Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acids Res ; 48(11): 5891-5906, 2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32421830

RESUMEN

Originally identified as an RNA polymerase II interactor, Che-1/AATF (Che-1) has now been recognized as a multifunctional protein involved in cell-cycle regulation and cancer progression, as well as apoptosis inhibition and response to stress. This protein displays a peculiar nucleolar localization and it has recently been implicated in pre-rRNA processing and ribosome biogenesis. Here, we report the identification of a novel function of Che-1 in the regulation of ribosomal RNA (rRNA) synthesis, in both cancer and normal cells. We demonstrate that Che-1 interacts with RNA polymerase I and nucleolar upstream binding factor (UBF) and promotes RNA polymerase I-dependent transcription. Furthermore, this protein binds to the rRNA gene (rDNA) promoter and modulates its epigenetic state by contrasting the recruitment of HDAC1. Che-1 downregulation affects RNA polymerase I and UBF recruitment on rDNA and leads to reducing rDNA promoter activity and 47S pre-rRNA production. Interestingly, Che-1 depletion induces abnormal nucleolar morphology associated with re-distribution of nucleolar proteins. Finally, we show that upon DNA damage Che-1 re-localizes from rDNA to TP53 gene promoter to induce cell-cycle arrest. This previously uncharacterized function of Che-1 confirms the important role of this protein in the regulation of ribosome biogenesis, cellular proliferation and response to stress.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , ADN Ribosómico/genética , Genes de ARNr/genética , ARN Polimerasa I/metabolismo , Proteínas Represoras/metabolismo , Transcripción Genética , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Puntos de Control del Ciclo Celular , Línea Celular , Nucléolo Celular/metabolismo , Nucléolo Celular/patología , Daño del ADN , ADN Ribosómico/metabolismo , Homeostasis , Humanos , Fosforilación , Proteínas del Complejo de Iniciación de Transcripción Pol1/metabolismo , Regiones Promotoras Genéticas , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Ribosomas/metabolismo
2.
Int J Mol Sci ; 21(8)2020 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-32316313

RESUMEN

Pharmacological treatment of colorectal carcinoma currently proceeds through the administration of a combination of different chemotherapeutic agents. In the case of rectal carcinoma, radiation therapy also represents a therapeutic strategy. In an attempt at translating much-needed new targeted therapy to the clinics, p38 mitogen activated protein kinase (MAPK) inhibitors have been tested in clinical trials involving colorectal carcinoma patients, especially in combination with chemotherapy; however, despite the high expectations raised by a clear involvement of the p38 MAPK pathway in the response to therapeutic treatments, poor results have been obtained so far. In this work, we review recent insights into the exact role of the p38 MAPK pathway in response to currently available therapies for colorectal carcinoma, depicting an intricate scenario in which the p38 MAPK node presents many opportunities, as well as many challenges, for its perspective exploitation for clinical purposes.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/radioterapia , Fluorouracilo/farmacología , Fluorouracilo/uso terapéutico , Humanos , Oxaliplatino/farmacología , Oxaliplatino/uso terapéutico , Isoformas de Proteínas/metabolismo , Transducción de Señal/efectos de los fármacos
3.
EMBO J ; 34(9): 1214-30, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25770584

RESUMEN

Mammalian target of rapamycin (mTOR) is a key protein kinase that regulates cell growth, metabolism, and autophagy to maintain cellular homeostasis. Its activity is inhibited by adverse conditions, including nutrient limitation, hypoxia, and DNA damage. In this study, we demonstrate that Che-1, a RNA polymerase II-binding protein activated by the DNA damage response, inhibits mTOR activity in response to stress conditions. We found that, under stress, Che-1 induces the expression of two important mTOR inhibitors, Redd1 and Deptor, and that this activity is required for sustaining stress-induced autophagy. Strikingly, Che-1 expression correlates with the progression of multiple myeloma and is required for cell growth and survival, a malignancy characterized by high autophagy response.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/fisiología , Mieloma Múltiple/patología , Proteínas Represoras/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Línea Celular Tumoral , Supervivencia Celular , Femenino , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones Desnudos , Mieloma Múltiple/metabolismo , Complejos Multiproteicos/metabolismo , Fosforilación , Proteínas Represoras/genética , Estrés Fisiológico , Serina-Treonina Quinasas TOR/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
Int J Mol Sci ; 20(2)2019 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-30658426

RESUMEN

Breast cancer (BC) is the most common tumor and the second cause for cancer-related death in women worldwide, although combined treatments are well-established interventions. Several effects seem to be responsible for poor outcomes in advanced or triple-negative BC patients. Focusing on the interaction of ionizing radiation with tumor and normal tissues, the role of cytokine modulation as a surrogate of immunomodulation must still be explored. In this work, we carried out an overview of studies published in the last five years involving the cytokine profile in BC patients undergoing radiotherapy. The goal of this review was to evaluate the profile and modulation of major cytokines and interleukins as potential biomarkers of survival, treatment response, and toxicity in BC patient undergoing radiotherapy. Out of 47 retrieved papers selected using PubMed search, 15 fulfilled the inclusion criteria. Different studies reported that the modulation of specific cytokines was time- and treatment-dependent. Radiotherapy (RT) induces the modulation of inflammatory cytokines up to 6 months for most of the analyzed cytokines, which in some cases can persist up to several years post-treatment. The role of specific cytokines as prognostic and predictive of radiotherapy outcome is critically discussed.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/radioterapia , Citocinas/metabolismo , Biomarcadores , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/mortalidad , Terapia Combinada , Femenino , Humanos , Inmunomodulación , Resultado del Tratamiento
5.
J Exp Clin Cancer Res ; 43(1): 234, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39164711

RESUMEN

BACKGROUND: Colorectal cancer (CRC) is the third most common type of cancer and the second leading cause of cancer-related deaths worldwide, with a survival rate near to 10% when diagnosed at an advanced stage. Hence, the identification of new molecular targets to design more selective and efficient therapies is urgently required. The Mitogen activated protein kinase kinase 3 (MKK3) is a dual-specificity threonine/tyrosine protein kinase that, activated in response to cellular stress and inflammatory stimuli, regulates a plethora of biological processes. Previous studies revealed novel MKK3 roles in supporting tumor malignancy, as its depletion induces autophagy and cell death in cancer lines of different tumor types, including CRC. Therefore, MKK3 may represent an interesting new therapeutic target in advanced CRC, however selective MKK3 inhibitors are currently not available. METHODS: The study involved transcriptomic based drug repurposing approach and confirmatory assays with CRC lines, primary colonocytes and a subset of CRC patient-derived organoids (PDO). Investigations in vitro and in vivo were addressed. RESULTS: The repurposing approach identified the multitargeted kinase inhibitor AT9283 as a putative compound with MKK3 depletion-mimicking activities. Indeed, AT9283 drops phospho- and total-MKK3 protein levels in tested CRC models. Likely the MKK3 silencing, AT9283 treatment: i) inhibited cell proliferation promoting autophagy and cell death in tested CRC lines and PDOs; ii) resulted well-tolerated by CCD-18Co colonocytes; iii) reduced cancer cell motility inhibiting CRC cell migration and invasion; iv) inhibited COLO205 xenograft tumor growth. Mechanistically, AT9283 abrogated MKK3 protein levels mainly through the inhibition of aurora kinase A (AURKA), impacting on MKK3/AURKA protein-protein interaction and protein stability therefore uncovering the relevance of MKK3/AURKA crosstalk in sustaining CRC malignancy in vitro and in vivo. CONCLUSION: Overall, we demonstrated that the anti-tumoral effects triggered by AT9283 treatment recapitulated the MKK3 depletion effects in all tested CRC models in vitro and in vivo, suggesting that AT9283 is a repurposed drug. According to its good tolerance when tested with primary colonocytes (CCD-18CO), AT9283 is a promising drug for the development of novel therapeutic strategies to target MKK3 oncogenic functions in late-stage and metastatic CRC patients.


Asunto(s)
Neoplasias Colorrectales , MAP Quinasa Quinasa 3 , Humanos , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/genética , Animales , Ratones , MAP Quinasa Quinasa 3/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Línea Celular Tumoral , Reposicionamiento de Medicamentos , Proliferación Celular/efectos de los fármacos , Autofagia/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico
6.
Int J Cancer ; 132(10): 2237-47, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23124354

RESUMEN

Leydig cell tumors (LCTs) are the most common tumors of the gonadal stroma and represent about 3% of all testicular neoplasms. In most cases, LCTs are benign; however, if the tumor is malignant, no effective treatments are currently available. We have recently reported that farnesoid X receptor (FXR) is expressed in R2C Leydig tumor cells, and it reduces the estrogen-dependent cell proliferation by negatively regulating aromatase expression. Here, we demonstrated that treatment with GW4064, a specific FXR agonist, markedly reduced Leydig tumor growth in vivo by inhibiting proliferation and inducing apoptosis. Indeed, the tumors from GW4064-treated mice exhibited a decrease in the expression of the proliferation marker Ki-67 and aromatase along with an increase in the apoptotic nuclei. FXR activation induced an enhanced poly(ADP-ribose) polymerase cleavage, a marked DNA fragmentation and a strong increase in TUNEL-positive R2C cells also in vitro. Moreover, in both in vivo and in vitro models, FXR ligands upregulated mRNA and protein levels of p53 and of its downstream effector p21(WAF1/Cip1) . Functional experiments showed that FXR ligands upregulated p53 promoter activity and this occurred through an increased binding of FXR/nuclear factor-kB (NF-kB) complex to the NF-kB site located within p53 promoter region as revealed by electrophoretic mobility shift assay and chromatin immunoprecipitation analysis. Taken together, results from our study show, for the first time, that treatment with FXR ligands induces Leydig tumor regression in vivo, suggesting that activation of FXR may represent a promising therapeutic strategy for LCTs.


Asunto(s)
Antineoplásicos/farmacología , Isoxazoles/farmacología , Tumor de Células de Leydig/tratamiento farmacológico , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/metabolismo , Neoplasias Testiculares/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Aromatasa/efectos de los fármacos , Aromatasa/metabolismo , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo , Electroforesis en Gel de Poliacrilamida , Ensayo de Cambio de Movilidad Electroforética , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Immunoblotting , Inmunoprecipitación , Etiquetado Corte-Fin in Situ , Isoxazoles/administración & dosificación , Tumor de Células de Leydig/metabolismo , Masculino , Ratones , Ratones Desnudos , FN-kappa B/metabolismo , Distribución Aleatoria , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Testiculares/metabolismo , Activación Transcripcional , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasas p21 Activadas/metabolismo
7.
Biomed Pharmacother ; 167: 115480, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37713993

RESUMEN

Colorectal cancer (CRC) patients with BRAF mutations develop resistance to BRAF inhibitors at a very early stage. Understanding the molecular mechanisms involved in BRAF inhibitor resistance is critical for the development of novel therapeutic opportunities for this subtype of CRC patients. CRC cells bearing BRAF mutations are mostly sensitive to the abrogation of Mitogen-Activated Protein Kinase Kinase 3 (MKK3), a specific activator of p38MAPKs signaling, suggesting that BRAF alterations might addict CRC cells to the MKK3/p38MAPK signaling. Interestingly, publicly available gene expression profiling data show significantly higher MKK3 transcript levels in CRC lines with acquired resistance to BRAF inhibitors. Herein, we investigated the roles of MKK3 in the response to BRAF targeting (dabrafenib) with COLO205 and HT29 BRAFV600E CRC lines and derived dabrafenib-resistant (DABR) sublines. Dabrafenib treatments reduce MKK3 activation by inducing autophagy in parental but not DABR cells. The MKK3 knockdown induces cell death in DABR cells, whereas ectopic MKK3 expression reduces dabrafenib sensitivity in parental cells. Mechanistically, activated MKK3 interacts and co-localizes with c-Myc oncoprotein (MYC), sustaining MYC protein stability and thus preventing the dabrafenib induced effects in CRC DABR cells both in vitro and in vivo. Overall, we identify a novel molecular mechanism beyond the dabrafenib resistance, shedding light on an uncovered vulnerability for the development of novel therapeutic opportunities in BRAFV600E CRC.


Asunto(s)
Neoplasias Colorrectales , MAP Quinasa Quinasa 3 , Proteínas Proto-Oncogénicas c-myc , Humanos , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Mutación/genética , Oximas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Transducción de Señal , Resistencia a Antineoplásicos
8.
Am J Pathol ; 179(2): 1030-40, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21704006

RESUMEN

Obesity is a major risk factor for the development and progression of breast cancer. Leptin, a cytokine mainly produced by adipocytes, plays a crucial role in mammary carcinogenesis and is elevated in hyperinsulinemia and insulin resistance. The antidiabetic thiazolidinediones inhibit leptin gene expression through ligand activation of the peroxisome proliferator-activated receptor-γ (PPARγ) and exert antiproliferative and apoptotic effects on breast carcinoma. In this study, we investigated the ability of PPARγ ligands to counteract leptin stimulatory effects on breast cancer growth in either in vivo or in vitro models. The results show that activation of PPARγ prevented the development of leptin-induced MCF-7 tumor xenografts and inhibited the increased cell-cell aggregation and proliferation observed on leptin exposure. PPARγ ligands abrogated the leptin-induced up-regulation of leptin gene expression and its receptors in breast cancer. PPARγ-mediated repression of leptin gene involved the recruitment of nuclear receptor corepressor protein and silencing mediator of retinoid and thyroid hormone receptors corepressors on the glucocorticoid responsive element site in the leptin gene expression regulatory region in the presence of glucocorticoid receptor and PPARγ. In addition, PPARγ ligands inhibited leptin signaling mediated by MAPK/STAT3/Akt phosphorylation and counteracted leptin stimulatory effect on estrogen signaling. These findings suggest that PPARγ ligands may have potential therapeutic benefits in the treatment of breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Leptina/metabolismo , PPAR gamma/metabolismo , Animales , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Femenino , Humanos , Técnicas In Vitro , Ligandos , Neoplasias Mamarias Animales/metabolismo , Ratones , Ratones Desnudos , Obesidad/complicaciones , Interferencia de ARN , Receptores de Glucocorticoides/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Factores de Riesgo , Transducción de Señal
9.
Cancers (Basel) ; 14(3)2022 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-35158751

RESUMEN

The role played by MKK3 in human cancer is controversial. MKK3 is an evolutionarily conserved protein kinase that activates in response to a variety of stimuli. Phosphorylates, specifically the p38MAPK family proteins, contribute to the regulation of a plethora of cellular processes such as proliferation, differentiation, apoptosis, invasion, and cell migration. Genes in carcinogenesis are classified as oncogenes and tumor suppressors; however, a clear distinction is not always easily made as it depends on the cell context and tissue specificity. The aim of this study is the examination of the potential contribution of MKK3 in cancer through a systematic analysis of the recent literature. The overall results reveal a complex scenario of MKK3's involvement in cancer. The oncogenic functions of MKK3 were univocally documented in several solid tumors, such as colorectal, prostate cancer, and melanoma, while its tumor-suppressing functions were described in glioblastoma and gastric cancer. Furthermore, a dual role of MKK3 as an oncogene as well as tumor a suppressor has been described in breast, cervical, ovarian, liver, esophageal, and lung cancer. However, overall, more evidence points to its role as an oncogene in these diseases. This review indicates that the oncogenic and tumor-suppressing roles of MKK3 are strictly dependent on the tumor type and further suggests that MKK3 could represent an efficient putative molecular target that requires contextualization within a specific tumor type in order to adequately evaluate its potential effectiveness in designing novel anticancer therapies.

10.
Oncol Rep ; 48(3)2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35801577

RESUMEN

Insights into the molecular and cellular biology of embryonal rhabdomyosarcoma (ERMS), an aggressive paediatric tumour, are required in order to identify new targets for novel treatments that may benefit patients with this disease. The present study examined the functional effects of MKK3 and MKK6, two upstream kinases of p38, and found that the ectopic expression of MKK6 led to rapid p38 activation and the myogenic differentiation of ERMS cells, whereas MKK3 failed to induce differentiation, while maintaining the proliferation state. Myogenin and myosin heavy chain were induced in MKK6­overexpressing ERMS cells and were inhibited by the p38 inhibitor, SB203580. The expression of Myc and ERK­PO4 increased under the effect of SB203580, whereas it decreased in MKK6­overexpressing cells. AKT activation was part of the myogenic program triggered by MKK6 overexpression alone. To the best of our knowledge, the present study demonstrates, for the first time, that the endogenous MKK6 pathway may be recovered by MEK/ERK inhibition (U0126 and trametinib) and that it concomitantly induces the reversal of the oncogenic pattern and the induction of the myogenic differentiation of ERMS cell lines. The effects of MEK/ERK inhibitors markedly increase the potential clinical applications in ERMS, particularly on account of the MEK inhibitor­induced early MKK6/p38 axis activation and of their anti­oncogenic effects. The findings presented herein lend further support to the antitumour effects of MKK6; MKK6 may thus represent a novel target for advanced personalised treatments against ERMS.


Asunto(s)
Rabdomiosarcoma Embrionario , Diferenciación Celular , Línea Celular Tumoral , Niño , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-akt , Rabdomiosarcoma Embrionario/tratamiento farmacológico , Rabdomiosarcoma Embrionario/genética , Rabdomiosarcoma Embrionario/patología , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
J Exp Clin Cancer Res ; 41(1): 148, 2022 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-35440004

RESUMEN

BACKGROUND: Despite the promise of dual BRAF/MEK inhibition as a therapy for BRAF-mutant (BRAF-mut) melanoma, heterogeneous responses have been observed in patients, thus predictors of benefit from therapy are needed. We have previously identified semaphorin 6A (SEMA6A) as a BRAF-mut-associated protein involved in actin cytoskeleton remodeling. The purpose of the present study is to dissect the role of SEMA6A in the biology of BRAF-mut melanoma, and to explore its predictive potential towards dual BRAF/MEK inhibition. METHODS: SEMA6A expression was assessed by immunohistochemistry in melanoma cohort RECI1 (N = 112) and its prognostic potential was investigated in BRAF-mut melanoma patients from DFCI and TCGA datasets (N = 258). The molecular mechanisms regulated by SEMA6A to sustain tumor aggressiveness and targeted therapy resistance were investigated in vitro by using BRAF-mut and BRAF-wt melanoma cell lines, an inducible SEMA6A silencing cell model and a microenvironment-mimicking fibroblasts-coculturing model. Finally, SEMA6A prediction of benefit from dual BRAF/MEK inhibition was investigated in melanoma cohort RECI2 (N = 14). RESULTS: Our results indicate higher protein expression of SEMA6A in BRAF-mut compared with BRAF-wt melanoma patients and show that SEMA6A is a prognostic indicator in BRAF-mut melanoma from TCGA and DFCI patients cohorts. In BRAF-mut melanoma cells, SEMA6A coordinates actin cytoskeleton remodeling by the RhoA-dependent activation of YAP and dual BRAF/MEK inhibition by dabrafenib+trametinib induces SEMA6A/RhoA/YAP axis. In microenvironment-mimicking co-culture condition, fibroblasts confer to melanoma cells a proliferative stimulus and protect them from targeted therapies, whereas SEMA6A depletion rescues the efficacy of dual BRAF/MEK inhibition. Finally, in BRAF-mut melanoma patients treated with dabrafenib+trametinib, high SEMA6A predicts shorter recurrence-free interval. CONCLUSIONS: Overall, our results indicate that SEMA6A contributes to microenvironment-coordinated evasion of melanoma cells from dual BRAF/MEK inhibition and it might be a good candidate predictor of short-term benefit from dual BRAF/MEK inhibition.


Asunto(s)
Melanoma , Semaforinas , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Humanos , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Mutación , Pronóstico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Semaforinas/genética , Semaforinas/metabolismo , Microambiente Tumoral , Proteína de Unión al GTP rhoA/metabolismo
12.
J Biol Chem ; 285(19): 14160-9, 2010 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-20223820

RESUMEN

Mitogen-activated protein kinase kinase 3 (MAP2K3) is a member of the dual specificity kinase group. Growing evidence links MAP2K3 to invasion and tumor progression. Here, we identify MAP2K3 as a transcriptional target of endogenous gain-of-function p53 mutants R273H, R175H, and R280K. We show that MAP2K3 modulation occurred at the mRNA and protein levels and that endogenous mutant p53 proteins are capable of binding to and activate the MAP2K3 promoter. In addition, we found that the studied p53 mutants regulate MAP2K3 gene expression through the involvement of the transcriptional cofactors NF-Y and NF-kappaB. Finally, functional studies showed that endogenous MAP2K3 knockdown inhibits proliferation and survival of human tumor cells, whereas the ectopic expression of MAP2K3 can rescue the proliferative defect induced by mutant p53 knockdown. Taken together, our findings define a novel player through which mutant p53 exerts its gain-of-function activity in cancer cells.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , MAP Quinasa Quinasa 3/genética , Mutación/genética , Neoplasias/genética , Proteína p53 Supresora de Tumor/genética , Western Blotting , Línea Celular Tumoral , Proliferación Celular , Inmunoprecipitación de Cromatina , Citometría de Flujo , Humanos , MAP Quinasa Quinasa 3/antagonistas & inhibidores , MAP Quinasa Quinasa 3/metabolismo , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Neoplasias/patología , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Activación Transcripcional , Regulación hacia Arriba
13.
J Biol Chem ; 285(38): 29434-45, 2010 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-20622260

RESUMEN

In metastatic cancer cells, the process of invasion is regulated by several transcription factors that induce changes required for migration and resistance to apoptosis. Slug (SNAI2, Snail2) is involved in epithelial mesenchymal transition in physiological and in pathological contexts. We show here that in embryonic kidney, colon carcinoma, chronic myeloid leukemia-blast crisis, and in neuroblastoma cells, expression of Slug is transcriptionally regulated by c-Myb via Myb binding sites in the 5'-flanking region and in the first intron of the slug gene. In embryonic kidney and neuroblastoma cells, c-Myb induced vimentin, fibronectin, and N-cadherin expression and membrane ruffling via actin polymerization consistent with the acquisition of a mesenchymal-like phenotype. Furthermore, down-regulation of endogenous c-Myb levels in colon carcinoma cells led to increased expression of E-cadherin and reduced levels of vimentin. Some of these changes are predominantly Slug-dependent as Slug silencing via RNA interference (RNAi) reverts the cells to a quasi-parental condition. Changes in gene expression and morphology induced by c-Myb-activated Slug correlated with increased ability to migrate (embryonic kidney) and to invade through a Matrigel membrane (embryonic kidney, colon carcinoma, neuroblastoma). c-Myb-dependent Slug expression was also essential for the homing of chronic myeloid leukemia K562 cells to the bone marrow. In summary, we show here that the proto-oncogene c-Myb controls Slug transcription in tumor cells of different origin. Such a regulatory pathway contributes to the acquisition of invasive properties that are important for the metastatic process.


Asunto(s)
Médula Ósea/patología , Proteínas Proto-Oncogénicas c-myb/metabolismo , Factores de Transcripción/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Western Blotting , Línea Celular , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Etopósido/farmacología , Citometría de Flujo , Humanos , Intrones/genética , Ratones , Ratones SCID , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/fisiopatología , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas/genética , Unión Proteica , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-myb/genética , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética
14.
Chemistry ; 17(12): 3474-81, 2011 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-21341330

RESUMEN

The ruthenium and osmium complexes [MCl(2)(diphosphane)(L)] (M=Ru, Os; L=bidentate amino ligand) and [MCl(CNN)(dppb)] (CNN=pincer ligand; dppb=1,4-bis-(diphenylphosphino)butane), containing the N−H moiety, have been found to catalyze the acceptorless dehydrogenation of alcohols in tBuOH and in the presence of KOtBu. The compounds trans-[MCl(2)(dppf)(en)] (M=Ru 7, Os 13; dppf=1,1'-bisdiphenylphosphino)ferrocene; en=ethylenediamine) display very high activity and different substrates, including cyclic and linear alcohols, are efficiently oxidized to ketones by using 0.8-0.04 mol % of catalyst. The effect of the base and the comparison of the catalytic activity of the Ru versus Os complexes are reported. The ruthenium complex 7 generally leads to a faster conversion into ketones with respect to the osmium complex 13, which displays better activity in the dehydrogenation of 5-en-3ß-hydroxy steroids. The synthesis of new Ru and Os complexes [MCl(2)(PP)(L)] (PP=dppb, dppf; L=(±)-trans-1,2-diaminocyclohexane,2-(aminomethyl)pyridine, and 2-aminoethanol) of trans and cis configuration is also reported.

15.
J Exp Clin Cancer Res ; 40(1): 89, 2021 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-33673859

RESUMEN

BACKGROUND: Recent developments in abscopal effect strongly support the use of radiotherapy for the treatment of metastatic disease. However, deeper understanding of the molecular mechanisms underlying the abscopal effect are required to best benefit a larger proportion of patients with metastasis. Several groups including ours, reported the involvement of wild-type (wt) p53 in radiation-induced abscopal effects, however very little is known on the role of wtp53 dependent molecular mechanisms. METHODS: We investigated through in vivo and in vitro approaches how wtp53 orchestrates radiation-induced abscopal effects. Wtp53 bearing (A549) and p53-null (H1299) NSCLC lines were xenotransplanted in nude mice, and cultured in 2D monolayers and 3D tumor spheroids. Extracellular vesicles (EVs) were isolated from medium cell culture by ultracentrifugation protocol followed by Nanoparticle Tracking Analysis. Gene expression was evaluated by RT-Real Time, digital qRT-PCR, and dot blot technique. Protein levels were determined by immunohistochemistry, confocal anlysis, western blot techniques, and immunoassay. RESULTS: We demonstrated that single high-dose irradiation (20 Gy) induces significant tumor growth inhibition in contralateral non-irradiated (NIR) A549 xenograft tumors but not in NIR p53-null H1299 or p53-silenced A549 (A549sh/p53) xenografts. We further demonstrates that irradiation of A549 cells in vitro induces a senescence-associated secretory phenotype (SASP) producing extracellular vesicles (EVs) expressing CD63 and carrying DNA:RNA hybrids and LINE-1 retrotransposon. IR-A549 EVs also hamper the colony-forming capability of recipient NIR A549 cells, induce senescent phenotype, nuclear expression of DNA:RNA hybrids, and M1 macrophage polarization. CONCLUSIONS: In our models, we demonstrate that high radiation dose in wtp53 tumors induce the onset of SASP and secretion of CD63+ EVs loaded with DNA:RNA hybrids and LINE-1 retrotransposons that convey senescence messages out of the irradiation field triggering abscopal effect in NIR tumors.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Células A549 , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular/fisiología , Senescencia Celular/fisiología , Femenino , Xenoinjertos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Células RAW 264.7
16.
Sci Rep ; 11(1): 8118, 2021 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-33854097

RESUMEN

A radiological or nuclear attack could involve such a large number of subjects as to overwhelm the emergency facilities in charge. Resources should therefore be focused on those subjects needing immediate medical attention and care. In such a scenario, for the triage management by first responders, it is necessary to count on efficient biological dosimetry tools capable of early detection of the absorbed dose. At present the validated assays for measuring the absorbed dose are dicentric chromosomes and micronuclei counts, which require more than 2-3 days to obtain results. To overcome this limitation the NATO SPS Programme funded an Italian-Egyptian collaborative project aimed at validating a fast, accurate and feasible tool for assessing the absorbed dose early after radiation exposure. Biomarkers as complete blood cell counts, DNA breaks and radio-inducible proteins were investigated on blood samples collected before and 3 h after the first fraction of radiotherapy in patients treated in specific target areas with doses/fraction of about: 2, 3.5 or > 5 Gy and compared with the reference micronuclei count. Based on univariate and multivariate multiple linear regression correlation, our results identify five early biomarkers potentially useful for detecting the extent of the absorbed dose 3 h after the exposure.


Asunto(s)
Biomarcadores/metabolismo , Radiación Ionizante , Adulto , Anciano , Anciano de 80 o más Años , Área Bajo la Curva , Biomarcadores/sangre , Recuento de Células Sanguíneas , Roturas del ADN de Doble Cadena/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Femenino , Humanos , Masculino , Persona de Mediana Edad , Curva ROC , Exposición a la Radiación , Radiometría
17.
Biochim Biophys Acta ; 1793(2): 368-77, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19046997

RESUMEN

HIPK2 has been implicated in restraining tumor progression by more than one mechanism, involving both its catalytic and transcriptional co-repressor functions. Starting from the finding that HIPK2 knockdown by RNA-interference (HIPK2i) induced significant up-regulation of HIF-1alpha mRNA and of its target VEGF in tumor cells, we evaluated the role of HIPK2 in transcriptional regulation of HIF-1alpha. We found that HIPK2 overexpression downmodulated both HIF-1alpha reporter activity and mRNA levels and showed that HIPK2 was bound in vivo to the HIF-1alpha promoter likely in a multiprotein co-repressor complex with histone deacetylase 1 (HDAC1). Thus, the HIF-1alpha promoter was strongly acetylated following HIPK2 knockdown. The HIF-1alpha-dependent VEGF transcription was evaluated by co-transfection of a dominant negative (DN) construct of HIF-1alpha that inhibited VEGF reporter activity induced by HIPK2 knockdown. HIF-1alpha and VEGF up-regulation in HIPK2i cells correlated with increased vascularity of tumor xenografts in vivo and tube formation in HUVEC in vitro. These findings provide the first evidence of HIPK2-mediated transcriptional regulation of HIF-1alpha that might play a critical role in VEGF expression.


Asunto(s)
Proteínas Portadoras/metabolismo , Regulación Neoplásica de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias/genética , Neoplasias/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Transcripción Genética , Animales , Línea Celular Tumoral , Proliferación Celular , Ensamble y Desensamble de Cromatina/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Neoplasias/irrigación sanguínea , Neoplasias/enzimología , Neovascularización Patológica/genética , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Represoras/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética
18.
Exp Cell Res ; 315(1): 67-75, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18996371

RESUMEN

The maintenance of p53 transactivation activity is important for p53 apoptotic function. We have shown that stable knockdown of HIPK2 induces p53 misfolding with inhibition of p53 target gene transcription. In this study we established a lentiviral-based system for doxycyclin (Dox)-induced conditional interference of HIPK2 expression to evaluate the molecular mechanisms involved in p53 deregulation. We found that HIPK2 knockdown induced metallothionein 2A (MT2A) upregulation as assessed by RT-PCR analysis, increased promoter acetylation, and increased promoter luciferase activity. The MT2A upregulation correlated with resistance to Adriamycin (ADR)-driven apoptosis and with p53 inhibition. Thus, acute knockdown of HIPK2 (HIPK2i) induced misfolded p53 protein in MCF7 breast cancer cells and inhibited p53 DNA-binding and transcription activities in response to ADR treatment. Previous works show that MT may modulate p53 activity through zinc exchange. Here, we found that inhibition of MT2A expression by siRNA in the HIPK2i cells restored p53 transcription activity. Similarly zinc supplementation to HIPK2i cells restored p53 transcription activity and drug-induced apoptosis. These data support the notion that MT2A is involved in p53 deregulation and strengthen the possibility that combination of chemotherapy and zinc might be useful to treat tumors with inactive wtp53.


Asunto(s)
Metalotioneína/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Proteína p53 Supresora de Tumor/metabolismo , Zinc/metabolismo , Apoptosis/efectos de los fármacos , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , ADN de Neoplasias/metabolismo , Doxorrubicina/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Lentivirus , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Unión Proteica/efectos de los fármacos , Conformación Proteica/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Transcripción Genética/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/genética , Regulación hacia Arriba/efectos de los fármacos , Zinc/farmacología
19.
Mol Cancer Ther ; 8(3): 543-51, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19258428

RESUMEN

We reported previously that the disruption of c-Myc through mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) inhibition blocks the expression of the transformed phenotype in the embryonal rhabdomyosarcoma (ERMS) cell line (RD), thereby inducing myogenic differentiation in vitro. In this article, we investigate whether MEK/ERK inhibition, by the MEK/ERK inhibitor U0126, affects c-Myc protein level and growth of RMS tumor in an in vivo xenograft model. U0126 significantly reduced RMS tumor growth in RD cell line-xenotransplanted mice. Immunobiochemical and immunohistochemical analysis showed (a) phospho-active ERK levels were reduced by U0126 therapy and unaltered in normal tissues, (b) phospho-Myc and c-Myc was reduced commensurate with phospho-ERK inhibition, and (c) reduction in Ki-67 and endothelial (CD31) marker expression. These results indicate that MEK/ERK inhibition affects growth and angiogenic signals in tumor. The RD-M1 cultured xenograft tumor-derived cell line and the ERMS cell line TE671 responded to U0126 by arresting growth, down-regulating c-Myc, and initiating myogenesis. All these results suggest a tight correlation of MEK/ERK inhibition with c-Myc down-regulation and arrest of tumor growth. Thus, MEK inhibitors may be investigated for a signal transduction-based targeting of the c-Myc as a therapeutic strategy in ERMS.


Asunto(s)
Butadienos/farmacología , Proliferación Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Neoplasias de los Músculos/patología , Nitrilos/farmacología , Rabdomiosarcoma Embrionario/patología , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Butadienos/uso terapéutico , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , Neoplasias de los Músculos/tratamiento farmacológico , Neoplasias de los Músculos/genética , Nitrilos/uso terapéutico , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Rabdomiosarcoma Embrionario/tratamiento farmacológico , Rabdomiosarcoma Embrionario/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Blood Adv ; 4(22): 5616-5630, 2020 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-33186461

RESUMEN

Multiple myeloma (MM) is a hematologic malignancy produced by a clonal expansion of plasma cells and characterized by abnormal production and secretion of monoclonal antibodies. This pathology exhibits an enormous heterogeneity resulting not only from genetic alterations but also from several epigenetic dysregulations. Here we provide evidence that Che-1/AATF (Che-1), an interactor of RNA polymerase II, promotes MM proliferation by affecting chromatin structure and sustaining global gene expression. We found that Che-1 depletion leads to a reduction of "active chromatin" by inducing a global decrease of histone acetylation. In this context, Che-1 directly interacts with histones and displaces histone deacetylase class I members from them. Strikingly, transgenic mice expressing human Che-1 in plasma cells develop MM with clinical features resembling those observed in the human disease. Finally, Che-1 downregulation decreases BRD4 chromatin accumulation to further sensitize MM cells to bromodomain and external domain inhibitors. These findings identify Che-1 as a promising target for MM therapy, alone or in combination with bromodomain and external domain inhibitors.


Asunto(s)
Mieloma Múltiple , Proteínas Nucleares , Proliferación Celular , Cromatina , Humanos , Mieloma Múltiple/genética , Proteínas Nucleares/genética , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA