Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biol Reprod ; 101(4): 686-694, 2019 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-31322170

RESUMO

The overexpression of hepatocyte nuclear factor-1 beta (HNF1ß) in endometriotic lesion has been demonstrated. However, the role of HNF1ß in endometriosis remains largely unknown. Human endometriotic 12Z cells showed higher level of HNF1ß when compared with normal endometrial HES cells. In human endometriotic 12Z cells, HNF1ß knockdown increased susceptibility to apoptotic cell death by oxidative stress, while HNF1ß overexpression suppressed apoptosis. In addition, HNF1ß knockdown and overexpression significantly decreased and increased, respectively, the expression of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)-dependent antiapoptotic genes. Knockdown of the antiapoptotic genes significantly reduced the HNF1ß-induced resistance against oxidative stress in 12Z cells. Furthermore, HNF1ß regulated the transcriptional activity of NF-κB, and an NF-κB inhibitor suppressed the HNF1ß-enhanced NF-κB-dependent antiapoptotic gene expression and the resistance of the 12Z cells against cell death. Taken together, these data suggest that HNF1ß overexpression may protect endometriotic cells against oxidative damage by augmenting antiapoptotic gene expression.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Apoptose/genética , Endometriose/patologia , Endométrio/patologia , Fator 1-beta Nuclear de Hepatócito/fisiologia , Proteínas Reguladoras de Apoptose/metabolismo , Proliferação de Células/genética , Células Cultivadas , Endometriose/genética , Endométrio/metabolismo , Endométrio/fisiologia , Feminino , Humanos , Estresse Oxidativo/genética , Regulação para Cima/genética
2.
Am J Physiol Renal Physiol ; 317(2): F478-F488, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31188030

RESUMO

The hepatocyte nuclear factor-1ß (Hnf1b) transcription factor is a key regulator of kidney tubule formation and is associated with a syndrome of renal cysts and early onset diabetes. To further our understanding of Hnf1b in the developing zebrafish kidney, we performed RNA sequencing analysis of proximal tubules from hnf1b-deficient larvae. This analysis revealed an enrichment of gene transcripts encoding transporters of the solute carrier (SLC) superfamily, including multiple members of slc2 and slc5 glucose transporters. An investigation of expression of slc2a1a, slc2a2, and slc5a2 as well as a poorly studied glucose/mannose transporter encoded by slc5a9 revealed that these genes undergo dynamic spatiotemporal changes during tubule formation and maturation. A comparative analysis of zebrafish SLC genes with those expressed in mouse proximal tubules showed a substantial overlap at the level of gene families, indicating a high degree of functional conservation between zebrafish and mammalian proximal tubules. Taken together, our findings are consistent with a role for Hnf1b as a critical determinant of proximal tubule transport function by acting upstream of a large number of SLC genes and validate the zebrafish as a physiologically relevant model of the mammalian proximal tubule.


Assuntos
Perfilação da Expressão Gênica , Fator 1-beta Nuclear de Hepatócito/genética , Fator 1-beta Nuclear de Hepatócito/fisiologia , Túbulos Renais Proximais/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Larva , Camundongos , RNA/biossíntese , RNA/genética , Especificidade da Espécie
3.
Proc Natl Acad Sci U S A ; 115(21): E4910-E4919, 2018 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-29735694

RESUMO

Mitochondrial dysfunction is increasingly recognized as a critical determinant of both hereditary and acquired kidney diseases. However, it remains poorly understood how mitochondrial metabolism is regulated to support normal kidney function and how its dysregulation contributes to kidney disease. Here, we show that the nuclear receptor estrogen-related receptor gamma (ERRγ) and hepatocyte nuclear factor 1 beta (HNF1ß) link renal mitochondrial and reabsorptive functions through coordinated epigenomic programs. ERRγ directly regulates mitochondrial metabolism but cooperatively controls renal reabsorption via convergent binding with HNF1ß. Deletion of ERRγ in renal epithelial cells (RECs), in which it is highly and specifically expressed, results in severe renal energetic and reabsorptive dysfunction and progressive renal failure that recapitulates phenotypes of animals and patients with HNF1ß loss-of-function gene mutations. Moreover, ERRγ expression positively correlates with renal function and is decreased in patients with chronic kidney disease (CKD). REC-ERRγ KO mice share highly overlapping renal transcriptional signatures with human patients with CKD. Together these findings reveal a role for ERRγ in directing independent and HNF1ß-integrated programs for energy production and use essential for normal renal function and the prevention of kidney disease.


Assuntos
Cistos/prevenção & controle , Metabolismo Energético , Epigenômica , Regulação da Expressão Gênica , Fator 1-beta Nuclear de Hepatócito/genética , Receptores de Estrogênio/genética , Insuficiência Renal Crônica/prevenção & controle , Animais , Cistos/metabolismo , Cistos/patologia , Fator 1-beta Nuclear de Hepatócito/metabolismo , Fator 1-beta Nuclear de Hepatócito/fisiologia , Humanos , Rim/metabolismo , Rim/patologia , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Regiões Promotoras Genéticas , Receptores de Estrogênio/metabolismo , Receptores de Estrogênio/fisiologia , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia
4.
J Am Soc Nephrol ; 28(11): 3205-3217, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28739648

RESUMO

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


Assuntos
Injúria Renal Aguda/metabolismo , Fator 1-beta Nuclear de Hepatócito/fisiologia , Túbulos Renais Proximais/metabolismo , Mitocôndrias/metabolismo , Injúria Renal Aguda/etiologia , Adulto , Animais , Fator 1-beta Nuclear de Hepatócito/antagonistas & inibidores , Fator 1-beta Nuclear de Hepatócito/genética , Humanos , Camundongos Endogâmicos C57BL , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/fisiologia
5.
J Am Soc Nephrol ; 28(10): 2887-2900, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28507058

RESUMO

The transcription factor hepatocyte nuclear factor-1ß (HNF-1ß) is essential for normal kidney development and function. Inactivation of HNF-1ß in mouse kidney tubules leads to early-onset cyst formation and postnatal lethality. Here, we used Pkhd1/Cre mice to delete HNF-1ß specifically in renal collecting ducts (CDs). CD-specific HNF-1ß mutant mice survived long term and developed slowly progressive cystic kidney disease, renal fibrosis, and hydronephrosis. Compared with wild-type littermates, HNF-1ß mutant mice exhibited polyuria and polydipsia. Before the development of significant renal structural abnormalities, mutant mice exhibited low urine osmolality at baseline and after water restriction and administration of desmopressin. However, mutant and wild-type mice had similar plasma vasopressin and solute excretion levels. HNF-1ß mutant kidneys showed increased expression of aquaporin-2 mRNA but mislocalized expression of aquaporin-2 protein in the cytoplasm of CD cells. Mutant kidneys also had decreased expression of the UT-A urea transporter and collectrin, which is involved in apical membrane vesicle trafficking. Treatment of HNF-1ß mutant mIMCD3 cells with hypertonic NaCl inhibited the induction of osmoregulated genes, including Nr1h4, which encodes the transcription factor FXR that is required for maximal urinary concentration. Chromatin immunoprecipitation and sequencing experiments revealed HNF-1ß binding to the Nr1h4 promoter in wild-type kidneys, and immunoblot analysis revealed downregulated expression of FXR in HNF-1ß mutant kidneys. These findings reveal a novel role of HNF-1ß in osmoregulation and identify multiple mechanisms, whereby mutations of HNF-1ß produce defects in urinary concentration.


Assuntos
Fator 1-beta Nuclear de Hepatócito/fisiologia , Túbulos Renais Coletores/fisiologia , Animais , Linhagem Celular , Feminino , Regulação da Expressão Gênica , Masculino , Camundongos Transgênicos , Poliúria/genética , Regiões Promotoras Genéticas , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Urina
6.
J Am Soc Nephrol ; 27(2): 345-53, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26319241

RESUMO

Hepatocyte nuclear factor 1ß (HNF1ß)-associated disease is a recently recognized clinical entity with a variable multisystem phenotype. Early reports described an association between HNF1B mutations and maturity-onset diabetes of the young. These patients often presented with renal cysts and renal function decline that preceded the diabetes, hence it was initially referred to as renal cysts and diabetes syndrome. However, it is now evident that many more symptoms occur, and diabetes and renal cysts are not always present. The multisystem phenotype is probably attributable to functional promiscuity of the HNF1ß transcription factor, involved in the development of the kidney, urogenital tract, pancreas, liver, brain, and parathyroid gland. Nephrologists might diagnose HNF1ß-associated kidney disease in patients referred with a suspected diagnosis of autosomal dominant polycystic kidney disease, medullary cystic kidney disease, diabetic nephropathy, or CKD of unknown cause. Associated renal or extrarenal symptoms should alert the nephrologist to HNF1ß-associated kidney disease. A considerable proportion of these patients display hypomagnesemia, which sometimes mimics Gitelman syndrome. Other signs include early onset diabetes, gout and hyperparathyroidism, elevated liver enzymes, and congenital anomalies of the urogenital tract. Because many cases of this disease are probably undiagnosed, this review emphasizes the clinical manifestations of HNF1ß-associated disease for the nephrologist.


Assuntos
Nefropatias Diabéticas/etiologia , Fator 1-beta Nuclear de Hepatócito/fisiologia , Insuficiência Renal Crônica/etiologia , Adulto , Nefropatias Diabéticas/genética , Feminino , Fator 1-beta Nuclear de Hepatócito/genética , Humanos , Doenças Renais Císticas/etiologia , Masculino , Pessoa de Meia-Idade , Mutação , Fenótipo , Insuficiência Renal Crônica/genética , Adulto Jovem
7.
J Am Soc Nephrol ; 27(8): 2408-21, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26712526

RESUMO

HNF-1ß is a tissue-specific transcription factor that is expressed in the kidney and other epithelial organs. Humans with mutations in HNF-1ß develop kidney cysts, and HNF-1ß regulates the transcription of several cystic disease genes. However, the complete spectrum of HNF-1ß-regulated genes and pathways is not known. Here, using chromatin immunoprecipitation/next generation sequencing and gene expression profiling, we identified 1545 protein-coding genes that are directly regulated by HNF-1ß in murine kidney epithelial cells. Pathway analysis predicted that HNF-1ß regulates cholesterol metabolism. Expression of dominant negative mutant HNF-1ß or kidney-specific inactivation of HNF-1ß decreased the expression of genes that are essential for cholesterol synthesis, including sterol regulatory element binding factor 2 (Srebf2) and 3-hydroxy-3-methylglutaryl-CoA reductase (Hmgcr). HNF-1ß mutant cells also expressed lower levels of cholesterol biosynthetic intermediates and had a lower rate of cholesterol synthesis than control cells. Additionally, depletion of cholesterol in the culture medium mitigated the inhibitory effects of mutant HNF-1ß on the proteins encoded by Srebf2 and Hmgcr, and HNF-1ß directly controlled the renal epithelial expression of proprotein convertase subtilisin-like kexin type 9, a key regulator of cholesterol uptake. These findings reveal a novel role of HNF-1ß in a transcriptional network that regulates intrarenal cholesterol metabolism.


Assuntos
Colesterol/metabolismo , Fator 1-beta Nuclear de Hepatócito/fisiologia , Rim/metabolismo , Animais , Colesterol/genética , Camundongos
8.
Nat Rev Nephrol ; 11(2): 102-12, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25536396

RESUMO

Heterozygous mutations in the gene that encodes the transcription factor hepatocyte nuclear factor 1ß (HNF1B) represent the most common known monogenic cause of developmental kidney disease. Renal cysts are the most frequently detected feature of HNF1B-associated kidney disease; however, other structural abnormalities, including single kidneys and renal hypoplasia, and electrolyte abnormalities can also occur. Extra-renal phenotypes might also be observed; consequently, HNF1B-associated disease is considered a multi-system disorder. Other clinical features include early-onset diabetes mellitus, pancreatic hypoplasia, genital tract malformations, abnormal liver function and early-onset gout. Heterozygous mutations in the coding region or splice sites of HNF1B, and complete gene deletion, each account for ∼50% of all cases of HNF1B-associated disease, respectively, and often arise spontaneously. There is no clear genotype-phenotype correlation, consistent with haploinsufficiency as the disease mechanism. Data from animal models suggest that HNF1B has an important function during several stages of nephrogenesis; however, the precise signalling pathways remain to be elucidated. This Review discusses the genetics and molecular pathways that lead to disease development, summarizes the reported renal and extra-renal phenotypes, and identifies areas for future research in HNF1B-associated disease.


Assuntos
Fator 1-beta Nuclear de Hepatócito/genética , Nefropatias/genética , Fator 1-beta Nuclear de Hepatócito/fisiologia , Humanos , Nefropatias/etiologia , Mutação , Fenótipo
9.
Am J Physiol Renal Physiol ; 307(3): F356-68, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24899057

RESUMO

Autosomal recessive polycystic kidney disease, an inherited disorder characterized by the formation of cysts in renal collecting ducts and biliary dysgenesis, is caused by mutations of the polycystic kidney and hepatic disease 1 (PKHD1) gene. Expression of PKHD1 is tissue specific and developmentally regulated. Here, we show that a 2.0-kb genomic fragment containing the proximal promoter of mouse Pkhd1 directs tissue-specific expression of a lacZ reporter gene in transgenic mice. LacZ is expressed in renal collecting ducts beginning during embryonic development but is not expressed in extrarenal tissues. The Pkhd1 promoter contains a binding site for the transcription factor hepatocyte nuclear factor (HNF)-1ß, which is required for activity in transfected cells. Mutation of the HNF-1ß-binding site abolishes the expression of the lacZ reporter gene in renal collecting ducts. Transgenes containing the 2.0-kb promoter and 2.7 kb of additional genomic sequence extending downstream to the second exon are expressed in the kidney, intrahepatic bile ducts, and male reproductive tract. This pattern overlaps with the endogenous expression of Pkhd1 and coincides with sites of expression of HNF-1ß. We conclude that the proximal 2.0-kb promoter is sufficient for tissue-specific expression of Pkhd1 in renal collecting ducts in vivo and that HNF-1ß is required for Pkhd1 promoter activity in collecting ducts. Additional genomic sequences located from exons 1-2 or elsewhere in the gene locus are required for expression in extrarenal tissues.


Assuntos
Túbulos Renais Coletores/fisiologia , Rim Policístico Autossômico Recessivo/fisiopatologia , Regiões Promotoras Genéticas/fisiologia , Receptores de Superfície Celular/fisiologia , Animais , Sistema Biliar/citologia , Sistema Biliar/fisiologia , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Fator 1-beta Nuclear de Hepatócito/fisiologia , Túbulos Renais Coletores/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Rim Policístico Autossômico Recessivo/genética , Regiões Promotoras Genéticas/genética , Receptores de Superfície Celular/genética , Sistema Urogenital/citologia , Sistema Urogenital/fisiologia
10.
Pediatr Nephrol ; 29(4): 659-64, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24190171

RESUMO

The nephron is the functional unit that executes the homeostatic roles of the kidney in vertebrates. Critical to this function is the physical arrangement of the glomerular blood filter attached to a tubular epithelium that is subdivided into specialized proximal and distal segments. During embryogenesis, nephron progenitors undergo a mesenchymal-epithelial transition (MET) and adopt different segment-specific cell fates along the proximo-distal axis of the nephron. The molecular basis of how these segments arise remains largely unknown. Recent studies using the zebrafish have identified the Hnf1beta transcription factor (Hnf1b) as a major regulator of tubular segmentation. In Hnf1b-deficient zebrafish embryos, nephron progenitors fail to adopt the proximo-distal segmentation pattern of the nephron, yet still undergo MET. This observation suggests that the functional segmentation of renal tubular epithelial cells is independent of pathways that induce their epithelialization. Here we review this new role of Hnf1b for nephron segmentation during zebrafish and mouse kidney development.


Assuntos
Fator 1-beta Nuclear de Hepatócito/metabolismo , Néfrons/embriologia , Néfrons/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Fator 1-beta Nuclear de Hepatócito/fisiologia , Humanos , Organogênese/fisiologia , Proteínas de Peixe-Zebra/fisiologia
11.
PLoS One ; 7(1): e30390, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22299039

RESUMO

We examined the association between HNF1B variants identified in a recent genome-wide association study and endometrial cancer in two large case-control studies nested in prospective cohorts: the Multiethnic Cohort Study (MEC) and the Women's Health Initiative (WHI) as part of the Population Architecture using Genomics and Epidemiology (PAGE) study. A total of 1,357 incident cases of invasive endometrial cancer and 7,609 controls were included in the analysis (MEC: 426 cases/3,854 controls; WHI: 931 cases/3,755 controls). The majority of women in the WHI were European American, while the MEC included sizable numbers of African Americans, Japanese and Latinos. We estimated the odds ratios (ORs) per allele and 95% confidence intervals (CIs) of each SNP using unconditional logistic regression adjusting for age, body mass index, and four principal components of ancestry informative markers. The combined ORs were estimated using fixed effect models. Rs4430796 and rs7501939 were associated with endometrial cancer risk in MEC and WHI with no heterogeneity observed across racial/ethnic groups (P ≥ 0.21) or between studies (P ≥ 0.70). The OR(per allele) was 0.82 (95% CI: 0.75, 0.89; P = 5.63 × 10(-6)) for rs4430796 (G allele) and 0.79 (95% CI: 0.73, 0.87; P = 3.77 × 10(-7)) for rs7501939 (A allele). The associations with the risk of Type I and Type II tumors were similar (P ≥ 0.19). Adjustment for additional endometrial cancer risk factors such as parity, oral contraceptive use, menopausal hormone use, and smoking status had little effect on the results. In conclusion, HNF1B SNPs are associated with risk of endometrial cancer and that the associated relative risks are similar for Type I and Type II tumors.


Assuntos
Carcinoma Endometrioide/genética , Neoplasias do Endométrio/genética , Fator 1-beta Nuclear de Hepatócito/fisiologia , Idoso , Carcinoma Endometrioide/etnologia , Carcinoma Endometrioide/patologia , Estudos de Casos e Controles , Estudos de Coortes , Neoplasias do Endométrio/etnologia , Neoplasias do Endométrio/patologia , Etnicidade/estatística & dados numéricos , Feminino , Genética Populacional , Fator 1-beta Nuclear de Hepatócito/genética , Humanos , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único/fisiologia
12.
J Pharmacol Exp Ther ; 340(3): 648-55, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22160269

RESUMO

We have reported previously that the kidney- and liver-specific expression of transporters in mice involves the coordinated regulation by hepatocyte nuclear factor 1 (HNF1) and DNA methylation. The present study was aimed at investigating the role of this cascade in the transcriptional regulation of renal organic anion transporters (OATs) yet to be characterized in human and mouse. Luciferase assays and electrophoretic mobility-shift assays demonstrated that HNF1α/ß enhances the promoter activity of OAT4/SLC22A11 via binding to the HNF1 motif located near the transcriptional start site (TSS). DNA methylation profiles of human OAT1, OAT3, OAT4, and urate transporter 1 (URAT1) were determined in human liver and kidney cortex by bisulfite sequencing. Most of the CpG dinucleotides around the TSSs of OAT1 and OAT3 were highly methylated in the liver compared with kidney cortex, being consistent with their tissue specificity, whereas the difference in the DNA methylation status was less remarkable between the two tissues for OAT4 and URAT1. Mouse Oat1 gene also contained CpG dinucleotides hypomethylated in the kidney and hypermethylated in the liver downstream its TSS, whereas two of the seven CpG dinucleotides around the TSS of mouse Oat3 were significantly methylated in the liver compared with the kidney. Taken together, these findings underscored the central role of HNF1α/ß in the transcriptional regulation of OATs and highlighted DNA methylation-dependent gene silencing as one of the mechanisms underlying the tissue-specific transactivation by this master regulator.


Assuntos
Metilação de DNA , Fator 1-alfa Nuclear de Hepatócito/fisiologia , Fator 1-beta Nuclear de Hepatócito/fisiologia , Rim/metabolismo , Transportadores de Ânions Orgânicos/genética , Animais , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Proteínas de Transporte de Cátions Orgânicos/genética , Regiões Promotoras Genéticas
13.
Histopathology ; 58(6): 934-43, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21438902

RESUMO

AIMS: Papillary renal cell tumours (RCT) are characterized by specific trisomies. The aim of this study was to identify small regions of duplication marking putative tumour genes. METHODS AND RESULTS: Full-tiling path bacterial artificial chromosome (BAC) array hybridization of 20 papillary RCTs confirmed the duplication of chromosomes 7 and 17 and also displayed smaller regions of gains/amplifications of 1.3-13.1 Mb in size. Detailed analysis showed a microamplification of BAC clones containing the MET at the 7q31.2 and also amplification of a DNA segment harbouring the transcription factor hepatocyte nuclear factor 1 beta (HNF1B) at chromosome 17q12. Nuclear expression of HNF1B protein was detected in 38 of 67 papillary RCTs, in five of five mucinous tubular and spindle cell carcinomas (MTSCC) and five of five metanephric adenomas by immunohistochemistry. Moreover, nine nephrogenic rests containing tubular differentiated structures and all 14 and five precursor lesions associated with papillary RCTs and MTSCCs, respectively, showed strong nuclear positivity when compared to the expression level in proximal tubules of the corresponding normal kidney. CONCLUSIONS: Our findings indicate a role of HNF1B in association with the high frequency of chromosome 17q duplication in the development of papillary RCTs and MTSCCs as well as in their precursor lesions.


Assuntos
Carcinoma de Células Renais/genética , DNA de Neoplasias/genética , Duplicação Gênica/genética , Perfilação da Expressão Gênica , Fator 1-beta Nuclear de Hepatócito/fisiologia , Neoplasias Renais/genética , Adenocarcinoma Mucinoso/genética , Adenocarcinoma Mucinoso/fisiopatologia , Adenoma/genética , Adenoma/fisiopatologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Renais/fisiopatologia , Cromossomos Artificiais Bacterianos/genética , Cromossomos Humanos Par 17/genética , Cromossomos Humanos Par 7/genética , Feminino , Humanos , Neoplasias Renais/fisiopatologia , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Trissomia
14.
J Invest Dermatol ; 131(6): 1291-9, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21346776

RESUMO

The prognosis of advanced melanoma remains poor in spite of treatment advances, emphasizing the importance of additional preventive measures. Flavonoids, natural components of our diet, are being investigated for their chemopreventive/therapeutic properties. Microphthalmia-associated transcription factor (Mitf), downstream of the Wnt/ß-catenin pathway, has become an important prognostic marker of melanoma. In this study, we show that treatment of 451Lu melanoma cells with the dietary flavonoid fisetin (3,7,3',4'-tetrahydroxyflavone) resulted in decreased cell viability with G1-phase arrest and disruption of Wnt/ß-catenin signaling. This was accompanied by a decrease in the expression of Wnt protein and its co-receptors, as well as by a parallel increase in the expression of endogenous Wnt inhibitors. Fisetin-treated cells showed increased cytosolic levels of Axin and ß-TrCP and decreased phosphorylation of glycogen synthase kinase 3ß associated with decreased ß-catenin stabilization. Fisetin-mediated interference with the functional cooperation between ß-catenin and T-cell factor (TCF)-2 resulted in the downregulation of positively regulated TCF targets, such as c-myc, Brn-2, and Mitf. Flow-cytometric analysis of Mitf-overexpressing cells showed that fisetin repressed Mitf-induced cell proliferation. Finally, administration of fisetin to 451Lu-xenografted nude mice resulted in the inhibition of tumor development and decreased Mitf expression. Our data suggest that fisetin can be developed as an effective agent against melanoma because of its potential inhibitory effect on ß-catenin/Mitf signaling.


Assuntos
Antineoplásicos/farmacologia , Flavonoides/farmacologia , Melanoma/tratamento farmacológico , Fator de Transcrição Associado à Microftalmia/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Proteínas Wnt/fisiologia , beta Catenina/fisiologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Flavonóis , Fase G1/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Fator 1-beta Nuclear de Hepatócito/fisiologia , Humanos , Melanoma/patologia , Camundongos , beta Catenina/análise , Proteínas Contendo Repetições de beta-Transducina/fisiologia
15.
Dev Biol ; 351(2): 229-41, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21215262

RESUMO

Planar cell polarity (PCP) establishes polarity within an epithelial sheet. Defects in PCP are associated with developmental defects involving directional cell growth, including defects in kidney tubule elongation that lead to formation of kidney cysts. Given the strong association between kidney cyst formation and developmental biliary defects in patients and in animal models, we investigated the importance of PCP in biliary development. Here we report that in zebrafish, morpholino antisense oligonucleotide-mediated knockdown of PCP genes including prickle-1a (pk1a) led to developmental biliary abnormalities, as well as localization defects of the liver and other digestive organs. The defects in biliary development appear to be mediated via downstream PCP targets such as Rho kinase, Jun kinase (JNK), and both actin and microtubule components of the cytoskeleton. Knockdown of pk1a led to decreased expression of vhnf1, a homeodomain gene previously shown to be involved in biliary development and in kidney cyst formation; forced expression of vhnf1 mRNA led to rescue of the pk1a morphant phenotype. Our results demonstrate that PCP plays an important role in vertebrate biliary development, interacting with other factors known to be involved in biliary morphogenesis.


Assuntos
Ductos Biliares/anormalidades , Proteínas de Transporte/fisiologia , Polaridade Celular , Fator 1-beta Nuclear de Hepatócito/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/embriologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Ductos Biliares/citologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas com Domínio LIM , Fígado/metabolismo
16.
Curr Opin Nephrol Hypertens ; 19(5): 456-62, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20625291

RESUMO

PURPOSE OF REVIEW: This review highlights recent advances in renal magnesium (Mg) handling. The understanding of the molecular processes of epithelial Mg transport has expanded considerably due to the identification of novel genes involved in hypomagnesemic disorders. RECENT FINDINGS: Mg deficiency remains one of the most common electrolyte disorders. Detailed genetic analysis of families with inherited forms of hypomagnesemia has led to the identification of new genes involved in Mg homeostasis. As such, familial hypomagnesemia has been linked to mutations in the claudin-16/19 complex located in the thick ascending limb. Moreover, the pro-epidermal growth factor, the potassium channels Kv1.1 and Kir4.1, and the hepatocyte nuclear factor 1B have recently been identified as causative factors in syndromes of hereditary hypomagnesemia. These proteins play key roles in regulating electrolyte balance within the distal convoluted tubule, either by directly affecting the epithelial Mg channel, transient receptor potential channel melastatin member 6, or by altering the driving force for Mg influx via the channel. SUMMARY: Recent genetic and molecular studies have further elucidated the processes that govern renal Mg transport and hence systemic Mg balance. This has provided us with new tools to understand the molecular pathology behind hypomagnesemia.


Assuntos
Magnésio/metabolismo , Néfrons/metabolismo , Fator 1-beta Nuclear de Hepatócito/fisiologia , Humanos , Transporte de Íons , Rim/metabolismo , Túbulos Renais Distais/metabolismo , Túbulos Renais Proximais/metabolismo , Canal de Potássio Kv1.1/fisiologia , Alça do Néfron/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , ATPase Trocadora de Sódio-Potássio/fisiologia , Canais de Cátion TRPM/fisiologia
17.
Urology ; 76(2): 507.e6-11, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20538322

RESUMO

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.


Assuntos
Carcinoma de Células Renais/secundário , Regulação para Baixo , Fator 1-beta Nuclear de Hepatócito/fisiologia , Neoplasias Renais/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/metabolismo , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Fator 1-beta Nuclear de Hepatócito/biossíntese , Fator 1-beta Nuclear de Hepatócito/genética , Humanos , Neoplasias Renais/genética , Neoplasias Renais/metabolismo , Masculino , Pessoa de Meia-Idade , Prognóstico
18.
Nat Med ; 16(1): 106-10, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19966811

RESUMO

Hepatocyte nuclear factor-1beta (HNF-1beta) is a transcription factor required for the expression of several renal cystic genes and whose prenatal deletion leads to polycystic kidney disease (PKD). We show here that inactivation of Hnf1b from postnatal day 10 onward does not elicit cystic dilations in tubules after their proliferative morphogenetic elongation is over. Cystogenic resistance is intrinsically linked to the quiescent state of cells. In fact, when Hnf1b deficient quiescent cells are forced to proliferate by an ischemia-reperfusion injury, they give rise to cysts, owing to loss of oriented cell division. Remarkably, in quiescent cells, the transcription of crucial cystogenic target genes is maintained even in the absence of HNF-1beta. However, their expression is lost as soon as cells proliferate and the chromatin of target genes acquires heterochromatin marks. These results unveil a previously undescribed aspect of gene regulation. It is well established that transcription is shut off during the mitotic condensation of chromatin. We propose that transcription factors such as HNF-1beta might be involved in reprogramming gene expression after transcriptional silencing is induced by mitotic chromatin condensation. Notably, HNF-1beta remains associated with the mitotically condensed chromosomal barrels. This association suggests that HNF-1beta is a bookmarking factor that is necessary for reopening the chromatin of target genes after mitotic silencing.


Assuntos
Fator 1-beta Nuclear de Hepatócito/fisiologia , Doenças Renais Policísticas/genética , Animais , Divisão Celular/genética , Cromatina/fisiologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Fator 1-beta Nuclear de Hepatócito/genética , Túbulos Renais/crescimento & desenvolvimento , Camundongos , Mitose/fisiologia , Doenças Renais Policísticas/etiologia , Ativação Transcricional/genética , Ativação Transcricional/efeitos da radiação
19.
Oncol Rep ; 22(2): 233-40, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19578761

RESUMO

Epithelial ovarian cancer (EOC) is the leading cause of death in women with gynecological malignancies. Among EOC, clear cell carcinoma (CCC) and endometrioid adenocarcinoma (EAC) differ from the other histological types with respect to their clinical characteristics and carcinogenesis. Both tumor types are often associated with endometriosis. EAC is recently reported to be characterized by K-RAS activation and PTEN dysfunction. However, the molecular changes in CCC remain largely unknown. The aim of this review is to summarize the current knowledge on the molecular mechanisms involved in CCC tumorigenesis. The present article reviews the English language literature for biological, pathogenetic and pathophysiological studies on endometriosis-associated CCC of the ovary. Several recent studies of loss of heterozygosity (LOH), allelic loss, comparative genomic hybridization, mutation, methylation status, microarray gene-expression profiling and proteomics are discussed in the context of CCC biology. Retrograde menstruation or ovarian hemorrhage carries highly pro-oxidant factors, such as heme and iron, into the peritoneal cavity or ovarian endometrioma. A histologically normal ectopic endometrium bears genetic damages caused by iron-dependent oxidative stress. DNA damage or LOH caused by oxidative stress is a critical factor in the carcinogenic process. LOH studies have implicated the involvement of specific chromosomal regions (5q, 6q, 9p, 10q, 11q, 17q and 22q). Furthermore, the PTEN and APC (early event), p53, polo-like kinases, Emi1 and K-RAS (late event) genes may be involved in CCC carcinogenesis. The molecular pathology of CCC is heterogeneous and involves various putative precursor lesions and multiple pathways of development, possibly via genetic alteration by oxidative stress.


Assuntos
Adenocarcinoma de Células Claras/etiologia , Endometriose/complicações , Neoplasias Ovarianas/etiologia , Proteínas de Ciclo Celular/fisiologia , Proteínas F-Box/fisiologia , Feminino , Genes Supressores de Tumor , Fator 1-beta Nuclear de Hepatócito/fisiologia , Humanos , Perda de Heterozigosidade , Instabilidade de Microssatélites , Estresse Oxidativo , Proteínas Quinases/fisiologia , Serina-Treonina Quinases TOR
20.
Cancer Res ; 69(11): 4843-50, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19487294

RESUMO

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


Assuntos
Carcinoma/genética , Regulação Neoplásica da Expressão Gênica , Fator 1-beta Nuclear de Hepatócito/fisiologia , Neoplasias Ovarianas/genética , Sulfotransferases/genética , Animais , Carcinoma/patologia , Linhagem Celular Tumoral , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Fator 1-beta Nuclear de Hepatócito/antagonistas & inibidores , Fator 1-beta Nuclear de Hepatócito/genética , Fator 1-beta Nuclear de Hepatócito/metabolismo , Humanos , Camundongos , Camundongos Nus , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Ovarianas/patologia , Ligação Proteica , RNA Interferente Pequeno/farmacologia , Elementos de Resposta , Sulfotransferases/fisiologia , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...