Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Breast Cancer Res ; 25(1): 143, 2023 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-37964360

RESUMEN

BACKGROUND: As in most solid cancers, the emergence of cells with oncogenic mutations in the mammary epithelium alters the tissue homeostasis. Some soluble factors, such as TGFß, potently modify the behavior of healthy stromal cells. A subpopulation of cancer-associated fibroblasts expressing a TGFß target, the SNAIL1 transcription factor, display myofibroblastic abilities that rearrange the stromal architecture. Breast tumors with the presence of SNAIL1 in the stromal compartment, and with aligned extracellular fiber, are associated with poor survival prognoses. METHODS: We used deep RNA sequencing and biochemical techniques to study alternative splicing and human tumor databases to test for associations (correlation t-test) between SNAIL1 and fibronectin isoforms. Three-dimensional extracellular matrices generated from fibroblasts were used to study the mechanical properties and actions of the extracellular matrices on tumor cell and fibroblast behaviors. A metastatic mouse model of breast cancer was used to test the action of fibronectin isoforms on lung metastasis. RESULTS: In silico studies showed that SNAIL1 correlates with the expression of the extra domain A (EDA)-containing (EDA+) fibronectin in advanced human breast cancer and other types of epithelial cancers. In TGFß-activated fibroblasts, alternative splicing of fibronectin as well as of 500 other genes was modified by eliminating SNAIL1. Biochemical analyses demonstrated that SNAIL1 favors the inclusion of the EDA exon by modulating the activity of the SRSF1 splicing factor. Similar to Snai1 knockout fibroblasts, EDA- fibronectin fibroblasts produce an extracellular matrix  that does not sustain TGFß-induced fiber organization, rigidity, fibroblast activation, or tumor cell invasion. The presence of EDA+ fibronectin changes the action of metalloproteinases on fibronectin fibers. Critically, in an mouse orthotopic breast cancer model, the absence of the fibronectin EDA domain completely prevents lung metastasis. CONCLUSIONS: Our results support the requirement of EDA+ fibronectin in the generation of a metastasis permissive stromal architecture in breast cancers and its molecular control by SNAIL1. From a pharmacological point of view, specifically blocking EDA+ fibronectin deposition could be included in studies to reduce the formation of a pro-metastatic environment.


Asunto(s)
Neoplasias de la Mama , Neoplasias Pulmonares , Animales , Femenino , Humanos , Ratones , Empalme Alternativo , Neoplasias de la Mama/genética , Fibronectinas/genética , Fibronectinas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
2.
Int J Cancer ; 145(11): 3064-3077, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31032902

RESUMEN

Myofibroblasts are a population of highly contractile fibroblasts that express and require the activity of the transcription factor Snail1. Cancer-associated fibroblasts (CAFs) correlate with low survival of cancer patients when present in the stroma of primary tumors. Remarkably, the presence of myofibroblastic CAFs (which express Snail1) creates mechanical properties in the tumor microenvironment that support metastasis. However, therapeutic blockage of fibroblast activity in patients with cancer is a double-edged sword, as normal fibroblast activities often restrict tumor cell invasion. We used fibroblasts depleted of Snail1 or protein arginine methyltransferases 1 and 4 (PRMT1/-4) to identify specific epigenetic modifications induced by TGFß/Snail1. Furthermore, we analyzed the in vivo efficiency of methyltransferase inhibitors using mouse models of wound healing and metastasis, as well as fibroblasts isolated from patients with idiopathic pulmonary fibrosis (IPF). Mechanistically, TGFß-induced Snail1 promotes the epigenetic mark of asymmetrically dimethylated arginine. Critically, we found that inhibitors of methyltransferases prevent myofibroblast activity (but not regular fibroblast activity) in the extracellular matrix, both in cell culture and in vivo. In a mouse breast cancer model, the inhibitor sinefungin reduces both the myofibroblast activity in the tumor stroma and the metastatic burden in the lung. Two distinct inhibitors effectively blocked the exacerbated myofibroblast activity of patient-derived IPF fibroblasts. Our data reveal epigenetic regulation of myofibroblast transdifferentiation in both wound healing and in disease (fibrosis and breast cancer). Thus, methyltransferase inhibitors are good candidates as therapeutic reagents for these diseases.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Inhibidores Enzimáticos/administración & dosificación , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Metiltransferasas/antagonistas & inhibidores , Miofibroblastos/efectos de los fármacos , Factores de Transcripción de la Familia Snail/genética , Adenosina/administración & dosificación , Adenosina/análogos & derivados , Adenosina/farmacología , Animales , Neoplasias de la Mama/enzimología , Fibroblastos Asociados al Cáncer/citología , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/metabolismo , Línea Celular Tumoral , Transdiferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Epigénesis Genética , Femenino , Eliminación de Gen , Humanos , Fibrosis Pulmonar Idiopática/enzimología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/enzimología , Metiltransferasas/genética , Ratones , Miofibroblastos/citología , Miofibroblastos/enzimología , Factores de Transcripción de la Familia Snail/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Biochim Biophys Acta ; 1856(1): 55-61, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26050961

RESUMEN

The transcription factor Snail1 induces epithelial-to-mesenchymal transition (EMT) in tumor epithelial cells, a process associated with the emergence of stemness, invasion and cancer malignancy. Here, we review recent reports indicating that Snail1 also regulates mesenchymal plasticity and paracrine signaling and propose that Snail1 orchestrates the generation of cancer stem cells (CSCs) and cancer-associated fibroblasts (CAFs). Our view supports the current models for tumorigenesis that consider stemness and tumor microenvironment as retroactive actors for metastasis formation, revealing Snail1 as a regulator of these metastatic forces. This view offers new perspectives for understanding and targeting metastasis.


Asunto(s)
Neoplasias/patología , Células Epiteliales/patología , Transición Epitelial-Mesenquimal , Humanos , Células Madre Mesenquimatosas/patología , Neoplasias/fisiopatología , Células Madre Neoplásicas/patología , Factores de Transcripción de la Familia Snail , Factores de Transcripción
4.
Biochim Biophys Acta ; 1825(2): 223-8, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22306657

RESUMEN

The epithelial to mesenchymal transition (EMT) consists of a rapid change of cell phenotype, characterized by the loss of epithelial traits and the acquisition of a more motile phenotype reminiscent of a fibroblast. The study of this process has received considerable attention because of its potential role in the acquisition of several cancer traits, particularly in cell invasion. In this article we describe the current knowledge of the molecular mechanisms governing this transition. In particular we discuss how initiation of EMT is dependent on the mutually exclusive levels of the transmembrane cell to cell adhesion molecule E-cadherin and its transcriptional repressor Snail1 and how Snail1 and other E-cadherin transcriptional repressors drive the EMT process. We focus on several new aspects of Snail1 regulation and propose a model for understanding the initiation and progression of this transition, based on the existence of feed-back mechanisms that limit or amplify the response to extracellular cues.


Asunto(s)
Cadherinas/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Factores de Transcripción/metabolismo , Cadherinas/genética , Transición Epitelial-Mesenquimal/genética , Retroalimentación Fisiológica , Humanos , Transducción de Señal , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética
5.
J Cell Sci ; 124(Pt 24): 4161-71, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22223884

RESUMEN

Snail1 is a transcriptional repressor of E-cadherin that triggers epithelial-mesenchymal transition (EMT). Here, we report assisted Snail1 interaction with the promoter of a typical mesenchymal gene, fibronectin (FN1), both in epithelial cells undergoing EMT and in fibroblasts. Together with Snail1, the p65 subunit of NF-κB and PARP1 bound to the FN1 promoter. We detected nuclear interaction of these proteins and demonstrated the requirement of all three for FN1 transcription. Moreover, other genes involved in cell movement mimic FN1 expression induced by Snail1 or TGF-ß1 treatment and recruit p65NF-κB and Snail1 to their promoters. The molecular cooperation between Snail1 and NF-κB in transcription activation provides a new insight into how Snail1 can modulate a variety of cell programs.


Asunto(s)
Fibronectinas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Factor de Transcripción ReIA/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional , Animales , Sitios de Unión , Línea Celular , Núcleo Celular/metabolismo , Proteínas de la Matriz Extracelular/genética , Fibronectinas/biosíntesis , Humanos , Mesodermo/citología , Ratones , Poli(ADP-Ribosa) Polimerasa-1 , Regiones Promotoras Genéticas , Factores de Transcripción de la Familia Snail
6.
Biochem J ; 435(3): 563-8, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21361876

RESUMEN

Fibronectins are cell-secreted glycoproteins that modulate cell attachment, spreading, migration, morphology, differentiation and oncogenic transformation. Fibronectin expression is activated during EMT (epithelial-mesenchymal transition) and is a hallmark of mesenchymal cells. It is shown in the present study that a transcription factor previously unrelated with EMT, TFCP2c/LSF/LBP-1c, was translocated to the nucleus and bound to the fibronectin promoter upon EMT induction by Snail1. Consequently, the interference of TFCP2c/LSF/LBP-1c's activity prevented fibronectin expression. Moreover, TFCP2c/LSF/LBP-1c was detected in nuclei of embryonic dermal mesenchymal cells adjacent to the hair bud, a cell population that expresses endogenous nuclear Snail1 and fibronectin. Therefore we indicate a new molecular role for TFCP2c/LSF/LBP-1c in fibronectin expression.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Fibronectinas/metabolismo , Regulación de la Expresión Génica/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Línea Celular , Embrión de Mamíferos/metabolismo , Fibronectinas/genética , Humanos , Ratones , Regiones Promotoras Genéticas , Unión Proteica , Transporte de Proteínas/fisiología , Factores de Transcripción de la Familia Snail
7.
J Biol Chem ; 285(6): 3794-3805, 2010 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-19955572

RESUMEN

The transcription factor SNAIL1 is a master regulator of epithelial to mesenchymal transition. SNAIL1 is a very unstable protein, and its levels are regulated by the E3 ubiquitin ligase beta-TrCP1 that interacts with SNAIL1 upon its phosphorylation by GSK-3beta. Here we show that SNAIL1 polyubiquitylation and degradation may occur in conditions precluding SNAIL1 phosphorylation by GSK-3beta, suggesting that additional E3 ligases participate in the control of SNAIL1 protein stability. In particular, we demonstrate that the F-box E3 ubiquitin ligase FBXl14 interacts with SNAIL1 and promotes its ubiquitylation and proteasome degradation independently of phosphorylation by GSK-3beta. In vivo, inhibition of FBXl14 using short hairpin RNA stabilizes both ectopically expressed and endogenous SNAIL1. Moreover, the expression of FBXl14 is potently down-regulated during hypoxia, a condition that increases the levels of SNAIL1 protein but not SNAIL1 mRNA. FBXL14 mRNA is decreased in tumors with a high expression of two proteins up-regulated in hypoxia, carbonic anhydrase 9 and TWIST1. In addition, Twist1 small interfering RNA prevents hypoxia-induced Fbxl14 down-regulation and SNAIL1 stabilization in NMuMG cells. Altogether, these results demonstrate the existence of an alternative mechanism controlling SNAIL1 protein levels relevant for the induction of SNAIL1 during hypoxia.


Asunto(s)
Proteínas F-Box/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Sitios de Unión , Western Blotting , Hipoxia de la Célula , Línea Celular , Línea Celular Tumoral , Regulación hacia Abajo , Proteínas F-Box/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Inmunoprecipitación , Ratones , Mutación , Células 3T3 NIH , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilación , Unión Proteica , Interferencia de ARN , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Transfección , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
8.
Cancer Res ; 81(2): 438-451, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33229340

RESUMEN

Tumors are complex tissues composed of transformed epithelial cells as well as cancer-activated fibroblasts (CAF) that facilitate epithelial tumor cell invasion. We show here that CAFs and other mesenchymal cells rely much more on glutamine than epithelial tumor cells; consequently, they are more sensitive to inhibition of glutaminase. Glutamine dependence drove CAF migration toward this amino acid when cultured in low glutamine conditions. CAFs also invaded a Matrigel matrix following a glutamine concentration gradient and enhanced the invasion of tumor cells when both cells were cocultured. Accordingly, glutamine directed invasion of xenografted tumors in immunocompromised mice. Stimulation of glutamine-driven epithelial tumor invasion by fibroblasts required previous CAF activation, which involved the TGFß/Snail1 signaling axis. CAFs moving toward Gln presented a polarized Akt2 distribution that was modulated by the Gln-dependent activity of TRAF6 and p62 in the migrating front, and depletion of these proteins prevented Akt2 polarization and Gln-driven CAF invasion. Our results demonstrate that glutamine deprivation promotes CAF migration and invasion, which in turn facilitates the movement of tumor epithelial cells toward nutrient-rich territories. These results provide a novel molecular mechanism for how metabolic stress enhances invasion and metastasis. SIGNIFICANCE: Cancer-associated fibroblasts migrate and invade toward free glutamine and facilitate invasion of tumor epithelial cells, accounting for their movement away from the hostile conditions of the tumor towards nutrient-rich adjacent tissues. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/2/438/F1.large.jpg.


Asunto(s)
Neoplasias de la Mama/patología , Fibroblastos Asociados al Cáncer/patología , Movimiento Celular , Transición Epitelial-Mesenquimal , Glutamina/farmacología , Neoplasias Glandulares y Epiteliales/patología , Animales , Apoptosis , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/metabolismo , Proliferación Celular , Femenino , Humanos , Ratones , Ratones Desnudos , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Glandulares y Epiteliales/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Clin Med ; 8(6)2019 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-31141910

RESUMEN

Snail1 transcriptional factor plays a key role in the control of epithelial to mesenchymal transition and fibroblast activation. As a consequence, Snail1 expression and function is regulated at multiple levels from gene transcription to protein modifications, affecting its interaction with specific cofactors. In this review, we describe the different elements that control Snail1 expression and its activity both as transcriptional repressor or activator.

10.
Nucleic Acids Res ; 34(7): 2077-84, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16617148

RESUMEN

The product of Snail1 gene is a transcriptional repressor of E-cadherin expression and an inductor of the epithelial-mesenchymal transition in several epithelial tumour cell lines. Transcription of Snail1 is induced when epithelial cells are forced to acquire a mesenchymal phenotype. In this work we demonstrate that Snail1 protein limits its own expression: Snail1 binds to an E-box present in its promoter (at -146 with respect to the transcription start) and represses its activity. Therefore, mutation of the E-box increases Snail1 transcription in epithelial and mesenchymal cells. Evidence of binding of ectopic or endogenous Snail1 to its own promoter was obtained by chromatin immunoprecipitation (ChIP) experiments. Studies performed expressing different forms of Snail1 under the control of its own promoter demonstrate that disruption of the regulatory loop increases the cellular levels of Snail protein. These results indicate that expression of Snail1 gene can be regulated by its product and evidence the existence of a fine-tuning feed-back mechanism of regulation of Snail1 transcription.


Asunto(s)
Elementos E-Box , Regulación de la Expresión Génica , Regiones Promotoras Genéticas , Proteínas Represoras/genética , Factores de Transcripción/genética , Animales , Sitios de Unión , Línea Celular , Regulación hacia Abajo , Homeostasis , Humanos , Ratones , ARN Mensajero/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo
11.
Mol Cell Biol ; 23(14): 5078-89, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12832491

RESUMEN

The Snail gene product is a transcriptional repressor of E-cadherin expression and an inducer of the epithelial-to-mesenchymal transition in several epithelial tumor cell lines. This report presents data indicating that Snail function is controlled by its intracellular location. The cytosolic distribution of Snail depended on export from the nucleus by a CRM1-dependent mechanism, and a nuclear export sequence (NES) was located in the regulatory domain of this protein. Export of Snail was controlled by phosphorylation of a Ser-rich sequence adjacent to this NES. Modification of this sequence released the restriction created by the zinc finger domain and allowed nuclear export of the protein. The phosphorylation and subcellular distribution of Snail are controlled by cell attachment to the extracellular matrix. Suspended cells presented higher levels of phosphorylated Snail and an augmented extranuclear localization with respect to cells attached to the plate. These findings show the existence in tumor cells of an effective and fine-tuning nontranscriptional mechanism of regulation of Snail activity dependent on the extracellular environment.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Receptores Citoplasmáticos y Nucleares , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Animales , Secuencia de Bases , Núcleo Celular/genética , Núcleo Celular/metabolismo , Citosol/metabolismo , Matriz Extracelular/metabolismo , Humanos , Carioferinas/metabolismo , Leucina/metabolismo , Ratones , Datos de Secuencia Molecular , Señales de Localización Nuclear/genética , Señales de Localización Nuclear/metabolismo , Fosforilación , Estructura Terciaria de Proteína , Transporte de Proteínas/fisiología , Serina/metabolismo , Factores de Transcripción de la Familia Snail , Fracciones Subcelulares , Células Tumorales Cultivadas , Proteína Exportina 1
12.
Mol Oncol ; 11(7): 847-859, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28544627

RESUMEN

Beyond inducing epithelial-to-mesenchymal transcription (EMT), transcriptional factors of the Snail, ZEB and Twist families (EMT-TFs) control global plasticity programmes affecting cell stemness and fate. Literature addressing the reactivation of these factors in adult tumour cells is very extensive, as they enable cancer cell plasticity and fuel both tumour initiation and metastatic spread. Incipient data reveal that EMT-TFs are also expressed in fibroblasts, providing these with additional properties. Here, I will review recent reports on the expression of EMT-TFs in cancer-associated fibroblasts (CAFs). The new model suggests that EMT-TFs can be envisioned as essential metastasis and chemoresistance-promoting molecules, thereby enabling coordinated plasticity programmes in parenchyma and stroma-tumour compartments.


Asunto(s)
Transición Epitelial-Mesenquimal , Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Factores de Transcripción/metabolismo , Microambiente Tumoral , Animales , Fibroblastos/patología , Humanos , Neoplasias/patología
14.
Cancer Res ; 76(21): 6205-6217, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27503928

RESUMEN

Snail1 transcriptional factor is essential for triggering epithelial-to-mesenchymal transition (EMT) and inducing tumor cell invasion. We report here an EMT-independent action of Snail1 on tumor invasion, as it is required for the activation of cancer-associated fibroblasts (CAF). Snail1 expression in fibroblasts requires signals derived from tumor cells, such as TGFß; reciprocally, in fibroblasts, Snail1 organizes a complex program that stimulates invasion of epithelial cells independent of the expression of Snail1 in these cells. Epithelial cell invasion is stimulated by the secretion by fibroblast of diffusible signaling molecules, such as prostaglandin E2 The capability of human or murine CAFs to promote tumor invasion is dependent on Snail1 expression. Inducible Snail1 depletion in mice decreases the invasion of breast tumors; moreover, epithelial tumor cells coxenografted with Snail1-depleted fibroblasts originated tumors with lower invasion than those transplanted with control fibroblasts. Therefore, these results demonstrate that the role of Snail1 in tumor invasion is not limited to EMT, but it is also dependent on its activity in stromal fibroblasts, where it orchestrates the cross-talk with epithelial tumor cells. Cancer Res; 76(21); 6205-17. ©2016 AACR.


Asunto(s)
Fibroblastos Asociados al Cáncer/fisiología , Neoplasias Glandulares y Epiteliales/patología , Factores de Transcripción de la Familia Snail/fisiología , Animales , Línea Celular Tumoral , Dinoprostona/metabolismo , Transición Epitelial-Mesenquimal , Humanos , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Factor de Crecimiento Transformador beta/farmacología
15.
Oncogene ; 23(44): 7345-54, 2004 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-15286702

RESUMEN

Expression of Snail transcriptional factor is a determinant in the acquisition of a mesenchymal phenotype by epithelial tumor cells. However, the regulation of the transcription of this gene is still unknown. We describe here the characterization of a human SNAIL promoter that contains the initiation of transcription and regulates the expression of this gene in tumor cells. This promoter was activated in cell lines in response to agents that induce Snail transcription and the mesenchymal phenotype, as addition of the phorbol ester PMA or overexpression of integrin-linked kinase (ILK) or oncogenes such as Ha-ras or v-Akt. Although other regions of the promoter were required for a complete stimulation by Akt or ILK, a minimal fragment (-78/+59) was sufficient to maintain the mesenchymal specificity. Activity of this minimal promoter and SNAIL RNA levels were dependent on ERK signaling pathway. NFkappaB/p65 also stimulated SNAIL transcription through a region located immediately upstream the minimal promoter, between -194 and -78. These results indicate that Snail transcription is driven by signaling pathways known to induce epithelial to mesenchymal transition, reinforcing the role of Snail in this process.


Asunto(s)
Proteínas de Unión al ADN/genética , Células Epiteliales/citología , Regulación Neoplásica de la Expresión Génica/genética , Mesodermo/citología , Factores de Transcripción/genética , Transcripción Genética/genética , Dedos de Zinc , Diferenciación Celular , Línea Celular Tumoral , Humanos , Factores de Transcripción de la Familia Snail
16.
Cancer Res ; 75(2): 284-95, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25488750

RESUMEN

Crosstalk between tumor and stromal cells in the tumor microenvironment alter its properties in ways that facilitate the invasive behavior of tumor cells. Here, we demonstrate that cancer-associated fibroblasts (CAF) increase the stiffness of the extracellular matrix (ECM) and promote anisotropic fiber orientation, two mechanical signals generated through a Snail1/RhoA/αSMA-dependent mechanism that sustains oriented tumor cell migration and invasiveness. Snail1-depleted CAF failed to acquire myofibroblastic traits in response to TGFß, including RhoA activation, αSMA-positive stress fibers, increased fibronectin fibrillogenesis, and production of a stiff ECM with oriented fibers. Snail1 expression in human tumor-derived CAF was associated with an ability to organize the ECM. In coculture, a relatively smaller number of Snail1-expressing CAF were capable of imposing an anisotropic ECM architecture, compared with nonactivated fibroblasts. Pathologically, human breast cancers with Snail1(+) CAF tended to exhibit desmoplastic areas with anisotropic fibers, lymph node involvement, and poorer outcomes. Snail1 involvement in driving an ordered ECM was further confirmed in wound-healing experiments in mice, with Snail1 depletion preventing the anisotropic organization of granulation tissue and delaying wound healing. Overall, our results showed that inhibiting Snail1 function in CAF could prevent tumor-driven ECM reorganization and cancer invasion.


Asunto(s)
Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Factores de Transcripción/biosíntesis , Animales , Humanos , Ratones , Ratones Noqueados , Metástasis de la Neoplasia , Factores de Transcripción de la Familia Snail , Microambiente Tumoral , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
17.
Oncoscience ; 2(11): 898-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26697515
18.
PLoS One ; 3(12): e4080, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19114997

RESUMEN

Presenilin1 (PS1) is a component of the gamma-secretase complex mutated in cases of Familial Alzheimer's disease (FAD). PS1 is synthesized as a 50 kDa peptide subsequently processed to two 29 and 20 kDa subunits that remain associated. Processing of PS1 is inhibited by several mutations detected in FAD patients. PS1 acts as negative modulator of beta-catenin.Tcf-4 transcriptional activity. In this article we show that in murine embryonic fibroblasts (MEFs) the mechanisms of action of the processed and non-processed forms of PS1 on beta-catenin.Tcf-4 transcription are different. Whereas non-processed PS1 inhibits beta-catenin.Tcf-4 activity through a mechanism independent of gamma-secretase and associated with the interaction of this protein with plakoglobin and Tcf-4, the effect of processed PS1 is prevented by gamma-secretase inhibitors, and requires its interaction with E- or N-cadherin and the generation of cytosolic terminal fragments of these two cadherins, which in turn destabilize the beta-catenin transcriptional cofactor CBP. Accordingly, the two forms of PS1 interact differently with E-cadherin or beta-catenin and plakoglobin: whereas processed PS1 binds E-cadherin with high affinity and beta-catenin or plakoglobin weakly, the non-processed form behaves inversely. Moreover, contrarily to processed PS1, that decreases the levels of c-fos RNA, non-processed PS1 inhibits the expression c-myc, a known target of beta-catenin.Tcf-4, and does not block the activity of other transcriptional factors requiring CBP. These results indicate that prevention of PS1 processing in FAD affects the mechanism of repression of the transcriptional activity dependent on beta-catenin.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Presenilina-1/metabolismo , Factores de Transcripción TCF/genética , Transcripción Genética , beta Catenina/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Humanos , Ratones , Factores de Transcripción TCF/metabolismo , Transfección , beta Catenina/genética
19.
J Cell Sci ; 121(Pt 13): 2224-34, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18565826

RESUMEN

E-cadherin and its transcriptional repressor Snail1 (Snai1) are two factors that control epithelial phenotype. Expression of Snail1 promotes the conversion of epithelial cells to mesenchymal cells, and occurs concomitantly with the downregulation of E-cadherin and the upregulation of expression of mesenchymal genes such as those encoding fibronectin and LEF1. We studied the molecular mechanism controlling the expression of these genes in mesenchymal cells. Forced expression of E-cadherin strongly downregulated fibronectin and LEF1 RNA levels, indicating that E-cadherin-sensitive factors are involved in the transcription of these genes. E-cadherin overexpression decreased the transcriptional activity of the fibronectin promoter and reduced the interaction of beta-catenin and NF-kappaB with this promoter. Similar to beta-catenin, NF-kappaB was found, by co-immunoprecipitation and pull-down assays, to be associated with E-cadherin and other cell-adhesion components. Interaction of the NF-kappaB p65 subunit with E-cadherin or beta-catenin was reduced when adherens junctions were disrupted by K-ras overexpression or by E-cadherin depletion using siRNA. These conditions did not affect the association of p65 with the NF-kappaB inhibitor IkappaBalpha. The functional significance of these results was stressed by the stimulation of NF-kappaB transcriptional activity, both basal and TNF-alpha-stimulated, induced by an E-cadherin siRNA. Therefore, these results demonstrate that E-cadherin not only controls the transcriptional activity of beta-catenin but also that of NF-kappaB. They indicate too that binding of this latter factor to the adherens junctional complex prevents the transcription of mesenchymal genes.


Asunto(s)
Cadherinas/genética , Fibronectinas/genética , Regulación de la Expresión Génica , Factor de Unión 1 al Potenciador Linfoide/genética , FN-kappa B/genética , Transcripción Genética , beta Catenina/genética , Cadherinas/metabolismo , Línea Celular , Fibronectinas/metabolismo , Humanos , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Mesodermo/metabolismo , FN-kappa B/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , beta Catenina/metabolismo
20.
J Biol Chem ; 277(42): 39209-16, 2002 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-12161443

RESUMEN

E-cadherin protein plays a key role in the establishment and maintenance of adherent junctions. Recent evidence implicates the transcription factor Snail in the blockage of E-cadherin expression in fibroblasts and some epithelial tumor cells through direct binding to three E-boxes in the E-cadherin promoter. Transfection of Snail into epithelial cells leads to a more fibroblastic phenotype. Cells expressing Snail presented a scattered flattened phenotype with low intercellular contacts. Other epithelial markers like Cytokeratin 18 or MUC1 were also repressed. The effects of Snail on MUC1 transcription were mediated by two E-boxes present in the proximal promoter. Snail also induced expression of the mesenchymal markers fibronectin and LEF1 and the transcription repressor ZEB1. ZEB1 and Snail had a similar pattern of expression in epithelial cell lines, and both were induced by overexpression of ILK1, a kinase that causes the loss of E-cadherin and the acquisition of a fibroblastic phenotype. Snail overexpression in several cell lines raised ZEB1 RNA levels and increased the activity of ZEB1 promoter. ZEB1 could also repress E-cadherin and MUC1 promoters but less strongly than Snail. However, since ZEB1 expression persisted after Snail was down-regulated, ZEB1 may regulate epithelial genes in several tumor cell lines.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Células Epiteliales/metabolismo , Mesodermo/metabolismo , Mucina-1/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Animales , Northern Blotting , Western Blotting , Línea Celular , Núcleo Celular/metabolismo , Clonación Molecular , ADN/metabolismo , ADN Complementario/metabolismo , Perros , Regulación hacia Abajo , Fibroblastos/metabolismo , Humanos , Queratinas/metabolismo , Ratones , Microscopía Electrónica , Fenotipo , Regiones Promotoras Genéticas , Unión Proteica , ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción de la Familia Snail , Tetraciclina/farmacología , Transcripción Genética , Transfección , Células Tumorales Cultivadas , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA