Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Angiogenesis ; 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38842752

RESUMEN

Conjunctival melanoma (CoM) is a rare but potentially lethal cancer of the eye, with limited therapeutic option for metastases. A better understanding how primary CoM disseminate to form metastases is urgently needed in order to develop novel therapies. Previous studies indicated that primary CoM tumors express Vascular Endothelial Growth Factor (VEGF) and may recruit pro-tumorigenic M2-like macrophages. However, due to a lack of proper models, the expected role of angiogenesis in the metastatic dissemination of CoM is still unknown. We show that cells derived from two CoM cell lines induce a strong angiogenic response when xenografted in zebrafish larvae. CoM cells are highly glycolytic and secrete lactate, which recruits and polarizes human and zebrafish macrophages towards a M2-like phenotype. These macrophages elevate the levels of proangiogenic factors such as VEGF, TGF-ß, and IL-10 in the tumor microenvironment to induce an angiogenic response towards the engrafted CoM cells in vivo. Chemical ablation of zebrafish macrophages or inhibition of glycolysis in CoM cells terminates this response, suggesting that attraction of lactate-dependent macrophages into engrafted CoM cells drives angiogenesis and serves as a possible dissemination mechanism for glycolytic CoM cells.

2.
Biochim Biophys Acta ; 1863(4): 607-16, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26748383

RESUMEN

Understanding of the regulation mechanisms of CXCR4 signaling is essential for revealing its role in physiological and pathological processes. Though biochemical pathways following CXCR4 activation by its ligand CXCL12 are well established, knowledge about the receptor dynamics on the plasma membrane remains limited. Here we used Ewing sarcoma-derived cells to unravel the processes that are involved in regulating CXCR4 dynamics on the plasma membrane during receptor signaling. Single-molecule epi-fluorescence microscopy showed that CXCR4 was present in monomeric state on the plasma membrane independent of receptor stimulation. However, upon activation freely diffusing receptors were immobilized in a ligand concentration-dependent manner. CXCR4 immobilization was strongly correlated with the ability for G-protein signaling and was a precursor of subsequent endocytotic events. Our data suggest that, a balanced regulation of G-protein dependent and independent pathways is required for controlling CXCR4 receptor mobility, and potentially subsequent controlled signal transduction.


Asunto(s)
Membrana Celular/metabolismo , Receptores CXCR4/metabolismo , Citoesqueleto de Actina/metabolismo , Endocitosis/genética , Proteínas de Unión al GTP/metabolismo , Humanos , Multimerización de Proteína , Transporte de Proteínas , Receptores CXCR4/genética , Transducción de Señal/genética , Vesículas Transportadoras/metabolismo , Células Tumorales Cultivadas
3.
Adv Exp Med Biol ; 916: 315-32, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27165360

RESUMEN

Zebrafish embryos can be obtained for research purposes in large numbers at low cost and embryos develop externally in limited space, making them highly suitable for high-throughput cancer studies and drug screens. Non-invasive live imaging of various processes within the larvae is possible due to their transparency during development, and a multitude of available fluorescent transgenic reporter lines.To perform high-throughput studies, handling large amounts of embryos and larvae is required. With such high number of individuals, even minute tasks may become time-consuming and arduous. In this chapter, an overview is given of the developments in the automation of various steps of large scale zebrafish cancer research for discovering important cancer pathways and drugs for the treatment of human disease. The focus lies on various tools developed for cancer cell implantation, embryo handling and sorting, microfluidic systems for imaging and drug treatment, and image acquisition and analysis. Examples will be given of employment of these technologies within the fields of toxicology research and cancer research.


Asunto(s)
Automatización , Modelos Animales de Enfermedad , Neoplasias/patología , Pez Cebra/embriología , Animales , Microfluídica , Microinyecciones
4.
J Pathol ; 233(4): 415-24, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24974828

RESUMEN

Translocations involving ETS-transcription factors, most commonly leading to the EWSR1-FLI1 fusion protein, are the hallmark of Ewing sarcoma. Despite knowledge of this driving molecular event, an effective therapeutic strategy is lacking. To test potential treatment regimes, we established a novel Ewing sarcoma zebrafish engraftment model allowing time-effective, dynamic quantification of Ewing sarcoma progression and tumour burden in vivo, applicable for screening of single and combined compounds. In Ewing sarcoma the tumour-suppressor gene TP53 is commonly found to be wild-type, thus providing an attractive target for treatment. Here, we study TP53 wild-type (EW7, CADO-ES1 and TC32) and TP53-deleted (SK-N-MC) Ewing sarcoma cell lines to investigate the potentiating effect of p53 reactivation by Nutlin-3 on treatment with YK-4-279 to block transcriptional activity of EWSR1-FLI1 protein. Blocking EWSR1-FLI1 transcriptional activity reduced Ewing sarcoma tumour cell burden irrespective of TP53 status. We show that simultaneous YK-4-279 treatment with Nutlin-3 to stabilize p53 resulted in an additive inhibition of TP53 wild-type Ewing sarcoma cell burden, whilst not affecting TP53-deleted Ewing sarcoma cells. Improved inhibition of proliferation and migration by combinatorial treatment was confirmed in vivo by zebrafish engraftments. Mechanistically, both compounds together additively induced apoptosis of tumour cells in vivo by engaging distinct pathways. We propose reactivation of the p53 pathway in combination with complementary targeted therapy by EWSR1-FLI1 transcriptional activity disruption as a valuable strategy against p53 wild-type Ewing sarcoma.


Asunto(s)
Neoplasias Óseas/prevención & control , Proteínas de Unión al ARN/genética , Sarcoma de Ewing/prevención & control , Transcripción Genética/fisiología , Proteína p53 Supresora de Tumor/fisiología , Proteínas de Pez Cebra/genética , Animales , Antineoplásicos/farmacología , Neoplasias Óseas/genética , Neoplasias Óseas/fisiopatología , Línea Celular Tumoral , Células Cultivadas , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Xenoinjertos , Humanos , Imidazoles/farmacología , Indoles/farmacología , Piperazinas/farmacología , Proteína EWS de Unión a ARN , Proteínas de Unión al ARN/efectos de los fármacos , Sarcoma de Ewing/genética , Sarcoma de Ewing/fisiopatología , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética , Proteína p53 Supresora de Tumor/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética , Pez Cebra , Proteínas de Pez Cebra/efectos de los fármacos
5.
Methods ; 62(3): 246-54, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23769806

RESUMEN

The increasing use of zebrafish larvae for biomedical research applications is resulting in versatile models for a variety of human diseases. These models exploit the optical transparency of zebrafish larvae and the availability of a large genetic tool box. Here we present detailed protocols for the robotic injection of zebrafish embryos at very high accuracy with a speed of up to 2000 embryos per hour. These protocols are benchmarked for several applications: (1) the injection of DNA for obtaining transgenic animals, (2) the injection of antisense morpholinos that can be used for gene knock-down, (3) the injection of microbes for studying infectious disease, and (4) the injection of human cancer cells as a model for tumor progression. We show examples of how the injected embryos can be screened at high-throughput level using fluorescence analysis. Our methods open up new avenues for the use of zebrafish larvae for large compound screens in the search for new medicines.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Larva/genética , Microinyecciones/métodos , Robótica/métodos , Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Benchmarking , Modelos Animales de Enfermedad , Embrión no Mamífero/inmunología , Embrión no Mamífero/microbiología , Embrión no Mamífero/ultraestructura , Técnicas de Silenciamiento del Gen , Ensayos Analíticos de Alto Rendimiento/instrumentación , Humanos , Larva/inmunología , Larva/microbiología , Larva/ultraestructura , Microscopía Fluorescente , Morfolinos/administración & dosificación , Mycobacterium tuberculosis/inmunología , Trasplante de Neoplasias , Oligonucleótidos Antisentido/administración & dosificación , Staphylococcus epidermidis/inmunología , Células Tumorales Cultivadas/trasplante , Pez Cebra/inmunología , Pez Cebra/microbiología
6.
Cancer Lett ; 587: 216657, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38336289

RESUMEN

Metastatic colonization by circulating cancer cells is a highly inefficient process. To colonize distant organs, disseminating cancer cells must overcome many obstacles in foreign microenvironments, and only a small fraction of them survives this process. How these disseminating cancer cells cope with stress and initiate metastatic process is not fully understood. In this study, we report that the metastatic onset of prostate cancer cells is associated with the dynamic conversion of metabolism signaling pathways governed by the energy sensors AMPK and mTOR. While in circulation in blood flow, the disseminating cancer cells display decreased mTOR and increased AMPK activities that protect them from stress-induced death. However, after metastatic onset, the mTOR-AMPK activities are reversed, enabling mTOR-dependent tumor growth. Suppression of this dynamic conversion by co-targeting of AMPK and mTOR signaling significantly suppresses prostate cancer cell and tumor organoid growth in vitro and experimental metastasis in vivo, suggesting that this can be a therapeutic approach against metastasizing prostate cancer.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Neoplasias de la Próstata , Masculino , Humanos , Proteínas Quinasas Activadas por AMP/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Transducción de Señal , Neoplasias de la Próstata/patología , Microambiente Tumoral
7.
Breast Cancer Res ; 15(6): R106, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24196484

RESUMEN

INTRODUCTION: The transforming growth factor beta (TGF-ß) signalling pathway is known to control human breast cancer invasion and metastasis. We demonstrate that the zebrafish xenograft assay is a robust and dependable animal model for examining the role of pharmacological modulators and genetic perturbation of TGF-ß signalling in human breast tumour cells. METHODS: We injected cancer cells into the embryonic circulation (duct of cuvier) and examined their invasion and metastasis into the avascular collagenous tail. Various aspects of the TGF-ß signalling pathway were blocked by chemical inhibition, small interfering RNA (siRNA), or small hairpin RNA (shRNA). Analysis was conducted using fluorescent microscopy. RESULTS: Breast cancer cells with different levels of malignancy, according to in vitro and in vivo mouse studies, demonstrated invasive and metastatic properties within the embryonic zebrafish model that nicely correlated with their differential tumourigenicity in mouse models. Interestingly, MCF10A M2 and M4 cells invaded into the caudal hematopoietic tissue and were visible as a cluster of cells, whereas MDA MB 231 cells invaded into the tail fin and were visible as individual cells. Pharmacological inhibition with TGF-ß receptor kinase inhibitors or tumour specific Smad4 knockdown disturbed invasion and metastasis in the zebrafish xenograft model and closely mimicked the results we obtained with these cells in a mouse metastasis model. Inhibition of matrix metallo proteinases, which are induced by TGF-ß in breast cancer cells, blocked invasion and metastasis of breast cancer cells. CONCLUSIONS: The zebrafish-embryonic breast cancer xenograft model is applicable for the mechanistic understanding, screening and development of anti-TGF-ß drugs for the treatment of metastatic breast cancer in a timely and cost-effective manner.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Factor de Crecimiento Transformador beta/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra/metabolismo , Animales , Benzamidas/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Cromonas/farmacología , Dioxoles/farmacología , Dipéptidos/farmacología , Modelos Animales de Enfermedad , Ensayos de Selección de Medicamentos Antitumorales/métodos , Embrión no Mamífero , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Morfolinas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , ARN Interferente Pequeño/farmacología , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Proteína Smad2/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Pez Cebra/embriología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
8.
Biochem Biophys Res Commun ; 435(1): 58-63, 2013 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-23618854

RESUMEN

TGF-ß plays a dual role in cancer; in early stages it inhibits tumor growth, whereas later it promotes invasion and metastasis. TGF-ß is thought to be pro-invasive by inducing epithelial-to-mesenchymal transition (EMT) via induction of transcriptional repressors, including Slug and Snail. In this study, we investigated the role of Snail and Slug in TGF-ß-induced invasion in an in vitro invasion assay and in an embryonic zebrafish xenograft model. Ectopic expression of Slug or Snail promoted invasion of single, rounded amoeboid cells in vitro. In an embryonic zebrafish xenograft model, forced expression of Slug and Snail promoted single cell invasion and metastasis. Slug and Snail are sufficient for the induction of single-cell invasion in an in vitro invasion assay and in an embryonic zebrafish xenograft model.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta3/farmacología , Animales , Western Blotting , Línea Celular , Movimiento Celular/genética , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Embrión no Mamífero/patología , Transición Epitelial-Mesenquimal/genética , Expresión Génica/efectos de los fármacos , Humanos , Ratones , Invasividad Neoplásica/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Pez Cebra/embriología , Pez Cebra/genética
9.
J Pathol ; 227(4): 431-45, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22374800

RESUMEN

Inhibition of VEGF signalling effectively suppresses localized tumour growth but accelerates tumour invasiveness and micrometastasis by unknown mechanisms. To study the dynamic and reciprocal interactions between tumour cells and their microenvironment during these processes, we established a xenograft model by injecting tumour cells into the blood circulation of transparent zebrafish embryos. This reproducibly results in rapid simultaneous formation of a localized tumour and experimental micrometastasis, allowing time-resolved imaging of both processes at single-cell resolution within 1 week. The tumour vasculature was initiated de novo by remodelling of primitive endothelial cells into a functional network. Roles of myeloid cells in critical tumourigenesis steps such as vascularization and invasion were revealed by genetic and pharmaceutical approaches. We discovered that the physiological migration of neutrophils controlled tumour invasion by conditioning the collagen matrix and forming the metastatic niche, as detected by two-photon confocal microscopy and second harmonic generation. Administration of VEGFR inhibitors blocked tumour vascularization and a localized tumour growth but enhanced migration of neutrophils, which in turn promoted tumour invasion and formation of micrometastasis. This demonstrates the in vivo cooperation between VEGF signalling and myeloid cells in metastasis and provides a new mechanism underlying the recent findings that VEGFR targeting can promote tumour invasiveness.


Asunto(s)
Modelos Animales de Enfermedad , Metástasis de la Neoplasia/fisiopatología , Neutrófilos/fisiología , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra/embriología , Animales , Beclometasona/farmacología , Neoplasias de la Mama/patología , Movimiento Celular/fisiología , Transformación Celular Neoplásica , Células Endoteliales/patología , Humanos , Indoles/farmacología , Ratones , Células Mieloides/patología , Células Mieloides/fisiología , Neutrófilos/patología , Pirroles/farmacología , Receptores de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Transducción de Señal/fisiología , Sunitinib
10.
Pharmaceuticals (Basel) ; 16(4)2023 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-37111355

RESUMEN

Uveal melanoma (UM) is a rare malignant cancer of the eye, with up to 50% of patients dying from metastasis, for which no effective treatment is available. Due to the rarity of the disease, there is a great need to harness the limited material available from primary tumors and metastases for advanced research and preclinical drug screening. We established a platform to isolate, preserve, and transiently recover viable tissues, followed by the generation of spheroid cultures derived from primary UM. All assessed tumor-derived samples formed spheroids in culture within 24 h and stained positive for melanocyte-specific markers, indicating the retention of their melanocytic origin. These short-lived spheroids were only maintained for the duration of the experiment (7 days) or re-established from frozen tumor tissue acquired from the same patient. Intravenous injection of fluorescently labeled UM cells derived from these spheroids into zebrafish yielded a reproducible metastatic phenotype and recapitulated molecular features of the disseminating UM. This approach allowed for the experimental replications required for reliable drug screening (at least 2 individual biological experiments, with n > 20). Drug treatments with navitoclax and everolimus validated the zebrafish patient-derived model as a versatile preclinical tool for screening anti-UM drugs and as a preclinical platform to predict personalized drug responses.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA