Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Front Oncol ; 13: 1091274, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37007076

RESUMO

Introduction: Wilms Tumor (WT), or nephroblastoma, is the most common pediatric kidney cancer. Most WTs display a "favorable" triphasic histology, in which the tumor is comprised of blastemal, stromal, and epithelial cell types. Blastemal predominance after neoadjuvant chemotherapy or diffuse anaplasia ("unfavorable" histology; 5-8%) portend a worse prognosis. Blastema likely provide the putative cancer stem cells (CSCs), which retain molecular and histologic features characteristic of nephron progenitor cells (NPCs), within WTs. NPCs arise in the metanephric mesenchyme (MM) and populate the cap mesenchyme (CM) in the developing kidney. WT blastemal cells, like NPCs, similarly express markers, SIX2 and CITED1. Tumor xenotransplantation is currently the only dependable method to propagate tumor tissue for research or therapeutic screening, since efforts to culture tumors in vitro as monolayers have invariably failed. Therefore, a critical need exists to propagate WT stem cells rapidly and efficiently for high-throughput, real-time drug screening. Methods: Previously, our lab developed niche conditions that support the propagation of murine NPCs in culture. Applying similar conditions to WTs, we assessed our ability to maintain key NPC "stemness" markers, SIX2, NCAM, and YAP1, and CSC marker ALDHI in cells from five distinct untreated patient tumors. Results: Accordingly, our culture conditions maintained the expression of these markers in cultured WT cells through multiple passages of rapidly dividing cells. Discussion: These findings suggest that our culture conditions sustain the WT blastemal population, as previously shown for normal NPCs. As a result, we have developed new WT cell lines and a multi-passage in vitro model for studying the blastemal lineage/CSCs in WTs. Furthermore, this system supports growth of heterogeneous WT cells, upon which potential drug therapies could be tested for efficacy and resistance.

2.
Differentiation ; 130: 7-15, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36527791

RESUMO

Fibroblast growth factors (Fgfs) have long been implicated in processes critical to embryonic development, such as cell survival, migration, and differentiation. Several mouse models of organ development ascribe a prosurvival requirement specifically to FGF8. Here, we explore the potential role of prosurvival FGF8 signaling in kidney development. We have previously demonstrated that conditional deletion of Fgf8 in the mesodermal progenitors that give rise to the kidney leads to renal aplasia in the mutant neonate. Deleterious consequences caused by loss of FGF8 begin to manifest by E14.5 when massive aberrant cell death occurs in the cortical nephrogenic zone in the rudimentary kidney as well as in the renal vesicles that give rise to the nephrons. To rescue cell death in the Fgf8 mutant kidney, we inactivate the genes encoding the pro-apoptotic factors BAK and BAX. In a wild-type background, the loss of Bak and Bax abrogates normal cell death and has minimal effect on renal development. However, in Fgf8 mutants, the combined loss of Bak and Bax rescues aberrant cell death in the kidneys and restores some measure of kidney development: 1) the nephron progenitor population is greatly increased; 2) some glomeruli form, which are rarely observed in Fgf8 mutants; and 3) kidney size is rescued by about 50% at E18.5. The development of functional nephrons, however, is not rescued. Thus, FGF8 signaling is required for nephron progenitor survival by regulating BAK/BAX and for subsequent steps involving, as yet, undefined roles in kidney development.


Assuntos
Rim , Néfrons , Camundongos , Animais , Feminino , Gravidez , Proteína X Associada a bcl-2/metabolismo , Néfrons/metabolismo , Apoptose , Diferenciação Celular , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Fator 8 de Crescimento de Fibroblasto/metabolismo
3.
Differentiation ; 130: 28-31, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36543010

RESUMO

The International Society of Differentiation was born from the First International Conference on Cell Differentiation conceived by D.V. and held in Nice, France in 1971. The conference also resulted in the creation of the journal of the Society named Differentiation. The Society advocates for the field of differentiation through the journal Differentiation, organizing and supporting international scientific conferences, honoring scientific achievements, and supporting trainees.


Assuntos
Diferenciação Celular , Sociedades Científicas , Sociedades Científicas/história
4.
PLoS One ; 13(5): e0197356, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29771971

RESUMO

Transplacental viral and parasitic infections have been shown to initiate an innate response in the mammalian embryo by increasing the expression of pro-inflammatory cytokines such as interferon-gamma (Ifng). However, the developmental consequences of an activated innate immunity and, in particular, the effects of induction of Ifng expression independent of infection have been largely overlooked. Here, we demonstrate in vivo that the conditional overexpression of Ifng in metanephric mesenchymal (MM) progenitors results in renal agenesis or hypoplasia. Cell death was observed in and around the MM region of E10.5-11.5 mutants where Ifng was constitutively expressed during early kidney development and resulted in a retardation of branching morphogenesis. Furthermore, isolated mutant or normal Ifng-treated metanephroi replicated this phenotype in culture, demonstrating the inherent nature of the aberrant morphogenesis. The expression of renal progenitor marker Sall1 was significantly decreased in the MM of mutant kidneys, suggesting that a reduction in Sall1 may be the cause of cell death in the MM during early kidney development and that, in turn, retards UB branching in the mutants. Therefore, the aberrant induction of Ifng expression, as part of an innate immune response, may contribute to renal agenesis or hypoplasia during early metanephric development by regulating the MM progenitor population.


Assuntos
Interferon gama/metabolismo , Rim/anormalidades , Rim/embriologia , Células-Tronco Mesenquimais/metabolismo , Fatores de Transcrição/metabolismo , Animais , Morte Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Interferon gama/genética , Rim/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Organogênese/fisiologia , Técnicas de Cultura de Tecidos
5.
PLoS Genet ; 13(2): e1006610, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28166224

RESUMO

To date, mutations within the coding region and translocations around the SOX9 gene both constitute the majority of genetic lesions underpinning human campomelic dysplasia (CD). While pathological coding-region mutations typically result in a non-functional SOX9 protein, little is known about what mechanism(s) controls normal SOX9 expression, and subsequently, which signaling pathways may be interrupted by alterations occurring around the SOX9 gene. Here, we report the identification of Stat3 as a key modulator of Sox9 expression in nascent cartilage and developing chondrocytes. Stat3 expression is predominant in tissues of mesodermal origin, and its conditional ablation using mesoderm-specific TCre, in vivo, causes dwarfism and skeletal defects characteristic of CD. Specifically, Stat3 loss results in the expansion of growth plate hypertrophic chondrocytes and deregulation of normal endochondral ossification in all bones examined. Conditional deletion of Stat3 with a Sox9Cre driver produces palate and tracheal irregularities similar to those described in Sox9+/- mice. Furthermore, mesodermal deletion of Stat3 causes global embryonic down regulation of Sox9 expression and function in vivo. Mechanistic experiments ex vivo suggest Stat3 can directly activate the expression of Sox9 by binding to its proximal promoter following activation. These findings illuminate a novel role for Stat3 in chondrocytes during skeletal development through modulation of a critical factor, Sox9. Importantly, they further provide the first evidence for the modulation of a gene product other than Sox9 itself which is capable of modeling pathological aspects of CD and underscore a potentially valuable therapeutic target for patients with the disorder.


Assuntos
Displasia Campomélica/genética , Fatores de Transcrição SOX9/genética , Fator de Transcrição STAT3/genética , Animais , Displasia Campomélica/patologia , Diferenciação Celular/genética , Condrócitos/metabolismo , Condrócitos/patologia , Condrogênese/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Mesoderma/crescimento & desenvolvimento , Mesoderma/patologia , Camundongos , Camundongos Transgênicos , Osteogênese/genética , Fenótipo , Fatores de Transcrição SOX9/biossíntese , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais
6.
Differentiation ; 94: 1-7, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27923152

RESUMO

The Wnt5a null mouse is a complex developmental model which, among its several posterior-localized axis defects, exhibits multiple kidney phenotypes, including duplex kidney and loss of the medullary zone. We previously reported that ablation of Wnt5a in nascent mesoderm causes duplex kidney formation as a result of aberrant development of the nephric duct and abnormal extension of intermediate mesoderm. However, these mice also display a loss of the medullary region late in gestation. We have now genetically isolated duplex kidney formation from the medullary defect by specifically targeting the progenitors for both the ureteric bud and metanephric mesenchyme. The conditional mutants fail to form a normal renal medulla but no longer exhibit duplex kidney formation. Approximately 1/3 of the mutants develop hydronephrosis in the kidneys either uni- or bilaterally when using Dll1Cre. The abnormal kidney phenotype becomes prominent at E16.5, which approximates the time when urine production begins in the mouse embryonic kidney, and is associated with a dramatic increase in apoptosis only in mutant kidneys with hydronephrosis. Methylene blue dye injection and histologic examination reveal that aberrant cell death likely results from urine toxicity due to an abnormal ureter-bladder connection. This study shows that Wnt5a is not required for development of the renal medulla and that loss of the renal medullary region in the Wnt5a-deleted kidney is caused by an abnormal ureter-bladder connection.


Assuntos
Diferenciação Celular/genética , Hidronefrose/genética , Rim/crescimento & desenvolvimento , Proteína Wnt-5a/genética , Animais , Hidronefrose/fisiopatologia , Rim/fisiopatologia , Camundongos , Camundongos Knockout , Morfogênese/genética , Transdução de Sinais/genética , Ureter/anormalidades , Ureter/crescimento & desenvolvimento , Bexiga Urinária/anormalidades , Bexiga Urinária/crescimento & desenvolvimento
7.
Cell Rep ; 15(4): 801-813, 2016 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-27149838

RESUMO

Nephron progenitors in the embryonic kidney propagate while generating differentiated nephrons. However, in mice, the progenitors terminally differentiate shortly after birth. Here, we report a method for selectively expanding nephron progenitors in vitro in an undifferentiated state. Combinatorial and concentration-dependent stimulation with LIF, FGF2/9, BMP7, and a WNT agonist is critical for expansion. The purified progenitors proliferated beyond the physiological limits observed in vivo, both for cell numbers and lifespan. Neonatal progenitors were maintained for a week, while progenitors from embryonic day 11.5 expanded 1,800-fold for nearly 20 days and still reconstituted 3D nephrons containing glomeruli and renal tubules. Furthermore, progenitors generated from mouse embryonic stem cells and human induced pluripotent cells could be expanded with retained nephron-forming potential. Thus, we have established in vitro conditions for promoting the propagation of nephron progenitors, which will be essential for dissecting the mechanisms of kidney organogenesis and for regenerative medicine.

8.
Differentiation ; 91(4-5): 152-8, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26856661

RESUMO

Significant recent advances in methodologies for the differentiation of pluripotent stem cells to renal progenitors as well as the definition of niche conditions for sustaining those progenitors have dramatically enhanced our understanding of their biology and developmental programing, prerequisites for establishing viable approaches to renal regeneration. In this article, we review the evolution of culture techniques and models for the study of metanephric development, describe the signaling mechanisms likely to be driving progenitor self-renewal, and discuss current efforts to generate de novo functional tissues, providing in depth protocols and niche conditions for the stabilization of the nephronic Six2+progenitor.


Assuntos
Diferenciação Celular/genética , Rim/citologia , Néfrons/citologia , Células-Tronco Pluripotentes/citologia , Humanos , Rim/crescimento & desenvolvimento , Néfrons/crescimento & desenvolvimento , Organogênese/genética , Regeneração , Transdução de Sinais
9.
Stem Cell Reports ; 5(3): 435-47, 2015 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-26321142

RESUMO

Understanding the mechanisms responsible for nephrogenic stem cell preservation and commitment is fundamental to harnessing the potential of the metanephric mesenchyme (MM) for nephron regeneration. Accordingly, we established a culture model that preferentially expands the MM SIX2+ progenitor pool using leukemia inhibitory factor (LIF), a Rho kinase inhibitor (ROCKi), and extracellular matrix. Passaged MM cells express the key stem cell regulators Six2 and Pax2 and remain competent to respond to WNT4 induction and form mature tubular epithelia and glomeruli. Mechanistically, LIF activates STAT, which binds to a Stat consensus sequence in the Six2 proximal promoter and sustains SIX2 levels. ROCKi, on the other hand, attenuates the LIF-induced differentiation activity of JNK. Concomitantly, the combination of LIF/ROCKi upregulates Slug expression and activates YAP, which maintains SIX2, PAX2, and SALL1. Using this novel model, our study underscores the pivotal roles of SIX2 and YAP in MM stem cell stability.


Assuntos
Proteínas de Homeodomínio/biossíntese , Fator Inibidor de Leucemia/farmacologia , Células-Tronco Mesenquimais/metabolismo , Néfrons/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Fatores de Transcrição/biossíntese , Quinases Associadas a rho/antagonistas & inibidores , Animais , Células-Tronco Mesenquimais/citologia , Camundongos , Néfrons/citologia , Fator de Transcrição PAX2/biossíntese , Fatores de Transcrição da Família Snail
10.
Lab Chip ; 15(5): 1320-8, 2015 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-25589423

RESUMO

This paper presents a continuous-flow microfluidic device for sorting stem cells and their differentiation progenies. The principle of the device is based on the accumulation of multiple dielectrophoresis (DEP) forces to deflect cells laterally in conjunction with the alternating on/off electric field to manipulate the cell trajectories. The microfluidic device containing a large array of oblique interdigitated electrodes was fabricated using a combination of standard and soft lithography techniques to generate a PDMS-gold electrode construct. Experimental testing with human mesenchymal stem cells (hMSC) and their differentiation progenies (osteoblasts) was carried out at different flow rates, and clear separation of the two populations was achieved. Most of the osteoblasts experiencing stronger DEP forces were deflected laterally and continuously, following zig-zag trajectories, and moved towards the desired collection outlet, whereas most of the hMSCs remained on the original trajectory due to weaker DEP forces. The experimental measurements were characterized and evaluated quantitatively, and consistent performance was demonstrated. Collection efficiency up to 92% and 67% for hMSCs and osteoblasts, respectively, along with purity up to 84% and 87% was obtained. The experimental results established the feasibility of our microfluidic DEP sorting device for continuous, label-free sorting of stem cells and their differentiation progenies.


Assuntos
Separação Celular/métodos , Eletroforese , Células-Tronco Mesenquimais/citologia , Técnicas Analíticas Microfluídicas/métodos , Diferenciação Celular , Separação Celular/instrumentação , Desenho de Equipamento , Humanos , Técnicas Analíticas Microfluídicas/instrumentação , Osteoblastos/citologia
11.
Hum Mol Genet ; 23(25): 6807-14, 2014 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-25082826

RESUMO

Congenital anomalies of the kidney and urinary tract (CAKUT) affect about 1 in 500 births and are a major cause of morbidity in infants. Duplex collecting systems rank among the most common abnormalities of CAKUT, but the molecular basis for this defect is poorly understood. In mice, conditional deletion of Wnt5a in mesoderm results in bilateral duplex kidney and ureter formation. The ureteric buds (UBs) in mutants emerge as doublets from the intermediate mesoderm (IM)-derived nephric duct (ND) without anterior expansion of the glial cell line-derived neurotrophic factor (Gdnf) expression domain in the surrounding mesenchyme. Wnt5a is normally expressed in a graded manner at the posterior end of the IM, but its expression is down-regulated prior to UB outgrowth at E10.5. Furthermore, ablation of Wnt5a in the mesoderm with an inducible Cre at E7.5 results in duplex UBs, whereas ablation at E8.5 yields normal UB outgrowth, demonstrating that Wnt5a functions in IM development well before the formation of the metanephros. In mutants, the posterior ND is duplicated and surrounding Pax2-positive mesenchymal cells persist in the nephric cord, suggesting that disruption of normal ND patterning prompts the formation of duplex ureters and kidneys. Ror2 homozygous mutants, which infrequently yield duplex collecting systems, show a dramatic increase in incidence with the additional deletion of one copy of Wnt5a, implicating this receptor in non-canonical Wnt5a signaling during IM development. This work provides the first evidence of a role of Wnt5a/Ror2 signaling in IM extension and offers new insights into the etiology of CAKUT and possible involvement of Wnt5a/Ror2 mutations.


Assuntos
Rim/metabolismo , Mesoderma/metabolismo , Morfogênese/genética , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Transdução de Sinais/genética , Proteínas Wnt/genética , Animais , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Homozigoto , Integrases/genética , Integrases/metabolismo , Rim/crescimento & desenvolvimento , Rim/patologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Mesoderma/crescimento & desenvolvimento , Mesoderma/patologia , Camundongos , Camundongos Transgênicos , Fator de Transcrição PAX2/genética , Fator de Transcrição PAX2/metabolismo , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Fatores de Tempo , Ureter/crescimento & desenvolvimento , Ureter/metabolismo , Ureter/patologia , Proteínas Wnt/deficiência , Proteína Wnt-5a , Ductos Mesonéfricos/crescimento & desenvolvimento , Ductos Mesonéfricos/metabolismo , Ductos Mesonéfricos/patologia
12.
J Am Soc Nephrol ; 25(11): 2584-95, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24744442

RESUMO

The balanced self-renewal and differentiation of nephron progenitors are critical for kidney development and controlled, in part, by the transcription factor Six2, which antagonizes canonical Wnt signaling-mediated differentiation. A nuclear factor, Sall1, is expressed in Six2-positive progenitors as well as differentiating nascent nephrons, and it is essential for kidney formation. However, the molecular functions and targets of Sall1, especially the functions and targets in the nephron progenitors, remain unknown. Here, we report that Sall1 deletion in Six2-positive nephron progenitors results in severe progenitor depletion and apoptosis of the differentiating nephrons in mice. Analysis of mice with an inducible Sall1 deletion revealed that Sall1 activates genes expressed in progenitors while repressing genes expressed in differentiating nephrons. Sall1 and Six2 co-occupied many progenitor-related gene loci, and Sall1 bound to Six2 biochemically. In contrast, Sall1 did not bind to the Wnt4 locus suppressed by Six2. Sall1-mediated repression was also independent of its binding to DNA. Thus, Sall1 maintains nephron progenitors and their derivatives by a unique mechanism, which partly overlaps but is distinct from that of Six2: Sall1 activates progenitor-related genes in Six2-positive nephron progenitors and represses gene expression in Six2-negative differentiating nascent nephrons.


Assuntos
Rim/embriologia , Néfrons/embriologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Apoptose/fisiologia , Diferenciação Celular/fisiologia , Feminino , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Rim/citologia , Rim/fisiologia , Camundongos Transgênicos , Néfrons/citologia , Néfrons/fisiologia , Técnicas de Cultura de Órgãos , Gravidez , Transdução de Sinais/fisiologia , Células-Tronco/citologia , Células-Tronco/fisiologia
13.
Oncotarget ; 5(2): 386-402, 2014 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-24481423

RESUMO

Wilms tumor (WT) is the most common childhood kidney cancer and retains gene expression profiles reminiscent of the embryonic kidney. We have shown previously that CITED1, a transcriptional regulator that labels the self-renewing, multipotent nephron progenitor population of the developing kidney, is robustly expressed across all major WT disease and patient characteristics. In this malignant context, CITED1 becomes enriched in the nucleus, which deviates from its cytosolic predominance in embryonic nephron progenitors. We designed the current studies to test the functional and mechanistic effects of differential CITED1 subcellular localization on WT behavior. To mimic its subcellular distribution observed in clinical WT specimens, CITED1 was misexpressed ectopically in the human WT cell line, WiT49, as either a wild-type (predominantly cytosolic) or a mutant, but transcriptionally active, protein (two point mutations in its nuclear export signal, CITED1ΔNES; nuclear-enriched). In vitro analyses showed that CITED1ΔNES enhanced WiT49 proliferation and colony formation in soft agar relative to wild-type CITED1 and empty vector controls. The nuclear-enriched CITED1ΔNES cell line showed the greatest tumor volumes after xenotransplantation into immunodeficient mice (n=15 animals per cell line). To elucidate CITED1 gene targets in this model, microarray profiling showed that wild-type CITED1 foremost upregulated LGR5 (stem cell marker), repressed CDH6 (early marker of epithelial commitment of nephron progenitors), and altered expression of specific WNT pathway participants. In summary, forced nuclear enrichment of CITED1 in a human WT cell line appears to enhance tumorigenicity, whereas ectopic cytosolic expression confers stem-like properties and an embryonic phenotype, analogous to the developmental context.


Assuntos
Núcleo Celular/metabolismo , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Células-Tronco Neoplásicas/patologia , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Tumor de Wilms/metabolismo , Tumor de Wilms/patologia , Animais , Proteínas Reguladoras de Apoptose , Carcinogênese , Modelos Animais de Doenças , Feminino , Células HEK293 , Humanos , Neoplasias Renais/genética , Camundongos , Camundongos SCID , Células-Tronco Neoplásicas/metabolismo , Proteínas Nucleares/genética , Transativadores , Fatores de Transcrição/genética , Ativação Transcricional , Transfecção , Tumor de Wilms/genética , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Methods Mol Biol ; 1092: 255-67, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24318826

RESUMO

While gene targeting methods have largely supplanted cell/explant culture models for studying developmental processes, they have not eliminated the need for or value of such approaches in the investigator's technical arsenal. Explant culture models, such as those devised for the metanephric kidney and its progenitors, remain invaluable as tools for screening regulatory factors involved in tissue induction or in the inhibition of progenitor specification. Thus, some factors capable of inducing tissue condensations or nephronic tubule formation in explants of metanephric mesenchyme have been identified through direct treatment of cultures rather than lengthy genetic engineering in animals. Unfortunately, renal progenitors are largely refractory to most contemporary methods for gene manipulation, including transfection and viral transduction, so the applications of explant culture have been rather limited. However, methods for protein or peptide transduction offer greatly improved efficiencies for uptake and expression/regulation of proteins within cells and tissues. Biologically active TAT- or penetratin-fusion proteins/peptides are readily taken up by most cells in metanephric explants or monolayer cultured cells (Plisov et al., J Am Soc Nephrol 16:1632-1644, 2005; Osafune et al., Development 133:151-161, 2006; Wang et al., Cell Signal 22:1717-1726, 2010; Tanigawa, Dev Biol 352:58-69, 2011), allowing a direct functional evaluation of theoretically any protein, including biologically active enzymes and transcription factors, or any targeted interactive domain within a protein.


Assuntos
Peptídeos Penetradores de Células/genética , Rim/citologia , Mesoderma/citologia , Técnicas de Cultura de Órgãos , Animais , Peptídeos Penetradores de Células/química , Biologia Molecular/métodos , Proteínas/química , Proteínas/genética , Transdução de Sinais
15.
Pediatr Blood Cancer ; 61(2): 198-205, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24115727

RESUMO

BACKGROUND: The Yes-associated-protein-1 (YAP1) is a novel, direct regulator of stem cell genes both in development and cancer. FAT4 is an upstream regulator that induces YAP1 cytosolic sequestering by phosphorylation (p-Ser 127) and therefore inhibits YAP1-dependent cellular proliferation. We hypothesized that loss of FAT4 signaling would result in expansion of the nephron progenitor population in kidney development and that YAP1 subcellular localization would be dysregulated in Wilms tumor (WT), an embryonal malignancy that retains gene expression profiles and histologic features reminiscent of the embryonic kidney. METHODS: Fetal kidneys from Fat4(-/-) mice were harvested at e18.5 and markers of nephron progenitors were investigated using immunohistochemical analysis. To examine YAP1 subcellular localization in WT, a primary WT cell line (VUWT30) was analyzed by immunofluorescence. Forty WT specimens evenly distributed between favorable and unfavorable histology (n = 20 each), and treatment failure or success (n = 20 each) was analyzed for total and phosphorylated YAP1 using immunohistochemistry and Western blot. RESULTS: Fat4(-/-) mouse fetal kidneys exhibit nuclear YAP1 with increased proliferation and expansion of nephron progenitor cells. In contrast to kidney development, subcellular localization of YAP1 is dysregulated in WT, with a preponderance of nuclear p-YAP1. By Western blot, median p-YAP1 quantity was 5.2-fold greater in unfavorable histology WT (P = 0.05). CONCLUSIONS: Fetal kidneys in Fat4(-/-) mice exhibit a phenotype reminiscent of nephrogenic rests, a WT precursor lesion. In WT, YAP1 subcellular localization is dysregulated and p-YAP1 accumulation is a novel biomarker of unfavorable histology.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Embrião de Mamíferos/patologia , Regulação da Expressão Gênica no Desenvolvimento , Rim/embriologia , Rim/patologia , Fosfoproteínas/metabolismo , Fosfoproteínas/fisiologia , Tumor de Wilms/patologia , Animais , Western Blotting , Proteínas de Ciclo Celular , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Proliferação de Células , Células Cultivadas , Pré-Escolar , Embrião de Mamíferos/metabolismo , Feminino , Células HeLa , Humanos , Técnicas Imunoenzimáticas , Rim/metabolismo , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Masculino , Camundongos , Camundongos Knockout , Néfrons/metabolismo , Néfrons/patologia , Fosforilação , Transporte Proteico , Células-Tronco/metabolismo , Células-Tronco/patologia , Frações Subcelulares , Fatores de Transcrição , Tumor de Wilms/metabolismo , Proteínas de Sinalização YAP
16.
Nat Cell Biol ; 15(9): 1035-44, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23974041

RESUMO

Present models suggest that the fate of the kidney epithelial progenitors is solely regulated by signals from the adjacent ureteric bud. The bud provides signals that regulate the survival, renewal and differentiation of these cells. Recent data suggest that Wnt9b, a ureteric-bud-derived factor, is sufficient for both progenitor cell renewal and differentiation. How the same molecule induces two seemingly contradictory processes is unknown. Here, we show that signals from the stromal fibroblasts cooperate with Wnt9b to promote differentiation of the progenitors. The atypical cadherin Fat4 encodes at least part of this stromal signal. Our data support a model whereby proper kidney size and function is regulated by balancing opposing signals from the ureteric bud and stroma to promote renewal and differentiation of the nephron progenitors.


Assuntos
Caderinas/metabolismo , Células Epiteliais/metabolismo , Fibroblastos/metabolismo , Rim/metabolismo , Células-Tronco/metabolismo , Células Estromais/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Wnt/metabolismo , Caderinas/genética , Comunicação Celular , Diferenciação Celular , Células Epiteliais/citologia , Fibroblastos/citologia , Regulação da Expressão Gênica , Humanos , Rim/citologia , Rim/crescimento & desenvolvimento , Tamanho do Órgão , Transdução de Sinais , Células-Tronco/citologia , Células Estromais/citologia , Proteínas Supressoras de Tumor/genética , Proteínas Wnt/genética
17.
Chem Res Toxicol ; 26(1): 96-105, 2013 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-23137061

RESUMO

Inorganic arsenic in the drinking water is a multisite human carcinogen that potentially targets the kidney. Recent evidence also indicates that developmental arsenic exposure impacts renal carcinogenesis in humans and mice. Emerging theory indicates that cancer may be a disease of stem cells (SCs) and that there are abundant active SCs during early life. Therefore, we hypothesized that inorganic arsenic targets SCs, or partially differentiated progenitor cells (PCs), for oncogenic transformation. Thus, a rat kidney SC/PC cell line, RIMM-18, was chronically exposed to low-level arsenite (500 nM) for up to 28 weeks. Multiple markers of acquired cancer phenotype were assessed biweekly during arsenic exposure, including secreted matrix metalloproteinase (MMP) activity, proliferation rate, colony formation in soft agar, and cellular invasiveness. Arsenic exposure by 10 weeks and after also induced marked and sustained increases in colony formation, indicative of the loss of contact inhibition, and increased invasiveness, both cancer cell characteristics. Compared to the passage-matched control, chronic arsenic exposure caused exposure-duration dependent increases in secreted MMP-2 and MMP-9 activity, Cox-2 expression, and more rapid proliferation (all >2-fold), characteristics typical of cancer cells. Dysregulation of SC maintenance genes and signaling pathways are common during oncogenesis. During arsenite exposure, expression of several genes associated with normal kidney development and SC regulation and differentiation (i.e., Wt-1, Wnt-4, Bmp-7, etc.) were aberrantly altered. Arsenic-exposed renal SCs produced more nonadherent spheroid bodies that grew much more aggressively in Matrigel, typical of cancer SCs (CSCs). The transformed cells also showed gene overexpression typical of renal SCs/CSCs (CD24, Osr1, Ncam) and arsenic adaptation such as overexpression of Mt-1, Mt2, Sod-1, and Abcc2. These data suggest that inorganic arsenic induced an acquired cancer phenotype in vitro in these rat kidney SCs potentially forming CSCs and, consistent with data in vivo, indicate that these multipotent SCs may be targets of arsenic during renal carcinogenesis.


Assuntos
Arsênio/toxicidade , Células-Tronco/efeitos dos fármacos , Antígeno AC133 , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Antígeno CD24/genética , Antígeno CD24/metabolismo , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Ciclo-Oxigenase 2/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Rim/citologia , Rim/metabolismo , Neoplasias Renais/etiologia , Neoplasias Renais/genética , Neoplasias Renais/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Metalotioneína/genética , Metalotioneína/metabolismo , Camundongos , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/metabolismo , Peptídeos/genética , Peptídeos/metabolismo , Ratos , Células-Tronco/citologia , Células-Tronco/metabolismo , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1
18.
J Pediatr Surg ; 47(6): 1239-49, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22703800

RESUMO

PURPOSE: SIX2 and CITED1 are transcriptional regulators that specify self-renewing nephronic progenitor cells of the embryonic kidney. We hypothesized that SIX2, which promotes and maintains this stem cell population, and CITED1 remain active in Wilms' tumor (WT). METHODS: To evaluate expression domains and the pathogenic significance of SIX2 and CITED1 across WT, the Children's Oncology Group provided 40 WT specimens of stages I to IV (n = 10 per stage), which were enriched for unfavorable histology (n = 20) and treatment failure (relapse or death, n = 20). SIX2 and CITED1 protein expression was evaluated qualitatively (immunohistochemistry) and quantitatively (Western blot, or WB). Gene transcription was estimated using quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS: SIX2 was visualized by immunohistochemistry in 36 (94.7%) of 38 specimens. Protein and messenger RNA expression of SIX2 were quantitatively similar across all stages of disease (P = .48 WB; P = 0.38 qPCR), in favorable or unfavorable histology (P = 0.51 WB; P = 0.58 qPCR), and in treatment failure or success (P = 0.86 WB; P = 0.49 qPCR). Although CITED1 expression paralleled SIX2 qualitatively, no quantitative correlation between SIX2 and CITED1 expression was observed (Spearman correlation coefficient, 0.28; P = 0.08). As in the fetal kidney, overlapping, but also distinct, WT cellular expression domains were observed between SIX2 and CITED1. CONCLUSION: SIX2 and CITED1 remain active across all disease characteristics of WT. Activity of these genes in WT potentially identifies a population of self-renewing cancer cells that exhibit an embryonic, stemlike phenotype. Taken together, these transcriptional regulators may be fundamental to WT cellular self-renewal and may represent targets for novel therapies that promote terminal differentiation.


Assuntos
Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/biossíntese , Neoplasias Renais/metabolismo , Proteínas de Neoplasias/biossíntese , Células-Tronco Neoplásicas/metabolismo , Néfrons/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Proteínas Nucleares/biossíntese , Fatores de Transcrição/biossíntese , Tumor de Wilms/metabolismo , Proteínas Reguladoras de Apoptose , Biomarcadores Tumorais , Western Blotting , Núcleo Celular/metabolismo , Citosol/metabolismo , Proteínas de Homeodomínio/genética , Humanos , Técnicas Imunoenzimáticas , Rim/embriologia , Rim/metabolismo , Neoplasias Renais/patologia , Microscopia de Fluorescência , Proteínas de Neoplasias/genética , Células-Tronco Neoplásicas/patologia , Néfrons/patologia , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Prognóstico , RNA Mensageiro/biossíntese , RNA Neoplásico/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Método Simples-Cego , Transativadores , Fatores de Transcrição/genética , Tumor de Wilms/patologia
19.
Neoplasia ; 14(12): 1153-63, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23308048

RESUMO

Hepatoblastoma, the most common pediatric liver cancer, consists of epithelial mixed embryonal/fetal (EMEF) and pure fetal histologic subtypes, with the latter exhibiting a more favorable prognosis. Few embryonal histology markers that yield insight into the biologic basis for this prognostic discrepancy exist. CBP/P-300 interacting transactivator 1 (CITED1), a transcriptional co-activator, is expressed in the self-renewing nephron progenitor population of the developing kidney and broadly in its malignant analog, Wilms tumor (WT). In this current study, CITED1 expression is detected in mouse embryonic liver initially on post-coitum day 10.5 (e10.5), begins to taper by e14.5, and is undetectable in e18.5 and adult livers. CITED1 expression is detected in regenerating murine hepatocytes following liver injury by partial hepatectomy and 3,5-diethoxycarbonyl-1,4-dihydrocollidine. Importantly, while CITED1 is undetectable in normal human adult livers, 36 of 41 (87.8%) hepatoblastoma specimens express CITED1, where it is enriched in EMEF specimens compared to specimens of pure fetal histology. CITED1 overexpression in Hep293TT human hepatoblastoma cells induces cellular proliferation and upregulates the Wnt inhibitors Kringle containing transmembrane protein 1 (KREMEN1) and CXXC finger protein 4 (CXXC4). CITED1 mRNA expression correlates with expression of CXXC4 and KREMEN1 in clinical hepatoblastoma specimens. These data show that CITED1 is expressed during a defined time course of liver development and is no longer expressed in the adult liver but is upregulated in regenerating hepatocytes following liver injury. Moreover, as in WT, this embryonic marker is reexpressed in hepatoblastoma and correlates with embryonal histology. These findings identify CITED1 as a novel marker of hepatic progenitor cells that is re-expressed following liver injury and in embryonic liver tumors.


Assuntos
Hepatoblastoma/metabolismo , Neoplasias Hepáticas/metabolismo , Regeneração Hepática , Fígado/metabolismo , Proteínas Nucleares/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Via de Sinalização Wnt , Animais , Proteínas Reguladoras de Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos/metabolismo , Hepatoblastoma/genética , Humanos , Fígado/lesões , Neoplasias Hepáticas/genética , Proteínas de Membrana/metabolismo , Camundongos , Mutação , RNA Mensageiro/metabolismo , Fatores de Tempo , beta Catenina/genética
20.
Development ; 138(24): 5369-78, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22110055

RESUMO

During development of the urogenital tract, fibroblast growth factor 8 (Fgf8) is expressed in mesonephric tubules, but its role in this tissue remains undefined. An evaluation of previously generated T-Cre-mediated Fgf8-deficient mice (T-Cre; Fgf8(flox/Δ2,3) mice), which lack Fgf8 expression in the mesoderm, revealed that the cranial region of the Wolffian duct degenerated prematurely and the cranial mesonephric tubules were missing. As a result, the epididymis, vas deferens and efferent ductules were largely absent in mutant mice. Rarb2-Cre was used to eliminate FGF8 from the mesonephric tubules but to allow expression in the adjacent somites. These mutants retained the cranial end of the Wolffian duct and formed the epididymis and vas deferens, but failed to elaborate the efferent ductules, indicating that Fgf8 expression by the mesonephric tubules is required specifically for the formation of the ductules. Ret knockout mice do not form the ureteric bud, a caudal outgrowth of the Wolffian duct and progenitor for the collecting duct network in the kidney, but they do develop the cranial end normally. This indicates that Fgf8, but not Ret, expression is essential to the outgrowth of the cranial mesonephric tubules from the Wolffian duct and to the development of major portions of the sex accessory tissues in the male reproductive tract. Mechanistically, FGF8 functions upstream of Lhx1 expression in forming the nephron, and analysis of Fgf8 mutants similarly shows deficient Lhx1 expression in the mesonephric tubules. These results demonstrate a multifocal requirement for FGF8 in establishing the male reproductive tract ducts and implicate Lhx1 signaling in tubule elongation.


Assuntos
Fator 8 de Crescimento de Fibroblasto/metabolismo , Genitália Masculina/crescimento & desenvolvimento , Ductos Mesonéfricos/crescimento & desenvolvimento , Animais , Regulação da Expressão Gênica no Desenvolvimento , Genitália Masculina/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Masculino , Mesoderma/crescimento & desenvolvimento , Mesoderma/metabolismo , Camundongos , Camundongos Knockout , Néfrons/crescimento & desenvolvimento , Néfrons/metabolismo , Proteínas Proto-Oncogênicas c-ret/metabolismo , Fatores de Transcrição/metabolismo , Sistema Urogenital/crescimento & desenvolvimento , Sistema Urogenital/metabolismo , Ductos Mesonéfricos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA