Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
BMC Cancer ; 20(1): 55, 2020 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31973710

RESUMEN

Following publication of the original article [1], the authors reported an error in Fig. 6c and in the figure legends for Fig. 5c and Fig. 6c.

2.
BMC Cancer ; 16: 142, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26908052

RESUMEN

BACKGROUND: Epithelial-mesenchymal transition (EMT) increases cell migration and is implicated in cancer cell invasion and metastasis. We have previously described the involvement of the transcription factors, nuclear factor I-C2 (NFI-C2) and Forkhead box F1 (FoxF1), in the regulation of EMT and invasion during breast tumor progression. NFI-C2 counteracts these processes and FoxF1 is a directly repressed target of NFI-C2. FoxF1 induces EMT and invasiveness and enhances xenograft tumorigenicity in nude mice. Here we identify oppositely regulated targets of NFI-C2 and FoxF1 involved in these processes and further study a possible role for FoxF1 in tumorigenesis. METHODS: We used Affymetrix microarray to detect changes in the transcriptome of a mouse mammary epithelial cell line upon overexpression of NFI-C2 or FoxF1. To elucidate the effects and signaling events following FoxF1 overexpression we investigated in vitro invasion capacity and changes in transcription and protein expression resulting from RNAi and inhibitor treatment. RESULTS: The extracellular matrix enzyme lysyl oxidase (LOX) was negatively regulated by NFI-C2 and positively regulated by FoxF1, and upregulation of LOX following FoxF1 overexpression in mouse mammary epithelial cells increased in vitro cell invasion. In the nuclei of FoxF1-overexpressing cells, the phosphorylation of Smad2 decreased, while that of p38 increased. Depletion of LOX by RNAi enhanced phosphorylation of Smad2 by a focal adhesion kinase (FAK)-dependent mechanism. In addition, induced expression of FoxF1 in a non-malignant human mammary epithelial cell line showed that the increase in LOX transcription and the suppression of Smad2 activity are early effects of FoxF1. CONCLUSION: These data show that FoxF1 enhances invasion in a LOX-dependent manner, is involved in the regulation of Smad2 signaling, and that FoxF1 overexpression ultimately leads to activation of p38 MAPK signaling. These findings provide new insights into the regulation of signaling pathways known to be important during breast tumor progression.


Asunto(s)
Neoplasias de la Mama/patología , Factores de Transcripción Forkhead/genética , Factores de Transcripción NFI/genética , Proteína-Lisina 6-Oxidasa/genética , Proteínas Smad/genética , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Movimiento Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Factores de Transcripción Forkhead/metabolismo , Humanos , Ratones , Factores de Transcripción NFI/metabolismo , Invasividad Neoplásica , Proteína-Lisina 6-Oxidasa/metabolismo , Transducción de Señal
3.
Genes Chromosomes Cancer ; 51(1): 77-82, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22034095

RESUMEN

DNA copy number aberrations (CNA) and subsequent altered gene expression profiles (mRNA levels) are characteristic features of cancerous cells. Integrative genomic analysis aims to identify recurrent CNA that may have a potential role in cancer development, assuming that gene amplification is accompanied by overexpression, while deletions give rise to downregulation of gene expression. We propose a segmented regression-based approach to identify CNA-driven alteration of gene expression profiles. Segmented regression allows to fit piecewise linear models in different domains of CNA joined by a change-point, where the mRNA-CNA relationship undergoes structural changes. Here, we illustrate the implementation and applicability of the proposed model using 1,161 chromosome fragments detected as DNA CNA in primary tumors from 97 breast cancer patients. We identified significant CNA-driven changes in gene expression levels for 341 chromosome fragments, of which 72 showed a nonlinear relationship to CNA. For 59 of 72 chromosome fragments (82%), we observed an initial increase in mRNA levels due to changes in CNA. After the change-point was passed, the mRNA levels reached a plateau, and a further increase in DNA copy numbers did not induce further elevation in mRNA levels. In contrast, for 13 chromosome fragments, the change-point marked the point where mRNA production accelerated. We conclude that segmented regression modeling may provide valuable insights into the impact CNA have on gene expression in cancer cells.


Asunto(s)
Neoplasias de la Mama/genética , Dosificación de Gen , Perfilación de la Expresión Génica , ARN Mensajero/metabolismo , Hibridación Genómica Comparativa , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Modelos Genéticos , Análisis de Regresión
4.
Dev Cell ; 12(1): 99-112, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17199044

RESUMEN

We show that removing the Shh signal tranducer Smoothened from skin epithelium secondarily results in excess Shh levels in the mesenchyme. Moreover, the phenotypes we observe reflect decreased epithelial Shh signaling, yet increased mesenchymal Shh signaling. For example, the latter contributes to exuberant hair follicle (HF) induction, while the former depletes the resulting follicular stem cell niches. This disruption of the niche apparently also allows the remaining stem cells to initiate hair formation at inappropriate times. Thus, the temporal structure of the hair cycle may depend on the physical structure of the niche. Finally, we find that the ablation of epithelial Shh signaling results in unexpected transformations: the follicular outer root sheath takes on an epidermal character, and certain HFs disappear altogether, having adopted a strikingly mammary gland-like fate. Overall, our study uncovers a multifaceted function for Shh in sculpting and maintaining the integrity and identity of the developing HF.


Asunto(s)
Folículo Piloso/anomalías , Folículo Piloso/embriología , Proteínas Hedgehog/metabolismo , Glándulas Mamarias Animales/patología , Transducción de Señal , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Línea Celular Transformada , Ectodermo/citología , Regulación del Desarrollo de la Expresión Génica , Folículo Piloso/patología , Proteínas Hedgehog/genética , Hiperplasia , Integrasas/metabolismo , Queratinocitos/citología , Glándulas Mamarias Animales/citología , Mesodermo/citología , Metaplasia , Ratones , Morfogénesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/metabolismo , Receptor Smoothened , Células Madre/citología , beta Catenina/metabolismo
5.
Mol Cell Biol ; 26(15): 5663-74, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16847321

RESUMEN

The classical mechanism by which prolactin transduces its signal in mammary epithelial cells is by activation of cytosolic signal transducer and activator of transcription 5 (Stat5) via a plasma membrane-associated prolactin receptor-Janus kinase 2 (Jak2) complex. Here we describe an alternative pathway through which prolactin via Jak2 localized in the nucleus activates the transcription factor nuclear factor 1-C2 (NF1-C2). Previous reports have demonstrated a nuclear localization of Jak2, but the physiologic importance of nuclear Jak2 has not been clear. We demonstrate that nuclear Jak2 regulates the amount of active NF1-C2 through tyrosine phosphorylation and proteasomal degradation. Our data also demonstrate a link between prolactin and p53 as well as the milk gene carboxyl ester lipase through nuclear Jak2 and NF1-C2. Hence, we describe a novel pathway through which nuclear Jak2 is subject to regulation by prolactin in mammary epithelial cells.


Asunto(s)
Núcleo Celular/metabolismo , Células Epiteliales/metabolismo , Glándulas Mamarias Humanas/anatomía & histología , Factores de Transcripción NFI/metabolismo , Prolactina/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal/fisiología , Animales , Línea Celular , Células Epiteliales/citología , Humanos , Janus Quinasa 2 , Lipasa/genética , Lipasa/metabolismo , Glándulas Mamarias Humanas/metabolismo , Ratones , Factores de Transcripción NFI/genética , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Técnicas del Sistema de Dos Híbridos , Tirosina/metabolismo
6.
Mol Endocrinol ; 19(4): 992-1003, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15637146

RESUMEN

We have previously demonstrated that the transcription factor nuclear factor (NF)1-C2 plays an important role in the mammary gland for the activation of the tumor suppressor gene p53. It also activates the milk genes carboxyl ester lipase and whey acidic protein, implying that NF1-C2 participates both in the establishment of a functional gland and in protection of the gland against tumorigenesis during proliferation. In this study, we have developed a new sensitive NF1-C2-specific antiserum for immunohistochemical analyses of the NF1-C2 distribution during mammary gland development. We show that the NF1-C2 protein is present in the epithelial compartment at the virgin stage and throughout mammary gland development. However, in the lactation stage the NF1-C2 protein levels strongly decreased, and many epithelial nuclei stained negative. In situ hybridization shows that NF1-C2 transcripts are expressed in the whole epithelium at pregnancy as well as the lactation stage, indicating that the reduction in protein levels is posttranscriptionally regulated. At involution, the NF1-C2 proteins are back to high levels. Based on studies using NMuMG cells and mammary tissue from heterozygous prolactin receptor knockout mice, we also demonstrate that prolactin has a direct effect in the maintenance of the NF1-C2 protein levels in the mammary epithelial nuclei at the virgin stage and during pregnancy. Hence, we have identified another transcription factor in the mammary gland, besides signal transducer and activator of transcription 5, through which prolactin may control mammary gland development. Furthermore, our data suggest a link between prolactin and p53 in the mammary gland, through NF1-C2.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Regulación del Desarrollo de la Expresión Génica , Glándulas Mamarias Animales/crecimiento & desarrollo , Prolactina/farmacología , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Potenciadoras de Unión a CCAAT/análisis , Proteínas Potenciadoras de Unión a CCAAT/genética , Núcleo Celular/química , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Humanos , Inmunoquímica , Lactancia/metabolismo , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Factores de Transcripción NFI , Embarazo , Prolactina/genética , Prolactina/metabolismo , Factores de Transcripción/análisis , Factores de Transcripción/genética , Transcripción Genética
7.
Oncogene ; 22(38): 6061-70, 2003 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-12955085

RESUMEN

The p53 tumor suppressor protein plays an important role in preventing cancer development by arresting or killing potential tumor cells. Downregulated p53 levels, or mutations within the p53 gene, leading to the loss of p53 activity, are found in many breast carcinomas. Here we demonstrate that the p53 gene is transcriptionally upregulated in the normal mouse mammary gland at midpregnancy. We show that the specific isoform nuclear factor 1-C2 (NF1-C2) plays an important role in this activation. Functional mutation of the NF1-binding site in the mouse p53 promoter resulted in a reduction of the gene expression to less than 30% in mammary epithelial cells. By the use of two powerful techniques, chromatin immunoprecipitation and oligonucleotide decoy, we verify the importance of NF1-C2 in p53 gene activation in vivo. These findings demonstrate a broader role for NF1-C2 in the mammary gland at midpregnancy, beyond its earlier reported activation of milk protein genes. We also demonstrate that NF1-A1 proteins are produced in the mouse mammary gland. However, due to their lower affinity to the NF1-binding site, these proteins are not involved in the transcriptional upregulation of p53 at midpregnancy. This paper constitutes the first report demonstrating the importance of NF1 proteins in the p53 gene activation in the mouse mammary gland. It is also the first time that p53 gene activation is coupled to a specific, endogenously expressed NF1 isoform.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas de Unión al ADN , Regulación de la Expresión Génica/fisiología , Glándulas Mamarias Animales/fisiología , Preñez/metabolismo , Proteína p53 Supresora de Tumor/genética , Animales , Sitios de Unión , Proteínas Potenciadoras de Unión a CCAAT/genética , Células Cultivadas , Células Epiteliales/metabolismo , Femenino , Técnicas Genéticas , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones , Ratones Endogámicos , Factores de Transcripción NFI , Proteínas Nucleares , Oligonucleótidos/genética , Embarazo , Regiones Promotoras Genéticas , Isoformas de Proteínas , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional , Proteína p53 Supresora de Tumor/metabolismo , Proteína 1 de Unión a la Caja Y
8.
Int J Oncol ; 45(1): 82-94, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24807161

RESUMEN

Recent research into the mechanisms of tumour cell invasiveness has highlighted the parallels between carcinogenesis and epithelial-mesenchymal transition (EMT), originally described as a developmental transdifferentiation program but also implicated in fibrosis and cancer. In a model system for mammary carcinogenesis, we previously observed that induced signalling from a homodimer of the c-erbB2 (HER2) receptor tyrosine kinase in an initially non-malignant mammary cell line caused EMT where i) cell scattering occurred before downregulation of the cell-cell adhesion molecule E-cadherin and ii) the progress of EMT was dramatically delayed when cells were grown at high density. Here, we have further analysed these phenomena. Ectopic expression of E-cadherin concomitant with c-erbB2 signalling was unable to impede the progression of EMT, suggesting that E-cadherin downregulation is not required for EMT. Furthermore, fibroblast-like cells isolated after EMT induced in the presence or absence of ectopic E-cadherin expression showed highly similar morphology and vimentin expression. E-cadherin expressed in these fibroblastic cells had a subcellular localisation similar to that found in epithelial cells, but it exhibited a much weaker attachment to the cytoskeleton, suggesting cytoskeletal rearrangements as an important mechanism in EMT-associated cell scattering. We also investigated whether density-dependent inhibition of EMT is mediated by E-cadherin as a sensor for cell-cell contact, by expressing dominant-negative E-cadherin. While expression of this mutant weakened cell-cell adhesion, it failed to facilitate EMT at high cell densities. These results indicate that loss of E-cadherin expression is a consequence rather than a cause of c-erbB2-induced EMT and that density­dependent inhibition of EMT is not mediated by E-cadherin signalling.


Asunto(s)
Cadherinas/metabolismo , Transición Epitelial-Mesenquimal , Fibroblastos/metabolismo , Glándulas Mamarias Humanas/metabolismo , Receptor ErbB-2/metabolismo , Adhesión Celular , Línea Celular , Citoesqueleto/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Fibroblastos/citología , Regulación de la Expresión Génica , Humanos , Glándulas Mamarias Humanas/citología , Transducción de Señal , Tetraciclina/farmacología , Vimentina/metabolismo
9.
PLoS One ; 9(5): e96007, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24789143

RESUMEN

Carbonic anhydrases (CAs) play fundamental roles in several physiological events, and emerging evidence points at their involvement in an array of disorders, including cancer. The expression of CAs in the different cells of teeth is unknown, let alone their expression patterns during odontogenesis. As a first step towards understanding the role of CAs during odontogenesis, we used immunohistochemistry, histochemistry and in situ hybridization to reveal hitherto unknown dynamic distribution patterns of eight CAs in mice. The most salient findings include expression of CAII/Car2 not only in maturation-stage ameloblasts (MA) but also in the papillary layer, dental papilla mesenchyme, odontoblasts and the epithelial rests of Malassez. We uncovered that the latter form lace-like networks around incisors; hitherto these have been known to occur only in molars. All CAs studied were produced by MA, however CAIV, CAIX and CARPXI proteins were distinctly enriched in the ruffled membrane of the ruffled MA but exhibited a homogeneous distribution in smooth-ended MA. While CAIV, CAVI/Car6, CAIX, CARPXI and CAXIV were produced by all odontoblasts, CAIII distribution displayed a striking asymmetry, in that it was virtually confined to odontoblasts in the root of molars and root analog of incisors. Remarkably, from initiation until near completion of odontogenesis and in several other tissues, CAXIII localized mainly in intracellular punctae/vesicles that we show to overlap with LAMP-1- and LAMP-2-positive vesicles, suggesting that CAXIII localizes within lysosomes. We showed that expression of CAs in developing teeth is not confined to cells involved in biomineralization, pointing at their participation in other biological events. Finally, we uncovered novel sites of CA expression, including the developing brain and eye, the olfactory epithelium, melanoblasts, tongue, notochord, nucleus pulposus and sebaceous glands. Our study provides important information for future single or multiple gene targeting strategies aiming at deciphering the function of CAs during odontogenesis.


Asunto(s)
Anhidrasas Carbónicas/genética , Anhidrasas Carbónicas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Odontogénesis/genética , Diente/metabolismo , Animales , Animales Recién Nacidos , Inmunohistoquímica , Hibridación in Situ , Isoenzimas , Lisosomas/metabolismo , Ratones , Especificidad de Órganos/genética , Transporte de Proteínas , Diente/embriología , Diente/crecimiento & desarrollo
10.
Cancer Res ; 70(5): 2020-9, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20145151

RESUMEN

Progression to metastasis is the proximal cause of most cancer-related mortality. Yet much remains to be understood about what determines the spread of tumor cells. This paper describes a novel pathway in breast cancer that regulates epithelial-to-mesenchymal transition (EMT), motility, and invasiveness. We identify two transcription factors, nuclear factor 1-C2 (NF1-C2) and Forkhead box F1 (FoxF1), downstream of prolactin/nuclear Janus-activated kinase 2, with opposite effects on these processes. We show that NF1-C2 is lost during mammary tumor progression and is almost invariably absent from lymph node metastases. NF1-C2 levels in primary tumors correlate with better patient survival. Manipulation of NF1-C2 levels by expression of a stabilized version or using small interfering RNA showed that NF1-C2 counteracts EMT, motility, invasiveness, and tumor growth. FoxF1 was found to be a direct repressed target of NF1-C2. We provide the first evidence for a role of FoxF1 in cancer and in the regulation of EMT in cells of epithelial origin. Overexpression of FoxF1 was associated with a mesenchymal phenotype, increased invasiveness in vitro, and enhanced growth of breast carcinoma xenografts in nude mice. The relevance of these findings is strengthened by the correlation between FoxF1 expression and a mesenchymal phenoype in breast cancer cell isolates, consistent with the interpretation that FoxF1 promotes invasion and metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Factores de Transcripción Forkhead/antagonistas & inhibidores , Janus Quinasa 2/metabolismo , Factores de Transcripción NFI/metabolismo , Animales , Neoplasias de la Mama/genética , Adhesión Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Células Epiteliales/patología , Femenino , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Humanos , Mesodermo/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Regiones Promotoras Genéticas
11.
J Biol Chem ; 277(20): 17589-96, 2002 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-11877413

RESUMEN

Members of the nuclear factor 1 (NF1) transcription factor family have been postulated to be involved in the regulation of milk genes. In this work we have been able to identify the splice variant NF1-C2 as an important member of a tissue-specific activating complex that regulates the milk gene encoding carboxyl ester lipase (CEL). Mutation of the NF1-binding site in the CEL gene promoter results in a drastic reduction of the gene expression to about 15% in mammary epithelial cells. Furthermore, we demonstrate that the NF1-C2 protein interacts with a higher affinity to the NF1-binding site in the CEL gene promoter than other NF1 family members do and that NF1-C2 in the mouse mammary gland is a phosphorylated protein. During development of the mouse mammary gland, binding of NF1-C2 to the CEL gene promoter is induced at midpregnancy, in correlation with the induction of CEL gene expression. The fact that the NF1-C2 involving complex remains throughout the lactation period and decreases during the weaning period, when the CEL gene is down-regulated, supports its importance in the regulation of CEL gene expression. To our knowledge, this is the first report identifying a specific, endogenously expressed NF1 isoform to be involved in the tissue-specific activation of a gene.


Asunto(s)
Mama/crecimiento & desarrollo , Proteínas Potenciadoras de Unión a CCAAT/fisiología , Hidrolasas de Éster Carboxílico/genética , Proteínas de Unión al ADN , Regulación de la Expresión Génica , Proteínas de la Leche/genética , Factores de Transcripción , Empalme Alternativo , Animales , Western Blotting , Carboxilesterasa , Diferenciación Celular , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Factores de Transcripción NFI , Proteínas Nucleares , Ratas , Transfección , Células Tumorales Cultivadas , Proteína 1 de Unión a la Caja Y
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA