Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Dermatol Sci ; 86(2): 162-169, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28153538

RESUMO

BACKGROUND: It is likely that skin is exposed to low concentrations of pollutants such as Polycyclic Aromatic Hydrocarbons (PAH) either through topical penetration by ultrafine particles or by systemic distribution. No precise estimation of pollutants in living skin is available, but literature has reported contamination of blood by PAH at concentrations in the nanomolar range. Some pollutants (PAH for example) are photo-reactive and phototoxic: sunlight and pollution might thus synergistically compromise skin health. OBJECTIVE: Here, the biological effects of particulate matter, PM extract and various PAH were compared in normal human epidermal keratinocytes (NHEK) and reconstructed skin model exposed to either daily UV (d-UV 300-400nm) or UVA1 (350-400nm). Impact of pollutants (PM, PAH or PM extract) combined to UV was studied on NHEK by measuring toxicity, redox homeostasis and GSH metabolism in NHEK. METHODS: NHEK were exposed to UV from solar simulator (either d-UV or UVA1) combined with pollutants. Viability, clonogenic efficiency, redox homeostasis and GSH metabolism were assessed. RESULTS: Pollutants (PAH, PM or PM extract) ±UVA1 irradiation was associated with a significant phototoxic effect that was equal to or greater than that produced by d-UV. This result is interesting considering that UVA1 represents around 80% of daily UV and reaches the dermal-epidermal junction with ease. Moreover, among PAH studied, benzo[a]pyrene and indeno[1,2,3-cd]pyrene were phototoxic at very low concentrations (nanomolar range) on cultured cells or in reconstructed epidermis and also impaired keratinocyte clonogenic potential at sub-toxic doses. ROS generation within cells and in the inner mitochondrial compartment, mitochondrial membrane depolarization and/or reduced ATP production were also noted. Meanwhile, intracellular glutathione concentrations transiently decreased several hours post-treatment and reduction of its synthesis by buthionine sulfoximine potentiated PAH phototoxicity. Consequently, expression of GSH neo-synthesis genes such as SLC7A11 or GCLc was upregulated several hours post-treatment. CONCLUSION: These results obtained using PAH concentrations in the range of those reported in blood of pollution-exposed people suggest that exposure to such a photo-pollution stress, particularly if chronic, may impair cutaneous homeostasis and aggravate sunlight-induced skin damage.


Assuntos
Poluentes Atmosféricos/toxicidade , Material Particulado/toxicidade , Hidrocarbonetos Policíclicos Aromáticos/toxicidade , Pele/efeitos dos fármacos , Pele/efeitos da radiação , Raios Ultravioleta/efeitos adversos , Linhagem Celular , Sobrevivência Celular , Epiderme/metabolismo , Fibroblastos/metabolismo , Glutationa/metabolismo , Homeostase , Humanos , Queratinócitos/citologia , Queratinócitos/efeitos da radiação , Luz , Potencial da Membrana Mitocondrial , Oxirredução , Fotoquímica , Pirenos/toxicidade , Pele/metabolismo , Luz Solar
2.
Pigment Cell Melanoma Res ; 28(4): 464-75, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25950383

RESUMO

Exosomes are important mediators in cell-to-cell communication and, recently, their role in melanoma progression has been brought to light. Here, we characterized exosomes secreted by seven melanoma cell lines with varying degrees of aggressivity. Extensive proteomic analysis of their exosomes confirmed the presence of characteristic exosomal markers as well as melanoma-specific antigens and oncogenic proteins. Importantly, the protein composition differed among exosomes from different lines. Exosomes from aggressive cells contained specific proteins involved in cell motility, angiogenesis, and immune response, while these proteins were less abundant or absent in exosomes from less aggressive cells. Interestingly, when exposed to exosomes from metastatic lines, less aggressive cells increased their migratory capacities, likely due to transfer of pro-migratory exosomal proteins to recipient cells. Hence, this study shows that the specific protein composition of melanoma exosomes depends on the cells' aggressivity and suggests that exosomes influence the behavior of other tumor cells and their microenvironment.


Assuntos
Exossomos/metabolismo , Melanoma/metabolismo , Proteoma/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Humanos , Espectrometria de Massas , Metástase Neoplásica , Proteínas de Neoplasias/metabolismo , Proteômica
3.
J Invest Dermatol ; 134(6): 1512-1518, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24573173

RESUMO

Epidermal melanocytes are particularly vulnerable to oxidative stress owing to the pro-oxidant state generated during melanin synthesis, and to the intrinsic antioxidant defenses that are compromised in pathologic conditions. Melanoma is thought to be oxidative stress driven, and melanocyte death in vitiligo is thought to be instigated by a highly pro-oxidant state in the epidermis. We review the current knowledge about melanin and the redox state of melanocytes, how paracrine factors help counteract oxidative stress, the role of oxidative stress in melanoma initiation and progression and in melanocyte death in vitiligo, and how this knowledge can be harnessed for melanoma and vitiligo treatment.


Assuntos
Epiderme/metabolismo , Regulação Neoplásica da Expressão Gênica , Melanócitos/citologia , Estresse Oxidativo , Animais , Antioxidantes/metabolismo , Humanos , Melaninas/química , Melanoma/metabolismo , Camundongos , Oxidantes/química , Oxirredução , Pigmentação , Espécies Reativas de Oxigênio , Vitiligo/metabolismo
4.
Mol Cell Biol ; 32(7): 1237-47, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22290434

RESUMO

MITF-M and PAX3 are proteins central to the establishment and transformation of the melanocyte lineage. They control various cellular mechanisms, including migration and proliferation. BRN2 is a POU domain transcription factor expressed in melanoma cell lines and is involved in proliferation and invasion, at least in part by regulating the expression of MITF-M and PAX3. The T361 and S362 residues of BRN2, both in the POU domain, are conserved throughout the POU protein family and are targets for phosphorylation, but their roles in vivo remain unknown. To examine the role of this phosphorylation, we generated mutant BRN2 in which these two residues were replaced with alanines (BRN2TS→BRN2AA). When expressed in melanocytes in vitro or in the melanocyte lineage in transgenic mice, BRN2TS induced proliferation and repressed migration, whereas BRN2AA repressed both proliferation and migration. BRN2TS and BRN2AA bound and repressed the MITF-M promoter, whereas PAX3 transcription was induced by BRN2TS but repressed by BRN2AA. Expression of the BRN2AA transgene in a Mitf heterozygous background and in a Pax3 mutant background enhanced the coat color phenotype. Our findings show that melanocyte migration and proliferation are controlled both through the regulation of PAX3 by nonphosphorylated BRN2 and through the regulation of MITF-M by the overall BRN2 level.


Assuntos
Proliferação de Células , Melanócitos/citologia , Proteínas do Tecido Nervoso/metabolismo , Fatores do Domínio POU/metabolismo , Fatores de Transcrição Box Pareados/genética , Animais , Linhagem Celular Tumoral , Movimento Celular , Humanos , Melanócitos/metabolismo , Melanoma/genética , Melanoma/metabolismo , Camundongos , Camundongos Transgênicos , Fator de Transcrição Associado à Microftalmia/genética , Mutação , Proteínas do Tecido Nervoso/genética , Fator de Transcrição PAX3 , Fatores do Domínio POU/genética , Fenótipo , Fosforilação , Regiões Promotoras Genéticas , Transcrição Gênica
5.
Development ; 138(18): 3943-54, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21862558

RESUMO

We aim to evaluate environmental and genetic effects on the expansion/proliferation of committed single cells during embryonic development, using melanoblasts as a paradigm to model this phenomenon. Melanoblasts are a specific type of cell that display extensive cellular proliferation during development. However, the events controlling melanoblast expansion are still poorly understood due to insufficient knowledge concerning their number and distribution in the various skin compartments. We show that melanoblast expansion is tightly controlled both spatially and temporally, with little variation between embryos. We established a mathematical model reflecting the main cellular mechanisms involved in melanoblast expansion, including proliferation and migration from the dermis to epidermis. In association with biological information, the model allows the calculation of doubling times for melanoblasts, revealing that dermal and epidermal melanoblasts have short but different doubling times. Moreover, the number of trunk founder melanoblasts at E8.5 was estimated to be 16, a population impossible to count by classical biological approaches. We also assessed the importance of the genetic background by studying gain- and loss-of-function ß-catenin mutants in the melanocyte lineage. We found that any alteration of ß-catenin activity, whether positive or negative, reduced both dermal and epidermal melanoblast proliferation. Finally, we determined that the pool of dermal melanoblasts remains constant in wild-type and mutant embryos during development, implying that specific control mechanisms associated with cell division ensure half of the cells at each cell division to migrate from the dermis to the epidermis. Modeling melanoblast expansion revealed novel links between cell division, cell localization within the embryo and appropriate feedback control through ß-catenin.


Assuntos
Diferenciação Celular , Crescimento e Desenvolvimento/fisiologia , Melanócitos/fisiologia , Modelos Biológicos , Modelos Teóricos , Animais , Animais Recém-Nascidos , Proliferação de Células , Células Cultivadas , Derme/citologia , Derme/embriologia , Embrião de Mamíferos , Células Epidérmicas , Epiderme/embriologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
6.
Pigment Cell Melanoma Res ; 24(4): 631-42, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21599871

RESUMO

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.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Fator 1 de Ligação ao Facilitador Linfoide/genética , Melanoma/genética , Melanoma/patologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Fatores de Transcrição/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Humanos , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Modelos Biológicos , Invasividade Neoplásica , Fenótipo , Fator de Transcrição 4 , Fatores de Transcrição/metabolismo , Proteínas Wnt/farmacologia , beta Catenina/metabolismo
7.
Cancer Res ; 68(19): 7788-94, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18829533

RESUMO

The origin of tumor heterogeneity is poorly understood, yet it represents a major barrier to effective therapy. In melanoma and in melanocyte development, the microphthalmia-associated transcription factor (Mitf) controls survival, differentiation, proliferation, and migration/metastasis. The Brn-2 (N-Oct-3, POU3F2) transcription factor also regulates melanoma proliferation and is up-regulated by BRAF and beta-catenin, two key melanoma-associated signaling molecules. Here, we show that Brn-2 also regulates invasiveness and directly represses Mitf expression. Remarkably, in melanoma biopsies, Mitf and Brn-2 each mark a distinct subpopulation of melanoma cells, providing a striking illustration of melanoma tumor heterogeneity with implications for melanoma therapy.


Assuntos
Biomarcadores Tumorais/genética , Proteínas de Homeodomínio/fisiologia , Melanoma/genética , Fator de Transcrição Associado à Microftalmia/genética , Fatores do Domínio POU/fisiologia , Animais , Sequência de Bases , Biomarcadores Tumorais/metabolismo , Regulação para Baixo , Feminino , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fator de Transcrição Associado à Microftalmia/metabolismo , Dados de Sequência Molecular , Invasividade Neoplásica , Fatores do Domínio POU/genética , Fatores do Domínio POU/metabolismo , Ligação Proteica , Transplante Heterólogo , Células Tumorais Cultivadas
8.
Genes Dev ; 21(22): 2923-35, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18006687

RESUMO

Tumor progression is a multistep process in which proproliferation mutations must be accompanied by suppression of senescence. In melanoma, proproliferative signals are provided by activating mutations in NRAS and BRAF, whereas senescence is bypassed by inactivation of the p16(Ink4a) gene. Melanomas also frequently exhibit constitutive activation of the Wnt/beta-catenin pathway that is presumed to induce proliferation, as it does in carcinomas. We show here that, contrary to expectations, stabilized beta-catenin reduces the number of melanoblasts in vivo and immortalizes primary skin melanocytes by silencing the p16(Ink4a) promoter. Significantly, in a novel mouse model for melanoma, stabilized beta-catenin bypasses the requirement for p16(Ink4a) mutations and, together with an activated N-Ras oncogene, leads to melanoma with high penetrance and short latency. The results reveal that synergy between the Wnt and mitogen-activated protein (MAP) kinase pathways may represent an important mechanism underpinning the genesis of melanoma, a highly aggressive and increasingly common disease.


Assuntos
Transformação Celular Neoplásica/efeitos dos fármacos , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Genes ras , Melanócitos/efeitos dos fármacos , Melanoma/genética , beta Catenina/farmacologia , Animais , Linhagem Celular Transformada , Células Cultivadas , Imunoprecipitação da Cromatina , Cruzamentos Genéticos , Ensaio de Desvio de Mobilidade Eletroforética , Inativação Gênica , Humanos , Luciferases/metabolismo , Melanócitos/citologia , Melanócitos/metabolismo , Melanoma/metabolismo , Camundongos , Camundongos Transgênicos , Transfecção , beta-Galactosidase/metabolismo
9.
Bull Cancer ; 94(1): 81-92, 2007 Jan.
Artigo em Francês | MEDLINE | ID: mdl-17237008

RESUMO

Mitf protein is a transcription factor involved all along the life of pigmented cells. This protein is located in the center of multiple signaling pathways which control differentiation, morphology, proliferation and survival of the various cells of the melanocyte lineage: melanoblasts, melanocytes and melanoma. Mitf plays a major role in melanoblasts differentiation, by inducing the key enzyme of melanogenesis, tyrosinase, and its secondary enzymes, Tyrp1 and Dct. Mitf regulates morphology and migration of melanocytes, particularly by regulating cytoskeleton organization and cell-cell adhesion. Mitf plays a double role of inducer/repressor of cellular proliferation. This protein inhibits cell cycle progression and prevents non-proper cell division. In few cases, Mitf can also induce cell cycle. A minimal quantity/activity of Mitf is necessary for melanoblast survival. Essential protein of the melanocyte lineage, Mitf was proposed as diagnostic/pronostic marker for cutaneous melanoma. However, could we then consider MITF as the unique marker of such a cancer?


Assuntos
Transformação Celular Neoplásica/patologia , Melanócitos/citologia , Melanoma/patologia , Fator de Transcrição Associado à Microftalmia/fisiologia , Neoplasias Cutâneas/patologia , Animais , Diferenciação Celular/fisiologia , Sobrevivência Celular/fisiologia , Humanos , Melanoma/metabolismo , Camundongos , Fator de Transcrição Associado à Microftalmia/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fosforilação , Transdução de Sinais/fisiologia , Neoplasias Cutâneas/metabolismo , Fatores de Transcrição/fisiologia
10.
Genes Dev ; 20(24): 3426-39, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17182868

RESUMO

It is widely held that cells with metastatic properties such as invasiveness and expression of matrix metalloproteinases arise through the stepwise accumulation of genetic lesions arising from genetic instability and "clonal evolution." By contrast, we show here that in melanomas invasiveness can be regulated epigenetically by the microphthalmia-associated transcription factor, Mitf, via regulation of the DIAPH1 gene encoding the diaphanous-related formin Dia1 that promotes actin polymerization and coordinates the actin cytoskeleton and microtubule networks at the cell periphery. Low Mitf levels lead to down-regulation of Dia1, reorganization of the actin cytoskeleton, and increased ROCK-dependent invasiveness, whereas increased Mitf expression leads to decreased invasiveness. Significantly the regulation of Dia1 by Mitf also controls p27(Kip1)-degradation such that reduced Mitf levels lead to a p27(Kip1)-dependent G1 arrest. Thus Mitf, via regulation of Dia1, can both inhibit invasiveness and promote proliferation. The results imply variations in the repertoire of environmental cues that determine Mitf activity will dictate the differentiation, proliferative, and invasive/migratory potential of melanoma cells through a dynamic epigenetic mechanism.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Melanoma/metabolismo , Fator de Transcrição Associado à Microftalmia/metabolismo , Invasividade Neoplásica , Neoplasias Cutâneas/metabolismo , Actinas/metabolismo , Animais , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Feminino , Forminas , Fase G1 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Melanoma/genética , Melanoma/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fator de Transcrição Associado à Microftalmia/genética , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/metabolismo , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Transfecção , Quinases Associadas a rho
11.
Nature ; 433(7027): 764-9, 2005 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-15716956

RESUMO

The controls that enable melanoblasts and melanoma cells to proliferate are likely to be related, but so far no key regulator of cell cycle progression specific to the melanocyte lineage has been identified. The microphthalmia-associated transcription factor Mitf has a crucial but poorly defined role in melanoblast and melanocyte survival and in differentiation. Here we show that Mitf can act as a novel anti-proliferative transcription factor able to induce a G1 cell-cycle arrest that is dependent on Mitf-mediated activation of the p21(Cip1) (CDKN1A) cyclin-dependent kinase inhibitor gene. Moreover, cooperation between Mitf and the retinoblastoma protein Rb1 potentiates the ability of Mitf to activate transcription. The results indicate that Mitf-mediated activation of p21Cip1 expression and consequent hypophosphorylation of Rb1 will contribute to cell cycle exit and activation of the differentiation programme. The mutation of genes associated with melanoma, such as INK4a or BRAF that would affect either Mitf cooperation with Rb1 or Mitf stability respectively, would impair Mitf-mediated cell cycle control.


Assuntos
Proteínas de Ciclo Celular/genética , Ciclo Celular , Proteínas de Ligação a DNA/metabolismo , Proteína do Retinoblastoma/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional , Animais , Sequência de Bases , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Imunoprecipitação da Cromatina , Inibidor de Quinase Dependente de Ciclina p21 , Proteínas de Ligação a DNA/genética , Humanos , Melanócitos/citologia , Melanócitos/metabolismo , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Fator de Transcrição Associado à Microftalmia , Células NIH 3T3 , Fosforilação , Ligação Proteica , Proteína do Retinoblastoma/genética , Fatores de Transcrição/genética
12.
Oncogene ; 23(40): 6726-35, 2004 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-15273735

RESUMO

We have determined the expression profiles of cdh7, and the related cdh20 during development. Both transcripts are found in the adult brain, but only cadherin-20 mRNA was detected during embryogenesis. In mouse embryos, cadherin-20 is synthesized by the forebrain, anterior neural ridge, developing visual system, primitive external granular layer of the cerebellum and a subset of neural crest cells likely to develop into melanoblasts. We found that the other embryonic tissues in which cadherin-20 was synthesized depended on genetic background. Melanoma cell lines contained transcripts for cadherin-7 but not for cadherin-20. The majority of the malignant melanoma cell lines produced N-cadherin (N-Cad) and/or cadherin-7 whereas melanocyte cell lines did not. The converse was observed for E-cadherin (E-Cad). Our data suggest that during development cadherin-20 is a key player in compartmentalization of the neural tube and establishment of neural circuitry. Finally, during oncogenesis, cadherin-7, N-cad and E-cad could be used as an efficient marker set for melanoma.


Assuntos
Caderinas/genética , Transformação Celular Neoplásica/genética , Desenvolvimento Embrionário e Fetal/genética , Melanócitos/fisiologia , Melanoma/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Encéfalo/embriologia , Caderinas/química , Linhagem Celular Tumoral , Clonagem Molecular , Sequência Conservada , Desenvolvimento Embrionário e Fetal/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Hibridização In Situ , Camundongos , Dados de Sequência Molecular , Especificidade de Órgãos , RNA Mensageiro/genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...