Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Rev Sci Instrum ; 95(5)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38717271

RESUMEN

Angle-resolved photoemission spectroscopy (ARPES) is a technique used to map the occupied electronic structure of solids. Recent progress in x-ray focusing optics has led to the development of ARPES into a microscopic tool, permitting the electronic structure to be spatially mapped across the surface of a sample. This comes at the expense of a time-consuming scanning process to cover not only a three-dimensional energy-momentum (E, kx, ky) space but also the two-dimensional surface area. Here, we implement a protocol to autonomously search both k- and real-space in order to find positions of particular interest, either because of their high photoemission intensity or because of sharp spectral features. The search is based on the use of Gaussian process regression and can easily be expanded to include additional parameters or optimization criteria. This autonomous experimental control is implemented on the SGM4 micro-focus beamline of the synchrotron radiation source ASTRID2.

2.
Nature ; 629(8012): 543-554, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38750233

RESUMEN

Metastasis is a multistep process by which cancer cells break away from their original location and spread to distant organs, and is responsible for the vast majority of cancer-related deaths. Preventing early metastatic dissemination would revolutionize the ability to fight cancer. Unfortunately, the relatively poor understanding of the molecular underpinnings of metastasis has hampered the development of effective anti-metastatic drugs. Although it is now accepted that disseminating tumour cells need to acquire multiple competencies to face the many obstacles they encounter before reaching their metastatic site(s), whether these competencies are acquired through an accumulation of metastasis-specific genetic alterations and/or non-genetic events is often debated. Here we review a growing body of literature highlighting the importance of both genetic and non-genetic reprogramming events during the metastatic cascade, and discuss how genetic and non-genetic processes act in concert to confer metastatic competencies. We also describe how recent technological advances, and in particular the advent of single-cell multi-omics and barcoding approaches, will help to better elucidate the cross-talk between genetic and non-genetic mechanisms of metastasis and ultimately inform innovative paths for the early detection and interception of this lethal process.


Asunto(s)
Metástasis de la Neoplasia , Neoplasias , Animales , Humanos , Metástasis de la Neoplasia/tratamiento farmacológico , Metástasis de la Neoplasia/genética , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Análisis de la Célula Individual , Multiómica , Tipificación Molecular , Reprogramación Celular
3.
Cell ; 187(1): 166-183.e25, 2024 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-38181739

RESUMEN

To better understand intrinsic resistance to immune checkpoint blockade (ICB), we established a comprehensive view of the cellular architecture of the treatment-naive melanoma ecosystem and studied its evolution under ICB. Using single-cell, spatial multi-omics, we showed that the tumor microenvironment promotes the emergence of a complex melanoma transcriptomic landscape. Melanoma cells harboring a mesenchymal-like (MES) state, a population known to confer resistance to targeted therapy, were significantly enriched in early on-treatment biopsies from non-responders to ICB. TCF4 serves as the hub of this landscape by being a master regulator of the MES signature and a suppressor of the melanocytic and antigen presentation transcriptional programs. Targeting TCF4 genetically or pharmacologically, using a bromodomain inhibitor, increased immunogenicity and sensitivity of MES cells to ICB and targeted therapy. We thereby uncovered a TCF4-dependent regulatory network that orchestrates multiple transcriptional programs and contributes to resistance to both targeted therapy and ICB in melanoma.


Asunto(s)
Melanoma , Humanos , Redes Reguladoras de Genes , Inmunoterapia , Melanocitos , Melanoma/tratamiento farmacológico , Melanoma/genética , Factor de Transcripción 4/genética , Microambiente Tumoral
4.
Trends Cancer ; 9(11): 897-910, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37453870

RESUMEN

Tumors are heterogeneous ecosystems in which cancer cells coexist within a complex tumor immune microenvironment (TIME). The malignant, stromal, and immune cell compartments establish a plethora of bidirectional cell-cell communication crosstalks that influence tumor growth and metastatic dissemination, which we are only beginning to understand. Cancer cells either co-opt or promote the formation of new blood and lymphatic vessels to cope with their need for nutrients and oxygen. Recent studies have highlighted additional key roles for the tumor vasculature and have identified the perivascular niche as a cellular hub, where intricate and dynamic cellular interactions promote cancer stemness, immune evasion, dormancy, and metastatic spreading. Here, we review these findings, and discuss how they may be exploited therapeutically.


Asunto(s)
Ecosistema , Neoplasias , Humanos , Neoplasias/patología , Comunicación Celular , Microambiente Tumoral
6.
Nature ; 610(7930): 190-198, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36131018

RESUMEN

Although melanoma is notorious for its high degree of heterogeneity and plasticity1,2, the origin and magnitude of cell-state diversity remains poorly understood. Equally, it is unclear whether growth and metastatic dissemination are supported by overlapping or distinct melanoma subpopulations. Here, by combining mouse genetics, single-cell and spatial transcriptomics, lineage tracing and quantitative modelling, we provide evidence of a hierarchical model of tumour growth that mirrors the cellular and molecular logic underlying the cell-fate specification and differentiation of the embryonic neural crest. We show that tumorigenic competence is associated with a spatially localized perivascular niche, a phenotype acquired through an intercellular communication pathway established by endothelial cells. Consistent with a model in which only a fraction of cells are fated to fuel growth, temporal single-cell tracing of a population of melanoma cells with a mesenchymal-like state revealed that these cells do not contribute to primary tumour growth but, instead, constitute a pool of metastatic initiating cells that switch cell identity while disseminating to secondary organs. Our data provide a spatially and temporally resolved map of the diversity and trajectories of melanoma cell states and suggest that the ability to support growth and metastasis are limited to distinct pools of cells. The observation that these phenotypic competencies can be dynamically acquired after exposure to specific niche signals warrant the development of therapeutic strategies that interfere with the cancer cell reprogramming activity of such microenvironmental cues.


Asunto(s)
Proliferación Celular , Melanoma , Metástasis de la Neoplasia , Animales , Comunicación Celular , Diferenciación Celular , Linaje de la Célula , Rastreo Celular , Reprogramación Celular , Células Endoteliales , Melanoma/genética , Melanoma/patología , Mesodermo/patología , Ratones , Metástasis de la Neoplasia/patología , Cresta Neural/embriología , Fenotipo , Análisis de la Célula Individual , Transcriptoma , Microambiente Tumoral
8.
Nat Cell Biol ; 24(6): 940-953, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35654839

RESUMEN

Bidirectional signalling between the tumour and stroma shapes tumour aggressiveness and metastasis. ATF4 is a major effector of the Integrated Stress Response, a homeostatic mechanism that couples cell growth and survival to bioenergetic demands. Using conditional knockout ATF4 mice, we show that global, or fibroblast-specific loss of host ATF4, results in deficient vascularization and a pronounced growth delay of syngeneic melanoma and pancreatic tumours. Single-cell transcriptomics of tumours grown in Atf4Δ/Δ mice uncovered a reduction in activation markers in perivascular cancer-associated fibroblasts (CAFs). Atf4Δ/Δ fibroblasts displayed significant defects in collagen biosynthesis and deposition and a reduced ability to support angiogenesis. Mechanistically, ATF4 regulates the expression of the Col1a1 gene and levels of glycine and proline, the major amino acids of collagen. Analyses of human melanoma and pancreatic tumours revealed a strong correlation between ATF4 and collagen levels. Our findings establish stromal ATF4 as a key driver of CAF functionality, malignant progression and metastasis.


Asunto(s)
Fibroblastos Asociados al Cáncer , Melanoma , Neoplasias Pancreáticas , Animales , Fibroblastos Asociados al Cáncer/metabolismo , Colágeno/metabolismo , Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Melanoma/genética , Ratones , Ratones Noqueados , Neovascularización Patológica/metabolismo , Neoplasias Pancreáticas/patología
9.
Nature ; 605(7911): 747-753, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35585241

RESUMEN

Cancer metastasis requires the transient activation of cellular programs enabling dissemination and seeding in distant organs1. Genetic, transcriptional and translational heterogeneity contributes to this dynamic process2,3. Metabolic heterogeneity has also been observed4, yet its role in cancer progression is less explored. Here we find that the loss of phosphoglycerate dehydrogenase (PHGDH) potentiates metastatic dissemination. Specifically, we find that heterogeneous or low PHGDH expression in primary tumours of patients with breast cancer is associated with decreased metastasis-free survival time. In mice, circulating tumour cells and early metastatic lesions are enriched with Phgdhlow cancer cells, and silencing Phgdh in primary tumours increases metastasis formation. Mechanistically, Phgdh interacts with the glycolytic enzyme phosphofructokinase, and the loss of this interaction activates the hexosamine-sialic acid pathway, which provides precursors for protein glycosylation. As a consequence, aberrant protein glycosylation occurs, including increased sialylation of integrin αvß3, which potentiates cell migration and invasion. Inhibition of sialylation counteracts the metastatic ability of Phgdhlow cancer cells. In conclusion, although the catalytic activity of PHGDH supports cancer cell proliferation, low PHGDH protein expression non-catalytically potentiates cancer dissemination and metastasis formation. Thus, the presence of PHDGH heterogeneity in primary tumours could be considered a sign of tumour aggressiveness.


Asunto(s)
Neoplasias de la Mama , Metástasis de la Neoplasia , Fosfoglicerato-Deshidrogenasa , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Femenino , Silenciador del Gen , Humanos , Ratones , Fosfoglicerato-Deshidrogenasa/genética , Serina/metabolismo
10.
Neuro Oncol ; 24(12): 2133-2145, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-35639831

RESUMEN

BACKGROUND: Glioblastoma (GBM) is the most aggressive primary brain tumor. Its cellular composition is very heterogeneous, with cells exhibiting stem-cell characteristics (GSCs) that co-determine therapy resistance and tumor recurrence. Bone Morphogenetic Protein (BMP)-4 promotes astroglial and suppresses oligodendrocyte differentiation in GSCs, processes associated with superior patient prognosis. We characterized variability in cell viability of patient-derived GBM cultures in response to BMP4 and, based on single-cell transcriptome profiling, propose predictive positive and early-response markers for sensitivity to BMP4. METHODS: Cell viability was assessed in 17 BMP4-treated patient-derived GBM cultures. In two cultures, one highly-sensitive to BMP4 (high therapeutic efficacy) and one with low-sensitivity, response to treatment with BMP4 was characterized. We applied single-cell RNA-sequencing, analyzed the relative abundance of cell clusters, searched for and identified the aforementioned two marker types, and validated these results in all 17 cultures. RESULTS: High variation in cell viability was observed after treatment with BMP4. In three cultures with highest sensitivity for BMP4, a substantial new cell subpopulation formed. These cells displayed decreased cell proliferation and increased apoptosis. Neuronal differentiation was reduced most in cultures with little sensitivity for BMP4. OLIG1/2 levels were found predictive for high sensitivity to BMP4. Activation of ribosomal translation (RPL27A, RPS27) was up-regulated within one day in cultures that were very sensitive to BMP4. CONCLUSION: The changes in composition of patient-derived GBM cultures obtained after treatment with BMP4 correlate with treatment efficacy. OLIG1/2 expression can predict this efficacy, and upregulation of RPL27A and RPS27 are useful early-response markers.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Humanos , Glioma/patología , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Proliferación Celular , Perfilación de la Expresión Génica , Biomarcadores/metabolismo , ARN/metabolismo , Células Madre Neoplásicas/metabolismo , Diferenciación Celular , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/farmacología , Proteína Morfogenética Ósea 4/metabolismo
11.
FEBS J ; 289(5): 1352-1368, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-33999497

RESUMEN

Epithelial-to-mesenchymal transition (EMT), a process through which epithelial tumor cells acquire mesenchymal phenotypic properties, contributes to both metastatic dissemination and therapy resistance in cancer. Accumulating evidence indicates that nonepithelial tumors, including melanoma, can also gain mesenchymal-like properties that increase their metastatic propensity and decrease their sensitivity to therapy. In this review, we discuss recent findings, illustrating the striking similarities-but also knowledge gaps-between the biology of mesenchymal-like state(s) in melanoma and mesenchymal state(s) from epithelial cancers. Based on this comparative analysis, we suggest hypothesis-driven experimental approaches to further deepen our understanding of the EMT-like process in melanoma and how such investigations may pave the way towards the identification of clinically relevant biomarkers for prognosis and new therapeutic strategies.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Melanoma/genética , Proteínas de Neoplasias/genética , Neoplasias Cutáneas/genética , Factores de Transcripción/genética , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Diferenciación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Melanoma/patología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Terapia Molecular Dirigida/métodos , Metástasis de la Neoplasia , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Factores de Transcripción/metabolismo
12.
Ann N Y Acad Sci ; 1506(1): 142-163, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34850398

RESUMEN

The test for the cancer stem cell (CSC) hypothesis is to find a target expressed on all, and only CSCs in a patient tumor, then eliminate all cells with that target that eliminates the cancer. That test has not yet been achieved, but CSC diagnostics and targets found on CSCs and some other cells have resulted in a few clinically relevant therapies. However, it has become apparent that eliminating the subset of tumor cells characterized by self-renewal properties is essential for long-term tumor control. CSCs are able to regenerate and initiate tumor growth, recapitulating the heterogeneity present in the tumor before treatment. As great progress has been made in identifying and elucidating the biology of CSCs as well as their interactions with the tumor microenvironment, the time seems ripe for novel therapeutic strategies that target CSCs to find clinical applicability. On May 19-21, 2021, researchers in cancer stem cells met virtually for the Keystone eSymposium "Cancer Stem Cells: Advances in Biology and Clinical Translation" to discuss recent advances in the understanding of CSCs as well as clinical efforts to target these populations.


Asunto(s)
Congresos como Asunto/tendencias , Neoplasias/genética , Células Madre Neoplásicas/fisiología , Informe de Investigación , Investigación Biomédica Traslacional/tendencias , Microambiente Tumoral/fisiología , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Humanos , Neoplasias/metabolismo , Investigación Biomédica Traslacional/métodos
13.
J Exp Med ; 218(9)2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34287642

RESUMEN

The ability to adapt to environmental stress, including therapeutic insult, contributes to tumor evolution and drug resistance. In suboptimal conditions, the integrated stress response (ISR) promotes survival by dampening cytosolic translation. We show that ISR-dependent survival also relies on a concomitant up-regulation of mitochondrial protein synthesis, a vulnerability that can be exploited using mitoribosome-targeting antibiotics. Accordingly, such agents sensitized to MAPK inhibition, thus preventing the development of resistance in BRAFV600E melanoma models. Additionally, this treatment compromised the growth of melanomas that exhibited elevated ISR activity and resistance to both immunotherapy and targeted therapy. In keeping with this, pharmacological inactivation of ISR, or silencing of ATF4, rescued the antitumoral response to the tetracyclines. Moreover, a melanoma patient exposed to doxycycline experienced complete and long-lasting response of a treatment-resistant lesion. Our study indicates that the repurposing of mitoribosome-targeting antibiotics offers a rational salvage strategy for targeted therapy in BRAF mutant melanoma and a therapeutic option for NRAS-driven and immunotherapy-resistant tumors.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Melanoma/tratamiento farmacológico , Melanoma/patología , Ribosomas Mitocondriales/efectos de los fármacos , Anciano , Animales , Línea Celular Tumoral , Doxiciclina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , Masculino , Melanoma/genética , Melanoma/mortalidad , Ratones Endogámicos C57BL , Ratones Desnudos , Inhibidores de Proteínas Quinasas/farmacología , Estrés Fisiológico/efectos de los fármacos , Tigeciclina/farmacología , Neoplasias de la Úvea/tratamiento farmacológico , Neoplasias de la Úvea/patología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cancer Cell ; 39(8): 1135-1149.e8, 2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34143978

RESUMEN

Therapy resistance arises from heterogeneous drug-tolerant persister cells or minimal residual disease (MRD) through genetic and nongenetic mechanisms. A key question is whether specific molecular features of the MRD ecosystem determine which of these two distinct trajectories will eventually prevail. We show that, in melanoma exposed to mitogen-activated protein kinase therapeutics, emergence of a transient neural crest stem cell (NCSC) population in MRD concurs with the development of nongenetic resistance. This increase relies on a glial cell line-derived neurotrophic factor-dependent signaling cascade, which activates the AKT survival pathway in a focal adhesion kinase (FAK)-dependent manner. Ablation of the NCSC population through FAK inhibition delays relapse in patient-derived tumor xenografts. Strikingly, all tumors that ultimately escape this treatment exhibit resistance-conferring genetic alterations and increased sensitivity to extracellular signal-regulated kinase inhibition. These findings identify an approach that abrogates the nongenetic resistance trajectory in melanoma and demonstrate that the cellular composition of MRD deterministically imposes distinct drug resistance evolutionary paths.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/fisiología , Melanoma/tratamiento farmacológico , Melanoma/genética , Animales , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Humanos , Imidazoles/farmacología , Melanoma/patología , Ratones SCID , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Recurrencia Local de Neoplasia/genética , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Cresta Neural/patología , Oximas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Piridonas/farmacología , Pirimidinonas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Genome Res ; 30(12): 1815-1834, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32732264

RESUMEN

Deciphering the genomic regulatory code of enhancers is a key challenge in biology because this code underlies cellular identity. A better understanding of how enhancers work will improve the interpretation of noncoding genome variation and empower the generation of cell type-specific drivers for gene therapy. Here, we explore the combination of deep learning and cross-species chromatin accessibility profiling to build explainable enhancer models. We apply this strategy to decipher the enhancer code in melanoma, a relevant case study owing to the presence of distinct melanoma cell states. We trained and validated a deep learning model, called DeepMEL, using chromatin accessibility data of 26 melanoma samples across six different species. We show the accuracy of DeepMEL predictions on the CAGI5 challenge, where it significantly outperforms existing models on the melanoma enhancer of IRF4 Next, we exploit DeepMEL to analyze enhancer architectures and identify accurate transcription factor binding sites for the core regulatory complexes in the two different melanoma states, with distinct roles for each transcription factor, in terms of nucleosome displacement or enhancer activation. Finally, DeepMEL identifies orthologous enhancers across distantly related species, where sequence alignment fails, and the model highlights specific nucleotide substitutions that underlie enhancer turnover. DeepMEL can be used from the Kipoi database to predict and optimize candidate enhancers and to prioritize enhancer mutations. In addition, our computational strategy can be applied to other cancer or normal cell types.


Asunto(s)
Biología Computacional/métodos , Melanoma/genética , Pez Cebra/genética , Animales , Aprendizaje Profundo , Perros , Elementos de Facilitación Genéticos , Regulación Neoplásica de la Expresión Génica , Caballos , Humanos , Ratones , Porcinos
16.
Cell Rep ; 31(11): 107765, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32553158

RESUMEN

Tumor cell plasticity, including transdifferentiation, is thought to be a key driver of therapy failure, tumor dormancy, and metastatic dissemination. Although melanoma cells have been shown to adopt various phenotypic features in vitro, direct in vivo evidence of metastatic cell plasticity remains sparse. Here, we combine lineage tracing in a spontaneous metastatic mouse model of melanoma, advanced imaging, and single-cell RNA sequencing approaches to search for pathophysiologically relevant melanoma cellular states. We identify melanoma cells in intravascular niches of various metastatic organs. These cells are quiescent, are negative for characteristic melanoma markers, and acquire endothelial cell features. We replicate the endothelial transdifferentiation (EndT) finding in another mouse model and provide evidence of EndT in BRAFV600E-metastatic biopsies from human lung, brain, and small intestine, thus highlighting the clinical relevance of these findings. The tumor-vasculature pattern described herein may contribute to melanoma dormancy within metastatic organs and represent a putative target for therapies.


Asunto(s)
Transdiferenciación Celular/fisiología , Células Endoteliales/citología , Melanoma/patología , Metástasis de la Neoplasia/patología , Microambiente Tumoral/fisiología , Animales , Biomarcadores de Tumor/genética , Diferenciación Celular/fisiología , Melanoma/metabolismo , Ratones Transgénicos
17.
Science ; 366(6468): 1029-1034, 2019 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-31754005

RESUMEN

The Hippo signaling pathway and its two downstream effectors, the YAP and TAZ transcriptional coactivators, are drivers of tumor growth in experimental models. Studying mouse models, we show that YAP and TAZ can also exert a tumor-suppressive function. We found that normal hepatocytes surrounding liver tumors displayed activation of YAP and TAZ and that deletion of Yap and Taz in these peritumoral hepatocytes accelerated tumor growth. Conversely, experimental hyperactivation of YAP in peritumoral hepatocytes triggered regression of primary liver tumors and melanoma-derived liver metastases. Furthermore, whereas tumor cells growing in wild-type livers required YAP and TAZ for their survival, those surrounded by Yap- and Taz-deficient hepatocytes were not dependent on YAP and TAZ. Tumor cell survival thus depends on the relative activity of YAP and TAZ in tumor cells and their surrounding tissue, suggesting that YAP and TAZ act through a mechanism of cell competition to eliminate tumor cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Colangiocarcinoma/metabolismo , Hepatocitos/metabolismo , Neoplasias Hepáticas Experimentales/metabolismo , Transactivadores/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Supervivencia Celular , Colangiocarcinoma/patología , Vía de Señalización Hippo , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/secundario , Neoplasias Hepáticas Experimentales/patología , Melanoma/metabolismo , Melanoma/secundario , Ratones Endogámicos C57BL , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Transactivadores/economía , Transactivadores/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Carga Tumoral , Proteínas Señalizadoras YAP
18.
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
19.
Cell ; 174(4): 843-855.e19, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-30017245

RESUMEN

Many patients with advanced cancers achieve dramatic responses to a panoply of therapeutics yet retain minimal residual disease (MRD), which ultimately results in relapse. To gain insights into the biology of MRD, we applied single-cell RNA sequencing to malignant cells isolated from BRAF mutant patient-derived xenograft melanoma cohorts exposed to concurrent RAF/MEK-inhibition. We identified distinct drug-tolerant transcriptional states, varying combinations of which co-occurred within MRDs from PDXs and biopsies of patients on treatment. One of these exhibited a neural crest stem cell (NCSC) transcriptional program largely driven by the nuclear receptor RXRG. An RXR antagonist mitigated accumulation of NCSCs in MRD and delayed the development of resistance. These data identify NCSCs as key drivers of resistance and illustrate the therapeutic potential of MRD-directed therapy. They also highlight how gene regulatory network architecture reprogramming may be therapeutically exploited to limit cellular heterogeneity, a key driver of disease progression and therapy resistance.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Melanoma/tratamiento farmacológico , Neoplasia Residual/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Receptor gamma X Retinoide/antagonistas & inhibidores , Animales , Biomarcadores de Tumor , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/genética , Masculino , Melanoma/metabolismo , Melanoma/patología , Ratones SCID , Mutación , Neoplasia Residual/metabolismo , Neoplasia Residual/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...