Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oncogene ; 38(20): 3765-3780, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30262865

RESUMEN

To improve cancer patient outcome significantly, we must understand the mechanisms regulating stem-like cancer cells, which have been implicated as a cause of metastasis and treatment resistance. The transcription factor C/EBPδ can exhibit pro- and anti-tumorigenic activities, but the mechanisms underlying the complexity of its functions are poorly understood. Here we identify a role for breast cancer cell intrinsic C/EBPδ in promoting phenotypes that have been associated with cancer stem cells (CSCs). While C/EBPδ expression is not abundant in most metastatic breast cancers, our data support a pro-tumorigenic role of C/EBPδ when expressed in subsets of tumor cells and/or through transient activation by the tumor microenvironment or loss of substrate adhesion. Using genetic mouse models and human breast cancer cell lines, we show that deletion or depletion of C/EBPδ reduced expression of stem cell factors and stemnness markers, sphere formation and self-renewal, along with growth of tumors and established experimental metastases in vivo. C/EBPδ is also known as a mediator of the innate immune response, which is enhanced by hypoxia and interleukin-6 (IL-6) signaling, two conditions that also play important roles in cancer progression. Our mechanistic data reveal C/EBPδ as a link that engages two positive feedback loops, in part by directly targeting the IL-6 receptor (IL6RA) gene, and, thus, amplifying IL-6 and HIF-1 signaling. This study provides a molecular mechanism for the synergism of tumor microenvironmental conditions in cancer progression with potential implications for the targeting of CSCs.


Asunto(s)
Neoplasias de la Mama/patología , Proteína delta de Unión al Potenciador CCAAT/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Interleucina-6/metabolismo , Células Madre Neoplásicas/patología , Animales , Neoplasias de la Mama/metabolismo , Proteína delta de Unión al Potenciador CCAAT/genética , Línea Celular Tumoral , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones Noqueados , Células Madre Neoplásicas/metabolismo , Receptores de Interleucina-6/genética , Transducción de Señal/genética , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Cell ; 175(7): 1972-1988.e16, 2018 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-30550791

RESUMEN

In vitro cancer cultures, including three-dimensional organoids, typically contain exclusively neoplastic epithelium but require artificial reconstitution to recapitulate the tumor microenvironment (TME). The co-culture of primary tumor epithelia with endogenous, syngeneic tumor-infiltrating lymphocytes (TILs) as a cohesive unit has been particularly elusive. Here, an air-liquid interface (ALI) method propagated patient-derived organoids (PDOs) from >100 human biopsies or mouse tumors in syngeneic immunocompetent hosts as tumor epithelia with native embedded immune cells (T, B, NK, macrophages). Robust droplet-based, single-cell simultaneous determination of gene expression and immune repertoire indicated that PDO TILs accurately preserved the original tumor T cell receptor (TCR) spectrum. Crucially, human and murine PDOs successfully modeled immune checkpoint blockade (ICB) with anti-PD-1- and/or anti-PD-L1 expanding and activating tumor antigen-specific TILs and eliciting tumor cytotoxicity. Organoid-based propagation of primary tumor epithelium en bloc with endogenous immune stroma should enable immuno-oncology investigations within the TME and facilitate personalized immunotherapy testing.


Asunto(s)
Modelos Inmunológicos , Neoplasias Experimentales/inmunología , Organoides/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Microambiente Tumoral/inmunología , Animales , Antígeno B7-H1/inmunología , Técnicas de Cocultivo , Femenino , Humanos , Inmunoterapia , Masculino , Ratones , Ratones Endogámicos BALB C , Proteínas de Neoplasias/inmunología , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Organoides/patología
3.
Int J Oncol ; 53(2): 579-591, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29901186

RESUMEN

Although high-risk human papillomavirus (HR­HPV) infection has a prominent role in the aetiology of cervical cancer (CC), sex steroid hormones may also be involved in this process; however, the cooperation between oestrogen and HR­HPV in the early stages of cervical carcinogenesis is poorly understood. Since 17ß-oestradiol (E2) and the HPV type 16­E7 oncoprotein induce CC in transgenic mice, a microarray analysis was performed in the present study to generate global gene expression profiles from 2­month­old FVB (non­transgenic) and K14E7 (transgenic) mice who were left untreated or were treated for 1 month with E2. Upregulation of cancer-related genes that have not been previously reported in the context of CC, including glycerophosphodiester phosphodiesterase domain containing 3, interleukin 1 receptor type II, natriuretic peptide type C, MGAT4 family member C, lecithin-retinol acyltransferase (phosphatidylcholine-retinol-O-acyltransferase) and glucoside xylosyltransferase 2, was observed. Notably, upregulation of the serine (or cysteine) peptidase inhibitor clade B member 9 gene and downregulation of the Granzyme gene family were observed; the repression of the Granzyme B pathway may be a novel mechanism of immune evasion by cancer cells. The present results provide the basis for further studies on early biomarkers of CC risk and synergistic interactions between HR­HPV and oestrogen.


Asunto(s)
Estradiol/efectos adversos , Perfilación de la Expresión Génica/métodos , Granzimas/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Proteínas E7 de Papillomavirus/genética , Neoplasias del Cuello Uterino/genética , Animales , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Péptido Natriurético Tipo-C/genética , Neoplasias Experimentales , Proteínas E7 de Papillomavirus/metabolismo , Hidrolasas Diéster Fosfóricas/genética , Receptores Tipo II de Interleucina-1/genética , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/virología
4.
Biomed Res Int ; 2017: 7868645, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29130045

RESUMEN

OBJECTIVE: The aim of this work was to compare the early gene expression profiles in the skin of HPV16-E6 transgenic mice regulated by the E6 PDZ-binding motif. MATERIALS AND METHODS: The global transcriptional profiles in dorsal skin biopsies from K14E6 and K14E6Δ146-151 transgenic mice were compared using microarrays. Relevant genes obtained from the most differentially expressed processes were further examined by RT-qPCR, in situ RT-PCR, Western blot, or immunofluorescence. RESULTS: The transcriptomic landscape of K14E6 versus K14E6Δ146-151 shows that the most affected expression profiles were those related to keratinocyte differentiation, stem cell maintenance, and keratinization. Additionally, downregulation of epidermal stemness markers such as K15 and CD34, as well as the upregulation of cytokeratin 6b, appeared to be dependent on the E6 PDZ-binding motif. Finally, wound healing, a physiological process linked to stemness, is impaired in the K14E6 mice compared to K14E6Δ146-151. CONCLUSION: The E6 PDZ-binding motif appears to affect stemness and keratinization during early stages of skin carcinogenesis. As E6 plays a significant role in HPV-induced skin carcinogenesis, the K14E6 versus K14E6Δ146-151 transcriptional profile provides a source of valuable data to uncover novel E6 functions in the skin.


Asunto(s)
Queratinas/metabolismo , Proteínas Oncogénicas Virales/química , Proteínas Oncogénicas Virales/metabolismo , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Células Madre/metabolismo , Transcripción Genética , Secuencias de Aminoácidos , Animales , Antígenos CD34/metabolismo , Biomarcadores/metabolismo , Cadherinas/metabolismo , Diferenciación Celular , Queratinocitos/citología , Queratinas/genética , Ratones Transgénicos , Dominios PDZ , Transporte de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Piel/metabolismo , Relación Estructura-Actividad , Transcriptoma , Cicatrización de Heridas , beta Catenina/metabolismo
5.
Adv Exp Med Biol ; 962: 353-368, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28299668

RESUMEN

A full understanding of RUNX gene function in different epithelial lineages has been thwarted by the lethal phenotypes observed when constitutively knocking out these mammalian genes. However temporal expression of the Runx genes throughout the different phases of mammary gland development is indicative of a functional role in this tissue. A few studies have emerged describing how these genes impact on the fate of mammary epithelial cells by regulating lineage differentiation and stem/progenitor cell potential, with implications for the transformed state. The importance of the RUNX/CBFß core factor binding complex in breast cancer has very recently been highlighted with both RUNX1 and CBFß appearing in a comprehensive gene list of predicted breast cancer driver mutations. Nonetheless, the evidence to date shows that the RUNX genes can have dualistic outputs with respect to promoting or constraining breast cancer phenotypes, and that this may be aligned to individual subtypes of the clinical disease. We take this opportunity to review the current literature on RUNX and CBFß in the normal and neoplastic mammary lineage while appreciating that this is likely to be the tip of the iceberg in our knowledge.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Linaje de la Célula/genética , Subunidades alfa del Factor de Unión al Sitio Principal/genética , Mamíferos/genética , Animales , Diferenciación Celular/genética , Femenino , Humanos , Mutación/genética
6.
Virology ; 447(1-2): 155-65, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24210110

RESUMEN

The HPV16 E7 oncoprotein and 17ß-estradiol are important factors for the induction of premalignant lesions and cervical cancer. The study of these factors is crucial for a better understanding of cervical tumorigenesis. Here, we assessed the global gene expression profiles induced by the HPV16 E7 oncoprotein and/or 17ß-estradiol in cervical tissue of FvB and K14E7 transgenic mice. We found that the most dramatic changes in gene expression occurred in K14E7 and FvB groups treated with 17ß-estradiol. A large number of differentially expressed genes involved in the immune response were observed in 17ß-estradiol treated groups. The E7 oncoprotein mainly affected the expression of genes involved in cellular metabolism. Our microarray data also identified differentially expressed genes that have not previously been reported in cervical cancer. The identification of genes regulated by E7 and 17ß-estradiol, provides the basis for further studies on their role in cervical carcinogenesis.


Asunto(s)
Estradiol/farmacología , Estrógenos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Interacciones Huésped-Patógeno , Proteínas E7 de Papillomavirus/metabolismo , Animales , Femenino , Perfilación de la Expresión Génica , Ratones , Ratones Transgénicos
7.
Proc Natl Acad Sci U S A ; 109(5): 1524-9, 2012 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-22307607

RESUMEN

Filamin A (FLNA) is an actin-binding protein with a well-established role in the cytoskeleton, where it determines cell shape and locomotion by cross-linking actin filaments. Mutations in FLNA are associated with a wide range of genetic disorders. Here we demonstrate a unique role for FLNA as a nucleolar protein that associates with the RNA polymerase I (Pol I) transcription machinery to suppress rRNA gene transcription. We show that depletion of FLNA by siRNAs increased rRNA expression, rDNA promoter activity and cell proliferation. Immunodepletion of FLNA from nuclear extracts resulted in a decrease in rDNA promoter-driven transcription in vitro. FLNA coimmunoprecipitated with the Pol I components actin, TIF-IA, and RPA40, and their occupancy of the rDNA promoter was increased in the absence of FLNA in vivo. The FLNA actin-binding domain is essential for the suppression of rRNA expression and for inhibiting recruitment of the Pol I machinery to the rDNA promoter. These findings reveal an additional role for FLNA as a regulator of rRNA gene expression and have important implications for our understanding of the role of FLNA in human disease.


Asunto(s)
Proteínas Contráctiles/fisiología , Proteínas del Citoesqueleto/fisiología , Proteínas de Microfilamentos/fisiología , ARN Ribosómico/genética , Transcripción Genética/fisiología , Actinas , Sitios de Unión , Línea Celular , Proteínas Contráctiles/metabolismo , Proteínas del Citoesqueleto/metabolismo , ADN Ribosómico/genética , Filaminas , Humanos , Proteínas de Microfilamentos/metabolismo , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Interferente Pequeño
8.
Breast Cancer Res ; 13(5): R106, 2011 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-22032690

RESUMEN

INTRODUCTION: Breast cancers frequently metastasise to the skeleton where they cause osteolytic bone destruction by stimulating osteoclasts to resorb bone and by preventing osteoblasts from producing new bone. The Runt-related transcription factor 2, Runx2, is an important determinant of bone metastasis in breast cancer. Runx2 is known to mediate activation of osteoclast activity and inhibition of osteoblast differentiation by metastatic breast cancer cells. However, while Runx2-regulated genes that mediate osteoclast activation have been identified, how Runx2 determines inhibition of osteoblasts is unknown. METHODS: The aim of this study was to determine how Runx2 mediates the ability of metastatic breast cancer cells to modulate the activity of bone cells. We have previously demonstrated that Runx2 requires the co-activator core binding factor beta (CBFß) to regulate gene expression in breast cancer cells. We, therefore, performed independent microarray analyses to identify target genes whose expression is dependent upon both Runx2 and CBFß. Common target genes, with a role in modulating bone-cell function, were confirmed using a combination of siRNA, quantitative reverse transcriptase PCR (qRT-PCR), ELISA, promoter reporter analysis, Electrophoretic Mobility Shift Assay (EMSA) and chromatin immunoprecipitation (ChIP) assays. The function of Runx2/CBFß-regulated genes in mediating the ability of MDA-MB-231 to inhibit osteoblast differentiation was subsequently established in primary bone marrow stromal cell cultures and MC-3T3 osteoblast cells. RESULTS: We show that Runx2/CBFß mediates inhibition of osteoblast differentiation by MDA-MB-231 cells through induction of the Wnt signaling antagonist, sclerostin. We demonstrate that MDA-MB-231 cells secrete sclerostin and that sclerostin-expression is critically dependent on both Runx2 and CBFß. We also identified the osteoclast activators IL-11 and granulocyte-macrophage colony-stimulating factor (GM-CSF) as new target genes of Runx2/CBFß in metastatic breast cancer cells. CONCLUSIONS: This study demonstrates that Runx2 and CBFß are required for the expression of genes that mediate the ability of metastatic breast cancer cells to directly modulate both osteoclast and osteoblast function. We also show that Runx2-dependent inhibition of osteoblast differentiation by breast cancer cells is mediated through the Wnt antagonist, sclerostin.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Neoplasias de la Mama/patología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , Marcadores Genéticos/genética , Osteoblastos/metabolismo , Osteoblastos/patología , Proteínas Wnt/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Neoplasias de la Mama/metabolismo , Diferenciación Celular/genética , Línea Celular Tumoral , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad beta del Factor de Unión al Sitio Principal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Interleucina-11/genética , Interleucina-11/metabolismo , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Células del Estroma/citología , Células del Estroma/metabolismo
9.
Mol Cancer ; 9: 171, 2010 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-20591170

RESUMEN

BACKGROUND: The transcription factor Runx2 has an established role in cancers that metastasize to bone. In metastatic breast cancer cells Runx2 is overexpressed and contributes to the invasive capacity of the cells by regulating the expression of several invasion genes. CBFbeta is a transcriptional co-activator that is recruited to promoters by Runx transcription factors and there is considerable evidence that CBFbeta is essential for the function of Runx factors. However, overexpression of Runx1 can partially rescue the lethal phenotype in CBFbeta-deficient mice, indicating that increased levels of Runx factors can, in some situations, overcome the requirement for CBFbeta. Since Runx2 is overexpressed in metastatic breast cancer cells, and there are no reports of CBFbeta expression in breast cells, we sought to determine whether Runx2 function in these cells was dependent on CBFbeta. Such an interaction might represent a viable target for therapeutic intervention to inhibit bone metastasis. RESULTS: We show that CBFbeta is expressed in the metastatic breast cancer cells, MDA-MB-231, and that it associates with Runx2. Matrigel invasion assays and RNA interference were used to demonstrate that CBFbeta contributes to the invasive capacity of these cells. Subsequent analysis of Runx2 target genes in MDA-MB-231 cells revealed that CBFbeta is essential for the expression of Osteopontin, Matrixmetalloproteinase-13, Matrixmetalloproteinase-9, and Osteocalcin but not for Galectin-3. Chromatin immunoprecipitation analysis showed that CBFbeta is recruited to both the Osteopontin and the Galectin-3 promoters. CONCLUSIONS: CBFbeta is expressed in metastatic breast cancer cells and is essential for cell invasion. CBFbeta is required for expression of several Runx2-target genes known to be involved in cell invasion. However, whilst CBFbeta is essential for invasion, not all Runx2-target genes require CBFbeta. We conclude that CBFbeta is required for a subset of Runx2-target genes that are sufficient to maintain the invasive phenotype of the cells. These findings suggest that the interaction between Runx2 and CBFbeta might represent a viable target for therapeutic intervention to inhibit bone metastasis.


Asunto(s)
Neoplasias de la Mama/patología , Subunidad beta del Factor de Unión al Sitio Principal/fisiología , Invasividad Neoplásica , Animales , Línea Celular Tumoral , Subunidad beta del Factor de Unión al Sitio Principal/genética , Femenino , Humanos , Ratones , Metástasis de la Neoplasia , Interferencia de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...