Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Pathol ; 259(1): 56-68, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36219477

RESUMEN

Melanoma is a heterogenous malignancy with an unpredictable clinical course. Most patients who present in the clinic are diagnosed with primary melanoma, yet large-scale sequencing efforts have focused primarily on metastatic disease. In this study we sequence-profiled 524 American Joint Committee on Cancer Stage I-III primary tumours. Our analysis of these data reveals recurrent driver mutations, mutually exclusive genetic interactions, where two genes were never or rarely co-mutated, and an absence of co-occurring genetic events. Further, we intersected copy number calls from our primary melanoma data with whole-genome CRISPR screening data to identify the transcription factor interferon regulatory factor 4 (IRF4) as a melanoma-associated dependency. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Melanoma , Humanos , Mutación , Melanoma/genética , Genoma , Genómica , Reino Unido
2.
Pharmacol Res ; 173: 105911, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34560251

RESUMEN

In melanomas, therapy resistance can arise due to a combination of genetic, epigenetic and phenotypic mechanisms. Due to its crucial role in DNA supercoil relaxation, TOP1 is often considered an essential chemotherapeutic target in cancer. However, how TOP1 expression and activity might differ in therapy sensitive versus resistant cell types is unknown. Here we show that TOP1 expression is increased in metastatic melanoma and correlates with an invasive gene expression signature. More specifically, TOP1 expression is highest in cells with the lowest expression of MITF, a key regulator of melanoma biology. Notably, TOP1 and DNA Single-Strand Break Repair genes are downregulated in BRAFi- and BRAFi/MEKi-resistant cells and TOP1 inhibition decreases invasion markers only in BRAFi/MEKi-resistant cells. Thus, we show three different phenotypes related to TOP1 levels: i) non-malignant cells with low TOP1 levels; ii) metastatic cells with high TOP1 levels and high invasiveness; and iii) BRAFi- and BRAFi/MEKi-resistant cells with low TOP1 levels and high invasiveness. Together, these results highlight the potential role of TOP1 in melanoma progression and resistance.


Asunto(s)
ADN-Topoisomerasas de Tipo I , Resistencia a Antineoplásicos , Melanoma , Neoplasias Cutáneas , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , ADN-Topoisomerasas de Tipo I/genética , ADN-Topoisomerasas de Tipo I/metabolismo , Progresión de la Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/metabolismo , Melanoma/mortalidad , Persona de Mediana Edad , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/mortalidad
3.
J Transl Med ; 18(1): 56, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-32024530

RESUMEN

BACKGROUND: Interaction between malignant cells and immune cells that reside within the tumor microenvironment (TME) modulate different aspects of tumor development and progression. Recent works showed the importance of miRNA-containing extracellular vesicles in this crosstalk. METHODS: Interested in understanding the interplay between melanoma and immune-related TME cells, we characterized the TCGA's metastatic melanoma samples according to their tumor microenvironment profiles, HLA-I neoepitopes, transcriptome profile and classified them into three groups. Moreover, we combined our results with melanoma single-cell gene expression and public miRNA data to better characterize the regulatory network of circulating miRNAs and their targets related to immune evasion and microenvironment response. RESULTS: The group associated with a worse prognosis showed phenotypic characteristics that favor immune evasion, including a strong signature of suppressor cells and less stable neoantigen:HLA-I complexes. Conversely, the group with better prognosis was marked by enrichment in lymphocyte and MHC signatures. By analyzing publicly available melanoma single-cell RNA and microvesicle microRNAs sequencing data we identified circulating microRNAs potentially involved in the crosstalk between tumor and TME cells. Candidate miRNA/target gene pairs with previously reported roles in tumor progression and immune escape mechanisms were further investigated and demonstrated to impact patient's overall survival not only in melanoma but across different tumor types. CONCLUSION: Our results underscore the impact of tumor-microenvironment interactions on disease outcomes and reveal potential non-invasive biomarkers of prognosis and treatment response.


Asunto(s)
Melanoma , MicroARNs , Humanos , Melanoma/genética , MicroARNs/genética , Pronóstico , Transcriptoma , Microambiente Tumoral
4.
Genes Dev ; 26(10): 1055-69, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22549727

RESUMEN

Human melanocytic nevi (moles) are benign lesions harboring activated oncogenes, including BRAF. Although this oncogene initially acts mitogenically, eventually, oncogene-induced senescence (OIS) ensues. Nevi can infrequently progress to melanomas, but the mechanistic relationship with OIS is unclear. We show here that PTEN depletion abrogates BRAF(V600E)-induced senescence in human fibroblasts and melanocytes. Correspondingly, in established murine BRAF(V600E)-driven nevi, acute shRNA-mediated depletion of PTEN prompted tumor progression. Furthermore, genetic analysis of laser-guided microdissected human contiguous nevus-melanoma specimens recurrently revealed identical mutations in BRAF or NRAS in adjacent benign and malignant melanocytes. The PI3K pathway was often activated through either decreased PTEN or increased AKT3 expression in melanomas relative to their adjacent nevi. Pharmacologic PI3K inhibition in melanoma cells suppressed proliferation and induced the senescence-associated tumor suppressor p15(INK4B). This treatment also eliminated subpopulations resistant to targeted BRAF(V600E) inhibition. Our findings suggest that a significant proportion of melanomas arise from nevi. Furthermore, these results demonstrate that PI3K pathway activation serves as a rate-limiting event in this setting, acting at least in part by abrogating OIS. The reactivation of senescence features and elimination of cells refractory to BRAF(V600E) inhibition by PI3K inhibition warrants further investigation into the therapeutic potential of simultaneously targeting these pathways in melanoma.


Asunto(s)
Senescencia Celular , Melanoma/patología , Nevo/patología , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias Cutáneas/patología , Sustitución de Aminoácidos , Proliferación Celular , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/metabolismo , Activación Enzimática , Fibroblastos/metabolismo , Fibroblastos/patología , Ácido Glutámico/genética , Ácido Glutámico/metabolismo , Humanos , Melanocitos/metabolismo , Melanocitos/patología , Melanoma/metabolismo , Nevo/metabolismo , Fosfohidrolasa PTEN/genética , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias Cutáneas/metabolismo , Valina/genética , Valina/metabolismo
5.
Biochem Biophys Res Commun ; 504(1): 270-276, 2018 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-30172372

RESUMEN

Protease-activated receptor 2 (PAR2) is a G-protein coupled receptor which is activated upon cleavage of its N-terminal region. PAR2 has been associated with many aspects regarding tumor progression, such as the production of pro-tumoral cytokines. Granulocyte colony-stimulating factor (G-CSF) is a cytokine essential to neutrophil production and maturation, and it is often overexpressed in tumors. In this study, we evaluated the ability of PAR2 to modulate G-CSF expression. PAR2 and G-CSF were significantly more expressed in metastatic (4T1 and MDA-MB-231) as compared to non-metastatic (67NR and MCF7) breast cancer cell lines. In addition, PAR2 stimulation by a synthetic agonist peptide significantly increased G-CSF gene expression in the metastatic cell lines. Knockdown of PAR2 in 4T1 cells decreased G-CSF expression and secretion. In addition, treatment of 4T1 with the commercial PAR2 antagonist, ENMD-1068, significantly decreased G-CSF expression. cBioPortal analyses of the TCGA database showed a significant co-occurrence of G-CSF and PAR2 gene overexpression in breast cancer samples. In conclusion, our data suggest that PAR2 contributes to G-CSF expression in breast cancer cells, possibly favoring tumor progression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos/metabolismo , Receptor PAR-2/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Línea Celular Tumoral , Citocinas/metabolismo , Femenino , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Ratones , Activación Transcripcional , Regulación hacia Arriba
6.
J Proteome Res ; 14(10): 4332-41, 2015 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-26317507

RESUMEN

Protein phosphorylation is an essential post-translational modification (PTM) regulating many biological processes at the cellular and multicellular level. Continuous improvements in phosphoproteomics technology allow the analysis of this PTM in an expanding biological content, yet up until now proteome data visualization tools are still very gene centric, hampering the ability to comprehensively map and study PTM dynamics. Here we present PhosphoPath, a Cytoscape app designed for the visualization and analysis of quantitative proteome and phosphoproteome data sets. PhosphoPath brings knowledge into the biological network by importing publically available data and enables PTM site-specific visualization of information from quantitative time series. To showcase PhosphoPath performance we use a quantitative proteomics data set comparing patient-derived melanoma cell lines grown in either conventional cell culture or xenografts.


Asunto(s)
Melanoma/metabolismo , Aplicaciones Móviles , Fosfoproteínas/metabolismo , Procesamiento Proteico-Postraduccional , Proteoma/metabolismo , Neoplasias Cutáneas/metabolismo , Línea Celular Tumoral , Gráficos por Computador , Redes Reguladoras de Genes , Humanos , Melanoma/genética , Melanoma/patología , Fosfoproteínas/genética , Fosfoproteínas/aislamiento & purificación , Fosforilación , Proteoma/genética , Proteoma/aislamiento & purificación , Proteómica , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Espectrometría de Masas en Tándem
7.
Mol Syst Biol ; 10: 772, 2014 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-25538140

RESUMEN

Treatment of BRAF mutant melanomas with specific BRAF inhibitors leads to tumor remission. However, most patients eventually relapse due to drug resistance. Therefore, we designed an integrated strategy using (phospho)proteomic and functional genomic platforms to identify drug targets whose inhibition sensitizes melanoma cells to BRAF inhibition. We found many proteins to be induced upon PLX4720 (BRAF inhibitor) treatment that are known to be involved in BRAF inhibitor resistance, including FOXD3 and ErbB3. Several proteins were down-regulated, including Rnd3, a negative regulator of ROCK1 kinase. For our genomic approach, we performed two parallel shRNA screens using a kinome library to identify genes whose inhibition sensitizes to BRAF or ERK inhibitor treatment. By integrating our functional genomic and (phospho)proteomic data, we identified ROCK1 as a potential drug target for BRAF mutant melanoma. ROCK1 silencing increased melanoma cell elimination when combined with BRAF or ERK inhibitor treatment. Translating this to a preclinical setting, a ROCK inhibitor showed augmented melanoma cell death upon BRAF or ERK inhibition in vitro. These data merit exploration of ROCK1 as a target in combination with current BRAF mutant melanoma therapies.


Asunto(s)
Melanoma/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Quinasas Asociadas a rho/metabolismo , Línea Celular Tumoral , Cromatografía Liquida , Regulación hacia Abajo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Humanos , Indazoles/farmacología , Indoles/farmacología , Terapia Molecular Dirigida , Mutación , Piperazinas/farmacología , Proteómica , Proteínas Proto-Oncogénicas B-raf/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptor ErbB-3/genética , Receptor ErbB-3/metabolismo , Sulfonamidas/farmacología , Espectrometría de Masas en Tándem , Vemurafenib , Quinasas Asociadas a rho/genética
8.
Neoplasia ; 46: 100947, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37913653

RESUMEN

Acral melanoma is a rare subtype of melanoma that arises on the non-hair bearing skin of the nail bed, palms of the hand and soles of the feet. It is unique among melanomas in not being linked to ultraviolet radiation (UVR) exposure from the sun, and, as such, its incidence is similar across populations who are of Asian, Hispanic, African and European origin. Although research into acral melanoma has lagged behind that of sun-exposed cutaneous melanoma, recent studies have begun to address the unique genetics and immune features of acral melanoma. In this review we will discuss the latest progress in understanding the biology of acral melanoma across different ethnic populations and will outline how these new discoveries can help to guide the therapeutic management of this rare tumor.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Humanos , Melanoma/genética , Melanoma/tratamiento farmacológico , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/tratamiento farmacológico , Rayos Ultravioleta/efectos adversos , Genómica , Melanoma Cutáneo Maligno
9.
J Pathol Clin Res ; 7(6): 531-541, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34213090

RESUMEN

Acral melanoma (AM) is a malignant cutaneous melanocytic tumour specifically located on the palms, soles, and nail apparatus, which are areas of glabrous (hairless) skin. Acral lentiginous melanoma, a subtype of AM, represents a histopathological subtype diagnosis of cutaneous melanoma with unique morphological and structural features. Despite clear definitions, the misuse of these terms and the inconsistency in reporting the histopathological features of AM cases have become a major obstacle to the study of the disease. In this review, we discuss the epidemiology, histopathological features, prognosis, and genetic profile of AM, highlighting the differences observed when histopathological subtypes are considered. The increasing global effort to characterise AM cases from ethnically diverse populations would benefit greatly from a more consistent classification of the disease.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Animales , Biomarcadores de Tumor/genética , Pie/patología , Mano/patología , Humanos , Melanoma/epidemiología , Melanoma/genética , Melanoma/patología , Uñas/patología , Pronóstico , Neoplasias Cutáneas/epidemiología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología
10.
Pigment Cell Melanoma Res ; 34(1): 59-71, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32330367

RESUMEN

Acral lentiginous melanoma is a histological subtype of cutaneous melanoma that occurs in the glabrous skin of the palms, soles and the nail unit. Although in some countries, particularly in Latin America, Africa and Asia, it represents the most frequently diagnosed subtype of the disease, it only represents a small proportion of melanoma cases in European-descent populations, which is partially why it has not been studied to the same extent as other forms of melanoma. As a result, its unique genomic drivers remain comparatively poorly explored, as well as its causes, with current evidence supporting a UV-independent path to tumorigenesis. In this review, we discuss current knowledge of the aetiology and diagnostic criteria of acral lentiginous melanoma, as well as its epidemiological and histopathological characteristics. We also describe what is known about the genomic landscape of this disease and review the available biological models to explore potential therapeutic targets.


Asunto(s)
Enfermedades del Pie/patología , Melanocitos/patología , Melanoma/patología , Enfermedades de la Uña/patología , Neoplasias Cutáneas/patología , Humanos
11.
J Leukoc Biol ; 108(4): 1101-1115, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32450618

RESUMEN

Melanoma is a very lethal tumor type that easily spreads and colonizes regional and distant tissues. Crucial phenotypic changes that favor melanoma metastasis are interposed by the tumor microenvironment (TME), representing a complex network in which malignant cells communicate not only with each other but also with stromal and immune cells. This cell-cell communication can be mediated by extracellular vesicles (EVs), which are lipid bilayer-delimited particles capable of carrying a wide variety of bioactive compounds. Both melanoma-derived or TME-derived EVs deliver important pro- and antitumor signals implicated in various stages of tumor progression, such as proliferation, metastasis, and treatment response. In this review, we highlight the recent advances in EV-mediated crosstalk between melanoma and immune cells and other important cells of the TME, and address different aspects of this bidirectional interaction as well as how this may hinder or trigger the development and progression of melanoma. We also discuss the potential of using EVs as biomarkers and therapeutic strategies for melanoma.


Asunto(s)
Biomarcadores de Tumor/inmunología , Comunicación Celular/inmunología , Proliferación Celular , Vesículas Extracelulares/inmunología , Melanoma/inmunología , Microambiente Tumoral/inmunología , Vesículas Extracelulares/patología , Humanos , Melanoma/patología , Melanoma/terapia , Metástasis de la Neoplasia
13.
Oncotarget ; 7(28): 42859-42872, 2016 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-27374095

RESUMEN

Breast cancer is the second most common cause of cancer-related deaths worldwide among women. Despite several therapeutic options, 15% of breast cancer patients succumb to the disease owing to tumor relapse and acquired therapy resistance. Particularly in triple-negative breast cancer (TNBC), developing effective treatments remains challenging owing to the lack of a common vulnerability that can be exploited by targeted approaches. We have previously shown that tumor cells have different requirements for growth in vivo than in vitro. Therefore, to discover novel drug targets for TNBC, we performed parallel in vivo and in vitro genetic shRNA dropout screens. We identified several potential drug targets that were required for tumor growth in vivo to a greater extent than in vitro. By combining pharmacologic inhibitors acting on a subset of these candidates, we identified a synergistic interaction between EGFR and ROCK inhibitors. This combination effectively reduced TNBC cell growth by inducing cell cycle arrest. These results illustrate the power of in vivo genetic screens and warrant further validation of EGFR and ROCK as combined pharmacologic targets for breast cancer.


Asunto(s)
Receptores ErbB/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Quinasas Asociadas a rho/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Sinergismo Farmacológico , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Células HEK293 , Humanos , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Interferencia de ARN , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
14.
Pigment Cell Melanoma Res ; 28(3): 307-17, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25728708

RESUMEN

No effective targeted therapy is currently available for NRAS mutant melanoma. Experimental MEK inhibition is rather toxic and has only limited efficacy in clinical trials. At least in part, this is caused by the emergence of drug resistance, which is commonly seen for single agent treatment and shortens clinical responses. Therefore, there is a dire need to identify effective companion drug targets for NRAS mutant melanoma. Here, we show that at concentrations where single drugs had little effect, ROCK inhibitors GSK269962A or Fasudil, in combination with either MEK inhibitor GSK1120212 (Trametinib) or ERK inhibitor SCH772984 cooperatively caused proliferation inhibition and cell death in vitro. Simultaneous inhibition of MEK and ROCK caused induction of BimEL , PARP, and Puma, and hence apoptosis. In vivo, MEK and ROCK inhibition suppressed growth of established tumors. Our findings warrant clinical investigation of the effectiveness of combinatorial targeting of MAPK/ERK and ROCK in NRAS mutant melanoma.


Asunto(s)
Apoptosis/efectos de los fármacos , GTP Fosfohidrolasas/genética , Melanoma/patología , Proteínas de la Membrana/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Mutación/genética , Quinasas Asociadas a rho/antagonistas & inhibidores , Línea Celular Tumoral , Humanos , Melanoma/enzimología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteoma/metabolismo , Quinasas Asociadas a rho/metabolismo
15.
Cell Rep ; 9(4): 1375-86, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25456132

RESUMEN

To identify factors preferentially necessary for driving tumor expansion, we performed parallel in vitro and in vivo negative-selection short hairpin RNA (shRNA) screens. Melanoma cells harboring shRNAs targeting several DNA damage response (DDR) kinases had a greater selective disadvantage in vivo than in vitro, indicating an essential contribution of these factors during tumor expansion. In growing tumors, DDR kinases were activated following hypoxia. Correspondingly, depletion or pharmacologic inhibition of DDR kinases was toxic to melanoma cells, including those that were resistant to BRAF inhibitor, and this could be enhanced by angiogenesis blockade. These results reveal that hypoxia sensitizes melanomas to targeted inhibition of the DDR and illustrate the utility of in vivo shRNA dropout screens for the identification of pharmacologically tractable targets.


Asunto(s)
Daño del ADN , Reparación del ADN , Pruebas Genéticas , Melanoma/genética , Melanoma/patología , Interferencia de ARN , Animales , Hipoxia de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Quinasa de Punto de Control 2/metabolismo , Reparación del ADN/efectos de los fármacos , Células HEK293 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/metabolismo , Estabilidad Proteica/efectos de los fármacos , Interferencia de ARN/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Reproducibilidad de los Resultados , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Nat Commun ; 5: 5712, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25502142

RESUMEN

Increased expression of the Microphthalmia-associated transcription factor (MITF) contributes to melanoma progression and resistance to BRAF pathway inhibition. Here we show that the lack of MITF is associated with more severe resistance to a range of inhibitors, while its presence is required for robust drug responses. Both in primary and acquired resistance, MITF levels inversely correlate with the expression of several activated receptor tyrosine kinases, most frequently AXL. The MITF-low/AXL-high/drug-resistance phenotype is common among mutant BRAF and NRAS melanoma cell lines. The dichotomous behaviour of MITF in drug response is corroborated in vemurafenib-resistant biopsies, including MITF-high and -low clones in a relapsed patient. Furthermore, drug cocktails containing AXL inhibitor enhance melanoma cell elimination by BRAF or ERK inhibition. Our results demonstrate that a low MITF/AXL ratio predicts early resistance to multiple targeted drugs, and warrant clinical validation of AXL inhibitors to combat resistance of BRAF and NRAS mutant MITF-low melanomas.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Melanoma/tratamiento farmacológico , Factor de Transcripción Asociado a Microftalmía/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Neoplasias Cutáneas/tratamiento farmacológico , Aminopiridinas/farmacología , Animales , Benzamidas/farmacología , Línea Celular Tumoral , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Mesilato de Imatinib , Imidazoles/farmacología , Indoles/farmacología , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , Ratones , Factor de Transcripción Asociado a Microftalmía/metabolismo , Oximas/farmacología , Piperazinas/farmacología , Pronóstico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Piridonas/farmacología , Pirimidinas/farmacología , Pirimidinonas/farmacología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Sulfonamidas/farmacología , Vemurafenib , Ensayos Antitumor por Modelo de Xenoinjerto , Tirosina Quinasa del Receptor Axl
17.
Endocr Relat Cancer ; 18(6): 743-57, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21937739

RESUMEN

Oncogene-induced senescence (OIS) is a growth arrest triggered by the enforced expression of cancer-promoting genes and acts as a barrier against malignant transformation in vivo. In this study, by a combination of in vitro and in vivo approaches, we investigate the role of OIS in tumours originating from the thyroid epithelium. We found that expression of different thyroid tumour-associated oncogenes in primary human thyrocytes triggers senescence, as demonstrated by the presence of OIS hallmarks: changes in cell morphology, accumulation of SA-ß-Gal and senescence-associated heterochromatic foci, and upregulation of transcription of the cyclin-dependent kinase inhibitors p16(INK4a) and p21(CIP1). Furthermore, immunohistochemical analysis of a panel of thyroid tumours characterised by different aggressiveness showed that the expression of OIS markers such as p16(INK4a), p21(CIP1) and IGFBP7 is upregulated at early stages, and lost during thyroid tumour progression. Taken together, our results suggest a role of OIS in thyroid carcinogenesis.


Asunto(s)
Envejecimiento , Proto-Oncogenes , Neoplasias de la Tiroides/metabolismo , Adulto , Anciano , Carcinoma , Carcinoma Papilar , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Femenino , Células HEK293 , Humanos , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/biosíntesis , Masculino , Persona de Mediana Edad , Proto-Oncogenes/genética , Cáncer Papilar Tiroideo , Carcinoma Anaplásico de Tiroides , Glándula Tiroides/citología , Transducción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA