Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Mol Carcinog ; 57(7): 831-841, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29528145

RESUMO

Polycomb group (PcG) protein BMI1 is an important regulator of oncogenic phenotype and is often overexpressed in several human malignancies including breast cancer. Aberrant expression of BMI1 is associated with metastasis and poor prognosis in cancer patients. At present, therapy reagents that can efficiently inhibit the expression of BMI1 are not very well known. Here, we report that Timosaponin A-III (TA-III), a steroidal saponin obtained from the rhizomes of an herb, Anemarrhena asphodeloides, strongly inhibits expression of BMI1 in breast cancer cells. Treatment of breast cancer cells with TA-III resulted in inhibition of oncogenic phenotypes such as proliferation, migration and invasion, and induction of cellular senescence. Inhibition of these oncogenic phenotypes was accompanied by downregulation of BMI1 expression and histone posttranslational modification activity of PRC1. The mechanistic analysis of TA-III-induced inhibition of oncogenic activity and BMI1 expression suggests that downregulation of c-Myc mediates TA-III effect on BMI1. We further show that exogenous BMI1 overexpression can overcome TA-III-induced inhibition of oncogenic phenotypes. We also show that TA-III induces expression of tumor suppressive miR-200c and miR-141, which are negatively regulated by BMI1. In summary, our data suggest that TA-III is a potent inhibitor of BMI1 and that it can be successfully used to inhibit the growth of tumors where PcG protein BMI1 and PcG activities are upregulated.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Carcinogênese/efeitos dos fármacos , Complexo Repressor Polycomb 1/genética , Proteínas do Grupo Polycomb/genética , Saponinas/farmacologia , Esteroides/farmacologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Senescência Celular/efeitos dos fármacos , Senescência Celular/genética , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Células MCF-7 , MicroRNAs/genética , Oncogenes/genética , Fenótipo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/genética
2.
J Biol Chem ; 290(5): 3033-44, 2015 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-25505268

RESUMO

The polycomb group protein BMI1 is an important regulator of cancer stem cell (CSC) phenotype and is often overexpressed in cancer cells. Its overexpression leads to increase in CSC fraction and therapy resistance in tumors. BMI1 functions via polycomb repressive complex 1 (PRC1)-mediated gene silencing and also via PRC1-independent transcriptional activities. At present, very little is known about the therapy reagents that can efficiently inhibit BMI1 expression, and the CSC phenotype. Here, we report that the polo-like kinase 1 (PLK1) regulates BMI1 expression, and that its inhibition can efficiently down-regulate BMI1 expression and PRC1 activity, and induce premature senescence in breast cancer cells. We also show that the exogenous BMI1 overexpression mitigates anti-oncogenic effects of PLK1 inhibition and overcomes senescence induction by PLK1 inhibitors. We further show that PLK1 inhibition down-regulates BMI1 by upregulating the miRNA-200c/141 cluster, which encodes miR-200c and miR-141, both of which are known to post-transcriptionally downregulate BMI1 expression. Thus, our data suggest that PLK1 inhibitors can be successfully used to inhibit growth of tumors in which PcG protein BMI1 is overexpressed or the PRC1 activity is deregulated.


Assuntos
Proteínas de Ciclo Celular/metabolismo , MicroRNAs/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Western Blotting , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Feminino , Humanos , MicroRNAs/genética , Complexo Repressor Polycomb 1/genética , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Quinase 1 Polo-Like
3.
J Biol Chem ; 290(16): 10555-67, 2015 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-25737447

RESUMO

MicroRNAs (miRNAs) have emerged as important regulators of tumorigenesis. Several miRNAs, which can function either as oncomiRs or tumor suppressive miRs are deregulated in cancer cells. The microRNA-31 (miR-31) has been shown to be overexpressed in metastatic breast cancer. It promotes multiple oncogenic phenotypes, including proliferation, motility, and invasion of cancer cells. Using a breast cancer-related miRNA array analysis, we identified miR-31 as a novel target of histone deacetylase inhibitors (HDACi) in breast cancer cells. Specifically, we show that sodium butyrate (NaB) and panobinostat (LBH589), two broad-spectrum HDAC inhibitors up-regulate hsa-miR-31 (miR-31). The up-regulation of miR-31 was accompanied by repression of the polycomb group (PcG) protein BMI1 and induction of cellular senescence. We further show that inhibition of miR-31 overcomes the senescence-inducing effect of HDACi, and restores expression of the PcG protein BMI1. Interestingly, BMI1 also acts as a repressor of miR-31 transcription, suggesting a cross-negative feedback loop between the expression of miR-31 and BMI1. Our data suggest that miR-31 is an important physiological target of HDACi, and that it is an important regulator of senescence relevant to cancer. These studies further suggest that manipulation of miR-31 expression can be used to modulate senescence-related pathological conditions such as cancer, and the aging process.


Assuntos
Ácido Butírico/farmacologia , Senescência Celular/genética , Regulação Neoplásica da Expressão Gênica , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Indóis/farmacologia , MicroRNAs/genética , Apoptose , Linhagem Celular Tumoral , Senescência Celular/efeitos dos fármacos , Retroalimentação Fisiológica , Feminino , Humanos , Células MCF-7 , MicroRNAs/agonistas , MicroRNAs/metabolismo , Análise em Microsséries , Análise de Sequência com Séries de Oligonucleotídeos , Panobinostat , Complexo Repressor Polycomb 1/antagonistas & inibidores , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/metabolismo , Transdução de Sinais , Transcrição Gênica
4.
J Pathol ; 235(4): 656-67, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25382750

RESUMO

The tumour suppressor p53 plays an important role in tumourigenesis. Besides inducing apoptosis, it regulates cellular senescence, which constitutes an important barrier to tumourigenesis. The mechanism of regulation of cellular senescence by p53 and its downstream pathway are poorly understood. Here, we report that the ubiquitin domain-containing 1 (UBTD1) gene, a new downstream target of p53, induces cellular senescence and acts as a novel tumour suppressor by a mechanism that depends on p53. Expression of UBTD1 increased upon cellular senescence induced by serial passageing of cultures, as well as by exposure to DNA-damageing drugs that induce premature senescence. Over-expression of UBTD1 induces senescence in human fibroblasts and cancer cells and attenuation of the transformed phenotype in cancer cells. UBTD1 is down-regulated in gastric and colorectal cancer tissues, and its lower expression correlates with a more aggressive phenotype and worse prognosis. Multivariate analysis revealed that UBTD1 expression was an independent prognostic factor for gastric cancer patients. Furthermore, UBTD1 increased the stability of p53 protein, by promoting the degradation of Mdm2 protein. Importantly, UBTD1 and p53 function mutually depend on each other in regulating cellular senescence and proliferation. Thus, our data suggest that, upon DNA damage, p53 induction by UBTD1 creates a positive feedback mechanism to further increase p53 expression. Our results establish UBTD1 as a regulator of cellular senescence that mediates p53 function, and provide insights into the mechanism of Mdm2 inhibition that impacts p53 dynamics during cellular senescence and tumourigenesis.


Assuntos
Senescência Celular , Neoplasias Colorretais/enzimologia , Fibroblastos/enzimologia , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Neoplasias Gástricas/enzimologia , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Ubiquitinas/metabolismo , Antineoplásicos/farmacologia , Sítios de Ligação , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Senescência Celular/efeitos dos fármacos , Neoplasias Colorretais/genética , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Dano ao DNA , Relação Dose-Resposta a Droga , Retroalimentação Fisiológica , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Fenótipo , Prognóstico , Regiões Promotoras Genéticas , Estabilidade Proteica , Proteólise , Proteínas Proto-Oncogênicas c-mdm2/genética , Interferência de RNA , Fatores de Risco , Transdução de Sinais , Neoplasias Gástricas/genética , Neoplasias Gástricas/mortalidade , Neoplasias Gástricas/patologia , Fatores de Tempo , Transfecção , Proteína Supressora de Tumor p53/genética , Ubiquitinação , Ubiquitinas/genética
5.
J Biol Chem ; 288(5): 3406-18, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23239878

RESUMO

Polycomb group protein BMI1 plays an important role in cellular homeostasis by maintaining a balance between proliferation and senescence. It is often overexpressed in cancer cells and is required for self-renewal of stem cells. At present, very little is known about the signaling pathways that regulate the expression of BMI1. Here, we report that BMI1 autoactivates its own promoter via an E-box present in its promoter. We show that BMI1 acts as an activator of the WNT pathway by repressing Dickkopf (DKK) family of WNT inhibitors. BMI1 mediated repression of DKK proteins; in particular, DKK1 led to up-regulation of WNT target c-Myc, which in turn further led to transcriptional autoactivation of BMI1. Thus, a positive feedback loop connected by the WNT signaling pathway regulates BMI1 expression. This positive feedback loop regulating BMI1 expression may be relevant to the role of BMI1 in promoting cancer and maintaining stem cell phenotype.


Assuntos
Retroalimentação Fisiológica , Regulação Neoplásica da Expressão Gênica , Complexo Repressor Polycomb 1/genética , Proteínas do Grupo Polycomb/genética , Via de Sinalização Wnt , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Modelos Biológicos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Fenótipo , Complexo Repressor Polycomb 1/metabolismo , Proteínas do Grupo Polycomb/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica/genética , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transcrição Gênica , Regulação para Cima/genética , Via de Sinalização Wnt/genética
6.
Methods Cell Biol ; 181: 59-72, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38302244

RESUMO

Cellular senescence, whereby cells cease to proliferate, is known to contribute to the aging process and age-related pathologies. It is elicited either by cell-intrinsic mechanisms such as progressive telomere shortening or due to the extrinsic stress-related factors, which via p53-p21 and p16-pRB tumor suppressor pathways signal cells to cease proliferation. A proper identification and characterization of senescent cells is necessary to understand the process of aging, age-related pathologies, and the development of therapeutics to treat age-related dysfunctions. The landmark discovery of Senescence-Associated-Beta-Galactosidase (SA-ß-Gal) marker, and a simple colorimetric method to detect SA-ß-Gal greatly facilitated identification of the senescent cells in human and rodent cells pertaining to age-related diseases (Dimri et al., 1995). Despite the availability of additional senescence biomarkers, the SA-ß-Gal marker and histochemical detection method remain the most widely used tool to identify senescent cells in vitro and in vivo. Here, we revisit the original colorimetric method to detect senescent cells that was first published in 1995 (Dimri et al., 1995).


Assuntos
Senescência Celular , Colorimetria , Humanos , Senescência Celular/genética , Envelhecimento/metabolismo , Biomarcadores/metabolismo , Transdução de Sinais
7.
Cancer Cell ; 7(6): 505-12, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15950900

RESUMO

Cancer therapeutics are primarily thought to work by inducing apoptosis in tumor cells. However, various tumor suppressors and oncogenes have been shown to regulate senescence in normal cells, and senescence bypass appears to be an important step in the development of cancer. Cellular senescence limits the replicative capacity of cells, thus preventing the proliferation of cells that are at different stages of malignancy. A recent body of evidence suggests that induction of senescence can be exploited as a basis for cancer therapy.


Assuntos
Antineoplásicos/uso terapêutico , Senescência Celular/fisiologia , Neoplasias/tratamento farmacológico , Antineoplásicos/farmacologia , Senescência Celular/efeitos dos fármacos , Humanos , Modelos Biológicos , Proteínas de Neoplasias/fisiologia , Neoplasias/fisiopatologia , Proteínas Nucleares/fisiologia , Proteína da Leucemia Promielocítica , Proteína do Retinoblastoma/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Telômero/fisiologia , Fatores de Transcrição/fisiologia , Proteína Supressora de Tumor p14ARF/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Proteínas Supressoras de Tumor/fisiologia
8.
Carcinogenesis ; 31(3): 489-95, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19969553

RESUMO

The polycomb group (PcG) protein, enhancer of zeste homologue 2 (EZH2), is overexpressed in several human malignancies including breast cancer. Aberrant expression of EZH2 has been associated with metastasis and poor prognosis in cancer patients. Despite the clear role of EZH2 in oncogenesis and therapy failure, not much is known about chemotherapeutics and chemopreventive agents that can suppress its expression and activity. Here, we show that dietary omega-3 (omega-3) polyunsaturated fatty acids (PUFAs) can regulate the expression of EZH2 in breast cancer cells. The treatment of breast cancer cells with omega-3 PUFAs, but not omega-6 PUFAs, led to downregulation of EZH2. Studies using proteosome inhibitor MG132 suggested that omega-3 PUFAs induce degradation of the PcG protein EZH2 through posttranslational mechanisms. Furthermore, downregulation of EZH2 by omega-3 PUFAs was accompanied by a decrease in histone 3 lysine 27 trimethylation (H3K27me3) activity of EZH2 and upregulation of E-cadherin and insulin-like growth factor binding protein 3, which are known targets of EZH2. Treatment with omega-3 PUFAs also led to decrease in invasion of breast cancer cells, an oncogenic phenotype that is known to be associated with EZH2. Thus, our studies suggest that the PcG protein EZH2 is an important target of omega-3 PUFAs and that downregulation of EZH2 may be involved in the mediation of anti-oncogenic and chemopreventive effects of omega-3 PUFAs.


Assuntos
Anticarcinógenos/farmacologia , Ácido Araquidônico/farmacologia , Neoplasias da Mama/patologia , Proteínas de Ligação a DNA/biossíntese , Gorduras na Dieta/farmacologia , Ácidos Docosa-Hexaenoicos/farmacologia , Ácido Eicosapentaenoico/farmacologia , Ácido Linoleico/farmacologia , Proteínas de Neoplasias/biossíntese , Fatores de Transcrição/biossíntese , Caderinas/biossíntese , Caderinas/genética , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/metabolismo , Proteínas de Ligação a DNA/genética , Regulação para Baixo/efeitos dos fármacos , Proteína Potenciadora do Homólogo 2 de Zeste , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Histonas/metabolismo , Humanos , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/biossíntese , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/genética , Metilação/efeitos dos fármacos , Invasividade Neoplásica , Proteínas de Neoplasias/genética , Complexo Repressor Polycomb 2 , Complexo de Endopeptidases do Proteassoma/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Fatores de Transcrição/genética
9.
Mol Cancer ; 9: 158, 2010 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-20569464

RESUMO

BACKGROUND: The polycomb group (PcG) protein BMI1 is an important regulator of development. Additionally, aberrant expression of BMI1 has been linked to cancer stem cell phenotype and oncogenesis. In particular, its overexpression has been found in several human malignancies including breast cancer. Despite its established role in stem cell maintenance, cancer and development, at present not much is known about the functional domains of BMI1 oncoprotein. In the present study, we carried out a deletion analysis of BMI1 to identify its negative regulatory domain. RESULTS: We report that deletion of the C-terminal domain of BMI1, which is rich in proline-serine (PS) residues and previously described as PEST-like domain, increased the stability of BMI1, and promoted its pro-oncogenic activities in human mammary epithelial cells (HMECs). Specifically, overexpression of a PS region deleted mutant of BMI1 increased proliferation of HMECs and promoted an epithelial-mesenchymal transition (EMT) phenotype in the HMECs. Furthermore, when compared to the wild type BMI1, exogenous expression of the mutant BMI1 led to a significant downregulation of p16INK4a and an efficient bypass of cellular senescence in human diploid fibroblasts. CONCLUSIONS: In summary, our data suggest that the PS domain of BMI1 is involved in its stability and that it negatively regulates function of BMI1 oncoprotein. Our results also suggest that the PS domain of BMI1 could be targeted for the treatment of proliferative disorders such as cancer and aging.


Assuntos
Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas/genética , Sequências Reguladoras de Ácido Nucleico , Proteínas Repressoras/genética , Linhagem Celular , Transição Epitelial-Mesenquimal , Meia-Vida , Humanos , Proteínas Nucleares/química , Proteínas Nucleares/fisiologia , Complexo Repressor Polycomb 1 , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Repressoras/química , Proteínas Repressoras/fisiologia , Deleção de Sequência
10.
Mol Cancer ; 9: 40, 2010 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-20170541

RESUMO

BACKGROUND: The BMI1 oncogene is overexpressed in several human malignancies including gastric cancer. In addition to BMI1, mammalian cells also express Mel-18, which is closely related to BMI1. We have reported that Mel-18 functions as a potential tumor suppressor by repressing the expression of BMI1 and consequent downregulation of activated AKT in breast cancer cells. However, the mechanisms of BMI1 overexpression and the role of Mel-18 in other cancers are still not clear. The purpose of this study is to investigate the role of BMI1 and Mel-18 in gastric cancer. RESULTS: BMI1 was found to be overexpressed in gastric cancer cell lines and gastric tumors. Overexpression of BMI1 correlated with advanced clinical stage and lymph node metastasis; while the expression of Mel-18 negatively correlated with BMI1. BMI1 but not Mel-18 was found to be an independent prognostic factor. Downregulation of BMI1 by Mel-18 overexpression or knockdown of BMI1 expression in gastric cancer cell lines led to upregulation of p16 (p16INK4a or CDKN2A) in p16 positive cell lines and reduction of phospho-AKT in both p16-positive and p16-negative cell lines. Downregulation of BMI1 was also accompanied by decreased transformed phenotype and migration in both p16- positive and p16-negative gastric cancer cell lines. CONCLUSIONS: In the context of gastric cancer, BMI1 acts as an oncogene and Mel-18 functions as a tumor suppressor via downregulation of BMI1. Mel-18 and BMI1 may regulate tumorigenesis, cell migration and cancer metastasis via both p16- and AKT-dependent growth regulatory pathways.


Assuntos
Progressão da Doença , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/metabolismo , Neoplasias Gástricas/patologia , Idoso , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Senescência Celular , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Regulação para Baixo/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Nucleares/genética , Fosforilação , Complexo Repressor Polycomb 1 , Prognóstico , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Repressoras/genética , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/enzimologia , Neoplasias Gástricas/genética
11.
Mol Biol Cell ; 18(2): 536-46, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17151361

RESUMO

Polycomb group (PcG) protein Bmi-1 is an important regulator of cell proliferation. It regulates cellular senescence and proliferation of cells via the transcriptional repression of INK4a/ARF locus and other target genes. Here, we report that Mel-18, a PcG ring finger protein (PCGF) transcriptionally down-regulates Bmi-1. Furthermore, the expression of Bmi-1 and Mel-18 inversely correlates in proliferating and senescent human fibroblasts. Bmi-1 down-regulation by Mel-18 results in accelerated senescence and shortening of the replicative life span in normal human cells. Importantly, using promoter-reporter, chromatin immunoprecipitation, and quantitative real-time primary transcript RT-PCR assays, and an RNA interference approach, we demonstrate that Bmi-1 is a bona fide target of c-Myc oncoprotein. Finally, our data suggest that Mel-18 regulates Bmi-1 expression during senescence via down-regulation of c-Myc. These studies link c-Myc and polycomb function in cell proliferation and senescence.


Assuntos
Proliferação de Células , Senescência Celular/genética , Regulação da Expressão Gênica , Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Sítios de Ligação , Células Cultivadas , Imunoprecipitação da Cromatina , Regulação para Baixo , Genes Reporter , Humanos , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Complexo Repressor Polycomb 1 , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Interferência de RNA , Proteínas Repressoras/antagonistas & inibidores
12.
Cancer Res ; 67(11): 5083-9, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17545584

RESUMO

The Bmi-1 oncogene is overexpressed in a number of malignancies including breast cancer. In addition to Bmi-1, mammalian cells also express four other polycomb group (PcG) proteins that are closely related to Bmi-1. Virtually nothing is known about the role of these PcG proteins in oncogenesis. We have recently reported that Mel-18, a Bmi-1-related PcG protein, negatively regulates Bmi-1 expression, and that its expression negatively correlates with Bmi-1 in proliferating and senescing human fibroblasts. Here, we report that the expression of Bmi-1 and Mel-18 inversely correlates in a number of breast cancer cell lines and in a significant number of breast tumor samples. Overexpression of Mel-18 results in repression of Bmi-1 and reduction of the transformed phenotype in malignant breast cancer cells. Furthermore, the repression of Bmi-1 by Mel-18 is accompanied by the reduction of Akt/protein kinase B (PKB) activity in breast cancer cells. Similarly, Bmi-1 knockdown using RNA interference approach results in down-regulation of Akt/PKB activity and reduction in transformed phenotype of MCF7 cells. Importantly, we show that overexpression of constitutively active Akt overrides tumor-suppressive effect of Mel-18 overexpression and the knockdown of Bmi-1 expression. Thus, our studies suggest that Mel-18 and Bmi-1 may regulate the Akt pathway in breast cancer cells, and that Mel-18 functions as a tumor suppressor by repressing the expression of Bmi-1 and consequently down-regulating Akt activity.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas de Ligação a DNA/biossíntese , Proteínas Nucleares/biossíntese , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Repressoras/biossíntese , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Adesão Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Proteínas de Ligação a DNA/genética , Regulação para Baixo , Genes Supressores de Tumor , Humanos , Proteínas Nucleares/genética , Complexo Repressor Polycomb 1 , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-akt/biossíntese , Proteínas Proto-Oncogênicas c-akt/genética , Interferência de RNA , Proteínas Repressoras/genética , Transfecção
13.
Cancer Res ; 67(9): 4164-72, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17483327

RESUMO

Epidermal growth factor receptor (EGFR), a member of the ErbB family of receptor tyrosine kinases, is overexpressed in as many as 60% cases of breast and other cancers. EGFR overexpression is a characteristic of highly aggressive molecular subtypes of breast cancer with basal-like and BRCA1 mutant phenotypes distinct from ErbB2-overexpressing breast cancers. Yet, EGFR is substantially weaker compared with ErbB2 in promoting the oncogenic transformation of nontumorigenic human mammary epithelial cells (human MEC), suggesting a role for cooperating oncogenes. Here, we have modeled the co-overexpression of EGFR and a biologically and clinically relevant potential modifier c-Src in two distinct immortal but nontumorigenic human MECs. Using a combination of morphologic analysis and confocal imaging of polarity markers in three-dimensional Matrigel culture together with functional analyses of early oncogenic traits, we show for the first time that EGFR and c-Src co-overexpression but not EGFR or c-Src overexpression alone unleashes an oncogenic signaling program that leads to hyperproliferation and loss of polarity in three-dimensional acinar cultures, marked enhancement of migratory and invasive behavior, and anchorage-independent growth. Our results establish that EGFR overexpression in an appropriate context (modeled here using c-Src overexpression) can initiate oncogenic transformation of nontumorigenic human MECs and provide a suitable in vitro model to interrogate human breast cancer-relevant oncogenic signaling pathways initiated by overexpressed EGFR and to identify modifiers of EGFR-mediated breast oncogenesis.


Assuntos
Neoplasias da Mama/enzimologia , Transformação Celular Neoplásica/metabolismo , Receptores ErbB/biossíntese , Proteínas Tirosina Quinases/biossíntese , Animais , Neoplasias da Mama/patologia , Proteína Tirosina Quinase CSK , Adesão Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Transformação Celular Neoplásica/patologia , Colágeno , Combinação de Medicamentos , Células Epiteliais/enzimologia , Células Epiteliais/patologia , Feminino , Humanos , Laminina , Camundongos , Camundongos Nus , Proteoglicanas , Células Tumorais Cultivadas , Quinases da Família src
14.
Cancer Res ; 66(12): 6225-32, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16778197

RESUMO

The Bmi-1 oncoprotein regulates proliferation and oncogenesis in human cells. Its overexpression leads to senescence bypass in human fibroblasts and immortalization of human mammary epithelial cells. In this study, we report that compared with normal nasopharyngeal epithelial cells (NPEC), Bmi-1 is overexpressed in nasopharyngeal carcinoma cell lines. Importantly, Bmi-1 was also found to be overexpressed in 29 of 75 nasopharyngeal carcinoma tumors (38.7%) by immunohistochemical analysis. In contrast to nasopharyngeal carcinoma, there was no detectable expression of Bmi-1 in noncancerous nasopharyngeal epithelium. Moreover, high Bmi-1 expression positively correlated with poor prognosis of nasopharyngeal carcinoma patients. We also report that the overexpression of Bmi-1 leads to bypass of senescence and immortalization of NPECs, which normally express p16(INK4a) and exhibit finite replicative life span. Overexpression of Bmi-1 in NPECs led to the induction of human telomerase reverse transcriptase activity and reduction of p16(INK4a) expression. Mutational analysis of Bmi-1 showed that both RING finger and helix-turn-helix domains of it are required for immortalization of NPECs. Our findings suggest that Bmi-1 plays an important role in the development and progression of nasopharyngeal carcinoma, and that Bmi-1 is a valuable marker for assessing the prognosis of nasopharyngeal carcinoma patients. Furthermore, this study provides the first cellular proto-oncogene immortalized nasopharyngeal epithelial cell line, which may serve as a cell model system for studying the mechanisms involved in the tumorigenesis of nasopharyngeal carcinoma.


Assuntos
Biomarcadores Tumorais/biossíntese , Neoplasias Nasofaríngeas/metabolismo , Proteínas Nucleares/biossíntese , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Repressoras/biossíntese , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Inibidor p16 de Quinase Dependente de Ciclina/biossíntese , Inibidor p16 de Quinase Dependente de Ciclina/genética , Dano ao DNA , Progressão da Doença , Regulação para Baixo , Células Epiteliais/enzimologia , Células Epiteliais/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Neoplasias Nasofaríngeas/enzimologia , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patologia , Estadiamento de Neoplasias , Proteínas Nucleares/genética , Complexo Repressor Polycomb 1 , Prognóstico , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas Repressoras/genética , Telomerase/metabolismo
15.
J Hematol Oncol ; 11(1): 17, 2018 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-29422082

RESUMO

BACKGROUND: Chromobox protein homolog 7 (CBX7), a member of the polycomb group (PcG) family of proteins, is involved in the regulation of cell proliferation and cancer progression. PcG family members, such as BMI, Mel-18, and EZH2, are integral constituents of the polycomb repressive complexes (PRCs) and have been known to regulate cancer stem cell (CSC) phenotype. However, the role of other PRCs' constituents such as CBX7 in the regulation of CSC phenotype remains largely elusive. This study was to investigate the role of CBX7 in regulating stem cell-like properties of gastric cancer and the underlying mechanisms. METHODS: Firstly, the role of CBX7 in regulating stem cell-like properties of gastric cancer was investigated using sphere formation, Western blot, and xenograft tumor assays. Next, RNA interference and ectopic CBX7 expression were employed to determine the impact of CBX7 on the expression of CSC marker proteins and CSC characteristics. The expression of CBX7, its downstream targets, and stem cell markers were analyzed in gastric stem cell spheres, common cancer cells, and gastric cancer tissues. Finally, the pathways by which CBX7 regulates stem cell-like properties of gastric cancer were explored. RESULTS: We found that CBX7, a constituent of the polycomb repressive complex 1 (PRC1), plays an important role in maintaining stem cell-like characteristics of gastric cancer cells via the activation of AKT pathway and the downregulation of p16. Spearman rank correlation analysis showed positive correlations among the expression of CBX7 and phospho-AKT (pAKT), stem cell markers OCT-4, and CD133 in gastric cancer tissues. In addition, CBX7 was found to upregulate microRNA-21 (miR-21) via the activation of AKT-NF-κB pathway, and miR-21 contributes to CBX7-mediated CSC characteristics. CONCLUSIONS: CBX7 positively regulates stem cell-like characteristics of gastric cancer cells by inhibiting p16 and activating AKT-NF-κB-miR-21 pathway.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/genética , Regulação Neoplásica da Expressão Gênica , Células-Tronco Neoplásicas/patologia , Complexo Repressor Polycomb 1/metabolismo , Transdução de Sinais , Neoplasias Gástricas/patologia , Linhagem Celular Tumoral , Humanos , MicroRNAs/genética , NF-kappa B/metabolismo , Células-Tronco Neoplásicas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo
16.
Mol Cell Biol ; 23(1): 389-401, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12482990

RESUMO

The polycomb protein Bmi-1 represses the INK4a locus, which encodes the tumor suppressors p16 and p14(ARF). Here we report that Bmi-1 is downregulated when WI-38 human fibroblasts undergo replicative senescence, but not quiescence, and extends replicative life span when overexpressed. Life span extension by Bmi-1 required the pRb, but not p53, tumor suppressor protein. Deletion analysis showed that the RING finger and helix-turn-helix domains of Bmi-1 were required for life span extension and suppression of p16. Furthermore, a RING finger deletion mutant exhibited dominant negative activity, inducing p16 and premature senescence. Interestingly, presenescent cultures of some, but not all, human fibroblasts contained growth-arrested cells expressing high levels of p16 and apparently arrested by a p53- and telomere-independent mechanism. Bmi-1 selectively extended the life span of these cultures. Low O(2) concentrations had no effect on p16 levels or life span extension by Bmi-1 but reduced expression of the p53 target, p21. We propose that some human fibroblast strains are more sensitive to stress-induced senescence and have both p16-dependent and p53/telomere-dependent pathways of senescence. Our data suggest that Bmi-1 extends the replicative life span of human fibroblasts by suppressing the p16-dependent senescence pathway.


Assuntos
Senescência Celular/fisiologia , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Fibroblastos/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Divisão Celular/genética , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina/genética , Replicação do DNA , Fibroblastos/citologia , Sequências Hélice-Volta-Hélice , Humanos , Mutação , Proteínas Nucleares/genética , Oxigênio/metabolismo , Complexo Repressor Polycomb 1 , Proteínas do Grupo Polycomb , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Telomerase/genética , Telomerase/metabolismo , Telômero/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
17.
Methods Mol Biol ; 371: 21-31, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17634571

RESUMO

Most normal human cells undergo cellular senescence after accruing a fixed number of cell divisions, or are challenged by a variety of potentially oncogenic stimuli, in culture and most likely in vivo. Cellular senescence is characterized by an irreversible growth arrest and certain altered functions. Senescent cells in culture are identified by their inability to undergo DNA synthesis, a property also shared by quiescent cells. Several years ago, we described a biomarker associated with the senescent phenotype, a senescence associated beta-galactosidase (SA-beta-gal), which is detected by histochemical staining of cells using the artificial substrate X-gal. The presence of the SA-beta-gal biomarker is independent of DNA synthesis and generally distinguishes senescent cells from quiescent cells. The method to detect SA-beta-gal is a convenient, single cell-based assay, which can identify senescent cells even in heterogeneous cell populations and aging tissues, such as skin biopsies from older individuals. Because it is easy to detect, SA-beta-gal is currently a widely used biomarker of senescence. Here we describe a method to detect SA-beta-gal in detail, including some recent modifications.


Assuntos
Envelhecimento/metabolismo , Senescência Celular/fisiologia , Fibroblastos/enzimologia , Pele/enzimologia , beta-Galactosidase/metabolismo , Biomarcadores/análise , Biomarcadores/metabolismo , Linhagem Celular , DNA/biossíntese , Fibroblastos/citologia , Galactosídeos/química , Humanos , Indóis/química , beta-Galactosidase/análise
18.
Cancer Res ; 62(16): 4736-45, 2002 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12183433

RESUMO

The vast majority of breast cancers are carcinomas that arise from mammary epithelial cells (MECs). One of the key early events in tumorigenic transformation is the ability of cells to overcome replicative senescence. However, the precise genetic changes that are responsible for this event in MECs is largely unknown. Here, we report that Bmi-1, originally identified as a c-Myc cooperating oncoprotein, can bypass senescence, extend the replicative life span, and immortalize MECs. Furthermore, Bmi-1 was overexpressed in immortal MECs and several breast cancer cell lines. Overexpression of Bmi-1 in MECs led to activation of human telomerase reverse transcriptase (hTERT) transcription and induction of telomerase activity. Telomerase induction by Bmi-1 was an early event in the extension of the replicative life span and immortalization. Bmi-1 was not overexpressed in hTERT-immortalized MECs, suggesting that Bmi-1 functions upstream of hTERT. Although, c-Myc has been reported to induce telomerase in MECs, Bmi-1 appeared to act independently of c-Myc binding sequences in the hTERT promoter. Deletion analysis of the Bmi-1 protein suggested that the RING finger, as well as a conserved helix-turn-helix-turn domain, were required for its ability to induce telomerase and immortalize MECs. These data suggest that Bmi-1 regulates telomerase expression in MECs and plays a role in the development of human breast cancer.


Assuntos
Mama/fisiologia , Transformação Celular Neoplásica/genética , Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Repressoras , Telomerase/metabolismo , Sítios de Ligação , Mama/citologia , Mama/enzimologia , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Transformação Celular Neoplásica/metabolismo , Células Cultivadas , Proteínas de Ligação a DNA , Regulação para Baixo , Indução Enzimática , Células Epiteliais/citologia , Células Epiteliais/enzimologia , Células Epiteliais/fisiologia , Humanos , Proteínas Nucleares/biossíntese , Proteínas Nucleares/fisiologia , Complexo Repressor Polycomb 1 , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Telomerase/biossíntese , Telomerase/genética , Ativação Transcricional , Células Tumorais Cultivadas , Proteína Supressora de Tumor p14ARF/biossíntese , Proteína Supressora de Tumor p14ARF/genética
19.
Oncotarget ; 7(24): 36220-36234, 2016 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-27105531

RESUMO

MicroRNAs (miRNAs) are known to function as oncomiRs or tumor suppressors and are important noncoding RNA regulators of oncogenesis. The miR-200c/141 locus on chromosome 12 encodes miR-200c and miR-141, two members of the miR-200 family, which have been shown to function as tumor suppressive miRNAs by targeting multiple oncogenic factors such as polycomb group protein BMI1. Here, we show that BMI1 reciprocally functions as a transcriptional repressor of the miR-200c/141 cluster and that BMI1 inhibitors upregulate expression of miR-200c and miR-141. Our data suggest that BMI1 binds to the miR-200c/141 promoter and regulates it through transcription factor binding motifs E-box 2 and Z-box 1 to repress expression of miR-200c/141 cluster. We also show that PTC-209, a small molecule inhibitor of BMI1 gene expression induces cellular senescence and transcriptionally upregulates expression of miR-200c/141 cluster in breast cancer cells. Furthermore, inhibition of expression of miR-200c or miR-141 overcomes tumor suppressive effects of PTC-209 including induction of cellular senescence and downregulation of breast cancer stem cell phenotype. Therefore, our studies suggest a reciprocal regulation between BMI1 and miR-200c/141 cluster, and that BMI1 inhibitory drugs can further amplify their inhibitory effects on BMI1 via multiple mechanisms including posttranscriptional regulation by upregulating BMI1 targeting miRNAs.


Assuntos
Regulação Neoplásica da Expressão Gênica/genética , MicroRNAs/genética , Complexo Repressor Polycomb 1/genética , Linhagem Celular , Linhagem Celular Tumoral , Senescência Celular/efeitos dos fármacos , Senescência Celular/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Compostos Heterocíclicos com 2 Anéis/farmacologia , Humanos , Células MCF-7 , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica , Tiazóis/farmacologia
20.
Sci Aging Knowledge Environ ; 2004(44): pe40, 2004 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-15525836

RESUMO

Although several biomarkers of aging have been described in the literature, it is only recently that gerontologists have started to search for molecular biomarkers of aging. A gene or a set of genes that are expressed in a wide range of tissues and exhibit an age-dependent, easily quantifiable increase in their expression represent a possible molecular biomarker of aging. Because the physiology of an organism is profoundly affected by the pattern of gene expression, it is hoped that molecular biomarkers of aging will more accurately predict the physiological age of an organism than the chronological age. A recent report from Sharpless's laboratory examines the possibility that the tumor suppressors p16 and ARF (encoded by the INK4a/ARF locus) represent molecular biomarkers of aging in rodent models.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Vermelho Congo/análogos & derivados , Chaperonas Moleculares/metabolismo , Vermelho Congo/farmacologia , Piperidinas/farmacologia , Proteínas de Ligação a Tacrolimo/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA