Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Pediatr Nephrol ; 34(5): 873-881, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30413946

RESUMO

BACKGROUND: Cystinosis is an ultrarare disorder caused by mutations of the cystinosin (CTNS) gene, encoding a cystine-selective efflux channel in the lysosomes of all cells of the body. Oral therapy with cysteamine reduces intralysosomal cystine accumulation and slows organ deterioration but cannot reverse renal Fanconi syndrome nor prevent the eventual need for renal transplantation. A definitive therapeutic remains elusive. About 15% of cystinosis patients worldwide carry one or more nonsense mutations that halt translation of the CTNS protein. Aminoglycosides such as geneticin (G418) can bind to the mammalian ribosome, relax translational fidelity, and permit readthrough of premature termination codons to produce full-length protein. METHODS: To ascertain whether aminoglycosides permit readthrough of the most common CTNS nonsense mutation, W138X, we studied the effect of G418 on patient fibroblasts. RESULTS: G418 treatment induced translational readthrough of CTNSW138X constructs transfected into HEK293 cells and expression of full-length endogenous CTNS protein in homozygous W138X fibroblasts. CONCLUSIONS: Reduction in intracellular cystine indicates that the CTNS protein produced is functional as a cystine transporter. Interestingly, similar effects were seen even in W138X compound heterozygotes. These studies establish proof-of-principle for the potential of aminoglycosides to treat cystinosis and possibly other monogenic diseases caused by nonsense mutations.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/genética , Cistinose/tratamento farmacológico , Fibroblastos/efeitos dos fármacos , Gentamicinas/farmacologia , Terminação Traducional da Cadeia Peptídica/efeitos dos fármacos , Códon sem Sentido , Cistina/metabolismo , Cistinose/genética , Fibroblastos/metabolismo , Vetores Genéticos/genética , Gentamicinas/uso terapêutico , Células HEK293 , Humanos , Terminação Traducional da Cadeia Peptídica/genética , Plasmídeos/genética , RNA Mensageiro/análise , Proteínas Recombinantes/genética , Transfecção
2.
J Biol Chem ; 290(4): 2279-88, 2015 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-25331950

RESUMO

The mammalian kidney is derived from progenitor cells in intermediate mesoderm. During embryogenesis, progenitor cells expressing the Wilms tumor suppressor gene, WT1, are induced to differentiate in response to WNT signals from the ureteric bud. In hereditary Wilms tumors, clonal loss of WT1 precludes the ß-catenin pathway response and leads to precancerous nephrogenic rests. We hypothesized that WT1 normally primes progenitor cells for differentiation by suppressing the enhancer of zeste2 gene (EZH2), involved in epigenetic silencing of differentiation genes. In human amniotic fluid-derived mesenchymal stem cells, we show that exogenous WT1B represses EZH2 transcription. This leads to a dramatic decrease in the repressive lysine 27 trimethylation mark on histone H3 that silences ß-catenin gene expression. As a result, amniotic fluid mesenchymal stem cells acquire responsiveness to WNT9b and increase expression of genes that mark the onset of nephron differentiation. Our observations suggest that biallelic loss of WT1 sustains the inhibitory histone methylation state that characterizes Wilms tumors.


Assuntos
Âmnio/metabolismo , Epigênese Genética , Histonas/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Proteínas WT1/metabolismo , beta Catenina/genética , Motivos de Aminoácidos , Células Cultivadas , Metilação de DNA , Proteína Potenciadora do Homólogo 2 de Zeste , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Inativação Gênica , Humanos , Células-Tronco Mesenquimais/citologia , Gravidez , Células-Tronco/citologia , Tumor de Wilms/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
3.
Pediatr Nephrol ; 29(4): 705-10, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24414605

RESUMO

The mammalian kidney arises from OSR1(+) progenitor cells in the intermediate mesoderm. However, these cells must acquire unique properties before they can respond to inductive signals that launch the differentiation program. Recent data indicate that the transcription factor, WT1, plays a master role in this transition. Interestingly, some of these embryonic nephron progenitor cells are retained in the adult organ where they may participate in tissue regeneration after acute kidney injury. A better understanding of the biology of these cells may one day allow progenitor cell-based therapeutic strategies to help regenerate damaged adult nephrons.


Assuntos
Diferenciação Celular/fisiologia , Rim/citologia , Rim/embriologia , Regeneração/fisiologia , Células-Tronco/citologia , Animais , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Humanos , Proteínas WT1/genética , Proteínas WT1/metabolismo
4.
Kidney Int ; 80(2): 146-53, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21389971

RESUMO

During murine kidney development, canonical WNT signaling is highly active in tubules until about embryonic days E16-E18. At this time, ß-catenin transcriptional activity is progressively restricted to the nephrogenic zone. The cilial protein genes PKD1 and PKD2 are known to be mutated in autosomal dominant polycystic kidney disease (ADPKD), and previous studies proposed that these mutations could lead to a failure to suppress canonical WNT signaling activity. Several in vitro studies have found a link between cilial signaling and ß-catenin regulation, suggesting that aberrant activity might contribute to the cystic phenotype. To study this, we crossed T-cell factor (TCF)/ß-catenin-lacZ reporter mice with mice having Pkd1 or Pkd2 mutations and found that there was no ß-galactosidase staining in cells lining the renal cysts. Thus, suppression of canonical WNT activity, defined by the TCF/ß-catenin-lacZ reporter, is normal in these two different models of polycystic kidney disease. Hence, excessive ß-catenin transcriptional activity may not contribute to cystogenesis in these models of ADPKD.


Assuntos
Cistos/etiologia , Rim Policístico Autossômico Dominante/metabolismo , Fatores de Transcrição TCF/genética , beta Catenina/metabolismo , Fatores Etários , Animais , Túbulos Renais/metabolismo , Camundongos , Camundongos Mutantes , Modelos Biológicos , Mutação , Rim Policístico Autossômico Dominante/genética , Fatores de Transcrição TCF/metabolismo , Canais de Cátion TRPP/genética , Transcrição Gênica , Proteínas Wnt/metabolismo
5.
Stem Cells Int ; 2015: 391043, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26089915

RESUMO

During development, nephron progenitor cells (NPC) are induced to differentiate by WNT9b signals from the ureteric bud. Although nephrogenesis ends in the perinatal period, acute kidney injury (AKI) elicits repopulation of damaged nephrons. Interestingly, embryonic NPC infused into adult mice with AKI are incorporated into regenerating tubules. Since WNT/ß-catenin signaling is crucial for primary nephrogenesis, we reasoned that it might also be needed for the endogenous repair mechanism and for integration of exogenous NPC. When we examined glycerol-induced AKI in adult mice bearing a ß-catenin/TCF reporter transgene, endogenous tubular cells reexpressed the NPC marker, CD24, and showed widespread ß-catenin/TCF signaling. We isolated CD24+ cells from E15 kidneys of mice with the canonical WNT signaling reporter. 40% of cells responded to WNT3a in vitro and when infused into glycerol-injured adult, the cells exhibited ß-catenin/TCF reporter activity when integrated into damaged tubules. When embryonic CD24+ cells were treated with a ß-catenin/TCF pathway inhibitor (IWR-1) prior to infusion into glycerol-injured mice, tubular integration of cells was sharply reduced. Thus, the endogenous canonical ß-catenin/TCF pathway is reactivated during recovery from AKI and is required for integration of exogenous embryonic renal progenitor cells into damaged tubules. These events appear to recapitulate the WNT-dependent inductive process which drives primary nephrogenesis.

6.
PLoS One ; 7(8): e42840, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22912749

RESUMO

Cystinosis is a rare disease caused by homozygous mutations of the CTNS gene, encoding a cystine efflux channel in the lysosomal membrane. In Ctns knockout mice, the pathologic intralysosomal accumulation of cystine that drives progressive organ damage can be reversed by infusion of wildtype bone marrow-derived stem cells, but the mechanism involved is unclear since the exogeneous stem cells are rarely integrated into renal tubules. Here we show that human mesenchymal stem cells, from amniotic fluid or bone marrow, reduce pathologic cystine accumulation in co-cultured CTNS mutant fibroblasts or proximal tubular cells from cystinosis patients. This paracrine effect is associated with release into the culture medium of stem cell microvesicles (100-400 nm diameter) containing wildtype cystinosin protein and CTNS mRNA. Isolated stem cell microvesicles reduce target cell cystine accumulation in a dose-dependent, Annexin V-sensitive manner. Microvesicles from stem cells expressing CTNS(Red) transfer tagged CTNS protein to the lysosome/endosome compartment of cystinotic fibroblasts. Our observations suggest that exogenous stem cells may reprogram the biology of mutant tissues by direct microvesicle transfer of membrane-associated wildtype molecules.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Cistina/metabolismo , Cistinose/metabolismo , Cistinose/patologia , Exossomos/metabolismo , Células-Tronco Mesenquimais/citologia , Sistemas de Transporte de Aminoácidos Neutros/genética , Animais , Cistinose/genética , Cistinose/cirurgia , Fibroblastos/metabolismo , Humanos , Lisossomos/metabolismo , Transplante de Células-Tronco Mesenquimais , Camundongos , Mutação , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
7.
Pediatr Dev Pathol ; 12(5): 347-54, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-18666806

RESUMO

Wilms tumor (WT) is the most frequent renal neoplasm of childhood; a myogenic component is observed in 5% to 10% of tumors. We demonstrate for the first time that myogenic WTs are associated with expression of PAX3, a transcription factor known to specify myoblast cell fate during muscle development. In a panel of 20 WTs, PAX3 was identified in 13 of 13 tumor samples with myogenic histopathology but was absent in 7 of 7 tumors lacking a myogenic component. Furthermore, we show that PAX3 is expressed in the metanephric mesenchyme and stromal compartment of developing mouse kidney. Modulation of endogenous PAX3 expression in human embryonic kidney (HEK293) cells influenced cell migration in in vitro assays. Mutations of WT1 were consistently associated with PAX3 expression in WTs, and modulation of WT1 expression in HEK293 cells was inversely correlated with the level of endogenous PAX3 protein. We demonstrate abundant PAX3 and absence of PAX2 expression in a novel cell line (WitP3) isolated from the stromal portion of a WT bearing a homozygous deletion of the WT1 gene. We hypothesize that PAX3 sets stromal cell fate in developing kidney but is normally suppressed by WT1 during the mesenchyme-to-epithelium transition leading to nephrogenesis. Loss of WT1 permits aberrant PAX3 expression in a subset of WTs with myogenic phenotype.


Assuntos
Neoplasias Renais/metabolismo , Rim/metabolismo , Fatores de Transcrição Box Pareados/biossíntese , Tumor de Wilms/metabolismo , Animais , Western Blotting , Diferenciação Celular , Linhagem Celular Tumoral , Matriz Extracelular/metabolismo , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Rim/embriologia , Neoplasias Renais/genética , Neoplasias Renais/patologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Mesoderma/metabolismo , Camundongos , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados/genética , Fenótipo , Reação em Cadeia da Polimerase , RNA Interferente Pequeno , Tumor de Wilms/genética , Tumor de Wilms/patologia
8.
Am J Physiol Renal Physiol ; 293(2): F494-500, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17494089

RESUMO

The canonical WNT signaling pathway plays a crucial role in patterning of the embryo during development, but little is known about the specific developmental events which are under WNT control. To understand more about how the WNT pathway orchestrates mammalian organogenesis, we studied the canonical beta-catenin-mediated WNT signaling pathway in kidneys of mice bearing a beta-catenin-responsive TCF/betaGal reporter transgene. In metanephric kidney, intense canonical WNT signaling was evident in epithelia of the branching ureteric bud and in nephrogenic mesenchyme during its transition into renal tubules. WNT signaling activity is rapidly downregulated in maturing nephrons and becomes undetectable in postnatal kidney. Sites of TCF/betaGal activity are in proximity to the known sites of renal WNT2b and WNT4 expression, and these WNTs stimulate TCF reporter activity in kidney cell lines derived from ureteric bud and metanephric mesenchyme lineages. When fetal kidney explants from HoxB7/GFP mice were exposed to the canonical WNT signaling pathway inhibitor, Dickkopf-1, arborization of the ureteric bud was significantly reduced. We conclude that restricted zones of intense canonical WNT signaling drive branching nephrogenesis in fetal kidney.


Assuntos
Rim/embriologia , Transdução de Sinais/fisiologia , Proteínas Wnt/fisiologia , Animais , Linhagem Celular , DNA Complementar/biossíntese , DNA Complementar/genética , Regulação para Baixo/fisiologia , Células Epiteliais/fisiologia , Genes Reporter/genética , Humanos , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Rim/metabolismo , Túbulos Renais Coletores/metabolismo , Óperon Lac/genética , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Fatores do Domínio POU/genética , Transfecção , Ureter/embriologia , beta Catenina/metabolismo
9.
Hum Mol Genet ; 15(24): 3520-8, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17082250

RESUMO

Mutations in PKD1 cause dominant polycystic kidney disease (PKD), characterized by large fluid-filled kidney cysts in adult life, but the molecular mechanism of cystogenesis remains obscure. Ostrom et al. [Dev. Biol., 219, 250-258 (2000)] showed that reduced dosage of Pax2 caused increased apoptosis, and ameliorated cystogenesis in Cpk mutant mice with recessive PKD. Pax2 is expressed in condensing metanephrogenic mesenchyme and arborizing ureteric bud, and plays an important role in kidney development. Transient Pax2 expression during fetal kidney mesenchyme-to-epithelial transition, as well as in nascent tubules, is followed by marked down-regulation of Pax2 expression. Here, we show that in humans with PKD, as well as in Pkd1(del34/del34) mutant mice, Pax2 was expressed in cyst epithelial cells, and facilitated cyst growth in Pkd1(del34/del34) mutant mice. In Pkd1(del34/del34) mutant kidneys, the expression of Pax2 persisted in nascent collecting ducts. In contrast, homozygous Pkd1(del34/del34) fetal mice carrying mutant Pax2 exhibited ameliorated cyst growth, although reduced cystogenesis was not associated with increased apoptosis. Pax2 expression was attenuated in nascent collecting ducts and absent from remnant cysts of Pkd1(del34/del34)/Pax2(1Neu/+) mutant mice. To investigate whether the Pkd1 gene product, Polycystin-1, regulates Pax2, MDCK cells were engineered constitutively expressing wild-type Pkd1; Pax2 protein levels and promoter activity were both repressed in MDCK cells over-expressing Pkd1, but not in cells without transgenic Pkd1. These data suggest that polycystin-1-deficient tubular epithelia persistently express Pax2 in ADPKD, and that Pax2 or its pathway may be an appropriate target for the development of novel therapies for ADPKD.


Assuntos
Dosagem de Genes , Fator de Transcrição PAX2/genética , Rim Policístico Autossômico Dominante/genética , Animais , Apoptose , Western Blotting , Linhagem Celular , Proliferação de Células , Heterozigoto , Homozigoto , Humanos , Imuno-Histoquímica , Rim/metabolismo , Rim/patologia , Camundongos , Camundongos Mutantes , Fator de Transcrição PAX2/metabolismo , Diester Fosfórico Hidrolases/genética , Diester Fosfórico Hidrolases/metabolismo , Rim Policístico Autossômico Dominante/metabolismo , Rim Policístico Autossômico Dominante/patologia , Pirofosfatases/genética , Pirofosfatases/metabolismo
10.
Medicina (B.Aires) ; 59(2): 133-7, 1999. ilus
Artigo em Espanhol | LILACS | ID: lil-234492

RESUMO

La poliquistosis renal autosómica (ADPKD) es una enfermedad hereditaria que presenta heterogeneidad genética. Al menos tres genes son responsables del desarrollo de la enfermedad: PKD1 en el cromosoma 16p 13.3, PKD2 en 4q21 y PKD3, aún no localizado. A partir de la descripción de la secuencia del gen PKD1, el interés general se volcó a la búsqueda de mutaciones causantes de la enfermedad. La mayoría de las mutaciones halladas es de diverso origen y localiza a lo largo del gen, no pudiendo hallarse correlación fenotípica alguna. En este trabajo se describe el hallazgo de una mutación en el exón 44 del gen PKD1 en una familia previamente caracterizada por análisis de ligamiento. La mutación consiste en una sustitución de una base C por una T en la posición 12220 originado un codón stop donde se produce la mutación. Esto llevaría a una terminación prematura en la traducción produciendo una proteína en la cual estaría ausente parte del extremo carboxílico.


Assuntos
Humanos , Adolescente , Recém-Nascido , Adulto , Ligação Genética , Mutação , Rim Policístico Autossômico Dominante/genética , Proteínas/genética , Códon de Terminação/genética , Reação em Cadeia da Polimerase , Análise de Sequência de DNA
11.
Medicina [B.Aires] ; 59(2): 133-7, 1999. ilus
Artigo em Espanhol | BINACIS | ID: bin-16206

RESUMO

La poliquistosis renal autosómica (ADPKD) es una enfermedad hereditaria que presenta heterogeneidad genética. Al menos tres genes son responsables del desarrollo de la enfermedad: PKD1 en el cromosoma 16p 13.3, PKD2 en 4q21 y PKD3, aún no localizado. A partir de la descripción de la secuencia del gen PKD1, el interés general se volcó a la búsqueda de mutaciones causantes de la enfermedad. La mayoría de las mutaciones halladas es de diverso origen y localiza a lo largo del gen, no pudiendo hallarse correlación fenotípica alguna. En este trabajo se describe el hallazgo de una mutación en el exón 44 del gen PKD1 en una familia previamente caracterizada por análisis de ligamiento. La mutación consiste en una sustitución de una base C por una T en la posición 12220 originado un codón stop donde se produce la mutación. Esto llevaría a una terminación prematura en la traducción produciendo una proteína en la cual estaría ausente parte del extremo carboxílico. (AU)


Assuntos
Humanos , Adolescente , Recém-Nascido , Adulto , Rim Policístico Autossômico Dominante/genética , Mutação , Proteínas/genética , Ligação Genética , Reação em Cadeia da Polimerase , Códon de Terminação/genética , Análise de Sequência de DNA
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa