Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Genet ; 8(11): e1003053, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23209428

RESUMO

Autosomal Dominant Polycystic Kidney Disease (ADPKD; MIM ID's 173900, 601313, 613095) leads to end-stage kidney disease, caused by mutations in PKD1 or PKD2. Inactivation of Pkd1 before or after P13 in mice results in distinct early- or late-onset disease. Using a mouse model of ADPKD carrying floxed Pkd1 alleles and an inducible Cre recombinase, we intensively analyzed the relationship between renal maturation and cyst formation by applying transcriptomics and metabolomics to follow disease progression in a large number of animals induced before P10. Weighted gene co-expression network analysis suggests that Pkd1-cystogenesis does not cause developmental arrest and occurs in the context of gene networks similar to those that regulate/maintain normal kidney morphology/function. Knowledge-based Ingenuity Pathway Analysis (IPA) software identifies HNF4α as a likely network node. These results are further supported by a meta-analysis of 1,114 published gene expression arrays in Pkd1 wild-type tissues. These analyses also predict that metabolic pathways are key elements in postnatal kidney maturation and early steps of cyst formation. Consistent with these findings, urinary metabolomic studies show that Pkd1 cystic mutants have a distinct profile of excreted metabolites, with pathway analysis suggesting altered activity in several metabolic pathways. To evaluate their role in disease, metabolic networks were perturbed by inactivating Hnf4α and Pkd1. The Pkd1/Hnf4α double mutants have significantly more cystic kidneys, thus indicating that metabolic pathways could play a role in Pkd1-cystogenesis.


Assuntos
Redes Reguladoras de Genes , Fator 4 Nuclear de Hepatócito , Rim Policístico Autossômico Dominante , Proteína Quinase C , Alelos , Animais , Proliferação de Células , Modelos Animais de Doenças , Regulação da Expressão Gênica , Fator 4 Nuclear de Hepatócito/genética , Fator 4 Nuclear de Hepatócito/metabolismo , Humanos , Camundongos , Mutação , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/metabolismo , Proteína Quinase C/genética , Proteína Quinase C/metabolismo
2.
Hum Mol Genet ; 21(26): 5456-71, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23001567

RESUMO

Autosomal-dominant polycystic kidney disease (ADPKD) and von Hippel-Lindau (VHL) disease lead to large kidney cysts that share pathogenetic features. The polycystin-1 (PC1) and pVHL proteins may therefore participate in the same key signaling pathways. Jade-1 is a pro-apoptotic and growth suppressive ubiquitin ligase for beta-catenin and transcriptional coactivator associated with histone acetyltransferase activity that is stabilized by pVHL in a manner that correlates with risk of VHL renal disease. Thus, a relationship between Jade-1 and PC1 was sought. Full-length PC1 bound, stabilized and colocalized with Jade-1 and inhibited Jade-1 ubiquitination. In contrast, the cytoplasmic tail or the naturally occurring C-terminal fragment of PC1 (PC1-CTF) promoted Jade-1 ubiquitination and degradation, suggesting a dominant-negative mechanism. ADPKD-associated PC1 mutants failed to regulate Jade-1, indicating a potential disease link. Jade-1 ubiquitination was mediated by Siah-1, an E3 ligase that binds PC1. By controlling Jade-1 abundance, PC1 and the PC1-CTF differentially regulate Jade-1-mediated transcriptional activity. A key target of PC1, the cyclin-dependent kinase inhibitor p21, is also up-regulated by Jade-1. Through Jade-1, PC1 and PC1 cleaved forms may exert fine control of beta-catenin and canonical Wnt signaling, a critical pathway in cystic renal disease. Thus, Jade-1 is a transcription factor and ubiquitin ligase whose activity is regulated by PC1 in a manner that is physiologic and may correlate with disease. Jade-1 may be an important therapeutic target in renal cystogenesis.


Assuntos
Regulação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Canais de Cátion TRPP/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Ubiquitinação , Sequência de Aminoácidos , Apoptose , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Células HEK293 , Meia-Vida , Proteínas de Homeodomínio/genética , Humanos , Rim/citologia , Rim/patologia , Dados de Sequência Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/patologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Canais de Cátion TRPP/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Regulação para Cima , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , Via de Sinalização Wnt , beta Catenina/genética , beta Catenina/metabolismo , Doença de von Hippel-Lindau/genética , Doença de von Hippel-Lindau/metabolismo
3.
Biochem Biophys Res Commun ; 444(4): 473-9, 2014 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-24472557

RESUMO

The PKD1 gene is essential for a number of biological functions, and its loss-of-function causes autosomal dominant polycystic kidney disease (ADPKD). The gene is developmentally regulated and believed to play an essential role in renal development. Previous studies have shown that manipulating murine renal organ cultures with dominant-negative forms of the Pkd1 gene impaired ureteric bud (UB) branching. In the current study, we analyzed different stages of renal development in two distinct mouse models carrying either a null mutation or inactivation of the last two exons of Pkd1. Surprisingly, metanephric explants from Pkd1-deleted kidneys harvested at day E11.5 did not show defects of UB branching and elongation, estimated by cytokeratin staining on fixed tissues or by Hoxb7-GFP time-lapse imaging. However, renal explants from Pkd1-mutants isolated at day E14.5 showed impaired nephrogenesis. Notably, we observed cell migratory defects in the developing endothelial compartment. Previous studies had implicated the Pkd1 gene in controlling cell migration and collagen deposition through PI3 kinases. In line with these studies, our results show that wild-type explants treated with PI3-kinase inhibitors recapitulate the endothelial defects observed in Pkd1 mutants, whereas treatment with VEGF only partially rescued the defects. Our data are consistent with a role for the Pkd1 gene in the endothelium that may be required for proper nephrogenesis.


Assuntos
Glomérulos Renais/embriologia , Glomérulos Renais/fisiopatologia , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/fisiopatologia , Canais de Cátion TRPP/genética , Animais , Movimento Celular , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Deleção de Genes , Glomérulos Renais/metabolismo , Camundongos , Mutação , Técnicas de Cultura de Órgãos , Inibidores de Fosfoinositídeo-3 Quinase , Canais de Cátion TRPP/metabolismo
4.
Dev Dyn ; 240(6): 1493-501, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21465620

RESUMO

Polycystic kidney diseases (PKD) are inherited disorders characterized by fluid-filled cysts primarily in the kidneys. We previously reported differences between the expression of Cux1, p21, and p27 in the cpk and Pkd1 null mouse models of PKD. Embryonic lethality of Pkd1 null mice limits its study to early stages of kidney development. Therefore, we examined mice with a collecting duct specific deletion in the Pkd1 gene. Cux1 was ectopically expressed in the cyst lining epithelial cells of newborn, P7 and P15 Pkd1(CD) mice. Cux1 expression correlated with cell proliferation in early stages of cystogenesis, however, as the disease progressed, fewer cyst lining cells showed increased cell proliferation. Rather, Cux1 expression in late stage cystogenesis was associated with increased apoptosis. Taken together, our results suggest that increased Cux1 expression associated with apoptosis is a common feature of late stage cyst progression in both the cpk and Pkd1(CD) mouse models of PKD.


Assuntos
Apoptose/genética , Inibidor de Quinase Dependente de Ciclina p27/genética , Proteínas de Homeodomínio/genética , Doenças Renais Císticas/genética , Proteínas Nucleares/genética , Proteínas Repressoras/genética , Canais de Cátion TRPP/genética , Animais , Animais Recém-Nascidos , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Progressão da Doença , Regulação para Baixo , Ativação Enzimática/genética , Feminino , Regulação da Expressão Gênica , Inativação Gênica , Doenças Renais Císticas/metabolismo , Doenças Renais Císticas/patologia , Túbulos Renais Coletores/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Canais de Cátion TRPP/metabolismo , Transfecção , Regulação para Cima/genética
5.
J Am Soc Nephrol ; 21(3): 489-97, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20075061

RESUMO

Aberrant activation of the mammalian target of rapamycin (mTOR) pathway occurs in polycystic kidney disease (PKD). mTOR inhibitors, such as rapamycin, are highly effective in several rodent models of PKD, but these models result from mutations in genes other than Pkd1 and Pkd2, which are the primary genes responsible for human autosomal dominant PKD. To address this limitation, we tested the efficacy of rapamycin in a mouse model that results from conditional inactivation of Pkd1. Mosaic deletion of Pkd1 resulted in PKD and replicated characteristic features of human PKD including aberrant mTOR activation, epithelial proliferation and apoptosis, and progressive fibrosis. Treatment with rapamycin was highly effective: It reduced cyst growth, preserved renal function, inhibited epithelial cell proliferation, increased apoptosis of cyst-lining cells, and inhibited fibrosis. These data provide in vivo evidence that rapamycin is effective in a human-orthologous mouse model of PKD.


Assuntos
Doenças Renais Policísticas/genética , Doenças Renais Policísticas/fisiopatologia , Sirolimo/farmacologia , Canais de Cátion TRPP/genética , Animais , Apoptose/fisiologia , Nitrogênio da Ureia Sanguínea , Divisão Celular/fisiologia , Modelos Animais de Doenças , Fibrose , Expressão Gênica/efeitos dos fármacos , Humanos , Imunossupressores/farmacologia , Proteínas de Filamentos Intermediários/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Túbulos Renais Coletores/patologia , Túbulos Renais Coletores/fisiopatologia , Túbulos Renais Distais/patologia , Túbulos Renais Distais/fisiopatologia , Camundongos , Mosaicismo , Proteínas do Tecido Nervoso/genética , Nestina , Fenótipo , Doenças Renais Policísticas/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Serina-Treonina Quinases TOR , Canais de Cátion TRPP/metabolismo
6.
Kidney Int ; 75(6): 626-33, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19145237

RESUMO

Renal cysts in autosomal dominant polycystic kidney disease arise from cells throughout the nephron, but there is an uncertainty as to whether both the intercalated cells (ICs) and principal cells (PCs) within the collecting duct give rise to cysts. To determine this, we crossed mice containing loxP sites within introns 1 and 4 of the Pkd1 gene with transgenic mice expressing Cre recombinase under control of the aquaporin-2 promoter or the B1 subunit of the proton ATPase promoter, thereby generating PC- or IC-specific knockout of Pkd1, respectively. Mice, that had Pkd1 deleted in the PCs, developed progressive cystic kidney disease evident during the first postnatal week and had an average lifespan of 8.2 weeks. There was no change in the cellular cAMP content or membrane aquaporin-2 expression in their kidneys. Cysts were present in the cortex and outer medulla but were absent in the papilla. Mice in which PKd1 was knocked out in the ICs had a very mild cystic phenotype as late as 13 weeks of age, limited to 1-2 cysts and confined to the outer rim of the kidney cortex. These mice lived to at least 1.5 years of age without evidence of early mortality. Our findings suggest that PCs are more important than ICs for cyst formation in polycystic kidney disease.


Assuntos
Doenças Renais Císticas/genética , Doenças Renais Císticas/patologia , Canais de Cátion TRPP/genética , Animais , Córtex Renal/patologia , Medula Renal/patologia , Túbulos Renais Coletores/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fenótipo
7.
J Vis Exp ; (112)2016 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-27341293

RESUMO

Exosome research in the last three years has greatly extended the scope towards identification and characterization of biomarkers and their therapeutic uses. Exosomes have recently been shown to contain microRNAs (miRs). MiRs themselves have arisen as valuable biomarkers for diagnostic purposes. As specimen collection in clinics and hospitals is quite variable, miRNA isolation from whole bile varies substantially. To achieve robust, accurate and reproducible miRNA profiles from collected bile samples in a simple manner required the development of a high-quality protocol to isolate and characterize exosomes from bile. The method requires several centrifugations and a filtration step with a final ultracentrifugation step to pellet the isolated exosomes. Electron microscopy, Western blots, flow cytometry and multi-parameter nanoparticle optical analysis, where available, are crucial characterization steps to validate the quality of the exosomes. For the isolation of miRNA from these exosomes, spiking the lysate with a non-specific, synthetic miRNA from a species like Caenorhabditis elegans, i.e., Cel-miR-39, is important for normalization of RNA extraction efficiency. The isolation of exosome from bile fluid following this method allows the successful miRNA profiling from bile samples stored for several years at -80 °C.


Assuntos
Bile , Exossomos , Biomarcadores , Humanos , MicroRNAs , Ultracentrifugação
8.
J Clin Med ; 4(9): 1713-28, 2015 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-26343737

RESUMO

Profound changes in microRNA (miR) expression levels are frequently found in liver cancers compared to the normal liver. In this study, we evaluate the expression of miR-224 in human HCC and CCA, as well as its downstream targets and affected pathways. We show that miR-224 is upregulated in a large cohort of human CCA, similar to its upregulation in human HCC. For the purpose of studying the roles of miR-224 in HCC and CCA, we enforced miR-224 expression in cells. mRNA arrays followed by Ingenuity Pathway Analysis (IPA)-identified putative molecules and pathways downstream of miR-224. Phenotypically, we report that enforced expression of miR-224 increases the growth rate of normal cholangiocytes, CCA cell lines, and HCC cell lines. In addition, we identified, in an unbiased fashion, that one of the major biologic processes affected by miR-224 is Gap1 (G1) to Synthesis (S) transition checkpoint release. We next identified p21, p15, and CCNE1 as downstream targets of miR-224 and confirmed the coordinated downregulation results in the increased phosphorylation of Retinoblastoma (Rb) with resulting G1/S checkpoint release. Our data suggest that miR-224 is a master regulator of cell cycle progression, and that its overexpression results in G1/S checkpoint release followed by accelerated cell growth.

9.
Oncol Rep ; 32(1): 419-24, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24841903

RESUMO

Cyclin-dependent kinase 5 (CDK5) is a potential target for prostate cancer treatment, the enzyme being essential for prostate tumor growth and formation of metastases. In the present study, we identified agents that target prostate cancer cells based on CDK5 expression. CDK5 activity was suppressed by transfection of PC3 prostate cancer cells with a dominant-negative construct (PC3 CDK5dn). PC3 CDK5dn and PC3 control cells were screened for compounds that selectively target cells based on CDK5 expression, utilizing the Johns Hopkins Drug Library. MTS proliferation, clonogenic and 3D growth assays were performed to validate the selected hits. Screening of 3,360 compounds identified rutilantin, ethacridine lactate and cetalkonium chloride as compounds that selectively target PC3 control cells and a tilorone analog as a selective inhibitor of PC3 CDK5dn cells. A PubMed literature study indicated that tilorone may have clinical use in patients. Validation experiments confirmed that tilorone treatment resulted in decreased PC3 cell growth and invasion; PC3 cells with inactive CDK5 were inhibited more effectively. Future studies are needed to unravel the mechanism of action of tilorone in CDK5 deficient prostate cancer cells and to test combination therapies with tilorone and a CDK5 inhibitor for its potential use in clinical practice.


Assuntos
Antineoplásicos/farmacologia , Quinase 5 Dependente de Ciclina/metabolismo , Invasividade Neoplásica/patologia , Neoplasias da Próstata/patologia , Tilorona/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Neoplasias da Próstata/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia
10.
PLoS One ; 5(9)2010 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-20862291

RESUMO

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD) is a common cause of inherited renal failure that results from mutations in PKD1 and PKD2. The disorder is characterized by focal cyst formation that involves somatic mutation of the wild type allele in a large fraction of cysts. Consistent with a two-hit mechanism, mice that are homozygous for inactivating mutations of either Pkd1 or Pkd2 develop cystic kidneys, edema and hemorrhage and typically die in midgestation. Cystic kidney disease is unlikely to be the cause of fetal loss since renal function is not required to complete gestation. One hypothesis is that embryonic demise is due to leaky vessels or cardiac pathology. METHODOLOGY/PRINCIPAL FINDINGS: In these studies we used a series of genetically modified Pkd1 and Pkd2 murine models to investigate the cause of embryonic lethality in mutant embryos. Since placental defects are a frequent cause of fetal loss, we conducted histopathologic analyses of placentas from Pkd1 null mice and detected abnormalities of the labyrinth layer beginning at E12.5. We performed placental rescue experiments using tetraploid aggregation and conditional inactivation of Pkd1 with the Meox2 Cre recombinase. We found that both strategies improved the viability of Pkd1 null embryos. Selective inactivation of Pkd1 and Pkd2 in endothelial cells resulted in polyhydramnios and abnormalities similar to those observed in Pkd1(-/-) placentas. However, endothelial cell specific deletion of Pkd1 or Pkd2 did not yield the dramatic vascular phenotypes observed in null animals. CONCLUSIONS/SIGNIFICANCE: Placental abnormalities contribute to the fetal demise of Pkd(-/-) embryos. Endothelial cell specific deletion of Pkd1 or Pkd2 recapitulates a subset of findings seen in Pkd null animals. Our studies reveal a complex role for polycystins in maintaining vascular integrity.


Assuntos
Placenta/embriologia , Placenta/metabolismo , Canais de Cátion TRPP/metabolismo , Animais , Células Endoteliais/metabolismo , Feminino , Masculino , Camundongos , Camundongos Knockout , Gravidez , Canais de Cátion TRPP/genética
11.
Mol Cell Biol ; 29(9): 2359-71, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19255143

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is a common genetic disease characterized by bilateral renal cyst formation. Both hyperproliferation and hypertrophy have been previously observed in ADPKD kidneys. Polycystin-1 (PC-1), a large orphan receptor encoded by the PKD1 gene and mutated in 85% of all cases, is able to inhibit proliferation and apoptosis. Here we show that overexpression of PC-1 in renal epithelial cells inhibits cell growth (size) in a cell cycle-independent manner due to the downregulation of mTOR, S6K1, and 4EBP1. Upregulation of the same pathway leads to increased cell size, as found in mouse embryonic fibroblasts derived from Pkd1-/- mice. We show that PC-1 controls the mTOR pathway in a Tsc2-dependent manner, by inhibiting the extracellular signal-regulated kinase (ERK)-mediated phosphorylation of tuberin in Ser664. We provide a detailed molecular mechanism by which PC-1 can inhibit the mTOR pathway and regulate cell size.


Assuntos
Proteínas de Transporte/metabolismo , Tamanho Celular , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fosfoproteínas/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Canais de Cátion TRPP/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/genética , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular , Células Cultivadas , Fatores de Iniciação em Eucariotos , MAP Quinases Reguladas por Sinal Extracelular/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfoproteínas/genética , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/genética , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR , Canais de Cátion TRPP/genética , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética
12.
Hum Mol Genet ; 16(16): 1940-50, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17575307

RESUMO

Polycystic kidney disease (PKD) describes a heterogeneous collection of disorders that differ significantly with respect to their etiology and clinical presentation. They share, however, abnormal tubular morphology as a common feature, leading to the hypothesis that their respective gene products may function cooperatively in a common pathway to maintain tubular integrity. To study the pathobiology of one major form of human PKD, we generated a mouse line with a floxed allele of Pkhd1, the orthologue of the gene mutated in human autosomal recessive PKD. Cre-mediated excision of exons 3-4 results in a probable hypomorphic allele. Pkhd1(del3-4/del3-4) developed a range of phenotypes that recapitulate key features of the human disease. Like in humans, abnormalities of the biliary tract were an invariant finding. Most mice 6 months or older also developed renal cysts. Subsets of animals presented with either perinatal respiratory failure or exhibited growth retardation that was not due to the renal disease. We then tested for genetic interaction between Pkhd1 and Pkd1, the mouse orthologue of the gene most commonly linked to human autosomal dominant PKD. Pkd1(+/-); Pkhd1(del3-4/del3-4) mice had markedly more severe disease than Pkd1(+/+); Pkhd1(del3-4/del3-4) littermates. These studies are the first to show genetic interaction between the major loci responsible for human renal cystic disease in a common PKD pathway.


Assuntos
Ligação Genética , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Recessivo/genética , Animais , Cruzamentos Genéticos , Modelos Animais de Doenças , Feminino , Homozigoto , Masculino , Camundongos , Camundongos Transgênicos , Modelos Genéticos , Mutação , Receptores de Superfície Celular/genética
13.
Hum Mol Genet ; 15(1): 11-21, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16301212

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations of PKD1 and PKD2. Murine gene targeting studies have shown that these genes play an essential role in development, with homozygous inactivation resulting in embryonic lethality. Recently, Pkd1-/- lethality has been linked to placental insufficiency. In this study, the placenta was used as a model to identify factors involved in these developmental abnormalities. Microarray analysis of Pkd1-/- placentae showed upregulation of a set of apolipoprotein-related genes. These changes were validated and were found to be associated with increased quantities of apolipoproteins in the amniotic fluid. Increased apolipoprotein gene expression was also observed in Pkd2-/-placentae and in cystic kidneys of Pkd1cond/-; Meox2cre/+ mice. Using chromatin immunoprecipitation assays, we determined that the activity of HNF-4alpha, a major regulator of apolipoprotein gene expression, was also increased in these organs. These findings suggest a potential role for dysregulation of nuclear hormone receptors in the pathogenesis of ADPKD.


Assuntos
Apolipoproteínas/metabolismo , Regulação da Expressão Gênica/genética , Placenta/metabolismo , Rim Policístico Autossômico Dominante/genética , Canais de Cátion TRPP/metabolismo , Líquido Amniótico/metabolismo , Animais , Northern Blotting , Western Blotting , Imunoprecipitação da Cromatina , Primers do DNA , Feminino , Fator 4 Nuclear de Hepatócito/metabolismo , Imuno-Histoquímica , Rim/metabolismo , Camundongos , Camundongos Knockout , Análise em Microsséries , Gravidez
14.
Proc Natl Acad Sci U S A ; 103(14): 5466-71, 2006 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-16567633

RESUMO

Autosomal-dominant polycystic kidney disease (ADPKD) is a common genetic disorder that frequently leads to renal failure. Mutations in polycystin-1 (PC1) underlie most cases of ADPKD, but the function of PC1 has remained poorly understood. No preventive treatment for this disease is available. Here, we show that the cytoplasmic tail of PC1 interacts with tuberin, and the mTOR pathway is inappropriately activated in cyst-lining epithelial cells in human ADPKD patients and mouse models. Rapamycin, an inhibitor of mTOR, is highly effective in reducing renal cystogenesis in two independent mouse models of PKD. Treatment of human ADPKD transplant-recipient patients with rapamycin results in a significant reduction in native polycystic kidney size. These results indicate that PC1 has an important function in the regulation of the mTOR pathway and that this pathway provides a target for medical therapy of ADPKD.


Assuntos
Rim/patologia , Doenças Renais Policísticas/prevenção & controle , Proteínas Quinases/metabolismo , Animais , Apoptose/efeitos dos fármacos , Humanos , Camundongos , Doenças Renais Policísticas/patologia , Proteínas Quinases/efeitos dos fármacos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Canais de Cátion TRPP
15.
J Am Soc Nephrol ; 15(12): 3035-43, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15579506

RESUMO

Gene targeting has been used to create a variety of lines of mice with Pkd1 mutations that share many common features. Homozygous Pkd1 mutants invariably develop pancreatic and renal cysts if they survive to day 15.5 post coitum and die in either the fetal or the perinatal period. In contrast, mice with heterozygous mutations of Pkd1 are generally normal and have few if any renal cysts. These features have limited the utility of these models as tools to study the pathogenesis of cyst formation and the effect of various therapeutic interventions on disease progression. This report describes a new line of mice with a floxed allele of Pkd1 (Pkd1(cond)) that has an FRT-flanked neomycin cassette inserted into intron 1 and lox P sites inserted into intron 1 and intron 4. The Pkd1(cond) allele is fully functional, and homozygotes are viable and healthy. It is shown that the lox P and FRT sites can be selectively induced to recombine to produce two new alleles, Pkd1(del2-4) and Pkd1(cond-Deltaneo), by crossing to animals that express either the cre or FLPe recombinase, respectively. It is found that Pkd1(del2-4) allele functions as a true null, whereas presence or absence of the neomycin gene has no functional effects. It also is shown that somatic loss of Pkd1 results in renal and hepatic cysts. This new line of mice will be invaluable in the study of Pkd1 biology and serve as a powerful new tool that can be used to study the pathogenesis of autosomal dominant polycystic kidney disease.


Assuntos
Mutagênese Insercional/métodos , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/fisiopatologia , Proteínas/genética , Proteínas/metabolismo , Alelos , Animais , Modelos Animais de Doenças , Rim/patologia , Fígado/patologia , Camundongos , Camundongos Mutantes , Neomicina , Rim Policístico Autossômico Dominante/patologia , Canais de Cátion TRPP
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA