Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
PLoS One ; 18(4): e0269324, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37011054

RESUMEN

INTRODUCTION: We are conducting a multicenter study to identify classifiers predictive of disease-specific survival in patients with primary melanomas. Here we delineate the unique aspects, challenges, and best practices for optimizing a study of generally small-sized pigmented tumor samples including primary melanomas of at least 1.05mm from AJTCC TNM stage IIA-IIID patients. We also evaluated tissue-derived predictors of extracted nucleic acids' quality and success in downstream testing. This ongoing study will target 1,000 melanomas within the international InterMEL consortium. METHODS: Following a pre-established protocol, participating centers ship formalin-fixed paraffin embedded (FFPE) tissue sections to Memorial Sloan Kettering Cancer Center for the centralized handling, dermatopathology review and histology-guided coextraction of RNA and DNA. Samples are distributed for evaluation of somatic mutations using next gen sequencing (NGS) with the MSK-IMPACTTM assay, methylation-profiling (Infinium MethylationEPIC arrays), and miRNA expression (Nanostring nCounter Human v3 miRNA Expression Assay). RESULTS: Sufficient material was obtained for screening of miRNA expression in 683/685 (99%) eligible melanomas, methylation in 467 (68%), and somatic mutations in 560 (82%). In 446/685 (65%) cases, aliquots of RNA/DNA were sufficient for testing with all three platforms. Among samples evaluated by the time of this analysis, the mean NGS coverage was 249x, 59 (18.6%) samples had coverage below 100x, and 41/414 (10%) failed methylation QC due to low intensity probes or insufficient Meta-Mixed Interquartile (BMIQ)- and single sample (ss)- Noob normalizations. Six of 683 RNAs (1%) failed Nanostring QC due to the low proportion of probes above the minimum threshold. Age of the FFPE tissue blocks (p<0.001) and time elapsed from sectioning to co-extraction (p = 0.002) were associated with methylation screening failures. Melanin reduced the ability to amplify fragments of 200bp or greater (absent/lightly pigmented vs heavily pigmented, p<0.003). Conversely, heavily pigmented tumors rendered greater amounts of RNA (p<0.001), and of RNA above 200 nucleotides (p<0.001). CONCLUSION: Our experience with many archival tissues demonstrates that with careful management of tissue processing and quality control it is possible to conduct multi-omic studies in a complex multi-institutional setting for investigations involving minute quantities of FFPE tumors, as in studies of early-stage melanoma. The study describes, for the first time, the optimal strategy for obtaining archival and limited tumor tissue, the characteristics of the nucleic acids co-extracted from a unique cell lysate, and success rate in downstream applications. In addition, our findings provide an estimate of the anticipated attrition that will guide other large multicenter research and consortia.


Asunto(s)
Melanoma , MicroARNs , Ácidos Nucleicos , Humanos , Fijación del Tejido/métodos , MicroARNs/análisis , Melanoma/genética , ADN/genética , Adhesión en Parafina/métodos , Formaldehído
2.
Clin Cancer Res ; 27(10): 2678-2697, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33414132

RESUMEN

Five years ago, the Melanoma Research Foundation (MRF) conducted an assessment of the challenges and opportunities facing the melanoma research community and patients with melanoma. Since then, remarkable progress has been made on both the basic and clinical research fronts. However, the incidence, recurrence, and death rates for melanoma remain unacceptably high and significant challenges remain. Hence, the MRF Scientific Advisory Council and Breakthrough Consortium, a group that includes clinicians and scientists, reconvened to facilitate intensive discussions on thematic areas essential to melanoma researchers and patients alike, prevention, detection, diagnosis, metastatic dormancy and progression, response and resistance to targeted and immune-based therapy, and the clinical consequences of COVID-19 for patients with melanoma and providers. These extensive discussions helped to crystalize our understanding of the challenges and opportunities facing the broader melanoma community today. In this report, we discuss the progress made since the last MRF assessment, comment on what remains to be overcome, and offer recommendations for the best path forward.


Asunto(s)
COVID-19/prevención & control , Oncología Médica/métodos , Melanoma/terapia , Guías de Práctica Clínica como Asunto , SARS-CoV-2/aislamiento & purificación , Neoplasias Cutáneas/terapia , Investigación Biomédica/métodos , Investigación Biomédica/tendencias , COVID-19/epidemiología , COVID-19/virología , Humanos , Oncología Médica/organización & administración , Oncología Médica/tendencias , Melanoma/diagnóstico , SARS-CoV-2/fisiología , Neoplasias Cutáneas/diagnóstico
3.
Nucleic Acids Res ; 48(7): 3789-3805, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-31980816

RESUMEN

By interacting with proteins and nucleic acids, the vast family of mammalian circRNAs is proposed to influence many biological processes. Here, RNA sequencing analysis of circRNAs differentially expressed during myogenesis revealed that circSamd4 expression increased robustly in mouse C2C12 myoblasts differentiating into myotubes. Moreover, silencing circSamd4, which is conserved between human and mouse, delayed myogenesis and lowered the expression of myogenic markers in cultured myoblasts from both species. Affinity pulldown followed by mass spectrometry revealed that circSamd4 associated with PURA and PURB, two repressors of myogenesis that inhibit transcription of the myosin heavy chain (MHC) protein family. Supporting the hypothesis that circSamd4 might complex with PUR proteins and thereby prevent their interaction with DNA, silencing circSamd4 enhanced the association of PUR proteins with the Mhc promoter, while overexpressing circSamd4 interfered with the binding of PUR proteins to the Mhc promoter. These effects were abrogated when using a mutant circSamd4 lacking the PUR binding site. Our results indicate that the association of PUR proteins with circSamd4 enhances myogenesis by contributing to the derepression of MHC transcription.


Asunto(s)
Regulación de la Expresión Génica , Desarrollo de Músculos/genética , ARN Circular/metabolismo , Proteínas Represoras/metabolismo , Transcripción Genética , Animales , Sitios de Unión , Diferenciación Celular , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Humanos , Ratones , Mioblastos/citología , Mioblastos/metabolismo , Cadenas Pesadas de Miosina/biosíntesis , Cadenas Pesadas de Miosina/genética , Proteínas del Tejido Nervioso/metabolismo , ARN Circular/química , Factores de Transcripción/metabolismo
4.
Cancer Cell ; 37(1): 55-70.e15, 2020 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-31935372

RESUMEN

Metastasis is the primary cause of death of cancer patients. Dissecting mechanisms governing metastatic spread may uncover important tumor biology and/or yield promising therapeutic insights. Here, we investigated the role of circular RNAs (circRNA) in metastasis, using melanoma as a model aggressive tumor. We identified silencing of cerebellar degeneration-related 1 antisense (CDR1as), a regulator of miR-7, as a hallmark of melanoma progression. CDR1as depletion results from epigenetic silencing of LINC00632, its originating long non-coding RNA (lncRNA) and promotes invasion in vitro and metastasis in vivo through a miR-7-independent, IGF2BP3-mediated mechanism. Moreover, CDR1as levels reflect cellular states associated with distinct therapeutic responses. Our study reveals functional, prognostic, and predictive roles for CDR1as and expose circRNAs as key players in metastasis.


Asunto(s)
Autoantígenos/genética , Epigénesis Genética , Silenciador del Gen , Melanoma/patología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Proteínas de Unión al ARN/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/genética , Humanos , MicroARNs/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Pronóstico , ARN sin Sentido/genética , ARN Circular/genética , ARN Largo no Codificante/genética , Proteínas de Unión al ARN/genética
5.
Pigment Cell Melanoma Res ; 33(3): 466-479, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31663663

RESUMEN

Next-generation sequencing has enabled genetic and genomic characterization of melanoma to an unprecedent depth. However, the high mutational background plus the limited depth of coverage of whole-genome sequencing performed on cutaneous melanoma samples make the identification of novel driver mutations difficult. We sought to explore the somatic mutation portfolio in exonic and gene regulatory regions in human melanoma samples, for which we performed targeted sequencing of tumors and matched germline DNA samples from 89 melanoma patients, identifying known and novel recurrent mutations. Two recurrent mutations found in the RPS27 promoter associated with decreased RPS27 mRNA levels in vitro. Data mining and IHC analyses revealed a bimodal pattern of RPS27 expression in melanoma, with RPS27-low patients displaying worse prognosis. In vitro characterization of RPS27-high and RPS27-low melanoma cell lines, as well as loss-of-function experiments, demonstrated that high RPS27 status provides increased proliferative and invasive capacities, while low RPS27 confers survival advantage in low attachment and resistance to therapy. Additionally, we demonstrate that 10 other cancer types harbor bimodal RPS27 expression, and in those, similarly to melanoma, RPS27-low expression associates with worse clinical outcomes. RPS27 promoter mutation could thus represent a mechanism of gene expression modulation in melanoma patients, which may have prognostic and predictive implications.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Melanoma/genética , Metaloproteínas/genética , Mutación/genética , Proteínas Nucleares/genética , Proteínas de Unión al ARN/genética , Proteínas Ribosómicas/genética , Animales , Antineoplásicos/farmacología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Sitios Genéticos , Genómica , Humanos , Metaloproteínas/metabolismo , Ratones , Invasividad Neoplásica , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Ribosómicas/metabolismo
6.
Pigment Cell Melanoma Res ; 30(3): 328-338, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28140520

RESUMEN

Melanoma patients with BRAFV600E -mutant tumors display striking responses to BRAF inhibitors (BRAFi); however, almost all invariably relapse with drug-resistant disease. Here, we report that microRNA-125a (miR-125a) expression is upregulated in human melanoma cells and patient tissues upon acquisition of BRAFi resistance. We show that miR-125a induction confers resistance to BRAFV600E melanoma cells to BRAFi by directly suppressing pro-apoptotic components of the intrinsic apoptosis pathway, including BAK1 and MLK3. Apoptotic suppression and prolonged survival favor reactivation of the MAPK and AKT pathways by drug-resistant melanoma cells. We demonstrate that miR-125a inhibition suppresses the emergence of resistance to BRAFi and, in a subset of resistant melanoma cell lines, leads to partial drug resensitization. Finally, we show that miR-125a upregulation is mediated by TGFß signaling. In conclusion, the identification of this novel role for miR-125a in BRAFi resistance exposes clinically relevant mechanisms of melanoma cell survival that can be exploited therapeutically.


Asunto(s)
Apoptosis/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , MicroARNs/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Apoptosis/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Melanoma/tratamiento farmacológico , Melanoma/genética , MicroARNs/genética , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo
7.
Sci Signal ; 9(457): ra118, 2016 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-27923913

RESUMEN

Maintenance of mammary functional capacity during cycles of proliferation and regression depends on appropriate cell fate decisions of mammary progenitor cells to populate an epithelium consisting of secretory luminal cells and contractile myoepithelial cells. It is well established that transforming growth factor-ß (TGFß) restricts mammary epithelial cell proliferation and that sensitivity to TGFß is decreased in breast cancer. We show that TGFß also exerts control of mammary progenitor self-renewal and lineage commitment decisions by stringent regulation of breast cancer associated 1 (BRCA1), which controls stem cell self-renewal and lineage commitment. Either genetic depletion of Tgfb1 or transient blockade of TGFß increased self-renewal of mammary progenitor cells in mice, cultured primary mammary epithelial cells, and also skewed lineage commitment toward the myoepithelial fate. TGFß stabilized the abundance of BRCA1 by reducing the abundance of microRNA-182 (miR-182). Ectopic expression of BRCA1 or antagonism of miR-182 in cultured TGFß-deficient mammary epithelial cells restored luminal lineage commitment. These findings reveal that TGFß modulation of BRCA1 directs mammary epithelial cell fate and, because stem or progenitor cells are thought to be the cell of origin for aggressive breast cancer subtypes, suggest that TGFß dysregulation during tumorigenesis may promote distinct breast cancer subtypes.


Asunto(s)
Diferenciación Celular , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/metabolismo , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Animales , Proteína BRCA1 , Femenino , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/patología , Ratones , Ratones Endogámicos BALB C , MicroARNs/genética , Células Madre Neoplásicas/patología , Factor de Crecimiento Transformador beta1/genética , Proteínas Supresoras de Tumor/genética
8.
Cancer Med ; 5(6): 1183-93, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27169980

RESUMEN

Despite major advances in the treatment of metastatic melanoma, treatment failure is still inevitable in most cases. Manipulation of key epigenetic regulators, including inhibition of Bromodomain and extra-terminal domain (BET) family members impairs cell proliferation in vitro and tumor growth in vivo in different cancers, including melanoma. Here, we investigated the effect of combining the BET inhibitor JQ1 with the BRAF inhibitor Vemurafenib in in vitro and in vivo models of BRAF-mutant melanoma. We performed cytotoxicity and apoptosis assays, and a xenograft mouse model to determine the in vitro and in vivo efficacy of JQ1 in combination with Vemurafenib against BRAF-mutant melanoma cell lines. Further, to investigate the molecular mechanisms underlying the effects of combined treatment, we conducted antibody arrays of in vitro drug-treated cell lines and RNA sequencing of drug-treated xenograft tumors. The combination of JQ1 and Vemurafenib acted synergistically in BRAF-mutant cell lines, resulting in marked apoptosis in vitro, with upregulation of proapoptotic proteins. In vivo, combination treatment suppressed tumor growth and significantly improved survival compared to either drug alone. RNA sequencing of tumor tissues revealed almost four thousand genes that were uniquely modulated by the combination, with several anti-apoptotic genes significantly down-regulated. Collectively, our data provide a rationale for combined BET and BRAF inhibition as a novel strategy for the treatment of melanoma.


Asunto(s)
Melanoma/genética , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Azepinas/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Humanos , Indoles/farmacología , Melanoma/tratamiento farmacológico , Melanoma/patología , Sulfonamidas/farmacología , Transcripción Genética/efectos de los fármacos , Triazoles/farmacología , Vemurafenib , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Cancer ; 121(23): 4108-23, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26308244

RESUMEN

The American Joint Committee on Cancer staging system for cutaneous melanoma is based on primary tumor thickness and the presence of ulceration, mitoses, lymph node spread, and distant metastases as determinants of prognosis. Although this cutaneous melanoma staging system has evolved over time to more accurately reflect patient prognosis, improvements are still needed, because current understanding of the particular factors (genetic mutation, expression alteration, host response, etc) that are critical for predicting patient outcomes is incomplete. Given the clinical and biologic heterogeneity of primary melanomas, new prognostic tools are needed to more precisely identify patients who are most likely to develop advanced disease. Such tools would affect clinical surveillance strategies and aid in patient selection for adjuvant therapy. The authors reviewed the literature on prognostic molecular and immunologic markers in primary cutaneous melanoma, their associations with clinicopathologic and survival outcomes, and their potential for incorporation into current staging models. Overall, the studies considered in this review did not define prognostic markers that could be readily incorporated into the current staging system. Therefore, efforts should be continued in these and other directions to maximize the likelihood of identifying clinically useful prognostic biomarkers for cutaneous melanoma.


Asunto(s)
Biomarcadores de Tumor/genética , Biomarcadores de Tumor/inmunología , Melanoma/patología , Neoplasias Cutáneas/patología , Humanos , Melanoma/genética , Melanoma/inmunología , Estadificación de Neoplasias , Pronóstico , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Análisis de Supervivencia
10.
J Invest Dermatol ; 132(7): 1860-8, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22551973

RESUMEN

We examined the microRNA signature that distinguishes the most common melanoma histological subtypes, superficial spreading melanoma (SSM) and nodular melanoma (NM). We also investigated the mechanisms underlying the differential expression of histology-specific microRNAs. MicroRNA array performed on a training cohort of 82 primary melanoma tumors (26 SSM, 56 NM), and nine congenital nevi (CN) revealed 134 microRNAs differentially expressed between SSM and NM (P<0.05). Out of 134 microRNAs, 126 remained significant after controlling for thickness and 31 were expressed at a lower level in SSM compared with both NM and CN. For seven microRNAs (let-7g, miR-15a, miR-16, miR-138, miR-181a, miR-191, and miR-933), the downregulation was associated with selective genomic loss in SSM cell lines and primary tumors, but not in NM cell lines and primary tumors. The lower expression level of six out of seven microRNAs in SSM compared with NM was confirmed by real-time PCR on a subset of cases in the training cohort and validated in an independent cohort of 97 melanoma cases (38 SSM, 59 NM). Our data support a molecular classification in which SSM and NM are two molecularly distinct phenotypes. Therapeutic strategies that take into account subtype-specific alterations might improve the outcome of melanoma patients.


Asunto(s)
Melanoma/genética , MicroARNs/análisis , Adulto , Anciano , Línea Celular Tumoral , Femenino , Humanos , Modelos Lineales , Masculino , Melanoma/patología , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Estudios Prospectivos
11.
Cancer Cell ; 20(1): 104-18, 2011 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-21741600

RESUMEN

To metastasize, a tumor cell must acquire abilities such as the capacity to colonize new tissue and evade immune surveillance. Recent evidence suggests that microRNAs can promote the evolution of malignant behaviors by regulating multiple targets. We performed a microRNA analysis of human melanoma, a highly invasive cancer, and found that miR-30b/30d upregulation correlates with stage, metastatic potential, shorter time to recurrence, and reduced overall survival. Ectopic expression of miR-30b/30d promoted the metastatic behavior of melanoma cells by directly targeting the GalNAc transferase GALNT7, resulted in increased synthesis of the immunosuppressive cytokine IL-10, and reduced immune cell activation and recruitment. These data support a key role of miR-30b/30d and GalNAc transferases in metastasis, by simultaneously promoting cellular invasion and immunosuppression.


Asunto(s)
Tolerancia Inmunológica/genética , MicroARNs/metabolismo , N-Acetilgalactosaminiltransferasas/metabolismo , Animales , Secuencia de Bases , Proliferación Celular , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Genoma Humano/genética , Humanos , Interleucina-10/metabolismo , Melanoma/enzimología , Melanoma/genética , Melanoma/patología , Ratones , MicroARNs/genética , Datos de Secuencia Molecular , N-Acetilgalactosaminiltransferasas/antagonistas & inhibidores , N-Acetilgalactosaminiltransferasas/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Polisacáridos/metabolismo , Regulación hacia Arriba , Polipéptido N-Acetilgalactosaminiltransferasa
12.
Pigment Cell Melanoma Res ; 24(1): 197-206, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20883453

RESUMEN

In this study, we investigated the mechanism(s) of altered expression of protooncogene SKP2 in metastatic melanoma and its clinical relevance in patients with metastatic melanoma. The genomic status of SKP2 was assessed in cell lines by sequencing, single nucleotide polymorphism array, and genomic PCR. Copy number status was then evaluated for concordance with SKP2 mRNA and protein expression. SKP2 protein was further evaluated by immunohistochemistry in 93 human metastatic tissues. No mutations were identified in SKP2. Increased copy number at the SKP2 locus was observed in 6/14 (43%) metastatic cell lines and in 9/22 (41%) human metastatic tissues which was associated with overexpression of SKP2 protein. Overexpression of SKP2 protein in human tissues was associated with worse survival in a multivariate model controlling for the site of metastasis. Copy number gain is a major contributing mechanism of SKP2 overexpression in metastatic melanoma. Results may have implications for the development of therapeutics that target SKP2.


Asunto(s)
Melanoma/genética , Melanoma/patología , Proteínas Quinasas Asociadas a Fase-S/genética , Proteínas Cdh1 , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Dosificación de Gen/genética , Regulación Neoplásica de la Expresión Génica , Sitios Genéticos/genética , Humanos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Nucleótidos/genética , Sistemas de Lectura Abierta/genética , Polimorfismo de Nucleótido Simple/genética , Recurrencia , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Supervivencia
13.
Clin Cancer Res ; 16(5): 1577-86, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20179230

RESUMEN

PURPOSE: To identify a melanoma microRNA (miRNA) expression signature that is predictive of outcome and then evaluate its potential to improve risk stratification when added to the standard-of-care staging criteria. EXPERIMENTAL DESIGN: Total RNA was extracted from 59 formalin-fixed paraffin-embedded melanoma metastases and hybridized to miRNA arrays containing 911 probes. We then correlated miRNA expression with post-recurrence survival and other clinicopathologic criteria. RESULTS: We identified a signature of 18 miRNAs whose overexpression was significantly correlated with longer survival, defined as more than 18 months post-recurrence survival. Subsequent cross-validation showed that a small subset of these miRNAs can predict post-recurrence survival in metastatic melanoma with an estimated accuracy of 80.2% (95% confidence interval, 79.8-80.6%). In contrast to standard-of-care staging criteria, a six-miRNA signature significantly stratified stage III patients into "better" and "worse" prognostic categories, and a multivariate Cox regression analysis revealed the signature to be an independent predictor of survival. Furthermore, we showed that most miRNAs from the signature also showed differential expression between patients with better and worse prognoses in the corresponding paired primary melanoma. CONCLUSIONS: MiRNA signatures have potential as clinically relevant biomarkers of prognosis in metastatic melanoma. Our data suggest that molecularly based models of risk assessment can improve the standard staging criteria and support the incorporation of miRNAs into such models.


Asunto(s)
Melanoma/genética , MicroARNs/análisis , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/mortalidad , Neoplasias Cutáneas/genética , Biomarcadores de Tumor/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Masculino , Melanoma/mortalidad , Melanoma/patología , MicroARNs/genética , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/patología
14.
Proc Natl Acad Sci U S A ; 106(6): 1814-9, 2009 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-19188590

RESUMEN

The highly aggressive character of melanoma makes it an excellent model for probing the mechanisms underlying metastasis, which remains one of the most difficult challenges in treating cancer. We find that miR-182, member of a miRNA cluster in a chromosomal locus (7q31-34) frequently amplified in melanoma, is commonly up-regulated in human melanoma cell lines and tissue samples; this up-regulation correlates with gene copy number in a subset of melanoma cell lines. Moreover, miR-182 ectopic expression stimulates migration of melanoma cells in vitro and their metastatic potential in vivo, whereas miR-182 down-regulation impedes invasion and triggers apoptosis. We further show that miR-182 over-expression promotes migration and survival by directly repressing microphthalmia-associated transcription factor-M and FOXO3, whereas enhanced expression of either microphthalmia-associated transcription factor-M or FOXO3 blocks miR-182's proinvasive effects. In human tissues, expression of miR-182 increases with progression from primary to metastatic melanoma and inversely correlates with FOXO3 and microphthalmia-associated transcription factor levels. Our data provide a mechanism for invasion and survival in melanoma that could prove applicable to metastasis of other cancers and suggest that miRNA silencing may be a worthwhile therapeutic strategy.


Asunto(s)
Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica , Melanoma/patología , MicroARNs/genética , Factor de Transcripción Asociado a Microftalmía/genética , Metástasis de la Neoplasia/genética , Animales , Movimiento Celular , Supervivencia Celular , Progresión de la Enfermedad , Proteína Forkhead Box O3 , Humanos , Ratones , MicroARNs/fisiología , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...