Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Cell ; 148(1-2): 349-61, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-22265420

RESUMEN

Metastasis is the leading cause of cancer-associated death but has been difficult to study because it involves a series of rare, stochastic events. To capture these events, we developed a sensitive method to tag and track pancreatic epithelial cells in a mouse model of pancreatic cancer. Tagged cells invaded and entered the bloodstream unexpectedly early, before frank malignancy could be detected by rigorous histologic analysis; this behavior was widely associated with epithelial-to-mesenchymal transition (EMT). Circulating pancreatic cells maintained a mesenchymal phenotype, exhibited stem cell properties, and seeded the liver. EMT and invasiveness were most abundant at inflammatory foci, and induction of pancreatitis increased the number of circulating pancreatic cells. Conversely, treatment with the immunosuppressive agent dexamethasone abolished dissemination. These results provide insight into the earliest events of cellular invasion in situ and suggest that inflammation enhances cancer progression in part by facilitating EMT and entry into the circulation.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Transición Epitelial-Mesenquimal , Invasividad Neoplásica , Neoplasias Pancreáticas/patología , Animales , Carcinoma Ductal Pancreático/inmunología , Modelos Animales de Enfermedad , Humanos , Ratones , Células Madre Neoplásicas/patología , Neoplasias Pancreáticas/inmunología , Pancreatitis/patología
2.
Genes Dev ; 27(7): 719-24, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23520387

RESUMEN

Cellular reprogramming-the ability to interconvert distinct cell types with defined factors-is transforming the field of regenerative medicine. However, this phenomenon has rarely been observed in vivo without exogenous factors. Here, we report that activation of Notch, a signaling pathway that mediates lineage segregation during liver development, is sufficient to reprogram hepatocytes into biliary epithelial cells (BECs). Moreover, using lineage tracing, we show that hepatocytes undergo widespread hepatocyte-to-BEC reprogramming following injuries that provoke a biliary response, a process requiring Notch. These results provide direct evidence that mammalian regeneration prompts extensive and dramatic changes in cellular identity under injury conditions.


Asunto(s)
Células Epiteliales/citología , Hepatocitos/citología , Regeneración Hepática/fisiología , Animales , Linaje de la Célula , Células Epiteliales/metabolismo , Hepatocitos/metabolismo , Ratones , Receptores Notch/metabolismo , Transducción de Señal , Células Madre/citología
4.
Gastroenterology ; 151(3): 540-553.e14, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27318148

RESUMEN

BACKGROUND & AIMS: The ETS-transcription factor ETV1 is involved in epithelial-mesenchymal transition during pancreatic development and is induced in mouse pancreatic intraepithelial neoplasia (PanIN) and pancreatic ductal adenocarcinoma (PDAC). We investigated the function of ETV1 in stromal expansion of PDAC and metastasis, as well as its effects on a novel downstream target Sparc, which encodes a matricellular protein found in PDAC stroma that has been associated with invasiveness, metastasis and poor patient outcomes. METHODS: Pancreatic ductal cells were isolated from Pdx1Cre;Kras(G12D/+) mice (PanIN), Pdx1Cre;Kras(G12D/+);p53(fl/+) and Pdx1Cre;Kras(G12D/+);p53(fl/+);Rosa26(YFP) mice (PDAC), and Pdx1Cre;Kras(G12D/+);p53(fl/+);Sparc(-/-) mice. Cells were grown in 3-dimensional organoid culture to analyze morphology, proliferation, and invasion. Human PanIN and PDAC tissues were evaluated for ETV1 expression. Orthotopic pancreatic transplants of ETV1-overexpressing PDAC and respective control cells were performed. RESULTS: ETV1 expression was significantly increased in human PanINs and, even more so, in primary and metastatic PDAC. Analyses of mouse orthotopic xenografts revealed that ETV1 induced significantly larger primary tumors than controls, with significantly increased stromal expansion, ascites and metastases. In 3-dimensional organoids, ETV1 disrupted cyst architecture, induced EMT, and increased invasive capacity. Furthermore, we identified Sparc as a novel functional gene target of Etv1 by luciferase assays, and SPARC and ETV1 proteins co-localized in vivo. Disruption of Sparc abrogates the phenotype of stromal expansion and metastasis found with ETV1 overexpression in vivo. We identified hyaluronan synthase 2 (Has2) as another novel downstream factor of Etv1; that may mediate ETV1's significant expansion of hyaluronic acid in PDAC stroma. Conversely, disruption of Etv1 in PDAC mice (Pdx1Cre;Kras(G12D/+);p53(fl/+);Rosa26(YFP);Cre;Etv1(fl/fl)) reduced levels of SPARC and hyaluronic acid in the stroma. CONCLUSIONS: ETV1 is critical in the desmoplastic stromal expansion and metastatic progression of pancreatic cancer in mice, mediated functionally in part through Sparc and Has2.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Proteínas de Unión al ADN/metabolismo , Conductos Pancreáticos/citología , Neoplasias Pancreáticas/genética , Células del Estroma/metabolismo , Factores de Transcripción/metabolismo , Animales , Carcinoma Ductal Pancreático/patología , Transición Epitelial-Mesenquimal/genética , Ratones , Metástasis de la Neoplasia/genética , Neoplasias Pancreáticas/patología
5.
J Exp Med ; 216(5): 1016-1026, 2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-30975895

RESUMEN

Epithelial-mesenchymal transition (EMT) is a developmental process whereby stationary, adherent cells acquire the ability to migrate. EMT is critical for dramatic cellular movements during embryogenesis; however, tumor cells can reactivate EMT programs, which increases their aggressiveness. In addition to motility, EMT is associated with enhanced stem cell properties and drug resistance; thus it can drive metastasis, tumor recurrence, and therapy resistance in the context of cancer. However, the precise requirements for EMT in metastasis have not been fully delineated, with different tumor types relying on discrete EMT effectors. Most tumor cells do not undergo a full EMT, but rather adopt some qualities of mesenchymal cells and maintain some epithelial characteristics. Emerging evidence suggests that partial EMT can drive distinct migratory properties and enhance the epithelial-mesenchymal plasticity of cancer cells as well as cell fate plasticity. This review discusses the diverse regulatory mechanisms and functional consequences of EMT, with an emphasis on the importance of partial EMT.


Asunto(s)
Transición Epitelial-Mesenquimal , Metástasis de la Neoplasia , Neoplasias/patología , Animales , Biomarcadores de Tumor , Movimiento Celular , Plasticidad de la Célula , Células Epiteliales/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones
6.
Nat Commun ; 10(1): 527, 2019 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-30692542

RESUMEN

The original version of this Article contained an error in the spelling of the author Daniel D. Liu, which was incorrectly given as Daniel Liu. This has now been corrected in both the PDF and HTML versions of the Article.

7.
Dev Cell ; 45(6): 681-695.e4, 2018 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-29920274

RESUMEN

Epithelial-mesenchymal transition (EMT) is strongly implicated in tumor cell invasion and metastasis. EMT is thought to be regulated primarily at the transcriptional level through the repressive activity of EMT transcription factors. However, these classical mechanisms have been parsed out almost exclusively in vitro, leaving questions about the programs driving EMT in physiological contexts. Here, using a lineage-labeled mouse model of pancreatic ductal adenocarcinoma to study EMT in vivo, we found that most tumors lose their epithelial phenotype through an alternative program involving protein internalization rather than transcriptional repression, resulting in a "partial EMT" phenotype. Carcinoma cells utilizing this program migrate as clusters, contrasting with the single-cell migration pattern associated with traditionally defined EMT mechanisms. Moreover, many breast and colorectal cancer cell lines utilize this alternative program to undergo EMT. Collectively, these results suggest that carcinoma cells have different ways of losing their epithelial program, resulting in distinct modes of invasion and dissemination.


Asunto(s)
Plasticidad de la Célula/fisiología , Células Epiteliales/fisiología , Transición Epitelial-Mesenquimal/fisiología , Animales , Cadherinas/metabolismo , Cadherinas/fisiología , Línea Celular Tumoral , Movimiento Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Ratones , Invasividad Neoplásica/genética , Neoplasias Pancreáticas/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Neoplasias Pancreáticas
8.
Nat Commun ; 9(1): 5005, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30479345

RESUMEN

Epithelial-mesenchymal transition (EMT) have been extensively characterized in development and cancer, and its dynamics have been modeled as a non-linear process. However, less is known about how such dynamics may affect its biological impact. Here, we use mathematical modeling and experimental analysis of the TGF-ß-induced EMT to reveal a non-linear hysteretic response of E-cadherin repression tightly controlled by the strength of the miR-200s/ZEBs negative feedback loop. Hysteretic EMT conveys memory state, ensures rapid and robust cellular response and enables EMT to persist long after withdrawal of stimuli. Importantly, while both hysteretic and non-hysteretic EMT confer similar morphological changes and invasive potential of cancer cells, only hysteretic EMT enhances lung metastatic colonization efficiency. Cells that undergo hysteretic EMT differentially express subsets of stem cell and extracellular matrix related genes with significant clinical prognosis value. These findings illustrate distinct biological impact of EMT depending on the dynamics of the transition.


Asunto(s)
Transición Epitelial-Mesenquimal , Metástasis de la Neoplasia/patología , Animales , Cadherinas/metabolismo , Línea Celular Tumoral , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Retroalimentación Fisiológica , Femenino , Ratones Endogámicos BALB C , MicroARNs/genética , MicroARNs/metabolismo , Modelos Biológicos , Metástasis de la Neoplasia/genética , Factor de Crecimiento Transformador beta/farmacología , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
9.
ACS Nano ; 11(11): 11182-11193, 2017 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-29019651

RESUMEN

Circulating exosomes contain a wealth of proteomic and genetic information, presenting an enormous opportunity in cancer diagnostics. While microfluidic approaches have been used to successfully isolate cells from complex samples, scaling these approaches for exosome isolation has been limited by the low throughput and susceptibility to clogging of nanofluidics. Moreover, the analysis of exosomal biomarkers is confounded by substantial heterogeneity between patients and within a tumor itself. To address these challenges, we developed a multichannel nanofluidic system to analyze crude clinical samples. Using this platform, we isolated exosomes from healthy and diseased murine and clinical cohorts, profiled the RNA cargo inside of these exosomes, and applied a machine learning algorithm to generate predictive panels that could identify samples derived from heterogeneous cancer-bearing individuals. Using this approach, we classified cancer and precancer mice from healthy controls, as well as pancreatic cancer patients from healthy controls, in blinded studies.


Asunto(s)
Exosomas/genética , Técnicas Analíticas Microfluídicas/métodos , Neoplasias Pancreáticas/diagnóstico , Proteómica , Animales , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Exosomas/patología , Humanos , Aprendizaje Automático , Ratones , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , ARN Mensajero/genética
10.
Dis Model Mech ; 9(2): 105-14, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26839398

RESUMEN

The hallmark of embryonic development is regulation - the tendency for cells to find their way into organized and 'well behaved' structures - whereas cancer is characterized by dysregulation and disorder. At face value, cancer biology and developmental biology would thus seem to have little to do with each other. But if one looks beneath the surface, embryos and cancers share a number of cellular and molecular features. Embryos arise from a single cell and undergo rapid growth involving cell migration and cell-cell interactions: features that are also seen in the context of cancer. Consequently, many of the experimental tools that have been used to study embryogenesis for over a century are well-suited to studying cancer. This article will review the similarities between embryogenesis and cancer progression and discuss how some of the concepts and techniques used to understand embryos are now being adapted to provide insight into tumorigenesis, from the origins of cancer cells to metastasis.


Asunto(s)
Desarrollo Embrionario , Neoplasias/patología , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor , Carcinogénesis , Transición Epitelial-Mesenquimal , Humanos , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/genética
11.
Cold Spring Harb Protoc ; 2016(1): pdb.prot078360, 2016 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-26729902

RESUMEN

Pancreatic ductal adenocarcinoma is an aggressive disease with a 5-yr survival rate of only 5%. The location of the pancreas in the abdomen, where it is obscured by other organs, makes it a difficult tissue to study and manipulate. This protocol describes in detail how to orthotopically inject cancer cells into the pancreas in mice. This technique is particularly useful when the cells must be manipulated in ways that cannot be modeled genetically.


Asunto(s)
Separación Celular/métodos , Células Epiteliales/fisiología , Transición Epitelial-Mesenquimal/fisiología , Neoplasias Pancreáticas/patología , Animales , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Ratones , Trasplante de Neoplasias
12.
Cold Spring Harb Protoc ; 2016(1): pdb.prot078352, 2016 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-26729901

RESUMEN

Transgenic mice that express conditional reporters allow for the isolation of specific cell lineages. These cells can be further stratified by gene expression and collected by fluorescence-activated cell sorting (FACS) for further analysis. Using Cre-recombinase (Cre) technology we have generated a transgenic mouse line termed PKCY in which all pancreatic epithelial cells and therefore all pancreatic cancer cells are constitutively labeled with yellow fluorescent protein (YFP). We have used immunofluorescent staining for E-cadherin to divide the YFP(+) tumor population into epithelial cells (E-cadherin positive) and cells that have undergone an epithelial-to-mesenchymal transition (EMT; E-cadherin negative). This protocol describes how to prepare a tumor sample for FACS, with an emphasis on separating epithelial and EMT populations. These cells can then be used for a number of applications including, but not limited to, the generation of cell lines, gene-expression analysis by quantitative polymerase chain reaction (qPCR) or RNA sequencing, DNA sequencing, chromatin immunoprecipitation, and western blots.


Asunto(s)
Movimiento Celular/fisiología , Células Epiteliales/fisiología , Transición Epitelial-Mesenquimal/fisiología , Citometría de Flujo/métodos , Animales , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Ratones
13.
Nat Commun ; 7: 12819, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27628423

RESUMEN

Most cancer-associated deaths result from metastasis. However, it remains unknown whether the size, microenvironment or other features of a metastatic lesion dictate its behaviour or determine the efficacy of chemotherapy in the adjuvant (micrometastatic) setting. Here we delineate the natural history of metastasis in an autochthonous model of pancreatic ductal adenocarcinoma (PDAC), using lineage tracing to examine the evolution of disseminated cancer cells and their associated microenvironment. With increasing size, lesions shift from mesenchymal to epithelial histology, become hypovascular and accumulate a desmoplastic stroma, ultimately recapitulating the primary tumours from which they arose. Moreover, treatment with gemcitabine and nab-paclitaxel significantly reduces the overall number of metastases by inducing cell death in lesions of all sizes, challenging the paradigm that PDAC stroma imposes a critical barrier to drug delivery. These results illuminate the cellular dynamics of metastatic progression and suggest that adjuvant chemotherapy affords a survival benefit by directly targeting micrometastases.


Asunto(s)
Carcinoma Ductal Pancreático/secundario , Neoplasias Hepáticas/secundario , Hígado/patología , Neoplasias Pancreáticas/patología , Microambiente Tumoral , Anciano , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carcinoma Ductal Pancreático/irrigación sanguínea , Carcinoma Ductal Pancreático/tratamiento farmacológico , Linaje de la Célula , Femenino , Humanos , Hígado/efectos de los fármacos , Neoplasias Hepáticas/irrigación sanguínea , Neoplasias Hepáticas/tratamiento farmacológico , Masculino , Ratones , Persona de Mediana Edad , Metástasis de la Neoplasia , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/tratamiento farmacológico
14.
Nat Cell Biol ; 17(6): 816-26, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25985394

RESUMEN

Pancreatic ductal adenocarcinomas (PDACs) are highly metastatic with poor prognosis, mainly due to delayed detection. We hypothesized that intercellular communication is critical for metastatic progression. Here, we show that PDAC-derived exosomes induce liver pre-metastatic niche formation in naive mice and consequently increase liver metastatic burden. Uptake of PDAC-derived exosomes by Kupffer cells caused transforming growth factor ß secretion and upregulation of fibronectin production by hepatic stellate cells. This fibrotic microenvironment enhanced recruitment of bone marrow-derived macrophages. We found that macrophage migration inhibitory factor (MIF) was highly expressed in PDAC-derived exosomes, and its blockade prevented liver pre-metastatic niche formation and metastasis. Compared with patients whose pancreatic tumours did not progress, MIF was markedly higher in exosomes from stage I PDAC patients who later developed liver metastasis. These findings suggest that exosomal MIF primes the liver for metastasis and may be a prognostic marker for the development of PDAC liver metastasis.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Exosomas/metabolismo , Neoplasias Hepáticas/patología , Factores Inhibidores de la Migración de Macrófagos/biosíntesis , Neoplasias Pancreáticas/patología , Animales , Secuencia de Bases , Células de la Médula Ósea/inmunología , Línea Celular Tumoral , Movimiento Celular , Femenino , Fibronectinas/biosíntesis , Regulación Neoplásica de la Expresión Génica , Células Estrelladas Hepáticas/patología , Humanos , Hígado/citología , Hígado/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundario , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Lesiones Precancerosas/patología , Interferencia de ARN , ARN Interferente Pequeño , Análisis de Secuencia de ARN , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
15.
Cell Metab ; 20(2): 306-319, 2014 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-24998913

RESUMEN

Histone acetylation plays important roles in gene regulation, DNA replication, and the response to DNA damage, and it is frequently deregulated in tumors. We postulated that tumor cell histone acetylation levels are determined in part by changes in acetyl coenzyme A (acetyl-CoA) availability mediated by oncogenic metabolic reprogramming. Here, we demonstrate that acetyl-CoA is dynamically regulated by glucose availability in cancer cells and that the ratio of acetyl-CoA:coenzyme A within the nucleus modulates global histone acetylation levels. In vivo, expression of oncogenic Kras or Akt stimulates histone acetylation changes that precede tumor development. Furthermore, we show that Akt's effects on histone acetylation are mediated through the metabolic enzyme ATP-citrate lyase and that pAkt(Ser473) levels correlate significantly with histone acetylation marks in human gliomas and prostate tumors. The data implicate acetyl-CoA metabolism as a key determinant of histone acetylation levels in cancer cells.


Asunto(s)
Histonas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Acetilcoenzima A/metabolismo , Acetilación/efectos de los fármacos , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Análisis por Conglomerados , Coenzima A/metabolismo , Glioma/metabolismo , Glioma/patología , Glucosa/farmacología , Humanos , Interleucina-3/farmacología , Masculino , Fosforilación/efectos de los fármacos , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas ras/genética , Proteínas ras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA