Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Am J Surg Pathol ; 43(3): 325-333, 2019 03.
Article in English | MEDLINE | ID: mdl-30608233

ABSTRACT

BACKGROUND: The Arias-Stella reaction (ASR) can mimic endometrial clear cell carcinoma (ECCC) in small biopsies, especially when drug or pregnancy history is unknown. A panel of immunohistochemical markers comprising napsin A, hepatocyte nuclear factor-1-beta (HNF-1ß), estrogen and progesterone receptors (ER, PR) has been found useful in confirming a diagnosis of ECCC. However, the detailed characterization of how expression of this combination of markers in the ECCC mimics ASR has yet to be thoroughly evaluated. DESIGN: The frequency and extent of napsin A, HNF-1ß, ER, and PR expression in ASR were assessed in a large series. For napsin A, any cytoplasmic staining was considered positive while only nuclear staining was deemed to be positive for HNF-1ß, ER, and PR. Immunohistochemical histoscores based on the intensity and extent of staining were calculated. RESULTS: Forty cases were gestational and 10 were nongestational ASR. In 19 (38%), the reaction was extensive and involved >50% of the glands. A stromal decidual change was found in 31 (77.5%) of the gestational and 3 (30%) of the nongestational cases. Napsin A was positive in all gestational and 8 of 10 (80%) nongestational ASR. All ASR showed HNF-1ß expression. ER expression was reduced in 37 (92.5%) and lost in 3 (7.5%) gestational ASR, and reduced in 9 (90%) and lost in 1 (10%) of nongestational ASR. None of the ASR in our series expressed PR. CONCLUSIONS: Naspin A and HNF-1ß were frequently expressed in both gestational and nongestational ASR, and ER expression was usually either reduced or loss. Interpretation of these markers in small biopsies containing atypical clear cells should be made with caution.


Subject(s)
Adenocarcinoma, Clear Cell/diagnosis , Biomarkers, Tumor/analysis , Endometrial Neoplasms/diagnosis , Uterine Diseases/diagnosis , Adolescent , Adult , Aspartic Acid Endopeptidases/analysis , Aspartic Acid Endopeptidases/biosynthesis , Diagnosis, Differential , Female , Hepatocyte Nuclear Factor 1-beta/analysis , Hepatocyte Nuclear Factor 1-beta/biosynthesis , Humans , Pregnancy , Receptors, Estrogen/analysis , Receptors, Estrogen/biosynthesis , Receptors, Progesterone/analysis , Receptors, Progesterone/biosynthesis , Young Adult
2.
Appl Immunohistochem Mol Morphol ; 27(4): e32-e38, 2019 04.
Article in English | MEDLINE | ID: mdl-29406331

ABSTRACT

Biliary atresia (BA) is a necroinflammatory occlusive cholangiopathy that affects infants. Genetic and environmental factors has been proposed for its occurrence. The objectives of this study was to investigate the protein expression of 2 important genes regulating ductal plate remodeling, hepatocyte nuclear factor 1-beta (Hnf1ß) and the fork head box protein A2 (FoxA2) in liver tissue from patients with BA and to compare their expression with other causes of neonatal cholestasis (NC). This retrospective study included 60 pediatric patients, 30 with BA and 30 with NC. Immunohistochemistry of Hnf1ß and FoxA2 was performed on liver tissues from studied patients as well as 20 healthy subjects. Statistical analysis between immunohistochemistry results and other parameters was performed. Liver tissue from patients with BA revealed reduced Hnf1ß and FoxA2 immunoexpression. A strong significant statistical difference between BA and NC group (P<0.0001) with regard to Hnf1ß and FoxA2 immunoexpression was evident. Moreover, Hnf1ß was significantly correlated with FoxA2 immunoexpression, stage of fibrosis, bile ductular proliferation, and bile plugs in bile ductules. Hnf1ß immunoreaction in BA cases showed 76.7% sensitivity, 90% specificity, 88.5% positive predictive value, 79.4% negative predictive value, and 83.4% accuracy. FoxA2 expression in BA cases revealed 70.0% sensitivity, 80.0% specificity, 77.8% positive predictive value, 72.7% negative predictive value, 75.0% accuracy. Hnf1ß and FoxA2 immunoexpression could differentiate between BA from other cause of NC.


Subject(s)
Biliary Atresia , Cholestasis , Gene Expression Regulation , Hepatocyte Nuclear Factor 1-beta/biosynthesis , Hepatocyte Nuclear Factor 3-beta/biosynthesis , Liver , Biliary Atresia/metabolism , Biliary Atresia/pathology , Cholestasis/metabolism , Cholestasis/pathology , Female , Humans , Immunohistochemistry , Infant , Liver/metabolism , Liver/pathology , Male , Retrospective Studies
3.
Development ; 145(12)2018 06 08.
Article in English | MEDLINE | ID: mdl-29769220

ABSTRACT

Retinoic acid (RA) is required for pancreas specification in Xenopus and other vertebrates. However, the gene network that is directly induced by RA signalling in this context remains to be defined. By RNA sequencing of in vitro-generated pancreatic explants, we identified the genes encoding the transcription factor Hnf1ß and the Wnt-receptor Fzd4/Fzd4s as direct RA target genes. Functional analyses of Hnf1b and Fzd4/Fzd4s in programmed pancreatic explants and whole embryos revealed their requirement for pancreatic progenitor formation and differentiation. Thus, Hnf1ß and Fzd4/Fzd4s appear to be involved in pre-patterning events of the embryonic endoderm that allow pancreas formation in Xenopus.


Subject(s)
Frizzled Receptors/biosynthesis , Hepatocyte Nuclear Factor 1-beta/biosynthesis , Organogenesis/genetics , Pancreas/embryology , Tretinoin/metabolism , Xenopus Proteins/biosynthesis , Xenopus laevis/embryology , Animals , Cell Differentiation/genetics , Frizzled Receptors/genetics , Gene Expression Regulation, Developmental/genetics , Gene Knockdown Techniques , Hepatocyte Nuclear Factor 1-beta/genetics , Morpholinos/genetics , Wnt Signaling Pathway/genetics , Xenopus Proteins/genetics
4.
Development ; 144(24): 4530-4539, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29113990

ABSTRACT

Notch signaling plays important roles during mammalian nephrogenesis. To investigate whether Notch regulates nephron segmentation, we performed Notch loss-of-function and gain-of-function studies in developing nephrons in mice. Contrary to the previous notion that Notch signaling promotes the formation of proximal tubules and represses the formation of distal tubules in the mammalian nephron, we show that inhibition of Notch blocks the formation of all nephron segments and that constitutive activation of Notch in developing nephrons does not promote or repress the formation of a specific segment. Cells lacking Notch fail to form the S-shaped body and show reduced expression of Lhx1 and Hnf1b Consistent with this, we find that constitutive activation of Notch in mesenchymal nephron progenitors causes ectopic expression of Lhx1 and Hnf1b and that these cells eventually form a heterogeneous population that includes proximal tubules and other types of cells. Our data suggest that Notch signaling is required for the formation of all nephron segments and that it primes nephron progenitors for differentiation rather than directing their cell fates into a specific nephron segment.


Subject(s)
Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Kidney Tubules, Proximal/embryology , Organogenesis/physiology , Receptors, Notch/metabolism , Animals , Cell Differentiation , Enzyme Activation/genetics , Hepatocyte Nuclear Factor 1-beta/biosynthesis , LIM-Homeodomain Proteins/biosynthesis , Mice , Mice, Transgenic , Receptors, Notch/genetics , Signal Transduction/physiology , Transcription Factors/biosynthesis , Wnt4 Protein/metabolism
5.
Tumour Biol ; 37(4): 4813-29, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26520442

ABSTRACT

Chemoresistance to platinum-based antineoplastic agents is a consistent feature among ovarian carcinomas; however, whereas high-grade serous carcinoma (OSC) acquires resistance during chemotherapy, ovarian clear cell carcinoma (OCCC) is intrinsically resistant. The main objective of this study was to explore, in vitro and in vivo, if hepatocyte nuclear factor 1ß (HNF1ß) and glutaminolysis contribute for the resistance of OCCC to carboplatin through the intrinsically increased GSH bioavailability. To disclose the role of HNF1ß, experiments were also performed in an OSC cell line, which does not express HNF1ß. Metabolic profiles, GSH quantification, HNF1ß, and γ-glutamylcysteine ligase catalytic subunit (GCLC) and modifier subunit (GCLM) expression, cell cycle, and death were assessed in ES2 cell line (OCCC) and OVCAR3 cell line (OSC); HNF1ß knockdown was performed in ES2 and murine model of subcutaneous and peritoneal OCCC tumors was established to test buthionine sulphoxamine (BSO), as a sensitizer to carboplatin. Glutaminolysis is activated in ES2 and OVCAR3, though ES2 exclusively synthesizes amino acids and GSH. ES2 cells are more resistant to carboplatin than OVCAR3 and the abrogation of GSH production by BSO sensitizes ES2 to carboplatin. HNF1ß regulates the expression of GCLC, but not GCLM, and consequently GSH production in ES2. In vivo, BSO prior to carboplatin reduces dramatically subcutaneous tumor size and GSH levels, as well as peritoneal dissemination. Our study discloses HNF1ß as the mediator of intrinsic OCCC chemoresistance and sheds a light to re-explore a cancer adjuvant therapeutic approach using BSO to overcome the lack of efficient therapy in OCCC.


Subject(s)
Adenocarcinoma, Clear Cell/drug therapy , Glutamate-Cysteine Ligase/biosynthesis , Glutamate-Cysteine Ligase/blood , Hepatocyte Nuclear Factor 1-beta/biosynthesis , Ovarian Neoplasms/drug therapy , Adenocarcinoma, Clear Cell/genetics , Adenocarcinoma, Clear Cell/pathology , Animals , Carboplatin/administration & dosage , Cell Line, Tumor , Cisplatin/administration & dosage , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Glutamate-Cysteine Ligase/genetics , Glutathione/biosynthesis , Hepatocyte Nuclear Factor 1-beta/genetics , Humans , Mice , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Xenograft Model Antitumor Assays
6.
J Gastrointest Surg ; 18(10): 1784-94, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25052070

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) frequently exhibits biliary differentiation, which is typically overlooked. Hepatocyte nuclear factor 1ß (HNF1ß), a bile duct-specific transcription factor expressed in bile ducts but not in the normal hepatocytes, is also expressed in HCC. MATERIALS AND METHODS: The expression of HNF1ß and the biliary differentiation marker cytokeratin 19 (CK19) were retrospectively evaluated using immunohistochemistry in 159 surgically resected primary HCCs. RESULTS: A significant correlation was observed between HNF1ß protein expression and younger age (p = 0.0293), high serum α-fetoprotein levels (p = 6 × 10(-4)), and high tumor grade (p = 0.0255). However, HNF1ß expression exhibited no correlation with tumor stage. Patients with HCCs and HNF1ß expression were more likely to exhibit early tumor recurrence (ETR; p = 0.0048) and a lower 5-year survival rate (p = 0.0001). A multivariate analysis indicated HNF1ß expression as an independent prognostic factor in HCC (p = 0.0048). A combinatorial analysis revealed additive adverse effects of HNF1ß when concomitant with CK19 expression and p53 mutation. Furthermore, HNF1ß expression can predict poor prognosis in patients with ETR. CONCLUSION: Our results indicated that HNF1ß expression is a crucial predictor of poor prognosis in HCC and is independent of tumor stage. Moreover, concomitant HNF1ß and CK19 expressions exhibited additive adverse effects in HCC, confirming that HCC with biliary differentiation has a poor prognosis.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Hepatocyte Nuclear Factor 1-beta/biosynthesis , Liver Neoplasms/metabolism , Neoplasm Recurrence, Local/metabolism , Neoplasm Staging/methods , Adolescent , Adult , Aged, 80 and over , Biomarkers, Tumor/biosynthesis , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/surgery , Female , Follow-Up Studies , Hepatectomy , Humans , Immunohistochemistry , Incidence , Keratin-19/biosynthesis , Liver Neoplasms/diagnosis , Liver Neoplasms/surgery , Male , Middle Aged , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/epidemiology , Prognosis , Retrospective Studies , Survival Rate/trends , Taiwan/epidemiology , Time Factors , Young Adult
7.
Gene ; 518(2): 246-55, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23370336

ABSTRACT

Human embryonic stem (hES) cells with the capacity of self-renewal and multilineage differentiation are promising sources for generation of pancreatic islet cells for cell replacement therapy in diabetes. Here we induced hES cells into insulin-producing cells (IPCs) in a stepwise process which recapitulated islet organogenesis by directing cells through the stages resembling definitive endoderm, gut-tube endoderm, pancreatic precursor and cells that expressed pancreatic endocrine hormones. The dynamic expression of microRNAs (miRNAs) during the differentiation was analyzed and was compared with that in the development of human pancreatic islets. We found that the dynamic expression patterns of miR-375 and miR-7 were similar to those seen in the development of human fetal pancreas, whereas the dynamic expression of miR-146a and miR-34a showed specific patterns during the differentiation. Furthermore, the expression of Hnf1ß and Pax6, the predicted target genes of miR-375 and miR-7, was reciprocal to that of miR-375 and miR-7. Over-expression of miR-375 down-regulated the expression of gut-endoderm/pancreatic progenitor specific markers Hnf1ß and Sox9. Therefore, the miRNAs may directly or indirectly regulate the expression of pancreatic islet organogenesis-specific transcription factors to control the differentiation and maturation of pancreatic islet cells.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/metabolism , Insulin-Secreting Cells/metabolism , MicroRNAs/genetics , Embryonic Stem Cells/cytology , Eye Proteins/biosynthesis , Eye Proteins/genetics , Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 1-beta/biosynthesis , Hepatocyte Nuclear Factor 1-beta/genetics , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Insulin/biosynthesis , Islets of Langerhans/metabolism , MicroRNAs/biosynthesis , PAX6 Transcription Factor , Paired Box Transcription Factors/biosynthesis , Paired Box Transcription Factors/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , SOX9 Transcription Factor/biosynthesis , SOX9 Transcription Factor/genetics
8.
Am J Physiol Renal Physiol ; 303(5): F689-99, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22759397

ABSTRACT

Hepatocyte nuclear factor-1ß (HNF-1ß) is a transcription factor expressed in the kidney, liver, pancreas, and other organs. Mutations of HNF-1ß cause maturity-onset diabetes of the young type 5 (MODY5). The aims of this study were to investigate the functional roles of the HNF-1ß/suppressor of cytokine signaling-3 (SOCS-3) pathway in tubule damage after acute kidney injury (AKI) both in vivo and in vitro and to examine the effect of HNF-1ß on renal tubule formation. To clarify the significance of the HNF-1ß/SOCS-3 pathway in AKI, we used a rat ischemia/reperfusion (I/R) AKI model and cultured renal tubular cells (NRK-52E cells). Western blot analysis showed that HNF-1ß and polycystic kidney disease 2 (PKD2) expressions were increased at 3-12 h and 12-24 h after I/R, respectively. The expression level of SOCS-3 was decreased at 3-48 h. Immunohistological examination revealed that expression of HNF-1ß was increased in proximal tubules. Overexpression of HNF-1ß resulted in decreased SOCS-3 expression, activation of signal transducer and activator of transcription 3 (STAT3) and Erk, and increased [(3)H]thymidine uptake in the presence of hepatocyte growth factor. Furthermore, tubule formation in three-dimensional gels was inhibited by dominant-negative HNF-1ß. Our study shows that HNF-1ß is upregulated after AKI in proximal tubular cells and that HNF-1ß controls cellular proliferation and tubule formation by regulating SOCS-3 expression and STAT3/Erk activation. Therefore, the current study unravels the physiological and pathological significance of the HNF-1ß pathway in AKI.


Subject(s)
Acute Kidney Injury/physiopathology , Hepatocyte Nuclear Factor 1-beta/genetics , Kidney Tubules/physiology , Regeneration , Suppressor of Cytokine Signaling Proteins/physiology , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Hepatocyte Nuclear Factor 1-beta/biosynthesis , Male , Rats , Rats, Sprague-Dawley , Reperfusion Injury/physiopathology , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/biosynthesis , TRPP Cation Channels/biosynthesis , Up-Regulation
9.
Proc Natl Acad Sci U S A ; 109(28): 11252-7, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22730461

ABSTRACT

One of the central goals of human genetics is to discover the genes and pathways driving human traits. To date, most of the common risk alleles discovered through genome-wide association studies (GWAS) map to nonprotein-coding regions. Because of our relatively poorer understanding of this part of the genome, the functional consequences of trait-associated variants pose a considerable challenge. To identify the genes through which risk loci act, we hypothesized that the risk variants are regulatory elements. For each of 12 known risk polymorphisms, we evaluated the correlation between risk allele status and transcript abundance for all annotated protein-coding transcripts within a 1-Mb interval. A total of 103 transcripts were evaluated in 662 prostate tissue samples [normal (n = 407) and tumor (n = 255)] from 483 individuals [European Americans (n = 233), Japanese (n = 127), and African Americans (n = 123)]. In a pooled analysis, 4 of the 12 risk variants were strongly associated with five transcripts (NUDT11, MSMB, NCOA4, SLC22A3, and HNF1B) in histologically normal tissue (P ≤ 0.001). Although associations were also observed in tumor tissue, they tended to be more attenuated. Previously, we showed that MSMB and NCOA4 participate in prostate cancer pathogenesis. Suppressing the expression of NUDT11, SLC22A3, and HNF1B influences cellular phenotypes associated with tumor-related properties in prostate cancer cells. Taken together, the data suggest that these transcripts contribute to prostate cancer pathogenesis.


Subject(s)
Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 1-beta/biosynthesis , Organic Cation Transport Proteins/biosynthesis , Prostatic Neoplasms/metabolism , Pyrophosphatases/biosynthesis , Alleles , Gene Expression Profiling , Genome-Wide Association Study , Humans , Male , Models, Genetic , Phenotype , Polymorphism, Genetic , Polymorphism, Single Nucleotide , Prostatic Neoplasms/genetics , Quantitative Trait Loci , Risk
10.
Hum Pathol ; 43(10): 1618-26, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22436624

ABSTRACT

The nature of "piling up" proliferation of clear cells in müllerian mucinous/mixed borderline tumor has not been well characterized. The purpose of this study was to clarify whether or not such clear cells represent concomitant clear cell neoplasms. First, we carefully reviewed hematoxylin and eosin slides taken from 139 ovarian tumors diagnosed as clear cell carcinoma (112 cases) and müllerian mucinous/mixed borderline tumor (27 cases) to clarify (1) the frequency of piling-up clear cells in müllerian mucinous/mixed borderline tumor and (2) the frequency of the coexistence of typical clear cell carcinoma and müllerian mucinous/mixed borderline tumor. Second, we investigated the immunohistochemical expression of estrogen receptor, hepatocyte nuclear factor-1ß, and glypican-3 in proliferating clear cells in both tumors. We identified piling-up clear cells in 56% of müllerian mucinous/mixed borderline tumors. Such clear cells lacked the severe nuclear atypia, complex branching, and dense hyalinized cores of typical clear cell carcinoma. We did not find coexistence of typical clear cell carcinoma and müllerian mucinous/mixed borderline tumor in any tumors. Piling-up clear cells and endocervical-like mucinous cells were positive for estrogen receptor but negative for hepatocyte nuclear factor-1ß and glypican-3. Most clear cell carcinomas showed a hepatocyte nuclear factor-1ß-positive/estrogen receptor-negative immunophenotype, and about half of them were glypican-3 positive. In conclusion, piling-up clear cells in müllerian mucinous/mixed borderline tumor do not represent concomitant clear cell neoplasms because clear cell carcinoma and müllerian mucinous/mixed borderline tumor hardly ever coexist and because such clear cells in both tumors are immunophenotypically distinct.


Subject(s)
Adenocarcinoma, Clear Cell/pathology , Adenocarcinoma, Mucinous/pathology , Ovarian Neoplasms/pathology , Adenocarcinoma, Clear Cell/metabolism , Adenocarcinoma, Mucinous/metabolism , Female , Glypicans/biosynthesis , Hepatocyte Nuclear Factor 1-beta/biosynthesis , Humans , Immunohistochemistry , Neoplasms, Multiple Primary/diagnosis , Neoplasms, Multiple Primary/metabolism , Ovarian Neoplasms/metabolism , Receptors, Estrogen/biosynthesis
11.
Hum Pathol ; 42(11): 1613-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21496868

ABSTRACT

The histogenesis of clear cell adenocarcinoma of the bladder/urethra is uncertain. Hepatocyte nuclear factor-1ß is a homeodomain protein that has been reported to be frequently overexpressed in ovarian clear cell adenocarcinoma in comparison with rare or no expression in other types of epithelial ovarian tumors. We assessed the expression of hepatocyte nuclear factor-1ß in a series of 18 clear cell adenocarcinomas of the bladder and urethra and compared it with that of invasive high-grade transitional/urothelial carcinoma (n = 35); adenocarcinomas of the bladder, urethra, and paraurethral glands (n = 21); as well as nephrogenic adenomas of the bladder (n = 8). Staining intensity and extent were evaluated using a 4-tiered grading system (0-3). A case was considered positive for hepatocyte nuclear factor-1ß if 10% or more of tumor cells showed at least weak nuclear staining or if any moderate or strong nuclear staining was observed. All 18 clear cell adenocarcinomas exhibited nuclear staining in at least 50% of tumor cells (16 strong, 1 moderate, and 1 weak with focal strong nuclear staining) in comparison with positive nuclear staining (moderate) in 1 of 21 bladder adenocarcinoma, 1 of 35 invasive high-grade transitional/urothelial carcinoma (weak to moderate staining), and 2 of 8 nephrogenic adenomas (1 weak and 1 moderate to strong staining). We concluded that hepatocyte nuclear factor-1ß is a useful marker in differentiating clear cell adenocarcinomas of the bladder/urethra from invasive high-grade transitional/urothelial carcinoma and other types of bladder adenocarcinomas and to a lesser extent from nephrogenic adenomas. Hepatocyte nuclear factor-1ß is of no diagnostic utility in discriminating primary bladder/urethral clear cell adenocarcinomas from metastatic clear cell adenocarcinomas of the female genital tract to the bladder/urethra. From a histogenesis standpoint, although the expression of hepatocyte nuclear factor-1ß in both gynecologic and urologic tract clear cell adenocarcinomas may point to a Müllerian derivation/differentiation, this immunohistochemical evidence is insufficient to completely exclude an urothelial association.


Subject(s)
Adenocarcinoma, Clear Cell/metabolism , Hepatocyte Nuclear Factor 1-beta/biosynthesis , Urethral Neoplasms/metabolism , Urinary Bladder Neoplasms/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma, Clear Cell/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Female , Humans , Immunohistochemistry , Male , Middle Aged , Urethra/metabolism , Urethra/pathology , Urethral Neoplasms/pathology , Urinary Bladder/metabolism , Urinary Bladder Neoplasms/pathology , Urothelium/metabolism
12.
Urology ; 76(2): 507.e6-11, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20538322

ABSTRACT

OBJECTIVES: The aim of this study was to identify new prognostic factors in metastatic renal cell carcinoma (RCC) based on the analysis of precisely defined metastatic tissue. METHODS: Expression profiling was done on 26 snap-frozen samples of clear-cell RCC metastases with complete follow-up (up to 116 months) using laser microdissection and oligonucleotide microarrays (Affymetrix). A prognosis-associated gene signature was determined using the semi-supervised principal components analysis method. Validation was performed with quantitative RT-PCR on samples of normal renal tissue (n = 6), RCC primary tumor (n = 57), and RCC metastases (n = 59). Immunohistochemistry (IHC) was done to localize HNF-1B. RESULTS: Analysis of expression data revealed a three-gene signature consisting of HNF-1B, KIAA1919, and SYDE1, which discriminated well between 2 prognosis groups (P < .05), independently of the TNMG classification. Expression of HNF-1B was analyzed in detail. HNF-1B mRNA expression correlated with malignant transformation and progression (normal renal tissue > primary tumor > metastasis; P < .0001). There was a significant correlation between high HNF-1B mRNA expression in primary tumor and better prognosis (P < .05). IHC showed a specific nuclear HNF-1B staining confined to the tumor cells of the primary tumors and of the metastases. CONCLUSIONS: The level of HNF-1B mRNA expression significantly decreases with tumor progression, and patients with high HNF-1B mRNA levels have a significantly better prognosis. HNF-1B might be a useful prognostic factor for metastatic RCC and also a potential therapeutic target in the future.


Subject(s)
Carcinoma, Renal Cell/secondary , Down-Regulation , Hepatocyte Nuclear Factor 1-beta/physiology , Kidney Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 1-beta/biosynthesis , Hepatocyte Nuclear Factor 1-beta/genetics , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Male , Middle Aged , Prognosis
13.
PLoS One ; 4(11): e7855, 2009 Nov 16.
Article in English | MEDLINE | ID: mdl-19924231

ABSTRACT

BACKGROUND: The HNF1A, HNF1B and HNF4A genes are part of an autoregulatory network in mammalian pancreas, liver, kidney and gut. The layout of this network appears to be similar in rodents and humans, but inactivation of HNF1A, HNF1B or HNF4A genes in animal models cause divergent phenotypes to those seen in man. We hypothesised that some differences may arise from variation in the expression profile of alternatively processed isoforms between species. METHODOLOGY/PRINCIPAL FINDINGS: We measured the expression of the major isoforms of the HNF1A, HNF1B and HNF4A genes in human and rodent pancreas, islet, liver and kidney by isoform-specific quantitative real-time PCR and compared their expression by the comparative Ct (DeltaDeltaCt) method. We found major changes in the expression profiles of the HNF genes between humans and rodents. The principal difference lies in the expression of the HNF1A gene, which exists as three isoforms in man, but as a single isoform only in rodents. More subtle changes were to the balance of HNF1B and HNF4A isoforms between species; the repressor isoform HNF1B(C) comprised only 6% in human islets compared with 24-26% in rodents (p = 0.006) whereas HNF4A9 comprised 22% of HNF4A expression in human pancreas but only 11% in rodents (p = 0.001). CONCLUSIONS/SIGNIFICANCE: The differences we note in the isoform-specific expression of the human and rodent HNF1A, HNF1B and HNF4A genes may impact on the absolute activity of these genes, and therefore on the activity of the pancreatic transcription factor network as a whole. We conclude that alterations to expression of HNF isoforms may underlie some of the phenotypic variation caused by mutations in these genes.


Subject(s)
Gene Expression Regulation , Hepatocyte Nuclear Factor 1-alpha/biosynthesis , Hepatocyte Nuclear Factor 1-beta/biosynthesis , Hepatocyte Nuclear Factor 4/biosynthesis , Adult , Alternative Splicing , Animals , Female , Gene Expression Profiling , Humans , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred BALB C , Middle Aged , Rats , Rats, Wistar , Species Specificity
14.
Exp Clin Endocrinol Diabetes ; 115(10): 654-61, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18058600

ABSTRACT

Hepatocyte nuclear factor-6 (HNF-6) is the ONECUT-homeodomain transcription factor that is enriched in liver and also present in pancreas and central nervous system. It is expressed in the pancreatic bud at E10.5. In adult pancreas, its expression is restricted to the exocrine pancreas and duct cells. Since duct cells are thought to be precursors of endocrine cells and HNF-6 is involved in the regulation of the expression of HNF-4alpha and -1beta, genes that cause maturity onset diabetes of the young (MODY), we hypothesized that the sustained expression of HNF-6 would affect beta-cell function. We generated transgenic mice over-expressing human HNF-6 using the mouse insulin I promoter (MIP). We obtained one female founder in which the transgene had been incorporated into two sites; the chromosome (Ch) 14 and the X chromosome. The integration site of the latter was within centromeric heterochromatin and the transgene was inactivated. Studies on mice in which the transgene was integrated into Ch14 showed beta-cell specific defects functionally and pathologically. The insulin secretory response to glucose and arginine in the in situ-perfused pancreas was also significantly impaired in these mice. Immunohistochemical analysis revealed that the islets were smaller and had an abnormal architecture with an inverted ratio of alpha- and beta-cells resulting from beta-cell loss to 30% by 6-wk of age. The decreased number of beta-cells was quantified first time by fluorescent activated cell sorting using entire pancreata from the transgenic mice crossed with MIP-green fluorescent protein (GFP) mice. This severe loss of beta-cells involved programmed cell death.


Subject(s)
Apoptosis/genetics , Gene Expression Regulation/genetics , Hepatocyte Nuclear Factor 6/biosynthesis , Insulin-Secreting Cells/metabolism , Transgenes , Animals , Central Nervous System/metabolism , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Female , Glucose/pharmacology , Hepatocyte Nuclear Factor 1-beta/biosynthesis , Hepatocyte Nuclear Factor 4/biosynthesis , Hepatocyte Nuclear Factor 6/genetics , Heterochromatin/genetics , Heterochromatin/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/pathology , Liver/metabolism , Male , Mice , Mice, Transgenic , Organ Specificity/physiology , Sweetening Agents/pharmacology , X Chromosome/genetics , X Chromosome/metabolism
15.
Mol Pharmacol ; 72(6): 1619-25, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17855651

ABSTRACT

Expression of Urate transporter 1 (URAT1/SLC22A12) is restricted to the proximal tubules in the kidney, where it is responsible for the tubular reabsorption of urate. To elucidate the mechanism underlying its tissue-specific expression, the transcriptional regulation of the hURAT1 and mUrat1 genes was investigated. Hepatocyte nuclear factor 1 alpha (HNF1alpha) and HNF1beta positively regulate minimal promoter activity of the URAT1 gene as shown by reporter gene assays. Electrophoretic mobility shift assays revealed binding of HNF1alpha and/or HNF1beta to the HNF1 motif in the hURAT1 promoter. Furthermore, the mRNA expression of Urat1 is reduced in the kidneys of Hnf1alpha-null mice compared with wild-type mice, confirming the indispensable role of HNF1alpha in the constitutive expression of URAT1 genes. It was also shown that the proximal promoter region of mUrat1 was hypermethylated in the liver and kidney medulla, whereas this region was relatively hypomethylated in the kidney cortex. These methylation profiles are in a good agreement with the proximal tubule-restricted expression of mUrat1 in the kidney cortex. Taken together, these results strongly suggest that tissue-specific expression of the URAT1 genes is coordinately regulated by the transcriptional activation by HNF1alpha/HNF1beta heterodimer and repression by DNA methylation.


Subject(s)
DNA Methylation , Hepatocyte Nuclear Factor 1-alpha/physiology , Hepatocyte Nuclear Factor 1-beta/physiology , Organic Anion Transporters/biosynthesis , Organic Cation Transport Proteins/biosynthesis , Animals , Base Sequence , Cell Line , Female , Gene Expression Regulation/physiology , Hepatocyte Nuclear Factor 1-alpha/biosynthesis , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-beta/biosynthesis , Hepatocyte Nuclear Factor 1-beta/genetics , Humans , Male , Mice , Mice, Knockout , Molecular Sequence Data , Organic Anion Transporters/genetics , Organic Cation Transport Proteins/genetics , Tissue Distribution/genetics , Transcriptional Activation/genetics
16.
Biochem Biophys Res Commun ; 353(3): 617-22, 2007 Feb 16.
Article in English | MEDLINE | ID: mdl-17194452

ABSTRACT

Sucrase-isomaltase (SI) gene expression is negatively regulated by glucose, but its molecular mechanism is not completely clear. The purpose of this study is to investigate whether HNF-1alpha and HNF-1beta contribute to glucose regulation of SI gene expression. To explore this question, we examined the association of gene expressions between SI and HNF-1alpha and HNF-1beta in Caco-2 cells cultured in medium containing 2.0 and 16.7 mM glucose. We found that gene expression of HNF-1alpha but not HNF-1beta exhibits a positive correlation with that of SI regulated by glucose. Moreover, to elucidate whether glucose regulation of SI gene expression is changed when HNF-1alpha and HNF-1beta are inhibited, we produced three stable cell lines, in which dominant-negative mutant HNF-1alphaT539fsdelC, mutant HNF-1betaR177X, and empty vector (as a control), respectively, were stably expressed. We found that the glucose regulation of SI gene expression was significantly attenuated in HNF-1alphaT539fsdelC cells, but it was well maintained in empty vector and HNF-1betaR177X cells. These results suggest that HNF-1alpha participates in glucose regulation of SI gene expression.


Subject(s)
Glucose/pharmacology , Hepatocyte Nuclear Factor 1-alpha/physiology , Hepatocyte Nuclear Factor 1-beta/physiology , Sucrase-Isomaltase Complex/biosynthesis , Caco-2 Cells , Enzyme Repression , Epithelial Cells/enzymology , Gene Expression Regulation, Enzymologic , Hepatocyte Nuclear Factor 1-alpha/biosynthesis , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-beta/biosynthesis , Hepatocyte Nuclear Factor 1-beta/genetics , Humans , Mutation , Sucrase-Isomaltase Complex/genetics , Transfection
17.
Br J Cancer ; 94(6): 914-21, 2006 Mar 27.
Article in English | MEDLINE | ID: mdl-16479257

ABSTRACT

Transcription factor 2 gene (TCF2) encodes hepatocyte nuclear factor 1beta (HNF1beta), a transcription factor associated with development and metabolism. Mutation of TCF2 has been observed in renal cell cancer, and by screening aberrantly methylated genes, we have now identified TCF2 as a target for epigenetic inactivation in ovarian cancer. TCF2 was methylated in 53% of ovarian cancer cell lines and 26% of primary ovarian cancers, resulting in loss of the gene's expression. TCF2 expression was restored by treating cells with a methyltransferase inhibitor, 5-aza-2'deoxycitidine (5-aza-dC). In addition, chromatin immunoprecipitation showed deacetylation of histone H3 in methylated cells and, when combined with 5-aza-dC, the histone deacetylase inhibitor trichostatin A synergistically induced TCF2 expression. Epigenetic inactivation of TCF2 was also seen in colorectal, gastric and pancreatic cell lines, suggesting general involvement of epigenetic inactivation of TCF2 in tumorigenesis. Restoration of TCF2 expression induced expression of HNF4alpha, a transcriptional target of HNF1beta, indicating that epigenetic silencing of TCF2 leads to alteration of the hepatocyte nuclear factor network in tumours. These results suggest that TCF2 is involved in the development of ovarian cancers and may represent a useful target for their detection and treatment.


Subject(s)
Hepatocyte Nuclear Factor 1-beta/biosynthesis , Hepatocyte Nuclear Factor 1-beta/metabolism , Ovarian Neoplasms/genetics , Base Sequence , DNA Methylation , Epigenesis, Genetic , Female , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/pathology , Gene Expression Profiling , Humans , Molecular Sequence Data , Ovarian Neoplasms/pathology , Tumor Cells, Cultured
18.
Mod Pathol ; 19(1): 83-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16258507

ABSTRACT

Clear cell tumors of the ovary are frequently associated with ovarian endometriosis. Clinicopathologically, it has been suggested that clear cell tumors develop from endometriosis, but there has been little molecular evidence supporting this speculation. Microarray analysis revealed recently that hepatocyte nuclear factor-1beta (HNF-1beta) was significantly upregulated in clear cell carcinoma of the ovary. In the present study, we examined 30 clear cell tumors (26 malignant, three borderline, and one benign) and 40 endometriotic cysts to clarify if differentiation into the clear cell lineage already begins in ovarian endometriosis. All of the 30 clear cell tumors, including borderline and benign ones, showed immunohistochemical expression of HNF-1beta in the nucleus, while other types of ovarian epithelial tumors (endometrioid, serous, mucinous, and Brenner tumors) rarely expressed it. Among 30 clear cell tumors, 17 (56%) cases were associated with endometriosis, and endometriotic epithelium was identified in 12 cases. In nine of the 12 cases, distinct nuclear immunostaining for HNF-1beta was detected in the endometriotic epithelium, as well as in the clear cell tumor. HNF-1beta expression was observed either in atypical endometriosis (four cases), or in endometriosis of a reactive nature (five cases). Furthermore, 16 of 40 (40%) endometriotic cysts without a neoplasm also expressed HNF-1beta, and the expression was almost exclusively observed in the epithelium showing inflammatory atypia. Our results indicate that HNF-1beta is an excellent molecular marker for ovarian clear cell tumors, including benign, borderline and malignant lesions. Early differentiation into the clear cell lineage takes place in ovarian endometriosis, not only in atypical endometriosis, but also in endometriosis with degenerative and regenerative changes, and this is probably responsible for the frequent occurrence of clear cell carcinoma in ovarian endometriosis.


Subject(s)
Adenocarcinoma, Clear Cell/pathology , Endometriosis/pathology , Hepatocyte Nuclear Factor 1-beta/biosynthesis , Ovarian Diseases/pathology , Ovarian Neoplasms/pathology , Adenocarcinoma, Clear Cell/metabolism , Adult , Aged , Endometriosis/metabolism , Epithelial Cells/chemistry , Epithelial Cells/pathology , Female , Humans , Immunohistochemistry , Middle Aged , Ovarian Cysts/metabolism , Ovarian Cysts/pathology , Ovarian Diseases/metabolism , Ovarian Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...