Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 80(2): 47, 2023 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-36658409

RESUMEN

Histone H3 trimethylation on lysine 9 (H3K9me3) is a defining feature of mammalian pericentromeres, loss of which results in genome instability. Here we show that CDYL2 is recruited to pericentromeres in an H3K9me3-dependent manner and is required for faithful mitosis and genome stability. CDYL2 RNAi in MCF-7 breast cancer cells and Hela cervical cancer cells inhibited their growth, induced apoptosis, and provoked both nuclear and mitotic aberrations. Mass spectrometry analysis of CDYL2-interacting proteins identified the neurodevelopmental disease-linked mitotic regulators CHAMP1 and POGZ, which are associated with a central non-conserved region of CDYL2. RNAi rescue assays identified both the CDYL2 chromodomain and the CHAMP1-POGZ interacting region as required and, together, sufficient for CDYL2 regulation of mitosis and genome stability. CDYL2 RNAi caused loss of CHAMP1 localization at pericentromeres. We propose that CDYL2 functions as an adaptor protein that connects pericentromeric H3K9me3 with CHAMP1 and POGZ to ensure mitotic fidelity and genome stability.


Asunto(s)
Proteínas Cromosómicas no Histona , Proteínas Co-Represoras , Histonas , Mitosis , Humanos , Proteínas Cromosómicas no Histona/metabolismo , Inestabilidad Genómica , Histonas/genética , Histonas/metabolismo , Lisina/metabolismo , Mitosis/genética , Fosfoproteínas/metabolismo , Interferencia de ARN , Células MCF-7 , Proteínas Co-Represoras/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo
2.
EMBO Mol Med ; 13(4): e12878, 2021 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-33719214

RESUMEN

The navigation cue netrin-1 is well-documented for its key role in cancer development and represents a promising therapeutic target currently under clinical investigation. Phase 1 and 2 clinical trials are ongoing with NP137, a humanized monoclonal antibody against netrin-1. Interestingly, the epitope recognized by NP137 in netrin-1 shares 90% homology with its counterpart in netrin-3, the closest member to netrin-1 in humans, for which little is known in the field of cancer. Here, we unveiled that netrin-3 appears to be expressed specifically in human neuroblastoma (NB) and small cell lung cancer (SCLC), two subtypes of neuroectodermal/neuroendocrine lineages. Netrin-3 and netrin-1 expression are mutually exclusive, and the former is driven by the MYCN oncogene in NB, and the ASCL-1 or NeuroD1 transcription factors in SCLC. Netrin-3 expression is correlated with disease stage, aggressiveness, and overall survival in NB. Mechanistically, we confirmed the high affinity of netrin-3 for netrin-1 receptors and we demonstrated that netrin-3 genetic silencing or interference using NP137, delayed tumor engraftment, and reduced tumor growth in animal models. Altogether, these data support the targeting of netrin-3 in NB and SCLC.


Asunto(s)
Neoplasias Pulmonares , Neuroblastoma , Carcinoma Pulmonar de Células Pequeñas , Animales , Humanos , Netrina-1 , Netrinas
3.
iScience ; 23(6): 101141, 2020 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-32450513

RESUMEN

Epigenetic deregulation of gene transcription is central to cancer cell plasticity and malignant progression but remains poorly understood. We found that the uncharacterized epigenetic factor chromodomain on Y-like 2 (CDYL2) is commonly over-expressed in breast cancer, and that high CDYL2 levels correlate with poor prognosis. Supporting a functional role for CDYL2 in malignancy, it positively regulated breast cancer cell migration, invasion, stem-like phenotypes, and epithelial-to-mesenchymal transition. CDYL2 regulation of these plasticity-associated processes depended on signaling via p65/NF-κB and STAT3. This, in turn, was downstream of CDYL2 regulation of MIR124 gene transcription. CDYL2 co-immunoprecipitated with G9a/EHMT2 and GLP/EHMT1 and regulated the chromatin enrichment of G9a and EZH2 at MIR124 genes. We propose that CDYL2 contributes to poor prognosis in breast cancer by recruiting G9a and EZH2 to epigenetically repress MIR124 genes, thereby promoting NF-κB and STAT3 signaling, as well as downstream cancer cell plasticity and malignant progression.

4.
J Virol ; 89(2): 1461-7, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25378492

RESUMEN

EVER1 and EVER2 are mutated in epidermodysplasia verruciformis patients, who are susceptible to human betapapillomavirus (HPV) infection. It is unknown whether their products control the infection of other viruses. Here, we show that the expression of both genes in B cells is activated immediately after Epstein-Barr virus (EBV) infection, whereas at later stages, it is strongly repressed via activation of the NF-κB signaling pathway by latent membrane protein 1 (LMP1). Ectopic expression of EVER1 impairs the ability of EBV to infect B cells.


Asunto(s)
Epidermodisplasia Verruciforme/patología , Regulación de la Expresión Génica , Herpesvirus Humano 4/fisiología , Interacciones Huésped-Patógeno , Proteínas de la Membrana/biosíntesis , Proteínas de la Matriz Viral/metabolismo , Linfocitos B/virología , Humanos , Proteínas de la Membrana/genética
5.
Nucleic Acids Res ; 42(15): 9700-16, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25092922

RESUMEN

The Epstein-Barr virus (EBV) nuclear antigen 3 family of protein is critical for the EBV-induced primary B-cell growth transformation process. Using a yeast two-hybrid screen we identified 22 novel cellular partners of the EBNA3s. Most importantly, among the newly identified partners, five are known to play direct and important roles in transcriptional regulation. Of these, the Myc-interacting zinc finger protein-1 (MIZ-1) is a transcription factor initially characterized as a binding partner of MYC. MIZ-1 activates the transcription of a number of target genes including the cell cycle inhibitor CDKN2B. Focusing on the EBNA3A/MIZ-1 interaction we demonstrate that binding occurs in EBV-infected cells expressing both proteins at endogenous physiological levels and that in the presence of EBNA3A, a significant fraction of MIZ-1 translocates from the cytoplasm to the nucleus. Moreover, we show that a trimeric complex composed of a MIZ-1 recognition DNA element, MIZ-1 and EBNA3A can be formed, and that interaction of MIZ-1 with nucleophosmin (NPM), one of its coactivator, is prevented by EBNA3A. Finally, we show that, in the presence of EBNA3A, expression of the MIZ-1 target gene, CDKN2B, is downregulated and repressive H3K27 marks are established on its promoter region suggesting that EBNA3A directly counteracts the growth inhibitory action of MIZ-1.


Asunto(s)
Inhibidor p15 de las Quinasas Dependientes de la Ciclina/genética , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Regulación de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Represoras/metabolismo , Transcripción Genética , Oxidorreductasas de Alcohol/metabolismo , Núcleo Celular/metabolismo , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/biosíntesis , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Antígenos Nucleares del Virus de Epstein-Barr/química , Células HEK293 , Células HeLa , Histonas/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/química , Proteínas Nucleares/metabolismo , Nucleofosmina , Regiones Promotoras Genéticas , Dominios y Motivos de Interacción de Proteínas , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Represoras/química
6.
PLoS Pathog ; 10(5): e1004125, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24809689

RESUMEN

The DOK1 tumor suppressor gene encodes an adapter protein that acts as a negative regulator of several signaling pathways. We have previously reported that DOK1 expression is up-regulated upon cellular stress, via the transcription factor E2F1, and down-regulated in a variety of human malignancies due to aberrant hypermethylation of its promoter. Here we show that Epstein Barr virus (EBV) infection of primary human B-cells leads to the down-regulation of DOK1 gene expression via the viral oncoprotein LMP1. LMP1 alone induces recruitment to the DOK1 promoter of at least two independent inhibitory complexes, one containing E2F1/pRB/DNMT1 and another containing at least EZH2. These events result in tri-methylation of histone H3 at lysine 27 (H3K27me3) of the DOK1 promoter and gene expression silencing. We also present evidence that the presence of additional EBV proteins leads to further repression of DOK1 expression with an additional mechanism. Indeed, EBV infection of B-cells induces DNA methylation at the DOK1 promoter region including the E2F1 responsive elements that, in turn, lose the ability to interact with E2F complexes. Treatment of EBV-infected B-cell-lines with the methyl-transferase inhibitor 5-aza-2'-deoxycytidine rescues DOK1 expression. In summary, our data show the deregulation of DOK1 gene expression by EBV and provide novel insights into the regulation of the DOK1 tumor suppressor in viral-related carcinogenesis.


Asunto(s)
Proteínas de Unión al ADN/genética , Infecciones por Virus de Epstein-Barr/genética , Herpesvirus Humano 4/fisiología , Fosfoproteínas/genética , Proteínas de Unión al ARN/genética , Linfocitos B/metabolismo , Linfocitos B/virología , Transformación Celular Viral/genética , Células Cultivadas , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/genética , Infecciones por Virus de Epstein-Barr/inmunología , Regulación de la Expresión Génica , Silenciador del Gen , Genes Supresores de Tumor , Humanos , Fosfoproteínas/metabolismo , Cultivo Primario de Células , Proteínas de Unión al ARN/metabolismo , Proteínas de la Matriz Viral/fisiología
7.
Mol Cell Biol ; 32(23): 4877-90, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23028047

RESUMEN

The expression of the tumor suppressor DOK1 is repressed in a variety of human tumors as a result of hypermethylation of its promoter region. However, the molecular mechanisms by which DOK1 expression is regulated have been poorly investigated. Here, we show that the expression of DOK1 is regulated mainly by the transcription factor E2F1. We identified three putative E2F1 response elements (EREs) in the DOK1 promoter region. E2F1 had a relatively higher binding affinity for the ERE located between bp -498 and -486 compared with the other two EREs. E2F1 gene silencing strongly inhibited DOK1 expression. E2F1-driven DOK1 transcription occurred in the presence of cellular stresses, such as accumulation of DNA damage induced by etoposide. DOK1 silencing promoted cell proliferation and protected against etoposide-induced apoptosis, indicating that DOK1 acts as a key mediator of cellular stress-induced cell death. Most importantly, we observed that DNA methylation of the DOK1 core promoter region found in head and neck cancer cell lines hampered the recruitment of E2F1 to the DOK1 promoter and compromised DOK1 expression. In summary, our data show that E2F1 is a key factor in DOK1 expression and provide novel insights into the regulation of these events in cancer cells.


Asunto(s)
Proteínas de Unión al ADN/genética , Factor de Transcripción E2F1/metabolismo , Fosfoproteínas/genética , Proteínas de Unión al ARN/genética , Elementos de Respuesta , Activación Transcripcional , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Azacitidina/análogos & derivados , Azacitidina/farmacología , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Decitabina , Factor de Transcripción E2F1/genética , Etopósido/farmacología , Silenciador del Gen , Células HEK293 , Humanos , Metiltransferasas/antagonistas & inhibidores , Activación Transcripcional/efectos de los fármacos
8.
Int J Cancer ; 130(11): 2484-94, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21796618

RESUMEN

The DOK1 gene is a putative tumour suppressor gene located on the human chromosome 2p13 which is frequently rearranged in leukaemia and other human tumours. We previously reported that the DOK1 gene can be mutated and its expression down-regulated in human malignancies. However, the mechanism underlying DOK1 silencing remains largely unknown. We show here that unscheduled silencing of DOK1 expression through aberrant hypermethylation is a frequent event in a variety of human malignancies. DOK1 was found to be silenced in nine head and neck cancer (HNC) cell lines studied and DOK1 CpG hypermethylation correlated with loss of gene expression in these cells. DOK1 expression could be restored via demethylating treatment using 5-aza-2'deoxycytidine. In addition, transduction of cancer cell lines with DOK1 impaired their proliferation, consistent with the critical role of epigenetic silencing of DOK1 in the development and maintenance of malignant cells. We further observed that DOK1 hypermethylation occurs frequently in a variety of primary human neoplasm including solid tumours (93% in HNC, 81% in lung cancer) and haematopoietic malignancy (64% in Burkitt's lymphoma). Control blood samples and exfoliated mouth epithelial cells from healthy individuals showed a low level of DOK1 methylation, suggesting that DOK1 hypermethylation is a tumour specific event. Finally, an inverse correlation was observed between the level of DOK1 gene methylation and its expression in tumour and adjacent non tumour tissues. Thus, hypermethylation of DOK1 is a potentially critical event in human carcinogenesis, and may be a potential cancer biomarker and an attractive target for epigenetic-based therapy.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/genética , Neoplasias de Cabeza y Cuello/genética , Fosfoproteínas/genética , Regiones Promotoras Genéticas , Proteínas de Unión al ARN/genética , Adulto , Anciano , Azacitidina/análogos & derivados , Azacitidina/farmacología , Línea Celular Tumoral , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Proteínas de Unión al ADN/antagonistas & inhibidores , Decitabina , Femenino , Genes Supresores de Tumor , Neoplasias de Cabeza y Cuello/patología , Humanos , Masculino , Persona de Mediana Edad , Fosfoproteínas/antagonistas & inhibidores , Proteínas de Unión al ARN/antagonistas & inhibidores , Factores de Riesgo , Proteínas Supresoras de Tumor/genética
9.
J Virol ; 85(17): 8477-94, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21697493

RESUMEN

We previously reported that the oncoproteins E6 and E7 from cutaneous human papillomavirus type 38 (HPV38) can immortalize primary human keratinocytes in vitro and sensitize transgenic mice to develop skin cancer in vivo. Immunofluorescence staining revealed that human keratinocytes immortalized by HPV38 E6 and E7 display fewer actin stress fibers than do control primary keratinocyte cells, raising the possibility of a role of the viral oncoproteins in the remodeling of the actin cytoskeleton. In this study, we show that HPV38 E7 induces actin stress fiber disruption and that this phenomenon correlates with its ability to downregulate Rho activity. The downregulation of Rho activity by HPV38 E7 is mediated through the activation of the CK2-MEK-extracellular signal-regulated kinase (ERK) pathway. In addition, HPV38 E7 is able to induce actin fiber disruption by binding directly to eukaryotic elongation factor 1A (eEF1A) and abolishing its effects on actin fiber formation. Finally, we found that the downregulation of Rho activity by HPV38 E7 through the CK2-MEK-ERK pathway facilitates cell growth proliferation. Taken together, our data support the conclusion that HPV38 E7 promotes keratinocyte proliferation in part by negatively regulating actin cytoskeleton fiber formation through the CK2-MEK-ERK-Rho pathway and by binding to eEF1A and inhibiting its effects on actin cytoskeleton remodeling.


Asunto(s)
Actinas/metabolismo , Quinasa de la Caseína II/metabolismo , Citoesqueleto/metabolismo , Factor 1 Eucariótico de Iniciación/antagonistas & inhibidores , Queratinocitos/virología , Proteínas Oncogénicas Virales/metabolismo , Papillomaviridae/patogenicidad , Línea Celular , Proliferación Celular , Humanos , Unión Proteica
10.
Carcinogenesis ; 32(7): 978-85, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21459755

RESUMEN

Hepatitis B virus (HBV) X protein (HBx) is a key player in HBV-induced hepatocellular carcinoma (HCC). HBx interacts with several cell signaling molecules, leading to activation of various transcription factors including nuclear factor-kappaB (NF-κB). Activated NF-κB signaling is implicated in many human cancers including HCC. Here, we present evidence that the NF-κB signaling activator, tumor necrosis factor (TNF)-α, induces the accumulation of HBx in cells by increasing protein stability due to reduced proteasomal degradation. The effects of TNF-α on HBx protein stability are mediated via activated NF-κB effector kinases IKKα and IKKß and p65. The non-IKK-phosphorylable p65-S534A mutant did not induce HBx protein stability; hence, phosphorylation of p65 by IKK is a key step in TNF-α-induced stabilization of HBx. Phospho-p65 showed higher affinity to HBx compared with the non-phosphorylable p65 mutant, suggesting that the interaction of phospho-p65 with HBx might be important for HBx stabilization. We also show that the increased level of HBx in cells cooperates with TNF-α toward activation of NF-κB and expression of NF-κB-regulated genes, indicating a positive feedback loop between HBx and NF-κB signaling. Overall, our study provides evidence for interplay between HBx and NF-κB signaling, which may account for HBV-mediated liver carcinogenesis.


Asunto(s)
FN-kappa B/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/fisiología , Secuencia de Bases , Línea Celular , Silenciador del Gen , Humanos , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño , Transactivadores , Proteínas Reguladoras y Accesorias Virales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...