Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Oncogene ; 40(38): 5718-5729, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34331013

RESUMEN

Melanomas driven by loss of the NF1 tumor suppressor have a high risk of treatment failure and effective therapies have not been developed. Here we show that loss-of-function mutations of nf1 and pten result in aggressive melanomas in zebrafish, representing the first animal model of NF1-mutant melanomas harboring PTEN loss. MEK or PI3K inhibitors show little activity when given alone due to cross-talk between the pathways, and high toxicity when given together. The mTOR inhibitors, sirolimus, everolimus, and temsirolimus, were the most active single agents tested, potently induced tumor-suppressive autophagy, but not apoptosis. Because addition of the BCL2 inhibitor venetoclax resulted in compensatory upregulation of MCL1, we established a three-drug combination composed of sirolimus, venetoclax, and the MCL1 inhibitor S63845. This well-tolerated drug combination potently and synergistically induces apoptosis in both zebrafish and human NF1/PTEN-deficient melanoma cells, providing preclinical evidence justifying an early-stage clinical trial in patients with NF1/PTEN-deficient melanoma.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes/administración & dosificación , Inhibidores mTOR/administración & dosificación , Melanoma/tratamiento farmacológico , Neurofibromina 1/genética , Fosfohidrolasa PTEN/genética , Pirimidinas/administración & dosificación , Sulfonamidas/administración & dosificación , Tiofenos/administración & dosificación , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Everolimus/administración & dosificación , Everolimus/farmacología , Humanos , Mutación con Pérdida de Función , Inhibidores mTOR/farmacología , Melanoma/genética , Melanoma/patología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Pirimidinas/farmacología , Sirolimus/administración & dosificación , Sirolimus/análogos & derivados , Sirolimus/farmacología , Sulfonamidas/farmacología , Tiofenos/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra
2.
Elife ; 102021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33527896

RESUMEN

Recent genomic and scRNA-seq analyses of melanoma demonstrated a lack of recurrent genetic drivers of metastasis, while identifying common transcriptional states correlating with invasion or drug resistance. To test whether transcriptional adaptation can drive melanoma progression, we made use of a zebrafish mitfa:BRAFV600E;tp53-/- model, in which malignant progression is characterized by minimal genetic evolution. We undertook an overexpression-screen of 80 epigenetic/transcriptional regulators and found neural crest-mesenchyme developmental regulator SATB2 to accelerate aggressive melanoma development. Its overexpression induces invadopodia formation and invasion in zebrafish tumors and human melanoma cell lines. SATB2 binds and activates neural crest-regulators, including pdgfab and snai2. The transcriptional program induced by SATB2 overlaps with known MITFlowAXLhigh and AQP1+NGFR1high drug-resistant states and functionally drives enhanced tumor propagation and resistance to Vemurafenib in vivo. In summary, we show that melanoma transcriptional rewiring by SATB2 to a neural crest mesenchyme-like program can drive invasion and drug resistance in autochthonous tumors.


Asunto(s)
Resistencia a Antineoplásicos/genética , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Melanoma/genética , Invasividad Neoplásica/genética , Factores de Transcripción/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Sistemas CRISPR-Cas , Línea Celular Tumoral , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Cresta Neural/citología , Factores de Transcripción/genética , Pez Cebra , Proteínas de Pez Cebra/genética
3.
Pigment Cell Melanoma Res ; 34(3): 641-647, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33064882

RESUMEN

A progressive increase in copy number variation (CNV) characterizes the natural history of cutaneous melanoma progression toward later disease stages, but our understanding of genetic drivers underlying chromosomal arm-level CNVs remains limited. To identify candidate progression drivers, we mined the TCGA SKCM dataset and identified HDGF as a recurrently amplified gene whose high mRNA expression correlates with poor patient survival. Using melanocyte-specific overexpression in the zebrafish BRAFV600E -driven MiniCoopR melanoma model, we show that HDGF accelerates melanoma development in vivo. Transcriptional analysis of HDGF compared to control EGFP tumors showed the activation of endothelial/angiogenic pathways. We validated this observation using an endothelial kdrl:mCherry reporter line which showed HDGF to increases tumor vasculature. HDGF is frequently co-altered with the established melanoma driver SETDB1. Both genes are located on chromosome 1q, and their co-amplification is observed in up to 13% of metastatic melanoma. TCGA patients with both genes amplified and/or overexpressed have a worse melanoma specific survival. We tested co-expression of HDGF and SETDB1 in the MiniCoopR model, which resulted in faster and more aggressive melanoma development than either gene individually. Our work identifies the co-amplification of HDGF and SETDB1 as a functional driver of melanoma progression and poor patient prognosis.


Asunto(s)
Biomarcadores de Tumor/genética , Cromosomas Humanos Par 1/genética , N-Metiltransferasa de Histona-Lisina/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Melanoma/mortalidad , Mutación , Neoplasias Cutáneas/mortalidad , Animales , Estudios de Seguimiento , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/genética , Melanoma/patología , Pronóstico , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Tasa de Supervivencia , Pez Cebra
4.
Genome Res ; 30(12): 1815-1834, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32732264

RESUMEN

Deciphering the genomic regulatory code of enhancers is a key challenge in biology because this code underlies cellular identity. A better understanding of how enhancers work will improve the interpretation of noncoding genome variation and empower the generation of cell type-specific drivers for gene therapy. Here, we explore the combination of deep learning and cross-species chromatin accessibility profiling to build explainable enhancer models. We apply this strategy to decipher the enhancer code in melanoma, a relevant case study owing to the presence of distinct melanoma cell states. We trained and validated a deep learning model, called DeepMEL, using chromatin accessibility data of 26 melanoma samples across six different species. We show the accuracy of DeepMEL predictions on the CAGI5 challenge, where it significantly outperforms existing models on the melanoma enhancer of IRF4 Next, we exploit DeepMEL to analyze enhancer architectures and identify accurate transcription factor binding sites for the core regulatory complexes in the two different melanoma states, with distinct roles for each transcription factor, in terms of nucleosome displacement or enhancer activation. Finally, DeepMEL identifies orthologous enhancers across distantly related species, where sequence alignment fails, and the model highlights specific nucleotide substitutions that underlie enhancer turnover. DeepMEL can be used from the Kipoi database to predict and optimize candidate enhancers and to prioritize enhancer mutations. In addition, our computational strategy can be applied to other cancer or normal cell types.


Asunto(s)
Biología Computacional/métodos , Melanoma/genética , Pez Cebra/genética , Animales , Aprendizaje Profundo , Perros , Elementos de Facilitación Genéticos , Regulación Neoplásica de la Expresión Génica , Caballos , Humanos , Ratones , Porcinos
5.
Cancer Discov ; 10(7): 980-997, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32269030

RESUMEN

Epigenetic regulators, when genomically altered, may become driver oncogenes that mediate otherwise unexplained pro-oncogenic changes lacking a clear genetic stimulus, such as activation of the WNT/ß-catenin pathway in melanoma. This study identifies previously unrecognized recurrent activating mutations in the G9a histone methyltransferase gene, as well as G9a genomic copy gains in approximately 26% of human melanomas, which collectively drive tumor growth and an immunologically sterile microenvironment beyond melanoma. Furthermore, the WNT pathway is identified as a key tumorigenic target of G9a gain-of-function, via suppression of the WNT antagonist DKK1. Importantly, genetic or pharmacologic suppression of mutated or amplified G9a using multiple in vitro and in vivo models demonstrates that G9a is a druggable target for therapeutic intervention in melanoma and other cancers harboring G9a genomic aberrations. SIGNIFICANCE: Oncogenic G9a abnormalities drive tumorigenesis and the "cold" immune microenvironment by activating WNT signaling through DKK1 repression. These results reveal a key druggable mechanism for tumor development and identify strategies to restore "hot" tumor immune microenvironments.This article is highlighted in the In This Issue feature, p. 890.


Asunto(s)
Carcinogénesis/genética , Mutación con Ganancia de Función/genética , Antígenos de Histocompatibilidad/genética , N-Metiltransferasa de Histona-Lisina/genética , Oncogenes/genética , Línea Celular Tumoral , Humanos , Mutación
7.
Cancer Cell ; 33(2): 322-336.e8, 2018 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-29438700

RESUMEN

Oncogene-induced senescence, e.g., in melanocytic nevi, terminates the expansion of pre-malignant cells via transcriptional silencing of proliferation-related genes due to decoration of their promoters with repressive trimethylated histone H3 lysine 9 (H3K9) marks. We show here that structurally distinct H3K9-active demethylases-the lysine-specific demethylase-1 (LSD1) and several Jumonji C domain-containing moieties (such as JMJD2C)-disable senescence and permit Ras/Braf-evoked transformation. In mouse and zebrafish models, enforced LSD1 or JMJD2C expression promoted Braf-V600E-driven melanomagenesis. A large subset of established melanoma cell lines and primary human melanoma samples presented with a collective upregulation of related and unrelated H3K9 demethylase activities, whose targeted inhibition restored senescence, even in Braf inhibitor-resistant melanomas, evoked secondary immune effects and controlled tumor growth in vivo.


Asunto(s)
Histona Demetilasas/genética , Histona Demetilasas con Dominio de Jumonji/genética , Melanoma/genética , Animales , Histonas/metabolismo , Humanos , Lisina/genética , Lisina/metabolismo , Metilación , Ratones Desnudos , Regiones Promotoras Genéticas/genética
8.
Nephron ; 138(4): 310-323, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29342457

RESUMEN

BACKGROUND: von Hippel-Lindau (VHL) disease is characterized by the development of benign and malignant tumours in many organ systems, including renal cysts and clear cell renal cell carcinoma. It is not completely understood what underlies the development of renal pathology, and the use of murine Vhl models has been challenging due to limitations in disease conservation. We previously described a zebrafish model bearing inactivating mutations in the orthologue of the human VHL gene. METHODS: We used histopathological and functional assays to investigate the pronephric and glomerular developmental defects in vhl mutant zebrafish, supported by human cell culture assays. RESULTS: Here, we report that vhl is required to maintain pronephric tubule and glomerulus integrity in zebrafish embryos. vhl mutant glomeruli are enlarged, cxcr4a+ capillary loops are dilated and the Bowman space is widened. While we did not observe pronephric cysts, the cells of the proximal convoluted and anterior proximal straight tubule are enlarged, periodic acid schiff (PAS) and Oil Red O positive, and display a clear cytoplasm after hematoxylin and eosine staining. Ultrastructural analysis showed the vhl-/- tubule to accumulate large numbers of vesicles of variable size and electron density. Microinjection of the endocytic fluorescent marker AM1-43 in zebrafish embryos revealed an accumulation of endocytic vesicles in the vhl mutant pronephric tubule, which we can recapitulate in human cells lacking VHL. CONCLUSIONS: Our data indicates that vhl is required to maintain pronephric tubule and glomerulus integrity during zebrafish development, and suggests a role for VHL in endocytic vesicle trafficking.


Asunto(s)
Glomérulos Renales/metabolismo , Túbulos Renales Proximales/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas de Pez Cebra/genética , Pez Cebra/fisiología , Animales , Desarrollo Embrionario/genética , Glomérulos Renales/anomalías , Glomérulos Renales/crecimiento & desarrollo , Túbulos Renales Proximales/anomalías , Túbulos Renales Proximales/crecimiento & desarrollo , Larva , Mutación , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
9.
Zebrafish ; 14(4): 379-382, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28557653

RESUMEN

The establishment of in vitro cultures of zebrafish cancer cells has expanded the potential of zebrafish as a disease model. However, the lack of effective methods for gene delivery and genetic manipulation has limited the experimental applications of these cultures. To overcome this barrier, we tested and optimized vesicular stomatitis virus glycoprotein (VSV-G) pseudotyped lentiviral and retroviral vector transduction protocols. We show that lentivirus successfully and efficiently transduced zebrafish melanoma cell lines in vitro, allowing antibiotic selection, fluorescence-based sorting, and in vivo allotransplantation. In addition, injection of concentrated lentiviral particles into embryos and tumors established the feasibility of in vivo gene delivery.


Asunto(s)
Vectores Genéticos/administración & dosificación , Lentivirus/genética , Melanoma/genética , Retroviridae/genética , Transducción Genética , Pez Cebra/embriología , Pez Cebra/genética , Animales , Melanoma/patología , Glicoproteínas de Membrana/genética , Células Tumorales Cultivadas , Proteínas del Envoltorio Viral/genética , Pez Cebra/crecimiento & desarrollo
10.
Pigment Cell Melanoma Res ; 30(4): 402-412, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28379616

RESUMEN

Melanoma is the most aggressive and deadliest form of skin cancer. A detailed knowledge of the cellular, molecular, and genetic events underlying melanoma progression is highly relevant to diagnosis, prognosis and risk stratification, and the development of new therapies. In the last decade, zebrafish have emerged as a valuable model system for the study of melanoma. Pathway conservation, coupled with the availability of robust genetic, transgenic, and chemical tools, has made the zebrafish a powerful model for identifying novel disease genes, visualizing cancer initiation, interrogating tumor-microenvironment interactions, and discovering new therapeutics that regulate melanocyte and melanoma development. In this review, we will give an overview of these studies, and highlight recent advancements that will help unravel melanoma pathogenesis and impact human disease.


Asunto(s)
Melanoma/patología , Melanoma/terapia , Investigación Biomédica Traslacional , Pez Cebra/fisiología , Animales , Carcinogénesis/patología , Modelos Animales de Enfermedad , Humanos , Melanocitos/patología
11.
Value Health ; 20(4): 627-636, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28408005

RESUMEN

OBJECTIVES: The aim of this article was to provide practical guidance in setting up patient registries to facilitate real-world data collection for health care decision making. METHODS: This guidance was based on our experiences and involvement in setting up patient registries in oncology in the Netherlands. All aspects were structured according to 1) mission and goals ("the Why"), 2) stakeholders and funding ("the Who"), 3) type and content ("the What"), and 4) identification and recruitment of patients, data handling, and pharmacovigilance ("the How"). RESULTS: The mission of most patient registries is improving patient health by improving the quality of patient care; monitoring and evaluating patient care is often the primary goal ("the Why"). It is important to align the objectives of the registry and agree on a clear and functional governance structure with all stakeholders ("the Who"). There is often a trade off between reliability, validity, and specificity of data elements and feasibility of data collection ("the What"). Patient privacy should be carefully protected, and address (inter-)national and local regulations. Patient registries can reveal unique safety information, but it can be challenging to comply with pharmacovigilance guidelines ("the How"). CONCLUSIONS: It is crucial to set up an efficient patient registry that serves its aims by collecting the right data of the right patient in the right way. It can be expected that patient registries will become the new standard alongside randomized controlled trials due to their unique value.


Asunto(s)
Recolección de Datos/métodos , Toma de Decisiones , Investigación sobre Servicios de Salud/métodos , Oncología Médica/métodos , Formulación de Políticas , Sistema de Registros , Confidencialidad , Exactitud de los Datos , Recolección de Datos/economía , Recolección de Datos/normas , Adhesión a Directriz , Guías como Asunto , Investigación sobre Servicios de Salud/economía , Investigación sobre Servicios de Salud/normas , Humanos , Oncología Médica/economía , Oncología Médica/normas , Países Bajos , Objetivos Organizacionales , Farmacovigilancia , Sistema de Registros/normas , Reproducibilidad de los Resultados , Apoyo a la Investigación como Asunto
12.
J Exp Med ; 214(3): 623-637, 2017 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-28148688

RESUMEN

We studied three patients with severe skeletal dysplasia, T cell immunodeficiency, and developmental delay. Whole-exome sequencing revealed homozygous missense mutations affecting exostosin-like 3 (EXTL3), a glycosyltransferase involved in heparan sulfate (HS) biosynthesis. Patient-derived fibroblasts showed abnormal HS composition and altered fibroblast growth factor 2 signaling, which was rescued by overexpression of wild-type EXTL3 cDNA. Interleukin-2-mediated STAT5 phosphorylation in patients' lymphocytes was markedly reduced. Interbreeding of the extl3-mutant zebrafish (box) with Tg(rag2:green fluorescent protein) transgenic zebrafish revealed defective thymopoiesis, which was rescued by injection of wild-type human EXTL3 RNA. Targeted differentiation of patient-derived induced pluripotent stem cells showed a reduced expansion of lymphohematopoietic progenitor cells and defects of thymic epithelial progenitor cell differentiation. These data identify EXTL3 mutations as a novel cause of severe immune deficiency with skeletal dysplasia and developmental delay and underline a crucial role of HS in thymopoiesis and skeletal and brain development.


Asunto(s)
Enfermedades del Desarrollo Óseo/etiología , Discapacidades del Desarrollo/etiología , Síndromes de Inmunodeficiencia/etiología , Mutación , N-Acetilglucosaminiltransferasas/genética , Animales , Preescolar , Femenino , Heparitina Sulfato/fisiología , Humanos , Células Madre Pluripotentes Inducidas/citología , Lactante , Linfocitos/fisiología , Pez Cebra
13.
Dev Neurorehabil ; 20(3): 173-178, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27055081

RESUMEN

OBJECTIVE: Determine healthcare costs of upper-extremity surgical correction in children with spastic cerebral palsy (CP). METHOD: This cohort study included 39 children with spastic CP who had surgery for their upper extremity at a Dutch hospital. A retrospective cost analysis was performed including both hospital and rehabilitation costs. Hospital costs were determined using microcosting methodology. Rehabilitation costs were estimated using reference prices. RESULTS: Hospital costs averaged €6813 per child. Labor (50%), overheads (29%), and medical aids (15%) were important cost drivers. Rehabilitation costs were estimated at €3599 per child. CONCLUSIONS: Surgery of the upper extremity is an important contributor to the healthcare costs of children with CP. Our study shows that labor is the most important cost driver for hospital costs, owing to the multidisciplinary approach and patient-specific treatment plan. A remarkable finding was the substantial amount of rehabilitation costs.


Asunto(s)
Parálisis Cerebral/economía , Parálisis Cerebral/cirugía , Costos y Análisis de Costo/economía , Extremidad Superior/cirugía , Adolescente , Niño , Femenino , Humanos , Masculino , Estudios Retrospectivos
14.
Mol Cell ; 62(1): 34-46, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27058786

RESUMEN

Studying cancer metabolism gives insight into tumorigenic survival mechanisms and susceptibilities. In melanoma, we identify HEXIM1, a transcription elongation regulator, as a melanoma tumor suppressor that responds to nucleotide stress. HEXIM1 expression is low in melanoma. Its overexpression in a zebrafish melanoma model suppresses cancer formation, while its inactivation accelerates tumor onset in vivo. Knockdown of HEXIM1 rescues zebrafish neural crest defects and human melanoma proliferation defects that arise from nucleotide depletion. Under nucleotide stress, HEXIM1 is induced to form an inhibitory complex with P-TEFb, the kinase that initiates transcription elongation, to inhibit elongation at tumorigenic genes. The resulting alteration in gene expression also causes anti-tumorigenic RNAs to bind to and be stabilized by HEXIM1. HEXIM1 plays an important role in inhibiting cancer cell-specific gene transcription while also facilitating anti-cancer gene expression. Our study reveals an important role for HEXIM1 in coupling nucleotide metabolism with transcriptional regulation in melanoma.


Asunto(s)
Melanoma/metabolismo , Factor B de Elongación Transcripcional Positiva/genética , Pirimidinas/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/genética , Melanoma/patología , Melanoma Experimental , Proteínas Oncogénicas/genética , Factores de Transcripción , Transcripción Genética , Proteínas Supresoras de Tumor/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
15.
Science ; 351(6272): aad2197, 2016 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-26823433

RESUMEN

The "cancerized field" concept posits that cancer-prone cells in a given tissue share an oncogenic mutation, but only discreet clones within the field initiate tumors. Most benign nevi carry oncogenic BRAF(V600E) mutations but rarely become melanoma. The zebrafish crestin gene is expressed embryonically in neural crest progenitors (NCPs) and specifically reexpressed in melanoma. Live imaging of transgenic zebrafish crestin reporters shows that within a cancerized field (BRAF(V600E)-mutant; p53-deficient), a single melanocyte reactivates the NCP state, revealing a fate change at melanoma initiation in this model. NCP transcription factors, including sox10, regulate crestin expression. Forced sox10 overexpression in melanocytes accelerated melanoma formation, which is consistent with activation of NCP genes and super-enhancers leading to melanoma. Our work highlights NCP state reemergence as a key event in melanoma initiation.


Asunto(s)
Carcinogénesis/genética , Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Melanoma Experimental/genética , Melanoma/genética , Cresta Neural/metabolismo , Neoplasias Cutáneas/genética , Pez Cebra , Animales , Animales Modificados Genéticamente , Células Madre Embrionarias/metabolismo , Elementos de Facilitación Genéticos , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Melanocitos/metabolismo , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas Proto-Oncogénicas B-raf/genética , Factores de Transcripción SOXE/genética , Proteína p53 Supresora de Tumor/genética , Proteínas de Pez Cebra/genética
16.
Cancer Res ; 75(20): 4272-4282, 2015 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-26282170

RESUMEN

Metastasis is the defining feature of advanced malignancy, yet remains challenging to study in laboratory environments. Here, we describe a high-throughput zebrafish system for comprehensive, in vivo assessment of metastatic biology. First, we generated several stable cell lines from melanomas of transgenic mitfa-BRAF(V600E);p53(-/-) fish. We then transplanted the melanoma cells into the transparent casper strain to enable highly quantitative measurement of the metastatic process at single-cell resolution. Using computational image analysis of the resulting metastases, we generated a metastasis score, µ, that can be applied to quantitative comparison of metastatic capacity between experimental conditions. Furthermore, image analysis also provided estimates of the frequency of metastasis-initiating cells (∼1/120,000 cells). Finally, we determined that the degree of pigmentation is a key feature defining cells with metastatic capability. The small size and rapid generation of progeny combined with superior imaging tools make zebrafish ideal for unbiased high-throughput investigations of cell-intrinsic or microenvironmental modifiers of metastasis. The approaches described here are readily applicable to other tumor types and thus serve to complement studies also employing murine and human cell culture systems.


Asunto(s)
Modelos Biológicos , Neoplasias/patología , Pez Cebra , Algoritmos , Animales , Animales Modificados Genéticamente , Línea Celular Tumoral , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , Mutación , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/metabolismo , Transcriptoma
17.
J Clin Invest ; 125(5): 1987-97, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25866969

RESUMEN

Patients with a germline mutation in von Hippel-Lindau (VHL) develop renal cell cancers and hypervascular tumors of the brain, adrenal glands, and pancreas as well as erythrocytosis. These phenotypes are driven by aberrant expression of HIF2α, which induces expression of genes involved in cell proliferation, angiogenesis, and red blood cell production. Currently, there are no effective treatments available for VHL disease. Here, using an animal model of VHL, we report a marked improvement of VHL-associated phenotypes following treatment with HIF2α inhibitors. Inactivation of vhl in zebrafish led to constitutive activation of HIF2α orthologs and modeled several aspects of the human disease, including erythrocytosis, pathologic angiogenesis in the brain and retina, and aberrant kidney and liver proliferation. Treatment of vhl(-/-) mutant embryos with HIF2α-specific inhibitors downregulated Hif target gene expression in a dose-dependent manner, improved abnormal hematopoiesis, and substantially suppressed erythrocytosis and angiogenic sprouting. Moreover, pharmacologic inhibition of HIF2α reversed the compromised cardiac contractility of vhl(-/-) embryos and partially rescued early lethality. This study demonstrates that small-molecule targeting of HIF2α improves VHL-related phenotypes in a vertebrate animal model and supports further exploration of this strategy for treating VHL disease.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Hidrazonas/uso terapéutico , Sulfonas/uso terapéutico , Enfermedad de von Hippel-Lindau/tratamiento farmacológico , Regiones no Traducidas 5' , Aminoácidos Dicarboxílicos/toxicidad , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Encéfalo/irrigación sanguínea , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Embrión no Mamífero , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hidrazonas/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/deficiencia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Riñón/patología , Hígado/patología , Contracción Miocárdica/efectos de los fármacos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/genética , Fenotipo , Policitemia/tratamiento farmacológico , Policitemia/genética , Vasos Retinianos/patología , Sulfonas/farmacología , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética , Enfermedad de von Hippel-Lindau/genética , Enfermedad de von Hippel-Lindau/patología , Enfermedad de von Hippel-Lindau/fisiopatología
18.
J Pathol ; 231(1): 117-29, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23744542

RESUMEN

Biallelic mutations of the von Hippel-Lindau (VHL) gene are the most common cause of sporadic and inherited renal cell carcinoma (RCC). Loss of VHL has been reported to affect cell proliferation by deregulating cell cycle-associated proteins. We report that the VHL gene product (pVHL) inhibits E2F1 expression at both mRNA and protein level in zebrafish and human RCC cells, while loss of VHL increases E2F1 expression in patient kidney tumour tissue and RCC cells, resulting in a delay of cell cycle progression. RCCs from von Hippel-Lindau patients with known germline VHL mutations express significantly more E2F1 compared to sporadic RCCs with either clear-cell (cc) or non-cc histology. Analysis of 138 primary RCCs reveals that E2F1 expression is significantly higher in tumours with a diameter ≤7 cm and with a favourable American Joint Committee on Cancer (AJCC) stage. The expression of E2F1 in RCC significantly correlates with p27 expression, suggesting that increased expression of E2F1 in RCC induces tumour cell senescence via p27. Cox regression analysis shows significant prediction of E2F1 expression for disease-free survival and overall survival, implying that E2F1 expression in kidney tumour is a novel prognostic factor for patients with RCC.


Asunto(s)
Carcinoma de Células Renales/mortalidad , Factor de Transcripción E2F1/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Neoplasias Renales/mortalidad , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/fisiología , Animales , Western Blotting , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Senescencia Celular , Modelos Animales de Enfermedad , Factor de Transcripción E2F1/metabolismo , Femenino , Humanos , Neoplasias Renales/genética , Neoplasias Renales/patología , Masculino , Persona de Mediana Edad , Organismos Modificados Genéticamente , Plásmidos , Pronóstico , Antígeno Nuclear de Célula en Proliferación/metabolismo , Modelos de Riesgos Proporcionales , Reacción en Cadena en Tiempo Real de la Polimerasa , Tasa de Supervivencia , Transfección , Células Tumorales Cultivadas , Pez Cebra
19.
PLoS Genet ; 9(4): e1003384, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23599692

RESUMEN

Seminoma is a subclass of human testicular germ cell tumors (TGCT), the most frequently observed cancer in young men with a rising incidence. Here we describe the identification of a novel gene predisposing specifically to seminoma formation in a vertebrate model organism. Zebrafish carrying a heterozygous nonsense mutation in Leucine-Rich Repeat Containing protein 50 (lrrc50 also called dnaaf1), associated previously with ciliary function, are found to be highly susceptible to the formation of seminomas. Genotyping of these zebrafish tumors shows loss of heterozygosity (LOH) of the wild-type lrrc50 allele in 44.4% of tumor samples, correlating with tumor progression. In humans we identified heterozygous germline LRRC50 mutations in two different pedigrees with a family history of seminomas, resulting in a nonsense Arg488* change and a missense Thr590Met change, which show reduced expression of the wild-type allele in seminomas. Zebrafish in vivo complementation studies indicate the Thr590Met to be a loss-of-function mutation. Moreover, we show that a pathogenic Gln307Glu change is significantly enriched in individuals with seminoma tumors (13% of our cohort). Together, our study introduces an animal model for seminoma and suggests LRRC50 to be a novel tumor suppressor implicated in human seminoma pathogenesis.


Asunto(s)
Seminoma , Pez Cebra , Animales , Genes Supresores de Tumor , Genotipo , Humanos , Mutación , Pez Cebra/genética
20.
Cancer Res ; 72(16): 4017-27, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22665266

RESUMEN

Hypoxic signaling is a central modulator of cellular physiology in cancer. Core members of oxygen-sensing pathway including the von Hippel-Lindau tumor suppressor protein (pVHL) and the hypoxia inducible factor (HIF) transcription factors have been intensively studied, but improved organismal models might speed advances for both pathobiologic understanding and therapeutic modulation. To study HIF signaling during tumorigenesis and development in zebrafish, we developed a unique in vivo reporter for hypoxia, expressing EGFP driven by prolyl hydroxylase 3 (phd3) promoter/regulatory elements. Modulation of HIF pathway in Tg(phd3::EGFP) embryos showed a specific role for hypoxic signaling in the transgene activation. Zebrafish vhl mutants display a systemic hypoxia response, reflected by strong and ubiquitous transgene expression. In contrast to human VHL patients, heterozygous Vhl mice and vhl zebrafish are not predisposed to cancer. However, upon exposure to dimethylbenzanthracene (DMBA), the vhl heterozygous fish showed an increase in the occurrence of hepatic and intestinal tumors, a subset of which exhibited strong transgene expression, suggesting loss of Vhl function in these tumor cells. Compared with control fish, DMBA-treated vhl heterozygous fish also showed an increase in proliferating cell nuclear antigen-positive renal tubules. Taken together, our findings establish Vhl as a genuine tumor suppressor in zebrafish and offer this model as a tool to noninvasively study VHL and HIF signaling during tumorigenesis and development.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Neoplasias Intestinales/metabolismo , Neoplasias Hepáticas Experimentales/metabolismo , 9,10-Dimetil-1,2-benzantraceno , Animales , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Transformación Celular Neoplásica/inducido químicamente , Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Intestinales/inducido químicamente , Neoplasias Hepáticas Experimentales/inducido químicamente , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...