Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Kidney Int ; 103(1): 87-99, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36283570

RESUMEN

Widespread aberrant gene expression is a pathological hallmark of polycystic kidney disease (PKD). Numerous pathogenic signaling cascades, including c-Myc, Fos, and Jun, are transactivated. However, the underlying epigenetic regulators are poorly defined. Here we show that H3K27ac, an acetylated modification of DNA packing protein histone H3 that marks active enhancers, is elevated in mouse and human samples of autosomal dominant PKD. Using comparative H3K27ac ChIP-Seq analysis, we mapped over 16000 active intronic and intergenic enhancer elements in Pkd1-mutant mouse kidneys. We found that the cystic kidney epigenetic landscape resembles that of a developing kidney, and over 90% of upregulated genes in Pkd1-mutant kidneys are co-housed with activated enhancers in the same topologically associated domains. Furthermore, we identified an evolutionarily conserved enhancer cluster downstream of the c-Myc gene and super-enhancers flanking both Jun and Fos loci in mouse and human models of autosomal dominant PKD. Deleting these regulatory elements reduced c-Myc, Jun, or Fos abundance and suppressed proliferation and 3D cyst growth of Pkd1-mutant cells. Finally, inhibiting glycolysis and glutaminolysis or activating Ppara in Pkd1-mutant cells lowerd global H3K27ac levels and its abundance on c-Myc enhancers. Thus, our work suggests that epigenetic rewiring mediates the transcriptomic dysregulation in PKD, and the regulatory elements can be targeted to slow cyst growth.


Asunto(s)
Elementos de Facilitación Genéticos , Epigénesis Genética , Riñón Poliquístico Autosómico Dominante , Animales , Humanos , Ratones , Quistes/patología , Histonas/metabolismo , Riñón/patología , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/patología , Transducción de Señal
2.
Cell Metab ; 33(6): 1234-1247.e7, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33852874

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is a common monogenic disorder marked by numerous progressively enlarging kidney cysts. Mettl3, a methyltransferase that catalyzes the abundant N6-methyladenosine (m6A) RNA modification, is implicated in development, but its role in most diseases is unknown. Here, we show that Mettl3 and m6A levels are increased in mouse and human ADPKD samples and that kidney-specific transgenic Mettl3 expression produces tubular cysts. Conversely, Mettl3 deletion in three orthologous ADPKD mouse models slows cyst growth. Interestingly, methionine and S-adenosylmethionine (SAM) levels are also elevated in ADPKD models. Moreover, methionine and SAM induce Mettl3 expression and aggravate ex vivo cyst growth, whereas dietary methionine restriction attenuates mouse ADPKD. Finally, Mettl3 activates the cyst-promoting c-Myc and cAMP pathways through enhanced c-Myc and Avpr2 mRNA m6A modification and translation. Thus, Mettl3 promotes ADPKD and links methionine utilization to epitranscriptomic activation of proliferation and cyst growth.


Asunto(s)
Adenosina/análogos & derivados , Metionina/metabolismo , Metiltransferasas/metabolismo , Enfermedades Renales Poliquísticas/genética , Adenosina/metabolismo , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
3.
J Biol Chem ; 295(51): 17560-17572, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33453998

RESUMEN

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


Asunto(s)
Factor Nuclear 1-beta del Hepatocito/metabolismo , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Vía de Señalización Wnt , Animales , Proteína Axina/genética , Proteína Axina/metabolismo , Sitios de Unión , Células Epiteliales/citología , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Factor Nuclear 1-beta del Hepatocito/deficiencia , Factor Nuclear 1-beta del Hepatocito/genética , Histonas/metabolismo , Riñón/citología , Factor de Unión 1 al Potenciador Linfoide/antagonistas & inhibidores , Factor de Unión 1 al Potenciador Linfoide/genética , Metilación , Ratones , Ratones Noqueados , Mutagénesis , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Elementos Reguladores de la Transcripción/genética , Proteína Wnt3A/metabolismo , beta Catenina/metabolismo
4.
J Am Soc Nephrol ; 29(2): 518-531, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29021386

RESUMEN

microRNAs (miRNAs) are sequence-specific inhibitors of post-transcriptional gene expression. The physiologic function of these noncoding RNAs in postnatal renal tubules still remains unclear. Surprisingly, they appear to be dispensable for mammalian proximal tubule (PT) function. Here, we examined the effects of miRNA suppression in collecting ducts (CDs). To conclusively evaluate the role of miRNAs, we generated three mouse models with CD-specific inactivation of key miRNA pathway genes Dicer, Dgcr8, and the entire Argonaute gene family (Ago1, 2, 3, and 4). Characterization of these three mouse models revealed that inhibition of miRNAs in CDs spontaneously evokes a renal tubule injury-like response, which culminates in progressive tubulointerstitial fibrosis (TIF) and renal failure. Global miRNA profiling of microdissected renal tubules showed that miRNAs exhibit segmental distribution along the nephron and CDs. In particular, the expression of miR-200c is nearly 70-fold higher in CDs compared with PTs. Accordingly, miR-200s are downregulated in Dicer-KO CDs, its direct target genes Zeb1, Zeb2, and Snail2 are upregulated, and miRNA-depleted CDs undergo partial epithelial-to-mesenchymal transition (EMT). Thus, miRNAs are essential for CD homeostasis. Downregulation of CD-enriched miRNAs and the subsequent induction of partial EMT may be a new mechanism for TIF progression.


Asunto(s)
Epitelio/metabolismo , Epitelio/patología , Túbulos Renales Colectores/metabolismo , Túbulos Renales Colectores/patología , MicroARNs/genética , Animales , Proteínas Argonautas/genética , Línea Celular , ARN Helicasas DEAD-box/genética , Regulación hacia Abajo , Transición Epitelial-Mesenquimal/genética , Factores Eucarióticos de Iniciación/genética , Femenino , Fibrosis , Expresión Génica , Homeostasis/genética , Túbulos Renales Proximales/metabolismo , Masculino , Ratones , Ratones Noqueados , MicroARNs/antagonistas & inhibidores , Fenotipo , Proteínas de Unión al ARN/genética , Ribonucleasa III/genética , Factores de Transcripción de la Familia Snail/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética
5.
Nat Commun ; 8: 14395, 2017 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-28205547

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is the most frequent genetic cause of renal failure. Here we identify miR-17 as a target for the treatment of ADPKD. We report that miR-17 is induced in kidney cysts of mouse and human ADPKD. Genetic deletion of the miR-17∼92 cluster inhibits cyst proliferation and PKD progression in four orthologous, including two long-lived, mouse models of ADPKD. Anti-miR-17 treatment attenuates cyst growth in short-term and long-term PKD mouse models. miR-17 inhibition also suppresses proliferation and cyst growth of primary ADPKD cysts cultures derived from multiple human donors. Mechanistically, c-Myc upregulates miR-17∼92 in cystic kidneys, which in turn aggravates cyst growth by inhibiting oxidative phosphorylation and stimulating proliferation through direct repression of Pparα. Thus, miR-17 family is a promising drug target for ADPKD, and miR-17-mediated inhibition of mitochondrial metabolism represents a potential new mechanism for ADPKD progression.


Asunto(s)
MicroARNs/metabolismo , Mitocondrias/metabolismo , Riñón Poliquístico Autosómico Dominante/metabolismo , Animales , Proliferación Celular/fisiología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Eliminación de Gen , Humanos , Masculino , Ratones , Ratones Noqueados , MicroARNs/genética , Fosforilación , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/patología , Riñón Poliquístico Autosómico Dominante/terapia , Regulación hacia Arriba
6.
J Am Soc Nephrol ; 27(8): 2319-30, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26677864

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD), one of the most common monogenetic disorders, is characterized by kidney failure caused by bilateral renal cyst growth. MicroRNAs (miRs) have been implicated in numerous diseases, but the role of these noncoding RNAs in ADPKD pathogenesis is still poorly defined. Here, we investigated the role of miR-21, an oncogenic miR, in kidney cyst growth. We found that transcriptional activation of miR-21 is a common feature of murine PKD. Furthermore, compared with renal tubules from kidney samples of normal controls, cysts in kidney samples from patients with ADPKD had increased levels of miR-21. cAMP signaling, a key pathogenic pathway in PKD, transactivated miR-21 promoter in kidney cells and promoted miR-21 expression in cystic kidneys of mice. Genetic deletion of miR-21 attenuated cyst burden, reduced kidney injury, and improved survival of an orthologous model of ADPKD. RNA sequencing analysis and additional in vivo assays showed that miR-21 inhibits apoptosis of cyst epithelial cells, likely through direct repression of its target gene programmed cell death 4 Thus, miR-21 functions downstream of the cAMP pathway and promotes disease progression in experimental PKD. Our results suggest that inhibiting miR-21 is a potential new therapeutic approach to slow cyst growth in PKD.


Asunto(s)
MicroARNs/fisiología , Riñón Poliquístico Autosómico Dominante/etiología , Riñón Poliquístico Autosómico Dominante/patología , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Índice de Severidad de la Enfermedad
7.
J Biol Chem ; 290(41): 24793-805, 2015 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-26292219

RESUMEN

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


Asunto(s)
Regulación de la Expresión Génica , Factor Nuclear 1-beta del Hepatocito/metabolismo , MicroARNs/genética , ARN Largo no Codificante/genética , Animales , Secuencia de Bases , Células Epiteliales/metabolismo , Técnicas de Inactivación de Genes , Genómica , Células HeLa , Factor Nuclear 1-beta del Hepatocito/deficiencia , Factor Nuclear 1-beta del Hepatocito/genética , Proteínas de Homeodominio/genética , Humanos , Riñón/citología , Ratones , Mutación , Proteínas Represoras/genética , Canales Catiónicos TRPP/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc
8.
Am J Physiol Renal Physiol ; 307(3): F356-68, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24899057

RESUMEN

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.


Asunto(s)
Túbulos Renales Colectores/fisiología , Riñón Poliquístico Autosómico Recesivo/fisiopatología , Regiones Promotoras Genéticas/fisiología , Receptores de Superficie Celular/fisiología , Animales , Sistema Biliar/citología , Sistema Biliar/fisiología , Células Cultivadas , Células Epiteliales/citología , Células Epiteliales/fisiología , Factor Nuclear 1-beta del Hepatocito/fisiología , Túbulos Renales Colectores/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Riñón Poliquístico Autosómico Recesivo/genética , Regiones Promotoras Genéticas/genética , Receptores de Superficie Celular/genética , Sistema Urogenital/citología , Sistema Urogenital/fisiología
9.
Proc Natl Acad Sci U S A ; 110(26): 10765-70, 2013 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-23759744

RESUMEN

Polycystic kidney disease (PKD), the most common genetic cause of chronic kidney failure, is characterized by the presence of numerous, progressively enlarging fluid-filled cysts in the renal parenchyma. The cysts arise from renal tubules and are lined by abnormally functioning and hyperproliferative epithelial cells. Despite recent progress, no Food and Drug Administration-approved therapy is available to retard cyst growth. MicroRNAs (miRNAs) are short noncoding RNAs that inhibit posttranscriptional gene expression. Dysregulated miRNA expression is observed in PKD, but whether miRNAs are directly involved in kidney cyst formation and growth is not known. Here, we show that miR-17∼92, an oncogenic miRNA cluster, is up-regulated in mouse models of PKD. Kidney-specific transgenic overexpression of miR-17∼92 produces kidney cysts in mice. Conversely, kidney-specific inactivation of miR-17∼92 in a mouse model of PKD retards kidney cyst growth, improves renal function, and prolongs survival. miR-17∼92 may mediate these effects by promoting proliferation and through posttranscriptional repression of PKD genes Pkd1, Pkd2, and hepatocyte nuclear factor-1ß. These studies demonstrate a pathogenic role of miRNAs in mouse models of PKD and identify miR-17∼92 as a therapeutic target in PKD. Our results also provide a unique hypothesis for disease progression in PKD involving miRNAs and regulation of PKD gene dosage.


Asunto(s)
MicroARNs/genética , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/patología , Animales , Secuencia de Bases , Proliferación Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Túbulos Renales/metabolismo , Túbulos Renales/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Riñón Poliquístico Autosómico Dominante/metabolismo , Canales Catiónicos TRPP/genética , Regulación hacia Arriba/genética
10.
Drug Discov Today Dis Models ; 10(3): e137-e1743, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-25221607

RESUMEN

Polycystic kidney disease (PKD), the most common genetic cause of chronic renal failure, is characterized by the presence of numerous fluid-filled cysts in renal parenchyma. Despite recent progress, no FDA-approved therapy is available to retard cyst growth. Here, we review current evidence implicating two groups of miRNAs - the miR-17~92 cluster and miR-200s - in the pathogenesis of PKD. We present a new hypothesis for cyst growth involving miRNAs and regulation of PKD gene dosage. We propose that manipulating miRNA function in an attempt to normalize PKD gene dosage represents a novel therapeutic strategy in PKD.

11.
J Am Soc Nephrol ; 23(12): 1941-8, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23138483

RESUMEN

MicroRNAs (miRNAs) contribute to the regulation of early kidney development, but their role during later stages of renal tubule maturation is not well understood. Here, we found that ablation of the miRNA-processing enzyme Dicer from maturing renal tubules produces tubular and glomerular cysts in mice. Inactivation of Dicer is associated with downregulation of miR-200, a kidney-enriched miRNA family, and upregulation of the polycystic kidney disease gene Pkd1. Inhibition of miR-200 in cultured renal epithelial cells disrupted tubulogenesis and led to upregulation of Pkd1. Using bioinformatic and in vitro approaches, we found that miR-200b/c/429 induce post-transcriptional repression of Pkd1 through two conserved binding sites in the 3'-Untranslated regions of Pkd1. Overexpression of PKD1 in renal epithelial cells was sufficient to disrupt tubulogenesis and produce cyst-like structures. In conclusion, miRNAs are essential for the maturation of renal tubules, and Pkd1 is a target of miR-200. These results also suggest that miRNAs may modulate PKD1 gene dosage and play a role in the initiation of cystogenesis.


Asunto(s)
Túbulos Renales/crecimiento & desarrollo , MicroARNs/metabolismo , Canales Catiónicos TRPP/metabolismo , Animales , Animales Recién Nacidos , ARN Helicasas DEAD-box/genética , Dosificación de Gen , Humanos , Ratones , Ratones Transgénicos , Enfermedades Renales Poliquísticas/etiología , Ribonucleasa III/genética , Canales Catiónicos TRPP/genética
12.
J Urol ; 187(1): 272-8, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22114815

RESUMEN

PURPOSE: Although the cause of prune belly syndrome is unknown, familial evidence suggests a genetic component. Recently 2 nonfamilial cases of prune belly syndrome with chromosome 17q12 deletions encompassing the HNF1ß gene have made this a candidate gene for prune belly syndrome. To date, there has been no large-scale screening of patients with prune belly syndrome for HNF1ß mutations. We assessed the role of HNF1ß in prune belly syndrome by screening for genomic mutations with functional characterization of any detected mutations. MATERIALS AND METHODS: We studied patients with prune belly syndrome who were prospectively enrolled in our Pediatric Genitourinary DNA Repository since 2001. DNA from patient samples was amplified by polymerase chain reaction, sequenced for coding and splice regions of the HNF1ß gene, and compared to control databases. We performed functional assay testing of the ability of mutant HNF1ß to activate a luciferase construct with an HNF1ß DNA binding site. RESULTS: From 32 prune belly syndrome probands (30 males, 2 females) HNF1ß sequencing detected a missense mutation (V61G) in 1 child with prune belly syndrome. Absent in control databases, V61G was previously reported in 2 patients without prune belly syndrome who had congenital genitourinary anomalies. Functional testing showed similar luciferase activity compared to wild-type HNF1ß, suggesting the V61G substitution does not disturb HNF1ß function. CONCLUSIONS: One genomic HNF1ß mutation was detected in 3% of patients with prune belly syndrome but found to be functionally normal. Thus, functionally significant HNF1ß mutations are uncommon in prune belly syndrome, despite case reports of HNF1ß deletions. Further genetic study is necessary, as identification of the genetic basis of prune belly syndrome may ultimately lead to prevention and improved treatments for this rare but severe syndrome.


Asunto(s)
Factor Nuclear 1-beta del Hepatocito/genética , Mutación , Síndrome del Abdomen en Ciruela Pasa/genética , Femenino , Humanos , Masculino , Estudios Prospectivos
13.
Am J Physiol Renal Physiol ; 301(5): F1066-77, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21795643

RESUMEN

Onset of metabolic acidosis leads to a rapid and pronounced increase in expression of phosphoenolpyruvate carboxykinase (PEPCK) in rat renal proximal convoluted tubules. This adaptive response is modeled by treating a clonal line of porcine LLC-PK(1)-F(+) cells with an acidic medium (pH 6.9, 9 mM HCO(3)(-)). Measurement of the half-lives of PEPCK mRNA in cells treated with normal (pH 7.4, 26 mM HCO(3)(-)) and acidic medium established that the observed increase is due in part to stabilization of the PEPCK mRNA. The pH-responsive stabilization was reproduced in a Tet-responsive chimeric reporter mRNA containing the 3'-UTR of PEPCK mRNA. This response was lost by mutation of a highly conserved AU sequence that binds AUF1 and is the primary element that mediates the rapid turnover of PEPCK mRNA. However, siRNA knockdown of AUF1 had little effect on the basal levels and the pH-responsive increases in PEPCK mRNA and protein. Electrophoretic mobility shift assays established that purified recombinant HuR, another AU element binding protein, also binds with high affinity and specificity to multiple sites within the final 92-nucleotides of the 3'-UTR of the PEPCK mRNA, including the highly conserved AU-rich element. siRNA knockdown of HuR caused pronounced decreases in basal expression and the pH-responsive increases in PEPCK mRNA and protein. Therefore, basal expression and the pH-responsive stabilization of PEPCK mRNA in LLC-PK(1)-F(+) cells, and possibly in the renal proximal tubule, may require the remodeling of HuR and AUF1 binding to the elements that mediate the rapid turnover of PEPCK mRNA.


Asunto(s)
Proteínas ELAV/fisiología , Ribonucleoproteína Heterogénea-Nuclear Grupo D/fisiología , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Regiones no Traducidas 3' , Animales , Antibióticos Antineoplásicos/farmacología , Secuencia de Bases , Western Blotting , Dactinomicina/farmacología , Doxorrubicina/farmacología , Ensayo de Cambio de Movilidad Electroforética , Ribonucleoproteína Nuclear Heterogénea D0 , Concentración de Iones de Hidrógeno , Riñón/enzimología , Células LLC-PK1 , Ratones , Datos de Secuencia Molecular , ARN Mensajero/biosíntesis , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/metabolismo , Porcinos
14.
Proc Natl Acad Sci U S A ; 108(26): 10679-84, 2011 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-21670265

RESUMEN

Polycystic kidney disease (PKD) is a genetic disorder that is characterized by cyst formation in kidney tubules. PKD arises from abnormalities of the primary cilium, a sensory organelle located on the cell surface. Here, we show that the primary cilium of renal epithelial cells contains a protein complex comprising adenylyl cyclase 5/6 (AC5/6), A-kinase anchoring protein 150 (AKAP150), and protein kinase A. Loss of primary cilia caused by deletion of Kif3a results in activation of AC5 and increased cAMP levels. Polycystin-2 (PC2), a ciliary calcium channel that is mutated in human PKD, interacts with AC5/6 through its C terminus. Deletion of PC2 increases cAMP levels, which can be corrected by reexpression of wild-type PC2 but not by a mutant lacking calcium channel activity. Phosphodiesterase 4C (PDE4C), which catabolizes cAMP, is also located in renal primary cilia and interacts with the AKAP150 complex. Expression of PDE4C is regulated by the transcription factor hepatocyte nuclear factor-1ß (HNF-1ß), mutations of which produce kidney cysts. PDE4C is down-regulated and cAMP levels are increased in HNF-1ß mutant kidney cells and mice. Collectively, these findings identify PC2 and PDE4C as unique components of an AKAP complex in primary cilia and reveal a common mechanism for dysregulation of cAMP signaling in cystic kidney diseases arising from different gene mutations.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Cilios/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Enfermedades Renales Quísticas/metabolismo , Canales Catiónicos TRPP/metabolismo , Proteínas de Anclaje a la Quinasa A/genética , Animales , AMP Cíclico/metabolismo , Técnicas para Inmunoenzimas , Ratones , Mutación , Transducción de Señal
15.
Am J Physiol Renal Physiol ; 290(2): F313-8, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16144962

RESUMEN

Phosphoenolpyruvate carboxykinase (PEPCK) catalyzes a rate-limiting step in hepatic and renal gluconeogenesis. In the kidney, PEPCK expression is enhanced during metabolic acidosis and in response to ANG II and parathyroid hormone. The effect of the latter hormone is mediated, in part, by cAMP. Treatment of subconfluent cultures of LLC-PK1-F+ cells, a gluconeogenic line of porcine proximal tubule-like cells, with cAMP produces a pronounced increase in the level of PEPCK mRNA. The luciferase activity of pLuc/3'-PCK-1, a reporter construct that contains the 3'-UTR of the PEPCK mRNA, was increased three- to fourfold by coexpression of the catalytic subunit of protein kinase A (PKA). This result indicates that cAMP-dependent stabilization may contribute to the increased expression of PEPCK mRNA in LLC-PK1-F+ cells. Various pLuc/3' constructs containing different segments of the 3'-UTR of PEPCK mRNA were used to map the cAMP response to two segments that were previously shown to bind AUF1 and to function as instability elements. A tetracycline-responsive promoter system was used to quantify the effect of forskolin on the half-lives of chimeric beta-globin-PEPCK (TbetaG-PCK) mRNAs. The half-life of the labile betaG-PCK-1 mRNA was increased eightfold by addition of forskolin. In contrast, the half-lives of the constructs containing the individual instability elements were increased only twofold. Therefore, the multiple instability elements present within the 3'-UTR may function synergistically to mediate both the rapid degradation and the cAMP-induced stabilization of PEPCK mRNA. The latter process may result from a PKA-dependent phosphorylation of AUF1.


Asunto(s)
AMP Cíclico/fisiología , Estabilidad de Enzimas/efectos de los fármacos , Fosfoenolpiruvato Carboxiquinasa (ATP)/genética , Fosfoenolpiruvato Carboxiquinasa (ATP)/metabolismo , Regiones no Traducidas 3' , Animales , Colforsina/farmacología , AMP Cíclico/química , Proteínas Quinasas Dependientes de AMP Cíclico , Globinas/metabolismo , Semivida , Ribonucleoproteína Nuclear Heterogénea D0 , Ribonucleoproteína Heterogénea-Nuclear Grupo D/metabolismo , Células LLC-PK1 , Proteínas Serina-Treonina Quinasas/farmacología , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Elementos de Respuesta , Mapeo Restrictivo , Porcinos , Transfección
16.
J Biol Chem ; 280(31): 28272-80, 2005 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-15951444

RESUMEN

Phosphoenolpyruvate carboxykinase (PEPCK) is regulated solely by alterations in gene expression that involve changes in rates of PEPCK mRNA transcription and degradation. A tetracycline-responsive promoter system was used to quantify the half-life of various chimeric beta-globin-PEPCK (betaG-PCK) mRNAs in LLC-PK -F(+) cells. The control betaG mRNA was extremely stable (t(1/2) = 5 days). However, betaG-PCK-1 mRNA, which contains the entire 3'-UTR of the PEPCK mRNA, was degraded with a half-life of 1.2 h. RNase H treatment indicated that rapid deadenylation occurred concomitant with degradation of the betaG-PCK-1 mRNA. Previous studies indicate that PCK-7, a 50-nucleotide segment at the 3'-end of the 3'-UTR, binds an unidentified protein that may contribute to the rapid decay of the PEPCK mRNA. However, the chimeric betaG-PCK-7 mRNA has a half-life of 17 h. Inclusion of the adjacent PCK-6 segment, a 23-bp AU-rich region, produced the betaG-PCK-6/7 mRNA, which has a half-life of 3.6 h. The betaG-PCK-3 mRNA that contains the 3'-half of 3'-UTR was degraded with the same half-life. Surprisingly, the betaG-PCK-2 mRNA, containing the 5'-end of the 3'-UTR, was also degraded rapidly (t((1/2)) = 5.4 h). RNA gel shift analyses established that AUF1 (hnRNP D) binds to the PCK-7, PCK-6, and PCK-2 segments with high affinity and specificity. Mutational analysis indicated that AUF1 binds to a UUAUUUUAU sequence within PCK-6 and the stem-loop structure and adjacent CU-region of PCK-7. Thus, AUF1 binds to multiple destabilizing elements within the 3'-UTR that participate in the rapid turnover of the PEPCK mRNA.


Asunto(s)
Regiones no Traducidas 3'/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo D/metabolismo , Fosfoenolpiruvato Carboxiquinasa (ATP)/genética , ARN Mensajero/genética , Animales , Secuencia de Bases , Estabilidad de Enzimas , Semivida , Ribonucleoproteína Nuclear Heterogénea D0 , Ribonucleoproteína Heterogénea-Nuclear Grupo D/genética , Cinética , Masculino , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Ratas , Ratas Sprague-Dawley , Mapeo Restrictivo , Alineación de Secuencia , Eliminación de Secuencia , Homología de Secuencia de Ácido Nucleico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA