Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 11(1): 14932, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34294755

RESUMEN

Whole-genome duplication (WGD) generates polyploid cells possessing more than two copies of the genome and is among the most common genetic abnormalities in cancer. The frequency of WGD increases in advanced and metastatic tumors, and WGD is associated with poor prognosis in diverse tumor types, suggesting a functional role for polyploidy in tumor progression. Experimental evidence suggests that polyploidy has both tumor-promoting and suppressing effects, but how polyploidy regulates tumor progression remains unclear. Using a genetically engineered mouse model of Her2-driven breast cancer, we explored the prevalence and consequences of whole-genome duplication during tumor growth and recurrence. While primary tumors in this model are invariably diploid, nearly 40% of recurrent tumors undergo WGD. WGD in recurrent tumors was associated with increased chromosomal instability, decreased proliferation and increased survival in stress conditions. The effects of WGD on tumor growth were dependent on tumor stage. Surprisingly, in recurrent tumor cells WGD slowed tumor formation, growth rate and opposed the process of recurrence, while WGD promoted the growth of primary tumors. These findings highlight the importance of identifying conditions that promote the growth of polyploid tumors, including the cooperating genetic mutations that allow cells to overcome the barriers to WGD tumor cell growth and proliferation.


Asunto(s)
Duplicación de Gen , Hibridación Fluorescente in Situ/métodos , Cariotipificación/métodos , Neoplasias Mamarias Experimentales/genética , Receptor ErbB-2/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Inestabilidad Cromosómica , Evolución Molecular , Femenino , Humanos , Ratones , Ratones Transgénicos , Poliploidía
2.
Cell Rep ; 33(5): 108341, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33147463

RESUMEN

Dysregulated gene expression is a common feature of cancer and may underlie some aspects of tumor progression, including tumor relapse. Here, we show that recurrent mammary tumors exhibit global changes in gene expression and histone modifications and acquire dependence on the G9a histone methyltransferase. Genetic ablation of G9a delays tumor recurrence, and pharmacologic inhibition of G9a slows the growth of recurrent tumors. Mechanistically, G9a activity is required to silence pro-inflammatory cytokines, including tumor necrosis factor (TNF), through H3K9 methylation at gene promoters. G9a inhibition induces re-expression of these cytokines, leading to p53 activation and necroptosis. Recurrent tumors upregulate receptor interacting protein kinase-3 (RIPK3) expression and are dependent upon RIPK3 activity. High RIPK3 expression renders recurrent tumors sensitive to necroptosis following G9a inhibition. These findings demonstrate that G9a-mediated silencing of pro-necroptotic proteins is a critical step in tumor recurrence and suggest that G9a is a targetable dependency in recurrent breast cancer.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/metabolismo , Inflamación/patología , Neoplasias Mamarias Animales/enzimología , Neoplasias Mamarias Animales/patología , Recurrencia Local de Neoplasia/patología , Animales , Muerte Celular , Supervivencia Celular , Epigénesis Genética , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Neoplasias Mamarias Animales/genética , Ratones Desnudos , Necroptosis , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Factores de Riesgo , Transcripción Genética , Factor de Necrosis Tumoral alfa/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
3.
Nat Commun ; 11(1): 5017, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-33024122

RESUMEN

The survival and recurrence of residual tumor cells following therapy constitutes one of the biggest obstacles to obtaining cures in breast cancer, but it remains unclear how the clonal composition of tumors changes during relapse. We use cellular barcoding to monitor clonal dynamics during tumor recurrence in vivo. We find that clonal diversity decreases during tumor regression, residual disease, and recurrence. The recurrence of dormant residual cells follows several distinct routes. Approximately half of the recurrent tumors exhibit clonal dominance with a small number of subclones comprising the vast majority of the tumor; these clonal recurrences are frequently dependent upon Met gene amplification. A second group of recurrent tumors comprises thousands of subclones, has a clonal architecture similar to primary tumors, and is dependent upon the Jak/Stat pathway. Thus the regrowth of dormant tumors proceeds via multiple routes, producing recurrent tumors with distinct clonal composition, genetic alterations, and drug sensitivities.


Asunto(s)
Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Animales , Línea Celular Tumoral , Crizotinib/farmacología , Doxiciclina/farmacología , Transición Epitelial-Mesenquimal/genética , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones Desnudos , Recurrencia Local de Neoplasia/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/genética , Receptor ErbB-2/genética , Análisis de la Célula Individual , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Nat Metab ; 2(4): 318-334, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32691018

RESUMEN

The survival and recurrence of dormant tumour cells following therapy is a leading cause of death in cancer patients. The metabolic properties of these cells are likely distinct from those of rapidly growing tumours. Here we show that Her2 down-regulation in breast cancer cells promotes changes in cellular metabolism, culminating in oxidative stress and compensatory upregulation of the antioxidant transcription factor, NRF2. NRF2 is activated during dormancy and in recurrent tumours in animal models and breast cancer patients with poor prognosis. Constitutive activation of NRF2 accelerates recurrence, while suppression of NRF2 impairs it. In recurrent tumours, NRF2 signalling induces a transcriptional metabolic reprogramming to re-establish redox homeostasis and upregulate de novo nucleotide synthesis. The NRF2-driven metabolic state renders recurrent tumour cells sensitive to glutaminase inhibition, which prevents reactivation of dormant tumour cells in vitro, suggesting that NRF2-high dormant and recurrent tumours may be targeted. These data provide evidence that NRF2-driven metabolic reprogramming promotes the recurrence of dormant breast cancer.


Asunto(s)
Factor 2 Relacionado con NF-E2/metabolismo , Nucleótidos/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Muerte Celular , Línea Celular Tumoral , Regulación hacia Abajo , Femenino , Homeostasis , Humanos , Ratones , Recurrencia Local de Neoplasia , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Receptor ErbB-2/metabolismo , Transducción de Señal , Transcripción Genética
5.
Cell Rep ; 20(4): 999-1015, 2017 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-28746882

RESUMEN

Combinatorial inhibition of effector and feedback pathways is a promising treatment strategy for KRAS mutant cancers. However, the particular pathways that should be targeted to optimize therapeutic responses are unclear. Using CRISPR/Cas9, we systematically mapped the pathways whose inhibition cooperates with drugs targeting the KRAS effectors MEK, ERK, and PI3K. By performing 70 screens in models of KRAS mutant colorectal, lung, ovarian, and pancreas cancers, we uncovered universal and tissue-specific sensitizing combinations involving inhibitors of cell cycle, metabolism, growth signaling, chromatin regulation, and transcription. Furthermore, these screens revealed secondary genetic modifiers of sensitivity, yielding a SRC inhibitor-based combination therapy for KRAS/PIK3CA double-mutant colorectal cancers (CRCs) with clinical potential. Surprisingly, acquired resistance to combinations of growth signaling pathway inhibitors develops rapidly following treatment, but by targeting signaling feedback or apoptotic priming, it is possible to construct three-drug combinations that greatly delay its emergence.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Apoptosis/genética , Apoptosis/fisiología , Línea Celular Tumoral , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Neoplasias Colorrectales/genética , Humanos , Mutación/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Transducción de Señal/genética , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...