Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Methods Cell Biol ; 181: 73-85, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38302245

RESUMO

The gut microbiota is a complex community of different microbial species that influence many aspects of health. Consequently, shifts in the composition of gut microbiome have been proposed to exert negative effects on the host physiology, leading to the pathogenesis of various age-related disorders, including cardiovascular and neurological diseases, type 2 diabetes, obesity, non-alcoholic liver disease, and other pathological conditions. Thus, understanding how the gut microbiota influences the aging-related decline is particularly topical. Advances in next-generation sequencing techniques, together with mechanistic experiments in animal models, have provided substantial improvements in microbiome analysis. However, standardization and best practices are needed to limit experimental variation between different studies. Here, we detail a simple method for microbiota composition analysis in mouse fecal samples using 16S rRNA next-generation sequencing.


Assuntos
Diabetes Mellitus Tipo 2 , Microbioma Gastrointestinal , Microbiota , Camundongos , Animais , RNA Ribossômico 16S/genética , Microbiota/genética , Fezes , Microbioma Gastrointestinal/genética
2.
Cell Death Dis ; 14(1): 60, 2023 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-36702832

RESUMO

The spindle assembly checkpoint (SAC) is an essential mechanism that ensures the accurate chromosome segregation during mitosis, thus preventing genomic instability. Deubiquitinases have emerged as key regulators of the SAC, mainly by determining the fate of proteins during cell cycle progression. Here, we identify USP49 deubiquitinase as a novel regulator of the spindle checkpoint. We show that loss of USP49 in different cancer cell lines impairs proliferation and increases aneuploidy. In addition, USP49-depleted cells overcome the arrest induced by the SAC in the presence of nocodazole. Finally, we report new binding partners of USP49, including ribophorin 1, USP44, and different centrins.


Assuntos
Pontos de Checagem da Fase M do Ciclo Celular , Fuso Acromático , Humanos , Fuso Acromático/metabolismo , Aneuploidia , Mitose , Enzimas Desubiquitinantes/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Ubiquitina Tiolesterase/genética , Ubiquitina Tiolesterase/metabolismo
3.
Nat Ecol Evol ; 3(1): 87-95, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30510174

RESUMO

Giant tortoises are among the longest-lived vertebrate animals and, as such, provide an excellent model to study traits like longevity and age-related diseases. However, genomic and molecular evolutionary information on giant tortoises is scarce. Here, we describe a global analysis of the genomes of Lonesome George-the iconic last member of Chelonoidis abingdonii-and the Aldabra giant tortoise (Aldabrachelys gigantea). Comparison of these genomes with those of related species, using both unsupervised and supervised analyses, led us to detect lineage-specific variants affecting DNA repair genes, inflammatory mediators and genes related to cancer development. Our study also hints at specific evolutionary strategies linked to increased lifespan, and expands our understanding of the genomic determinants of ageing. These new genome sequences also provide important resources to help the efforts for restoration of giant tortoise populations.


Assuntos
Envelhecimento/genética , Genoma , Tartarugas/genética , Animais , Reparo do DNA/genética , Evolução Molecular , Células HEK293 , Humanos , Mediadores da Inflamação , Masculino , Neoplasias/genética , Filogenia , Densidade Demográfica
4.
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
5.
Methods Mol Biol ; 1731: 261-269, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29318560

RESUMO

Proteases play key roles in the execution and regulation of most if not all biological functions, and alterations in their activity, expression, or location are associated with multiple pathological conditions, including cancer and aging. In this regard, the use of RNA interference-based approaches to specifically target the expression of individual proteases constitutes an invaluable tool to identify enzymes involved in central aspects of these processes and to explore their potential as targets of therapeutic interventions. Here we describe simple protocols to optimize and monitor the specific silencing of cancer- and aging-related proteases.


Assuntos
Envelhecimento/metabolismo , Proteínas de Membrana/genética , Metaloendopeptidases/genética , Neoplasias/patologia , Interferência de RNA , Proteases Específicas de Ubiquitina/genética , Envelhecimento/genética , Apoptose/genética , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes/instrumentação , Técnicas de Silenciamento de Genes/métodos , Vetores Genéticos/genética , Humanos , Lentivirus/genética , Proteínas de Membrana/metabolismo , Metaloendopeptidases/metabolismo , Neoplasias/genética , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Proteases Específicas de Ubiquitina/metabolismo
6.
J Biol Chem ; 293(6): 2183-2194, 2018 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-29273634

RESUMO

Deubiquitinases are proteases with a wide functional diversity that profoundly impact multiple biological processes. Among them, the ubiquitin-specific protease 36 (USP36) has been implicated in the regulation of nucleolar activity. However, its functional relevance in vivo has not yet been fully described. Here, we report the generation of an Usp36-deficient mouse model to examine the function of this enzyme. We show that Usp36 depletion is lethal in preimplantation mouse embryos, where it blocks the transition from morula to blastocyst during embryonic development. USP36 reduces the ubiquitination levels and increases the stability of the DEAH-box RNA helicase DHX33, which is critically involved in ribosomal RNA synthesis and mRNA translation. In agreement with this finding, O-propargyl-puromycin incorporation experiments, Northern blot, and electron microscopy analyses demonstrated the role of USP36 in ribosomal RNA and protein synthesis. Finally, we show that USP36 down-regulation alters cell proliferation in human cancer cells by inducing both apoptosis and cell cycle arrest, and that reducing DHX33 levels through short hairpin RNA interference has the same effect. Collectively, these results support that Usp36 is essential for cell and organism viability because of its role in ribosomal RNA processing and protein synthesis, which is mediated, at least in part, by regulating DHX33 stability.


Assuntos
Blastocisto , RNA Helicases DEAD-box/química , Enzimas Desubiquitinantes/fisiologia , RNA Helicases/química , Ubiquitina Tiolesterase/fisiologia , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Enzimas Desubiquitinantes/genética , Perda do Embrião , Humanos , Camundongos , Camundongos Knockout , Biossíntese de Proteínas , Estabilidade Proteica , RNA Ribossômico , Ubiquitina Tiolesterase/genética
7.
J Biol Chem ; 292(10): 4164-4175, 2017 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-28154181

RESUMO

KRAS is the most frequently mutated oncogene in human cancer, but its therapeutic targeting remains challenging. Here, we report a synthetic lethal screen with a library of deubiquitinases and identify USP39, which encodes an essential splicing factor, as a critical gene for the viability of KRAS-dependent cells. We show that splicing fidelity inhibitors decrease preferentially the proliferation rate of KRAS-active cells. Moreover, depletion of DHX38, encoding an USP39-interacting splicing factor, also reduces the viability of these cells. In agreement with these results, USP39 depletion caused a significant reduction in pre-mRNA splicing efficiency, as demonstrated through RNA-seq experiments. Furthermore, we show that USP39 is up-regulated in lung and colon carcinomas and its expression correlates with KRAS levels and poor clinical outcome. Accordingly, our work provides critical information for the development of splicing-directed antitumor treatments and supports the potential of USP39-targeting strategies as the basis of new anticancer therapies.


Assuntos
Neoplasias do Colo/patologia , Neoplasias Pulmonares/patologia , Mutação/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteases Específicas de Ubiquitina/metabolismo , Animais , Apoptose , Western Blotting , Proliferação de Células , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Nus , Prognóstico , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Taxa de Sobrevida , Células Tumorais Cultivadas , Proteases Específicas de Ubiquitina/genética , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Oncotarget ; 7(46): 74427-74434, 2016 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-27769071

RESUMO

Ubiquitin-Specific Proteases (USPs) are deubiquitinating enzymes frequently deregulated in human malignancies. Here, we show that USP54 is overexpressed in intestinal stem cells and demonstrate that its downregulation in colorectal carcinoma cells impedes tumorigenesis. We have generated mutant mice deficient for this deubiquitinase, which are viable and fertile, and protected against chemically-induced colorectal carcinoma. Furthermore, we show that USP54 is upregulated in human colon cancer and associates with poor prognosis. In agreement with these results, Usp54 downregulation in mouse melanoma cells inhibits lung metastasis formation. Collectively, this work has uncovered the pro-tumorigenic properties of USP54, highlighting the importance of deubiquitinating enzymes as promising targets for the development of specific anti-cancer therapies.


Assuntos
Neoplasias Colorretais/genética , Expressão Gênica , Células-Tronco Neoplásicas/metabolismo , Proteases Específicas de Ubiquitina/genética , Animais , Biomarcadores Tumorais , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Progressão da Doença , Desenvolvimento Embrionário/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Knockout , Invasividade Neoplásica , Metástase Neoplásica , Proteases Específicas de Ubiquitina/metabolismo
10.
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
11.
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.

12.
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
13.
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
14.
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
15.
J Am Soc Nephrol ; 23(1): 63-72, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22021707

RESUMO

ß-Catenin/Wnt signaling is essential during early inductive stages of kidney development, but its role during postinductive stages of nephron development and maturation is not well understood. In this study, we used Pax8Cre mice to target ß-catenin deficiency to renal epithelial cells at the late S-shaped body stage and the developing collecting ducts. The conditional ß-catenin knockout mice formed abnormal kidneys and had reduced renal function. The kidneys were hypoplastic with a thin cortex; a superficial layer of tubules was missing. A high proportion of glomeruli had small, underdeveloped capillary tufts. In these glomeruli, well differentiated podocytes replaced parietal epithelial cells in Bowman's capsule; capillaries toward the outer aspect of these podocytes mimicked the formation of glomerular capillaries. Tracing nephrogenesis in embryonic conditional ß-catenin knockout mice revealed that these "parietal podocytes" derived from precursor cells in the parietal layer of the S-shaped body by direct lineage switch. Taken together, these findings demonstrate that ß-catenin/Wnt signaling is important during the late stages of nephrogenesis and for the lineage specification of parietal epithelial cells.


Assuntos
Rim/embriologia , Organogênese , Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Diferenciação Celular , Linhagem da Célula , Células Epiteliais/citologia , Rim/citologia , Rim/metabolismo , Camundongos , Camundongos Knockout , beta Catenina/genética
16.
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
17.
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
18.
Pediatr Nephrol ; 24(7): 1313-9, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19294427

RESUMO

We report on a child with X-linked nephrogenic diabetes insipidus (NDI) who developed Wilms tumor (WT). Nephrogenic diabetes insipidus is caused by mutations of the arginine vasopressin receptor (AVPR2) or aquaporin-II (AQP2) genes. Wilms tumor is also genetically heterogeneous and is associated with mutations of WT1 (15-20%), WTX (20-30%) and other loci. The boy presented at 5 months with failure to thrive, polyuria, hypernatremia and abdominal mass. Analysis of leukocyte DNA showed a novel missense mutation (Q174H) of the AVPR2 gene, which was not present in his mother. In cells (WitS) isolated from the tumor, WTX mRNA expression and coding sequence were intact. However, we identified a 44-kb homozygous deletion of the WT1 gene spanning exons 4 to 10. The WT1 deletion was not present in leukocyte DNA from the patient or his mother. We also noted strong beta-catenin (CTNNB1) expression in the tumor cells and identified a heterozygote missense Ser45Cys mutation of exon 3 of CTNNB1. However, the mutation was absent both in the constitutional DNA of the patient and his mother. The concurrence of WT and NDI has not been previously reported and may be unrelated. Nevertheless, this case nicely illustrates the sequence of events leading to sporadic Wilms tumor.


Assuntos
Diabetes Insípido Nefrogênico/complicações , Doenças Genéticas Ligadas ao Cromossomo X/complicações , Neoplasias Renais/complicações , Tumor de Wilms/complicações , Sequência de Bases , Aberrações Cromossômicas , Análise Mutacional de DNA , Diabetes Insípido Nefrogênico/genética , Diabetes Insípido Nefrogênico/patologia , Genes do Tumor de Wilms , Doenças Genéticas Ligadas ao Cromossomo X/genética , Doenças Genéticas Ligadas ao Cromossomo X/patologia , Humanos , Imuno-Histoquímica , Lactente , Neoplasias Renais/genética , Neoplasias Renais/patologia , Masculino , Mutação de Sentido Incorreto , Receptores de Vasopressinas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tumor de Wilms/genética , Tumor de Wilms/patologia , beta Catenina/genética
19.
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
20.
Cancer Lett ; 265(1): 148-55, 2008 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-18439754

RESUMO

PAX genes are frequently overexpressed in human cancer tissue and appear to contribute to the tumor phenotype, suggesting that they may be potential targets for cancer therapy. In particular, aberrant PAX2 expression has been reported in a high proportion of primary tumors, including the majority of renal cell carcinomas (RCC). We recently demonstrated that PAX2 suppresses cisplatin-induced apoptosis in cultured RCC cells. We hypothesized that silencing of PAX2 expression might partially overcome the notorious resistance of renal cell carcinomas to chemotherapy in vivo. In this report, we show that a PAX2 shRNA successfully knocks down PAX2 mRNA and protein levels in an RCC cell line (ACHN). ACHN cells stably transfected with shRNAs targeted against the PAX2 homeodomain are 3-6-fold more susceptible to cisplatin-induced caspase-3 activation than control ACHN cells line. Furthermore, growth of subcutaneous ACHN/shPAX2 xenografts in nude mice is significantly more responsive to cisplatin therapy than control ACHN cell tumors. Our observations validate PAX2 as a potential therapeutic gene target in renal cancer and suggest that adjunctive PAX2 knockdown may enhance the efficacy of other chemotherapeutic agents.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma de Células Renais/terapia , Cisplatino/uso terapêutico , Neoplasias Renais/terapia , Fator de Transcrição PAX2/metabolismo , Animais , Apoptose/efeitos dos fármacos , Sequência de Bases , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Ativação Enzimática , Inativação Gênica , Humanos , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Transplante de Neoplasias , Fator de Transcrição PAX2/genética , RNA Interferente Pequeno/genética , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...