Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
EMBO Rep ; 25(4): 1886-1908, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38413734

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a tumor with a dismal prognosis that arises from precursor lesions called pancreatic intraepithelial neoplasias (PanINs). Progression from low- to high-grade PanINs is considered as tumor initiation, and a deeper understanding of this switch is needed. Here, we show that synaptic molecule neuroligin-2 (NLGN2) is expressed by pancreatic exocrine cells and plays a crucial role in the regulation of contact inhibition and epithelial polarity, which characterize the switch from low- to high-grade PanIN. NLGN2 localizes to tight junctions in acinar cells, is diffusely distributed in the cytosol in low-grade PanINs and is lost in high-grade PanINs and in a high percentage of advanced PDACs. Mechanistically, NLGN2 is necessary for the formation of the PALS1/PATJ complex, which in turn induces contact inhibition by reducing YAP function. Our results provide novel insights into NLGN2 functions outside the nervous system and can be used to model PanIN progression.


Asunto(s)
Carcinoma in Situ , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Neuroliginas , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Carcinoma in Situ/patología , Transformación Celular Neoplásica
2.
Bio Protoc ; 13(9): e4662, 2023 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-37188107

RESUMEN

X-ray computed microtomography (µCT) is a powerful tool to reveal the 3D structure of tissues and organs. Compared with the traditional sectioning, staining, and microscopy image acquisition, it allows a better understanding of the morphology and a precise morphometric analysis. Here, we describe a method for 3D visualization and morphometric analysis by µCT scanning of the embryonic heart of iodine-stained E15.5 mouse embryos.

3.
Pharmacol Ther ; 246: 108418, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37088448

RESUMEN

Endothelial cells (ECs) that line vascular and lymphatic vessels are being increasingly recognized as important to organ function in health and disease. ECs participate not only in the trafficking of gases, metabolites, and cells between the bloodstream and tissues but also in the angiocrine-based induction of heterogeneous parenchymal cells, which are unique to their specific tissue functions. The molecular mechanisms regulating EC heterogeneity between and within different tissues are modeled during embryogenesis and become fully established in adults. Any changes in adult tissue homeostasis induced by aging, stress conditions, and various noxae may reshape EC heterogeneity and induce specific transcriptional features that condition a functional phenotype. Heterogeneity is sustained via specific genetic programs organized through the combinatory effects of a discrete number of transcription factors (TFs) that, at the single tissue-level, constitute dynamic networks that are post-transcriptionally and epigenetically regulated. This review is focused on outlining the TF-based networks involved in EC specialization and physiological and pathological stressors thought to modify their architecture.


Asunto(s)
Células Endoteliales , Factores de Transcripción , Células Endoteliales/metabolismo , Factores de Transcripción/metabolismo
4.
Nat Commun ; 13(1): 5191, 2022 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-36057632

RESUMEN

Epithelial-mesenchymal transition (EMT) is a complex and pivotal process involved in organogenesis and is related to several pathological processes, including cancer and fibrosis. During heart development, EMT mediates the conversion of epicardial cells into vascular smooth muscle cells and cardiac interstitial fibroblasts. Here, we show that the oncogenic transcription factor EB (TFEB) is a key regulator of EMT in epicardial cells and that its genetic overexpression in mouse epicardium is lethal due to heart defects linked to impaired EMT. TFEB specifically orchestrates the EMT-promoting function of transforming growth factor (TGF) ß, and this effect results from activated transcription of thymine-guanine-interacting factor (TGIF)1, a TGFß/Smad pathway repressor. The Tgif1 promoter is activated by TFEB, and in vitro and in vivo findings demonstrate its increased expression when Tfeb is overexpressed. Furthermore, Tfeb overexpression in vitro prevents TGFß-induced EMT, and this effect is abolished by Tgif1 silencing. Tfeb loss of function, similar to that of Tgif1, sensitizes cells to TGFß, inducing an EMT response to low doses of TGFß. Together, our findings reveal an unexpected function of TFEB in regulating EMT, which might provide insights into injured heart repair and control of cancer progression.


Asunto(s)
Transición Epitelial-Mesenquimal , Factor de Crecimiento Transformador beta , Animales , Células Cultivadas , Transición Epitelial-Mesenquimal/fisiología , Ratones , Organogénesis , Pericardio/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
5.
FEBS Lett ; 596(16): 1973-1980, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35781277

RESUMEN

Transcription factor EB (TFEB) belongs to the microphthalmia family of bHLH-leucine zipper transcription factors and was first identified as an oncogene in a subset of renal cell carcinomas. In addition to exhibiting oncogenic activity, TFEB coordinates genetic programs connected with the cellular response to stress conditions, including roles in lysosome biogenesis, autophagy, and modulation of metabolism. As is the case for other transcription factors, the activities of TFEB are not limited to a specific cellular condition such as the response to stress, and recent findings indicate that TFEB has more widespread functions. Here, we review the emerging roles of TFEB in regulating cellular proliferation and motility. The well-established and emerging roles of TFEB suggest that this protein serves as a hub of signaling networks involved in many non-communicable diseases, such as cancer, ischaemic diseases and immune disorders, drug resistance mechanisms, and tissue generation.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Neoplasias , Autofagia , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Puntos de Control del Ciclo Celular , Humanos , Lisosomas , Factores de Transcripción
6.
Angiogenesis ; 25(4): 471-492, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35545719

RESUMEN

The dynamic integrin-mediated adhesion of endothelial cells (ECs) to the surrounding ECM is fundamental for angiogenesis both in physiological and pathological conditions, such as embryonic development and cancer progression. The dynamics of EC-to-ECM adhesions relies on the regulation of the conformational activation and trafficking of integrins. Here, we reveal that oncogenic transcription factor EB (TFEB), a known regulator of lysosomal biogenesis and metabolism, also controls a transcriptional program that influences the turnover of ECM adhesions in ECs by regulating cholesterol metabolism. We show that TFEB favors ECM adhesion turnover by promoting the transcription of genes that drive the synthesis of cholesterol, which promotes the aggregation of caveolin-1, and the caveolin-dependent endocytosis of integrin ß1. These findings suggest that TFEB might represent a novel target for the pharmacological control of pathological angiogenesis and bring new insights in the mechanism sustaining TFEB control of endocytosis.


Asunto(s)
Células Endoteliales , Integrinas , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Caveolina 1/metabolismo , Adhesión Celular/genética , Colesterol , Células Endoteliales/metabolismo , Humanos , Integrina beta1/metabolismo , Integrinas/metabolismo , Neovascularización Patológica/metabolismo
7.
Angiogenesis ; 25(1): 113-128, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34478025

RESUMEN

Embryonic stem cells (ES) are a valuable source of endothelial cells. By co-culturing ES cells with the stromal PA6 cells, the endothelial commitment can be achieved by adding exogenous FGF2 or BMP4. In this work, the molecular pathways that direct the differentiation of ES cells toward endothelium in response to FGF2 are evaluated and compared to those activated by BMP4. To this purpose the genes expression profiles of both ES/PA6 co-cultures and of pure cultures of PA6 cells were obtained by microarray technique at different time points. The bioinformatics processing of the data indicated TGFß1 as the most represented upstream regulator in FGF2-induced endothelial commitment while WNT pathway as the most represented in BMP4-activated endothelial differentiation. Loss of function experiments were performed to validate the importance of TGFß1 and WNT6 respectively in FGF2 and BMP4-induced endothelial differentiation. The loss of TGFß1 expression significantly impaired the accomplishment of the endothelial commitment unless exogenous recombinant TGFß1 was added to the culture medium. Similarly, silencing WNT6 expression partially affected the endothelial differentiation of the ES cells upon BMP4 stimulation. Such dysfunction was recovered by the addition of recombinant WNT6 to the culture medium. The ES/PA6 co-culture system recreates an in vitro complete microenvironment in which endothelial commitment is accomplished in response to alternative signals through different mechanisms. Given the importance of WNT and TGFß1 in mediating the crosstalk between tumor and stromal cells this work adds new insights in the mechanism of tumor angiogenesis and of its possible inhibition.


Asunto(s)
Células Endoteliales , Factor 2 de Crecimiento de Fibroblastos , Factor de Crecimiento Transformador beta1/fisiología , Animales , Proteína Morfogenética Ósea 4 , Diferenciación Celular , Células Madre Embrionarias , Factor 2 de Crecimiento de Fibroblastos/farmacología , Ratones , Proteínas Proto-Oncogénicas , Células del Estroma , Factor de Crecimiento Transformador beta1/genética , Proteínas Wnt
8.
Int J Mol Sci ; 22(9)2021 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-34067060

RESUMEN

Recent findings suggest that epithelial to mesenchymal transition (EMT), a key step during heart development, is involved in cardiac tissue repair following myocardial infarction (MI). MicroRNAs (miRNAs) act as key regulators in EMT processes; however, the mechanisms by which miRNAs target epicardial EMT remain largely unknown. Here, by using an in vitro model of epicardial EMT, we investigated the role of miRNAs as regulators of this process and their potential targets. EMT was induced in murine epicardial-mesothelial cells (EMCs) through TGF ß1 treatment for 48, 72, and 96 h as indicated by the expression of EMT-related genes by qRT-PCR, WB, and immunofluorescence. Further, enhanced expression of stemness genes was also detected. Among several EMT-related miRNAs, miR-200c-3p expression resulted as the most strongly suppressed. Interestingly, we also found a significant upregulation of Follistatin-related protein 1 (FSTL1), a miR-200c predicted target already identified as a potent cardiogenic factor produced by epicardial cells that promotes regeneration following MI. Dual-luciferase reporter assay demonstrated that miR-200c-3p directly targeted the 3'-untranslated region of FSTL1 in EMCs. Consistently, WB analysis showed that knockdown of miR-200c-3p significantly increased FSTL1 expression, whereas overexpression of miR-200c-3p counteracted TGF ß1-mediated FSTL1 upregulation. Importantly, FSTL1 silencing maintained epithelial features in EMCs, despite EMT induction by TGF ß1, and attenuated EMT-associated traits, including migration and stemness. In conclusion, epicardial FSTL1, an important cardiogenic factor in its secreted form, induces EMT, stemness, and migration of EMCs in a miR-200c-3p dependent pathway.


Asunto(s)
Transición Epitelial-Mesenquimal , Epitelio/metabolismo , Proteínas Relacionadas con la Folistatina/metabolismo , MicroARNs/metabolismo , Pericardio/patología , Animales , Biomarcadores/metabolismo , Transición Epitelial-Mesenquimal/genética , Femenino , Mesodermo/patología , Ratones Endogámicos C57BL , MicroARNs/genética , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Factor de Crecimiento Transformador beta1/farmacología
9.
Front Physiol ; 12: 640061, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33912071

RESUMEN

Transcription factor EB (TFEB) represents an emerging player in vascular biology. It belongs to the bHLH-leucine zipper transcription factor microphthalmia family, which includes microphthalmia-associated transcription factor, transcription factor E3 and transcription factor EC, and is known to be deregulated in cancer. The canonical transcriptional pathway orchestrated by TFEB adapts cells to stress in all kinds of tissues by supporting lysosomal and autophagosome biogenesis. However, emerging findings highlight that TFEB activates other genetic programs involved in cell proliferation, metabolism, inflammation and immunity. Here, we first summarize the general principles and mechanisms by which TFEB activates its transcriptional program. Then, we analyze the current knowledge of TFEB in the vascular system, placing particular emphasis on its regulatory role in angiogenesis and on the involvement of the vascular unit in inflammation and atherosclerosis.

10.
Mol Oncol ; 15(2): 327-346, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33252196

RESUMEN

Transcription factor EB (TFEB) represents an emerging player in cancer biology. Together with microphthalmia-associated transcription factor, transcription factor E3 and transcription factor EC, TFEB belongs to the microphthalmia family of bHLH-leucine zipper transcription factors that may be implicated in human melanomas, renal and pancreatic cancers. TFEB was originally described as being translocated in a juvenile subset of pediatric renal cell carcinoma; however, whole-genome sequencing reported that somatic mutations were sporadically found in many different cancers. Besides its oncogenic activity, TFEB controls the autophagy-lysosomal pathway by recognizing a recurrent motif present in the promoter regions of a set of genes that participate in lysosome biogenesis; furthermore, its dysregulation was found to have a crucial pathogenic role in different tumors by modulating the autophagy process. Other than regulating cancer cell-autonomous responses, recent findings indicate that TFEB participates in the regulation of cellular functions of the tumor microenvironment. Here, we review the emerging role of TFEB in regulating cancer cell behavior and choreographing tumor-microenvironment interaction. Recognizing TFEB as a hub of network of signals exchanged within the tumor between cancer and stroma cells provides a fresh perspective on the molecular principles of tumor self-organization, promising to reveal numerous new and potentially druggable vulnerabilities.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Melanoma/metabolismo , Proteínas de Neoplasias/metabolismo , Microambiente Tumoral , Humanos , Melanoma/patología
11.
EMBO J ; 38(3)2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30591554

RESUMEN

Transcription factor TFEB is thought to control cellular functions-including in the vascular bed-primarily via regulation of lysosomal biogenesis and autophagic flux. Here, we report that TFEB also orchestrates a non-canonical program that controls the cell cycle/VEGFR2 pathway in the developing vasculature. In endothelial cells, TFEB depletion halts proliferation at the G1-S transition by inhibiting the CDK4/Rb pathway. TFEB-deficient cells attempt to compensate for this limitation by increasing VEGFR2 levels at the plasma membrane via microRNA-mediated mechanisms and controlled membrane trafficking. TFEB stimulates expression of the miR-15a/16-1 cluster, which limits VEGFR2 transcript stability and negatively modulates expression of MYO1C, a regulator of VEGFR2 trafficking to the cell surface. Altered levels of miR-15a/16-1 and MYO1C in TFEB-depleted cells cause increased expression of plasma membrane VEGFR2, but in a manner associated with low signaling strength. An endothelium-specific Tfeb-knockout mouse model displays defects in fetal and newborn mouse vasculature caused by reduced endothelial proliferation and by anomalous function of the VEGFR2 pathway. These previously unrecognized functions of TFEB expand its role beyond regulation of the autophagic pathway in the vascular system.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/fisiología , Proliferación Celular , Embrión de Mamíferos/citología , Endotelio Vascular/citología , Regulación del Desarrollo de la Expresión Génica , Neovascularización Fisiológica , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Células Cultivadas , Embrión de Mamíferos/fisiología , Endotelio Vascular/fisiología , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
12.
Nat Commun ; 7: 13546, 2016 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-27876801

RESUMEN

Basolateral polymerization of cellular fibronectin (FN) into a meshwork drives endothelial cell (EC) polarity and vascular remodelling. However, mechanisms coordinating α5ß1 integrin-mediated extracellular FN endocytosis and exocytosis of newly synthesized FN remain elusive. Here we show that, on Rab21-elicited internalization, FN-bound/active α5ß1 is recycled to the EC surface. We identify a pathway, comprising the regulators of post-Golgi carrier formation PI4KB and AP-1A, the small GTPase Rab11B, the surface tyrosine phosphatase receptor PTPRF and its adaptor PPFIA1, which we propose acts as a funnel combining FN secretion and recycling of active α5ß1 integrin from the trans-Golgi network (TGN) to the EC surface, thus allowing FN fibrillogenesis. In this framework, PPFIA1 interacts with active α5ß1 integrin and localizes close to EC adhesions where post-Golgi carriers are targeted. We show that PPFIA1 is required for FN polymerization-dependent vascular morphogenesis, both in vitro and in the developing zebrafish embryo.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Fibronectinas/metabolismo , Regulación de la Expresión Génica/fisiología , Integrina alfa5beta1/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células Cultivadas , Embrión no Mamífero , Fibronectinas/genética , Aparato de Golgi/fisiología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrina alfa5beta1/genética , Neovascularización Fisiológica/fisiología , Pez Cebra
13.
Trends Mol Med ; 20(10): 589-98, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25154329

RESUMEN

During organogenesis, patterning is primarily achieved by the combined actions of morphogens. Among these, semaphorins represent a general system for establishing the appropriate wiring architecture of biological nets. Originally discovered as evolutionarily conserved steering molecules for developing axons, subsequent studies on semaphorins expanded their functions to the cardiovascular and immune systems. Semaphorins participate in cardiac organogenesis and control physiological vasculogenesis and angiogenesis, which result from a balance between pro- and anti-angiogenic signals. These signals are altered in several diseases. In this review, we discuss the role of semaphorins in vascular biology, emphasizing the mechanisms by which these molecules control vascular patterning and lymphangiogenesis, as well as in genetically inherited and degenerative vascular diseases.


Asunto(s)
Corazón/embriología , Linfangiogénesis , Semaforinas/fisiología , Animales , Aterosclerosis/metabolismo , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Corazón/inervación , Humanos , Isquemia/metabolismo , Ratones , Terapia Molecular Dirigida , Neovascularización Fisiológica , Semaforinas/genética , Semaforinas/metabolismo
14.
EMBO J ; 32(5): 609-11, 2013 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-23361316

RESUMEN

The lymphatic system is indispensable for the collection and cycling of tissue-extravasated fluids, macromolecules and immune cells into the bloodstream. Different mechanisms, including sprouting, ballooning and budding of lymphatic endothelial cells from the cardinal vein, have been proposed for lymphatic vessel formation during mammalial embryogenesis. Hägerling et al (2013) now provide a cell-scale model of lymphoangiogenesis by applying selective plane illumination-based ultramicroscopy (Becker et al, 2008) to wholemount-immunostained mouse embryos. They describe VEGFR-3, VEGF-C and CCBE1 as key regulators of lymphatic endothelial cell budding and migration at the early emergence of lymphatics from venous endothelium.


Asunto(s)
Proteínas de Unión al Calcio/fisiología , Embrión de Mamíferos/citología , Endotelio Linfático/citología , Endotelio Vascular/citología , Linfangiogénesis , Proteínas Supresoras de Tumor/fisiología , Factor C de Crecimiento Endotelial Vascular/fisiología , Receptor 3 de Factores de Crecimiento Endotelial Vascular/fisiología , Venas/citología , Animales , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/ultraestructura , Endotelio Linfático/metabolismo , Endotelio Linfático/ultraestructura , Endotelio Vascular/metabolismo , Endotelio Vascular/ultraestructura , Ratones , Ratones Noqueados , Venas/metabolismo , Venas/ultraestructura
15.
PLoS Biol ; 7(1): e25, 2009 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-19175293

RESUMEN

Neuropilin 1 (Nrp1) is a coreceptor for vascular endothelial growth factor A165 (VEGF-A165, VEGF-A164 in mice) and semaphorin 3A (SEMA3A). Nevertheless, Nrp1 null embryos display vascular defects that differ from those of mice lacking either VEGF-A164 or Sema3A proteins. Furthermore, it has been recently reported that Nrp1 is required for endothelial cell (EC) response to both VEGF-A165 and VEGF-A121 isoforms, the latter being incapable of binding Nrp1 on the EC surface. Taken together, these data suggest that the vascular phenotype caused by the loss of Nrp1 could be due to a VEGF-A164/SEMA3A-independent function of Nrp1 in ECs, such as adhesion to the extracellular matrix. By using RNA interference and rescue with wild-type and mutant constructs, we show here that Nrp1 through its cytoplasmic SEA motif and independently of VEGF-A165 and SEMA3A specifically promotes alpha5beta1-integrin-mediated EC adhesion to fibronectin that is crucial for vascular development. We provide evidence that Nrp1, while not directly mediating cell spreading on fibronectin, interacts with alpha5beta1 at adhesion sites. Binding of the homomultimeric endocytic adaptor GAIP interacting protein C terminus, member 1 (GIPC1), to the SEA motif of Nrp1 selectively stimulates the internalization of active alpha5beta1 in Rab5-positive early endosomes. Accordingly, GIPC1, which also interacts with alpha5beta1, and the associated motor myosin VI (Myo6) support active alpha5beta1 endocytosis and EC adhesion to fibronectin. In conclusion, we propose that Nrp1, in addition to and independently of its role as coreceptor for VEGF-A165 and SEMA3A, stimulates through its cytoplasmic domain the spreading of ECs on fibronectin by increasing the Rab5/GIPC1/Myo6-dependent internalization of active alpha5beta1. Nrp1 modulation of alpha5beta1 integrin function can play a causal role in the generation of angiogenesis defects observed in Nrp1 null mice.


Asunto(s)
Proteínas Portadoras/metabolismo , Endotelio Vascular/metabolismo , Integrina alfa5beta1/metabolismo , Neuropéptidos/metabolismo , Neuropilina-1/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Portadoras/genética , Adhesión Celular , Endotelio Vascular/citología , Fibronectinas/genética , Fibronectinas/metabolismo , Humanos , Integrina alfa5beta1/genética , Ratones , Ratones Noqueados , Neovascularización Fisiológica , Neuropéptidos/genética , Neuropilina-1/antagonistas & inhibidores , Neuropilina-1/genética , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Arterias Umbilicales/citología , Arterias Umbilicales/metabolismo
16.
FASEB J ; 22(8): 2702-14, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18443366

RESUMEN

The events occurring during tumor formation and progression display similarities to some of the steps in embryonic morphogenesis. The family of AP-2 proteins consists of five different transcription factors (alpha, beta, gamma, delta, and epsilon) that play relevant roles in embryonic development, as demonstrated by the phenotypes of the corresponding knockout mice. Here, we show that AP-2alpha and AP-2gamma proteins play an essential role in tumorigenesis. Down-modulation of AP-2 expression in tumor cells by RNA interference (RNAi) led to enhanced tumor growth and reduced chemotherapy-induced cell death, as well as migration and invasion. Most of these biological modulations were rescued by AP-2 overexpression. We observed that increased xenotransplant growth was mostly due to highly enhanced proliferation of the tumor cells together with reduced innate immune cell recruitment. Moreover, we showed that migration impairment was mediated, at least in part, by secreted factors. To identify the genetic programs involved in tumorigenesis, we performed whole genome microarray analysis of AP-2alpha knockdown cells and observed that AP-2alpha regulates specific genes involved in cell cycle, cell death, adhesion, and migration. In particular, we showed that ESDN, EREG, and CXCL2 play a major role in AP-2 controlled migration, as ablation of any of these genes severely altered migration.


Asunto(s)
Neoplasias/genética , Factor de Transcripción AP-2/genética , Animales , Secuencia de Bases , Neoplasias de la Mama/etiología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Muerte Celular , División Celular , Línea Celular Tumoral , Movimiento Celular , Quimiocina CXCL2/genética , Factor de Crecimiento Epidérmico/genética , Epirregulina , Femenino , Células HeLa , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Desnudos , Invasividad Neoplásica , Trasplante de Neoplasias , Neoplasias/etiología , Neoplasias/metabolismo , Neoplasias/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Interferencia de ARN , ARN Interferente Pequeño/genética , Factor de Transcripción AP-2/antagonistas & inhibidores , Factor de Transcripción AP-2/metabolismo , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...