Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Oncogenesis ; 12(1): 55, 2023 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-37973791

RESUMEN

Among follicular-derived thyroid cancers (TC), those with aggressive behavior and resistance to current treatments display poor prognosis. NF-κB signaling pathways are involved in tumor progression of various cancers. Here, we finely characterize the NF-κB pathways and their involvement in TC. By using immunoblot and gel shift assays, we demonstrated that both classical and alternative NF-κB pathways are activated in ten TC-derived cell lines, leading to activated RelA/p50 and RelB/p50 NF-κB dimers. By analyzing the RNAseq data of the large papillary thyroid carcinoma (PTC) cohort from The Cancer Genome Atlas (TCGA) project, we identified a tumor progression-related NF-κB signature in BRAFV600E mutated-PTCs. That corroborated with the role of RelA and RelB in cell migration and invasion processes that we demonstrated specifically in BRAFV600E mutated-cell lines, together with their role in the control of expression of genes implicated in invasiveness (MMP1, PLAU, LCN2 and LGALS3). We also identified NF-κB-inducing kinase (NIK) as a novel actor of the constitutive activation of the NF-κB pathways in TC-derived cell lines. Finally, its implication in invasiveness and its overexpression in PTC samples make NIK a potential therapeutic target for advanced TC treatment.

3.
Oncotarget ; 10(23): 2320-2334, 2019 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-31040922

RESUMEN

In thyroid cancers, MET receptor overexpression has been associated with higher risk of metastatic progression. In this study, it was shown that the anaplastic thyroid cancer (ATC)-derived TTA1 cell line overexpressed MET. By using FISH and relative quantification by qPCR, it was demonstrated that this overexpression resulted from a MET amplification with more than 20 copies. As expected, MET overexpression led to its constitutive activation and upregulated signaling towards the MAPK, PI3K/AKT, STAT3 and NF-κB pathways. Since the usual feature of MET-amplified cell lines is the "MET addiction" for their cell proliferation, the effect of the highly selective ATP competitive MET inhibitor PHA665752 was analyzed. While PHA665752 strongly inhibited the MAPK pathway, it did not reduce cell proliferation in TTA1 cells (IC50 = 4100 nM). This resistance to PHA665752 of the TTA1 cell line was demonstrated to be related to EGFR-MET functional cross-talk and PI3K/AKT and NF-κB signaling. Nevertheless, PHA665752 suppressed the anchorage-independent growth capacity of the TTA1 cell line and reduced cell migration and invasion in a transwell assay. The role of activated MET in these neoplastic properties of the TTA1 cells was also proved with si-MET-RNA targeting. Thus, this work highlights the TTA1 cell line as the first model of MET amplification in an ATC cell line, which leads to MET constitutive activation and underlies its neoplastic properties. Besides being a useful model for MET inhibitors screening, the TTA1 cell line also supports the argument for searching for MET amplification in ATC, as it could have therapeutic implications.

4.
PLoS One ; 12(9): e0184861, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28910386

RESUMEN

BACKGROUND: Molecular alterations of the MAPK pathway are frequently observed in papillary thyroid carcinomas (PTCs). It leads to a constitutive activation of the signalling pathway through an increase in MEK and ERK phosphorylation. ERK is negatively feedback-regulated by Dual Specificity Phosphatases (DUSPs), especially two ERK-specific DUSPs, DUSP5 (nuclear) and DUSP6 (cytosolic). These negative MAPK regulators may play a role in thyroid carcinogenesis. METHODS: MAPK pathway activation was analyzed in 11 human thyroid cancer cell lines. Both phosphatases were studied in three PCCL3 rat thyroid cell lines that express doxycycline inducible PTC oncogenes (RET/PTC3, H-RASV12 or BRAFV600E). Expression levels of DUSP5 and DUSP6 were quantified in 39 human PTCs. The functional role of DUSP5 and DUSP6 was investigated through their silencing in two human BRAFV600E carcinoma cell lines. RESULTS: BRAFV600E human thyroid cancer cell lines expressed higher phospho-MEK levels but not higher phospho-ERK levels. DUSP5 and DUSP6 are specifically induced by the MEK-ERK pathway in the three PTC oncogenes inducible thyroid cell lines. This negative feedback loop explains the tight regulation of p-ERK levels. DUSP5 and DUSP6 mRNA are overexpressed in human PTCs, especially in BRAFV600E mutated PTCs. DUSP5 and/or DUSP6 siRNA inactivation did not affect proliferation in two BRAFV600E mutated cell lines, which may be explained by a compensatory increase in other phosphatases. In the light of this, we observed a marked DUSP6 upregulation upon DUSP5 inactivation. Despite this, DUSP5 and DUSP6 positively control cell migration and invasion. CONCLUSIONS: Our results are in favor of a stronger activation of the MAPK pathway in BRAFV600E PTCs. DUSP5 and DUSP6 have pro-tumorigenic properties in two BRAFV600E PTC cell line models.


Asunto(s)
Carcinoma/genética , Fosfatasa 6 de Especificidad Dual/genética , Fosfatasas de Especificidad Dual/genética , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias de la Tiroides/genética , Animales , Carcinoma/metabolismo , Carcinoma Papilar , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Fosfatasa 6 de Especificidad Dual/metabolismo , Fosfatasas de Especificidad Dual/metabolismo , Retroalimentación Fisiológica , Regulación Neoplásica de la Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas , Invasividad Neoplásica , Fosforilación , Ratas , Cáncer Papilar Tiroideo , Neoplasias de la Tiroides/metabolismo , Regulación hacia Arriba
5.
Sci Rep ; 6: 25765, 2016 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-27166427

RESUMEN

Acute or chronic metabolic complications such as diabetic ketoacidosis are often associated with extracellular acidification and pancreatic ß-cell dysfunction. However, the mechanisms by which human ß-cells sense and respond to acidic pH remain elusive. In this study, using the recently developed human ß-cell line EndoC-ßH2, we demonstrate that ß-cells respond to extracellular acidification through GPR68, which is the predominant proton sensing receptor of human ß-cells. Using gain- and loss-of-function studies, we provide evidence that the ß-cell enriched transcription factor RFX6 is a major regulator of GPR68. Further, we show that acidic pH stimulates the production and secretion of the chemokine IL-8 by ß-cells through NF-кB activation. Blocking of GPR68 or NF-кB activity severely attenuated acidification induced IL-8 production. Thus, we provide mechanistic insights into GPR68 mediated ß-cell response to acidic microenvironment, which could be a new target to protect ß-cell against acidosis induced inflammation.


Asunto(s)
Ácidos/metabolismo , Espacio Extracelular/química , Células Secretoras de Insulina/metabolismo , Interleucina-8/biosíntesis , Receptores Acoplados a Proteínas G/metabolismo , Línea Celular , AMP Cíclico/biosíntesis , Humanos , Concentración de Iones de Hidrógeno , Mediadores de Inflamación/metabolismo , Fosfatos de Inositol/metabolismo , FN-kappa B/metabolismo , Neutrófilos/metabolismo , Protones , Factores de Transcripción del Factor Regulador X/metabolismo
6.
PLoS One ; 10(12): e0145484, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26691724

RESUMEN

Serum stimulation of mammalian cells induces, via the MAPK pathway, the nuclear protein DUSP5 (dual-specificity phosphatase 5), which specifically interacts with and inactivates the ERK1/2 MAP kinases. However, molecular mechanisms underlying DUSP5 induction are not well known. Here, we found that the DUSP5 mRNA induction depends on a transcriptional regulation by the MAPK pathway, without any modification of the mRNA stability. Two contiguous CArG boxes that bind serum response factor (SRF) were found in a 1 Kb promoter region, as well as several E twenty-six transcription factor family binding sites (EBS). These sites potentially bind Elk-1, a transcription factor activated by ERK1/2. Using wild type or mutated DUSP5 promoter reporters, we demonstrated that SRF plays a crucial role in serum induction of DUSP5 promoter activity, the proximal CArG box being important for SRF binding in vitro and in living cells. Moreover, in vitro and in vivo binding data of Elk-1 to the same promoter region further demonstrate a role for Elk-1 in the transcriptional regulation of DUSP5. SRF and Elk-1 form a ternary complex (Elk-1-SRF-DNA) on DUSP5 promoter, consequently providing a link to an important negative feedback tightly regulating phosphorylated ERK levels.


Asunto(s)
Fosfatasas de Especificidad Dual/genética , Fosfatasas de Especificidad Dual/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Factor de Respuesta Sérica/metabolismo , Proteína Elk-1 con Dominio ets/metabolismo , Animales , Sitios de Unión , Regulación Enzimológica de la Expresión Génica , Semivida , Ratones , Mutación , Células 3T3 NIH , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Elemento de Respuesta al Suero , Factor de Respuesta Sérica/genética , Transducción de Señal , Proteína Elk-1 con Dominio ets/genética
7.
Oncotarget ; 6(4): 2524-37, 2015 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-25426554

RESUMEN

Vav family members function as remarkable scaffold proteins that exhibit both GDP/GTP exchange activity for Rho/Rac GTPases and numerous protein-protein interactions via three adaptor Src-homology domains. The exchange activity is under the unique regulation by phosphorylation of tyrosine residues hidden by intra-molecular interactions. Deletion of the autoinhibitory N-terminal region results in an oncogenic protein, onco-Vav, leading to a potent activation of Rac GTPases whereas the proto-oncogene barely leads to transformation. Substitution of conserved residues of the SH2-SH3 adaptor region in onco-Vav reverses oncogenicity. While a unique substitution D797N did not affect transformation induced by onco-Vav, we demonstrate that this single substitution leads to transformation in the Vav1 proto-oncogene highlighting the pivotal role of the adaptor region. Moreover, we identified the cell junction protein ß-catenin as a new Vav1 interacting partner. We show that the oncogenicity of activated Vav1 proto-oncogene is associated with a non-degradative phosphorylation of ß-catenin at residues important for its functions and its redistribution along the cell membrane in fibroblasts. In addition, a similar interaction is evidenced in epithelial lung cancer cells expressing ectopically Vav1. In these cells, Vav1 is also involved in the modulation of ß-catenin phosphorylation. Altogether, our data highlight that only a single mutation in the proto-oncogene Vav1 enhances tumorigenicity.


Asunto(s)
Transformación Celular Neoplásica/genética , Mutación , Proteínas Proto-Oncogénicas c-vav/genética , Dominios Homologos src/genética , Animales , Antracenos/farmacología , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Western Blotting , Línea Celular Tumoral , Transformación Celular Neoplásica/metabolismo , Fibroblastos/metabolismo , Células HEK293 , Humanos , MAP Quinasa Quinasa 4/antagonistas & inhibidores , MAP Quinasa Quinasa 4/metabolismo , Ratones , Ratones Desnudos , Células 3T3 NIH , Fosforilación , Unión Proteica , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-vav/metabolismo , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , beta Catenina/genética , beta Catenina/metabolismo
8.
PLoS One ; 8(3): e59127, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23555623

RESUMEN

The NF-κB family of transcription factors has emerged as a key player in the pathogenesis of multiple myeloma (MM). NF-κB is activated by at least two major signaling pathways. The classical pathway results in the activation of mainly RelA containing dimers, whereas the alternative pathway leads to the activation of RelB/p52 and RelB/p50 heterodimers. Activating mutations in regulators of the alternative pathway have been identified in 17% of MM patients. However, the status of RelB activation per se and its role in the regulation of cell survival in MM has not been investigated. Here, we reveal that 40% of newly diagnosed MM patients have a constitutive RelB DNA-binding activity in CD138(+) tumor cells, and we show an association with increased expression of a subset of anti-apoptotic NF-κB target genes, such as cIAP2. Furthermore, we demonstrate that RelB exerts a crucial anti-apoptotic activity in MM cells. Our findings indicate that RelB activation is key for promoting MM cell survival through the upregulation of anti-apoptotic proteins. Altogether, our study provides the framework for the development of new molecules targeting RelB in the treatment of MM.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteínas Inhibidoras de la Apoptosis/genética , Mieloma Múltiple/genética , Factor de Transcripción ReIB/genética , Transcripción Genética , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis , Proteína 3 que Contiene Repeticiones IAP de Baculovirus , Supervivencia Celular , ADN/metabolismo , Femenino , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Masculino , Persona de Mediana Edad , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Cultivo Primario de Células , Transducción de Señal , Sindecano-1/genética , Sindecano-1/metabolismo , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIB/metabolismo , Ubiquitina-Proteína Ligasas
9.
Int J Pept ; 2012: 745027, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22187571

RESUMEN

Recent studies have highlighted the AT1 receptor as a potential therapeutic target in breast cancer, while the role of the AT2 subtype in this disease has remained largely neglected. The present study describes the generation and characterization of a new cellular model of human invasive breast cancer cells (D3H2LN-AT2) stably expressing high levels of Flag-tagged human AT2 receptor (Flag-hAT2). These cells exhibit high-affinity binding sites for AngII, and total binding can be displaced by the AT2-selective antagonist PD123319 but not by the AT1-selective antagonist losartan. Of interest, high levels of expression of luciferase and green fluorescent protein make these cells suitable for bioluminescence and fluorescence studies in vitro and in vivo. We provide here a novel tool to investigate the AT2 receptor functions in breast cancer cells, independently of AT1 receptor activation.

10.
PLoS One ; 6(7): e21589, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21754991

RESUMEN

BACKGROUND: Endothelial dysfunction has been implicated in the pathogenesis of diverse pathologies ranging from vascular and immune diseases to cancer. TNF-α is one of the mediators of endothelial dysfunction through the activation of transcription factors, including NF-κB. While HUVEC (macrovascular cells) have been largely used in the past, here, we documented an NF-κB gene signature in TNFα-stimulated microvascular endothelial cells HMEC often used in tumor angiogenesis studies. METHODOLOGY/PRINCIPAL FINDINGS: We measured mRNA expression of 55 NF-κB related genes using quantitative RT-PCR in HUVEC and HMEC. Our study identified twenty genes markedly up-regulated in response to TNFα, including adhesion molecules, cytokines, chemokines, and apoptosis regulators, some of them being identified as TNF-α-inducible genes for the first time in endothelial cells (two apoptosis regulators, TNFAIP3 and TNFRSF10B/Trail R2 (DR5), the chemokines GM-CSF/CSF2 and MCF/CSF1, and CD40 and TNF-α itself, as well as NF-κB components (RELB, NFKB1 or 50/p105 and NFKB2 or p52/p100). For eight genes, the fold induction was much higher in HMEC, as compared to HUVEC. Most importantly, our study described for the first time a connection between NF-κB activation and the induction of most, if not all, of these genes in HMEC as evaluated by pharmacological inhibition and RelA expression knock-down by RNA interference. Moreover, since TNF-α is highly expressed in tumors, we further applied the NF-κB gene signature documented in TNFα-stimulated endothelial cells to human breast tumors. We found a significant positive correlation between TNF and the majority (85 %) of the identified endothelial TNF-induced genes in a well-defined series of 96 (48 ERα positive and 48 ERα negative) breast tumors. CONCLUSION/SIGNIFICANCE: Taken together these data suggest the potential use of this NF-κB gene signature in analyzing the role of TNF-α in the endothelial dysfunction, as well as in breast tumors independently of the presence of ERα.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , FN-kappa B/genética , Factor de Necrosis Tumoral alfa/farmacología , Anciano , Biopsia , Western Blotting , Células Endoteliales/efectos de los fármacos , Receptor alfa de Estrógeno/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Cinética , Microvasos/patología , Persona de Mediana Edad , FN-kappa B/metabolismo , Pronóstico , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Factores de Tiempo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
11.
PLoS One ; 3(7): e2555, 2008 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-18596915

RESUMEN

BACKGROUND: The Rel/NF-kappaB transcription factors are often activated in solid or hematological malignancies. In most cases, NF-kappaB activation is found in malignant cells and results from activation of the canonical NF-kappaB pathway, leading to RelA and/or c-Rel activation. Recently, NF-kappaB activity in inflammatory cells infiltrating solid tumors has been shown to contribute to solid tumor initiation and progression. Noncanonical NF-kappaB activation, which leads to RelB activation, has also been reported in breast carcinoma, prostate cancer, and lymphoid leukemia. METHODOLOGY/PRINCIPAL FINDINGS: Here we report a novel role for RelB in stromal cells that promote T-cell leukemogenesis. RelB deficiency delayed leukemia onset in the TEL-JAK2 transgenic mouse model of human T acute lymphoblastic leukemia. Bone marrow chimeric mouse experiments showed that RelB is not required in the hematopoietic compartment. In contrast, RelB plays a role in radio-resistant stromal cells to accelerate leukemia onset and increase disease severity. CONCLUSIONS/SIGNIFICANCE: The present results are the first to uncover a role for RelB in the crosstalk between non-hematopoietic stromal cells and leukemic cells. Thus, besides its previously reported role intrinsic to specific cancer cells, the noncanonical NF-kappaB pathway may also play a pro-oncogenic role in cancer microenvironmental cells.


Asunto(s)
Leucemia-Linfoma de Células T del Adulto/metabolismo , Células del Estroma/metabolismo , Factor de Transcripción ReIB/metabolismo , Animales , Citometría de Flujo , Leucemia-Linfoma de Células T del Adulto/genética , Leucemia-Linfoma de Células T del Adulto/patología , Ratones , Ratones Transgénicos , Subunidad p50 de NF-kappa B/genética , Linfocitos T/metabolismo , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIB/genética
12.
Blood ; 109(9): 3972-81, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17192390

RESUMEN

The TEL-JAK2 gene fusion, which has been identified in human leukemia, encodes a chimeric protein endowed with constitutive tyrosine kinase activity. TEL-JAK2 transgenic expression in the mouse lymphoid lineage results in fatal and rapid T-cell leukemia/lymphoma. In the present report we show that T-cell leukemic cells from EmuSRalpha-TEL-JAK2 transgenic mice present an aberrant CD8(+) differentiation phenotype, as determined by the expression of stage-specific cell surface markers and lineage-specific genes. TEL-JAK2 transforms immature CD4(-)CD8(-) double-negative thymocytes, as demonstrated by the development of T-cell leukemia with full penetrance in a Rag2-deficient genetic background. This disease is similar to the bona fide TEL-JAK2 disease as assessed by phenotypic and gene profiling analyses. Pre-TCR signaling synergizes with TEL-JAK2 to transform immature thymocytes and initiate leukemogenesis as shown by (1) the delayed leukemia onset in Rag2-, CD3epsilon- and pTalpha-deficient mice, (2) the occurrence of recurrent chromosomal alterations in pre-TCR-deficient leukemia, and (3) the correction of delayed leukemia onset in Rag2-deficient TEL-JAK2 mice by an H-Y TCRalphabeta transgene that mimics pre-TCR signaling. Although not affecting leukemia incidence and mouse survival, TCRalphabeta expression was shown to facilitate leukemic cell expansion in secondary lymphoid organs.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Transformación Celular Neoplásica/metabolismo , Leucemia de Células T/metabolismo , Proteínas de Fusión Oncogénica/biosíntesis , Receptores de Antígenos de Linfocitos T alfa-beta/biosíntesis , Animales , Linfocitos T CD8-positivos/patología , Diferenciación Celular/genética , Proliferación Celular , Transformación Celular Neoplásica/genética , Regulación Leucémica de la Expresión Génica/genética , Humanos , Leucemia de Células T/genética , Leucemia de Células T/patología , Ratones , Ratones Noqueados , Proteínas de Fusión Oncogénica/genética , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Transducción de Señal/genética , Timo/metabolismo , Timo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...