Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 12(1): 1434, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33664264

RESUMEN

Although melanoma is initiated by acquisition of point mutations and limited focal copy number alterations in melanocytes-of-origin, the nature of genetic changes that characterise lethal metastatic disease is poorly understood. Here, we analyze the evolution of human melanoma progressing from early to late disease in 13 patients by sampling their tumours at multiple sites and times. Whole exome and genome sequencing data from 88 tumour samples reveals only limited gain of point mutations generally, with net mutational loss in some metastases. In contrast, melanoma evolution is dominated by whole genome doubling and large-scale aneuploidy, in which widespread loss of heterozygosity sculpts the burden of point mutations, neoantigens and structural variants even in treatment-naïve and primary cutaneous melanomas in some patients. These results imply that dysregulation of genomic integrity is a key driver of selective clonal advantage during melanoma progression.


Asunto(s)
Aneuploidia , Variaciones en el Número de Copia de ADN/genética , Genoma Humano/genética , Melanoma/genética , Neoplasias Cutáneas/genética , Progresión de la Enfermedad , Exoma/genética , Humanos , Mutación INDEL/genética , Melanocitos/patología , Mutación Puntual/genética , Polimorfismo de Nucleótido Simple/genética , Secuenciación del Exoma , Secuenciación Completa del Genoma , Melanoma Cutáneo Maligno
2.
Oncotarget ; 7(47): 77163-77174, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27791198

RESUMEN

Acquired chemotherapeutic resistance of cancer cells can result from a Darwinistic evolution process in which heterogeneity plays an important role. In order to understand the impact of genetic heterogeneity on acquired resistance and second line therapy selection in metastatic melanoma, we sequenced the exomes of 27 lesions which were collected from 3 metastatic melanoma patients treated with targeted or non-targeted inhibitors. Furthermore, we tested the impact of a second NRAS mutation in 7 BRAF inhibitor resistant early passage cell cultures on the selection of second line therapies.We observed a rapid monophyletic evolution of melanoma subpopulations in response to targeted therapy that was not observed in non-targeted therapy. We observed the acquisition of NRAS mutations in the BRAF mutated patient treated with a BRAF inhibitor in 1 of 5 of his post-resistant samples. In an additional cohort of 5 BRAF-inhibitor treated patients we detected 7 NRAS mutations in 18 post-resistant samples. No NRAS mutations were detected in pre-resistant samples. By sequencing 65 single cell clones we prove that NRAS mutations co-occur with BRAF mutations in single cells. The double mutated cells revealed a heterogeneous response to MEK, ERK, PI3K, AKT and multi RTK - inhibitors.We conclude that BRAF and NRAS co-mutations are not mutually exclusive. However, the sole finding of double mutated cells in a resistant tumor is not sufficient to determine follow-up therapy. In order to target the large pool of heterogeneous cells in a patient, we think combinational therapy targeting different pathways will be necessary.


Asunto(s)
Resistencia a Antineoplásicos , GTP Fosfohidrolasas/genética , Melanoma/genética , Proteínas de la Membrana/genética , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Línea Celular Tumoral , Evolución Molecular , Femenino , Humanos , Masculino , Melanoma/tratamiento farmacológico , Metástasis de la Neoplasia , Inhibidores de Proteínas Quinasas/uso terapéutico , Análisis de la Célula Individual , Secuenciación del Exoma
3.
Clin Cancer Res ; 22(8): 2020-31, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26660520

RESUMEN

PURPOSE: Cutaneous T-cell lymphomas (CTCL) are a heterogeneous group of malignancies that despite available therapies commonly relapse. The emergence of combination epigenetic therapies in other hematologic malignancies have made investigation of such combinations in CTCL a priority. Here, we explore the synergistic antiproliferative effects of romidepsin, an HDAC inhibitor, and azacitidine, a demethylating agent, combination in CTCL. EXPERIMENTAL DESIGN: The growth inhibition under combination treatment and single agent was explored by the MTT cell viability assay and the Annexin V/propidium iodide (PI) apoptosis assay in different CTCL cell lines and tumor cells derived from Sézary syndrome patients. Quantitative analysis of a dose-effect relationship of romidepsin and azacitidine was done by the CompuSyn software. Investigation of mechanism of action was performed by flow cytometry, immunoblotting, qRT-PCR arrays, and chromatin immunoprecipitation. Global CpG methylation sequencing was utilized to study genome methylation alteration under the treatment modalities. RESULTS: The combination of romidepsin and azacitidine exerts synergistic antiproliferative effects and induction of apoptosis involving activation of the caspase cascade in CTCL cell lines and tumor cells derived from Sézary syndrome patients. We identified genes that were selectively induced by the combination treatment, such as the tumor suppressor geneRhoBthat is linked to enhanced histone acetylation at its promoter region in parallel with pronounced expression of p21. Global CpG methylation sequencing in a CTCL cell line and tumor cells demonstrated a subset of genes with a unique change in methylation profile in the combination treatment. CONCLUSIONS: The synergistic antiproliferative effects of romidepsin and azacitidine combination treatment justify further exploration in clinical trials for advanced CTCL.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Azacitidina/farmacología , Depsipéptidos/farmacología , Epigénesis Genética/efectos de los fármacos , Linfoma Cutáneo de Células T/genética , Acetilación , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Análisis por Conglomerados , Islas de CpG , Metilación de ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Histonas/metabolismo , Humanos , Linfoma Cutáneo de Células T/tratamiento farmacológico , Linfoma Cutáneo de Células T/patología , Regiones Promotoras Genéticas , Unión Proteica , Síndrome de Sézary/genética , Síndrome de Sézary/patología
5.
Genome Biol ; 16: 42, 2015 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-25885555

RESUMEN

BACKGROUND: Melanoma is the most fatal skin cancer displaying a high degree of molecular heterogeneity. Phenotype switching is a mechanism that contributes to melanoma heterogeneity by altering transcription profiles for the transition between states of proliferation/differentiation and invasion/stemness. As phenotype switching is reversible, epigenetic mechanisms, like DNA methylation, could contribute to the changes in gene expression. RESULTS: Integrative analysis of methylation and gene expression datasets of five proliferative and five invasion melanoma cell cultures reveal two distinct clusters. SOX9 is methylated and lowly expressed in the highly proliferative group. SOX9 overexpression results in decreased proliferation but increased invasion in vitro. In a B16 mouse model, sox9 overexpression increases the number of lung metastases. Transcriptional analysis of SOX9-overexpressing melanoma cells reveals enrichment in epithelial to mesenchymal transition (EMT) pathways. Survival analysis of The Cancer Genome Atlas melanoma dataset shows that metastatic patients with high expression levels of SOX9 have significantly worse survival rates. Additional survival analysis on the targets of SOX9 reveals that most SOX9 downregulated genes have survival benefit for metastatic patients. CONCLUSIONS: Our genome-wide DNA methylation and gene expression study of 10 early passage melanoma cell cultures reveals two phenotypically distinct groups. One of the genes regulated by DNA methylation between the two groups is SOX9. SOX9 induces melanoma cell invasion and metastasis and decreases patient survival. A number of genes downregulated by SOX9 have a negative impact on patient survival. In conclusion, SOX9 is an important gene involved in melanoma invasion and negatively impacts melanoma patient survival.


Asunto(s)
Melanoma/genética , Invasividad Neoplásica/genética , Factor de Transcripción SOX9/biosíntesis , Neoplasias Cutáneas/genética , Anciano , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Metilación de ADN/genética , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Melanoma/patología , Ratones , Persona de Mediana Edad , Invasividad Neoplásica/patología , Estadificación de Neoplasias , Pronóstico , Factor de Transcripción SOX9/genética , Transducción de Señal , Neoplasias Cutáneas/patología , Análisis de Supervivencia
6.
Exp Dermatol ; 24(5): 377-80, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25739758

RESUMEN

Fibroblast contamination can make establishing primary melanoma cell cultures from native biopsies a major challenge, due to fibroblasts overgrowing the melanoma cells. Standard protocols therefore enrich for highly proliferative melanoma cells that grow well in vitro but may not represent the full range of in vivo tumor heterogeneity. Here we apply conditional methods that more effectively retrieve melanoma cells by differential trypsinization or by inducing fibroblast senescence through contact inhibition, serum starvation or deprivation of adhesion. Simple mixing experiments of melanoma and fibroblast cells demonstrated the efficacy of the new protocols in retrieving slow-growing melanoma cells. Applying our protocols to 20 cultures that had failed to grow by conventional methods, we could retrieve 12 (60%) validated melanoma cell cultures. Further application of the protocols in the live-cell biobank of 124 early passage cultures significantly improved recovery rates from 13% using standard protocols to 70% overall for the new workflow.


Asunto(s)
Bancos de Muestras Biológicas , Melanoma/patología , Cultivo Primario de Células/métodos , Neoplasias Cutáneas/patología , Biopsia , Separación Celular/métodos , Fibroblastos/patología , Humanos , Melanoma/genética , Melanoma/secundario , Mutación , Neoplasias Cutáneas/genética , Células Tumorales Cultivadas , Flujo de Trabajo
7.
Tissue Eng Part A ; 21(5-6): 960-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25300246

RESUMEN

It has been shown in vitro that melanocyte proliferation and function in palmoplantar skin is regulated by mesenchymal factors derived from fibroblasts. In this study, we investigated in vivo the influence of mesenchymal-epithelial interactions in human tissue-engineered skin substitutes reconstructed from palmar- and nonpalmoplantar-derived fibroblasts. Tissue-engineered dermo-epidermal analogs based on collagen type I hydrogels were populated with either human palmar or nonpalmoplantar fibroblasts and seeded with human nonpalmoplantar-derived melanocytes and keratinocytes. These skin substitutes were transplanted onto full-thickness skin wounds of immunoincompetent rats. Four weeks after transplantation the development of skin color was measured and grafts were excised and analyzed with regard to epidermal characteristics, in particular melanocyte number and function. Skin substitutes containing palmar-derived fibroblasts in comparison to nonpalmoplantar-derived fibroblasts showed (a) a significantly lighter pigmentation; (b) a reduced amount of epidermal melanin granules; and (c) a distinct melanosome expression. However, the number of melanocytes in the basal layer remained similar in both transplantation groups. These findings demonstrate that human palmar fibroblasts regulate the function of melanocytes in human pigmented dermo-epidermal skin substitutes after transplantation, whereas the number of melanocytes remains constant. This underscores the influence of site-specific stromal cells and their importance when constructing skin substitutes for clinical application.


Asunto(s)
Dermis/trasplante , Epidermis/trasplante , Pigmentación , Trasplante de Piel , Ingeniería de Tejidos/métodos , Adolescente , Animales , Diferenciación Celular , Niño , Preescolar , Dermis/citología , Femenino , Fibroblastos/citología , Humanos , Lactante , Queratina-9/metabolismo , Masculino , Melanosomas/metabolismo , Ratas Desnudas , Piel Artificial , Células del Estroma/citología
8.
Exp Dermatol ; 24(1): 22-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25363503

RESUMEN

Epithelial to mesenchymal transition (EMT) is a programme defined in epithelial cells and recognized as playing a critical role in cancer progression. Although melanoma is not a cancer of epithelial cells, hallmarks of EMT have been described to play a critical role in melanoma progression. Here, we demonstrate that long-term TGFß exposure can induce a dedifferentiated EMT-like state resembling a previously described invasive phenotype (EMT-like). TGFß-induced EMT-like is marked by the downregulation of melanocyte differentiation markers, such as MITF, and the upregulation of mesenchymal markers, such as N-cadherin, and an increase in melanoma cell migration and cell invasion. Pharmacological interference shows the dependency of TGFß-induced EMT-like on the activation of the PDGF signalling pathway and the subsequent activation of PI3K in human melanoma cells. Together, the data provide novel insights into the transcriptional plasticity of melanoma cells that might contribute to tumor progression in patients and propose avenues to therapeutic interventions.


Asunto(s)
Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Melanoma/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Movimiento Celular , Progresión de la Enfermedad , Humanos , Neovascularización Patológica , Fenotipo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , ARN Interferente Pequeño/metabolismo , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Células Tumorales Cultivadas
9.
Exp Dermatol ; 24(1): 16-21, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25346346

RESUMEN

In our laboratory, we have been using human pigmented dermo-epidermal skin substitutes for short-term experiments since several years. Little is known, however, about the long-term biology of such constructs after transplantation. We constructed human, melanocyte-containing dermo-epidermal skin substitutes of different (light and dark) pigmentation types and studied them in a long-term animal experiment. Developmental and maturational stages of the epidermal and dermal compartment as well as signs of homoeostasis were analysed 15 weeks after transplantation. Keratinocytes, melanocytes and fibroblasts from human skin biopsies were isolated and assembled into dermo-epidermal skin substitutes. These were transplanted onto immuno-incompetent rats and investigated 15 weeks after transplantation. Chromameter evaluation showed a consistent skin colour between 3 and 4 months after transplantation. Melanocytes resided in the epidermal basal layer in physiological numbers and melanin accumulated in keratinocytes in a supranuclear position. Skin substitutes showed a mature epidermis in a homoeostatic state and the presence of dermal components such as Fibrillin and Tropoelastin suggested advanced maturation. Overall, pigmented dermo-epidermal skin substitutes show a promising development towards achieving near-normal skin characteristics and epidermal and dermal tissue homoeostasis. In particular, melanocytes function correctly over several months whilst remaining in a physiological, epidermal position and yield a pigmentation resembling original donor skin colour.


Asunto(s)
Melanocitos/citología , Pigmentación de la Piel , Piel Artificial , Piel/metabolismo , Ingeniería de Tejidos/métodos , Animales , Biopsia , Trasplante de Células , Fibrilinas , Fibroblastos/metabolismo , Homeostasis , Humanos , Queratinocitos/metabolismo , Melanocitos/metabolismo , Proteínas de Microfilamentos/metabolismo , Pigmentación , Ratas , Piel/patología , Tropoelastina/metabolismo
10.
Transl Oncol ; 7(2): 206-12, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24746711

RESUMEN

Despite existing vaccination strategies targeting TRP-2, its function is not yet fully understood. TRP-2 is an enzyme involved in melanin biosynthesis and therefore discussed as a differentiation antigen. However, in mice Trp-2 was shown to be expressed in melanocyte stem cells of the hair follicle and therefore also considered as an indicator of stemness. A proper understanding of the TRP-2 function is crucial, considering a vaccination targeting cells with stemness properties would be highly effective in contrast to a therapy targeting differentiated melanoma cells. Analysing over 200 melanomas including primaries, partly matched metastases and patients' cell cultures we show that TRP-2 is correlated with Melan A expression and decreases with tumor progression. In mice it is expressed in differentiated melanocytes as well as in stem cells. Furthermore, we identify a TRP-2 negative, proliferative, hypoxia related cell subpopulation which is significantly associated with tumor thickness and diseases progression. Patients with a higher percentage of those cells have a less favourable tumor specific survival. Our findings underline that TRP-2 is a differentiation antigen, highlighting the importance to combine TRP-2 vaccination with other strategies targeting the aggressive undifferentiated hypoxia related subpopulation.

11.
J Invest Dermatol ; 133(10): 2436-2443, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23474946

RESUMEN

We have previously reported a model for melanoma progression in which oscillation between melanoma cell phenotypes characterized by invasion or proliferation is fundamental to tumor heterogeneity and disease progression. In this study we examine the possible role of hypoxia as one of the microenvironmental influences driving metastatic progression by promoting a switch from a proliferative to an invasive phenotype. Immunohistochemistry on primary human cutaneous melanoma biopsies showed intratumoral heterogeneity for cells expressing melanocytic markers, and a loss of these markers correlated with hypoxic regions. Furthermore, we show that the downregulation of melanocytic markers is dependent on hypoxia inducible factor 1α (HIF1α), a known regulator of the hypoxic response. In vitro invasion assays showed that a hypoxic environment increases the invasiveness of proliferative melanoma cell cultures in a HIF1α-dependent manner. In contrast, invasive phenotype melanoma cells showed no increase in invasive potential upon exposure to hypoxia. Thus, exposure of proliferative melanoma cells to hypoxic microenvironments is sufficient, in a HIF1α-dependent manner, to downregulate melanocytic marker expression and increase their invasive potential.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/patología , Melanoma/patología , Neoplasias Cutáneas/patología , Microambiente Tumoral/fisiología , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Heterogeneidad Genética , Humanos , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Antígeno MART-1/genética , Antígeno MART-1/metabolismo , Melanoma/genética , Melanoma/metabolismo , Invasividad Neoplásica , Fenotipo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Células Tumorales Cultivadas
12.
Immunotherapy ; 5(2): 169-82, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23413908

RESUMEN

The idea of cancer immunotherapy has been around for more than a century; however, the first immunotherapeutic ipilimumab, an anti-CTLA-4 antibody, has only recently been approved by the US FDA for melanoma. With an increasing understanding of the immune response, it is expected that more therapies will follow. This review aims to provide a general overview of immunotherapy in melanoma. We first explain the development of cancer immunotherapy more than a century ago and the general opinions about it over time. This is followed by a general overview of the immune reaction in order to give insight into the possible targets for therapy. Finally, we will discuss the current therapies for melanoma, their shortcomings and why it is important to develop patient stratification criteria. We conclude with an overview of recent discoveries and possible future therapies.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Inmunoterapia/métodos , Melanoma/terapia , Neoplasias Cutáneas/terapia , Anticuerpos Monoclonales/inmunología , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/inmunología , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Humanos , Inmunoterapia/tendencias , Inmunoterapia Adoptiva/métodos , Inmunoterapia Adoptiva/tendencias , Ipilimumab , Melanoma/inmunología , Neoplasias Cutáneas/inmunología , Linfocitos T/inmunología , Linfocitos T/trasplante
13.
J Invest Dermatol ; 133(2): 316-24, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22971848

RESUMEN

Recently, Biedermann et al. (2010) have demonstrated that human eccrine sweat gland cells can develop a multilayered epidermis. The question still remains whether these cells can fulfill exclusive and very specific functional properties of epidermal keratinocytes, such as the incorporation of melanin, a feature absent in sweat gland cells. We added human melanocytes to eccrine sweat gland cells to let them develop into an epidermal analog in vivo. The interaction between melanocytes and sweat gland-derived keratinocytes was investigated. The following results were gained: (1) macroscopically, a pigmentation of the substitutes was seen 2-3 weeks after transplantation; (2) we confirmed the development of a multilayered, stratified epidermis with melanocytes distributed evenly throughout the basal layer; (3) melanocytic dendrites projected to suprabasal layers; and (4) melanin was observed to be integrated into former eccrine sweat gland cells. These skin substitutes were similar or equal to skin substitutes cultured from human epidermal keratinocytes. The only differences observed were a delay in pigmentation and less melanin uptake. These data suggest that eccrine sweat gland cells can form a functional epidermal melanin unit, thereby providing striking evidence that they can assume one of the most characteristic keratinocyte properties.


Asunto(s)
Queratinocitos/citología , Melaninas/metabolismo , Melanocitos/citología , Trasplante de Piel/métodos , Piel Artificial , Glándulas Sudoríparas/citología , Adolescente , Animales , Células Cultivadas , Niño , Preescolar , Dermis/citología , Dermis/metabolismo , Células Epidérmicas , Epidermis/metabolismo , Femenino , Humanos , Lactante , Queratinocitos/metabolismo , Masculino , Melanocitos/metabolismo , Ratones , Ratones Desnudos , Técnicas de Cultivo de Órganos , Ratas , Receptor PAR-2/metabolismo , Pigmentación de la Piel/fisiología , Glándulas Sudoríparas/metabolismo , Trasplante Heterólogo
14.
Pigment Cell Melanoma Res ; 25(3): 343-53, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22336146

RESUMEN

There is growing evidence that the metastatic spread of melanoma is driven not by a linear increase in tumorigenic aggressiveness, but rather by switching back and forth between two different phenotypes of metastatic potential. In vitro these phenotypes are respectively defined by the characteristics of strong proliferation/weak invasiveness and weak proliferation/strong invasiveness. Melanoma cell phenotype is tightly linked to gene expression. Taking advantage of this, we have developed a gene expression-based tool for predicting phenotype called Heuristic Online Phenotype Prediction. We demonstrate the predictive utility of this tool by comparing phenotype-specific signatures with measurements of characteristics of melanoma phenotype-specific biology in different melanoma cell lines and short-term cultures. We further show that 86% of 536 tested melanoma lines and short-term cultures are significantly associated with the phenotypes we describe. These findings reinforce the concept that a two-state system, as described by the phenotype switching model, underlies melanoma progression.


Asunto(s)
Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Perfilación de la Expresión Génica , Melanoma/clasificación , Neoplasias Cutáneas/clasificación , Proliferación Celular , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Genes Relacionados con las Neoplasias/genética , Estudios de Asociación Genética , Humanos , Melanoma/genética , Melanoma/patología , Análisis por Micromatrices , Invasividad Neoplásica , Fenotipo , Reproducibilidad de los Resultados , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Distribución Tisular , Células Tumorales Cultivadas/clasificación
15.
J Invest Dermatol ; 132(1): 144-53, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21881586

RESUMEN

To overcome the lack of effective therapeutics for aggressive melanoma, new research models closely resembling the human disease are required. Here we report the development of a fully orthotopic, humanized in vivo model for melanoma, faithfully recapitulating human disease initiation and progression. To this end, human melanoma cells were seeded into engineered human dermo-epidermal skin substitutes. Transplantation onto the back of immunocompromised rats consistently resulted in the development of melanoma, displaying the hallmarks of their parental tumors. Importantly, all initial steps of disease progression were recapitulated, including the incorporation of the tumor cells into their physiological microenvironment, transition of radial to vertical growth, and establishment of highly vascularized, aggressive tumors with dermal involvement. Because all cellular components can be individually accessed using this approach, it allows manipulation of the tumor cells, as well as of the keratinocyte and stromal cell populations. Therefore, in one defined model system, tumor cell-autonomous and non-autonomous pathways regulating human disease progression can be investigated in a humanized, clinically relevant context.


Asunto(s)
Melanoma/patología , Técnicas de Cultivo de Órganos/métodos , Neoplasias Cutáneas/patología , Piel Artificial , Ingeniería de Tejidos/métodos , Microambiente Tumoral/fisiología , Animales , Células Cultivadas , Dermis/citología , Modelos Animales de Enfermedad , Células Epidérmicas , Fibroblastos/citología , Humanos , Hipoxia/metabolismo , Hipoxia/patología , Huésped Inmunocomprometido , Queratinocitos/citología , Melanocitos/citología , Melanoma/irrigación sanguínea , Melanoma/metabolismo , Ratas , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/metabolismo , Trasplante de Piel , Trasplante Heterólogo
16.
Pigment Cell Melanoma Res ; 24(4): 631-42, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21599871

RESUMEN

Recent observations suggest that melanoma cells drive disease progression by switching back and forth between phenotypic states of proliferation and invasion. Phenotype switching has been linked to changes in Wnt signalling, and we therefore looked for cell phenotype-specific differences in the levels and activity of ß-catenin and its LEF/TCF co-factors. We found that while cytosolic ß-catenin distribution is phenotype-specific (membrane-associated in proliferative cells and cytosolic in invasive cells), its nuclear distribution and activity is not. Instead, the expression patterns of two ß-catenin co-factors, LEF1 and TCF4, are both phenotype-specific and inversely correlated. LEF1 is preferentially expressed by differentiated/proliferative phenotype cells and TCF4 by dedifferentiated/invasive phenotype cells. Knock-down experiments confirmed that these co-factors are important for the phenotype-specific expression of M-MITF, WNT5A and other genes and that LEF1 suppresses TCF4 expression independently of ß-catenin. Our data show that melanoma cell phenotype switching behaviour is regulated by differential LEF1/TCF4 activity.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factor de Unión 1 al Potenciador Linfoide/genética , Melanoma/genética , Melanoma/patología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Factores de Transcripción/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Modelos Biológicos , Invasividad Neoplásica , Fenotipo , Factor de Transcripción 4 , Factores de Transcripción/metabolismo , Proteínas Wnt/farmacología , beta Catenina/metabolismo
17.
Pigment Cell Melanoma Res ; 24(2): 326-33, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21176117

RESUMEN

Oncogenic mutations within the MAPK pathway are frequent in melanoma, and targeting of MAPK signaling has yielded spectacular responses in a significant number of patients that last for several months before relapsing. We investigated the effects of two different inhibitors of MAPK signaling in proliferative and invasive melanoma cell cultures with various mutations in the MAPK pathway. Proliferative melanoma cells were more susceptible to pathway inhibition than invasive phenotype cells, irrespective of BRAF mutation status, while invasive phenotype cell response was dependent on BRAF mutation status. Critically, MAPK pathway inhibition of proliferative phenotype cells resulted in acquisition of invasive phenotype characteristics. These results show that melanoma cell phenotype is an important factor in MAPK pathway inhibition response. This suggests that while current therapeutic strategies target proliferative melanoma cells, future approaches should also account for the invasive phenotype population.


Asunto(s)
Melanoma/genética , Melanoma/patología , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Fenotipo , Proteínas Proto-Oncogénicas B-raf/genética , Quinasas raf/antagonistas & inhibidores , Adulto , Anciano , Proliferación Celular , Inhibidores Enzimáticos/uso terapéutico , Femenino , Perfilación de la Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Análisis por Micromatrices , Persona de Mediana Edad , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Mutación , Invasividad Neoplásica , Proteínas Proto-Oncogénicas B-raf/metabolismo
18.
Pigment Cell Melanoma Res ; 21(6): 665-76, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19067971

RESUMEN

Malignant melanoma is a chemotherapy-resistant cancer with high mortality. Recent advances in our understanding of the disease at the molecular level have indicated that it shares many characteristics with developmental precursors to melanocytes, the mature pigment-producing cells of the skin and hair follicles. The development of melanocytes absolutely depends on the action of the microphthalmia-associated transcription factor (MITF). MITF has been shown to regulate a broad variety of genes, whose functions range from pigment production to cell-cycle regulation, migration and survival. However, the existing list of targets is not sufficient to explain the role of MITF in melanocyte development and melanoma progression. DNA microarray analysis of gene expression offers a straightforward approach to identify new target genes, but standard analytical procedures are susceptible to the generation of false positives and require additional experimental steps for validation. Here, we introduce a new strategy where two DNA microarray-based approaches for identifying transcription factor targets are combined in a cross-validation protocol designed to help control false-positive generation. We use this two-step approach to successfully re-identify thirteen previously recorded targets of MITF-mediated upregulation, as well as 71 novel targets. Many of these new targets have known relevance to pigmentation and melanoma biology, and further emphasize the critical role of MITF in these processes.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Regulación Neoplásica de la Expresión Génica , Melanoma/genética , Melanoma/patología , Factor de Transcripción Asociado a Microftalmía/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Biomarcadores de Tumor/genética , Western Blotting , Humanos , Melanoma/metabolismo , Factor de Transcripción Asociado a Microftalmía/genética , Transcripción Genética , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...