Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Cell ; 51(4): 409-22, 2013 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-23973372

RESUMEN

The individuals carrying melanocortin-1 receptor (MC1R) variants, especially those associated with red hair color, fair skin, and poor tanning ability (RHC trait), are more prone to melanoma; however, the underlying mechanism is poorly defined. Here, we report that UVB exposure triggers phosphatase and tensin homolog (PTEN) interaction with wild-type (WT), but not RHC-associated MC1R variants, which protects PTEN from WWP2-mediated degradation, leading to AKT inactivation. Strikingly, the biological consequences of the failure of MC1R variants to suppress PI3K/AKT signaling are highly context dependent. In primary melanocytes, hyperactivation of PI3K/AKT signaling leads to premature senescence; in the presence of BRAF(V600E), MC1R deficiency-induced elevated PI3K/AKT signaling drives oncogenic transformation. These studies establish the MC1R-PTEN axis as a central regulator for melanocytes' response to UVB exposure and reveal the molecular basis underlying the association between MC1R variants and melanomagenesis.


Asunto(s)
Regulación de la Expresión Génica/efectos de la radiación , Melanocitos/metabolismo , Melanoma Experimental/patología , Fosfohidrolasa PTEN/metabolismo , Receptor de Melanocortina Tipo 1/metabolismo , Pigmentación de la Piel/fisiología , Rayos Ultravioleta , Animales , Western Blotting , Células Cultivadas , Humanos , Técnicas para Inmunoenzimas , Melanocitos/efectos de la radiación , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Ratones , Mutación/genética , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Melanocortina Tipo 1/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Pigmentación de la Piel/efectos de la radiación , alfa-MSH/genética , alfa-MSH/metabolismo
2.
Lab Invest ; 97(6): 725-736, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28165469

RESUMEN

Melanoma is among the most virulent cancers, owing to its propensity to metastasize and its resistance to current therapies. The treatment failure is largely attributed to tumor heterogeneity, particularly subpopulations possessing stem cell-like properties, ie, melanoma stem-like cells (MSLCs). Evidence indicates that the MSLC phenotype is malleable and may be acquired by non-MSLCs through phenotypic switching upon appropriate stimuli, the so-called 'dynamic stemness'. Since the phenotypic characteristics and functional integrity of MSLCs depend on their vascular niche, using a two-dimensional (2D) melanoma-endothelium co-culture model, where the MSLC niche is recapitulated in vitro, we identified Notch3 signaling pathway as a micro-environmental cue governing MSLC phenotypic plasticity via pathway-specific gene expression arrays. Accordingly, lentiviral shRNA-mediated Notch3 knockdown (KD) in melanoma cell lines exhibiting high levels of endogenous Notch3 led to retarded/abolished tumorigenicity in vivo through both depleting MSLC fractions, evinced by MSLC marker downregulation (eg, CD133 and CD271); and impeding the MSLC niche, corroborated by the attenuated tumor angiogenesis as well as vasculogenic mimicry. In contrast, Notch3 KD affected neither tumor growth nor MSLC subsets in a melanoma cell line with relatively low endogenous Notch3 expression. Thus, Notch3 signaling may facilitate MSLC plasticity and niche morphogenesis in a cell context-dependent manner. Our findings illustrate Notch3 as a molecular switch driving melanoma heterogeneity, and provide the biological rationale for Notch inhibition as a promising therapeutic option.


Asunto(s)
Melanoma/metabolismo , Células Madre Neoplásicas/metabolismo , Receptor Notch3/metabolismo , Nicho de Células Madre/fisiología , Microambiente Tumoral/fisiología , Animales , Línea Celular Tumoral , Técnicas de Cocultivo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Transducción de Señal
3.
Cancer Cell ; 3(6): 525-30, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12842081

RESUMEN

Since the gene encoding Id1 was cloned in 1990, Id proteins have been implicated in regulating a variety of cellular processes, including cellular growth, senescence, differentiation, apoptosis, angiogenesis, and neoplastic transformation. The development of knockout and transgenic animal models for many members of the Id gene family has been particularly useful in sorting out the biologic relevance of these genes and their expression during normal development, malignant transformation, and tumor progression. Here we review the current understanding of Id gene function, the biologic consequences of Id gene expression, and the implications for Id gene regulation of cell growth and tumorigenesis.


Asunto(s)
Neoplasias/metabolismo , Proteínas Represoras , Factores de Transcripción/metabolismo , Animales , Apoptosis , Ciclo Celular , Diferenciación Celular , División Celular/fisiología , Transformación Celular Neoplásica , Senescencia Celular , Regulación de la Expresión Génica , Secuencias Hélice-Asa-Hélice , Humanos , Proteína 1 Inhibidora de la Diferenciación , Ratones
4.
Cancer Cell ; 4(4): 291-9, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14585356

RESUMEN

Id genes regulate tumor angiogenesis and loss of Id1 inhibits tumor xenograft growth in mice. Here we evaluate the role of Id1 in a more clinically relevant tumor model system using a two-step chemical carcinogenesis protocol. Remarkably, we find that Id1-/- mice are more susceptible to skin tumorigenesis compared to their wild-type counterparts. Cutaneous neoplasms in Id1-/- mice show increased proliferation without alterations in tumor angiogenesis; however, Id1-/- mice possess 50% fewer cutaneous gammadelta T cells than their wild-type counterparts due to an intrinsic migration defect associated with loss of expression of the chemokine receptor, CXCR4. We suggest that there are important differences between the mechanisms of angiogenesis in transplanted and autochthonous tumors and that these findings will have significant implications for the potential utility of antiangiogenic therapies in cancer.


Asunto(s)
Melanoma/metabolismo , Neovascularización Patológica/fisiopatología , Proteínas Represoras , Neoplasias Cutáneas/fisiopatología , Factores de Transcripción/metabolismo , Trasplante Heterólogo , Animales , Carcinógenos/toxicidad , Endotelio Vascular/fisiopatología , Citometría de Flujo , Proteína 1 Inhibidora de la Diferenciación , Melanoma/inducido químicamente , Ratones , Ratones Noqueados , Microscopía Fluorescente , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/fisiopatología , Receptores CXCR4/metabolismo , Neoplasias Cutáneas/inducido químicamente , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factores de Transcripción/genética
5.
Mol Cancer Res ; 20(2): 217-230, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34610961

RESUMEN

Aberrant epigenetic transcriptional regulation is linked to metastasis, a primary cause of cancer-related death. Dissecting the epigenetic mechanisms controlling metastatic progression may uncover important insights to tumor biology and potential therapeutic targets. Here, we investigated the role of the SIN3A histone deacetylase 1 and 2 (SIN3A-HDAC1/2) complex in cancer metastasis. Using a mouse model of melanoma metastasis, we found that the SIN3A-HDAC1/2 transcription repressor complex silences BMP6 expression, causing increased metastatic dissemination and tumor growth via suppression of BMP6-activated SMAD5 signaling. We further discovered that FAM83G/PAWS1, a downstream effector of BMP6-SMAD5 signaling, contributes critically to metastatic progression by promoting actin-dependent cytoskeletal dynamics and cell migration. Pharmacologic inhibition of the SIN3A-HDAC1/2 complex reduced the numbers of melanoma cells in the circulation and inhibited metastatic tumor growth by inducing disseminated cell dormancy, highlighting the SIN3A-HDAC1/2 repressor complex as a potential therapeutic target for blocking cancer metastasis. IMPLICATIONS: This study identifies the novel molecular links in the metastatic progression to target cytoskeletal dynamics in melanoma and identifies the SIN3A-HDAC1/2 complex and FAM83G/PAWS1 as potential targets for melanoma adjuvant therapy.


Asunto(s)
Proteína Morfogenética Ósea 6/metabolismo , Epigénesis Genética/genética , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Proteínas/metabolismo , Animales , Humanos , Melanoma , Ratones , Ratones Endogámicos NOD , Ratones SCID , Metástasis de la Neoplasia
6.
Cancer Res ; 79(10): 2649-2661, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30910803

RESUMEN

Histone modifications, largely regulated by histone acetyltransferases (HAT) and histone deacetylases, have been recognized as major regulatory mechanisms governing human diseases, including cancer. Despite significant effort and recent advances, the mechanism by which the HAT and transcriptional coactivator p300 mediates tumorigenesis remains unclear. Here, we use a genetic and chemical approach to identify the microphthalmia-associated transcription factor (MITF) as a critical downstream target of p300 driving human melanoma growth. Direct transcriptional control of MITF by p300-dependent histone acetylation within proximal gene regulatory regions was coupled to cellular proliferation, suggesting a significant growth regulatory axis. Further analysis revealed forkhead box M1 (FOXM1) as a key effector of the p300-MITF axis driving cell growth that is selectively activated in human melanomas. Targeted chemical inhibition of p300 acetyltransferase activity using a potent and selective catalytic p300/CBP inhibitor demonstrated significant growth inhibitory effects in melanoma cells expressing high levels of MITF. Collectively, these data confirm the critical role of the p300-MITF-FOXM1 axis in melanoma and support p300 as a promising novel epigenetic therapeutic target in human melanoma. SIGNIFICANCE: These results show that MITF is a major downstream target of p300 in human melanoma whose expression is predictive of melanoma response to small-molecule inhibition of p300 HAT activity.


Asunto(s)
Proteína p300 Asociada a E1A/genética , Melanoma/genética , Factor de Transcripción Asociado a Microftalmía/genética , Acetilación , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Senescencia Celular/genética , Proteína Forkhead Box M1/genética , Regulación Neoplásica de la Expresión Génica/genética , Histona Acetiltransferasas/genética , Humanos
7.
Mol Carcinog ; 47(9): 653-9, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18240291

RESUMEN

The Id family of helix-loop-helix transcription factors is upregulated in a variety of human malignancies and has been implicated in promoting tumorigenesis through effects on cell growth, differentiation, and tumor angiogenesis. While expression of Id proteins has been associated with tumorigenesis, the precise mechanistic relationship between Id expression and carcinogenesis has not been clearly delineated. We have previously shown that Id1 delays cellular senescence in primary mammalian cells through inhibition of the cell cycle regulatory protein and familial melanoma gene, p16/INK4a. We have also demonstrated that Id1 expression is upregulated in early stage primary human melanomas and may be an important marker for early malignancy. In order to further define the role of Id1 in human melanoma development, we have evaluated the function of Id1 in primary human melanocytes. Here we show that constitutive expression of Id1 in primary human melanocytes leads to delayed cellular senescence and decreased expression of the familial melanoma gene, p16/INK4a. Although melanocytes constitutively expressing Id1 are shown to possess extended lifespans, this is not associated with an appreciable change in cell growth or telomere length. We conclude that Id1 delays cellular senescence in primary human melanocytes through inhibition of p16/INK4a expression and suggest that Id1 may contribute to the malignant conversion of primary human melanocytes through extension of cellular lifespan.


Asunto(s)
División Celular/fisiología , Transformación Celular Neoplásica/patología , Senescencia Celular/fisiología , Proteína 1 Inhibidora de la Diferenciación/fisiología , Melanocitos/citología , Melanocitos/fisiología , Movimiento Celular , Transformación Celular Neoplásica/genética , Células Cultivadas , Prepucio/citología , Prepucio/fisiología , Humanos , Hibridación Fluorescente in Situ , Recién Nacido , Proteína 1 Inhibidora de la Diferenciación/genética , Masculino , Metilación , Regiones Promotoras Genéticas , Telómero/genética , Telómero/fisiología
8.
Melanoma Res ; 28(1): 71-75, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29227334

RESUMEN

There is a significant need for the development of diagnostic tools that can precisely distinguish Spitz nevi and spitzoid melanomas. Here, we report the development of a PCR-based quantitative diagnostic assay for spitzoid melanocytic lesions utilizing the expression ratio of neuropilin-2 and melan-A genes in primary tumor specimens. We find that the expression ratio of neuropilin-2/melan-A is significantly increased in spitzoid melanomas compared with Spitz nevi. The diagnostic potential of this quantitative assay was validated in two independent sets of patient samples as demonstrated in a receiver operating characteristic curve analysis showing an area under the curve value of 91.8%. Furthermore, the assay was found to quantitatively distinguish the clinical nature of atypical spitzoid melanocytic lesions that were diagnostically undetermined using histopathologic criteria alone. Our data indicate that this quantitative assay may be used as a tool in determining the diagnostic classification of histologically challenging spitzoid tumors.


Asunto(s)
Biomarcadores de Tumor/genética , Pruebas Diagnósticas de Rutina/métodos , Melanocitos/patología , Melanoma/diagnóstico , Neuropilina-2/genética , Nevo de Células Epitelioides y Fusiformes/diagnóstico , Biomarcadores de Tumor/análisis , Diagnóstico Diferencial , Humanos , Melanocitos/metabolismo , Melanoma/genética , Nevo de Células Epitelioides y Fusiformes/genética
9.
Mol Diagn Ther ; 22(2): 203-218, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29411301

RESUMEN

Earlier identification of aggressive melanoma remains a goal in the field of melanoma research. With new targeted and immune therapies that have revolutionized the care of patients with melanoma, the ability to predict progression and monitor or predict response to therapy has become the new focus of research into biomarkers in melanoma. In this review, promising biomarkers are highlighted. These biomarkers have been used to diagnose melanoma as well as predict progression to advanced disease and response to therapy. The biomarkers take various forms, including protein expression at the level of tissue, genetic mutations of cancer cells, and detection of circulating DNA. First, a brief description is provided about the conventional tissue markers used to stage melanoma, including tumor depth. Next, protein biomarkers, which provide both diagnostic and prognostic information, are described. This is followed by a discussion of important genetic mutations, microRNA, and epigenetic modifications that can provide therapeutic and prognostic material. Finally, emerging serologic biomarkers are reviewed, including circulating melanoma cells and exosomes. Overall the goal is to identify biomarkers that aid in the earlier identification and improved treatment of aggressive melanoma.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Melanoma/metabolismo , Neoplasias Cutáneas/metabolismo , Biomarcadores de Tumor/sangre , Humanos , Melanoma/diagnóstico , Melanoma/genética , Melanoma/patología , Terapia Molecular Dirigida , Pronóstico , Neoplasias Cutáneas/diagnóstico , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Resultado del Tratamiento , Melanoma Cutáneo Maligno
10.
Nat Commun ; 9(1): 53, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29302039

RESUMEN

Here we report corin, a synthetic hybrid agent derived from the class I HDAC inhibitor (entinostat) and an LSD1 inhibitor (tranylcypromine analog). Enzymologic analysis reveals that corin potently targets the CoREST complex and shows more sustained inhibition of CoREST complex HDAC activity compared with entinostat. Cell-based experiments demonstrate that corin exhibits a superior anti-proliferative profile against several melanoma lines and cutaneous squamous cell carcinoma lines compared to its parent monofunctional inhibitors but is less toxic to melanocytes and keratinocytes. CoREST knockdown, gene expression, and ChIP studies suggest that corin's favorable pharmacologic effects may rely on an intact CoREST complex. Corin was also effective in slowing tumor growth in a melanoma mouse xenograft model. These studies highlight the promise of a new class of two-pronged hybrid agents that may show preferential targeting of particular epigenetic regulatory complexes and offer unique therapeutic opportunities.


Asunto(s)
Benzamidas/farmacología , Proteínas Co-Represoras/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Melanoma/tratamiento farmacológico , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Piridinas/farmacología , Tranilcipromina/farmacología , Anciano , Animales , Antineoplásicos , Línea Celular Tumoral , Proliferación Celular , Proteínas Co-Represoras/metabolismo , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Histona Desacetilasas/química , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Proteínas del Tejido Nervioso/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Neoplasias Cutáneas/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Cancer Res ; 62(3): 819-26, 2002 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11830538

RESUMEN

Pancreatic adenocarcinoma is among the most fatal of cancers, in part because of late diagnosis and a lack of effective therapies. Comprehensive studies are needed to better understand and address the cellular mechanisms and pathways of tumorigenesis. Serial analysis of gene expression was used to analyze gene expression profiles of pancreatic cancer cell lines, short-term cultures of normal pancreatic ductal epithelium, and primary pancreatic cancer tissue. A total of 294,920 tags representing 77,746 genes in 10 serial analysis of gene expression libraries were analyzed. A pancreatic cancer cell line (Hs766T) that exhibited a "normoid" profile of gene expression was identified. Several genes that may be involved in the fundamental nature of malignant changes in pancreatic ductal epithelium were suggested from those differentially and highly expressed in pancreatic cancer cells as compared with normal epithelium. Some overexpressed genes, such as S100A4, prostate stem cell antigen, carcinoembryonic antigen-related cell adhesion molecule 6, and mesothelin, suggest potential use as diagnostic markers. Others suggest potential novel therapeutic targets.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Neoplasias Pancreáticas/genética , Carcinoma Ductal Pancreático/metabolismo , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Humanos , Mutación , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
12.
J Invest Dermatol ; 135(9): 2283-2291, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25910030

RESUMEN

Aberrant HGF-MET (hepatocyte growth factor-met proto-oncogene) signaling activation via interactions with surrounding stromal cells in tumor microenvironment has significant roles in malignant tumor progression. However, extracellular proteolytic regulation of HGF activation, which is influenced by the tumor microenvironment, and its consequential effects on melanoma malignancy remain uncharacterized. In this study, we identified SPINT2 (serine peptidase inhibitor Kunitz type 2), a proteolytic inhibitor of hepatocyte growth factor activator (HGFA), which has a significant role in the suppression of the HGF-MET pathway and malignant melanoma progression. SPINT2 expression is significantly lower in metastatic melanoma tissues compared with those in early-stage primary melanomas, which also corresponded with DNA methylation levels isolated from tissue samples. Treatment with the DNA-hypomethylating agent decitabine in cultured melanoma cells induced transcriptional reactivation of SPINT2, suggesting that this gene is epigenetically silenced in malignant melanomas. Furthermore, we show that ectopically expressed SPINT2 in melanoma cells inhibits the HGF-induced MET-AKT (v-Akt murine thymoma viral oncogene) signaling pathway and decreases malignant phenotype potential such as cell motility and invasive growth of melanoma cells. These results suggest that SPINT2 is associated with tumor-suppressive functions in melanoma by inhibiting an extracellular signal regulator of HGF, which is typically activated by tumor-stromal interactions. These findings indicate that epigenetic impairment of the tightly regulated cytokine-receptor communications in tumor microenvironment may contribute to malignant tumor progression.


Asunto(s)
Movimiento Celular/genética , Regulación Neoplásica de la Expresión Génica , Factor de Crecimiento de Hepatocito/metabolismo , Melanoma/genética , Glicoproteínas de Membrana/genética , Neoplasias Cutáneas/genética , Animales , Proliferación Celular/genética , Metilación de ADN , Epigénesis Genética , Silenciador del Gen , Factor de Crecimiento de Hepatocito/genética , Humanos , Immunoblotting , Melanoma/patología , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa/métodos , Regiones Promotoras Genéticas , Proto-Oncogenes Mas , Serina Endopeptidasas , Transducción de Señal/genética , Neoplasias Cutáneas/patología , Células Tumorales Cultivadas
13.
Biofabrication ; 8(1): 015001, 2015 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-26716792

RESUMEN

How metastatic cancer lesions survive and grow in secondary locations is not fully understood. There is a growing appreciation for the importance of tumor components, i.e. microenvironmental cells, in this process. Here, we used a simple microfabricated dual cell culture platform with a 500 µm gap to assess interactions between two different metastatic melanoma cell lines (1205Lu isolated from a lung lesion established through a mouse xenograft; and WM852 derived from a stage III metastatic lesion of skin) and microenvironmental cells derived from either skin (fibroblasts), lung (epithelial cells) or liver (hepatocytes). We observed differential bi-directional migration between microenvironmental cells and melanoma, depending on the melanoma cell line. Lung epithelial cells and skin fibroblasts, but not hepatocytes, stimulated higher 1205Lu migration than without microenvironmental cells; in the opposite direction, 1205Lu cells induced hepatocytes to migrate, but had no effect on skin fibroblasts and slightly inhibited lung epithelial cells. In contrast, none of the microenvironments had a significant effect on WM852; in this case, skin fibroblasts and hepatocytes--but not lung epithelial cells--exhibited directed migration toward WM852. These observations reveal significant effects a given microenvironmental cell line has on the two different melanoma lines, as well as how melanoma effects different microenvironmental cell lines. Our simple platform thus has potential to provide complex insights into different strategies used by cancerous cells to survive in and colonize metastatic sites.


Asunto(s)
Comunicación Celular , Técnicas de Cocultivo/instrumentación , Melanoma/fisiopatología , Melanoma/secundario , Ingeniería de Tejidos/métodos , Microambiente Tumoral/fisiología , Animales , Línea Celular Tumoral/clasificación , Diseño de Equipo , Análisis de Falla de Equipo , Dispositivos Laboratorio en un Chip , Melanoma/patología , Ratones , Impresión Tridimensional , Vísceras/patología
14.
Cancer Biol Ther ; 2(2): 164-70, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12750556

RESUMEN

The binding of activin and TGFbeta to their respective receptors initiates signals that are carried by common intermediates (Smad proteins) to induce transcriptional activation of downstream genes. Mutations in tumors indicate that both receptor types convey tumorsuppressive signals, among other biologic roles, but their respective sets of transcriptional targets (transcriptomes) and the shared degree of transcriptome similarity are not well explored in these cells. Transcriptome changes were analyzed by gene expression profiling after expression of constitutively active activin type I (ALK4m) and TGFbeta type I (ALK5m) receptors and by variation of Smad4 expression in cancer cells. Eleven of 15 previously reported TGFbeta downstream genes were confirmed to be responsive to TGFb and activin receptors in cancer cells. Expression profiling detected eight of these 11, as well as 13 new Smad4-dependent transcripts. Although Smad4-dependent CDKN1A/p21 induction represents the sole known effector of TGFbeta and activin tumor-suppressor effects, many downstream genes have not yet been evaluated for a suppressive role. A high similarity of TGFbeta and activin responses among the known and new transcriptional target genes indicated an essential redundancy of the two related inputs. This similarity helps relate the mutations seen in both receptor systems and their Smad mediators in human cancers.


Asunto(s)
Receptores de Activinas Tipo I/genética , Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Pancreáticas/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Adenoviridae/genética , Neoplasias de la Mama/metabolismo , Cartilla de ADN/química , Proteínas de Unión al ADN/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Luciferasas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Pancreáticas/metabolismo , Reacción en Cadena de la Polimerasa , Proteínas Serina-Treonina Quinasas , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Proteína Smad4 , Transactivadores/metabolismo , Transfección , Células Tumorales Cultivadas
15.
J Invest Dermatol ; 133(5): 1137-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23594536

RESUMEN

Despite the growing focus on microRNAs (miRNAs) as novel diagnostic tools and therapeutic targets in cancer, global characterization of miRNA expression patterns and their specific targets in melanoma has lagged. In this issue, Reuland et al. (2012) identify miR-26a as being specifically downregulated in human melanoma cells. They further establish Silencer of Death Domains as a novel target for miR-26a, which functionally mediates melanoma cell death. These findings suggest that miR-26a may serve as a promising novel therapy for subsets of melanoma.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Apoptosis/fisiología , Regulación hacia Abajo/fisiología , Melanoma/metabolismo , MicroARNs/metabolismo , Neoplasias Cutáneas/metabolismo , Humanos
16.
J Invest Dermatol ; 133(8): 2041-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23344460

RESUMEN

Vemurafenib (PLX4032), a selective inhibitor of Braf, has been approved by the US Food and Drug Administration for the treatment of unresectable or metastatic melanoma in patients with Braf(V600E) mutations. Many patients treated with vemurafenib initially display dramatic improvement, with decreases in both risk of death and tumor progression. Acquired resistance, however, rapidly arises in previously sensitive cells. We attempted to overcome this resistance by targeting the signal transducer and activator of transcription 3 (STAT3)-paired box homeotic gene 3 (PAX3)-signaling pathway, which is upregulated, owing to fibroblast growth factor 2 (FGF2) secretion or increased kinase activity, with the Braf(V600E) mutation. We found that activation of Stat3 or overexpression of PAX3 induced resistance to vemurafenib in melanoma cells. In addition, PAX3 or Stat3 silencing inhibited the growth of melanoma cells with acquired resistance to vemurafenib. Furthermore, treatment with the Stat3 inhibitor, WP1066, resulted in growth inhibition in both vemurafenib-sensitive and -resistant melanoma cells. Significantly, vemurafenib stimulation induced FGF2 secretion from keratinocytes and fibroblasts, which might uncover, at least in part, the mechanisms underlying targeting Stat3-PAX3 signaling to overcome the acquired resistance to vemurafenib. Our results suggest that Stat3-targeted therapy is a new therapeutic strategy to overcome the acquired resistance to vemurafenib in the treatment of melanoma.


Asunto(s)
Resistencia a Antineoplásicos/fisiología , Indoles/farmacología , Queratinocitos/efectos de los fármacos , Melanoma/tratamiento farmacológico , Factor de Transcripción STAT3/antagonistas & inhibidores , Neoplasias Cutáneas/tratamiento farmacológico , Sulfonamidas/farmacología , Antineoplásicos/farmacología , Línea Celular Transformada , Línea Celular Tumoral , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Prepucio/citología , Humanos , Queratinocitos/citología , Masculino , Melanoma/metabolismo , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Cultivo Primario de Células , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Piridinas/farmacología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Neoplasias Cutáneas/metabolismo , Tirfostinos/farmacología , Vemurafenib
17.
J Invest Dermatol ; 133(10): 2444-2452, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23698071

RESUMEN

Epigenetic events, including covalent post-translational modifications of histones, have been demonstrated to have critical roles in tumor development and progression. The transcriptional coactivator p300/CBP possesses both histone acetyltransferase (HAT) activity and scaffolding properties that directly influence the transcriptional activation of targeted genes. We have used a potent and specific inhibitor of p300/CBP HAT activity, C646, in order to evaluate the functional contributions of p300/CBP HAT to tumor development and progression. Here we report that C646 inhibits the growth of human melanoma and other tumor cells and promotes cellular senescence. Global assessment of the p300 HAT transcriptome in human melanoma identified functional roles in promoting cell cycle progression, chromatin assembly, and activation of DNA repair pathways through direct transcriptional regulatory mechanisms. In addition, C646 is shown to promote sensitivity to DNA damaging agents, leading to the enhanced apoptosis of melanoma cells after combination treatment with cisplatin. Together, our data suggest that p300 HAT activity mediates critical growth regulatory pathways in tumor cells and may serve as a potential therapeutic target for melanoma and other malignancies by promoting cellular responses to DNA damaging agents that are currently ineffective against specific cancers.


Asunto(s)
Benzoatos/farmacología , Senescencia Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Melanoma/patología , Pirazoles/farmacología , Neoplasias Cutáneas/patología , Factores de Transcripción p300-CBP/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Línea Celular Tumoral , Senescencia Celular/fisiología , Daño del ADN/fisiología , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/fisiología , Histona Acetiltransferasas/antagonistas & inhibidores , Histona Acetiltransferasas/metabolismo , Humanos , Melanoma/genética , Nitrobencenos , Pirazolonas , Neoplasias Cutáneas/genética , Factores de Transcripción p300-CBP/genética , Factores de Transcripción p300-CBP/metabolismo
18.
Cancer Res ; 71(7): 2433-44, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21324919

RESUMEN

Tumor-endothelium interactions are critical for tumor survival and metastasis. Melanomas can rapidly metastasize early in tumor progression, but the dependence of this aggressive behavior on tumor-stromal interaction is poorly understood. To probe the mechanisms involved, we developed a heterotypic coculture methodology, allowing simultaneous tracking of genomic and phenotypic changes in interacting tumor and endothelial cells in vitro. We found a dramatic rearrangement of endothelial cell networks into patterns reminiscent of vascular beds, even on plastic and glass. Multiple genes were upregulated in the process, many coding for cell surface and secreted proteins, including Neuropilin-2 (NRP2). A critical role of NRP2 in coordinated cell patterning and growth was confirmed using the coculture system. We conclude that NRP2 represents an important mediator of melanoma-endothelial interactions. Furthermore, the described methodology represents a powerful yet simple system to elucidate heterotypic intercellular interactions mediating diverse physiological and pathological processes.


Asunto(s)
Comunicación Celular/fisiología , Melanoma/genética , Melanoma/patología , Neuropilina-2/genética , Células del Estroma/patología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Técnicas de Cocultivo , Células Endoteliales/patología , Regulación Neoplásica de la Expresión Génica , Genotipo , Humanos , Ligandos , Melanoma/metabolismo , Neuropilina-2/biosíntesis , Fenotipo , Regulación hacia Arriba
20.
PLoS One ; 2(7): e594, 2007 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-17611626

RESUMEN

BACKGROUND: Gene expression profiling has revolutionized our ability to molecularly classify primary human tumors and significantly enhanced the development of novel tumor markers and therapies; however, progress in the diagnosis and treatment of melanoma over the past 3 decades has been limited, and there is currently no approved therapy that significantly extends lifespan in patients with advanced disease. Profiling studies of melanoma to date have been inconsistent due to the heterogeneous nature of this malignancy and the limited availability of informative tissue specimens from early stages of disease. METHODOLOGY/PRINCIPLE FINDINGS: In order to gain an improved understanding of the molecular basis of melanoma progression, we have compared gene expression profiles from a series of melanoma cell lines representing discrete stages of malignant progression that recapitulate critical characteristics of the primary lesions from which they were derived. Here we describe the unsupervised hierarchical clustering of profiling data from melanoma cell lines and melanocytes. This clustering identifies two distinctive molecular subclasses of melanoma segregating aggressive metastatic tumor cell lines from less-aggressive primary tumor cell lines. Further analysis of expression signatures associated with melanoma progression using functional annotations categorized these transcripts into three classes of genes: 1) Upregulation of activators of cell cycle progression, DNA replication and repair (CDCA2, NCAPH, NCAPG, NCAPG2, PBK, NUSAP1, BIRC5, ESCO2, HELLS, MELK, GINS1, GINS4, RAD54L, TYMS, and DHFR), 2) Loss of genes associated with cellular adhesion and melanocyte differentiation (CDH3, CDH1, c-KIT, PAX3, CITED1/MSG-1, TYR, MELANA, MC1R, and OCA2), 3) Upregulation of genes associated with resistance to apoptosis (BIRC5/survivin). While these broad classes of transcripts have previously been implicated in the progression of melanoma and other malignancies, the specific genes identified within each class of transcripts are novel. In addition, the transcription factor NF-KB was specifically identified as being a potential "master regulator" of melanoma invasion since NF-KB binding sites were identified as consistent consensus sequences within promoters of progression-associated genes. CONCLUSIONS/SIGNIFICANCE: We conclude that tumor cell lines are a valuable resource for the early identification of gene signatures associated with malignant progression in tumors with significant heterogeneity like melanoma. We further conclude that the development of novel data reduction algorithms for analysis of microarray studies is critical to allow for optimized mining of important, clinically-relevant datasets. It is expected that subsequent validation studies in primary human tissues using such an approach will lead to more rapid translation of such studies to the identification of novel tumor biomarkers and therapeutic targets.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Melanoma/genética , Adhesión Celular/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Reparación del ADN , Replicación del ADN , ADN de Neoplasias/genética , Progresión de la Enfermedad , Humanos , Melanocitos/citología , Melanocitos/fisiología , Melanoma/patología , Invasividad Neoplásica/genética , Proteínas de Neoplasias/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA