Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Biol Chem ; 291(46): 24017-24028, 2016 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-27650497

RESUMEN

Activation of the blood vessel endothelium is a critical step during inflammation. Endothelial cells stimulated by pro-inflammatory cytokines play an essential part in the adhesion and extravasation of circulating leukocytes into inflamed tissues. The endothelial egfl7 gene (VE-statin) represses endothelial cell activation in tumors, and prior observations suggested that it could also participate in the regulation of endothelial cell activation during inflammation. We show here that Egfl7 expression is strongly repressed in mouse lung endothelial cells during LPS- and TNFα-induced inflammation in vivo LPS have a limited effect on Egfl7 expression by endothelial cells in vitro, whereas the pro-inflammatory cytokine TNFα strongly represses Egfl7 expression in endothelial cells. TNFα regulates the egfl7 gene promoter through regions located between -7585 and -5550 bp ahead of the main transcription start site and via an NF-κB-dependent mechanism. Conversely, Egfl7 regulates the response of endothelial cells to TNFα by restraining the induced expression of intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and E-selectin, resulting in a decreased adhesion of leukocytes onto endothelial cells stimulated by TNFα. Egfl7 regulates the expression of these adhesion molecules through the NF-κB and MEK/Erk pathways, in particular by preventing the proteasome-mediated degradation of IkBα both in non-activated endothelial cells and during activation. Egfl7 is thus an endogenous and constitutive repressor of blood vessel endothelial cell activation in normal and inflammatory conditions and participates in a loop of regulation of activation of these cells by pro-inflammatory cytokines.


Asunto(s)
Factores de Crecimiento Endotelial/biosíntesis , Regulación de la Expresión Génica , Sistema de Señalización de MAP Quinasas , Elementos de Respuesta , Animales , Proteínas de Unión al Calcio , Familia de Proteínas EGF , Factores de Crecimiento Endotelial/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Células Jurkat , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
2.
J Biol Chem ; 287(8): 5366-78, 2012 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-22184117

RESUMEN

The tumor suppressor gene hypermethylated in cancer 1 (HIC1), which encodes a transcriptional repressor, is epigenetically silenced in many human tumors. Here, we show that ectopic expression of HIC1 in the highly malignant MDA-MB-231 breast cancer cell line severely impairs cell proliferation, migration, and invasion in vitro. In parallel, infection of breast cancer cell lines with a retrovirus expressing HIC1 also induces decreased mRNA and protein expression of the tyrosine kinase receptor EphA2. Moreover, chromatin immunoprecipitation (ChIP) and sequential ChIP experiments demonstrate that endogenous HIC1 proteins are bound, together with the MTA1 corepressor, to the EphA2 promoter in WI38 cells. Taken together, our results identify EphA2 as a new direct target gene of HIC1. Finally, we observe that inactivation of endogenous HIC1 through RNA interference in normal breast epithelial cells results in the up-regulation of EphA2 and is correlated with increased cellular migration. To conclude, our results involve the tumor suppressor HIC1 in the transcriptional regulation of the tyrosine kinase receptor EphA2, whose ligand ephrin-A1 is also a HIC1 target gene. Thus, loss of the regulation of this Eph pathway through HIC1 epigenetic silencing could be an important mechanism in the pathogenesis of epithelial cancers.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/metabolismo , Receptor EphA2/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Progresión de la Enfermedad , Regulación hacia Abajo/genética , Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Células HEK293 , Histona Desacetilasas/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/deficiencia , Factores de Transcripción de Tipo Kruppel/genética , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , Regiones Promotoras Genéticas/genética , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/metabolismo , Transactivadores
3.
Biochem Biophys Res Commun ; 440(3): 424-30, 2013 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-24076391

RESUMEN

The tumor suppressor gene HIC1 (Hypermethylated In Cancer 1) is located in 17p13.3 a region frequently hypermethylated or deleted in tumors and in a contiguous-gene syndrome, the Miller-Dieker syndrome which includes classical lissencephaly (smooth brain) and severe developmental defects. HIC1 encodes a transcriptional repressor involved in the regulation of growth control, DNA damage response and cell migration properties. We previously demonstrated that the membrane-associated G-protein-coupled receptors CXCR7, ADRB2 and the tyrosine kinase receptor EphA2 are direct target genes of HIC1. Here we show that ectopic expression of HIC1 in U2OS and MDA-MB-231 cell lines decreases expression of the ApoER2 and VLDLR genes, encoding two canonical tyrosine kinase receptors for Reelin. Conversely, knock-down of endogenous HIC1 in BJ-Tert normal human fibroblasts through RNA interference results in the up-regulation of these two Reelin receptors. Finally, through chromatin immunoprecipitation (ChIP) in BJ-Tert fibroblasts, we demonstrate that HIC1 is a direct transcriptional repressor of ApoER2 and VLDLR. These data provide evidence that HIC1 is a new regulator of the Reelin pathway which is essential for the proper migration of neuronal precursors during the normal development of the cerebral cortex, of Purkinje cells in the cerebellum and of mammary epithelial cells. Deregulation of this pathway through HIC1 inactivation or deletion may contribute to its role in tumor promotion. Moreover, HIC1, through the direct transcriptional repression of ATOH1 and the Reelin receptors ApoER2 and VLDLR, could play an essential role in normal cerebellar development.


Asunto(s)
Movimiento Celular/genética , Regulación de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Relacionadas con Receptor de LDL/genética , Células de Purkinje/fisiología , Receptores de Superficie Celular/genética , Receptores de LDL/genética , Secuencia de Bases , Línea Celular Tumoral , Cerebelo/citología , Cerebelo/crecimiento & desarrollo , Metilación de ADN , Humanos , Células de Purkinje/metabolismo , Proteína Reelina
4.
Stem Cells ; 30(5): 845-53, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22331796

RESUMEN

Presence in glioblastomas of cancer cells with normal neural stem cell (NSC) properties, tumor initiating capacity, and resistance to current therapies suggests that glioblastoma stem-like cells (GSCs) play central roles in glioblastoma development. We cultured human GSCs endowed with all features of tumor stem cells, including tumor initiation after xenograft and radio-chemoresistance. We established proteomes from four GSC cultures and their corresponding whole tumor tissues (TTs) and from human NSCs. Two-dimensional difference gel electrophoresis and tandem mass spectrometry revealed a twofold increase of hepatoma-derived growth factor (HDGF) in GSCs as compared to TTs and NSCs. Western blot analysis confirmed HDGF overexpression in GSCs as well as its presence in GSC-conditioned medium, while, in contrast, no HDGF was detected in NSC secretome. At the functional level, GSC-conditioned medium induced migration of human cerebral endothelial cells that can be blocked by anti-HDGF antibodies. In vivo, GSC-conditioned medium induced neoangiogenesis, whereas HDGF-targeting siRNAs abrogated this effect. Altogether, our results identify a novel candidate, by which GSCs can support neoangiogenesis, a high-grade glioma hallmark. Our strategy illustrates the usefulness of comparative proteomic analysis to decipher molecular pathways, which underlie GSC properties.


Asunto(s)
Inductores de la Angiogénesis/metabolismo , Glioblastoma/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neovascularización Patológica/metabolismo , Células-Madre Neurales/metabolismo , Proteómica , Adulto , Animales , Movimiento Celular , Medios de Cultivo Condicionados , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Glioblastoma/patología , Humanos , Masculino , Ratones , Trasplante de Neoplasias , Neovascularización Patológica/patología , Células-Madre Neurales/patología , Trasplante Heterólogo , Células Tumorales Cultivadas
5.
Biochem Biophys Res Commun ; 421(2): 384-8, 2012 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-22510409

RESUMEN

HIC1 (Hypermethylated in Cancer 1) is a tumor suppressor gene frequently epigenetically silenced in human cancers. HIC1 encodes a transcriptional repressor involved in the regulation of growth control, cell survival and DNA damage response. The deacetylase SIRT1 regulates the repressive capacity of HIC1 in several fashions. First SIRT1 interacts with the BTB/POZ domain of HIC1 to form a transcriptional repression complex that prevents the transcription of SIRT1 itself. SIRT1 is also responsible of the deacetylation of the lysine 314 of HIC1 that allows its subsequent SUMOylation which in turn favors its interaction with the NuRD complex. To better understand the interplay between HIC1 and SIRT1, we performed co-immunoprecipitation experiments to define the domains essential for the HIC1/SIRT1 interaction. We demonstrated that the isolated four last zinc fingers of HIC1 were capable to interact with SIRT1 and that the amino-acids 610-677 of SIRT1 encompassing the ESA region of the deacetylase were crucial for the HIC1/SIRT1 interaction and HIC1 deacetylation. Finally we demonstrated that this interaction mainly depends on CKII-mediated phosphorylation of SIRT1 serine 659/661 which occurs upon DNA damage. Therefore, our results demonstrate that the activating acetylation to SUMOylation switch of HIC1 is favored by genotoxic stresses to regulate the DNA damage response.


Asunto(s)
Daño del ADN , Factores de Transcripción de Tipo Kruppel/metabolismo , Sirtuina 1/metabolismo , Acetilación , Células HEK293 , Humanos , Inmunoprecipitación , Factores de Transcripción de Tipo Kruppel/genética , Dominios y Motivos de Interacción de Proteínas , Serina/genética , Serina/metabolismo , Sirtuina 1/genética , Sumoilación , Dedos de Zinc
6.
Nat Struct Mol Biol ; 14(2): 147-54, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17220898

RESUMEN

Telomeres can fold into t-loops that may result from the invasion of the 3' overhang into duplex DNA. Their formation is facilitated in vitro by the telomeric protein TRF2, but very little is known regarding the mechanisms involved. Here we reveal that TRF2 generates positive supercoiling and condenses DNA. Using a variety of TRF2 mutants, we demonstrate a strong correlation between this topological activity and the ability to stimulate strand invasion. We also report that these properties require the combination of the TRF-homology (TRFH) domain of TRF2 with either its N- or C-terminal DNA-binding domains. We propose that TRF2 complexes, by constraining DNA around themselves in a right-handed conformation, can induce untwisting of the neighboring DNA, thereby favoring strand invasion. Implications of this topological model in t-loop formation and telomere homeostasis are discussed.


Asunto(s)
ADN/química , Telómero/química , Proteína 2 de Unión a Repeticiones Teloméricas/química , ADN Superhelicoidal/química , Humanos , Microscopía de Fuerza Atómica , Conformación de Ácido Nucleico , Proteína 1 de Unión a Repeticiones Teloméricas/química
7.
Oncol Rep ; 47(1)2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34738625

RESUMEN

The tumor blood vessel endothelium forms a barrier that must be crossed by circulating immune cells in order for them to reach and kill cancer cells. Epidermal growth factor­like domain 7 (Egfl7) represses this immune infiltration by lowering the expression levels of leukocyte adhesion receptors on the surface of endothelial cells. However, the protein domains involved in these properties are not completely understood. Egfl7 is structurally composed of the predicted EMI­, EGF­ and C­terminal domains. The present study aimed to investigate the roles of these different domains in tumor development by designing retroviruses coding for deletion mutants and then infecting 4T1 breast cancer cell populations, which consequently overexpressed the variants. By performing in vitro soft­agar assays, it was found that Egfl7 and its deletion variants did not affect cell proliferation or anchorage­independent growth. When 4T1 cells expressing either the wild­type Egfl7 protein or Egfl7 domain variants were implanted in mice, Egfl7 expression markedly promoted tumor development and deletion of the EGF repeats decreased the tumor growth rate. By contrast, deleting any other domain displayed no significant effect on tumor development. The overexpression of Egfl7 also decreased T cell and natural killer cell infiltration in tumors, as determined by immunofluorescence staining of tumor sections, whereas deletion of the EGF repeats inhibited this effect. Reverse transcription­quantitative PCR analysis of the mechanisms involved revealed that deleting the EGF repeats partially restored the expression levels of vascular cell adhesion molecule 1 and E­selectin, which were suppressed by overexpression of Egfl7 in endothelial cells in vitro. This resulted in a higher number of lymphocytes bound to HUVEC expressing Egfl7­ΔEGF compared with HUVEC expressing wild­type Egfl7, as assessed by fluorescent­THP­1 adhesion assays onto endothelial cells. Overall, the present study demonstrated that the EGF repeats may participate in the protumoral and anti­inflammatory effects of Egfl7.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Familia de Proteínas EGF/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Sistema Inmunológico/metabolismo , Escape del Tumor/genética , Animales , Proteínas de Unión al Calcio/genética , Proliferación Celular/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo , Familia de Proteínas EGF/genética , Femenino , Ratones , Ratones Endogámicos BALB C
8.
J Biol Chem ; 284(31): 20927-35, 2009 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-19525223

RESUMEN

The tumor suppressor gene HIC1 (Hypermethylated in Cancer 1) that is epigenetically silenced in many human tumors and is essential for mammalian development encodes a sequence-specific transcriptional repressor. The few genes that have been reported to be directly regulated by HIC1 include ATOH1, FGFBP1, SIRT1, and E2F1. HIC1 is thus involved in the complex regulatory loops modulating p53-dependent and E2F1-dependent cell survival and stress responses. We performed genome-wide expression profiling analyses to identify new HIC1 target genes, using HIC1-deficient U2OS human osteosarcoma cells infected with adenoviruses expressing either HIC1 or GFP as a negative control. These studies identified several putative direct target genes, including CXCR7, a G-protein-coupled receptor recently identified as a scavenger receptor for the chemokine SDF-1/CXCL12. CXCR7 is highly expressed in human breast, lung, and prostate cancers. Using quantitative reverse transcription-PCR analyses, we demonstrated that CXCR7 was repressed in U2OS cells overexpressing HIC1. Inversely, inactivation of endogenous HIC1 by RNA interference in normal human WI38 fibroblasts results in up-regulation of CXCR7 and SIRT1. In silico analyses followed by deletion studies and luciferase reporter assays identified a functional and phylogenetically conserved HIC1-responsive element in the human CXCR7 promoter. Moreover, chromatin immunoprecipitation (ChIP) and ChIP upon ChIP experiments demonstrated that endogenous HIC1 proteins are bound together with the C-terminal binding protein corepressor to the CXCR7 and SIRT1 promoters in WI38 cells. Taken together, our results implicate the tumor suppressor HIC1 in the transcriptional regulation of the chemokine receptor CXCR7, a key player in the promotion of tumorigenesis in a wide variety of cell types.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/metabolismo , Receptores CXCR/genética , Adenoviridae/genética , Oxidorreductasas de Alcohol/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Proliferación Celular , Inmunoprecipitación de Cromatina , Secuencia Conservada , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/genética , Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Genes Relacionados con las Neoplasias , Vectores Genéticos/genética , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteosarcoma/genética , Osteosarcoma/patología , Filogenia , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores CXCR/metabolismo , Sirtuina 1 , Sirtuinas/genética , Sirtuinas/metabolismo
9.
Mol Cell Biol ; 27(7): 2661-75, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17283066

RESUMEN

Tumor suppressor HIC1 (hypermethylated in cancer 1) is a gene that is essential for mammalian development, epigenetically silenced in many human tumors, and involved in a complex pathway regulating P53 tumor suppression activity. HIC1 encodes a sequence-specific transcriptional repressor containing five Krüppel-like C(2)H(2) zinc fingers and an N-terminal BTB/POZ repression domain. Here, we show that endogenous HIC1 is SUMOylated in vivo on a phylogenetically conserved lysine, K314, located in the central region which is a second repression domain. K314R mutation does not influence HIC1 subnuclear localization but significantly reduces its transcriptional repression potential, as does the mutation of the other conserved residue in the psiKXE consensus, E316A, or the overexpression of the deSUMOylase SSP3/SENP2. Furthermore, HIC1 is acetylated in vitro by P300/CBP. Strikingly, the K314R mutant is less acetylated than wild-type HIC1, suggesting that this lysine is a target for both SUMOylation and acetylation. We further show that HIC1 transcriptional repression activity is positively controlled by two types of deacetylases, SIRT1 and HDAC4, which increase the deacetylation and SUMOylation, respectively, of K314. Knockdown of endogenous SIRT1 by the transfection of short interfering RNA causes a significant loss of HIC1 SUMOylation. Thus, this dual-deacetylase complex induces either a phosphorylation-dependent acetylation-SUMOylation switch through a psiKXEXXSP motif, as previously shown for MEF2, or a phosphorylation-independent switch through a psiKXEP motif, as shown here for HIC1, since P317A mutation severely impairs HIC1 acetylation. Finally, our results demonstrate that HIC1 is a target of the class III deacetylase SIRT1 and identify a new posttranslational modification step in the P53-HIC1-SIRT1 regulatory loop.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteína SUMO-1/metabolismo , Sirtuinas/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Acetilación , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Línea Celular , Núcleo Celular/metabolismo , Secuencia Conservada , Proteínas de Unión al ADN/genética , Histona Desacetilasas/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel , Lisina/metabolismo , Datos de Secuencia Molecular , Mutación , Fosforilación , Filogenia , ARN Interferente Pequeño/genética , Sirtuina 1 , Sirtuinas/genética , Factores de Transcripción/genética , Transcripción Genética
10.
Mol Cell Biol ; 22(13): 4890-901, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12052894

RESUMEN

HIC1 (hypermethylated in cancer) and its close relative HRG22 (HIC1-related gene on chromosome 22) encode transcriptional repressors with five C(2)H(2) zinc fingers and an N-terminal BTB/POZ autonomous transcriptional repression domain that is unable to recruit histone deacetylases (HDACs). Alignment of the HIC1 and HRG22 proteins from various species highlighted a perfectly conserved GLDLSKK/R motif highly related to the consensus CtBP interaction motif (PXDLSXK/R), except for the replacement of the virtually invariant proline by a glycine. HIC1 strongly interacts with mCtBP1 both in vivo and in vitro through this conserved GLDLSKK motif, thus extending the CtBP consensus binding site. The BTB/POZ domain does not interact with mCtBP1, but the dimerization of HIC1 through this domain is required for the interaction with mCtBP1. When tethered to DNA by fusion with the Gal4 DNA-binding domain, the HIC1 central region represses transcription through interactions with CtBP in a trichostatin A-sensitive manner. In conclusion, our results demonstrate that HIC1 mediates transcriptional repression by both HDAC-independent and HDAC-dependent mechanisms and show that CtBP is a HIC1 corepressor that is recruited via a variant binding site.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Fosfoproteínas/metabolismo , Factores de Transcripción/metabolismo , Oxidorreductasas de Alcohol , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células COS/metabolismo , Estructuras del Núcleo Celular/metabolismo , Células Cultivadas , Secuencia Conservada , Cricetinae , Proteínas de Unión al ADN/genética , Dimerización , Evolución Molecular , Genes Supresores de Tumor , Histona Desacetilasas/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel , Fosfoproteínas/genética , Conejos , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción/genética , Técnicas del Sistema de Dos Híbridos
11.
Nat Commun ; 7: 10399, 2016 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-26822533

RESUMEN

The main characteristic of senescence is its stability which relies on the persistence of DNA damage. We show that unlike fibroblasts, senescent epithelial cells do not activate an ATM-or ATR-dependent DNA damage response (DDR), but accumulate oxidative-stress-induced DNA single-strand breaks (SSBs). These breaks remain unrepaired because of a decrease in PARP1 expression and activity. This leads to the formation of abnormally large and persistent XRCC1 foci that engage a signalling cascade involving the p38MAPK and leading to p16 upregulation and cell cycle arrest. Importantly, the default in SSB repair also leads to the emergence of post-senescent transformed and mutated precancerous cells. In human-aged skin, XRCC1 foci accumulate in the epidermal cells in correlation with a decline of PARP1, whereas DDR foci accumulate mainly in dermal fibroblasts. These findings point SSBs as a DNA damage encountered by epithelial cells with aging which could fuel the very first steps of carcinogenesis.


Asunto(s)
Senescencia Celular , Roturas del ADN de Cadena Simple , Células Epiteliales/citología , Neoplasias/genética , Daño del ADN , Reparación del ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/metabolismo , Humanos , Neoplasias/metabolismo , Neoplasias/fisiopatología , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X
12.
Oncogene ; 23(22): 4023-31, 2004 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-15007385

RESUMEN

The BTB/POZ transcriptional repressor HIC1 (Hypermethylated in Cancer 1) is a tumor suppressor gene located at chromosome 17p13.3, a region frequently hypermethylated or deleted in human tumors and in a contiguous-gene syndrome, the Miller-Dieker syndrome. The human and murine HIC1 genes are composed of two alternative 5' exons, 1a and 1b fused to a large second coding exon 2. Exon 1a is a noncoding exon associated with a major G-C-rich promoter whereas exon 1b is a downstream coding exon associated with a minor TATA box promoter. By human-mouse genome comparison, we have identified a short upstream conserved sequence containing G-C boxes which were shown to be functional. Transcripts initiating from this new promoter were detected in various human and mouse tissues and contained a long 5'-UTR sequence, called 1c which encompass the G-C-rich promoter associated with exon 1a and uses the same splice donor site. RT-PCR analyses of two primary breast epithelial cell lines identified two other 5'-UTRs generated by alternative splicing within exon 1c. Our results thus highlight the existence of an unexpected complex transcriptional regulation of HIC1.


Asunto(s)
Secuencia Conservada , Genes Supresores de Tumor , Regiones Promotoras Genéticas , Factores de Transcripción/genética , Animales , Secuencia de Bases , Femenino , Humanos , Factores de Transcripción de Tipo Kruppel , Ratones , Datos de Secuencia Molecular , Ovario/metabolismo , Conejos , Ratas
13.
Biochim Biophys Acta ; 1678(1): 57-66, 2004 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-15093138

RESUMEN

Hypermethylated in Cancer 1 (HIC1) is a human tumor suppressor gene located at chromosome 17p13.3 which is frequently hypermethylated and transcriptionally silent in many types of tumors. In addition, its location in the Miller-Dieker syndrome's (MDS) deletion region, its embryonic expression pattern in mice and the phenotype of the HIC1-deficient mice have provided strong evidence for its implication in this contiguous-gene syndrome. HIC1 encodes a five C2H2-type zinc finger transcriptional repressor belonging to the BTB/POZ family. We have isolated the true zebrafish orthologue of human HIC1 since it has a comparable intron-exon structure and since its predicted gene product, ZfHIC1 displays much higher sequence similarities in its overall sequence (737 residues) with human HIC1 (714 residues) than the 454 residues encoded by the only zebrafish HIC1 sequence (AF111712) described so far, which has been renamed ZfHIC1alpha. Notably, the C-terminal end and one zinc finger in the DNA-binding domain are missing in ZfHIC1alpha. As a consequence, ZfHIC1 proteins bind the human HIC1 consensus DNA-binding sequence in vitro, whereas ZfHIC1alpha cannot. Analyses of the expression pattern of ZfHIC1 and of its paralogue ZfHRG22 (HIC1 related gene on chromosome 22) show that they share expression domains with their respective orthologous vertebrate genes. ZfHRG22 is prominently expressed in the brain and in neural tissues. Interestingly, the predominant expression of ZfHIC1 in the mesenchyme of the head, around the nose and the eye and in the branchial arches is possibly consistent with some of the abnormalities seen in the HIC1-deficient mice and provides another clue for the implication of HIC1 in MDS.


Asunto(s)
Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Clonación Molecular , Proteínas de Unión al ADN , Genes Supresores de Tumor , Factores de Transcripción de Tipo Kruppel , Mesodermo/metabolismo , Datos de Secuencia Molecular , Alineación de Secuencia , Factores de Transcripción/genética , Proteínas Supresoras de Tumor , Pez Cebra/embriología , Proteínas de Pez Cebra/genética
14.
Int J Oncol ; 42(4): 1367-75, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23404186

RESUMEN

Egfl7 (VE-statin) is specifically expressed by endothelial cells of normal tissues but its expression is deregulated in human cancers. Analysis of expression of Egfl7 protein and transcripts in 211 human breast cancer samples shows that Egfl7 is strongly expressed by breast tumor cells. Egfl7 expression is significantly higher in invasive ductal than in invasive lobular carcinoma. Expression of Egfl7 transcripts is also higher in lower SBR grade lesions and in lesions which are not associated with lymph node invasion. Within the invasive ductal carcinoma sub-population, expression of Egfl7 transcripts is correlated with the SBR score and with the ER+ status. High transcript and Egfl7 protein levels significantly correlate with the absence of axillary lymph node invasion. In lymph nodes, the levels of Egfl7 are correlated with the histological type of the primary lesions; they are higher in ductal than in lobular carcinoma. Egfl7 expression is thus associated with better prognosis factors and with the absence of lymph node invasion in human breast cancer lesions.


Asunto(s)
Neoplasias de la Mama Masculina/metabolismo , Carcinoma Ductal de Mama/metabolismo , Carcinoma Intraductal no Infiltrante/metabolismo , Carcinoma Lobular/metabolismo , Factores de Crecimiento Endotelial/metabolismo , Células 3T3 , Adulto , Anciano , Anciano de 80 o más Años , Animales , Axila/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Neoplasias de la Mama Masculina/mortalidad , Neoplasias de la Mama Masculina/patología , Proteínas de Unión al Calcio , Carcinoma Ductal de Mama/mortalidad , Carcinoma Ductal de Mama/secundario , Carcinoma Intraductal no Infiltrante/mortalidad , Carcinoma Intraductal no Infiltrante/secundario , Carcinoma Lobular/mortalidad , Carcinoma Lobular/secundario , Células Cultivadas , Supervivencia sin Enfermedad , Familia de Proteínas EGF , Factores de Crecimiento Endotelial/genética , Femenino , Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Estimación de Kaplan-Meier , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Metástasis Linfática , Masculino , Ratones , Persona de Mediana Edad , Invasividad Neoplásica , Especificidad de Órganos
15.
Nat Cell Biol ; 15(7): 818-28, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23792691

RESUMEN

Dysfunctional telomeres suppress tumour progression by activating cell-intrinsic programs that lead to growth arrest. Increased levels of TRF2, a key factor in telomere protection, are observed in various human malignancies and contribute to oncogenesis. We demonstrate here that a high level of TRF2 in tumour cells decreased their ability to recruit and activate natural killer (NK) cells. Conversely, a reduced dose of TRF2 enabled tumour cells to be more easily eliminated by NK cells. Consistent with these results, a progressive upregulation of TRF2 correlated with decreased NK cell density during the early development of human colon cancer. By screening for TRF2-bound genes, we found that HS3ST4--a gene encoding for the heparan sulphate (glucosamine) 3-O-sulphotransferase 4--was regulated by TRF2 and inhibited the recruitment of NK cells in an epistatic relationship with TRF2. Overall, these results reveal a TRF2-dependent pathway that is tumour-cell extrinsic and regulates NK cell immunity.


Asunto(s)
Neoplasias de la Mama/prevención & control , Neoplasias del Colon/prevención & control , Células Asesinas Naturales/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental/prevención & control , Sulfotransferasas/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Animales , Apoptosis , Western Blotting , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Adhesión Celular , Proliferación Celular , Neoplasias del Colon/inmunología , Neoplasias del Colon/metabolismo , Cartilla de ADN/química , Receptor con Dominio Discoidina 1 , Femenino , Citometría de Flujo , Células HeLa , Humanos , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/patología , Linfocitos Infiltrantes de Tumor/patología , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Ratones , Ratones Desnudos , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sulfotransferasas/genética , Proteína 2 de Unión a Repeticiones Teloméricas/antagonistas & inhibidores , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Células Tumorales Cultivadas
16.
Oncoimmunology ; 1(3): 375-376, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22737620

RESUMEN

Egfl7 is an endothelial-specific gene which expression is deregulated in human cancers. We showed that Egfl7 promotes tumor escape from immunity by downregulating the expression of leukocyte adhesion molecules in endothelial cells, thus repressing immune cell extravasation into tumors.

17.
PLoS One ; 7(4): e34386, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22536324

RESUMEN

Mammalian telomeres stabilize chromosome ends as a result of their assembly into a peculiar form of chromatin comprising a complex of non-histone proteins named shelterin. TRF2, one of the shelterin components, binds to the duplex part of telomeric DNA and is essential to fold the telomeric chromatin into a protective cap. Although most of the human telomeric DNA is organized into tightly spaced nucleosomes, their role in telomere protection and how they interplay with telomere-specific factors in telomere organization is still unclear. In this study we investigated whether TRF2 can regulate nucleosome assembly at telomeres.By means of chromatin immunoprecipitation (ChIP) and Micrococcal Nuclease (MNase) mapping assay, we found that the density of telomeric nucleosomes in human cells was inversely proportional to the dosage of TRF2 at telomeres. This effect was not observed in the G1 phase of the cell cycle but appeared coincident of late or post-replicative events. Moreover, we showed that TRF2 overexpression altered nucleosome spacing at telomeres increasing internucleosomal distance. By means of an in vitro nucleosome assembly system containing purified histones and remodeling factors, we reproduced the short nucleosome spacing found in telomeric chromatin. Importantly, when in vitro assembly was performed in the presence of purified TRF2, nucleosome spacing on a telomeric DNA template increased, in agreement with in vivo MNase mapping.Our results demonstrate that TRF2 negatively regulates the number of nucleosomes at human telomeres by a cell cycle-dependent mechanism that alters internucleosomal distance. These findings raise the intriguing possibility that telomere protection is mediated, at least in part, by the TRF2-dependent regulation of nucleosome organization.


Asunto(s)
Puntos de Control del Ciclo Celular , Nucleosomas/metabolismo , Telómero/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/fisiología , Línea Celular Tumoral , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Inmunoprecipitación de Cromatina , Expresión Génica , Humanos , Unión Proteica , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo
18.
Cancer Res ; 71(23): 7176-86, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22037871

RESUMEN

Downregulating the leukocyte adhesion molecules expressed by endothelial cells that line tumor blood vessels can limit the entry of immune effector cells into the tumor mass, thereby contributing to tumoral immune escape. Egfl7 (also known as VE-statin) is a secreted protein specifically expressed by endothelial cells in normal tissues and by cancer cells in various human tumors. High levels of Egfl7 correlate with higher tumor grade and poorer prognosis. Here we show that expression of Egfl7 in breast and lung carcinoma cells accelerates tumor growth and metastasis in immunocompetent mice but not in immunodeficient mice. Tumors expressing Egfl7 were infiltrated relatively poorly by immune cells and were characterized by reduced levels of immunostimulatory cytokines [IFN-γ, interleukin-12 (IL-12)] and fewer endothelial adhesion molecules [intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1)]. In vitro studies revealed that Egfl7 inhibited the expression of leukocyte adhesion molecules by endothelial cells, preventing lymphocyte adhesion. In contrast, Egfl7 did not exert any effects on immune cell activation. Human breast cancer lesions expressing high levels of Egfl7 also expressed less ICAM-1 and VCAM-1 in their blood vessels, also indicating an inverse correlation between expression levels of Egfl7 and IFN-γ. Thus, Egfl7 expression in tumors promotes tumor progression by reducing the expression of endothelial molecules that mediate immune cell infiltration. Our findings highlight a novel mechanism through which tumors escape immune control.


Asunto(s)
Células Endoteliales/inmunología , Factores de Crecimiento Endotelial/inmunología , Proteínas/inmunología , Escape del Tumor/inmunología , Animales , Proteínas de Unión al Calcio , Adhesión Celular/genética , Adhesión Celular/inmunología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/inmunología , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Cultivadas , Progresión de la Enfermedad , Familia de Proteínas EGF , Células Endoteliales/patología , Factores de Crecimiento Endotelial/genética , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/inmunología , Interferón gamma/genética , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-12/genética , Interleucina-12/inmunología , Interleucina-12/metabolismo , Células Jurkat , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones SCID , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/inmunología , Metástasis de la Neoplasia/patología , Proteínas/genética , Escape del Tumor/genética , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/inmunología
19.
Mol Cell Biol ; 30(16): 4045-59, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20547755

RESUMEN

The tumor suppressor gene HIC1 encodes a transcriptional repressor involved in regulatory loops modulating P53-dependent and E2F1-dependent cell survival, growth control, and stress responses. Despite its importance, few HIC1 corepressors and target genes have been characterized thus far. Using a yeast two-hybrid approach, we identify MTA1, a subunit of the NuRD complex, as a new HIC1 corepressor. This interaction is regulated by two competitive posttranslational modifications of HIC1 at lysine 314, promotion by SUMOylation, and inhibition by acetylation. Consistent with the role of HIC1 in growth control, we demonstrate that HIC1/MTA1 complexes bind on two new target genes, Cyclin D1 and p57KIP2 in quiescent but not in growing WI38 cells. In addition, HIC1/MTA1 and HIC1/CtBP complexes differentially bind on two mutually exclusive HIC1 binding sites (HiRE) on the SIRT1 promoter. SIRT1 transcriptional activation induced by short-term serum starvation coincides with loss of occupancy of the distal sites by HIC1/MTA1 and HIC1/CtBP. Upon longer starvation, both complexes are found but on a newly identified proximal HiRE that is evolutionarily conserved and specifically enriched with repressive histone marks. Our results decipher a mechanistic link between two competitive posttranslational modifications of HIC1 and corepressor recruitment to specific genes, leading to growth control.


Asunto(s)
Oxidorreductasas de Alcohol/metabolismo , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Acetilación , Oxidorreductasas de Alcohol/genética , Animales , Secuencia de Bases , Sitios de Unión/genética , Línea Celular , Proliferación Celular , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , ADN/genética , ADN/metabolismo , Proteínas de Unión al ADN/genética , Genes bcl-1 , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Técnicas In Vitro , Interfase , Factores de Transcripción de Tipo Kruppel/química , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Ratones , Modelos Biológicos , Células 3T3 NIH , Regiones Promotoras Genéticas , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Sirtuina 1/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Transactivadores , Activación Transcripcional , Técnicas del Sistema de Dos Híbridos
20.
J Biol Chem ; 280(33): 29625-36, 2005 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-15985428

RESUMEN

The imprinted H19 gene has riboregulatory functions. We show here that H19 transcription is up-regulated during the S-phase of growth-stimulated cells and that the H19 promoter is activated by E2F1 in breast cancer cells. H19 repression by pRb and E2F6 confirms the E2F1-dependent control of the H19 promoter. Consistently, we demonstrate by chromatin immunoprecipitation assays that endogenous E2F1 is recruited to the H19 promoter in vivo. The functionality of E2F promoter sites was further confirmed by gel shift and mutagenesis experiments, revealing that these sites are required for binding and promoter response to E2F1 exogenous expression and serum stimulation. Furthermore, we show that H19 overexpression confers a growth advantage on breast cancer cells released from growth arrest as well as in asynchronously growing cells. The H19 knockdown by small interfering RNA duplexes impedes S-phase entry in both wild-type and stably H19-transfected cells. Based on these findings, we conclude that the H19 RNA is actively linked to E2F1 to promote cell cycle progression of breast cancer cells. This clearly supports the H19 oncogenic function in breast tumor genesis.


Asunto(s)
Neoplasias de la Mama/patología , Proteínas de Ciclo Celular/fisiología , Proteínas de Unión al ADN/fisiología , ARN no Traducido/fisiología , Factores de Transcripción/fisiología , Secuencia de Bases , Neoplasias de la Mama/etiología , Línea Celular Tumoral , Proliferación Celular , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Factor de Transcripción E2F6 , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , ARN Largo no Codificante , Fase S
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA