Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Invest Dermatol ; 143(6): 1042-1051.e3, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36566878

RESUMEN

Phakomatosis pigmentovascularis is a diagnosis that denotes the coexistence of pigmentary and vascular birthmarks of specific types, accompanied by variable multisystem involvement, including CNS disease, asymmetrical growth, and a predisposition to malignancy. Using a tight phenotypic group and high-depth next-generation sequencing of affected tissues, we discover here clonal mosaic variants in gene PTPN11 encoding SHP2 phosphatase as a cause of phakomatosis pigmentovascularis type III or spilorosea. Within an individual, the same variant is found in distinct pigmentary and vascular birthmarks and is undetectable in blood. We go on to show that the same variants can cause either the pigmentary or vascular phenotypes alone, and drive melanoma development within pigmentary lesions. Protein structure modeling highlights that although variants lead to loss of function at the level of the phosphatase domain, resultant conformational changes promote longer ligand binding. In vitro modeling of the missense variants confirms downstream MAPK pathway overactivation and widespread disruption of human endothelial cell angiogenesis. Importantly, patients with PTPN11 mosaicism theoretically risk passing on the variant to their children as the germline RASopathy Noonan syndrome with lentigines. These findings improve our understanding of the pathogenesis and biology of nevus spilus and capillary malformation syndromes, paving the way for better clinical management.


Asunto(s)
Lentigo , Melanoma , Síndromes Neurocutáneos , Niño , Humanos , Síndromes Neurocutáneos/genética , Síndromes Neurocutáneos/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Mosaicismo , Melanoma/genética
2.
Dis Model Mech ; 15(9)2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35929478

RESUMEN

Melanoma heterogeneity and plasticity underlie therapy resistance. Some tumour cells possess innate resistance, while others reprogramme during drug exposure and survive to form persister cells, a source of potential cancer cells for recurrent disease. Tracing individual melanoma cell populations through tumour regression and into recurrent disease remains largely unexplored, in part, because complex animal models are required for live imaging of cell populations over time. Here, we applied tamoxifen-inducible creERt2/loxP lineage tracing to a zebrafish model of MITF-dependent melanoma regression and recurrence to image and trace cell populations in vivo through disease stages. Using this strategy, we show that melanoma persister cells at the minimal residual disease site originate from the primary tumour. Next, we fate mapped rare MITF-independent persister cells and demonstrate that these cells directly contribute to progressive disease. Multiplex immunohistochemistry confirmed that MITF-independent persister cells give rise to Mitfa+ cells in recurrent disease. Taken together, our work reveals a direct contribution of persister cell populations to recurrent disease, and provides a resource for lineage-tracing methodology in adult zebrafish cancer models.


Asunto(s)
Melanoma , Pez Cebra , Animales , Melanoma/patología , Factor de Transcripción Asociado a Microftalmía/genética , Tamoxifeno/farmacología , Proteínas de Pez Cebra
3.
Cell Rep ; 38(2): 110234, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35021087

RESUMEN

Melanocytes, the pigment-producing cells, are replenished from multiple stem cell niches in adult tissue. Although pigmentation traits are known risk factors for melanoma, we know little about melanocyte stem cell (McSC) populations other than hair follicle McSCs and lack key lineage markers with which to identify McSCs and study their function. Here we find that Tfap2b and a select set of target genes specify an McSC population at the dorsal root ganglia in zebrafish. Functionally, Tfap2b is required for only a few late-stage embryonic melanocytes, and is essential for McSC-dependent melanocyte regeneration. Fate mapping data reveal that tfap2b+ McSCs have multifate potential, and are the cells of origin for large patches of adult melanocytes, two other pigment cell types (iridophores and xanthophores), and nerve-associated cells. Hence, Tfap2b confers McSC identity in early development, distinguishing McSCs from other neural crest and pigment cell lineages, and retains multifate potential in the adult zebrafish.


Asunto(s)
Melanocitos/metabolismo , Células Madre/clasificación , Factor de Transcripción AP-2/metabolismo , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Melanocitos/fisiología , Pigmentación/genética , Piel/metabolismo , Pigmentación de la Piel/genética , Células Madre/metabolismo , Factor de Transcripción AP-2/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética
4.
Sci Rep ; 11(1): 10123, 2021 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-33980872

RESUMEN

In vitro, depending on extracellular matrix (ECM) architecture, macrophages migrate either in amoeboid or mesenchymal mode; while the first is a general trait of leukocytes, the latter is associated with tissue remodelling via Matrix Metalloproteinases (MMPs). To assess whether these stereotyped migrations could be also observed in a physiological context, we used the zebrafish embryo and monitored macrophage morphology, behaviour and capacity to mobilise haematopoietic stem/progenitor cells (HSPCs), as a final functional readout. Morphometric analysis identified 4 different cell shapes. Live imaging revealed that macrophages successively adopt all four shapes as they migrate through ECM. Treatment with inhibitors of MMPs or Rac GTPase to abolish mesenchymal migration, suppresses both ECM degradation and HSPC mobilisation while differently affecting macrophage behaviour. This study depicts real time macrophage behaviour in a physiological context and reveals extreme reactivity of these cells constantly adapting and switching migratory shapes to achieve HSPCs proper mobilisation.


Asunto(s)
Movimiento Celular , Plasticidad de la Célula , Macrófagos/citología , Macrófagos/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rac/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Humanos , Macrófagos/efectos de los fármacos , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Transducción de Señal/efectos de los fármacos , Pez Cebra
5.
Elife ; 102021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33438577

RESUMEN

The microphthalmia-associated transcription factor (MITF) is a critical regulator of melanocyte development and differentiation. It also plays an important role in melanoma where it has been described as a molecular rheostat that, depending on activity levels, allows reversible switching between different cellular states. Here, we show that MITF directly represses the expression of genes associated with the extracellular matrix (ECM) and focal adhesion pathways in human melanoma cells as well as of regulators of epithelial-to-mesenchymal transition (EMT) such as CDH2, thus affecting cell morphology and cell-matrix interactions. Importantly, we show that these effects of MITF are reversible, as expected from the rheostat model. The number of focal adhesion points increased upon MITF knockdown, a feature observed in drug-resistant melanomas. Cells lacking MITF are similar to the cells of minimal residual disease observed in both human and zebrafish melanomas. Our results suggest that MITF plays a critical role as a repressor of gene expression and is actively involved in shaping the microenvironment of melanoma cells in a cell-autonomous manner.


Asunto(s)
Transición Epitelial-Mesenquimal , Matriz Extracelular/metabolismo , Adhesiones Focales/metabolismo , Factor de Transcripción Asociado a Microftalmía/genética , Línea Celular Tumoral , Humanos , Melanoma/metabolismo , Factor de Transcripción Asociado a Microftalmía/metabolismo
6.
Pigment Cell Melanoma Res ; 34(2): 150-162, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32910840

RESUMEN

Oncogenic BRAF and NRAS mutations drive human melanoma initiation. We used transgenic zebrafish to model NRAS-mutant melanoma, and the rapid tumor onset allowed us to study candidate tumor suppressors. We identified P38α-MAPK14 as a potential tumor suppressor in The Cancer Genome Atlas melanoma cohort of NRAS-mutant melanomas, and overexpression significantly increased the time to tumor onset in transgenic zebrafish with NRAS-driven melanoma. Pharmacological activation of P38α-MAPK14 using anisomycin reduced in vitro viability of melanoma cultures, which we confirmed by stable overexpression of p38α. We observed that the viability of MEK inhibitor resistant melanoma cells could be reduced by combined treatment of anisomycin and MEK inhibition. Our study demonstrates that activating the p38α-MAPK14 pathway in the presence of oncogenic NRAS abrogates melanoma in vitro and in vivo. SIGNIFICANCE: The significance of our study is in the accountability of NRAS mutations in melanoma. We demonstrate here that activation of p38α-MAPK14 pathway can abrogate NRAS-mutant melanoma which is contrary to the previously published role of p38α-MAPK14 pathway in BRAF mutant melanoma. These results implicate that BRAF and NRAS-mutant melanoma may not be identical biologically. We also demonstrate the translational benefit of our study by using a small molecule compound-anisomycin (already in use for other diseases in clinical trials) to activate p38α-MAPK14 pathway.


Asunto(s)
GTP Fosfohidrolasas/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Melanoma/prevención & control , Proteínas de la Membrana/genética , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Mutación , Animales , Anisomicina/farmacología , Apoptosis , Proliferación Celular , Humanos , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 14 Activada por Mitógenos/genética , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de la Síntesis de la Proteína/farmacología , Células Tumorales Cultivadas , Pez Cebra
7.
J Invest Dermatol ; 141(6): 1389-1394, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33340501

RESUMEN

Dynamic cellular heterogeneity underlies melanoma progression and therapy resistance. Advances in single-cell technologies have revealed an increasing number of tumor and microenvironment cell states in melanoma, but little is understood about their function in vivo. Zebrafish models are a powerful system for discovery, live imaging, and functional investigation of cell states throughout melanoma progression and treatment. By capturing dynamic melanoma states in living animals, zebrafish have the potential to resolve the complexity of melanoma heterogeneity from a single cell through disease processes within the context of the whole body, revealing novel cancer biology and therapeutic targets.


Asunto(s)
Plasticidad de la Célula/genética , Transformación Celular Neoplásica/genética , Melanoma/genética , Neoplasias Cutáneas/genética , Animales , Animales Modificados Genéticamente , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Plasticidad de la Célula/efectos de los fármacos , Transformación Celular Neoplásica/patología , Modelos Animales de Enfermedad , Ensayos de Selección de Medicamentos Antitumorales , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/tratamiento farmacológico , Melanoma/patología , Mutación , RNA-Seq , Análisis de la Célula Individual , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Pez Cebra
8.
Cell Mol Life Sci ; 77(17): 3453-3464, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31732791

RESUMEN

During embryogenesis of all vertebrates, haematopoietic stem/progenitor cells (HSPCs) extrude from the aorta by a complex process named endothelial-to-haematopoietic transition (EHT). HSPCs will then colonize haematopoietic organs allowing haematopoiesis throughout adult life. The mechanism underlying EHT including the role of each aortic endothelial cell (EC) within the global aorta dynamics remains unknown. In the present study, we show for the first time that EHT involves the remodelling of individual cells within a collective migration of ECs which is tightly orchestrated, resulting in HSPCs extrusion in the sub-aortic space without compromising aorta integrity. By performing a cross-disciplinary study which combines high-resolution 4D imaging and theoretical analysis based on the concepts of classical mechanics, we propose that this complex developmental process is dependent on mechanical instabilities of the aorta preparing and facilitating the extrusion of HSPCs.


Asunto(s)
Aorta/fisiología , Células Madre Hematopoyéticas/metabolismo , Amidas/farmacología , Aminoquinolinas/farmacología , Animales , Animales Modificados Genéticamente/metabolismo , Embrión no Mamífero/citología , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Células Endoteliales/citología , Hematopoyesis , Células Madre Hematopoyéticas/citología , Microscopía Fluorescente , Piridinas/farmacología , Pirimidinas/farmacología , Imagen de Lapso de Tiempo , Pez Cebra/crecimiento & desarrollo
9.
Cancer Res ; 79(22): 5769-5784, 2019 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-31582381

RESUMEN

The melanocyte-inducing transcription factor (MITF)-low melanoma transcriptional signature is predictive of poor outcomes for patients, but little is known about its biological significance, and animal models are lacking. Here, we used zebrafish genetic models with low activity of Mitfa (MITF-low) and established that the MITF-low state is causal of melanoma progression and a predictor of melanoma biological subtype. MITF-low zebrafish melanomas resembled human MITF-low melanomas and were enriched for stem and invasive (mesenchymal) gene signatures. MITF-low activity coupled with a p53 mutation was sufficient to promote superficial growth melanomas, whereas BRAFV600E accelerated MITF-low melanoma onset and further promoted the development of MITF-high nodular growth melanomas. Genetic inhibition of MITF activity led to rapid regression; recurrence occurred following reactivation of MITF. At the regression site, there was minimal residual disease that was resistant to loss of MITF activity (termed MITF-independent cells) with very low-to-no MITF activity or protein. Transcriptomic analysis of MITF-independent residual disease showed enrichment of mesenchymal and neural crest stem cell signatures similar to human therapy-resistant melanomas. Single-cell RNA sequencing revealed MITF-independent residual disease was heterogeneous depending on melanoma subtype. Further, there was a shared subpopulation of residual disease cells that was enriched for a neural crest G0-like state that preexisted in the primary tumor and remained present in recurring melanomas. These findings suggest that invasive and stem-like programs coupled with cellular heterogeneity contribute to poor outcomes for MITF-low melanoma patients and that MITF-independent subpopulations are an important therapeutic target to achieve long-term survival outcomes. SIGNIFICANCE: This study provides a useful model for MITF-low melanomas and MITF-independent cell populations that can be used to study the mechanisms that drive these tumors as well as identify potential therapeutic options.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/22/5769/F1.large.jpg.


Asunto(s)
Melanoma/genética , Factor de Transcripción Asociado a Microftalmía/genética , Neoplasia Residual/genética , Transcripción Genética/genética , Pez Cebra/genética , Animales , Resistencia a Medicamentos/genética , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/genética , Melanocitos/patología , Melanoma/patología , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Neoplasia Residual/patología , Cresta Neural/patología , Proteínas Proto-Oncogénicas B-raf/genética , Células Madre/patología
10.
Cell Death Dis ; 9(6): 709, 2018 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-29899536

RESUMEN

Correction to: Cell Death Dis. 8, e2979 (2017); https://doi.org/10.1038/cddis.2017.374 ; published online 10th August 2017.

11.
Cell Death Dis ; 8(8): e2979, 2017 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-28796253

RESUMEN

Macrophages are essential for appendage regeneration after amputation in regenerative species. The molecular mechanisms through which macrophages orchestrate blastema formation and regeneration are still unclear. Here, we use the genetically tractable and transparent zebrafish larvae to study the functions of polarized macrophage subsets during caudal fin regeneration. After caudal fin amputation, we show an early and transient accumulation of pro-inflammatory macrophages concomitant with the accumulation of non-inflammatory macrophages which, in contrast to pro-inflammatory macrophages, remain associated to the fin until the end of the regeneration. Chemical and genetic depletion of macrophages suggested that early recruited macrophages that express TNFα are critical for blastema formation. Combining parabiosis and morpholino knockdown strategies, we show that TNFα/TNFR1 signaling pathway is required for the fin regeneration. Our study reveals that TNFR1 has a necessary and direct role in blastema cell activation suggesting that macrophage subset balance provides the accurate TNFα signal to prime regeneration in zebrafish.


Asunto(s)
Larva/metabolismo , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Extremidades/embriología , Extremidades/fisiología , Larva/genética , Macrófagos/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Regeneración/genética , Regeneración/fisiología , Transducción de Señal/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
12.
Elife ; 4: e07288, 2015 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-26154973

RESUMEN

While the mammalian macrophage phenotypes have been intensively studied in vitro, the dynamic of their phenotypic polarization has never been investigated in live vertebrates. We used the zebrafish as a live model to identify and trail macrophage subtypes. We generated a transgenic line whose macrophages expressing tumour necrosis factor alpha (tnfa), a key feature of classically activated (M1) macrophages, express fluorescent proteins Tg(mpeg1:mCherryF/tnfa:eGFP-F). Using 4D-confocal microscopy, we showed that both aseptic wounding and Escherichia coli inoculation triggered macrophage recruitment, some of which started to express tnfa. RT-qPCR on Fluorescence Activated Cell Sorting (FACS)-sorted tnfa(+) and tnfa(-) macrophages showed that they, respectively, expressed M1 and alternatively activated (M2) mammalian markers. Fate tracing of tnfa(+) macrophages during the time-course of inflammation demonstrated that pro-inflammatory macrophages converted into M2-like phenotype during the resolution step. Our results reveal the diversity and plasticity of zebrafish macrophage subsets and underline the similarities with mammalian macrophages proposing a new system to study macrophage functional dynamic.


Asunto(s)
Macrófagos/clasificación , Macrófagos/inmunología , Pez Cebra/inmunología , Animales , Animales Modificados Genéticamente , Infecciones por Escherichia coli/inmunología , Citometría de Flujo , Perfilación de la Expresión Génica , Genes Reporteros , Microscopía Confocal , Datos de Secuencia Molecular , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Factor de Necrosis Tumoral alfa/biosíntesis , Heridas y Lesiones/inmunología
13.
Nat Commun ; 6: 6227, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25686881

RESUMEN

In vertebrates, haematopoietic stem/progenitor cells (HSPCs) first emerge in the aorta-gonad-mesonephros (AGM) before colonizing transitory and subsequently definitive haematopoietic organs allowing haematopoiesis throughout adult life. Here we identify an unexpected primitive macrophage population accumulated in the dorsal mesenteric mesoderm surrounding the dorsal aorta of the human embryo and study its function in the transparent zebrafish embryo. Our study reveals dynamic interactions occurring between the HSPCs and primitive macrophages in the AGM. Specific chemical and inducible genetic depletion of macrophages or inhibition of matrix metalloproteinases (Mmps) leads to an accumulation of HSPCs in the AGM and a decrease in the colonization of haematopoietic organs. Finally, in vivo zymography demonstrates the function of primitive macrophages in extracellular matrix degradation, which allows HSPC migration through the AGM stroma, their intravasation, leading to the colonization of haematopoietic organs and the establishment of definitive haematopoiesis.


Asunto(s)
Aorta/embriología , Hematopoyesis , Células Madre Hematopoyéticas/citología , Macrófagos/citología , Células Madre/citología , Animales , Animales Modificados Genéticamente , Linaje de la Célula , Biología Evolutiva , Matriz Extracelular/metabolismo , Gónadas/embriología , Humanos , Macrófagos/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Mesonefro/embriología , Microscopía Fluorescente , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...