Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nat Commun ; 12(1): 1434, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33664264

ABSTRACT

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.


Subject(s)
Aneuploidy , DNA Copy Number Variations/genetics , Genome, Human/genetics , Melanoma/genetics , Skin Neoplasms/genetics , Disease Progression , Exome/genetics , Humans , INDEL Mutation/genetics , Melanocytes/pathology , Point Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Exome Sequencing , Whole Genome Sequencing , Melanoma, Cutaneous Malignant
2.
Oncotarget ; 7(47): 77163-77174, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27791198

ABSTRACT

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.


Subject(s)
Drug Resistance, Neoplasm , GTP Phosphohydrolases/genetics , Melanoma/genetics , Membrane Proteins/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Cell Line, Tumor , Evolution, Molecular , Female , Humans , Male , Melanoma/drug therapy , Neoplasm Metastasis , Protein Kinase Inhibitors/therapeutic use , Single-Cell Analysis , Exome Sequencing
3.
Clin Cancer Res ; 22(8): 2020-31, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26660520

ABSTRACT

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.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Azacitidine/pharmacology , Depsipeptides/pharmacology , Epigenesis, Genetic/drug effects , Lymphoma, T-Cell, Cutaneous/genetics , Acetylation , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cluster Analysis , CpG Islands , DNA Methylation/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Humans , Lymphoma, T-Cell, Cutaneous/drug therapy , Lymphoma, T-Cell, Cutaneous/pathology , Promoter Regions, Genetic , Protein Binding , Sezary Syndrome/genetics , Sezary Syndrome/pathology
5.
Genome Biol ; 16: 42, 2015 Feb 22.
Article in English | MEDLINE | ID: mdl-25885555

ABSTRACT

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.


Subject(s)
Melanoma/genetics , Neoplasm Invasiveness/genetics , SOX9 Transcription Factor/biosynthesis , Skin Neoplasms/genetics , Aged , Animals , Cell Line, Tumor , Cell Proliferation/genetics , DNA Methylation/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Melanoma/pathology , Mice , Middle Aged , Neoplasm Invasiveness/pathology , Neoplasm Staging , Prognosis , SOX9 Transcription Factor/genetics , Signal Transduction , Skin Neoplasms/pathology , Survival Analysis
6.
Exp Dermatol ; 24(5): 377-80, 2015 May.
Article in English | MEDLINE | ID: mdl-25739758

ABSTRACT

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.


Subject(s)
Biological Specimen Banks , Melanoma/pathology , Primary Cell Culture/methods , Skin Neoplasms/pathology , Biopsy , Cell Separation/methods , Fibroblasts/pathology , Humans , Melanoma/genetics , Melanoma/secondary , Mutation , Skin Neoplasms/genetics , Tumor Cells, Cultured , Workflow
7.
Tissue Eng Part A ; 21(5-6): 960-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25300246

ABSTRACT

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.


Subject(s)
Dermis/transplantation , Epidermis/transplantation , Pigmentation , Skin Transplantation , Tissue Engineering/methods , Adolescent , Animals , Cell Differentiation , Child , Child, Preschool , Dermis/cytology , Female , Fibroblasts/cytology , Humans , Infant , Keratin-9/metabolism , Male , Melanosomes/metabolism , Rats, Nude , Skin, Artificial , Stromal Cells/cytology
8.
Exp Dermatol ; 24(1): 22-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25363503

ABSTRACT

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.


Subject(s)
Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Melanoma/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Movement , Disease Progression , Humans , Neovascularization, Pathologic , Phenotype , Platelet-Derived Growth Factor/metabolism , RNA, Small Interfering/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tumor Cells, Cultured
9.
Exp Dermatol ; 24(1): 16-21, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25346346

ABSTRACT

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.


Subject(s)
Melanocytes/cytology , Skin Pigmentation , Skin, Artificial , Skin/metabolism , Tissue Engineering/methods , Animals , Biopsy , Cell Transplantation , Fibrillins , Fibroblasts/metabolism , Homeostasis , Humans , Keratinocytes/metabolism , Melanocytes/metabolism , Microfilament Proteins/metabolism , Pigmentation , Rats , Skin/pathology , Tropoelastin/metabolism
10.
Transl Oncol ; 7(2): 206-12, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24746711

ABSTRACT

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.
Article in English | MEDLINE | ID: mdl-23474946

ABSTRACT

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.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/pathology , Melanoma/pathology , Skin Neoplasms/pathology , Tumor Microenvironment/physiology , Biomarkers, Tumor/metabolism , Cell Proliferation , Disease Progression , Gene Expression Regulation, Neoplastic , Genetic Heterogeneity , Humans , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , MART-1 Antigen/genetics , MART-1 Antigen/metabolism , Melanoma/genetics , Melanoma/metabolism , Neoplasm Invasiveness , Phenotype , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Tumor Cells, Cultured
12.
Immunotherapy ; 5(2): 169-82, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23413908

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunotherapy/methods , Melanoma/therapy , Skin Neoplasms/therapy , Antibodies, Monoclonal/immunology , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Humans , Immunotherapy/trends , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/trends , Ipilimumab , Melanoma/immunology , Skin Neoplasms/immunology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation
13.
J Invest Dermatol ; 133(2): 316-24, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22971848

ABSTRACT

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.


Subject(s)
Keratinocytes/cytology , Melanins/metabolism , Melanocytes/cytology , Skin Transplantation/methods , Skin, Artificial , Sweat Glands/cytology , Adolescent , Animals , Cells, Cultured , Child , Child, Preschool , Dermis/cytology , Dermis/metabolism , Epidermal Cells , Epidermis/metabolism , Female , Humans , Infant , Keratinocytes/metabolism , Male , Melanocytes/metabolism , Mice , Mice, Nude , Organ Culture Techniques , Rats , Receptor, PAR-2/metabolism , Skin Pigmentation/physiology , Sweat Glands/metabolism , Transplantation, Heterologous
14.
Pigment Cell Melanoma Res ; 25(3): 343-53, 2012 May.
Article in English | MEDLINE | ID: mdl-22336146

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Gene Expression Profiling , Melanoma/classification , Skin Neoplasms/classification , Cell Proliferation , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genes, Neoplasm/genetics , Genetic Association Studies , Humans , Melanoma/genetics , Melanoma/pathology , Microarray Analysis , Neoplasm Invasiveness , Phenotype , Reproducibility of Results , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Tissue Distribution , Tumor Cells, Cultured/classification
15.
J Invest Dermatol ; 132(1): 144-53, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21881586

ABSTRACT

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.


Subject(s)
Melanoma/pathology , Organ Culture Techniques/methods , Skin Neoplasms/pathology , Skin, Artificial , Tissue Engineering/methods , Tumor Microenvironment/physiology , Animals , Cells, Cultured , Dermis/cytology , Disease Models, Animal , Epidermal Cells , Fibroblasts/cytology , Humans , Hypoxia/metabolism , Hypoxia/pathology , Immunocompromised Host , Keratinocytes/cytology , Melanocytes/cytology , Melanoma/blood supply , Melanoma/metabolism , Rats , Skin Neoplasms/blood supply , Skin Neoplasms/metabolism , Skin Transplantation , Transplantation, Heterologous
16.
Pigment Cell Melanoma Res ; 24(4): 631-42, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21599871

ABSTRACT

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.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Lymphoid Enhancer-Binding Factor 1/genetics , Melanoma/genetics , Melanoma/pathology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing/drug effects , Humans , Lymphoid Enhancer-Binding Factor 1/metabolism , Models, Biological , Neoplasm Invasiveness , Phenotype , Transcription Factor 4 , Transcription Factors/metabolism , Wnt Proteins/pharmacology , beta Catenin/metabolism
17.
Pigment Cell Melanoma Res ; 24(2): 326-33, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21176117

ABSTRACT

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.


Subject(s)
Melanoma/genetics , Melanoma/pathology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Phenotype , Proto-Oncogene Proteins B-raf/genetics , raf Kinases/antagonists & inhibitors , Adult , Aged , Cell Proliferation , Enzyme Inhibitors/therapeutic use , Female , Gene Expression Profiling , Humans , MAP Kinase Signaling System/physiology , Male , Melanoma/drug therapy , Melanoma/metabolism , Microarray Analysis , Middle Aged , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Neoplasm Invasiveness , Proto-Oncogene Proteins B-raf/metabolism
18.
Pigment Cell Melanoma Res ; 21(6): 665-76, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19067971

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Melanoma/genetics , Melanoma/pathology , Microphthalmia-Associated Transcription Factor/metabolism , Oligonucleotide Array Sequence Analysis , Biomarkers, Tumor/genetics , Blotting, Western , Humans , Melanoma/metabolism , Microphthalmia-Associated Transcription Factor/genetics , Transcription, Genetic , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL