Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Genes Dev ; 27(3): 288-300, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23355395

RESUMEN

Pancreatic exocrine cell plasticity can be observed during development, pancreatitis with subsequent regeneration, and also transformation. For example, acinar-ductal metaplasia (ADM) occurs during acute pancreatitis and might be viewed as a prelude to pancreatic intraepithelial neoplasia (PanIN) and pancreatic ductal adenocarcinoma (PDAC) development. To elucidate regulatory processes that overlap ductal development, ADM, and the progression of normal cells to PanIN lesions, we undertook a systematic approach to identify the Prrx1 paired homeodomain Prrx1 transcriptional factor as a highly regulated gene in these processes. Prrx1 annotates a subset of pancreatic ductal epithelial cells in Prrx1creER(T2)-IRES-GFP mice. Furthermore, sorted Prrx1(+) cells have the capacity to self-renew and expand during chronic pancreatitis. The two isoforms, Prrx1a and Prrx1b, regulate migration and invasion, respectively, in pancreatic cancer cells. In addition, Prrx1b is enriched in circulating pancreatic cells (Pdx1cre;LSL-Kras(G12D/+);p53(fl/+);R26YFP). Intriguingly, the Prrx1b isoform, which is also induced in ADM, binds the Sox9 promoter and positively regulates Sox9 expression. This suggests a new hierarchical scheme whereby a Prrx1-Sox9 axis may influence the emergence of acinar-ductal metaplasia and regeneration. Furthermore, our data provide a possible explanation of why pancreatic cancer is skewed toward a ductal fate.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Páncreas/patología , Páncreas/fisiología , Neoplasias Pancreáticas/fisiopatología , Regeneración/fisiología , Animales , Línea Celular Tumoral , Células Cultivadas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos C57BL , Páncreas/citología , Regiones Promotoras Genéticas/genética , Unión Proteica , Isoformas de Proteínas/metabolismo , Factor de Transcripción SOX9/genética
2.
Mol Carcinog ; 56(3): 936-944, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27583552

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) remains one of the most deadly types of cancer, and the majority of pancreatic cancer deaths is caused by metastasis. Therapeutic options for systemic disease are limited, in particular due to the heterogeneous events leading to tumor dissemination. Previous studies highlighted an association of the homeodomain transcription factor MEIS1 with a ductal phenotype in pancreatic tissue architecture. Using immunohistochemistry, we demonstrate that MEIS1 is expressed in aberrant duct structures of Ela-TGFα transgenic mice as well as in pancreatic intraepithelial neoplasia (PanINs), primary PDAC, and metastatic disease in Ptf1aCre/+ ;LSL-KrasG12D/+ mice. To assess a putative role of MEIS1 in the progression of PDAC, MEIS1 overexpressing pancreatic cancer cell lines were generated by retroviral gene delivery and assessed for proliferation and migration. MEIS1 did not affect proliferation but increased migration in a subset of cell lines tested. Subsequent genome wide expression analysis identified upregulation of the pro-metastatic gene melanoma cell adhesion molecule (Mcam) in migrating cells. Employing DNA-pulldown and chromatin immunoprecipitation (ChIP) assays we reveal interaction of MEIS1 with the enhancer-DNA of Mcam and its transcriptional activation to facilitate migration of pancreatic cancer cells in vitro. Activation of Mcam through MEIS1 occurs in a cell type dependent fashion, reflecting the different routes that lead to metastasis in vivo. To our knowledge, these results show for the first time a pro-metastatic function of Mcam in pancreatic cancer. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/metabolismo , Animales , Antígeno CD146/genética , Carcinoma Ductal Pancreático/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Ratones , Ratones Transgénicos , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Neoplasias Pancreáticas/genética
3.
Proc Natl Acad Sci U S A ; 108(24): 9945-50, 2011 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-21628592

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a fatal disease with poor patient outcome often resulting from late diagnosis in advanced stages. To date methods to diagnose early-stage PDAC are limited and in vivo detection of pancreatic intraepithelial neoplasia (PanIN), a preinvasive precursor of PDAC, is impossible. Using a cathepsin-activatable near-infrared probe in combination with flexible confocal fluorescence lasermicroscopy (CFL) in a genetically defined mouse model of PDAC we were able to detect and grade murine PanIN lesions in real time in vivo. Our diagnostic approach is highly sensitive and specific and proved superior to clinically established fluorescein-enhanced imaging. Translation of this endoscopic technique into the clinic should tremendously improve detection of pancreatic neoplasia, thus reforming management of patients at risk for PDAC.


Asunto(s)
Carcinoma Ductal Pancreático/diagnóstico , Imagen Molecular/métodos , Neoplasias Pancreáticas/diagnóstico , Lesiones Precancerosas/diagnóstico , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Catepsinas/genética , Catepsinas/metabolismo , Femenino , Colorantes Fluorescentes/metabolismo , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Ratones , Microscopía Confocal , Microscopía Fluorescente , Estadificación de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
4.
Gastroenterology ; 137(1): 361-71, 371.e1-5, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19362090

RESUMEN

BACKGROUND & AIMS: Early metastasis is a hallmark of pancreatic ductal adenocarcinoma and responsible for >90% of pancreatic cancer death. Because little is known about the biology and genetics of the metastatic process, we desired to elucidate molecular pathways mediating pancreatic cancer metastasis in vivo by an unbiased forward genetic approach. METHODS: Highly metastatic pancreatic cancer cell populations were selected by serial in vivo passaging of parental cells with low metastatic potential and characterized by global gene expression profiling, chromatin immunoprecipitation, and in vivo metastatic assay. RESULTS: In vivo selection of highly metastatic pancreatic cancer cells induced epithelial-mesenchymal transition (EMT), loss of E-cadherin expression, and up-regulation of mesenchymal genes such as Snail. Genetic inactivation of E-cadherin in parental cells induced EMT and increased metastasis in vivo. Silencing of E-cadherin in highly metastatic cells is mediated by a transcriptional repressor complex containing Snail and histone deacetylase 1 (HDAC1) and HDAC2. In line, mesenchymal pancreatic cancer specimens and primary cell lines from genetically engineered Kras(G12D) mice showed HDAC-dependent down-regulation of E-cadherin and high metastatic potential. Finally, transforming growth factor beta-driven E-cadherin silencing and EMT of human pancreatic cancer cells depends on HDAC activity. CONCLUSIONS: We provide the first in vivo evidence that HDACs and Snail play an essential role in silencing E-cadherin during the metastatic process of pancreatic cancer cells. These data link the epigenetic HDAC machinery to EMT and metastasis and provide preclinical evidence that HDACs are promising targets for antimetastatic therapy.


Asunto(s)
Cadherinas/metabolismo , Histona Desacetilasas/metabolismo , Neoplasias Pulmonares/enzimología , Neoplasias Pancreáticas/enzimología , Factores de Transcripción/metabolismo , Animales , Antígenos CD , Antineoplásicos/farmacología , Cadherinas/genética , Línea Celular Tumoral , Transdiferenciación Celular , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Histona Desacetilasa 1 , Histona Desacetilasa 2 , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/genética , Humanos , Ácidos Hidroxámicos/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundario , Ratones , Ratones Desnudos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Regiones Promotoras Genéticas , Interferencia de ARN , Proteínas Represoras/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Transfección
5.
Int J Cancer ; 123(9): 2138-47, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18709639

RESUMEN

Pancreatic cancer is a serious disease with poor patient outcome, often as a consequence of late diagnosis in advanced stages. This is in large part due to the lack of diagnostic tools for early detection. To address this deficiency, we have investigated novel molecular near-infrared fluorescent (NIRF) in vivo imaging techniques in clinically relevant mouse models of pancreatic cancer. Genome wide gene expression profiling was used to identify cathepsin cystein proteases and matrix metalloproteinases (MMP) as targets for NIRF imaging. Appropriate protease activatable probes were evaluated for detection of early-stage pancreatic cancer in mice with orthotopically implanted pancreatic cancer cell lines. Mice with pancreatitis served as controls. Whole body in vivo NIRF imaging using activatable cathepsin sensitive probes specifically detected pancreatic tumors as small as 1-2 mm diameter. Imaging of MMP activity demonstrated high specificity for MMP positive tumors. Intravital flexible confocal fluorescence lasermicroscopy of protease activity enabled specific detection of pancreatic tumors at the cellular level. Importantly, topical application of NIRF-probes markedly reduced background without altering signal intensity. Taken together, macroscopic and confocal lasermicroscopic molecular in vivo imaging of protease activity is highly sensitive, specific and allows discrimination between normal pancreatic tissue, inflammation and pancreatic cancer. Translation of this approach to the clinic could significantly improve endoscopic and laparoscopic detection of early-stage pancreatic cancer.


Asunto(s)
Metaloproteinasas de la Matriz/análisis , Neoplasias Pancreáticas/diagnóstico , Espectroscopía Infrarroja Corta/métodos , Animales , Catepsinas/análisis , Catepsinas/genética , Línea Celular Tumoral , Diagnóstico Precoz , Fluorescencia , Perfilación de la Expresión Génica , Humanos , Metaloproteinasas de la Matriz/genética , Ratones , Estadificación de Neoplasias , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/patología , Sensibilidad y Especificidad
6.
Mol Cancer Res ; 13(5): 863-9, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25724428

RESUMEN

UNLABELLED: Pancreatic cancer is one of the deadliest cancers with poor survival rates and limited therapeutic options. To improve the understanding of this disease's biology, a prerequisite for the generation of novel therapeutics, new platforms for rapid and efficient genetic and therapeutic screening are needed. Therefore, a combined in vitro/in vivo hybrid shRNA assay was developed using isolated murine primary pancreatic ductal cells (PDCs), in which oncogenic Kras(G12D) could be activated in vitro by genomic recombination through 4OH-tamoxifen-induced nuclear translocation of Cre-ERT2 expressed under control of the ROSA26 promoter. Further genetic manipulation was achieved through selective and stable RNAi against the tumor suppressors p16(Ink4a) (CDKN2A) or Trp53 (TP53) using lentiviral gene delivery. Treatment of PDCs with 4OH-tamoxifen increased phosphorylation of ERK downstream of KRAS, and subsequent lentiviral transduction resulted in sustained target gene repression. Double-mutant PDCs were then reintroduced into the pancreata of NOD-SCID-gamma (NSG) mice and monitored for tumor growth. Orthotopic implantation of PDCs carrying the activated Kras(G12D)-allele and shRNA against p16(Ink4a) or Trp53 resulted in tumor growth, metastasis, and reduced survival of NSG mice. In contrast, Kras(G12D) alone was not sufficient to induce tumor growth. IMPLICATIONS: The combinatory in vitro/in vivo approach described in this study allows for rapid and efficient identification of genes involved in carcinogenesis and opens new avenues for the development of therapeutic strategies to improve cancer treatment.


Asunto(s)
Adenocarcinoma/genética , Carcinoma Ductal Pancreático/genética , Genes Supresores de Tumor , ARN Interferente Pequeño/genética , Adenocarcinoma/patología , Animales , Carcinoma Ductal Pancreático/patología , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , ARN Interferente Pequeño/metabolismo , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología
7.
Cell Stem Cell ; 16(6): 627-38, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-26046762

RESUMEN

Epithelium of the colon and intestine are renewed every 3 days. In the intestine there are at least two principal stem cell pools. The first contains rapid cycling crypt-based columnar (CBC) Lgr5(+) cells, and the second is composed of slower cycling Bmi1-expressing cells at the +4 position above the crypt base. In the colon, however, the identification of Lgr5(-) stem cell pools has proven more challenging. Here, we demonstrate that the intermediate filament keratin-19 (Krt19) marks long-lived, radiation-resistant cells above the crypt base that generate Lgr5(+) CBCs in the colon and intestine. In colorectal cancer models, Krt19(+) cancer-initiating cells are also radioresistant, while Lgr5(+) stem cells are radiosensitive. Moreover, Lgr5(+) stem cells are dispensable in both the normal and neoplastic colonic epithelium, as ablation of Lgr5(+) stem cells results in their regeneration from Krt19-expressing cells. Thus, Krt19(+) stem cells are a discrete target relevant for cancer therapy.


Asunto(s)
Colon/patología , Intestinos/patología , Queratina-19/metabolismo , Células Madre Neoplásicas/metabolismo , Tolerancia a Radiación , Receptores Acoplados a Proteínas G/metabolismo , Animales , Queratina-19/genética , Ratones Transgénicos , Células Madre Neoplásicas/patología , Transporte de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo
8.
PLoS One ; 5(8): e12311, 2010 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-20808819

RESUMEN

BACKGROUND: Pancreas organogenesis is the result of well-orchestrated and balanced activities of transcription factors. The homeobox transcription factor PDX-1 plays a crucial role in the development and function of the pancreas, both in the maintenance of progenitor cells and in determination and maintenance of differentiated endocrine cells. However, the activity of homeobox transcription factors requires coordination with co-factors, such as PBX and MEIS proteins. PBX and MEIS proteins belong to the family of three amino acid loop extension (TALE) homeodomain proteins. In a previous study we found that PDX-1 negatively regulates the transcriptional activity of the ductal specific keratin 19 (Krt19). In this study, we investigate the role of different domains of PDX-1 and elucidate the functional interplay of PDX-1 and MEIS1 necessary for Krt19 regulation. METHODOLOGY/PRINCIPAL FINDINGS: Here, we demonstrate that PDX-1 exerts a dual manner of regulation of Krt19 transcriptional activity. Deletion studies highlight that the NH(2)-terminus of PDX-1 is functionally relevant for the down-regulation of Krt19, as it is required for DNA binding of PDX-1 to the Krt19 promoter. Moreover, this effect occurs independently of PBX. Second, we provide insight on how PDX-1 regulates the Hox co-factor MEIS1 post-transcriptionally. We find specific binding of MEIS1 and MEIS2 to the Krt19 promoter using IP-EMSA, and siRNA mediated silencing of Meis1, but not Meis2, reduces transcriptional activation of Krt19 in primary pancreatic ductal cells. Over-expression of PDX-1 leads to a decreased level of MEIS1 protein, and this decrease is prevented by inhibition of the proteasome. CONCLUSIONS/SIGNIFICANCE: Taken together, our data provide evidence for a dual mechanism of how PDX-1 negatively regulates Krt19 ductal specific gene expression. These findings imply that transcription factors may efficiently regulate target gene expression through diverse, non-redundant mechanisms.


Asunto(s)
ADN/metabolismo , Proteínas de Homeodominio/metabolismo , Queratina-19/metabolismo , Proteínas de Neoplasias/metabolismo , Conductos Pancreáticos/metabolismo , Transactivadores/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Regulación hacia Abajo , Genes Reporteros/genética , Proteínas de Homeodominio/química , Queratina-19/genética , Ratones , Ratones Endogámicos C57BL , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Especificidad de Órganos , Regiones Promotoras Genéticas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Especificidad por Sustrato , Transactivadores/química , Transcripción Genética , Activación Transcripcional
9.
Mol Biol Cell ; 20(22): 4838-44, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19793922

RESUMEN

Embryonic development of the pancreas is marked by an early phase of dramatic morphogenesis, in which pluripotent progenitor cells of the developing pancreatic epithelium give rise to the full array of mature exocrine and endocrine cell types. The genetic determinants of acinar and islet cell lineages are somewhat well defined; however, the molecular mechanisms directing ductal formation and differentiation remain to be elucidated. The complex ductal architecture of the pancreas is established by a reiterative program of progenitor cell expansion and migration known as branching morphogenesis, or tubulogenesis, which proceeds in mouse development concomitantly with peak Pdx1 transcription factor expression. We therefore evaluated Pdx1 expression with respect to lineage-specific markers in embryonic sections of the pancreas spanning this critical period of duct formation and discovered an unexpected population of nonislet Pdx1-positive cells displaying physical traits of branching. We then established a 3D cell culture model of branching morphogenesis using primary pancreatic duct cells and identified a transient surge of Pdx1 expression exclusive to branching cells. From these observations we propose that Pdx1 might be involved temporally in a program of gene expression sufficient to facilitate the biochemical and morphological changes necessary for branching morphogenesis.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Morfogénesis/fisiología , Conductos Pancreáticos/embriología , Transactivadores/metabolismo , Animales , Biomarcadores/metabolismo , Linaje de la Célula , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Ratones , Ratones Noqueados , Conductos Pancreáticos/citología , Conductos Pancreáticos/fisiología , Células Madre/fisiología , Transactivadores/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA