Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Exp Cell Res ; 422(1): 113412, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36370852

RESUMO

The overexpression of BMI1, a polycomb protein, correlates with cancer development and aggressiveness. We previously reported that MYCN-induced BMI1 positively regulated neuroblastoma (NB) cell proliferation via the transcriptional inhibition of tumor suppressors in NB cells. To assess the potential of BMI1 as a new target for NB therapy, we examined the effects of reductions in BMI1 on NB cells. BMI1 knockdown (KD) in NB cells significantly induced their differentiation for up to 7 days. BMI1 depletion significantly induced apoptotic NB cell death for up to 14 days along with the activation of p53, increases in p73, and induction of p53 family downstream molecules and pathways, even in p53 mutant cells. BMI1 depletion in vivo markedly suppressed NB xenograft tumor growth. BMI1 reductions activated ATM and increased γ-H2AX in NB cells. These DNA damage signals and apoptotic cell death were not canceled by the transduction of the polycomb group molecules EZH2 and RING1B. Furthermore, EZH2 and RING1B KD did not induce apoptotic NB cell death to the same extent as BMI1 KD. Collectively, these results suggest the potential of BMI1 as a target of molecular therapy for NB and confirmed, for the first time, the shared role of PcG proteins in the DNA damage response of NB cells.


Assuntos
Neuroblastoma , Proteína Supressora de Tumor p53 , Humanos , Proteínas do Grupo Polycomb/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Neuroblastoma/patologia , Apoptose/genética , Dano ao DNA/genética , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/metabolismo
2.
Biochem Biophys Res Commun ; 529(2): 462-466, 2020 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-32703452

RESUMO

Aging promotes polarization of M2-like macrophages to M1-like macrophages and reduces their phagocytic ability. However, the molecular mechanisms underlying these aging-related changes remain poorly understood. Here, we demonstrate that p53 regulates phagocytic activity in macrophages from young mice but not in those from old ones. Macrophages from both old and young mice expressed functional p53 to induce target genes including p21 and Mdm2. In macrophages from young mice, chemically induced p53 decreased phagocytic activity and c-Myc levels, with the latter change reducing M2-related genes. However, in macrophages from old mice, phagocytic activity and c-Myc expression were independent of p53 activity. Furthermore, c-Myc suppression did not affect M2-related genes in old-mouse macrophages. These results demonstrate that dysregulation of p53 function is a molecular mechanism underlying reduced phagocytic activity in aged-mouse macrophages.


Assuntos
Macrófagos/imunologia , Fagocitose , Proteína Supressora de Tumor p53/imunologia , Envelhecimento , Animais , Células Cultivadas , Ativação de Macrófagos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células RAW 264.7
3.
Biochem Biophys Res Commun ; 518(2): 311-318, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31427086

RESUMO

TAp63 is an isoform of p63 gene, a p53 family gene that suppresses tumorigenesis via transcriptional regulation. TAp63 represses transcription of MYC oncogene in glioblastomas; however, its role in another MYC family gene, MYCN, has remained elusive. In this study, we showed that TAp63 repressed transcription of the MYCN gene in human cancer cells. Overexpression of TAp63 in HeLa cells suppressed MYCN expression, whereas knockdown of TAp63 had the opposite effect. By binding to exon 1 of MYCN gene, TAp63 suppressed the promoter activities of MYCN and its cis-antisense gene, NCYM. Other p53 family members, p53 and TAp73, showed lesser ability to suppress MYCN/NCYM promoter activities compared with that of TAp63. All-trans-retinoic acid (ATRA) treatment of MYCN/NCYM-amplified neuroblastoma CHP134 cells induced TAp63 and reduced p53 expressions, accompanied by downregulation of MYCN/NCYM expressions. Meanwhile, TAp63 knockdown inhibited ATRA-induced repression of NCYM gene expression. Blocking the p53 family binding sites by CRISPR-dCas9 system in CHP134 cells induced MYCN/NCYM expression and promoted apoptotic cell death. Expression levels of TAp63 mRNA inversely correlated with those of MYCN/NCYM expression in primary neuroblastomas, which was associated with a favorable prognosis. Collectively, TAp63 repressed MYCN/NCYM bidirectional transcription, contributing to the suppression of neuroblastoma growth.


Assuntos
Proteína Proto-Oncogênica N-Myc/genética , Proteínas de Neoplasias/genética , Neuroblastoma/genética , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Proliferação de Células/genética , Humanos , Neuroblastoma/metabolismo , Neuroblastoma/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica/genética , Proteínas Supressoras de Tumor/metabolismo
4.
Int Immunopharmacol ; 65: 44-49, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30273916

RESUMO

Exposure to an enriched environment (EE) affects not only brain functions but also immune responses upon viral or bacterial infections. In this study, we examined changes in the phagocytic response and chemokine production of resident peritoneal macrophages after mice had been housed under EE conditions for 6 or 8 weeks, and then explored the possibility that EE could cause a change in the macrophage phenotype by means of flow cytometry as well as quantitative RT-PCR. The percentages of EE macrophages phagocytosing S. aureus and apoptotic neutrophils were significantly larger than those of standard environment (SE) macrophages. After coculturing with S. aureus, EE macrophages tended to produce greater amounts of chemokines such as MIP-2, KC and MCP-1 than SE ones, although the increases for MIP-2 and KC were not statistically significant. As compared with SE macrophages, EE macrophages included more CD40-positive cells (M1 marker), and expressed more mRNAs of IL-6 (M1 marker) and IRF4 (M2 marker), and less mRNA of CD38 (M1 marker), suggesting either the possibility that EE macrophages are a mixed population of M1 and M2 macrophages or the possibility that they are a unique population with a mixed M1 and M2 macrophage phenotype.


Assuntos
Abrigo para Animais , Macrófagos Peritoneais , Animais , Biomarcadores , Quimiocinas/genética , Quimiocinas/metabolismo , Técnicas de Cocultura , Regulação da Expressão Gênica , Macrófagos Peritoneais/classificação , Macrófagos Peritoneais/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos , Fagocitose , Fenótipo , Staphylococcus aureus
5.
PLoS One ; 12(11): e0187927, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29121094

RESUMO

Islet transplantation is a useful cell replacement therapy that can restore the glycometabolic function of severe diabetic patients. It is known that many transplanted islets failed to engraft, and thus, new approaches for overcoming graft loss that may improve the outcome of future clinical islet transplantations are necessary. Pleckstrin homology-like domain family A, member 3 (PHLDA3) is a known suppressor of neuroendocrine tumorigenicity, yet deficiency of this gene increases islet proliferation, prevents islet apoptosis, and improves their insulin-releasing function without causing tumors. In this study, we examined the potential use of PHLDA3-deficient islets in transplantation. We observed that: 1) transplanting PHLDA3-deficient islets into diabetic mice significantly improved their glycometabolic condition, 2) the improved engraftment of PHLDA3-deficient islets resulted from increased cell survival during early transplantation, and 3) Akt activity was elevated in PHLDA3-deficient islets, especially under hypoxic conditions. Thus, we determined that PHLDA3-deficient islets are more resistant against stresses induced by islet isolation and transplantation. We conclude that use of islets with suppressed PHLDA3 expression could be a novel and promising treatment for improving engraftment and consequent glycemic control in islet transplantation.


Assuntos
Diabetes Mellitus Experimental/terapia , Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Proteínas Nucleares/deficiência , Animais , Hipóxia Celular , Sobrevivência Celular , Células Cultivadas , Diabetes Mellitus Experimental/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Transplante das Ilhotas Pancreáticas , Camundongos , Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo
6.
Eur J Cancer ; 50(8): 1555-65, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24559687

RESUMO

Neuroblastoma (NB) is a paediatric solid tumour which originates from sympathetic nervous tissues. Deletions in chromosome 1p are frequently found in unfavourable NBs and are correlated with v-myc avian myelocytomatosis viral oncogene neuroblastoma derived homolog (MYCN) amplification; however, it remains to be elucidated how the 1p loss contributes to MYCN-related oncogenic processes in NB. In this study, we identified the role of Dnmt1-associated protein 1 (DMAP1), coded on chromosome 1p34, in the processes. We studied the expression and function of DMAP1 in NB and found that low-level expression of DMAP1 related to poor prognosis, unfavourable histology and 1p Loss of heterozygosity (LOH) of primary NB samples. Intriguingly, DMAP1 induced ataxia telangiectasia mutated (ATM) phosphorylation and focus formation in the presence of a DNA damage reagent, doxorubicin. By DMAP1 expression in NB and fibroblasts, p53 was activated in an ATM-dependent manner and p53-downstream pro-apoptotic Bcl-2 family molecules were induced at the mRNA level, resulting in p53-induced apoptotic death. BAX and p21(Cip1/Waf1) promoter activity dependent on p53 was clearly up-regulated by DMAP1. Further, MYCN transduction in MYCN single-copy NB cells accelerated doxorubicin (Doxo)-induced apoptotic cell death; MYCN is implicated in DMAP1 protein stabilisation and ATM phosphorylation in these situations. DMAP1 knockdown attenuated MYCN-dependent ATM phosphorylation and NB cell apoptosis. Together, DMAP1 appears to be a new candidate for a 1p tumour suppressor and its reduction contributes to NB tumourigenesis via inhibition of MYCN-related ATM/p53 pathway activation.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Ataxia Telangiectasia/metabolismo , Neuroblastoma/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas/metabolismo , Proteínas Repressoras/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Apoptose , Linhagem Celular Tumoral , Pré-Escolar , Fibroblastos/metabolismo , Humanos , Lactente , Proteína Proto-Oncogênica N-Myc , Fosforilação , Prognóstico , Transdução de Sinais , Análise de Sobrevida
7.
Eur J Cancer ; 46(12): 2324-34, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20591651

RESUMO

HDM2, a human homologue of MDM2, is a major negative regulator of p53 function, and increased expression of HDM2 by its promoter polymorphism SNP309 resulted in p53 inactivation and an increased risk of several tumours, including neuroblastoma (NB). Herein, we show that increased expression of HDM2 is related to a worse prognosis in MYCN-amplified NB patients. HDM2 plays an important role in the expression of Noxa, a pro-apoptotic molecule of the Bcl-2 family, which induces NB cell apoptotic death after doxorubcin (Doxo) treatment. Knockdown of HDM2 by siRNA resulted in the upregulation of Noxa at mRNA/protein levels and improved the sensitivity of Doxo-resistant NB cells, although these were not observed in p53-mutant NB cells. Noxa-knockdown abolished the recovered Doxo-induced cell death by HDM2 reduction. Intriguingly, resistance to Doxo was up-regulated by over-expression of HDM2 in Doxo-sensitive NB cells. By HDM2 expression, p53 was inactivated but its degradation was not accelerated, suggesting that p53 was degraded in a proteasome-independent manner in NB cells; downstream effectors of p53, p21(Cip1/Waf1) and Noxa were suppressed by HDM2. Noxa transcription was considerably regulated by both p53 and p73 in NB cells. Furthermore, in vivo binding of p53 and p73 to Noxa promoter was suppressed and Noxa promoter activation was inhibited by HDM2. Taken together, our results may indicate that the HDM2-related resistance to chemotherapeutic drugs of NB is regulated by p53/p73-dependent Noxa expression in NB.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Neuroblastoma/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Antibióticos Antineoplásicos/uso terapêutico , Apoptose/fisiologia , Doxorrubicina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Neuroblastoma/tratamento farmacológico , RNA Mensageiro/metabolismo , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas , Proteína Tumoral p73 , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA