Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Biol Chem ; 285(1): 242-54, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19880519

RESUMEN

MAP2 is a neuron-specific microtubule-associated protein that binds and stabilizes dendritic microtubules. Previously, we showed that MAP2 expression is (a) activated in cutaneous primary melanoma and (b) inversely associated with melanoma tumor progression. We also showed that ectopic expression of MAP2 in metastatic melanoma cells inhibits cell growth by inducing mitotic spindle defects and apoptosis. However, molecular mechanisms of regulation of MAP2 gene expression in melanoma are not understood. Here, we show that in melanoma cells MAP2 expression is induced by the demethylating agent 5-aza-2'-cytidine, and MAP2 promoter is progressively methylated during melanoma progression, indicating that epigenetic mechanisms are involved in silencing of MAP2 in melanoma. In support of this, methylation of MAP2 promoter DNA in vitro inhibits its activity. Because MAP2 promoter activity levels in melanoma cell lines also correlated with activating mutation in BRAF, a gene that is highly expressed in neurons, we hypothesized that BRAF signaling is involved in MAP2 expression. We show that hyperactivation of BRAF-MEK signaling activates MAP2 expression in melanoma cells by two independent mechanisms, promoter demethylation or down-regulation of neuronal transcription repressor HES1. Our data suggest that BRAF oncogene levels can regulate melanoma neuronal differentiation and tumor progression.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Metilación de ADN/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/genética , Melanoma/genética , Proteínas Asociadas a Microtúbulos/genética , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas B-raf/genética , Sustitución de Aminoácidos/efectos de los fármacos , Sustitución de Aminoácidos/genética , Animales , Azacitidina/farmacología , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Islas de CpG/genética , Metilación de ADN/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Melanoma/enzimología , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Mutantes/metabolismo , Neuronas/citología , Elementos de Respuesta/genética , Factor de Transcripción HES-1 , Transcripción Genética/efectos de los fármacos
2.
Arch Biochem Biophys ; 508(2): 227-33, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21291857

RESUMEN

By virtue of the presence of multiple protein-protein interaction and signaling domains, PDZ proteins play important roles in assembling protein complexes that participate in diverse cell biological processes. GIPC is a versatile PDZ protein that binds a variety of target proteins in different cell types. In previous studies we showed that, in epidermal melanocytes, GIPC interacts with newly synthesized melanosomal protein TRP1 in the Golgi region and proposed that this interaction may facilitate intracellular trafficking of TRP1. However, since GIPC contains a single PDZ domain and no other known protein interaction motifs, it is not known how GIPC-TRP1 interaction affects melanosome biogenesis and/or melanin pigmentation. Here, we show that in human primary melanocytes GIPC interacts with AKT-binding protein APPL (adaptor protein containing pleckstrin homology, leucine zipper and phosphotyrosine binding domains), which readily co-precipitates with newly synthesized TRP1. Knockdown of either GIPC or APPL inhibits melanogenesis by decreasing tyrosinase protein levels and enzyme activity. In melanocytes, APPL exists in a complex with GIPC and phospho-AKT. Inhibition of AKT phosphorylation using a PI3-kinase inhibitor abolishes this interaction and results in retardation TRP1 in the Golgi. These data suggest that interactions between TRP1-GIPC and GIPC-APPL-AKT provide a potential link between melanogenesis and PI3 kinase signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Melaninas/biosíntesis , Melanosomas/metabolismo , Tripsina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Humanos , Melanosomas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica , Transporte de Proteínas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/genética
3.
Am J Physiol Cell Physiol ; 297(3): C679-87, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19587221

RESUMEN

Transient receptor potential melastatin (TRPM) is a subfamily of ion channels that are involved in sensing taste, ambient temperature, low pH, osmolarity, and chemical ligands. Melastatin 1/TRPM1, the founding member, was originally identified as melanoma metastasis suppressor based on its expression in normal pigment cells in the skin and the eye but not in aggressive, metastasis-competent melanomas. The role of TRPM1 and its regulation in normal melanocytes and in melanoma progression is not understood. Here, we studied the relationship of TRPM1 expression to growth and differentiation of human epidermal melanocytes. TRPM1 expression and intracellular Ca(2+) levels are significantly lower in rapidly proliferating melanocytes compared to the slow growing, differentiated melanocytes. We show that lentiviral short hairpin RNA (shRNA)-mediated knockdown of TRPM1 results in reduced intracellular Ca(2+) and decreased Ca(2+) uptake suggesting a role for TRPM1 in Ca(2+) homeostasis in melanocytes. TRPM1 knockdown also resulted in a decrease in tyrosinase activity and intracellular melanin pigment. Expression of the tumor suppressor p53 by transfection or induction of endogenous p53 by ultraviolet B radiation caused repression of TRPM1 expression accompanied by decrease in mobilization of intracellular Ca(2+) and uptake of extracellular Ca(2+). These data suggest a role for TRPM1-mediated Ca(2+) homeostasis, which is also regulated by ultraviolet B, in melanogenesis.


Asunto(s)
Calcio/metabolismo , Homeostasis/fisiología , Melanocitos/metabolismo , Canales Catiónicos TRPM/metabolismo , Rayos Ultravioleta , Células Cultivadas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/fisiología , Regulación de la Expresión Génica/efectos de la radiación , Silenciador del Gen , Humanos , Melaninas/biosíntesis , Melanocitos/efectos de la radiación
4.
Biochem Biophys Res Commun ; 388(3): 576-82, 2009 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-19683515

RESUMEN

Recently we showed Lupeol, a triterpene, found in fruits and vegetables inhibits the growth of tumors originated from human androgen-sensitive prostate cancer (CaP) cells and decreases the serum-PSA levels in a mouse model. Here, we provide evidence that Lupeol inhibits the growth of androgen-sensitive as well as androgen-insensitive CaP cells by inducing G2/M cell cycle arrest without exhibiting any toxicity to normal human prostate epithelial cells (PrEC) at the doses at which it kills cancer cells. We observed that Lupeol treatment to LNCaP and DU145 cells resulted in a dose-dependent (i) decrease in the protein levels of Cyclins-A, -B1, -D1, -D2, -E2, cyclin-dependent kinase (cdk)-2 and (ii) increase in the protein level of CDK-inhibitor p21. Since G2/M cell cycle phase is regulated by microtubule assembly, we investigated effect of Lupeol on microtubule assembly, its regulation and down-stream targets in CaP cells. Lupeol treatment significantly modulated the level of (i) microtubule components alpha-tubulin and beta-tubulin, (ii) microtubule-regulatory protein stathmin, and (iii) microtubule-regulatory down-stream target/pro-survival protein survivin. Lupeol treatment also decreased the level of anti-apoptotic protein cFLIP. Finally, Lupeol was observed to significantly decrease the transcriptional activation of survivin and cFLIP genes in CaP cells. We conclude that the Lupeol-induced growth inhibition of CaP cells is a net outcome of simultaneous effects on stathmin, cFLIP, and survivin which results in the disruption of microtubule assembly. We suggest that Lupeol alone or as an adjuvant to other microtubule agents could be developed as a potential agent for the treatment of human CaP.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/antagonistas & inhibidores , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Microtúbulos/efectos de los fármacos , Neoplasias de la Próstata/metabolismo , Triterpenos/farmacología , Moduladores de Tubulina/farmacología , Animales , Apoptosis/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/biosíntesis , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Dieta , Humanos , Proteínas Inhibidoras de la Apoptosis , Masculino , Ratones , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Triterpenos Pentacíclicos , Survivin , Activación Transcripcional
5.
Clin Cancer Res ; 14(7): 2119-27, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18381953

RESUMEN

PURPOSE: Poor prognosis of metastatic melanoma mandates the development of novel strategies for its treatment and prevention. In this study, the effect of lupeol, a diet-based triterpene, was determined on the growth and tumorigenicity of human melanoma cells in vitro and in vivo. EXPERIMENTAL DESIGN: Normal human melanocytes, and human metastatic (451Lu) and nonmetastatic (WM35) cells were treated with lupeol; its effect on growth, proliferation, and apoptosis were evaluated. Further athymic nude mice bearing 451Lu cell-originated tumors were administered with lupeol thrice a week, and its effect on tumor growth and surrogate biomarkers was evaluated. RESULTS: Lupeol significantly decreased the viability of 451Lu and WM35 melanoma cells but had only a marginal effect on normal human melanocyte cells at similar doses. Lupeol treatment of 451Lu cells caused (a) G(1)-S phase cell cycle arrest and apoptosis; (b) down-regulation of Bcl2 and up-regulation of Bax; (c) activation of caspase-3 and induction of poly(ADP)ribose polymerase cleavage; (d) decreased expression of cyclin D1, cyclin D2, and cdk2; and (e) increased expression of p21 protein. Next, lupeol significantly reduced 451Lu tumor growth in athymic nude mice and modulated the expression of proliferation markers, apoptotic markers, and cell cycle regulatory molecules in tumor xenografts. CONCLUSION: Our findings showed the anticancer efficacy of lupeol with mechanistic rationale against metastatic human melanoma cells. We suggest that lupeol, alone or as an adjuvant to current therapies, could be useful for the management of human melanoma.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Melanoma Experimental/tratamiento farmacológico , Triterpenos/farmacología , Animales , Western Blotting , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citometría de Flujo , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Triterpenos Pentacíclicos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Photochem Photobiol ; 84(2): 528-36, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18248502

RESUMEN

Malignant melanoma arises from epidermal melanocytes, the cells responsible for the production of the skin pigment melanin. The photoprotective role of melanin, which is transferred to neighboring keratinocytes, in UV-induced skin carcinogenesis, specifically in nonmelanoma skin cancers, has been well documented. Although melanocyte-resident melanin is expected to offer similar protection to melanocytes from UV-induced damage, UV radiation has long been suspected to have an etiologic role in cutaneous melanoma. However, nearly three decades of efforts using a variety of in vitro and in vivo models of human skin and mouse genetic models have produced conflicting data. Epidemiologic studies have also failed to establish a definitive association between UV exposure and risk of melanoma. In this review, we evaluate the dual role of the melanin pigment as a photoprotector as well as a photosensitizer and examine the evidence for association between melanin levels (constitutive and induced) and melanoma risk. We also discuss possible reasons for the lack of signature UV mutations in melanoma oncogenes known to date and potential alternative mechanisms to explain the role of UV in melanomagenesis.


Asunto(s)
Melanoma/etiología , Neoplasias Inducidas por Radiación/etiología , Neoplasias Cutáneas/etiología , Rayos Ultravioleta
7.
Nucleic Acids Res ; 34(13): 3819-32, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16916793

RESUMEN

Microtubule-associated protein 2 (MAP2), a neuron-specific protein, stabilizes microtubules and is critical for neurite outgrowth and dendrite development. Although MAP2 is widely used as a marker of neuronal differentiation, regulation of its transcription has not been investigated. We showed that MAP2 is frequently activated in human cutaneous melanoma. Here, we identified a 2.2 kb region that is sufficient for neuronal-specific expression in vitro and in vivo. Comparative analysis of the mouse, rat and human MAP2 promoter sequences showed the presence of a conserved bHLH factor binding sites. Electrophoretic mobility shift analysis, promoter mutagenesis and co-transfection experiments showed that NeuroD, a pro-neuronal differentiation factor, and Hairy and Enhancer of Split (HES1), a transcription repressor, are involved in the regulation of MAP2 promoter activity. Melanoma cells express both NeuroD and HES1. Chromatin immunoprecipitation showed that in metastatic melanoma cells N-box region of the MAP2 promoter is occupied by endogenous HES1. We show that the inhibition of Notch signaling, a regulator of HES1 gene expression, and/or shRNA knockdown of HES1 results in the upregulation of MAP2 promoter activity. Thus, our data suggest that Notch signaling, which is implicated in melanoma progression, and HES1 play a role in MAP2 gene regulation during melanoma progression.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación Neoplásica de la Expresión Génica , Melanoma/genética , Proteínas Asociadas a Microtúbulos/genética , Receptor Notch1/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular Tumoral , Secuencia Conservada , Células HeLa , Proteínas de Homeodominio/metabolismo , Humanos , Melanoma/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo , Células PC12 , Regiones Promotoras Genéticas , Ratas , Proteínas Represoras/metabolismo , Transducción de Señal , Factor de Transcripción HES-1 , Activación Transcripcional
8.
Biosens Bioelectron ; 68: 508-515, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25636023

RESUMEN

An electrochemical immunosensing method was developed to detect melanoma cells based on the affinity between cell surface melanocortin 1 receptor (MC1R) antigen and anti-MC1R antibody (MC1R-Ab). The MC1R-Abs were immobilized in amino-functionalized silica nanoparticles (n-SiNPs)-polypyrrole (PPy) nanocomposite modified on working electrode surface of screen-printed electrode (SPE). Cyclic voltammetry was employed, with the help of redox mediator ([Fe(CN)6](3-)), to measure the change in anodic oxidation peak current arising due to the specific interaction between MC1R antigens and MC1R-Abs when the target melanoma cells are present in the sample. Various factors affecting the sensor performance, such as the amount of MC1R-Abs loaded, incubation time with the target melanoma cells, the presence of interfering non-melanoma cells, were tested and optimized over different expected melanoma cell loads in the range of 50-7500 cells/2.5 mL. The immunosensor is highly sensitive (20 cells/mL), specific, and reproducible, and the antibody-loaded electrode in ready-to-use stage is stable over two weeks. Thus, in conjunction with a microfluidic lab-on-a-chip device our electrochemical immunosensing approach may be suitable for highly sensitive, selective, and rapid detection of circulating tumor cells (CTCs) in blood samples.


Asunto(s)
Técnicas Biosensibles , Inmunoensayo/métodos , Melanoma/sangre , Receptor de Melanocortina Tipo 1/aislamiento & purificación , Anticuerpos/inmunología , Anticuerpos Inmovilizados/inmunología , Oro/química , Humanos , Dispositivos Laboratorio en un Chip , Melanoma/patología , Nanopartículas del Metal/química , Técnicas Analíticas Microfluídicas , Células Neoplásicas Circulantes/inmunología , Receptor de Melanocortina Tipo 1/sangre , Receptor de Melanocortina Tipo 1/inmunología
9.
Am J Stem Cells ; 2(1): 52-61, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23671816

RESUMEN

Highly malignant tumors mostly consist of rapidly proliferating cells. However, tumors also contain a few cells in a quiescent state that can be characterized as slow-cycling, expressing markers of stem cells and possessing the ability to initiate new tumors. These quiescent cells, now generally termed 'cancer stem cells' (CSC) (or 'cancer initiating cells'), are capable of regenerating the entire tumor--as it occurs in metastatic spread. This process of tumor initiation by stem-like cells presumably involves differentiation of quiescent CSC into rapidly proliferating tumor cells. An important implication of the presence of slow cycling, quiescent stem-like cells in the tumor and their ability to initiate tumors is that they contribute to the resistance to treatments by conventional chemo- and radiotherapy directed toward killing rapidly dividing cells. However, similar to normal stem cells, the CSC could also potentially transdifferentiate into cell lineages other than the original lineage from which the tumor arose. Therefore, transdifferentiation of CSC offers a possible therapeutic strategy which has not yet been fully exploited. In this article, we provide a comprehensive review of the concepts in tumor cell transdifferentiation and discuss the mechanisms of transdifferentiation with emphasis on their relevance to potential novel treatment strategies.

10.
Pigment Cell Melanoma Res ; 26(3): 348-56, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23452348

RESUMEN

Mutations in TRPM1, a calcium channel expressed in retinal bipolar cells and epidermal melanocytes, cause complete congenital stationary night blindness with no discernible skin phenotype. In the retina, TRPM1 activity is negatively coupled to metabotropic glutamate receptor 6 (mGluR6) signaling through Gαo and TRPM1 mutations result in the loss of responsiveness of TRPM1 to mGluR6 signaling. Here, we show that human melanocytes express mGluR6, and treatment of melanocytes with L-AP4, a type III mGluR-selective agonist, enhances Ca(2+) uptake. Knockdown of TRPM1 or mGluR6 by shRNA abolished L-AP4-induced Ca(2+) influx and TRPM1 currents, showing that TRPM1 activity in melanocytes is positively coupled to mGluR6 signaling. Gαo protein is absent in melanocytes. However, forced expression of Gαo restored negative coupling of TRPM1 to mGluR6 signaling, but treatment with pertussis toxin, an inhibitor of Gi /Go proteins, did not affect basal or mGluR6-induced Ca(2+) uptake. Additionally, chronic stimulation of mGluR6 altered melanocyte morphology and increased melanin content. These data suggest differences in coupling of TRPM1 function to mGluR6 signaling explain different cellular responses to glutamate in the retina and the skin.


Asunto(s)
Melaninas/metabolismo , Melanocitos/metabolismo , Receptores de Glutamato/metabolismo , Canales Catiónicos TRPM/metabolismo , Calcio/metabolismo , Proliferación Celular/efectos de los fármacos , Células Epidérmicas , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Ácido Glutámico/farmacología , Humanos , Recién Nacido , Masculino , Melanocitos/citología , Melanocitos/efectos de los fármacos , Técnicas de Placa-Clamp , Propionatos/farmacología , Receptores de Glutamato/genética , Transducción de Señal/efectos de los fármacos
11.
PLoS One ; 8(10): e78280, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24167615

RESUMEN

Leopard complex spotting is a group of white spotting patterns in horses caused by an incompletely dominant gene (LP) where homozygotes (LP/LP) are also affected with congenital stationary night blindness. Previous studies implicated Transient Receptor Potential Cation Channel, Subfamily M, Member 1 (TRPM1) as the best candidate gene for both CSNB and LP. RNA-Seq data pinpointed a 1378 bp insertion in intron 1 of TRPM1 as the potential cause. This insertion, a long terminal repeat (LTR) of an endogenous retrovirus, was completely associated with LP, testing 511 horses (χ(2)=1022.00, p<<0.0005), and CSNB, testing 43 horses (χ(2)=43, p<<0.0005). The LTR was shown to disrupt TRPM1 transcription by premature poly-adenylation. Furthermore, while deleterious transposable element insertions should be quickly selected against the identification of this insertion in three ancient DNA samples suggests it has been maintained in the horse gene pool for at least 17,000 years. This study represents the first description of an LTR insertion being associated with both a pigmentation phenotype and an eye disorder.


Asunto(s)
Enfermedades de los Caballos/genética , Mutagénesis Insercional , Ceguera Nocturna/genética , Ceguera Nocturna/veterinaria , Retroviridae/genética , Pigmentación de la Piel/genética , Canales Catiónicos TRPM/genética , Animales , Femenino , Caballos , Masculino , Ceguera Nocturna/metabolismo , Retroelementos , Canales Catiónicos TRPM/metabolismo
12.
Photochem Photobiol ; 88(5): 1075-82, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22404235

RESUMEN

The term barrier function as applied to human skin often connotes the physical properties of this organ that provides protection from its surrounding environment. This term does not generally include skin pigmentation. However, skin pigmentation, which is the result of melanin produced in melanocytes residing in the basal layer of the skin and exported to the keratinocytes in the upper layers, serves equally important protective function. Indeed, changes in skin pigmentation are often the most readily recognized indicators of exposure of skin to damaging agents, especially to natural and artificial radiation in the environment. Several recent studies have shed new light on (1) the mechanisms involved in selective effects of subcomponents of UV radiation on human skin pigmentation and (2) the interactive influences between keratinocytes and melanocytes, acting as "epidermal melanin unit," that manifest as changes in skin pigmentation in response to exposure to various forms of radiation. This article provides a concise review of our current understanding of the effects of the nonionizing solar radiation, at cellular and molecular levels, on human skin pigmentation.


Asunto(s)
Queratinocitos/efectos de la radiación , Melaninas/metabolismo , Melanocitos/efectos de la radiación , Melanosomas/efectos de la radiación , Piel/efectos de la radiación , Comunicación Celular/fisiología , Comunicación Celular/efectos de la radiación , ADN/metabolismo , Daño del ADN , Humanos , Queratinocitos/citología , Queratinocitos/metabolismo , Melanocitos/citología , Melanocitos/metabolismo , Melanosomas/fisiología , Piel/metabolismo , Pigmentación de la Piel/fisiología , Luz Solar/efectos adversos , Rayos Ultravioleta/efectos adversos
13.
J Invest Dermatol ; 131(6): 1291-9, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21346776

RESUMEN

The prognosis of advanced melanoma remains poor in spite of treatment advances, emphasizing the importance of additional preventive measures. Flavonoids, natural components of our diet, are being investigated for their chemopreventive/therapeutic properties. Microphthalmia-associated transcription factor (Mitf), downstream of the Wnt/ß-catenin pathway, has become an important prognostic marker of melanoma. In this study, we show that treatment of 451Lu melanoma cells with the dietary flavonoid fisetin (3,7,3',4'-tetrahydroxyflavone) resulted in decreased cell viability with G1-phase arrest and disruption of Wnt/ß-catenin signaling. This was accompanied by a decrease in the expression of Wnt protein and its co-receptors, as well as by a parallel increase in the expression of endogenous Wnt inhibitors. Fisetin-treated cells showed increased cytosolic levels of Axin and ß-TrCP and decreased phosphorylation of glycogen synthase kinase 3ß associated with decreased ß-catenin stabilization. Fisetin-mediated interference with the functional cooperation between ß-catenin and T-cell factor (TCF)-2 resulted in the downregulation of positively regulated TCF targets, such as c-myc, Brn-2, and Mitf. Flow-cytometric analysis of Mitf-overexpressing cells showed that fisetin repressed Mitf-induced cell proliferation. Finally, administration of fisetin to 451Lu-xenografted nude mice resulted in the inhibition of tumor development and decreased Mitf expression. Our data suggest that fisetin can be developed as an effective agent against melanoma because of its potential inhibitory effect on ß-catenin/Mitf signaling.


Asunto(s)
Antineoplásicos/farmacología , Flavonoides/farmacología , Melanoma/tratamiento farmacológico , Factor de Transcripción Asociado a Microftalmía/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Proteínas Wnt/fisiología , beta Catenina/fisiología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Flavonoles , Fase G1/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Factor Nuclear 1-beta del Hepatocito/fisiología , Humanos , Melanoma/patología , Ratones , beta Catenina/análisis , Proteínas con Repetición de beta-Transducina/fisiología
14.
Cancers (Basel) ; 2(2): 989-99, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22545195

RESUMEN

Cutaneous malignant melanomas share a number of molecular attributes such as limitless replicative potential that define capabilities acquired by most malignancies. Accordingly, much effort has been focused on evaluating and validating protein markers related to these capabilities to function as melanoma prognostic markers. However, a few studies have also highlighted the prognostic value of markers that define melanocytic differentiation and the plasticity of melanoma cells to trans-differentiate along several other cellular pathways. Here, we provide a comprehensive review and evaluation of the prognostic significance of melanocyte-lineage markers such as MITF and melanogenic proteins, as well as markers of vascular epithelial and neuronal differentiation.

15.
J Invest Dermatol ; 130(6): 1657-67, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20182446

RESUMEN

Activating mutations in NRAS and BRAF are found frequently in cutaneous melanomas. Because concurrent mutations of both BRAF and RAS are extremely rare, it is thought that transformation by RAS and BRAF occurs through a common mechanism. Also, there is evidence for a relationship of synthetic lethality between NRAS and BRAF oncogenes that leads to selection against cells with a hyperactive mitogen-activated protein kinase (MAPK) pathway. However, it is not known whether the hyperactivation of the MAPK pathway by overexpression of either oncogene alone could also inhibit melanoma tumorigenesis. Here, we show that in melanoma cells with oncogenic BRAF (mBRAF), high levels of mBRAF induce hyperactivation of ERK and senescence-like phenotype and trigger autophagy by inhibiting the mammalian target of rapamycin complex signaling. Growth inhibition and cell death caused by high mBRAF levels are partially rescued by downregulation of BRAF protein or inhibition of autophagy, but not by inhibition of the MAPK or apoptotic pathways. In nude mice, growth of mBRAF-overexpressing tumors is inhibited. Quantitative immunohistochemical analysis of human melanomas and cell lines showed a significant positive correlation between the levels of BRAF protein and autophagy marker light chain 3. Our data suggest that high oncogenic BRAF levels trigger autophagy, which may have a role in melanoma tumor progression.


Asunto(s)
Autofagia/fisiología , Proliferación Celular , Melanoma/patología , Proteínas Proto-Oncogénicas B-raf/fisiología , Neoplasias Cutáneas/patología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Autofagia/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/fisiología , Masculino , Melanoma/fisiopatología , Ratones , Plásmidos/genética , Plásmidos/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas B-raf/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Neoplasias Cutáneas/fisiopatología , Serina-Treonina Quinasas TOR , Ensayos Antitumor por Modelo de Xenoinjerto
16.
J Invest Dermatol ; 127(9): 2123-8, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17495964

RESUMEN

The Class I MAGE proteins include the MAGE-A, MAGE-B, and MAGE-C antigens, which are normally expressed only in male germ cells but may be aberrantly expressed in melanomas and other tumors. It is known that MAGE gene expression is epigenetically repressed by promoter region methylation in most cells but factors controlling MAGE gene promoter methylation have not been identified. Using transcript microarray analysis and immunoblotting we found that MAGE-A and MAGE-C mRNA and protein are selectively downregulated by pharmacologic inhibition of KIT in KIT-dependent mast cell lines. Methylation-specific polymerase chain reaction studies showed that the MAGE-A3 and MAGE-C2 gene promoter regions were de-methylated in the presence of activated KIT but became methylated on inhibition of KIT, consistent with the downregulation of mRNA and protein. This is early evidence of a tyrosine kinase affecting MAGE gene promoter region methylation and expression, and represents early evidence of a tyrosine kinase in the epigenetic control of gene expression. MAGE proteins suppress apoptosis and promote tumor survival, and are novel targets for functional manipulation and immunotherapy. Understanding the factors controlling MAGE gene expression may allow more effective therapeutic strategies targeting MAGE antigens.


Asunto(s)
Antígenos de Neoplasias/fisiología , Epigénesis Genética , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/fisiología , Proteínas Proto-Oncogénicas c-kit/fisiología , Antígenos de Neoplasias/metabolismo , Antineoplásicos/farmacología , Apoptosis , Benzamidas , Línea Celular Tumoral , Metilación de ADN , Humanos , Mesilato de Imatinib , Proteínas de Neoplasias/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Piperazinas/farmacología , Regiones Promotoras Genéticas , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Pirimidinas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA