Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cell Reports ; 8(5): 1379-1391, 2017 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-28392221

RESUMO

A point mutation in the BRAF gene, leading to a constitutively active form of the protein, is present in 45%-60% of patients and acts as a key driver in melanoma. Shortly after therapy induction, resistance to MAPK pathway-specific inhibitors develops, indicating that pathway inhibition is circumvented by epigenetic mechanisms. Here, we mimicked epigenetic modifications in melanoma cells by reprogramming them into metastable induced pluripotent cancer cells (iPCCs) with the ability to terminally differentiate into non-tumorigenic lineages. iPCCs and their differentiated progeny were characterized by an increased resistance against targeted therapies, although the cells harbor the same oncogenic mutations and signaling activity as the parental melanoma cells. Furthermore, induction of a pluripotent state allowed the melanoma-derived cells to acquire a non-tumorigenic cell fate, further suggesting that tumorigenicity is influenced by the cell state.


Assuntos
Carcinogênese/patologia , Reprogramação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Melanoma/patologia , Células-Tronco Neoplásicas/citologia , Animais , Antineoplásicos/farmacologia , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Linhagem Celular , Linhagem da Célula , Células Cultivadas , Epigênese Genética , Fibroblastos/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/transplante , Neurônios/citologia , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo
2.
Sci Rep ; 6: 28891, 2016 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-27387763

RESUMO

Lineage-specific transcription factors determine the cell fate during development. Direct conversion of several cell types into other lineages has been achieved by the overexpression of specific transcription factors. Even cancer cells have been demonstrated to be amenable to transdifferentiation. Here, we identified a distinct set of transcription factors, which are sufficient to transform cells of the keratinocytic lineage to melanocyte-like cells. Melanocyte marker expression was induced and melanosome formation was observed in non-tumorigenic keratinocytes (HaCaT) and tumorigenic squamous cell carcinoma (MET-4) cells. Moreover, reduced proliferation, cell metabolism, invasion and migration were measured in vitro in transdifferentiated MT-MET-4 cells. A loss of tumorigenic potential of squamous cell carcinoma cells could be due to the upregulation of the melanocyte differentiation associated gene IL-24. Our data show that cells from the keratinocytic lineage can be transdifferented into the melanocytic lineage and provide a proof of principle for a potential new therapeutic strategy.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Transdiferenciação Celular , Queratinócitos/citologia , Melanócitos/citologia , Neoplasias Cutâneas/metabolismo , Animais , Carcinogênese/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Linhagem da Célula , Movimento Celular , Proliferação de Células , Aberrações Cromossômicas , Clonagem Molecular , Hibridização Genômica Comparativa , Metilação de DNA , Perfilação da Expressão Gênica , Humanos , Interleucinas/metabolismo , Camundongos , Invasividade Neoplásica , Transplante de Neoplasias , Fenótipo , Polimorfismo de Nucleotídeo Único , Transdução de Sinais
3.
Pigment Cell Melanoma Res ; 29(4): 453-8, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27105574

RESUMO

The sry-related high-mobility box (SOX)-2 protein has recently been proven to play a significant role in progression, metastasis, and clinical prognosis spanning several cancer types. Research on the role of SOX2 in melanoma is limited and currently little is known about the mechanistic function of this gene in this context. Here, we observed high expression of SOX2 in both human melanoma cell lines and primary melanomas in contrast to melanocytic nevi. This overexpression in melanoma can, in part, be explained by extra gene copy numbers of SOX2 in primary samples. Interestingly, we were able to induce SOX2 expression, mediated by SOX4, via TGF-ß1 stimulation in a time-dependent manner. Moreover, the knockdown of SOX2 impaired TGF-ß-induced invasiveness. This phenotype switch can be explained by SOX2-mediated cross talk between TGF-ß and non-canonical Wnt signaling. Thus, we propose that SOX2 is involved in the critical TGF-ß signaling pathway, which has been shown to correlate with melanoma aggressiveness and metastasis. In conclusion, we have identified a novel downstream factor of TGF-ß signaling in melanoma, which may have further implications in the clinic.


Assuntos
Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Melanoma/metabolismo , Nevo Pigmentado/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Neoplasias Cutâneas/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Células Cultivadas , Progressão da Doença , Humanos , Melanoma/tratamento farmacológico , Melanoma/patologia , Nevo Pigmentado/tratamento farmacológico , Nevo Pigmentado/patologia , Fatores de Transcrição SOXC/metabolismo , Transdução de Sinais/efeitos dos fármacos , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/patologia , Fatores de Tempo
4.
BMC Med Genomics ; 9: 10, 2016 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-26927636

RESUMO

BACKGROUND: Melanoma is a cancer with rising incidence and new therapeutics are needed. For this, it is necessary to understand the molecular mechanisms of melanoma development and progression. Melanoma differs from other cancers by its ability to produce the pigment melanin via melanogenesis; this biosynthesis is essentially regulated by microphthalmia-associated transcription factor (MITF). MITF regulates various processes such as cell cycling and differentiation. MITF shows an ambivalent role, since high levels inhibit cell proliferation and low levels promote invasion. Hence, well-balanced MITF homeostasis is important for the progression and spread of melanoma. Therefore, it is difficult to use MITF itself for targeted therapy, but elucidating its complex regulation may lead to a promising melanoma-cell specific therapy. METHOD: We systematically analyzed the regulation of MITF with a novel established transcription factor based gene regulatory network model. Starting from comparative transcriptomics analysis using data from cells originating from nine different tumors and a melanoma cell dataset, we predicted the transcriptional regulators of MITF employing ChIP binding information from a comprehensive set of databases. The most striking regulators were experimentally validated by functional assays and an MITF-promoter reporter assay. Finally, we analyzed the impact of the expression of the identified regulators on clinically relevant parameters of melanoma, i.e. the thickness of primary tumors and patient overall survival. RESULTS: Our model predictions identified SOX10 and SOX5 as regulators of MITF. We experimentally confirmed the role of the already well-known regulator SOX10. Additionally, we found that SOX5 knockdown led to MITF up-regulation in melanoma cells, while double knockdown with SOX10 showed a rescue effect; both effects were validated by reporter assays. Regarding clinical samples, SOX5 expression was distinctively up-regulated in metastatic compared to primary melanoma. In contrast, survival analysis of melanoma patients with predominantly metastatic disease revealed that low SOX5 levels were associated with a poor prognosis. CONCLUSION: MITF regulation by SOX5 has been shown only in murine cells, but not yet in human melanoma cells. SOX5 has a strong inhibitory effect on MITF expression and seems to have a decisive clinical impact on melanoma during tumor progression.


Assuntos
Regulação Neoplásica da Expressão Gênica , Melanoma/genética , Melanoma/patologia , Fator de Transcrição Associado à Microftalmia/genética , Fatores de Transcrição SOXD/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Simulação por Computador , Fluorescência , Técnicas de Silenciamento de Genes , Proteínas de Fluorescência Verde/metabolismo , Humanos , Fator de Transcrição Associado à Microftalmia/metabolismo , Invasividade Neoplásica , Fenótipo , Programação Linear , RNA Interferente Pequeno/metabolismo , Reprodutibilidade dos Testes , Fatores de Transcrição SOXE/metabolismo , Análise de Sobrevida , Transfecção
5.
Stem Cells ; 34(4): 832-46, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26753613

RESUMO

The combination of cancer-focused studies and research related to nuclear reprogramming has gained increasing importance since both processes-reprogramming towards pluripotency and malignant transformation-share essential features. Studies have revealed that incomplete reprogramming of somatic cells leads to malignant transformation indicating that epigenetic regulation associated with iPSC generation can drive cancer development [J Mol Cell Biol 2011;341-350; Cell 2012;151:1617-1632; Cell 2014;156:663-677]. However, so far it is unclear whether incomplete reprogramming also affects cancer cells and their function. In the context of melanoma, dedifferentiation correlates to therapy resistance in mouse studies and has been documented in melanoma patients [Nature 2012;490:412-416; Clin Cancer Res 2014;20:2498-2499]. Therefore, we sought to investigate directed dedifferentiation using incomplete reprogramming of melanoma cells. Using a murine model we investigated the effects of partial reprogramming on the cellular plasticity of melanoma cells. We demonstrate for the first time that induced partial reprogramming results in a reversible phenotype switch in melanoma cells. Partially reprogrammed cells at day 12 after transgene induction display elevated invasive potential in vitro and increased lung colonization in vivo. Additionally, using global gene expression analysis of partially reprogrammed cells, we identified SNAI3 as a novel invasion-related marker in human melanoma. SNAI3 expression correlates with tumor thickness in primary melanomas and thus, may be of prognostic value. In summary, we show that investigating intermediate states during the process of reprogramming melanoma cells can reveal novel insights into the pathogenesis of melanoma progression. We propose that deeper analysis of partially reprogrammed melanoma cells may contribute to identification of yet unknown signaling pathways that can drive melanoma progression.


Assuntos
Reprogramação Celular/genética , Células-Tronco Pluripotentes Induzidas/patologia , Melanoma/genética , Neoplasias Experimentais/genética , Fatores de Transcrição da Família Snail/biossíntese , Animais , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Desdiferenciação Celular/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Melanoma/patologia , Camundongos , Camundongos Transgênicos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Proteínas de Neoplasias/biossíntese , Neoplasias Experimentais/patologia , Transdução de Sinais , Fatores de Transcrição da Família Snail/genética
6.
Pigment Cell Melanoma Res ; 28(4): 407-16, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25824590

RESUMO

Neurofibromatosis type 1 (NF1) is a frequent genetic disease leading to the development of Schwann cell-derived neurofibromas or melanocytic lesions called café-au-lait macules (CALMs). The molecular mechanisms involved in CALMs formation remain largely unknown. In this report, we show for the first time pathophysiological mechanisms of abnormal melanocyte differentiation in a human NF1(+/-) -induced pluripotent stem cell (iPSC)-based model. We demonstrate that NF1 patient-derived fibroblasts can be successfully reprogrammed in NF1(+/-) iPSCs with active RAS signaling and that NF1 loss induces senescence during melanocyte differentiation as well as in patient's-derived CALMs, revealing a new role for NF1 in the melanocyte lineage.


Assuntos
Diferenciação Celular , Senescência Celular , Células-Tronco Pluripotentes Induzidas/patologia , Melanócitos/metabolismo , Melanócitos/patologia , Neurofibromina 1/deficiência , Adulto , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Melanócitos/ultraestrutura , Modelos Biológicos , Mutação/genética , Neurofibromina 1/metabolismo , Transdução de Sinais , Proteínas ras/metabolismo
8.
Biotechnol J ; 7(6): 810-21, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22589234

RESUMO

The discovery that overexpression of the transcription factors Oct4, Sox2, Klf4 and c-Myc reprograms differentiated cells into "induced pluripotent stem cells" (iPSCs) has extended our understanding of mechanisms required to maintain stem cell pluripotency and to drive differentiation. Subsequently, additional factors have been discovered that are able to induce a pluripotent state. Recently several groups have succeeded in reprogramming cancer cells to iPSC-like induced pluripotent cancer cells by use of the method established for the generation of iPSCs. This discovery highlighted several striking similarities between pluripotent stem cells and cancer cells, in turn implying that tumorigenesis and reprogramming are partly promoted by overlapping mechanisms. Thus, research on reprogramming might help unravel the mechanisms of carcinogenesis, and vice versa. This review gives an overview of the common features of pluripotent stem cells and cancer cells and summarizes the present state of knowledge in the field of cancer cell reprogramming.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Neoplasias/metabolismo , Diferenciação Celular/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Proteína Homeobox Nanog , Neoplasias/genética , Neoplasias/patologia , Fator 3 de Transcrição de Octâmero/genética , Pseudogenes , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...