Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 537(7620): 422-426, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27580028

RESUMEN

Melanoma is the deadliest form of commonly encountered skin cancer because of its rapid progression towards metastasis. Although metabolic reprogramming is tightly associated with tumour progression, the effect of metabolic regulatory circuits on metastatic processes is poorly understood. PGC1α is a transcriptional coactivator that promotes mitochondrial biogenesis, protects against oxidative stress and reprograms melanoma metabolism to influence drug sensitivity and survival. Here, we provide data indicating that PGC1α suppresses melanoma metastasis, acting through a pathway distinct from that of its bioenergetic functions. Elevated PGC1α expression inversely correlates with vertical growth in human melanoma specimens. PGC1α silencing makes poorly metastatic melanoma cells highly invasive and, conversely, PGC1α reconstitution suppresses metastasis. Within populations of melanoma cells, there is a marked heterogeneity in PGC1α levels, which predicts their inherent high or low metastatic capacity. Mechanistically, PGC1α directly increases transcription of ID2, which in turn binds to and inactivates the transcription factor TCF4. Inactive TCF4 causes downregulation of metastasis-related genes, including integrins that are known to influence invasion and metastasis. Inhibition of BRAFV600E using vemurafenib, independently of its cytostatic effects, suppresses metastasis by acting on the PGC1α-ID2-TCF4-integrin axis. Together, our findings reveal that PGC1α maintains mitochondrial energetic metabolism and suppresses metastasis through direct regulation of parallel acting transcriptional programs. Consequently, components of these circuits define new therapeutic opportunities that may help to curb melanoma metastasis.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Melanoma/genética , Melanoma/patología , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/prevención & control , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Transcripción Genética , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación hacia Abajo , Metabolismo Energético , Humanos , Indoles/farmacología , Indoles/uso terapéutico , Proteína 2 Inhibidora de la Diferenciación/genética , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Integrinas/genética , Integrinas/metabolismo , Masculino , Ratones , Mitocondrias/metabolismo , Invasividad Neoplásica/genética , Metástasis de la Neoplasia/tratamiento farmacológico , Biogénesis de Organelos , Estrés Oxidativo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/deficiencia , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Factor de Transcripción 4 , Factores de Transcripción/metabolismo , Vemurafenib
2.
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
3.
Proc Natl Acad Sci U S A ; 114(17): E3434-E3443, 2017 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-28396387

RESUMEN

Oncogenic PIK3CA mutations are found in a significant fraction of human cancers, but therapeutic inhibition of PI3K has only shown limited success in clinical trials. To understand how mutant PIK3CA contributes to cancer cell proliferation, we used genome scale loss-of-function screening in a large number of genomically annotated cancer cell lines. As expected, we found that PIK3CA mutant cancer cells require PIK3CA but also require the expression of the TCA cycle enzyme 2-oxoglutarate dehydrogenase (OGDH). To understand the relationship between oncogenic PIK3CA and OGDH function, we interrogated metabolic requirements and found an increased reliance on glucose metabolism to sustain PIK3CA mutant cell proliferation. Functional metabolic studies revealed that OGDH suppression increased levels of the metabolite 2-oxoglutarate (2OG). We found that this increase in 2OG levels, either by OGDH suppression or exogenous 2OG treatment, resulted in aspartate depletion that was specifically manifested as auxotrophy within PIK3CA mutant cells. Reduced levels of aspartate deregulated the malate-aspartate shuttle, which is important for cytoplasmic NAD+ regeneration that sustains rapid glucose breakdown through glycolysis. Consequently, because PIK3CA mutant cells exhibit a profound reliance on glucose metabolism, malate-aspartate shuttle deregulation leads to a specific proliferative block due to the inability to maintain NAD+/NADH homeostasis. Together these observations define a precise metabolic vulnerability imposed by a recurrently mutated oncogene.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I , Complejo Cetoglutarato Deshidrogenasa , Mutación , Proteínas de Neoplasias , Neoplasias , Animales , Línea Celular Tumoral , Ciclo del Ácido Cítrico/genética , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Glucólisis/genética , Humanos , Complejo Cetoglutarato Deshidrogenasa/biosíntesis , Complejo Cetoglutarato Deshidrogenasa/genética , Ratones , Ratones Desnudos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/enzimología , Neoplasias/genética , Neoplasias/patología
4.
Am J Pathol ; 185(1): 252-65, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25447045

RESUMEN

Microphthalmia-associated transcription factor (MITF) acts via pigment epithelium-derived factor (PEDF), an antiangiogenic protein, to regulate retinal pigment epithelium migration. PEDF expression and/or regulation during melanoma development have not been investigated previously. Using immunohistochemistry, we determined expression of PEDF in common and dysplastic melanocytic nevi, melanoma in situ, invasive melanoma, and metastatic melanoma (n = 102). PEDF expression was consistently decreased in invasive and metastatic melanoma, compared with nevi and melanoma in situ (P < 0.0001). PEDF was lost in thicker melanomas (P = 0.003), and correlated with depth of invasion (P = 0.003) and distant metastasis (P = 0.0331), but only marginally with mitotic index, AJCC stage, nodal metastasis, or blood vascular density (0.05 < P < 0.10). Quantitative real-time PCR and microarray analyses confirmed PEDF down-regulation at the mRNA level in several melanoma lines, compared with melanocytes. MITF positively correlated with PEDF expression in invasive melanomas (P = 0.0003). Searching for PEDF regulatory mechanisms revealed two occupied conserved E-boxes (DNA recognition elements) in the first intron of the human and mouse PEDF promoter regions, confirmed by binding assays. Dominant-negative and siRNA approaches in vivo demonstrated direct transcriptional influence of MITF on PEDF, establishing the PEDF gene (SERPINF1) as a MITF target in melanocytes and melanoma cells. These findings suggest that loss of PEDF expression promotes early invasive melanoma growth.


Asunto(s)
Proteínas del Ojo/metabolismo , Regulación Neoplásica de la Expresión Génica , Melanoma/metabolismo , Factor de Transcripción Asociado a Microftalmía/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Serpinas/metabolismo , Neoplasias Cutáneas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Secuencia de Bases , Línea Celular Tumoral , Femenino , Silenciador del Gen , Humanos , Inmunohistoquímica , Masculino , Melanocitos , Ratones , Microscopía Fluorescente , Persona de Mediana Edad , Datos de Secuencia Molecular , Invasividad Neoplásica , Metástasis de la Neoplasia , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño/metabolismo , Homología de Secuencia de Ácido Nucleico , Adulto Joven
5.
Nature ; 459(7250): 1085-90, 2009 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-19553991

RESUMEN

Genome-wide copy number analyses of human cancers identified a frequent 5p13 amplification in several solid tumour types, including lung (56%), ovarian (38%), breast (32%), prostate (37%) and melanoma (32%). Here, using integrative analysis of a genomic profile of the region, we identify a Golgi protein, GOLPH3, as a candidate targeted for amplification. Gain- and loss-of-function studies in vitro and in vivo validated GOLPH3 as a potent oncogene. Physically, GOLPH3 localizes to the trans-Golgi network and interacts with components of the retromer complex, which in yeast has been linked to target of rapamycin (TOR) signalling. Mechanistically, GOLPH3 regulates cell size, enhances growth-factor-induced mTOR (also known as FRAP1) signalling in human cancer cells, and alters the response to an mTOR inhibitor in vivo. Thus, genomic and genetic, biological, functional and biochemical data in yeast and humans establishes GOLPH3 as a new oncogene that is commonly targeted for amplification in human cancer, and is capable of modulating the response to rapamycin, a cancer drug in clinical use.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Proteínas de la Membrana/metabolismo , Neoplasias/fisiopatología , Proteínas Quinasas/metabolismo , Transducción de Señal , Sirolimus/farmacología , Animales , Línea Celular Tumoral/efectos de los fármacos , Proteínas de Unión al ADN/genética , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Desnudos , Proteínas Quinasas/genética , Saccharomyces cerevisiae/genética , Serina-Treonina Quinasas TOR , Factores de Transcripción/genética
6.
Cancer Causes Control ; 25(1): 125-32, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24158781

RESUMEN

PURPOSE: Genetic predisposition plays a major role in the etiology of melanoma, but known genetic markers only account for a limited fraction of family-history-associated melanoma cases. Expression microarrays have offered the opportunity to identify further genomic profiles correlated with family history of melanoma. We aimed to distinguish mRNA expression signatures between melanoma cases with and without a family history of melanoma. METHODS: Based on the Nurses' Health Study, family history was defined as having one or more first-degree family members diagnosed with melanoma. Melanoma diagnosis was confirmed by reviewing pathology reports, and tumor blocks were collected by mail from across the USA. Genomic interrogation was accomplished through evaluating expression profiling of formalin-fixed paraffin-embedded tissues from 78 primary cutaneous invasive melanoma cases, on either a 6K or whole-genome (24K) Illumina gene chip. Gene set enrichment analysis was performed for each batch to determine the differentially enriched pathways and key contributing genes. RESULTS: The CXC chemokine receptor 4 (CXCR4) pathway was consistently up-regulated within cases of familial melanoma in both platforms. Leading edge analysis showed four genes from the CXCR4 pathway, including MAPK1, PLCG1, CRK, and PTK2, were among the core members that contributed to the enrichment of this pathway. There was no association between the enrichment of CXCR4 pathway and NRAS, BRAF mutation, or Breslow thickness of the primary melanoma cases. CONCLUSIONS: We found that the CXCR4 pathway might constitute a novel susceptibility pathway associated with family history of melanoma in first-degree relatives.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Melanoma/genética , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Adulto , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Masculino , Melanoma/etiología , Melanoma/metabolismo , Persona de Mediana Edad , Mutación/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , ARN Mensajero/genética , Regulación hacia Arriba/genética
7.
Cancer Cell ; 9(6): 473-84, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16766266

RESUMEN

Clear cell sarcoma (CCS) harbors a pathognomonic chromosomal translocation fusing the Ewing's sarcoma gene (EWS) to the CREB family transcription factor ATF1 and exhibits melanocytic features. We show that EWS-ATF1 occupies the MITF promoter, mimicking melanocyte-stimulating hormone (MSH) signaling to induce expression of MITF, the melanocytic master transcription factor and an amplified oncogene in melanoma. Knockdown/rescue studies revealed that MITF mediates the requirement of EWS-ATF1 for CCS survival in vitro and in vivo as well as for melanocytic differentiation. Moreover, MITF and TFE3 reciprocally rescue one another in lines derived from CCS or pediatric renal carcinoma. Seemingly unrelated tumors thus employ distinct strategies to oncogenically dysregulate the MiT family, collectively broadening the definition of MiT-associated human cancers.


Asunto(s)
Factor de Transcripción Activador 1/metabolismo , Proteínas de Unión al ADN/metabolismo , Factor de Transcripción Asociado a Microftalmía/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Fusión Oncogénica/fisiología , Proteína EWS de Unión a ARN/genética , Sarcoma de Células Claras/metabolismo , Factor de Transcripción Activador 1/genética , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Proteínas del Grupo de Alta Movilidad/biosíntesis , Humanos , Hormonas Estimuladoras de los Melanocitos/fisiología , Melanocitos/metabolismo , Melanocitos/patología , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Desnudos , Factor de Transcripción Asociado a Microftalmía/genética , Trasplante de Neoplasias , Proteínas Nucleares/genética , Proteínas de Fusión Oncogénica/genética , Regiones Promotoras Genéticas , Factores de Transcripción del Factor Regulador X , Factores de Transcripción SOXE , Sarcoma de Células Claras/patología , Transducción de Señal , Factores de Transcripción/biosíntesis
8.
Proc Natl Acad Sci U S A ; 108(37): E699-708, 2011 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-21876152

RESUMEN

The PI3K pathway is frequently activated in cancer; therefore, considerable effort is focused on identifying compounds that can inhibit specific pathway components, particularly the hallmark oncogene PIK3CA. Although targeted inhibition of a cancer survival gene holds significant promise, there are concerns that drug resistance may emerge within the cancerous cells, thus limiting clinical efficacy. Using genetically defined human mammary epithelial cells, we evolved resistance to the PI3K/mammalian target of rapamycin (mTOR) inhibitor BEZ235, and by genome-wide copy number analyses, we identified MYC and eIF4E amplification within the resistant cells. Importantly, either MYC or eukaryotic translation initiation factor 4E (eIF4E) was required to bypass pharmacological PI3K/mTOR inhibition in resistant cells. Furthermore, these cells displayed elevated 5' cap-dependent protein translation. Collectively, these findings suggest that analysis of drivers of protein translation could facilitate the identification of cancer lesions that confer resistance to PI3K pathway-targeted drugs.


Asunto(s)
Factor 4E Eucariótico de Iniciación/genética , Amplificación de Genes , Terapia Molecular Dirigida , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-myc/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Factor 4E Eucariótico de Iniciación/metabolismo , Femenino , Amplificación de Genes/efectos de los fármacos , Dosificación de Gen/genética , Genoma Humano/genética , Humanos , Imidazoles/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Mutación Puntual/genética , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/metabolismo , Quinolinas/farmacología , Caperuzas de ARN/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Regulación hacia Arriba/efectos de los fármacos
9.
Proc Natl Acad Sci U S A ; 108(43): E924-33, 2011 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-21949374

RESUMEN

Microphthalmia-associated transcription factor (MITF) regulates normal melanocyte development and is also a lineage-selective oncogene implicated in melanoma and clear-cell sarcoma (i.e., melanoma of soft parts). We have observed that MITF expression is potently reduced under hypoxic conditions in primary melanocytes and melanoma and clear cell sarcoma cells through hypoxia inducible factor 1 (HIF1)-mediated induction of the transcriptional repressor differentially expressed in chondrocytes protein 1 (DEC1) (BHLHE40), which subsequently binds and suppresses the promoter of M-MITF (melanocyte-restricted MITF isoform). Correspondingly, hypoxic conditions or HIF1α stabilization achieved by using small-molecule prolyl-hydroxylase inhibitors reduced M-MITF expression, leading to melanoma cell growth arrest that was rescued by ectopic expression of M-MITF in vitro. Prolyl hydroxylase inhibition also potently suppressed melanoma growth in a mouse xenograft model. These studies illuminate a physiologic hypoxia response in pigment cells leading to M-MITF suppression, one that suggests a potential survival advantage mechanism for MITF amplification in metastatic melanoma and offers a small-molecule strategy for suppression of the MITF oncogene in vivo.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación de la Expresión Génica/fisiología , Proteínas de Homeodominio/metabolismo , Factor 1 Inducible por Hipoxia/metabolismo , Melanocitos/metabolismo , Melanoma/metabolismo , Factor de Transcripción Asociado a Microftalmía/metabolismo , Análisis de Varianza , Animales , Western Blotting , Hipoxia de la Célula/fisiología , Inmunoprecipitación de Cromatina , Cartilla de ADN/genética , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Plásmidos/genética , Interferencia de ARN , Reacción en Cadena en Tiempo Real de la Polimerasa
10.
Am J Pathol ; 180(6): 2462-78, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22546478

RESUMEN

Lesions displaying a variety of dysplastic changes precede invasive oral and epidermal squamous cell carcinoma (SCC); however, there are no histopathological criteria for either confirming or staging premalignancy. SCCs and dysplasias frequently contain cells that abnormally express the γ2 subunit of laminin-332. We developed cell culture models to investigate γ2 dysregulation. Normal human keratinocytes displayed density-dependent repression of γ2, whereas premalignant keratinocytes and SCC cells overexpressed γ2 and secreted laminin assembly intermediates. Neoplastic cells had hyperactive EGFR/MAPK(ERK) signaling coordinate with overexpressed γ2, and EGFR and MEK inhibitors normalized γ2 expression. Keratinocytes engineered to express HPV16 E6 or activated mutant HRAS, cRAF1, or MEK1 lost density repression of γ2 and shared with neoplastic cells signaling abnormalities downstream of ERK, including increased phosphorylation of S6 and eIF4 translation factors. Notably, qPCR results revealed that γ2 overexpression was not accompanied by increased γ2 mRNA levels, consistent with ERK-dependent, eIF4B-mediated translation initiation of the stem-looped, 5'-untranslated region of γ2 mRNA in neoplastic cells. Inhibitors of MEK, but not of TORC1/2, blocked S6 and eIF4B phosphorylation and γ2 overexpression. Immunostaining of oral dysplasias identified γ2 overexpression occurring within fields of basal cells that had elevated p-S6 levels. These results reveal a causal relationship between ERK-dependent translation factor activation and laminin γ2 dysregulation and identify new markers of preinvasive neoplastic change during progression to SCC.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Laminina/biosíntesis , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Neoplasias de la Boca/metabolismo , Lesiones Precancerosas/metabolismo , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/patología , Progresión de la Enfermedad , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Queratinocitos/metabolismo , Laminina/genética , Sistema de Señalización de MAP Quinasas/fisiología , Neoplasias de la Boca/enzimología , Neoplasias de la Boca/patología , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Lesiones Precancerosas/enzimología , Lesiones Precancerosas/patología , Modificación Traduccional de las Proteínas/fisiología , Transducción de Señal/fisiología , Células Tumorales Cultivadas , Quinasas raf/fisiología , Proteínas ras/fisiología
11.
bioRxiv ; 2023 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-37873273

RESUMEN

Targeting of specific metabolic pathways in tumor cells has the potential to sensitize them to immune-mediated attack. Here we provide evidence for a specific means of mitochondrial respiratory Complex I (CI) inhibition that improves tumor immunogenicity and sensitivity to immune checkpoint blockade (ICB). Targeted genetic deletion of the CI subunits Ndufs4 and Ndufs6 , but not other subunits, induces an immune-dependent tumor growth attenuation in mouse melanoma models. We show that deletion of Ndufs4 induces expression of the transcription factor Nlrc5 and genes in the MHC class I antigen presentation and processing pathway. This induction of MHC-related genes is driven by an accumulation of pyruvate dehydrogenase-dependent mitochondrial acetyl-CoA downstream of CI subunit deletion. This work provides a novel functional modality by which selective CI inhibition restricts tumor growth, suggesting that specific targeting of Ndufs4 , or related CI subunits, increases T-cell mediated immunity and sensitivity to ICB.

12.
Nat Commun ; 14(1): 3251, 2023 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-37277330

RESUMEN

While targeted treatment against BRAF(V600E) improve survival for melanoma patients, many will see their cancer recur. Here we provide data indicating that epigenetic suppression of PGC1α defines an aggressive subset of chronic BRAF-inhibitor treated melanomas. A metabolism-centered pharmacological screen further identifies statins (HMGCR inhibitors) as a collateral vulnerability within PGC1α-suppressed BRAF-inhibitor resistant melanomas. Lower PGC1α levels mechanistically causes reduced RAB6B and RAB27A expression, whereby their combined re-expression reverses statin vulnerability. BRAF-inhibitor resistant cells with reduced PGC1α have increased integrin-FAK signaling and improved extracellular matrix detached survival cues that helps explain their increased metastatic ability. Statin treatment blocks cell growth by lowering RAB6B and RAB27A prenylation that reduces their membrane association and affects integrin localization and downstream signaling required for growth. These results suggest that chronic adaptation to BRAF-targeted treatments drive novel collateral metabolic vulnerabilities, and that HMGCR inhibitors may offer a strategy to treat melanomas recurring with suppressed PGC1α expression.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas , Melanoma , Humanos , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Sensibilidad Colateral al uso de Fármacos , Recurrencia Local de Neoplasia , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/patología , Inhibidores de Proteínas Quinasas/farmacología , Integrinas/metabolismo , Epigénesis Genética , Línea Celular Tumoral , Mutación , Hidroximetilglutaril-CoA Reductasas/metabolismo
13.
Lab Invest ; 92(3): 362-70, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22184093

RESUMEN

The mechanisms of melanoma invasion are poorly understood despite extensive inquiry. SRY (sex determining region Y)-box 2 (SOX2) is an embryonic stem cell transcription factor that has recently been discovered to be expressed in human melanoma where it is associated with dermal invasion and primary tumor thickness. To assess the potential role of SOX2 expression in melanoma invasion, we examined patient melanomas and humanized melanoma xenografts, and noted preferential SOX2 expression in cells that interfaced and infiltrated dermal stroma. Experimental knockdown (KD) of SOX2 mRNA and protein in A2058 melanoma cells with high constitutive SOX2 expression resulted in 4.5-fold decreased invasiveness in vitro compared with controls (P<0.0001). Conversely, when G361 cells that normally express low SOX2 were transduced to overexpress SOX2 mRNA and protein, a 3.8-fold increase in invasiveness was observed (P=0.0004). Among 84 invasion-related genes, RT-PCR screening revealed that SOX2 KD resulted in striking decrease in matrix metalloproteinase-3 (MMP-3), an endopeptidase associated with cleavage of the extracellular matrix. Quantitatively, SOX2 KD diminished MMP-3 mRNA by 87.8%. MMP-3 KD in SOX2-expressing A2058 cells served to inhibit invasion, although to a lesser degree than SOX2 KD. Finally, immunostaining of patient and xenograft melanomas revealed coordinate SOX2 and MMP-3 expression in regions of stromal infiltration. These data implicate SOX2 expression in melanoma invasion, and suggest a role for MMP-3 as one potential mediator of this process.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Melanoma/metabolismo , Factores de Transcripción SOXB1/metabolismo , Animales , Línea Celular Tumoral , Humanos , Melanoma/patología , Ratones , Ratones SCID , Invasividad Neoplásica , Trasplante de Neoplasias
14.
Cancer Cell ; 6(6): 565-76, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15607961

RESUMEN

The genomic organization of the CDK2 gene, which overlaps the melanocyte-specific gene SILV/PMEL17, poses an interesting regulatory challenge. We show that, despite its ubiquitous expression, CDK2 exhibits tissue-specific regulation by the essential melanocyte lineage transcription factor MITF. In addition, functional studies revealed this regulation to be critical for maintaining CDK2 kinase activity and growth of melanoma cells. Expression levels of MITF and CDK2 are tightly correlated in primary melanoma specimens and predict susceptibility to the CDK2 inhibitor roscovitine. CDK2 depletion suppressed growth and cell cycle progression in melanoma, but not other cancers, corroborating previous results. Collectively, these data indicate that CDK2 activity in melanoma is largely maintained at the transcriptional level by MITF, and unlike other malignancies, it may be a suitable drug target in melanoma.


Asunto(s)
Quinasas CDC2-CDC28/fisiología , Proteínas de Unión al ADN/fisiología , Melanoma/patología , Factores de Transcripción/fisiología , Western Blotting , Quinasas CDC2-CDC28/genética , Quinasas CDC2-CDC28/metabolismo , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Inmunoprecipitación de Cromatina , Quinasa 2 Dependiente de la Ciclina , Quinasas Ciclina-Dependientes/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Elementos E-Box/fisiología , Fibroblastos/metabolismo , Citometría de Flujo , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes Reporteros/genética , Humanos , Melanocitos/metabolismo , Melanocitos/patología , Melanoma/metabolismo , Glicoproteínas de Membrana , Factor de Transcripción Asociado a Microftalmía , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas/genética , Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Purinas/farmacología , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Roscovitina , Fase S/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Transfección , Proteína bcl-X , Antígeno gp100 del Melanoma
15.
Nature ; 436(7047): 117-22, 2005 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-16001072

RESUMEN

Systematic analyses of cancer genomes promise to unveil patterns of genetic alterations linked to the genesis and spread of human cancers. High-density single-nucleotide polymorphism (SNP) arrays enable detailed and genome-wide identification of both loss-of-heterozygosity events and copy-number alterations in cancer. Here, by integrating SNP array-based genetic maps with gene expression signatures derived from NCI60 cell lines, we identified the melanocyte master regulator MITF (microphthalmia-associated transcription factor) as the target of a novel melanoma amplification. We found that MITF amplification was more prevalent in metastatic disease and correlated with decreased overall patient survival. BRAF mutation and p16 inactivation accompanied MITF amplification in melanoma cell lines. Ectopic MITF expression in conjunction with the BRAF(V600E) mutant transformed primary human melanocytes, and thus MITF can function as a melanoma oncogene. Reduction of MITF activity sensitizes melanoma cells to chemotherapeutic agents. Targeting MITF in combination with BRAF or cyclin-dependent kinase inhibitors may offer a rational therapeutic avenue into melanoma, a highly chemotherapy-resistant neoplasm. Together, these data suggest that MITF represents a distinct class of 'lineage survival' or 'lineage addiction' oncogenes required for both tissue-specific cancer development and tumour progression.


Asunto(s)
Linaje de la Célula , Proteínas de Unión al ADN/genética , Amplificación de Genes/genética , Genómica , Melanoma/genética , Melanoma/patología , Oncogenes/genética , Factores de Transcripción/genética , Línea Celular Tumoral , Supervivencia Celular , Cromosomas Humanos Par 3/genética , Progresión de la Enfermedad , Dosificación de Gen , Regulación Neoplásica de la Expresión Génica , Humanos , Hibridación Fluorescente in Situ , Factor de Transcripción Asociado a Microftalmía , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple/genética
16.
Oncogene ; 40(1): 112-126, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33082558

RESUMEN

Tuberous sclerosis complex (TSC) is an autosomal dominant tumor suppressor syndrome, characterized by tumor development in multiple organs, including renal angiomyolipoma. Biallelic loss of TSC1 or TSC2 is a known genetic driver of angiomyolipoma development, however, whether an altered transcriptional repertoire contributes to TSC-associated tumorigenesis is unknown. RNA-seq analyses showed that MITF A isoform (MITF-A) was consistently highly expressed in angiomyolipoma, immunohistochemistry showed microphthalmia-associated transcription factor nuclear localization, and Chromatin immuno-Precipitation Sequencing analysis showed that the MITF-A transcriptional start site was highly enriched with H3K27ac marks. Using the angiomyolipoma cell line 621-101, MITF knockout (MITF.KO) and MITF-A overexpressing (MITF.OE) cell lines were generated. MITF.KO cells showed markedly reduced growth and invasion in vitro, and were unable to form xenografted tumors. In contrast, MITF.OE cells grew faster in vitro and as xenografted tumors compared to control cells. RNA-Seq analysis showed that both ID2 and Cysteine-rich angiogenic inducer 61 (CYR61) expression levels were increased in the MITF.OE cells and reduced in the MITF.KO cells, and luciferase assays showed this was due to transcriptional effects. Importantly, CYR61 overexpression rescued MITF.KO cell growth in vitro and tumor growth in vivo. These findings suggest that MITF-A is a transcriptional oncogenic driver of angiomyolipoma tumor development, acting through regulation of CYR61.


Asunto(s)
Angiomiolipoma/patología , Proteína 61 Rica en Cisteína/genética , Proteína 2 Inhibidora de la Diferenciación/genética , Neoplasias Renales/patología , Factor de Transcripción Asociado a Microftalmía/genética , Regulación hacia Arriba , Angiomiolipoma/genética , Angiomiolipoma/metabolismo , Animales , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Ratones , Factor de Transcripción Asociado a Microftalmía/metabolismo , Invasividad Neoplásica , Trasplante de Neoplasias , Isoformas de ARN/genética , Análisis de Secuencia de ARN , Sitio de Iniciación de la Transcripción
17.
J Clin Invest ; 130(2): 853-862, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31929186

RESUMEN

Oncogene-targeted and immune checkpoint therapies have revolutionized the clinical management of malignant melanoma and now offer hope to patients with advanced disease. Intimately connected to patients' overall clinical risk is whether the initial primary melanoma lesion will metastasize and cause advanced disease, but underlying mechanisms are not entirely understood. A subset of melanomas display heightened peroxisome proliferator-activated receptor γ coactivator 1-α (PGC1α) expression that maintains cell survival cues by promoting mitochondrial function, but also suppresses metastatic spread. Here, we show that PGC1α expression in melanoma cells was silenced by chromatin modifications that involve promoter H3K27 trimethylation. Pharmacological EZH2 inhibition diminished H3K27me3 histone markers, increased PGC1α expression, and functionally suppressed invasion within PGC1α-silenced melanoma cells. Mechanistically, PGC1α silencing activated transcription factor 12 (TCF12), to increase expression of WNT5A, which in turn stabilized YAP protein levels to promote melanoma migration and metastasis. Accordingly, inhibition of components of this transcription-signaling axis, including TCF12, WNT5A, or YAP, blocked melanoma migration in vitro and metastasis in vivo. These results indicate that epigenetic control of melanoma metastasis involved altered expression of PGC1α and an association with the inherent metabolic state of the tumor.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Histonas/metabolismo , Melanoma Experimental/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/biosíntesis , Factores de Transcripción/metabolismo , Proteína Wnt-5a/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Línea Celular Tumoral , Células HEK293 , Histonas/genética , Humanos , Melanoma Experimental/genética , Melanoma Experimental/patología , Ratones , Ratones Desnudos , Invasividad Neoplásica , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Factores de Transcripción/genética , Proteína Wnt-5a/genética , Proteínas Señalizadoras YAP
18.
Cancer Immunol Res ; 8(5): 660-671, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32161110

RESUMEN

We assessed the contribution of IL1 signaling molecules to malignant tumor growth using IL1ß-/-, IL1α-/-, and IL1R1-/- mice. Tumors grew progressively in IL1R-/- and IL1α-/- mice but were often absent in IL1ß-/- mice. This was observed whether tumors were implanted intradermally or injected intravenously and was true across multiple distinct tumor lineages. Antibodies to IL1ß prevented tumor growth in wild-type (WT) mice but not in IL1R1-/- or IL1α-/- mice. Antibodies to IL1α promoted tumor growth in IL1ß-/- mice and reversed the tumor-suppressive effect of anti-IL1ß in WT mice. Depletion of CD8+ T cells and blockade of lymphocyte mobilization abrogated the IL1ß-/- tumor suppressive effect, as did crossing IL1ß-/- mice to SCID or Rag1-/- mice. Finally, blockade of IL1ß synergized with blockade of PD-1 to inhibit tumor growth in WT mice. These results suggest that IL1ß promotes tumor growth, whereas IL1α inhibits tumor growth by enhancing T-cell-mediated antitumor immunity.


Asunto(s)
Inmunidad Adaptativa , Anticuerpos Monoclonales/farmacología , Linfocitos T CD8-positivos/inmunología , Interleucina-1alfa/inmunología , Interleucina-1beta/inmunología , Neoplasias/terapia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Neoplasias/inmunología , Microambiente Tumoral
19.
J Cell Biol ; 158(6): 1079-87, 2002 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-12235125

RESUMEN

The transcription factor Microphthalmia-associated transcription factor (MITF) is a lineage-determination factor, which modulates melanocyte differentiation and pigmentation. MITF was recently shown to reside downstream of the canonical Wnt pathway during melanocyte differentiation from pluripotent neural crest cells in zebrafish as well as in mammalian melanocyte lineage cells. Although expression of many melanocytic/pigmentation markers is lost in human melanoma, MITF expression remains intact, even in unpigmented tumors, suggesting a role for MITF beyond its role in differentiation. A significant fraction of primary human melanomas exhibit deregulation (via aberrant nuclear accumulation) of beta-catenin, leading us to examine its role in melanoma growth and survival. Here, we show that beta-catenin is a potent mediator of growth for melanoma cells in a manner dependent on its downstream target MITF. Moreover, suppression of melanoma clonogenic growth by disruption of beta-catenin-T-cell transcription factor/LEF is rescued by constitutive MITF. This rescue occurs largely through a prosurvival mechanism. Thus, beta-catenin regulation of MITF expression represents a tissue-restricted pathway that significantly influences the growth and survival behavior of this notoriously treatment-resistant neoplasm.


Asunto(s)
Proteínas del Citoesqueleto/fisiología , Proteínas de Unión al ADN/fisiología , Regulación Neoplásica de la Expresión Génica , Melanoma/genética , Transactivadores/fisiología , Factores de Transcripción/fisiología , Animales , Apoptosis , División Celular , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/metabolismo , Factor de Unión 1 al Potenciador Linfoide , Melanoma/metabolismo , Melanoma/patología , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Factor de Transcripción Asociado a Microftalmía , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo , Activación Transcripcional , Transfección , Células Tumorales Cultivadas , beta Catenina
20.
Clin Cancer Res ; 14(23): 7726-32, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19047099

RESUMEN

PURPOSE: Melanoma subtypes based on anatomic location and UV light exposure can be further classified based on genetic alterations recently identified. Mutations and gene amplification in KIT have been described in a significant percentage of mucosal and acral melanomas. We recently reported a patient with metastatic mucosal melanoma harboring a known KIT mutation treated with imatinib mesylate who experienced a major response. Biological effects of KIT inhibition in these melanomas remain poorly understood. We sought to investigate further the effects of imatinib in these melanoma subsets. EXPERIMENTAL DESIGN: Mucosal melanoma cells were analyzed for KIT aberrations by genomic sequencing, quantitative PCR, and single nucleotide polymorphism analyses. Imatinib effects were assayed by viability measurements and apoptotic cytotoxicity. Tumor cell lysates were assayed by Western blots to determine effects on multiple signaling pathways after imatinib exposure. RESULTS: Mucosal melanoma cells exhibited imatinib sensitivity correlating with KIT mutational status. Imatinib dramatically decreased proliferation and was cytotoxic to a KIT mutated and amplified cell culture. Exposure to drug affected the mitogen-activated protein kinase, phosphatidylinositol 3-kinase/AKT, JAK-STAT, and antiapoptotic pathways. CONCLUSIONS: Rational targeting of KIT in melanoma offers a unique and potent clinical opportunity. In vitro analyses revealed major sensitivity to KIT kinase inhibition by imatinib, with potent induction of melanoma cell apoptosis. Biochemical studies identified changes in signaling molecules regulating proliferation and survival responses, which may serve as mediators and/or biomarkers of in vivo treatment efficacy. Pathways affected by KIT inhibition provide a model for understanding components in effective melanoma cell death and insights into targeting for resistance mechanisms.


Asunto(s)
Resistencia a Antineoplásicos/genética , Melanoma/genética , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-kit/genética , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Antineoplásicos/farmacología , Secuencia de Bases , Benzamidas , Western Blotting , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Mesilato de Imatinib , Melanoma/tratamiento farmacológico , Membrana Mucosa/patología , Mutación , Polimorfismo de Nucleótido Simple , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA