Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Immunity ; 56(1): 143-161.e11, 2023 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-36630913

RESUMEN

Although T cells can exert potent anti-tumor immunity, a subset of T helper (Th) cells producing interleukin-22 (IL-22) in breast and lung tumors is linked to dismal patient outcome. Here, we examined the mechanisms whereby these T cells contribute to disease. In murine models of lung and breast cancer, constitutional and T cell-specific deletion of Il22 reduced metastases without affecting primary tumor growth. Deletion of the IL-22 receptor on cancer cells decreases metastasis to a degree similar to that seen in IL-22-deficient mice. IL-22 induced high expression of CD155, which bound to the activating receptor CD226 on NK cells. Excessive activation led to decreased amounts of CD226 and functionally impaired NK cells, which elevated the metastatic burden. IL-22 signaling was also associated with CD155 expression in human datasets and with poor patient outcomes. Taken together, our findings reveal an immunosuppressive circuit activated by T cell-derived IL-22 that promotes lung metastasis.


Asunto(s)
Interleucinas , Neoplasias , Receptores Virales , Linfocitos T Colaboradores-Inductores , Animales , Humanos , Ratones , Antígenos de Diferenciación de Linfocitos T/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Células Asesinas Naturales/metabolismo , Neoplasias/metabolismo , Unión Proteica , Linfocitos T Colaboradores-Inductores/metabolismo , Interleucina-22
2.
Nat Commun ; 13(1): 2760, 2022 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-35589749

RESUMEN

Autophagy has vasculoprotective roles, but whether and how it regulates lymphatic endothelial cells (LEC) homeostasis and lymphangiogenesis is unknown. Here, we show that genetic deficiency of autophagy in LEC impairs responses to VEGF-C and injury-driven corneal lymphangiogenesis. Autophagy loss in LEC compromises the expression of main effectors of LEC identity, like VEGFR3, affects mitochondrial dynamics and causes an accumulation of lipid droplets (LDs) in vitro and in vivo. When lipophagy is impaired, mitochondrial ATP production, fatty acid oxidation, acetyl-CoA/CoA ratio and expression of lymphangiogenic PROX1 target genes are dwindled. Enforcing mitochondria fusion by silencing dynamin-related-protein 1 (DRP1) in autophagy-deficient LEC fails to restore LDs turnover and lymphatic gene expression, whereas supplementing the fatty acid precursor acetate rescues VEGFR3 levels and signaling, and lymphangiogenesis in LEC-Atg5-/- mice. Our findings reveal that lipophagy in LEC by supporting FAO, preserves a mitochondrial-PROX1 gene expression circuit that safeguards LEC responsiveness to lymphangiogenic mediators and lymphangiogenesis.


Asunto(s)
Linfangiogénesis , Vasos Linfáticos , Animales , Autofagia/genética , Células Endoteliales/metabolismo , Ácidos Grasos/metabolismo , Gotas Lipídicas/metabolismo , Linfangiogénesis/genética , Vasos Linfáticos/metabolismo , Ratones , Mitocondrias , Factores de Transcripción/metabolismo
4.
EMBO Mol Med ; 13(12): e12924, 2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34762341

RESUMEN

Long-range communication between tumor cells and the lymphatic vasculature defines competency for metastasis in different cancer types, particularly in melanoma. Nevertheless, the discovery of selective blockers of lymphovascular niches has been compromised by the paucity of experimental systems for whole-body analyses of tumor progression. Here, we exploit immunocompetent and immunodeficient mouse models for live imaging of Vegfr3-driven neolymphangiogenesis, as a versatile platform for drug screening in vivo. Spatiotemporal analyses of autochthonous melanomas and patient-derived xenografts identified double-stranded RNA mimics (dsRNA nanoplexes) as potent inhibitors of neolymphangiogenesis, metastasis, and post-surgical disease relapse. Mechanistically, dsRNA nanoplexes were found to exert a rapid dual action in tumor cells and in their associated lymphatic vasculature, involving the transcriptional repression of the lymphatic drivers Midkine and Vegfr3, respectively. This suppressive function was mediated by a cell-autonomous type I interferon signaling and was not shared by FDA-approved antimelanoma treatments. These results reveal an alternative strategy for targeting the tumor cell-lymphatic crosstalk and underscore the power of Vegfr3-lymphoreporters for pharmacological testing in otherwise aggressive cancers.


Asunto(s)
Melanoma , ARN Bicatenario , Animales , Humanos , Melanoma/tratamiento farmacológico , Melanoma/patología , Ratones , Ratones Desnudos , Transducción de Señal
5.
Adv Drug Deliv Rev ; 175: 113833, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34147531

RESUMEN

Imaging of the lymphatic vasculature has gained great attention in various fields, not only because lymphatic vessels act as a key draining system in the body, but also for their implication in autoimmune diseases, organ transplant, inflammation and cancer. Thus, neolymphangiogenesis, or the generation of new lymphatics, is typically an early event in the development of multiple tumor types, particularly in aggressive ones such as malignant melanoma. Still, the understanding of how lymphatic endothelial cells get activated at distal (pre)metastatic niches and their impact on therapy is still unclear. Addressing these questions is of particular interest in the case of immune modulators, because endothelial cells may favor or halt inflammatory processes depending on the cellular context. Therefore, there is great interest in visualizing the lymphatic vasculature in vivo. Here, we review imaging tools and mouse models used to analyze the lymphatic vasculature during tumor progression. We also discuss therapeutic approaches based on nanomedicines to target the lymphatic system and the potential use of extracellular vesicles to track and target sentinel lymph nodes. Finally, we summarize main pre-clinical models developed to visualize the lymphatic vasculature in vivo, discussing their applications with a particular focus in metastatic melanoma.


Asunto(s)
Vesículas Extracelulares/metabolismo , Sistema Linfático/diagnóstico por imagen , Sistema de Administración de Fármacos con Nanopartículas , Animales , Vesículas Extracelulares/patología , Humanos , Ganglios Linfáticos/diagnóstico por imagen , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/patología , Sistema Linfático/efectos de los fármacos , Sistema Linfático/patología , Vasos Linfáticos/diagnóstico por imagen , Vasos Linfáticos/efectos de los fármacos , Vasos Linfáticos/patología , Ganglio Linfático Centinela/diagnóstico por imagen , Ganglio Linfático Centinela/patología
6.
Cancer Cell ; 39(5): 610-631, 2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33545064

RESUMEN

There is a lack of appropriate melanoma models that can be used to evaluate the efficacy of novel therapeutic modalities. Here, we discuss the current state of the art of melanoma models including genetically engineered mouse, patient-derived xenograft, zebrafish, and ex vivo and in vitro models. We also identify five major challenges that can be addressed using such models, including metastasis and tumor dormancy, drug resistance, the melanoma immune response, and the impact of aging and environmental exposures on melanoma progression and drug resistance. Additionally, we discuss the opportunity for building models for rare subtypes of melanomas, which represent an unmet critical need. Finally, we identify key recommendations for melanoma models that may improve accuracy of preclinical testing and predict efficacy in clinical trials, to help usher in the next generation of melanoma therapies.


Asunto(s)
Modelos Animales de Enfermedad , Melanoma/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Microambiente Tumoral/inmunología , Animales , Humanos , Inmunidad/inmunología , Inmunoterapia/métodos , Melanoma/patología , Neoplasias Cutáneas/patología
7.
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
8.
Nat Med ; 26(12): 1865-1877, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33077955

RESUMEN

An open question in aggressive cancers such as melanoma is how malignant cells can shift the immune system to pro-tumorigenic functions. Here we identify midkine (MDK) as a melanoma-secreted driver of an inflamed, but immune evasive, microenvironment that defines poor patient prognosis and resistance to immune checkpoint blockade. Mechanistically, MDK was found to control the transcriptome of melanoma cells, allowing for coordinated activation of nuclear factor-κB and downregulation of interferon-associated pathways. The resulting MDK-modulated secretome educated macrophages towards tolerant phenotypes that promoted CD8+ T cell dysfunction. In contrast, genetic targeting of MDK sensitized melanoma cells to anti-PD-1/anti-PD-L1 treatment. Emphasizing the translational relevance of these findings, the expression profile of MDK-depleted tumors was enriched in key indicators of a good response to immune checkpoint blockers in independent patient cohorts. Together, these data reveal that MDK acts as an internal modulator of autocrine and paracrine signals that maintain immune suppression in aggressive melanomas.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Melanoma Experimental/terapia , Midkina/genética , Microambiente Tumoral/genética , Animales , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/genética , Linfocitos T CD8-positivos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Terapia Genética , Humanos , Melanoma Experimental/genética , Melanoma Experimental/patología , Ratones , Midkina/farmacología , FN-kappa B/genética , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Transcriptoma/genética
9.
Cancer Cell ; 38(4): 446-448, 2020 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-32916127

RESUMEN

Lymph nodes are typically the first clinically detectable site of metastasis in melanoma, but the mechanisms that determine this preference are not well understood. An article in Nature reports that the unique composition of lymph may protect melanoma cells from ferroptosis-a form of iron-dependent cell death, thereby increasing metastatic efficiency.


Asunto(s)
Ferroptosis , Melanoma , Humanos , Ganglios Linfáticos , Metástasis Linfática , Melanoma/genética
11.
Oncogene ; 38(19): 3585-3597, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30664687

RESUMEN

Although antioxidants promote melanoma metastasis, the role of reactive oxygen species (ROS) in other stages of melanoma progression is controversial. Moreover, genes regulating ROS have not been functionally characterized throughout the entire tumor progression in mouse models of cancer. To address this question, we crossed mice-bearing knock-out of Klf9, an ubiquitous transcriptional regulator of oxidative stress, with two conditional melanocytic mouse models: BrafCA mice, where BrafV600E causes premalignant melanocytic hyperplasia, and BrafCA/Pten-/- mice, where BrafV600E and loss of Pten induce primary melanomas and metastases. Klf9 deficiency inhibited premalignant melanocytic hyperplasia in BrafCA mice but did not affect formation and growth of BrafCA/Pten-/- primary melanomas. It also, as expected, promoted BrafCA/Pten-/- metastasis. Treatment with antioxidant N-acetyl cysteine phenocopied loss of Klf9 including suppression of melanocytic hyperplasia. We were interested in a different role of Klf9 in regulation of cell proliferation in BrafCA and BrafCA/Pten-/- melanocytic cells. Mechanistically, we demonstrated that BRAFV600E signaling transcriptionally upregulated KLF9 and that KLF9-dependent ROS were required for full-scale activation of ERK1/2 and induction of cell proliferation by BRAFV600E. PTEN depletion in BRAFV600E-melanocytes did not further activate ERK1/2 and cell proliferation, but rendered these phenotypes insensitive to KLF9 and ROS. Our data identified an essential role of KLF9-dependent ROS in BRAFV600E signaling in premalignant melanocytes, offered an explanation to variable role of ROS in premalignant and transformed melanocytic cells and suggested a novel mechanism for suppression of premalignant growth by topical antioxidants.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/metabolismo , Melanoma/patología , Especies Reactivas de Oxígeno/metabolismo , Neoplasias Cutáneas/patología , Acetilcisteína/efectos adversos , Adulto , Anciano , Anciano de 80 o más Años , Animales , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Melanocitos/efectos de los fármacos , Melanocitos/metabolismo , Melanocitos/patología , Melanoma/genética , Melanoma/metabolismo , Melanoma Experimental/inducido químicamente , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones Noqueados , Persona de Mediana Edad , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias Cutáneas/metabolismo
12.
Cancer Cell ; 35(1): 46-63.e10, 2019 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-30581152

RESUMEN

Modulators of mRNA stability are not well understood in melanoma, an aggressive tumor with complex changes in the transcriptome. Here we report the ability of p62/SQSTM1 to extend mRNA half-life of a spectrum of pro-metastatic factors. These include FERMT2 and other transcripts with no previous links to melanoma. Transcriptomic, proteomic, and interactomic analyses, combined with validation in clinical biopsies and mouse models, identified a selected set of RNA-binding proteins (RBPs) recruited by p62, with IGF2BP1 as a key partner. This p62-RBP interaction distinguishes melanoma from other tumors where p62 controls autophagy or oxidative stress. The relevance of these data is emphasized by follow-up analyses of patient prognosis revealing p62 and FERMT2 as adverse determinants of disease-free survival.


Asunto(s)
Melanoma/metabolismo , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética , ARN Mensajero/química , Proteínas de Unión al ARN/metabolismo , Proteína Sequestosoma-1/metabolismo , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/genética , Proteínas de la Membrana/química , Ratones , Proteínas de Neoplasias/química , Trasplante de Neoplasias , Mapas de Interacción de Proteínas , Proteómica/métodos , Estabilidad del ARN , Análisis de Matrices Tisulares
13.
Nat Commun ; 8(1): 2249, 2017 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-29269732

RESUMEN

Melanomas are well-known for their altered mRNA expression profiles. Yet, the specific contribution of mRNA binding proteins (mRBPs) to melanoma development remains unclear. Here we identify a cluster of melanoma-enriched genes under the control of CUGBP Elav-like family member 1 (CELF1). CELF1 was discovered with a distinct prognostic value in melanoma after mining the genomic landscape of the 692 known mRBPs across different cancer types. Genome-wide transcriptomic, proteomic, and RNA-immunoprecipitation studies, together with loss-of-function analyses in cell lines, and histopathological evaluation in clinical biopsies, revealed an intricate repertoire of CELF1-RNA interactors with minimal overlap with other malignancies. This systems approach uncovered the oncogene DEK as an unexpected target and downstream effector of CELF1. Importantly, CELF1 and DEK were found to represent early-induced melanoma genes and adverse indicators of overall patient survival. These results underscore novel roles of CELF1 in melanoma, illustrating tumor type-restricted functions of RBPs in cancer.


Asunto(s)
Proteínas CELF1/fisiología , Melanoma/genética , Oncogenes , Biología de Sistemas , Regiones no Traducidas 3' , Biopsia , Proteínas CELF1/genética , Proteínas CELF1/metabolismo , Línea Celular Tumoral , Proliferación Celular , Proteínas Cromosómicas no Histona/metabolismo , Humanos , Inmunoprecipitación , Melanoma/patología , Proteínas Oncogénicas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Pronóstico , Proteómica , ARN Neoplásico/genética , Análisis de Supervivencia , Transcriptoma
14.
Cell Stem Cell ; 21(5): 559-561, 2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-29100006

RESUMEN

It is unclear whether melanoma initiates from mature melanocytes or stem cell precursors. In this issue of Cell Stem Cell, Moon et al. (2017) and Köhler et al. (2017) use in vivo lineage tracing to demonstrate that these two possibilities may occur downstream of the same pro-tumorigenic lesions, depending on environmental factors or the anatomical location.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Señales (Psicología) , Humanos , Melanocitos/citología , Células Madre/citología
15.
Nat Cell Biol ; 19(11): 1311-1312, 2017 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-29087386

RESUMEN

A variety of non-coding RNAs have been reported as endogenous sponges for cancer-modulating miRNAs. However, miRNA trapping by transcripts with protein-coding functions is less understood. The mRNA of TYRP1 is now found to sequester the tumour suppressor miR-16 on non-canonical miRNA response elements in melanoma, thereby promoting malignant growth.


Asunto(s)
Melanoma/genética , Glicoproteínas de Membrana/genética , MicroARNs/genética , Oxidorreductasas/genética , Animales , Proliferación Celular/genética , Humanos , Proteínas Supresoras de Tumor/genética
16.
Cancer Res ; 77(21): 5873-5885, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28887323

RESUMEN

Autophagy mediates resistance to various anticancer agents. In melanoma, resistance to targeted therapy has been linked to expression of Wnt5A, an intrinsic inhibitor of ß-catenin, which also promotes invasion. In this study, we assessed the interplay between Wnt5A and autophagy by combining expression studies in human clinical biopsies with functional analyses in cell lines and mouse models. Melanoma cells with high Wnt5A and low ß-catenin displayed increased basal autophagy. Genetic blockade of autophagy revealed an unexpected feedback loop whereby knocking down the autophagy factor ATG5 in Wnt5Ahigh cells decreased Wnt5A and increased ß-catenin. To define the physiologic relevance of this loop, melanoma cells with different Wnt status were treated in vitro and in vivo with the potent lysosomotropic compound Lys05. Wnt5Ahigh cells were less sensitive to Lys05 and could be reverted by inducing ß-catenin activity. Our results suggest the efficacy of autophagy inhibitors might be improved by taking the Wnt signature of melanoma cells into account. Cancer Res; 77(21); 5873-85. ©2017 AACR.


Asunto(s)
Proteína 5 Relacionada con la Autofagia/genética , Autofagia/genética , Melanoma/genética , Vía de Señalización Wnt/genética , Aminoquinolinas/farmacología , Animales , Autofagia/efectos de los fármacos , Proteína 5 Relacionada con la Autofagia/metabolismo , Western Blotting , Línea Celular Tumoral , Retroalimentación Fisiológica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/metabolismo , Melanoma/patología , Ratones , Poliaminas/farmacología , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Vía de Señalización Wnt/efectos de los fármacos , Proteína Wnt-5a/genética , Proteína Wnt-5a/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
17.
Nature ; 546(7660): 676-680, 2017 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-28658220

RESUMEN

Cutaneous melanoma is a type of cancer with an inherent potential for lymph node colonization, which is generally preceded by neolymphangiogenesis. However, sentinel lymph node removal does not necessarily extend the overall survival of patients with melanoma. Moreover, lymphatic vessels collapse and become dysfunctional as melanomas progress. Therefore, it is unclear whether (and how) lymphangiogenesis contributes to visceral metastasis. Soluble and vesicle-associated proteins secreted by tumours and/or their stroma have been proposed to condition pre-metastatic sites in patients with melanoma. Still, the identities and prognostic value of lymphangiogenic mediators remain unclear. Moreover, our understanding of lymphangiogenesis (in melanomas and other tumour types) is limited by the paucity of mouse models for live imaging of distal pre-metastatic niches. Injectable lymphatic tracers have been developed, but their limited diffusion precludes whole-body imaging at visceral sites. Vascular endothelial growth factor receptor 3 (VEGFR3) is an attractive 'lymphoreporter' because its expression is strongly downregulated in normal adult lymphatic endothelial cells, but is activated in pathological situations such as inflammation and cancer. Here, we exploit this inducibility of VEGFR3 to engineer mouse melanoma models for whole-body imaging of metastasis generated by human cells, clinical biopsies or endogenously deregulated oncogenic pathways. This strategy revealed early induction of distal pre-metastatic niches uncoupled from lymphangiogenesis at primary lesions. Analyses of the melanoma secretome and validation in clinical specimens showed that the heparin-binding factor midkine is a systemic inducer of neo-lymphangiogenesis that defines patient prognosis. This role of midkine was linked to a paracrine activation of the mTOR pathway in lymphatic endothelial cells. These data support the use of VEGFR3 reporter mice as a 'MetAlert' discovery platform for drivers and inhibitors of metastasis.


Asunto(s)
Citocinas/metabolismo , Vasos Linfáticos/metabolismo , Metástasis de la Neoplasia/diagnóstico por imagen , Metástasis de la Neoplasia/patología , Imagen de Cuerpo Entero/métodos , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Endoteliales/metabolismo , Femenino , Genes Reporteros , Humanos , Linfangiogénesis , Vasos Linfáticos/patología , Masculino , Melanoma/diagnóstico por imagen , Melanoma/metabolismo , Melanoma/patología , Ratones , Midkina , Comunicación Paracrina , Pronóstico , Recurrencia , Reproducibilidad de los Resultados , Serina-Treonina Quinasas TOR/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/análisis , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Pigment Cell Melanoma Res ; 30(2): 194-202, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27893188

RESUMEN

DEK is an oncoprotein involved in a variety of cellular functions, such as DNA repair, replication, and transcriptional control. DEK is preferentially expressed in actively proliferating and malignant cells, including melanoma cell lines in which DEK was previously demonstrated to play a critical role in proliferation and chemoresistance. Still, the impact of this protein in melanoma progression remains unclear. Thus, we performed a comprehensive analysis of DEK expression in different melanocytic tumors. The immunostaining results of 303 tumors demonstrated negligible DEK expression in benign lesions. Conversely, malignant lesions, particularly in metastatic cases, were largely positive for DEK expression, which was partially associated with genomic amplification. Importantly, DEK overexpression was correlated with histological features of aggressiveness in primary tumors and poor prognosis in melanoma patients. In conclusion, our study provides new insight into the involvement of DEK in melanoma progression, as well as proof of concept for its potential application as a marker and therapeutic target of melanoma.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Regulación Neoplásica de la Expresión Génica , Melanoma/patología , Proteínas Oncogénicas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Biomarcadores de Tumor/genética , Proliferación Celular , Proteínas Cromosómicas no Histona/genética , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Técnicas para Inmunoenzimas , Metástasis Linfática , Masculino , Melanoma/genética , Melanoma/metabolismo , Persona de Mediana Edad , Estadificación de Neoplasias , Proteínas Oncogénicas/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Pronóstico , Tasa de Supervivencia , Células Tumorales Cultivadas
19.
Cancer Cell ; 30(5): 694-707, 2016 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-27908735

RESUMEN

RNA binding proteins (RBPs) modulate cancer progression through poorly understood mechanisms. Here we show that the RBP UNR/CSDE1 is overexpressed in melanoma tumors and promotes invasion and metastasis. iCLIP sequencing, RNA sequencing, and ribosome profiling combined with in silico studies unveiled sets of pro-metastatic factors coordinately regulated by UNR as part of RNA regulons. In addition to RNA steady-state levels, UNR was found to control many of its targets at the level of translation elongation/termination. Key pro-oncogenic targets of UNR included VIM and RAC1, as validated by loss- and gain-of-function studies. Our results identify UNR as an oncogenic modulator of melanoma progression, unravel the underlying molecular mechanisms, and identify potential targets for this therapeutically challenging malignancy.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Melanoma/patología , Proteínas de Unión al ARN/metabolismo , Regulación hacia Arriba , Vimentina/genética , Proteína de Unión al GTP rac1/genética , Animales , Proteínas de Unión al ADN/genética , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/genética , Melanoma/metabolismo , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Trasplante de Neoplasias , Procesamiento Postranscripcional del ARN , Proteínas de Unión al ARN/genética , Ribosomas/genética , Análisis de Secuencia de ARN/métodos
20.
Nat Commun ; 7: 13418, 2016 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-27857118

RESUMEN

Nuclear 3'-end-polyadenylation is essential for the transport, stability and translation of virtually all eukaryotic mRNAs. Poly(A) tail extension can also occur in the cytoplasm, but the transcripts involved are incompletely understood, particularly in cancer. Here we identify a lineage-specific requirement of the cytoplasmic polyadenylation binding protein 4 (CPEB4) in malignant melanoma. CPEB4 is upregulated early in melanoma progression, as defined by computational and histological analyses. Melanoma cells are distinct from other tumour cell types in their dependency on CPEB4, not only to prevent mitotic aberrations, but to progress through G1/S cell cycle checkpoints. RNA immunoprecipitation, sequencing of bound transcripts and poly(A) length tests link the melanoma-specific functions of CPEB4 to signalling hubs specifically enriched in this disease. Essential in these CPEB4-controlled networks are the melanoma drivers MITF and RAB7A, a feature validated in clinical biopsies. These results provide new mechanistic links between cytoplasmic polyadenylation and lineage specification in melanoma.


Asunto(s)
Melanoma/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Ciclo Celular , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Melanoma/genética , Ratones , Neoplasias Experimentales , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...