Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Mol Oncol ; 16(17): 3128-3145, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35398967

RESUMEN

As precision medicine increases the response rate of treatment, tumors frequently bypass inhibition, and reoccur. In order for treatment to be effective long term, the mechanisms enabling treatment adaptation need to be understood. Here, we report a mouse model that, in the absence of p53 and the presence of oncogenic KrasG12D , develops breast tumors. Upon inactivation of KrasG12D , tumors initially regress and enter remission. Subsequently, the majority of tumors adapt to the withdrawal of KrasG12D expression and return. KrasG12D -independent tumor cells show a strong mesenchymal profile with active RAS-RAF-MEK-ERK (MAPK/ERK) signaling. Both KrasG12D -dependent and KrasG12D -independent tumors display a high level of genomic instability, and KrasG12D -independent tumors harbor numerous amplified genes that can activate the MAPK/ERK signaling pathway. Our study identifies both epithelial-mesenchymal transition (EMT) and active MAPK/ERK signaling in tumors that adapt to oncogenic KrasG12D withdrawal in a novel Trp53-/- breast cancer mouse model. To achieve long-lasting responses in the clinic to RAS-fueled cancer, treatment will need to focus in parallel on obstructing tumors from adapting to oncogene inhibition.


Asunto(s)
Transición Epitelial-Mesenquimal , Genes ras , Animales , Carcinogénesis/genética , Transición Epitelial-Mesenquimal/genética , Sistema de Señalización de MAP Quinasas , Ratones , Mutación/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Transducción de Señal
2.
Sci Adv ; 8(45): eabm3548, 2022 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-36351009

RESUMEN

Metastasis is responsible for most breast cancer-related deaths; however, identifying the cellular determinants of metastasis has remained challenging. Here, we identified a minority population of immature THY1+/VEGFA+ tumor epithelial cells in human breast tumor biopsies that display angiogenic features and are marked by the expression of the oncogene, LMO2. Higher abundance of LMO2+ basal cells correlated with tumor endothelial content and predicted poor distant recurrence-free survival in patients. Using MMTV-PyMT/Lmo2CreERT2 mice, we demonstrated that Lmo2 lineage-traced cells integrate into the vasculature and have a higher propensity to metastasize. LMO2 knockdown in human breast tumors reduced lung metastasis by impairing intravasation, leading to a reduced frequency of circulating tumor cells. Mechanistically, we find that LMO2 binds to STAT3 and is required for STAT3 activation by tumor necrosis factor-α and interleukin-6. Collectively, our study identifies a population of metastasis-initiating cells with angiogenic features and establishes the LMO2-STAT3 signaling axis as a therapeutic target in breast cancer metastasis.


Asunto(s)
Neoplasias de la Mama , Neoplasias Pulmonares , Humanos , Ratones , Animales , Femenino , Neoplasias de la Mama/patología , Neoplasias Pulmonares/metabolismo , Transducción de Señal , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo
3.
Science ; 367(6476): 405-411, 2020 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-31974247

RESUMEN

Single-cell RNA sequencing (scRNA-seq) is a powerful approach for reconstructing cellular differentiation trajectories. However, inferring both the state and direction of differentiation is challenging. Here, we demonstrate a simple, yet robust, determinant of developmental potential-the number of expressed genes per cell-and leverage this measure of transcriptional diversity to develop a computational framework (CytoTRACE) for predicting differentiation states from scRNA-seq data. When applied to diverse tissue types and organisms, CytoTRACE outperformed previous methods and nearly 19,000 annotated gene sets for resolving 52 experimentally determined developmental trajectories. Additionally, it facilitated the identification of quiescent stem cells and revealed genes that contribute to breast tumorigenesis. This study thus establishes a key RNA-based feature of developmental potential and a platform for delineation of cellular hierarchies.


Asunto(s)
Diferenciación Celular/genética , Neoplasias/genética , ARN Citoplasmático Pequeño/genética , RNA-Seq/métodos , Análisis de la Célula Individual/métodos , Transcripción Genética , Animales , Secuencia de Bases , Variación Genética , Humanos , Ratones
4.
Cell Stem Cell ; 27(2): 284-299.e8, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32693087

RESUMEN

SMAD pathways govern epithelial proliferation, and transforming growth factor ß (TGF-ß and BMP signaling through SMAD members has distinct effects on mammary development and homeostasis. Here, we show that LEFTY1, a secreted inhibitor of NODAL/SMAD2 signaling, is produced by mammary progenitor cells and, concomitantly, suppresses SMAD2 and SMAD5 signaling to promote long-term proliferation of normal and malignant mammary epithelial cells. In contrast, BMP7, a NODAL antagonist with context-dependent functions, is produced by basal cells and restrains progenitor cell proliferation. In normal mouse epithelium, LEFTY1 expression in a subset of luminal cells and rare basal cells opposes BMP7 to promote ductal branching. LEFTY1 binds BMPR2 to suppress BMP7-induced activation of SMAD5, and this LEFTY1-BMPR2 interaction is specific to tumor-initiating cells in triple-negative breast cancer xenografts that rely on LEFTY1 for growth. These results suggest that LEFTY1 is an endogenous dual-SMAD inhibitor and that suppressing its function may represent a therapeutic vulnerability in breast cancer.


Asunto(s)
Transducción de Señal , Factor de Crecimiento Transformador beta , Animales , Carcinogénesis , Transformación Celular Neoplásica , Ratones
5.
Nat Commun ; 8(1): 1669, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29162812

RESUMEN

Previous studies have proposed that epithelial to mesenchymal transition (EMT) in breast cancer cells regulates metastasis, stem cell properties and chemo-resistance; most studies were based on in vitro culture of cell lines and mouse transgenic cancer models. However, the identity and function of cells expressing EMT-associated genes in normal murine mammary gland homeostasis and human breast cancer still remains under debate. Using in vivo lineage tracing and triple negative breast cancer (TNBC) patient derived xenografts we demonstrate that the repopulating capacity in normal mammary epithelial cells and tumorigenic capacity in TNBC is independent of expression of EMT-associated genes. In breast cancer, while a subset of cells with epithelial and mesenchymal phenotypes have stem cell activity, in many cells that have lost epithelial characteristics with increased expression of mesenchymal genes, have decreased tumor-initiating capacity and plasticity. These findings have implications for the development of effective therapeutic agents targeting tumor-initiating cells.


Asunto(s)
Mama/metabolismo , Transformación Celular Neoplásica/genética , Transición Epitelial-Mesenquimal/genética , Perfilación de la Expresión Génica , Neoplasias de la Mama Triple Negativas/genética , Animales , Mama/citología , Mama/fisiología , Células Epiteliales/metabolismo , Femenino , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Regeneración/genética , Trasplante Heterólogo , Neoplasias de la Mama Triple Negativas/patología
6.
Cell Stem Cell ; 20(2): 247-260.e5, 2017 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-28041896

RESUMEN

Stem cells in many tissues sustain themselves by entering a quiescent state to avoid genomic insults and to prevent exhaustion caused by excessive proliferation. In the mammary gland, the identity and characteristics of quiescent epithelial stem cells are not clear. Here, we identify a quiescent mammary epithelial cell population expressing high levels of Bcl11b and located at the interface between luminal and basal cells. Bcl11bhigh cells are enriched for cells that can regenerate mammary glands in secondary transplants. Loss of Bcl11b leads to a Cdkn2a-dependent exhaustion of ductal epithelium and loss of epithelial cell regenerative capacity. Gain- and loss-of-function studies show that Bcl11b induces cells to enter the G0 phase of the cell cycle and become quiescent. Taken together, these results suggest that Bcl11b acts as a central intrinsic regulator of mammary epithelial stem cell quiescence and exhaustion and is necessary for long-term maintenance of the mammary gland.


Asunto(s)
Ciclo Celular , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Proteínas Represoras/metabolismo , Células Madre/citología , Células Madre/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Antígenos CD/metabolismo , Linaje de la Célula , Proliferación Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Epiteliales/metabolismo , Femenino , Eliminación de Gen , Homeostasis , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones Endogámicos C57BL , Ratones Noqueados , Regeneración/fisiología
7.
Elife ; 32014 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-25406066

RESUMEN

MicroRNAs (miRNAs) are important regulators of stem and progenitor cell functions. We previously reported that miR-142 and miR-150 are upregulated in human breast cancer stem cells (BCSCs) as compared to the non-tumorigenic breast cancer cells. In this study, we report that miR-142 efficiently recruits the APC mRNA to an RNA-induced silencing complex, activates the canonical WNT signaling pathway in an APC-suppression dependent manner, and activates the expression of miR-150. Enforced expression of miR-142 or miR-150 in normal mouse mammary stem cells resulted in the regeneration of hyperproliferative mammary glands in vivo. Knockdown of endogenous miR-142 effectively suppressed organoid formation by BCSCs and slowed tumor growth initiated by human BCSCs in vivo. These results suggest that in some tumors, miR-142 regulates the properties of BCSCs at least in part by activating the WNT signaling pathway and miR-150 expression.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinogénesis/patología , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Vía de Señalización Wnt , Proteína de la Poliposis Adenomatosa del Colon/genética , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Proteínas Argonautas/metabolismo , Secuencia de Bases , Carcinogénesis/genética , Proliferación Celular , Células Clonales , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Hiperplasia , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Ratones , MicroARNs/genética , Datos de Secuencia Molecular , Organoides/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Complejo Silenciador Inducido por ARN/metabolismo , Transcripción Genética , Regulación hacia Arriba/genética , Vía de Señalización Wnt/genética
8.
Nat Cell Biol ; 16(12): 1238-48, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25362351

RESUMEN

It has been postulated that there is a link between inflammation and cancer. Here we describe a role for cell-intrinsic toll-like receptor-2 (TLR2; which is involved in inflammatory response) signalling in normal intestinal and mammary epithelial cells and oncogenesis. The downstream effectors of TLR2 are expressed by normal intestinal and mammary epithelia, including the stem/progenitor cells. Deletion of MYD88 or TLR2 in the intestinal epithelium markedly reduces DSS-induced colitis regeneration and spontaneous tumour development in mice. Limiting dilution transplantations of breast epithelial cells devoid of TLR2 or MYD88 revealed a significant decrease in mammary repopulating unit frequency compared with the control. Inhibition of TLR2, its co-receptor CD14, or its downstream targets MYD88 and IRAK1 inhibits growth of human breast cancers in vitro and in vivo. These results suggest that inhibitors of the TLR2 pathway merit investigation as possible therapeutic and chemoprevention agents.


Asunto(s)
Neoplasias de la Mama/patología , Mama/patología , Carcinogénesis/metabolismo , Neoplasias del Colon/patología , Mucosa Intestinal/patología , Factor 88 de Diferenciación Mieloide/metabolismo , Receptor Toll-Like 2/metabolismo , Animales , Antígenos de Neoplasias/metabolismo , Mama/metabolismo , Neoplasias de la Mama/genética , Carcinogénesis/genética , Moléculas de Adhesión Celular/metabolismo , Colitis/inducido químicamente , Colitis/patología , Neoplasias del Colon/genética , Sulfato de Dextran , Molécula de Adhesión Celular Epitelial , Epitelio/patología , Femenino , Células HEK293 , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/antagonistas & inhibidores , Quinasas Asociadas a Receptores de Interleucina-1/genética , Mucosa Intestinal/metabolismo , Antígenos Comunes de Leucocito/genética , Receptores de Lipopolisacáridos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/antagonistas & inhibidores , Factor 88 de Diferenciación Mieloide/genética , Trasplante de Neoplasias , Interferencia de ARN , ARN Interferente Pequeño , Receptores Acoplados a Proteínas G/metabolismo , Regeneración/genética , Transducción de Señal , Receptor Toll-Like 2/antagonistas & inhibidores , Receptor Toll-Like 2/genética , Células Tumorales Cultivadas
9.
Cancer Discov ; 2(6): 540-53, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22628407

RESUMEN

Colorectal cancer is a classic example of a tumor that progresses through multiple distinct stages in its evolution. To understand the mechanisms regulating the transition from indolent to invasive disease, we profiled somatic copy number alterations in noninvasive adenomas and invasive adenocarcinomas from Apc and DNA mismatch repair (MMR) mutant mouse models. We identified a recurrent amplicon on mouse chromosome 8 that encodes microRNA (miRNA) 23a and -27a (miR). miR-23a and -27a levels are upregulated in mouse intestinal adenocarcinomas, primary tumors from patients with stage I/II colorectal cancers, as well as in human colorectal cancer cell lines and cancer stem cells. Functionally, miR-23a promotes the migration and invasion of colorectal cancer cells and stem cells, whereas miR-27a primarily promotes proliferation. We computationally and experimentally validated that metastasis suppressor 1 (MTSS1) is a direct miR-23a target and similarly validated that the ubiquitin ligase FBXW7 is a direct miR-27a target. Analyses of computationally predicted target genes in microarray data sets of patients with colorectal cancers are consistent with a role for miR-23a, but not miR-27a, specifically in invasive colorectal cancers.


Asunto(s)
Adenocarcinoma/genética , Adenoma/genética , Neoplasias Colorrectales/genética , MicroARNs/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adenoma/metabolismo , Adenoma/patología , Animales , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Variaciones en el Número de Copia de ADN , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Invasividad Neoplásica/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
10.
Cancer Res ; 70(4): 1469-78, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20145124

RESUMEN

NOTCH signaling is critical for specifying the intestinal epithelial cell lineage and for initiating colorectal adenomas and colorectal cancers (CRC). Based on evidence that NOTCH is important for the maintenance and self-renewal of cancer-initiating cells in other malignancies, we studied the role of NOTCH signaling in colon cancer-initiating cells (CCIC). Tumors formed by CCICs maintain many properties of the primary CRCs from which they were derived, such as glandular organization, cell polarity, gap junctions, and expression of characteristic CRC molecular markers. Furthermore, CCICs have the property of self-renewal. In this study, we show that NOTCH signaling is 10- to 30-fold higher in CCIC compared with widely used colon cancer cell lines. Using small-molecule inhibition and short hairpin RNA knockdown, we show that NOTCH prevents CCIC apoptosis through repression of cell cycle kinase inhibitor p27 and transcription factor ATOH1. NOTCH is also critical to intrinsic maintenance of CCIC self-renewal and the repression of secretory cell lineage differentiation genes such as MUC2. Our findings describe a novel human cell system to study NOTCH signaling in CRC tumor initiation and suggest that inhibition of NOTCH signaling may improve CRC chemoprevention and chemotherapy.


Asunto(s)
Carcinoma/genética , Diferenciación Celular/genética , Proliferación Celular , Transformación Celular Neoplásica/genética , Neoplasias del Colon/genética , Células Enteroendocrinas/fisiología , Células Madre Neoplásicas/fisiología , Receptor Notch1/fisiología , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/fisiología , Carcinoma/patología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Neoplasias del Colon/patología , Células Enteroendocrinas/efectos de los fármacos , Células Enteroendocrinas/metabolismo , Células Enteroendocrinas/patología , Células HCT116 , Células HT29 , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , ARN Interferente Pequeño/farmacología , Receptor Notch1/antagonistas & inhibidores , Receptor Notch1/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA