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










Base de dados
Intervalo de ano de publicação
1.
Clin Transl Radiat Oncol ; 41: 100649, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37346275

RESUMO

Background and purpose: MAPKs are among the most relevant signalling pathways involved in coordinating cell responses to different stimuli. This group includes p38MAPKs, constituted by 4 different proteins with a high sequence homology: MAPK14 (p38α), MAPK11 (p38ß), MAPK12 (p38γ) and MAPK13 (p38δ). Despite their high similarity, each member shows unique expression patterns and even exclusive functions. Thus, analysing protein-specific functions of MAPK members is necessary to unequivocally uncover the roles of this signalling pathway. Here, we investigate the possible role of MAPK11 in the cell response to ionizing radiation (IR). Materials and methods: We developed MAPK11/14 knockdown through shRNA and CRISPR interference gene perturbation approaches and analysed the downstream effects on cell responses to ionizing radiation in A549, HCT-116 and MCF-7 cancer cell lines. Specifically, we assessed IR toxicity by clonogenic assays; DNA damage response activity by immunocytochemistry; apoptosis and cell cycle by flow cytometry (Annexin V and propidium iodide, respectively); DNA repair by comet assay; and senescence induction by both X-Gal staining and gene expression of senescence-associated genes by RT-qPCR. Results: Our findings demonstrate a critical role of MAPK11 in the cellular response to IR by controlling the associated senescent phenotype, and without observable effects on DNA damage response, apoptosis, cell cycle or DNA damage repair. Conclusion: Our results highlight MAPK11 as a novel mediator of the cellular response to ionizing radiation through the control exerted onto IR-associated senescence.

2.
Radiother Oncol ; 156: 136-144, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33310004

RESUMO

BACKGROUND AND PURPOSE: Gemcitabine is an antitumour agent currently used in the treatment of several types of cancer with known properties as a radiosensitizer. p38MAPK signalling pathway has been shown to be a major determinant in the cellular response to gemcitabine in different experimental models. However, the molecular mechanism implicated in gemcitabine-associated radiosensitivity remains unknown. MATERIALS AND METHODS: The human sarcoma cell lines A673 and HT1080, and a mouse cell line derived from a 3-methylcholanthrene induced sarcoma were used as experimental models. Modulation of p38MAPKs was performed by pharmacological approaches (SB203580) and genetic interference using lentiviral vectors coding for specific shRNAs. Viability was assessed by MTT. Gene expression was evaluated by western blot and RT-qPCR. Induction of apoptosis was monitored by caspase 3/7 activity. Response to ionizing radiation was evaluated by clonogenic assays. RESULTS: Our data demonstrate that chemical inhibition of p38MAPK signalling pathway blocks gemcitabine radiosensitizing potential. Genetic interference of MAPK14 (p38α), the most abundantly expressed and best characterized p38MAPK, despite promoting resistance to gemcitabine, it does not affect its radiosensitizing potential. Interestingly, specific knockdown of MAPK11 (p38ß) induces a total loss of the radiosensitivity associated to gemcitabine, as well as a marked increase in the resistance to the drug. CONCLUSION: The present work identifies p38ß as a major determinant of the radiosensitizing potential of gemcitabine without implication of p38α, suggesting that p38ß status should be analysed in those cases in which gemcitabine is combined with ionizing radiation.


Assuntos
Proteína Quinase 11 Ativada por Mitógeno , Sarcoma , Apoptose , Linhagem Celular Tumoral , Desoxicitidina/análogos & derivados , Humanos , Modelos Teóricos , Tolerância a Radiação/genética , Gencitabina
3.
Cancer Lett ; 451: 23-33, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30872077

RESUMO

Targeting cell cycle has become one of the major challenges in cancer therapy, being Palbociclib, a CDK4/6 inhibitor, an excellent example. Recently, it has been reported that Palbociclib could be a novel radiosensitizer agent. In an attempt to clarify the molecular basis of this effect we have used cell lines from colorectal (HT29, HCT116) lung (A549, H1299) and breast cancer (MCF-7). Our results indicate that the presence of a p53 wild type is strictly required for Palbociclib to exert its radiosensitizing effect, independently of the inhibitory effect exerted on CDK4/6. In fact, abrogation of p53 in cells with functional p53 blocks the radiosensitizing effect of Palbociclib. Moreover, no radiosensitizing effect is observed in cells with non-functional p53, but restoration of p53 function promotes radiosensitivity associated to Palbociclib. Furthermore, the presence of Palbociclib blocks the transcriptional activity of p53 in an ATM-dependent-fashion after ionizing radiation exposure, as the blockage of p21/WAF1 expression demonstrates. These observations are a proof of concept for a more selective therapy, based on the combination of CDK4/6 inhibition and radiotherapy, which would only benefit to those patients with a functional p53 pathway.


Assuntos
Piperazinas/farmacologia , Piridinas/farmacologia , Radiossensibilizantes/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Humanos , Transdução de Sinais/efeitos dos fármacos
4.
Clin Transl Oncol ; 21(9): 1280-1285, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30680609

RESUMO

PURPOSE: Autophagy has lately emerged as an important biological process with implications in several hematological pathologies. Recently, a growing body of evidence supports a putative role of autophagy in chronic lymphocytic leukemia; however, no definitive clue has been established so far. To elucidate this issue, we have developed a pilot study to measure autophagic flux in peripheral blood mononuclear cells from chronic lymphocytic leukemia patients, and explored its correlation with classical clinical/analytical parameters. METHODS/PATIENTS: Thirty-three chronic lymphocytic leukemia patients participated in the study. Autophagic flux in peripheral blood mononuclear cells was determined by western blot measuring the levels of the proteins p62 and lipidated LC3. Moreover, p62 mRNA levels were analyzed by RT-qPCR. RESULTS: Lymphocytosis and the percentage of tumoral lymphocytes in chronic lymphocytic leukemia patients statistically correlate with a blocked autophagic flux. CONCLUSION: Alterations in autophagic flux could play an important role in the physiopathology of chronic lymphocytic leukemia.


Assuntos
Autofagia , Biomarcadores Tumorais/metabolismo , Leucemia Linfocítica Crônica de Células B/patologia , Leucócitos Mononucleares/patologia , Linfocitose/patologia , Idoso , Idoso de 80 Anos ou mais , Feminino , Seguimentos , Humanos , Leucemia Linfocítica Crônica de Células B/metabolismo , Leucócitos Mononucleares/metabolismo , Linfocitose/metabolismo , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Prognóstico
5.
Oncogene ; 29(3): 411-20, 2010 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-19802006

RESUMO

The hairy/enhancer-of-split related with YRPW motif 1 (HEY1) is a member of the basic-helix-loop-helix-Orange (bHLH-O) family of transcriptional repressors that mediate Notch signaling. Several cancer-related pathways also regulate HEY1 expression, and HEY1 itself acts as an indirect positive regulator of the p53 tumor suppressor protein and a negative regulator of androgen receptor activity. In this study we show how a naturally occurring non-synonymous polymorphism at codon 94 of HEY1, which results in a substitution of leucine by methionine (Leu94Met), converts HEY1 from an androgen receptor corepressor to an androgen receptor co-activator without affecting its intrinsic transcriptional repressive domains. The polymorphism Leu94Met also abolishes HEY1-mediated activation of p53 and suppresses the ability of HEY1 to induce p53-dependent cell-cycle arrest and aberrant cell differentiation in human osteosarcoma U2OS cells. Moreover, expression of HEY1, but not of the variant Leu94Met, confers sensitivity to p53-activating chemotherapeutic drugs on U2OS cells. In addition, we have identified motifs in HEY1 that are critical for the regulation of its subcellular localization and analysed how mutations in those motifs affect both HEY1 and HEY1-Leu94Met functions. These findings suggest that the polymorphism Leu94Met in HEY1 radically alters its biological activities and may affect oncogenic processes.


Assuntos
Substituição de Aminoácidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteínas de Ciclo Celular/genética , Polimorfismo de Nucleotídeo Único , Receptores Androgênicos/genética , Sequência de Aminoácidos , Animais , Antineoplásicos/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Sítios de Ligação/genética , Células COS , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Humanos , Immunoblotting , Leucina/genética , Metionina/genética , Dados de Sequência Molecular , Mutação , Ligação Proteica , Receptores Androgênicos/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Ativação Transcricional , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
6.
Curr Cancer Drug Targets ; 8(7): 566-80, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18991567

RESUMO

The Notch pathway and the endocrine system constitute two key biological signaling mechanisms, responsible for cell-to-cell communication between adjacent cells and long-distance hormonal signals respectively. They play central roles during the development of higher eukaryotic organisms but they also take part in the regulation of many aspects of adult physiology and homeostasis. The contribution of defects in the normal transmission of hormone-dependent signals to the development of endocrine cancers has been widely analyzed and the knowledge derived from these studies has allowed us to develop many successful therapeutic strategies. However, in many cases these hormonal treatments become ineffective despite the fact that cancer cells maintain normal expression levels of wild-type hormone nuclear receptors. Less is known about the involvement of altered Notch signaling in the origin and progression of cancer, although there is clear evidence indicating that deregulation of Notch activity occurs in several types of tumors, including highly prevalent hormone-dependent types of cancer such as breast, ovarian and prostate cancer. This review will summarize accumulating data suggesting that Notch signaling plays a key role in the control of proliferation, differentiation and survival of prostate epithelial cells. Notch signals are required for normal prostate development and homeostasis, and abnormalities in Notch signaling may be critical during the development of prostate cancer. We will also discuss the possible oncogenic role for alterations in the crosstalk mechanisms between Notch and androgen-dependent signals during tumorigenesis in the prostate and how they could influence the outcome of anti-cancer hormonal treatments.


Assuntos
Antineoplásicos/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Neoplasias da Próstata/metabolismo , Receptores Notch/fisiologia , Transdução de Sinais/fisiologia , Animais , Proliferação de Células/efeitos dos fármacos , Humanos , Masculino , Neoplasias da Próstata/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos
7.
Biochem Soc Trans ; 34(Pt 6): 1124-7, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17073766

RESUMO

Anti-androgens used in prostate cancer therapy inhibit AR (androgen receptor) activity via largely unknown mechanisms. Although initially successful in most cases, they eventually fail and the disease progresses. We need to elucidate how anti-androgens work to understand why they fail, and prolong their effects or design further therapies. Using a cellular model, we found different anti-androgens have diverse effects on subcellular localization of AR, revealing that they work via different mechanisms and suggesting that an informed sequential treatment regime may benefit patients. In the presence of the anti-androgens bicalutamide and hydroxyflutamide, a significant proportion of the AR is translocated to the nucleus but remains inactive. Receptor inhibition under these conditions is likely to involve recruitment of co-repressor proteins, which interact with antagonist-occupied receptor but inhibit receptor-dependent transcription. Which co-repressors are required in vivo for AR repression by anti-androgens is not clear, but one candidate is the Notch effector Hey1. This inhibits ligand-dependent activity of the AR but not other steroid receptors. Further, it is excluded from the nucleus in most human prostate cancers, suggesting that abnormal subcellular distribution of co-repressors may contribute to the aberrant hormonal responses observed in prostate cancer. A decrease in co-repressor function is one possible explanation for the development of anti-androgen-resistant prostate cancer, and this suggests that it may not occur at the gross level of protein expression.


Assuntos
Antagonistas de Androgênios/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias da Próstata/fisiopatologia , Humanos , Masculino , Modelos Biológicos , Hiperplasia Prostática/patologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia
8.
J Biol Chem ; 275(40): 30801-5, 2000 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-10934189

RESUMO

SRC1, initially identified as a nuclear receptor coactivator, was found to interact with a member of the transcriptional enhancer factor (TEF) family of transcription factors, TEF-4. The interaction, which occurs in both intact cells and in a cell-free system, is mediated by the highly conserved basic helix-loop-helix/Per-Arnt-Sim (bHLH-PAS) domain present in the N-terminal region of SRC1. Moreover, all three members of the p160 family of nuclear receptor coactivators, SRC1, TIF2, and RAC3, are able to potentiate transcription from a TEF response element in transient transfection experiments, and this activation requires the presence of the bHLH-PAS domain. These results suggest that the p160 proteins could be bona fide coactivators of the TEF family of transcription factors.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Animais , Células COS , Sistema Livre de Células , DNA Complementar/metabolismo , Biblioteca Gênica , Glutationa Transferase/metabolismo , Células HeLa , Histona Acetiltransferases , Humanos , Immunoblotting , Camundongos , Modelos Genéticos , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Coativador 1 de Receptor Nuclear , Coativador 2 de Receptor Nuclear , Coativador 3 de Receptor Nuclear , Plasmídeos/metabolismo , Estrutura Terciária de Proteína , Elementos de Resposta , Saccharomyces cerevisiae/genética , Fatores de Transcrição de Domínio TEA , Transativadores/química , Transativadores/metabolismo , Fatores de Transcrição/química , Transfecção , Técnicas do Sistema de Duplo-Híbrido
9.
J Biol Chem ; 273(46): 30366-71, 1998 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-9804800

RESUMO

The expression of the beta-amyloid precursor protein (APP), which plays a key role in the development of Alzheimer's disease, is regulated by a variety of cellular mediators in a cell-dependent manner. In the present study, we present evidence that thyroid hormones negatively regulate the expression of the APP gene in neuroblastoma cells. Transient transfection studies using plasmids that contain progressive deletions of the 5' region of the gene demonstrate that triiodothyronine (T3), the more active form of the thyroid hormones, represses APP promoter activity by a mechanism that requires binding of the nuclear T3 receptor (TR) to a specific sequence located in the first exon. The unliganded receptor increases promoter activity, and T3 reverses that activity to basal levels. The repressive effect of T3 does not exhibit TR isoform specificity, and it is equally mediated by TRalpha and TRbeta. Gel mobility shift assays using in vitro synthesized nuclear receptors and nuclear extracts led to the identification of a negative thyroid hormone response element, at nucleotide position +80/+96, that preferentially binds heterodimers of TR with the retinoid X receptor. Insertion of sequences containing this element confers negative regulation by T3 to a heterologous TK promoter, thus indicating the functionality of the element.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Transcrição Gênica , Tri-Iodotironina/fisiologia , Animais , Éxons , Regulação da Expressão Gênica , Camundongos , RNA Mensageiro/metabolismo , Transfecção , Células Tumorais Cultivadas
10.
Endocrinology ; 139(6): 2692-8, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9607774

RESUMO

The beta-amyloid protein (Abeta), the major component of the senile plaques found in Alzheimer brains, derives from a larger beta-amyloid precursor protein (APP). Alternative splicing of the APP gene yields three major APP messenger RNAs (mRNAs), which, in turn, give rise to the APP770, APP751, and APP695 protein isoforms. In this study we examined the effects of thyroid hormone on APP expression in N2a-beta neuroblastoma cells. T3 caused a significant increase in the APP770 mRNA band, in detriment of the APP695 mRNA, which was proportionately reduced. In agreement with these results, T3 markedly altered the relative ratio of intracellular APP isoforms, increasing the amount of APP770 and causing an equivalent reduction of the immature APP695 isoform. In accordance with these results, the soluble APP695-derived form was specifically reduced in the culture medium obtained from T3-treated cells. In contrast, the increase in intracellular APP770 was not followed by an enhanced release of soluble derivatives of this isoform. These results suggest that T3 regulates not only APP gene splicing, but also the processing and secretion of the APP peptides. According to our results, thyroid hormone might play a role in the development of Alzheimer's disease by modulating the intracellular and extracellular contents of APP isoforms.


Assuntos
Processamento Alternativo , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Neuroblastoma/genética , Neuroblastoma/metabolismo , Tri-Iodotironina/farmacologia , Processamento Alternativo/efeitos dos fármacos , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Meios de Cultura/metabolismo , Isomerismo , Camundongos , Neuroblastoma/patologia , RNA Mensageiro/metabolismo , Células Tumorais Cultivadas/efeitos dos fármacos
11.
Biochem Biophys Res Commun ; 215(3): 889-95, 1995 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-7488057

RESUMO

Treatment of the human myeloid leukemia K562 cells with the protein phosphatase inhibitors okadaic acid or calyculin A resulted in down-regulation of both c-myc and max genes at the mRNA and protein levels. The extent of the down-regulation was similar for both genes and was dependent on the dose and on the treatment time. Interestingly, c-myc and max down-regulation was concomitant with apoptosis induced by okadaic acid and calyculin A in K562 cells. The expression of c-myc and max returned to control levels after the removal of okadaic acid from the media, although apoptosis was irreversible. These effects were observed at okadaic acid concentrations (15 nM) that inhibited the activity of protein phosphatase type 2A but not of phosphatase type 1. We conclude that the inhibition of protein phosphatase 2A is associated to decreased levels of c-Myc/Max heterodimers in K562 cells.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Regulação Neoplásica da Expressão Gênica , Genes myc , Apoptose/efeitos dos fármacos , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Fatores de Transcrição de Zíper de Leucina Básica , Northern Blotting , Linhagem Celular , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Éteres Cíclicos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Cinética , Leucemia Eritroblástica Aguda , Toxinas Marinhas , Ácido Okadáico , Oxazóis/farmacologia , Biossíntese de Proteínas , Proteína Fosfatase 2 , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-myc/biossíntese , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Fatores de Tempo , Fatores de Transcrição/biossíntese , Transcrição Gênica , Células Tumorais Cultivadas
12.
Mol Cell Biol ; 14(1): 200-6, 1994 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8264587

RESUMO

In chicken embryo fibroblasts, phosphorylation of the 40S ribosomal protein S6 increases during G1 but returns to basal level by mitosis. In contrast, in Rous sarcoma virus (RSV)-transformed fibroblasts, S6 remains highly phosphorylated throughout mitosis. This study investigated the mechanism by which RSV alters the pattern of S6 phosphorylation. Pulse-chase experiments demonstrate that phosphate turnover in S6 is rapid in normal cells and in cells infected with an RSV transformation-defective virus. In contrast, phosphate turnover in S6 is severely reduced in cells infected with temperature-sensitive RSV at a temperature permissive for transformation, indicating a diminished S6 phosphatase activity. Fractionation of cell lysates by DEAE chromatography showed an almost threefold lower S6 phosphatase activity in RSV-transformed versus normal cells. The S6 phosphatase was sensitive to inhibitor 2 and specifically recognized by an antibody to type 1 phosphatase (PP1). The S6 phosphatase activity recovered by immunoprecipitation of PP1 was threefold lower in transformed cells, but the steady-state level of expression and the rate of synthesis of PP1 were not altered by oncogenic transformation. Together, the results show that transformation by RSV reduced the S6-PP1 activity.


Assuntos
Transformação Celular Viral , Fibroblastos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Vírus do Sarcoma Aviário , Células Cultivadas , Embrião de Galinha , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Proteína S6 Ribossômica , Proteínas Quinases S6 Ribossômicas , Proteínas Ribossômicas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...