Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Int J Mol Sci ; 22(9)2021 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-33925224

RESUMEN

BACKGROUND: Cancer stem cells' (CSCs) self-maintenance is regulated via the pluripotency pathways promoting the most aggressive tumor phenotype. This study aimed to use the activity of these pathways for the CSCs' subpopulation enrichment and separating cells characterized by the OCT4 and SOX2 expression. METHODS: To select and analyze CSCs, we used the SORE6x lentiviral reporter plasmid for viral transduction of colon adenocarcinoma cells. Additionally, we assessed cell chemoresistance, clonogenic, invasive and migratory activity and the data of mRNA-seq and intrinsic disorder predisposition protein analysis (IDPPA). RESULTS: We obtained the line of CSC-like cells selected on the basis of the expression of the OCT4 and SOX2 stem cell factors. The enriched CSC-like subpopulation had increased chemoresistance as well as clonogenic and migration activities. The bioinformatic analysis of mRNA seq data identified the up-regulation of pluripotency, development, drug resistance and phototransduction pathways, and the downregulation of pathways related to proliferation, cell cycle, aging, and differentiation. IDPPA indicated that CSC-like cells are predisposed to increased intrinsic protein disorder. CONCLUSION: The use of the SORE6x reporter construct for CSCs enrichment allows us to obtain CSC-like population that can be used as a model to search for the new prognostic factors and potential therapeutic targets for colon cancer treatment.


Asunto(s)
Adenocarcinoma/patología , Neoplasias del Colon/patología , Células Madre Neoplásicas/metabolismo , Adenocarcinoma/genética , Adulto , Biomarcadores de Tumor/aislamiento & purificación , Técnicas de Cultivo de Célula/métodos , Ciclo Celular , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias del Colon/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Masculino , Persona de Mediana Edad , Células Madre Neoplásicas/patología , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Cultivo Primario de Células , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
2.
Biochem Biophys Res Commun ; 503(3): 2180-2185, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30098786

RESUMEN

Resveratrol is a natural polyphenol with several therapeutic effects, in particular, inducing p53-dependent cell cycle arrest and/or apoptosis in tumor cells. Resveratrol-induced p53 activation may trigger differentiation and apoptosis in embryonic stem cells (ESCs). We show that resveratrol activates p53 that is negatively regulated by SIRT1 deacetylation on Lys379 and positively by AMPK phosphorylation on Ser15 in mouse ESCs (mESCs). Surprisingly, the resveratrol-activated p53 is not associated with either G1/S cell cycle checkpoint or apoptosis in mESCs. Instead, it stimulates autophagy in a transcriptional-dependent manner involving up-regulation of dram1 gene expression. This study demonstrates a novel mechanism of resveratrol-dependent p53 activation in mESCs.


Asunto(s)
Autofagia/efectos de los fármacos , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/efectos de los fármacos , Resveratrol/farmacología , Proteína p53 Supresora de Tumor/agonistas , Animales , Puntos de Control del Ciclo Celular/efectos de los fármacos , Células Cultivadas , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba/efectos de los fármacos
3.
Methods Mol Biol ; 2445: 139-169, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34972991

RESUMEN

Anticancer therapy is complicated by the ability of malignant cells to activate cytoprotective autophagy that rescues treated cells. This protocol describes methods for analysis of autophagic process in apoptosis-resistant tumor cells treated with damaging agents. Induction of autophagy in these cells can activate apoptotic death. Protocol provides methods for Western blotting, immunofluorescent analysis, and transfection of cells with fluorescent protein-tagged LC3-encoding plasmids to analyze autophagy. Different approaches to change autophagy in tumor cells are suggested. A special approach is connected with induction of cellular senescence. Senescent cells, which are resistant to apoptosis, are vulnerable to certain damaging agents, in particular, to kinase inhibitors. Methods to induce and analyze senescence are considered. They include detection of proliferation arrest by different ways, mTORC1 activity assay and fluorescent analysis of mTORC1 and lysosome localization as a novel senescence hallmark. Incapability of senescent cells to complete autophagy after damage allows to force them to apoptosis. To demonstrate apoptotic cell death, analysis of caspase activity, Annexin V-FITC binding, DNA fragmentation, and mitochondria and lysosome damage are suggested. The methods described can be applied in studies aimed on developing different strategies of tumor cell elimination through changing autophagy.


Asunto(s)
Apoptosis , Autofagia , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular Tumoral , Senescencia Celular , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo
4.
Biol Cell ; 102(10): 549-60, 2010 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-20626347

RESUMEN

BACKGROUND: GSK3beta (glycogen synthase kinase 3beta) regulates the expression level and activity of various target proteins, including beta-catenin. beta-Catenin is a co-activator of Wnt-dependent genes as well as a partner for transmembrane cadherins to mediate cell-to-cell adhesion. In some cases, inhibition of GSK3beta activity was shown to promote self-renewal of ESCs (embryonic stem cells), but immediate effects of GSK3beta inhibitors in these cells still remain elusive. RESULTS: Here, we address the effects of GSK3beta inhibitors BIO (6-bromoindirubin-3'-oxime) and CHIR99021 on mESCs (mouse ESCs), focusing on modulation of beta-catenin activities. We found that, upon GSK3beta inhibition, the colonies of undifferentiated mESCs acquire a more compact morphology. This change is paralleled by two somewhat polar effects: (i) the accumulation of the beta-catenin, which is co-localized with E-cadherin at the plasma membrane, and the cytoplasmic, tyrosine unphosphorylated beta-catenin, which is able to bind the GST (glutathione transferase)-fused cytoplasmic domain of E-cadherin; and (ii) the accumulation of the tyrosine phosphorylated beta-catenin and its nuclear translocation that is accompanied by activation of the Tcf (T-cell factor)/beta-catenin-dependent transcription of Top-Flash reporter. The Tcf-mediated activation, however, does not affect most of the analysed Wnt-responsive genes involved in EMT (epithelial-mesenchymal transition) or cell-cycle progression, suggesting that the adhesive function of beta-catenin is dominant over transcription in undifferentiated mESCs. Treatment with BIO decreases proliferation rates of mESCs. This is not due to apoptosis, but rather to accumulation of cells in G1 phase of the cell cycle and is accompanied by down-regulation of the c-myc mRNA content. CONCLUSION: Our results suggest that inhibition of GSK3beta activity in mESCs enhances both the beta-catenin/E-cadherin-mediated adhesion and the Tcf/beta-catenin-dependent transcription, but does not activate transcription in most of the examined genes involved in EMT and cell cycle progression.


Asunto(s)
Células Madre Embrionarias/citología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Transducción de Señal , beta Catenina/metabolismo , Animales , Cadherinas/metabolismo , Adhesión Celular , Células Cultivadas , Células Madre Embrionarias/metabolismo , Transición Epitelial-Mesenquimal , Técnica del Anticuerpo Fluorescente , Glucógeno Sintasa Quinasa 3 beta , Ratones
5.
Biochem Biophys Res Commun ; 391(1): 142-6, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19900401

RESUMEN

HDAC inhibitors induce cell cycle arrest of E1A+Ras-transformed cells accompanied by e2f1 gene down-regulation and activation of Wnt pathway. Here we show that e2f1 expression is regulated through the Wnt/Tcf-pathway: e2f1 promoter activity is inhibited by sodium butyrate (NaB) and by overexpression of beta-catenin/Tcf. The e2f1 promoter was found to contain two putative Tcf-binding elements: the proximal one competes well with canonical Tcf element in DNA-binding assay. Being inserted into luciferase reporter vector, the identified element provides positive transcriptional regulation in response to beta-catenin/Tcf co-transfection and NaB treatment. Thus we have firstly demonstrated that e2f1 belongs to genes regulated through Wnt/beta-catenin/Tcf pathway.


Asunto(s)
Factor de Transcripción E2F1/genética , Regulación de la Expresión Génica , Factores de Transcripción TCF/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Secuencia de Bases , Butiratos/farmacología , Línea Celular , Inhibidores de Histona Desacetilasas/farmacología , Ratones , Datos de Secuencia Molecular , Regiones Promotoras Genéticas
6.
Cell Death Discov ; 5: 61, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30729040

RESUMEN

Resveratrol, a natural polyphenolic compound, shows many beneficial effects in various animal models. It increases efficiency of somatic cell reprograming into iPSCs and contributes to cell differentiation. Here, we studied the effect of resveratrol on proliferation and pluripotency of mouse embryonic stem cells (mESCs). Our results demonstrate that resveratrol induces autophagy in mESCs that is provided by the activation of the AMPK/Ulk1 pathway and the concomitant suppression of the activity of the mTORC1 signaling cascade. These events correlate with the enhanced expression of pluripotency markers Oct3/4, Sox2, Nanog, Klf4, SSEA-1 and alkaline phosphatase. Pluripotency is retained under resveratrol-caused retardation of cell proliferation. Given that the Ulk1 overexpression enhances pluripotency of mESCs, the available data evidence that mTOR/Ulk1/AMPK-autophagy network provides the resveratrol-mediated regulation of mESC pluripotency. The capability of resveratrol to support the mESC pluripotency provides a new approach for developing a defined medium for ESC culturing as well as for better understanding signaling events that govern self-renewal and pluripotency.

7.
J Cell Biochem ; 103(3): 1005-12, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17647273

RESUMEN

We have previously shown that transcription of immediate-early c-fos protooncogene is becoming strongly repressed in rat embryo fibroblasts transformed by oncogenes E1A and cHa-ras, so that serum only slightly stimulated c-fos transcription in these cells in contrast to high level of c-fos activation in non-transformed REF52 cells. Here we showed that stress-inducing agent anisomycin was able to override the c-fos repression and to induce c-fos transcription in E1A + ras transformants. In vitro kinase assay data demonstrated that anisomycin increased phosphorylation of transactivation domain of Elk-1 transcription factor--a key regulator of inducible c-fos transcription. Importantly, this activation was mediated through up-regulation of MEK/ERK but not stress-kinase cascades JNK or p38. The activating effect of anisomycin on c-fos transcription could be abrogated by a prior treatment with N-acetyl-L-cysteine. This indicates that anisomycin potentiates generation of reactive oxygen species (ROS), which, in turn, can modulate the activity of MAP kinase-specific phosphatases (MKPs). As anisomycin did not cause acetylation of nucleosome core histones, the present work focuses on the molecular mechanisms mediating the HDAC-independent induction of IEG c-fos by anisomycin in E1A + cHa-ras-transformed fibroblasts.


Asunto(s)
Proteínas E1A de Adenovirus/biosíntesis , Anisomicina/farmacología , Transformación Celular Neoplásica/metabolismo , Genes fos/efectos de los fármacos , Proteínas Inmediatas-Precoces/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Proteínas Proto-Oncogénicas c-fos/sangre , Animales , Línea Celular Transformada , Transformación Celular Neoplásica/genética , Activación Enzimática , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen/efectos de los fármacos , Silenciador del Gen/fisiología , Genes ras/efectos de los fármacos , Proteínas Inmediatas-Precoces/genética , Proteínas Quinasas Activadas por Mitógenos , Fosforilación , Ratas , Activación Transcripcional/efectos de los fármacos , Proteína Elk-1 con Dominio ets/efectos de los fármacos
8.
Aging (Albany NY) ; 9(11): 2352-2375, 2017 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-29140794

RESUMEN

The Ras-Raf-MEK-ERK pathway plays a central role in tumorigenesis and is a target for anticancer therapy. The successful strategy based on the activation of cell death in Ras-expressing cells is associated with the suppression of kinases involved in Ras pathway. However, activation of cytoprotective autophagy overcomes antiproliferative effect of the inhibitors and develops drug resistance. We studied whether cellular senescence induced by HDAC inhibitor sodium butyrate in E1a+cHa-Ras-transformed rat embryo fibroblasts (ERas) and A549 human Ki-Ras mutated lung adenocarcinoma cells would enhance the tumor suppressor effect of MEK/ERK inhibition. Treatment of control ERas cells with PD0325901 for 24 h results in mitochondria damage and apoptotic death of a part of cellular population. However, the activation of AMPK-dependent autophagy overcomes pro-apoptotic effects of MEK/ERK inhibitor and results in restoration of the mitochondria and rescue of viability. Senescent ERas cells do not develop cytoprotective autophagy upon inhibition of MEK/ERK pathway due to spatial dissociation of lysosomes and autophagosomes in the senescent cells. Senescent cells are unable to form the autophagolysosomes and to remove the damaged mitochondria resulting in apoptotic death. Our data show that suppression of MEK/ERK pathway in senescent cells provides a new strategy for elimination of Ras-expressing cells.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Senescencia Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibroblastos/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Quinasas Quinasa Quinasa PAM/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Células A549 , Proteínas Quinasas Activadas por AMP/metabolismo , Adenocarcinoma/enzimología , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Fibroblastos/enzimología , Fibroblastos/patología , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Ratas , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
9.
Cell Cycle ; 15(1): 52-63, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26636245

RESUMEN

Mouse embryonic stem cells (mESCs) lack of G1 checkpoint despite that irradiation (IR) activates ATM/ATR-mediated DDR signaling pathway. The IR-induced p53 localizes in the nuclei and up-regulates p21/Waf1 transcription but that does not lead to accumulation of p21/Waf1 protein. The negative control of the p21Waf1 expression appears to occur at 2 levels of regulation. First, both p21/Waf1 gene transcription and the p21/Waf1 protein content increase in mESCs treated with histone-deacetylase inhibitors, implying its epigenetic regulation. Second, proteasome inhibitors cause the p21/Waf1 accumulation, indicating that the protein is a subject of proteasome-dependent degradation in ESСs. Then, the dynamics of IR-induced p21Waf1 protein show its accumulation at long-term time points (3 and 5 days) that coincides with an increase in the proportion of G1-phase cells, down-regulation of Oct4 and Nanog pluripotent gene transcription and activation of endoderm-specific genes sox17 and afp. In addition, nutlin-dependent stabilization of p53 in mESC was also accompanied by the accumulation of p21/Waf1 as well as restoration of G1 checkpoint and an onset of differentiation. Thus, the lack of functional p21/Waf1 is indispensable for maintaining self-renewal and pluripotency of mESCs.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Madre Embrionarias/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/fisiología , Transducción de Señal/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Supervivencia Celular/fisiología , Ratones , Células 3T3 NIH
10.
Oncogene ; 21(5): 719-30, 2002 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-11850800

RESUMEN

REF cells transformed by oncogenes E1A and cHa-ras reveal high and constitutive DNA-binding activity of AP-1 factor lacking in c-Fos protein. Consistently, the transcription of c-fos gene has been found to be downregulated. To elucidate the mechanisms of c-fos downregulation in E1A+cHa-ras transformants, we studied the levels of activity of ERK, JNK/SAPK and p38 kinases and phosphorylation state of Elk-1 transcription factor involved in regulation of c-fos gene. Using two approaches, Western blot analysis with phospho-specific antibodies to MAP kinases and in vitro kinase assay with specific substrates, we show here that ectopic expression of E1A and ras oncogenes leads to a sustained activation of ERK and p38 kinases, whereas JNK/SAPK kinase activity is similar to that in non-transformed REF52 cells. Due to sustained activity of the MAP kinase cascades, Elk-1 transcription factor is being phosphorylated even in serum-starved E1A+cHa-ras cells; moreover, serum does not additionally increase phosphorylation of Elk-1, which is predominant TCF protein bound to SRE region of c-fos gene promoter in these cells. Although the amount of ternary complexes SRE/SRF/TCF estimated by EMSA was similar both in serum-starved and serum-stimulated transformed cells, serum addition still caused a modest activation of c-fos gene transcription at the level of 20% to normal REF cells. In attempt to determine how serum caused the stimulatory effect, we found that PD98059, an inhibitor of MEK/ERK kinase cascade, completely suppressed serum-induced c-fos transcription both in REF and E1A+cHa-ras cells, implicating the ERK as primary kinase for c-fos transcription in these cells. In contrast, SB203580, an inhibitor of p38 kinase, augmented noticeably serum-stimulated transcription of c-fos gene in REF cells, implying the involvement of p38 kinase in negative regulation of c-fos. Furthermore, sodium butyrate, an inhibitor of histone deacetylase activity, was capable of activating c-fos transcription both in serum-stimulated and even in serum-starved E1A+cHa-ras cells. Conversely, serum-starved REF cells fail to respond to sodium butyrate treatment by c-fos activation confirming necessity of prior Elk-1 phosphorylation. Taken together, these data suggest that downregulation of c-fos in E1A+cHa-ras cells seems to occur due to a maintenance of a refractory state that arises in normal REF cells after serum-stimulation. The refractory state of c-fos in E1A+cHa-ras cells is likely a consequence of Ras-induced sustained activation of MAPK (ERK) cascade and persistent phosphorylation of TCF (Elk-1) bound to SRE. Combination of these events eventually does contribute to formation of an inactive chromatin structure at c-fos promoter mediated through recruitment of histone deacetylase activity.


Asunto(s)
Proteínas E1A de Adenovirus/genética , Proteínas de Unión al ADN , Regulación hacia Abajo , Genes fos , Genes ras , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Factores de Transcripción , Animales , Butiratos/farmacología , Línea Celular , Línea Celular Transformada , Cromatina/metabolismo , Cromatina/ultraestructura , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Inhibidores de Histona Desacetilasas , Humanos , Isobutiratos , Cinética , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/metabolismo , ARN Mensajero/biosíntesis , Ratas , Elemento de Respuesta al Suero , Transcripción Genética , Proteína Elk-1 con Dominio ets , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA