Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
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 ; 291(8): 3785-95, 2016 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-26655220

RESUMO

Hereditary forms of Wilms arise from developmentally arrested clones of renal progenitor cells with biallelic mutations of WT1; recently, it has been found that Wilms tumors may also be associated with biallelic mutations in DICER1 or DROSHA, crucial for miRNA biogenesis. We have previously shown that a critical role for WT1 during normal nephrogenesis is to suppress transcription of the Polycomb group protein, EZH2, thereby de-repressing genes in the differentiation cascade. Here we show that WT1 also suppresses translation of EZH2. All major WT1 isoforms induce an array of miRNAs, which target the 3' UTR of EZH2 and other Polycomb-associated transcripts. We show that the WT1(+KTS) isoform binds to the 5' UTR of EZH2 and interacts directly with the miRNA-containing RISC to enhance post-transcriptional inhibition. These observations suggest a novel mechanism through which WT1 regulates the transition from resting stem cell to activated progenitor cell during nephrogenesis. Our findings also offer a plausible explanation for the fact that Wilms tumors can arise either from loss of WT1 or loss of miRNA processing enzymes.


Assuntos
Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , Complexo Repressor Polycomb 2/biossíntese , Biossíntese de Proteínas , RNA Neoplásico/metabolismo , Proteínas WT1/metabolismo , Regiões 3' não Traduzidas , Regiões 5' não Traduzidas , Proteína Potenciadora do Homólogo 2 de Zeste , Humanos , Células-Tronco Mesenquimais/patologia , MicroRNAs/genética , Complexo Repressor Polycomb 2/genética , RNA Neoplásico/genética , Proteínas WT1/genética , Tumor de Wilms/genética , Tumor de Wilms/metabolismo , Tumor de Wilms/patologia
3.
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.

4.
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
5.
Hum Mol Genet ; 20(21): 4167-74, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21821672

RESUMO

Human nephrons are formed during fetal life through an interaction between the branching ureteric bud and progenitor cells. The wide variation in final nephron number has been attributed to allelic variants of genes regulating ureteric bud arborization. Here, we hypothesize that dysfunctional variants of the Odd-Skipped Related 1 (OSR1) gene which compromise the renal progenitor cell pool might also limit newborn kidney size and function. We show that OSR1 is expressed in human mesenchymal stem cells, the blastemal component of Wilms tumors and CD24+/CD133+ progenitor cells isolated from the mature kidney. We identified an OSR1(rs12329305(T)) allele in 6% of normal Caucasians which alters an exon2 splice enhancer. This variant is predicted to reduce spliceosome-binding affinity and stability of the OSR1 mRNA. In cultured cells, the OSR1(rs12329305)(T) allele produced no identifiable transcript. Normal Caucasian newborns from Montreal with the OSR1(rs12329305)(T) allele had kidney volume 11.8% smaller (P= 0.006) and cord blood cystatin C levels 12.6% higher (P = 0.005) than those with wild-type genotype. Effects of the OSR1(rs12329305)(T) allele are additive with genes that alter ureteric bud branching. Kidney volume was reduced more in newborns bearing both RET(rs1800860)(A) and OSR1(rs12329305)(T) alleles (22%, P= 0.0008) and cystatin C was increased by 17% (P= 0.006) versus newborns with wild-type alleles. Although only two subjects had PAX2(rs11599825)(A) and OSR1(rs12329305)(T) alleles, kidney size was reduced by 27% and cystatin C was increased by 14% versus wild-types (P= NS).


Assuntos
Alelos , Rim/crescimento & desenvolvimento , Rim/patologia , Células-Tronco Mesenquimais/enzimologia , Mutação/genética , Proteínas Serina-Treonina Quinases/genética , Fatores de Transcrição/genética , Adulto , Animais , Animais Recém-Nascidos , Sequência de Bases , Separação Celular , Cistatina C/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Heterozigoto , Humanos , Hibridização In Situ , Recém-Nascido , Rim/enzimologia , Camundongos , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Tamanho do Órgão , Proteínas Serina-Treonina Quinases/metabolismo , RNA Mensageiro/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição/metabolismo , Cordão Umbilical/metabolismo , Tumor de Wilms/enzimologia , Tumor de Wilms/genética , Tumor de Wilms/patologia
6.
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
7.
J Am Soc Nephrol ; 21(4): 573-8, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20133479

RESUMO

The ureteric bud (UB) expresses high levels of the EGF receptor (EGFR) during kidney development, but its function in this setting is unclear. Here, Egfr mRNA was abundant in medullary portions of the UB trunk but absent from the branching UB tips during embryogenesis. Homozygous Egfr knockout did not affect the pattern of UB arborization, but renal papillae were hypoplastic and exhibited widespread apoptosis of tubular cells. Because these EGFR-deficient mice die within 1 week of life, we targeted Egfr inactivation to the renal collecting ducts using Cre-lox technology with a Hoxb7-Cre transgene. This targeted inactivation of Egfr led to a thin renal medulla, and at 7 weeks of age, the mice had moderate polyuria and reduced urine-concentrating ability. At 30 to 33 weeks, water deprivation demonstrated a continued urine-concentrating defect despite similar levels of vasopressin between knockout mice and littermate controls. Taken together, these results suggest that unlike other tyrosine kinases expressed at the UB tip, EGFR functions primarily to drive elongation of the emerging collecting ducts and to optimize urine-concentrating ability.


Assuntos
Receptores ErbB/fisiologia , Túbulos Renais Coletores/embriologia , Animais , Regulação da Expressão Gênica no Desenvolvimento , Inativação Gênica , Camundongos , Camundongos Knockout
8.
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
9.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...