RPCs produce many cell types, other RPCs produce restricted types of daughter cells, such as a cone photoreceptor and a horizontal cell (HC). We used genome-wide assays of chromatin structure to compare the profiles of a restricted cone/HC RPC and those of other RPCs in chicks. These data nominated regions of regulatory activity, which were tested in tissue, leading to the identification of many cis-regulatory modules (CRMs) active in cone/HC RPCs and developing cones. Two transcription factors, Otx2 and Oc1, were found to bind to many of these CRMs, including those near genes important for cone development and function, and their binding sites were required for activity. We also found that Otx2 has a predicted autoregulatory CRM. These results suggest that Otx2, Oc1 and possibly other Onecut proteins have a broad role in coordinating cone development and function. The many newly discovered CRMs for cones are potentially useful reagents for gene therapy of cone diseases.


Subject(s)
Dissection , Hepatocyte Nuclear Factor 6/metabolism , Otx Transcription Factors/metabolism , Retina/growth & development , Retinal Cone Photoreceptor Cells/metabolism , Transcription Factors/metabolism , Animals , Binding Sites , Chickens , Chromatin , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Hepatocyte Nuclear Factor 6/genetics , Otx Transcription Factors/genetics , Retina/metabolism , Stem Cells
2.
Genes Dev ; 30(14): 1636-44, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27445394

ABSTRACT

Hepatocyte nuclear factor 6 (HNF6) is required for liver development, but its role in adult liver metabolism is not known. Here we show that deletion of HNF6 in livers of adult C57Bl/6 mice leads to hepatic steatosis in mice fed normal laboratory chow. Although HNF6 is known mainly as a transcriptional activator, hepatic loss of HNF6 up-regulated many lipogenic genes bound directly by HNF6. Many of these genes are targets of the circadian nuclear receptor Rev-erbα, and binding of Rev-erbα at these sites was lost when HNF6 was ablated in the liver. While HNF6 and Rev-erbα coordinately regulate hepatic lipid metabolism, each factor also affects additional gene sets independently. These findings highlight a novel mechanism of transcriptional repression by HNF6 and demonstrate how overlapping and distinct mechanisms of transcription factor function contribute to the integrated physiology of the liver.


Subject(s)
Gene Expression Regulation/genetics , Hepatocyte Nuclear Factor 6/genetics , Hepatocyte Nuclear Factor 6/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Liver/physiopathology , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Animals , Fatty Liver/genetics , Gene Deletion , Gene Knockout Techniques , Male , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Protein Binding/genetics
3.
BMC Genomics ; 24(1): 252, 2023 May 10.
Article in English | MEDLINE | ID: mdl-37165305

ABSTRACT

BACKGROUND: The long non-coding RNAs (lncRNAs) are critical regulators of diverse biological processes. Nevertheless, a global view of its expression and function in the mouse retina, a crucial model for neurogenesis study, still needs to be made available. RESULTS: Herein, by integrating the established gene models and the result from ab initio prediction using short- and long-read sequencing, we characterized 4,523 lncRNA genes (MRLGs) in developing mouse retinas (from the embryonic day of 12.5 to the neonatal day of P28), which was so far the most comprehensive collection of retinal lncRNAs. Next, derived from transcriptomics analyses of different tissues and developing retinas, we found that the MRLGs were highly spatiotemporal specific in expression and played essential roles in regulating the genesis and function of mouse retinas. In addition, we investigated the expression of MRLGs in some mouse mutants and revealed that 97 intergenic MRLGs might be involved in regulating differentiation and development of retinal neurons through Math5, Isl1, Brn3b, NRL, Onecut1, or Onecut2 mediated pathways. CONCLUSIONS: In summary, this work significantly enhanced our knowledge of lncRNA genes in mouse retina development and provided valuable clues for future exploration of their biological roles.


Subject(s)
RNA, Long Noncoding , Animals , Mice , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Retina/metabolism , Gene Expression Profiling , Hepatocyte Nuclear Factor 6/genetics , Hepatocyte Nuclear Factor 6/metabolism
4.
Gastric Cancer ; 24(1): 103-116, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32705446

ABSTRACT

BACKGROUND: Gastric intestinal metaplasia (IM) is considered a precancerous lesion, and bile acids (BA) play a critical role in the induction of IM. Ectopic expression of HNF4α was observed in a BA-induced IM cell model. However, the mechanisms underlying the upregulation of the protein in IM cells remains to be elucidated. METHODS: The effects of HNF4α on gastric mucosal cells in vivo were identified by a transgenic mouse model and RNA-seq was used to screen downstream targets of deoxycholic acid (DCA). The expression of pivotal molecules and miR-1 was detected by immunohistochemistry and in situ hybridization in normal, gastritis and IM tissue slides or microarrays. The transcriptional regulation of HDAC6 was investigated by chromatin immunoprecipitation (ChIP) and luciferase reporter assays. RESULTS: The transgenic mouse model validated that HNF4α stimulated the HDAC6 expression and mucin secretion in gastric mucosa. Increased HDAC6 and HNF4α expression was also detected in the gastric IM cell model and patient specimens. HNF4α could bind to and activate HDAC6 promoter. In turn, HDAC6 enhanced the HNF4α protein level in GES-1 cells. Furthermore, miR-1 suppressed the expression of downstream intestinal markers by targeting HDAC6 and HNF4α. CONCLUSIONS: Our findings show that the HDAC6/HNF4α loop regulated by miR-1 plays a critical role in gastric IM. Blocking the activation of this loop could be a potential approach to preventing BA-induced gastric IM or even gastric cancer (GC).


Subject(s)
Gastric Mucosa/pathology , Hepatocyte Nuclear Factor 6/metabolism , Histone Deacetylase 6/metabolism , MicroRNAs/metabolism , Stomach Neoplasms/genetics , Animals , Bile Acids and Salts/metabolism , Disease Models, Animal , Gastric Mucosa/metabolism , Gastritis/genetics , Gene Expression Regulation/genetics , Humans , Immunohistochemistry , In Situ Hybridization , Metaplasia/genetics , Mice , Precancerous Conditions/genetics , Promoter Regions, Genetic/genetics , Stomach Neoplasms/pathology , Transcription, Genetic/genetics
5.
Nucleic Acids Res ; 47(11): 5587-5602, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31049588

ABSTRACT

Remodeling of chromatin accessibility is necessary for successful reprogramming of fibroblasts to neurons. However, it is still not fully known which transcription factors can induce a neuronal chromatin accessibility profile when overexpressed in fibroblasts. To identify such transcription factors, we used ATAC-sequencing to generate differential chromatin accessibility profiles between human fibroblasts and iNeurons, an in vitro neuronal model system obtained by overexpression of Neurog2 in induced pluripotent stem cells (iPSCs). We found that the ONECUT transcription factor sequence motif was strongly associated with differential chromatin accessibility between iNeurons and fibroblasts. All three ONECUT transcription factors associated with this motif (ONECUT1, ONECUT2 and ONECUT3) induced a neuron-like morphology and expression of neuronal genes within two days of overexpression in fibroblasts. We observed widespread remodeling of chromatin accessibility; in particular, we found that chromatin regions that contain the ONECUT motif were in- or lowly accessible in fibroblasts and became accessible after the overexpression of ONECUT1, ONECUT2 or ONECUT3. There was substantial overlap with iNeurons, still, many regions that gained accessibility following ONECUT overexpression were not accessible in iNeurons. Our study highlights both the potential and challenges of ONECUT-based direct neuronal reprogramming.


Subject(s)
Cellular Reprogramming , Chromatin/genetics , Gene Expression Regulation , Induced Pluripotent Stem Cells/metabolism , Neurons/metabolism , Onecut Transcription Factors/genetics , Cell Differentiation , Cell Line , Chromatin/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Profiling , Gene Ontology , Hepatocyte Nuclear Factor 6/genetics , Hepatocyte Nuclear Factor 6/metabolism , Homeodomain Proteins , Humans , Induced Pluripotent Stem Cells/cytology , Neurons/cytology , Onecut Transcription Factors/metabolism , Transcription Factors
6.
Bull Exp Biol Med ; 170(1): 40-45, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33222081

ABSTRACT

Hepatocyte nuclear factor (HNF-6) is a liver-specific protein and a key component in the differentiation process during the development of mature liver. The immunohistochemical staining and RT-PCR techniques were employed to examine the expression of HNF-6 and proliferation of Ki-67+ cells during the early regeneration of the liver on postsurgery in 3, 6, 12, and 24 h in original model of partial hepatectomy in rats. The earliest proliferating (Ki-67+) cells were observed in 3 h after surgery in liver sinusoids (liver macrophages) and then in liver parenchyma. Expression of HNF-6 in hepatocytes and epithelial cells of the bile ducts attained maximum in 6 h after surgery. At later terms, this parameter somewhat decreased, but still surpassed the control level.


Subject(s)
Hepatocyte Nuclear Factor 6/genetics , Hepatocytes/metabolism , Kupffer Cells/metabolism , Liver Regeneration/genetics , Liver/metabolism , Animals , Bile Ducts/metabolism , Bile Ducts/surgery , Cell Proliferation , Female , Gene Expression Regulation , Hepatectomy/methods , Hepatocyte Nuclear Factor 6/metabolism , Hepatocytes/cytology , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Kupffer Cells/cytology , Liver/surgery , Male , Rats , Rats, Sprague-Dawley
7.
J Cell Physiol ; 234(4): 3675-3684, 2019 04.
Article in English | MEDLINE | ID: mdl-30256389

ABSTRACT

Hepatocyte nuclear factor 6 (HNF6), as a transcription factor, has been reported to be involved in cell proliferation, carcinogenesis, and tumor metastasis. Here, we demonstrated the role of HNF6 in tumor growth and liver metastasis in colorectal cancer (CRC). Through bioinformatics and clinical samples analysis, we found HNF6 messenger RNA was upregulated both in CRC primary sites and liver metastases, and its high expression indicated poor survival in CRC patients. In vitro studies confirmed that HNF6 promoted cell proliferation and colony formation. What is more, in mouse models, the xenografts grew significantly faster and liver metastasis rate was nearly 45% higher in mice injected with HNF6-overexpressing cells. Further mechanism exploration showed that HNF6 expression affected cell adhesion and conferred resistance to anoikis in CRC cells. Taken together, HNF6 expression was upregulated in CRC and closely correlated with poor survival. HNF6 promoted CRC cell proliferation and tumor growth, and may contribute to liver metastasis via conferring cell resistance to anoikis.


Subject(s)
Cell Movement , Cell Proliferation , Colorectal Neoplasms/metabolism , Hepatocyte Nuclear Factor 6/metabolism , Liver Neoplasms/metabolism , Animals , Anoikis , Cell Adhesion , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 6/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Mice, Inbred BALB C , Mice, Nude , Signal Transduction , Tumor Burden , Up-Regulation
8.
Am J Physiol Endocrinol Metab ; 314(4): E308-E321, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29351489

ABSTRACT

The transcription factors pancreatic and duodenal homeobox 1 (Pdx1) and onecut1 (Oc1) are coexpressed in multipotent pancreatic progenitors (MPCs), but their expression patterns diverge in hormone-expressing cells, with Oc1 expression being extinguished in the endocrine lineage and Pdx1 being maintained at high levels in ß-cells. We previously demonstrated that cooperative function of these two factors in MPCs is necessary for proper specification and differentiation of pancreatic endocrine cells. In those studies, we observed a persistent decrease in expression of the ß-cell maturity factor MafA. We therefore hypothesized that Pdx1 and Oc1 cooperativity in MPCs impacts postnatal ß-cell maturation and function. Here our model of Pdx1-Oc1 double heterozygosity was used to investigate the impact of haploinsufficiency for both of these factors on postnatal ß-cell maturation, function, and adaptability. Examining mice at postnatal day (P) 14, we observed alterations in pancreatic insulin content in both Pdx1 heterozygotes and double heterozygotes. Gene expression analysis at this age revealed significantly decreased expression of many genes important for glucose-stimulated insulin secretion (e.g., Glut2, Pcsk1/2, Abcc8) exclusively in double heterozygotes. Analysis of P14 islets revealed an increase in the number of mixed islets in double heterozygotes. We predicted that double-heterozygous ß-cells would have an impaired ability to respond to stress. Indeed, we observed that ß-cell proliferation fails to increase in double heterozygotes in response to either high-fat diet or placental lactogen. We thus report here the importance of cooperation between regulatory factors early in development for postnatal islet maturation and adaptability.


Subject(s)
Hepatocyte Nuclear Factor 6/physiology , Homeodomain Proteins/physiology , Insulin-Secreting Cells/physiology , Islets of Langerhans/growth & development , Multipotent Stem Cells/metabolism , Trans-Activators/physiology , Adaptation, Physiological/drug effects , Adaptation, Physiological/genetics , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Diet, High-Fat , Gene Expression Regulation, Developmental/drug effects , Glucose/pharmacology , Hepatocyte Nuclear Factor 6/genetics , Homeodomain Proteins/genetics , Insulin-Secreting Cells/drug effects , Islets of Langerhans/drug effects , Islets of Langerhans/physiology , Male , Mice , Mice, Transgenic , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/physiology , Organogenesis/drug effects , Organogenesis/genetics , Trans-Activators/genetics
9.
Hum Mol Genet ; 25(22): 5017-5026, 2016 11 15.
Article in English | MEDLINE | ID: mdl-28159992

ABSTRACT

Genetic diseases associated with defects in primary cilia are classified as ciliopathies. Pancreatic lesions and ductal cysts are found in patients with ciliopathic polycystic kidney diseases suggesting a close connection between pancreatic defects and primary cilia. Here we investigate the role of two genes whose deletion is known to cause primary cilium defects, namely Hnf6 and Lkb1, in pancreatic ductal homeostasis. We find that mice with postnatal duct-specific deletion of Hnf6 or Lkb1 show duct dilations. Cells lining dilated ducts present shorter cilia with swollen tips, suggesting defective intraciliary transport. This is associated with signs of chronic pancreatitis, namely acinar-to-ductal metaplasia, acinar proliferation and apoptosis, presence of inflammatory infiltrates, fibrosis and lipomatosis. Our data reveal a tight association between ductal ciliary defects and pancreatitis with perturbed acinar homeostasis and differentiation. Such injuries can account for the increased risk to develop pancreatic cancer in Peutz-Jeghers patients who carry LKB1 loss-of-function mutations.


Subject(s)
Cilia/pathology , Hepatocyte Nuclear Factor 6/metabolism , Pancreatitis, Chronic/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Animals , Apoptosis/physiology , Cell Differentiation , Cilia/genetics , Epithelial Cells/pathology , Hepatocyte Nuclear Factor 6/genetics , Lipomatosis/genetics , Lipomatosis/metabolism , Metaplasia/genetics , Metaplasia/metabolism , Mice , Pancreas/pathology , Pancreatic Ducts/metabolism , Pancreatic Neoplasms/genetics , Pancreatitis, Chronic/genetics , Protein Serine-Threonine Kinases/genetics
10.
Development ; 142(5): 858-70, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25715394

ABSTRACT

In human, mutations in bicaudal C1 (BICC1), an RNA binding protein, have been identified in patients with kidney dysplasia. Deletion of Bicc1 in mouse leads to left-right asymmetry randomization and renal cysts. Here, we show that BICC1 is also expressed in both the pancreatic progenitor cells that line the ducts during development, and in the ducts after birth, but not in differentiated endocrine or acinar cells. Genetic inactivation of Bicc1 leads to ductal cell over-proliferation and cyst formation. Transcriptome comparison between WT and Bicc1 KO pancreata, before the phenotype onset, reveals that PKD2 functions downstream of BICC1 in preventing cyst formation in the pancreas. Moreover, the analysis highlights immune cell infiltration and stromal reaction developing early in the pancreas of Bicc1 knockout mice. In addition to these functions in duct morphogenesis, BICC1 regulates NEUROG3(+) endocrine progenitor production. Its deletion leads to a late but sustained endocrine progenitor decrease, resulting in a 50% reduction of endocrine cells. We show that BICC1 functions downstream of ONECUT1 in the pathway controlling both NEUROG3(+) endocrine cell production and ductal morphogenesis, and suggest a new candidate gene for syndromes associating kidney dysplasia with pancreatic disorders, including diabetes.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hepatocyte Nuclear Factor 6/metabolism , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Blotting, Western , Fluorescent Antibody Technique , Genotype , Hepatocyte Nuclear Factor 6/genetics , In Situ Nick-End Labeling , Mice , Nerve Tissue Proteins/genetics , RNA-Binding Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA , Stem Cells/cytology , Stem Cells/metabolism , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism
11.
Proc Natl Acad Sci U S A ; 111(39): E4086-95, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25228773

ABSTRACT

Previously, we have shown that Onecut1 (Oc1) and Onecut2 (Oc2) are expressed in retinal progenitor cells, developing retinal ganglion cells (RGCs), and horizontal cells (HCs). However, in Oc1-null mice, we only observed an 80% reduction in HCs, but no defects in other cell types. We postulated that the lack of defects in other cell types in Oc1-null retinas was a result of redundancy with Oc2. To test this theory, we have generated Oc2-null mice and now show that their retinas also only have defects in HCs, with a 50% reduction in their numbers. However, when both Oc1 and Oc2 are knocked out, the retinas exhibit more profound defects in the development of all early retinal cell types, including completely failed genesis of HCs, compromised generation of cones, reduced production (by 30%) of RGCs, and absence of starburst amacrine cells. Cone subtype diversification and RGC subtype composition also were affected in the double-null retina. Using RNA-Seq expression profiling, we have identified downstream genes of Oc1 and Oc2, which not only confirms the redundancy between the two factors and renders a molecular explanation for the defects in the double-null retinas, but also shows that the onecut factors suppress the production of the late cell type, rods, indicating that the two factors contribute to the competence of retinal progenitor cells for the early retinal cell fates. Our results provide insight into how onecut factors regulate the creation of cellular diversity in the retina and, by extension, in the central nervous system in general.


Subject(s)
Hepatocyte Nuclear Factor 6/metabolism , Homeodomain Proteins/metabolism , Retina/cytology , Retina/embryology , Transcription Factors/metabolism , Amacrine Cells/cytology , Amacrine Cells/metabolism , Animals , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 6/deficiency , Hepatocyte Nuclear Factor 6/genetics , Homeodomain Proteins/genetics , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Pregnancy , Retina/metabolism , Retinal Cone Photoreceptor Cells/cytology , Retinal Cone Photoreceptor Cells/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Retinal Horizontal Cells/cytology , Retinal Horizontal Cells/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics
12.
Dev Biol ; 402(1): 48-60, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25794677

ABSTRACT

Genetic studies of the last decades strongly indicated that generation of particular retinal cell types is governed by gene regulatory networks of transcription factors and their target genes. The paired and homeodomain transcription factor Pax6 plays a pivotal role in retinal development as its inactivation in the retinal progenitor cell population leads to abolished differentiation of all retinal cell types. However, until now, only a few transcription factors operating downstream of Pax6 responsible for generation of individual retinal cell types have been identified. In this study, we identified two transcription factors of the Onecut family, Onecut1 and Onecut2, as Pax6 downstream-acting factors. Onecut1 and Onecut2 were previously shown to be expressed in developing horizontal cells, retinal ganglion cells and cone photoreceptors; however, their role in differentiation of these cell types is poorly understood. In this study, we show that the horizontal cell genesis is severely disturbed in Onecut-deficient retinae. In single Onecut1 and Onecut2 mutants, the number of horizontal cells is dramatically reduced while horizontal cells are completely missing in the Onecut1/Onecut2 compound mutant. Analysis of genes involved in the horizontal cell genesis such as Foxn4, Ptf1a, Prox1 and Lim1 showed that although horizontal cells are initially formed, they are not maintained in Onecut-deficient retinae. Taken together, this study suggests the model in which Pax6 regulates the maintenance of horizontal cells through the activation of Onecut1 and Onecut2 transcription factors.


Subject(s)
Eye Proteins/metabolism , Hepatocyte Nuclear Factor 6/metabolism , Homeodomain Proteins/metabolism , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , Retina/embryology , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites , Female , Gene Expression Regulation, Developmental , In Situ Hybridization , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , PAX6 Transcription Factor , Phenotype , Retina/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Sequence Homology, Nucleic Acid , Stem Cells/cytology
13.
Am J Hum Genet ; 92(3): 460-7, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23472758

ABSTRACT

Hemophilia B, or the "royal disease," arises from mutations in coagulation factor IX (F9). Mutations within the F9 promoter are associated with a remarkable hemophilia B subtype, termed hemophilia B Leyden, in which symptoms ameliorate after puberty. Mutations at the -5/-6 site (nucleotides -5 and -6 relative to the transcription start site, designated +1) account for the majority of Leyden cases and have been postulated to disrupt the binding of a transcriptional activator, the identity of which has remained elusive for more than 20 years. Here, we show that ONECUT transcription factors (ONECUT1 and ONECUT2) bind to the -5/-6 site. The various hemophilia B Leyden mutations that have been reported in this site inhibit ONECUT binding to varying degrees, which correlate well with their associated clinical severities. In addition, expression of F9 is crucially dependent on ONECUT factors in vivo, and as such, mice deficient in ONECUT1, ONECUT2, or both exhibit depleted levels of F9. Taken together, our findings establish ONECUT transcription factors as the missing hemophilia B Leyden regulators that operate through the -5/-6 site.


Subject(s)
Factor IX/genetics , Hemophilia B/genetics , Hepatocyte Nuclear Factor 6/metabolism , Homeodomain Proteins/metabolism , Mutation , Transcription Factors/metabolism , Animals , Binding Sites , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Genetic Predisposition to Disease , Hep G2 Cells , Humans , Liver/metabolism , Male , Mice , Mice, Knockout , Promoter Regions, Genetic , Transcription, Genetic
14.
J Virol ; 89(8): 4345-55, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25653429

ABSTRACT

UNLABELLED: Hepatitis B virus (HBV), a small enveloped DNA virus, chronically infects more than 350 million people worldwide and causes liver diseases from hepatitis to cirrhosis and liver cancer. Here, we report that hepatocyte nuclear factor 6 (HNF6), a liver-enriched transcription factor, can inhibit HBV gene expression and DNA replication. Overexpression of HNF6 inhibited, while knockdown of HNF6 expression enhanced, HBV gene expression and replication in hepatoma cells. Mechanistically, the SP2 promoter was inhibited by HNF6, which partly accounts for the inhibition on S mRNA. Detailed analysis showed that a cis element on the HBV genome (nucleotides [nt] 3009 to 3019) was responsible for the inhibition of the SP2 promoter by HNF6. Moreover, further analysis showed that HNF6 reduced viral pregenomic RNA (pgRNA) posttranscriptionally via accelerating the degradation of HBV pgRNA independent of La protein. Furthermore, by using truncated mutation experiments, we demonstrated that the N-terminal region of HNF6 was responsible for its inhibitory effects. Importantly, introduction of an HNF6 expression construct with the HBV genome into the mouse liver using hydrodynamic injection resulted in a significant reduction in viral gene expression and DNA replication. Overall, our data demonstrated that HNF6 is a novel host factor that can restrict HBV replication via both transcriptional and posttranscriptional mechanisms. IMPORTANCE: HBV is a major human pathogen whose replication is regulated by host factors. Liver-enriched transcription factors are critical for many liver functions, including metabolism, development, and cell proliferation, and some of them have been shown to regulate HBV gene expression or replication in different manners. In this study, we showed that HNF6 could inhibit the gene expression and DNA replication of HBV via both transcriptional and posttranscriptional mechanisms. As HNF6 is differentially expressed in men and women, the current results may suggest a role of HNF6 in the gender dimorphism of HBV infection.


Subject(s)
DNA Replication/genetics , Gene Expression Regulation, Viral/genetics , Hepatitis B virus/metabolism , Hepatocyte Nuclear Factor 6/metabolism , Sex Characteristics , Animals , Blotting, Northern , Blotting, Western , Cell Line, Tumor , DNA Primers/genetics , Female , Genetic Vectors/genetics , HEK293 Cells , Hepatitis B virus/genetics , Hepatocyte Nuclear Factor 6/genetics , Humans , Immunohistochemistry , Luciferases , Male , Mice , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Transfection/methods
15.
Nucleic Acids Res ; 42(20): e156, 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-25249628

ABSTRACT

Understanding the role of a given transcription factor (TF) in regulating gene expression requires precise mapping of its binding sites in the genome. Chromatin immunoprecipitation-exo, an emerging technique using λ exonuclease to digest TF unbound DNA after ChIP, is designed to reveal transcription factor binding site (TFBS) boundaries with near-single nucleotide resolution. Although ChIP-exo promises deeper insights into transcription regulation, no dedicated bioinformatics tool exists to leverage its advantages. Most ChIP-seq and ChIP-chip analytic methods are not tailored for ChIP-exo, and thus cannot take full advantage of high-resolution ChIP-exo data. Here we describe a novel analysis framework, termed MACE (model-based analysis of ChIP-exo) dedicated to ChIP-exo data analysis. The MACE workflow consists of four steps: (i) sequencing data normalization and bias correction; (ii) signal consolidation and noise reduction; (iii) single-nucleotide resolution border peak detection using the Chebyshev Inequality and (iv) border matching using the Gale-Shapley stable matching algorithm. When applied to published human CTCF, yeast Reb1 and our own mouse ONECUT1/HNF6 ChIP-exo data, MACE is able to define TFBSs with high sensitivity, specificity and spatial resolution, as evidenced by multiple criteria including motif enrichment, sequence conservation, direct sequence pileup, nucleosome positioning and open chromatin states. In addition, we show that the fundamental advance of MACE is the identification of two boundaries of a TFBS with high resolution, whereas other methods only report a single location of the same event. The two boundaries help elucidate the in vivo binding structure of a given TF, e.g. whether the TF may bind as dimers or in a complex with other co-factors.


Subject(s)
Chromatin Immunoprecipitation/methods , Sequence Analysis, DNA/methods , Transcription Factors/metabolism , Algorithms , Animals , Binding Sites , CCCTC-Binding Factor , Computer Simulation , DNA-Binding Proteins/metabolism , Exodeoxyribonucleases , Genome , Hepatocyte Nuclear Factor 6/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism
16.
Nat Genet ; 39(6): 730-2, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17529977

ABSTRACT

We demonstrate that the binding sites for highly conserved transcription factors vary extensively between human and mouse. We mapped the binding of four tissue-specific transcription factors (FOXA2, HNF1A, HNF4A and HNF6) to 4,000 orthologous gene pairs in hepatocytes purified from human and mouse livers. Despite the conserved function of these factors, from 41% to 89% of their binding events seem to be species specific. When the same protein binds the promoters of orthologous genes, approximately two-thirds of the binding sites do not align.


Subject(s)
Conserved Sequence/genetics , Gene Expression Regulation , Regulatory Sequences, Nucleic Acid , Transcription Factors/genetics , Transcription, Genetic , Animals , Genetic Variation , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 6/genetics , Humans , Mice , Sequence Homology
17.
Dev Biol ; 396(2): 201-13, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25446530

ABSTRACT

UNLABELLED: Notch signaling plays an acknowledged role in bile-duct development, but its involvement in cholangiocyte-fate determination remains incompletely understood. We investigated the effects of early Notch2 deletion in Notch2(fl/fl)/Alfp-Cre(tg/-) ("Notch2-cKO") and Notch2(fl/fl)/Alfp-Cre(-/-) ("control") mice. Fetal and neonatal Notch2-cKO livers were devoid of cytokeratin19 (CK19)-, Dolichos-biflorus agglutinin (DBA)-, and SOX9-positive ductal structures, demonstrating absence of prenatal cholangiocyte differentiation. Despite extensive cholestatic hepatocyte necrosis and growth retardation, mortality was only ~15%. Unexpectedly, a slow process of secondary cholangiocyte differentiation and bile-duct formation was initiated around weaning that histologically resembled the ductular reaction. Newly formed ducts varied from rare and non-connected, to multiple, disorganized tubular structures that connected to the extrahepatic bile ducts. Jaundice had disappeared in ~30% of Notch2-cKO mice by 6 months. The absence of NOTCH2 protein in postnatally differentiating cholangiocyte nuclei of Notch2-cKO mice showed that these cells had not originated from non-recombined precursor cells. Notch2 and Hnf6 mRNA levels were permanently decreased in Notch2-cKO livers. Perinatally, Foxa1, Foxa2, Hhex, Hnf1ß, Cebpα and Sox9 mRNA levels were all significantly lower in Notch2-cKO than control mice, but all except Foxa2 returned to normal or increased levels after weaning, coincident with the observed secondary bile-duct formation. Interestingly, Hhex and Sox9 mRNA levels remained elevated in icteric 6 months old Notch2-cKOs, but decreased to control levels in non-icteric Notch2-cKOs, implying a key role in secondary bile-duct formation. CONCLUSION: Cholangiocyte differentiation becomes progressively less dependent on NOTCH2 signaling with age, suggesting that ductal-plate formation is dependent on NOTCH2, but subsequent cholangiocyte differentiation is not.


Subject(s)
Bile Ducts/abnormalities , Bile Ducts/growth & development , Liver/metabolism , Organogenesis/genetics , Receptor, Notch2/deficiency , Analysis of Variance , Animals , DNA Primers/genetics , Hepatocyte Nuclear Factor 6/metabolism , Histological Techniques , Immunohistochemistry , Mice , Mice, Knockout , Organogenesis/physiology , Polymerase Chain Reaction , Regression Analysis , Weaning
18.
Development ; 139(1): 179-90, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22115757

ABSTRACT

The spinal cord contains a diverse array of physiologically distinct interneuron cell types that subserve specialized roles in somatosensory perception and motor control. The mechanisms that generate these specialized interneuronal cell types from multipotential spinal progenitors are not known. In this study, we describe a temporally regulated transcriptional program that controls the differentiation of Renshaw cells (RCs), an anatomically and functionally discrete spinal interneuron subtype. We show that the selective activation of the Onecut transcription factors Oc1 and Oc2 during the first wave of V1 interneuron neurogenesis is a key step in the RC differentiation program. The development of RCs is additionally dependent on the forkhead transcription factor Foxd3, which is more broadly expressed in postmitotic V1 interneurons. Our demonstration that RCs are born, and activate Oc1 and Oc2 expression, in a narrow temporal window leads us to posit that neuronal diversity in the developing spinal cord is established by the composite actions of early spatial and temporal determinants.


Subject(s)
Cell Differentiation/physiology , Gene Expression Regulation, Developmental/physiology , Hepatocyte Nuclear Factor 6/metabolism , Homeodomain Proteins/metabolism , Interneurons/cytology , Spinal Cord/cytology , Spinal Cord/embryology , Transcription Factors/metabolism , Animals , Bromodeoxyuridine , Crosses, Genetic , Electrophysiology , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Interneurons/metabolism , Interneurons/physiology , Mice , Time Factors
19.
Am J Pathol ; 184(5): 1479-88, 2014 May.
Article in English | MEDLINE | ID: mdl-24631193

ABSTRACT

The potential for intrahepatic bile duct (IHBD) regeneration in patients with bile duct insufficiency diseases is poorly understood. Notch signaling and Hnf6 have each been shown to be important for the morphogenesis of IHBDs in mice. One congenital pediatric liver disease characterized by reduced numbers of IHBDs, Alagille syndrome, is associated with mutations in Notch signaling components. Therefore, we investigated whether liver cell plasticity could contribute to IHBD regeneration in mice with disruptions in Notch signaling and Hnf6. We studied a mouse model of bile duct insufficiency with liver epithelial cell-specific deficiencies in Hnf6 and Rbpj, a mediator of canonical Notch signaling. Albumin-Cre Hnf6(flox/flox)Rbpj(flox/flox) mice initially developed no peripheral bile ducts. The evolving postnatal liver phenotype was analyzed using IHBD resin casting, immunostaining, and serum chemistry. With age, Albumin-Cre Hnf6(flox/flox)Rbpj(flox/flox) mice mounted a ductular reaction extending through the hepatic tissue and then regenerated communicating peripheral IHBD branches. Rbpj and Hnf6 were determined to remain absent from biliary epithelial cells constituting the ductular reaction and the regenerated peripheral IHBDs. We report the expression of Sox9, a marker of biliary epithelial cells, in cells expressing hepatocyte markers. Tissue analysis indicates that reactive ductules did not arise directly from preexisting hilar IHBDs. We conclude that liver cell plasticity is competent for regeneration of IHBDs independent of Notch signaling via Rbpj and Hnf6.


Subject(s)
Bile Ducts, Intrahepatic/physiology , Hepatocyte Nuclear Factor 6/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Receptors, Notch/metabolism , Regeneration/physiology , Animals , Epithelial Cells/metabolism , Hepatocyte Nuclear Factor 6/deficiency , Hepatocytes/metabolism , Imaging, Three-Dimensional , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunohistochemistry , Keratin-19/metabolism , Mice, Knockout , Plant Lectins/metabolism , Portal Vein/metabolism , SOX9 Transcription Factor/metabolism
20.
J Neurosci ; 33(32): 13053-65, 13065a, 2013 Aug 07.
Article in English | MEDLINE | ID: mdl-23926259

ABSTRACT

Horizontal cells are interneurons that synapse with photoreceptors in the outer retina. Their genesis during development is subject to regulation by transcription factors in a hierarchical manner. Previously, we showed that Onecut 1 (Oc1), an atypical homeodomain transcription factor, is expressed in developing horizontal cells (HCs) and retinal ganglion cells (RGCs) in the mouse retina. Herein, by knocking out Oc1 specifically in the developing retina, we show that the majority (∼80%) of HCs fail to form during early retinal development, implying that Oc1 is essential for HC genesis. However, no other retinal cell types, including RGCs, were affected in the Oc1 knock-out. Analysis of the genetic relationship between Oc1 and other transcription factor genes required for HC development revealed that Oc1 functions downstream of FoxN4, in parallel with Ptf1a, but upstream of Lim1 and Prox1. By in utero electroporation, we found that Oc1 and Ptf1a together are not only essential, but also sufficient for determination of HC fate. In addition, the synaptic connections in the outer plexiform layer are defective in Oc1-null mice, and photoreceptors undergo age-dependent degeneration, indicating that HCs are not only an integral part of the retinal circuitry, but also are essential for the survival of photoreceptors. In sum, these results demonstrate that Oc1 is a critical determinant of HC fate, and reveal that HCs are essential for photoreceptor viability, retinal integrity, and normal visual function.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Hepatocyte Nuclear Factor 6/metabolism , Neurogenesis/genetics , Retina/cytology , Retinal Horizontal Cells/metabolism , Animals , Cell Count , Cell Differentiation/genetics , Cell Survival , Embryo, Mammalian , Eye Proteins/genetics , Green Fluorescent Proteins/genetics , Hepatocyte Nuclear Factor 6/genetics , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Pathways/metabolism , Neural Pathways/ultrastructure , Neuroglia/metabolism , Neuroglia/physiology , Neurons/classification , Neurons/metabolism , Neurons/ultrastructure , Protein Kinase C-alpha/metabolism , Retina/embryology , Retinal Horizontal Cells/ultrastructure , Synapses/metabolism , Synapses/ultrastructure , Transcription Factors/genetics , Transcription Factors/metabolism , Homeobox Protein SIX3
SELECTION OF CITATIONS
SEARCH DETAIL