Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
1.
Nucleic Acids Res ; 46(12): 6188-6205, 2018 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-29873780

RESUMO

p53 as an effector of nucleolar stress is well defined, but p53 independent mechanisms are largely unknown. Like p53, the NF-κB transcription factor plays a critical role in maintaining cellular homeostasis under stress. Many stresses that stimulate NF-κB also disrupt nucleoli. However, the link between nucleolar function and activation of the NF-κB pathway is as yet unknown. Here we demonstrate that artificial disruption of the PolI complex stimulates NF-κB signalling. Unlike p53 nucleolar stress response, this effect does not appear to be linked to inhibition of rDNA transcription. We show that specific stress stimuli of NF-κB induce degradation of a critical component of the PolI complex, TIF-IA. This degradation precedes activation of NF-κB and is associated with increased nucleolar size. It is mimicked by CDK4 inhibition and is dependent upon a novel pathway involving UBF/p14ARF and S44 of the protein. We show that blocking TIF-IA degradation blocks stress effects on nucleolar size and NF-κB signalling. Finally, using ex vivo culture, we show a strong correlation between degradation of TIF-IA and activation of NF-κB in freshly resected, human colorectal tumours exposed to the chemopreventative agent, aspirin. Together, our study provides compelling evidence for a new, TIF-IA-NF-κB nucleolar stress response pathway that has in vivo relevance and therapeutic implications.


Assuntos
Nucléolo Celular/metabolismo , NF-kappa B/metabolismo , Proteínas Pol1 do Complexo de Iniciação de Transcrição/metabolismo , Estresse Fisiológico , Transporte Ativo do Núcleo Celular , Linhagem Celular , Linhagem Celular Tumoral , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Humanos , Proteínas Pol1 do Complexo de Iniciação de Transcrição/química , RNA Polimerase I/metabolismo , Serina/metabolismo , Transdução de Sinais , Fator de Transcrição RelA/metabolismo , Proteína Supressora de Tumor p14ARF/fisiologia
2.
Mol Cells ; 41(5): 381-389, 2018 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-29665672

RESUMO

ARF is a tumor suppressor protein that has a pivotal role in the prevention of cancer development through regulating cell proliferation, senescence, and apoptosis. As a factor that induces senescence, the role of ARF as a tumor suppressor is closely linked to the p53-MDM2 axis, which is a key process that restrains tumor formation. Thus, many cancer cells either lack a functional ARF or p53, which enables them to evade cell oncogenic stress-mediated cycle arrest, senescence, or apoptosis. In particular, the ARF gene is a frequent target of genetic and epigenetic alterations including promoter hyper-methylation or gene deletion. However, as many cancer cells still express ARF, pathways that negatively modulate transcriptional or post-translational regulation of ARF could be potentially important means for cancer cells to induce cellular proliferation. These recent findings of regulators affecting ARF protein stability along with its low levels in numerous human cancers indicate the significance of an ARF post-translational mechanism in cancers. Novel findings of regulators stimulating or suppressing ARF function would provide new therapeutic targets to manage cancer- and senescence-related diseases. In this review, we present the current knowledge on the regulation and alterations of ARF expression in human cancers, and indicate the importance of regulators of ARF as a prognostic marker and in potential therapeutic strategies.


Assuntos
Transformação Celular Neoplásica/genética , Inibidor de Quinase Dependente de Ciclina p18/fisiologia , Proteínas de Neoplasias/fisiologia , Proteína Supressora de Tumor p14ARF/fisiologia , Apoptose , Divisão Celular , Senescência Celular , Inibidor p16 de Quinase Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p18/deficiência , Inibidor de Quinase Dependente de Ciclina p18/genética , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Genes p16 , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Processamento de Proteína Pós-Traducional , Estabilidade Proteica , Proteínas Proto-Oncogênicas c-mdm2/fisiologia , Proteína Supressora de Tumor p14ARF/deficiência , Proteína Supressora de Tumor p14ARF/genética , Proteína Supressora de Tumor p53/fisiologia , Ubiquitinação
3.
FEBS J ; 285(5): 832-847, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29283500

RESUMO

The tumor suppressor p14arf interacts, in response to oncogenic signals, with the p53 E3-ubiquitin ligase HDM2, thereby resulting in p53 stabilization and activation. In addition, it also exerts tumor-suppressive functions in p53-independent contexts. The activities of p14arf are regulated by the nucleolar chaperone nucleophosmin (NPM1), which controls its levels and cellular localization. In acute myeloid leukemia with mutations in the NPM1 gene, mutated NPM1 aberrantly translocates in the cytosol carrying with itself p14arf that is subsequently degraded, thus impairing the p14arf-HDM2-p53 axis. In this work we investigated the complex between these two proteins by means of NMR and other techniques. We identified a novel NPM1-interacting motif in the C-terminal region of p14arf, which corresponds to its predicted nucleolar localization signal. This motif recognizes a specific region of the NPM1 N-terminal domain and, upon binding, the two proteins form soluble high molecular weight complexes. By NMR, we identified critical residues on both proteins involved in the interaction. Collectively, our data provide a structural framework to rationalize the overall assembly of the p14arf-NPM1 supramolecular complexes. A number of p14arf cancer-associated mutations cluster in this motif and their effect on the interaction with NPM1 was also analyzed.


Assuntos
Proteínas Nucleares/química , Proteína Supressora de Tumor p14ARF/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Camundongos , Modelos Moleculares , Terapia de Alvo Molecular , Mutação , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Ressonância Magnética Nuclear Biomolecular , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Nucleofosmina , Agregados Proteicos , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Proteínas Recombinantes de Fusão/química , Alinhamento de Sequência , Deleção de Sequência , Homologia de Sequência de Aminoácidos , Espectrometria de Fluorescência , Proteína Supressora de Tumor p14ARF/genética , Proteína Supressora de Tumor p14ARF/fisiologia
4.
Cancer Res ; 77(4): 1035-1046, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28082400

RESUMO

Muscle-invasive bladder cancer (MIBC) generally responds poorly to treatment and tends to exhibit significant mortality. Here we show that expression of the tumor suppressor p14ARF (ARF) is upregulated in aggressive subtypes of MIBC. Accumulation of ARF in the nucleolus is associated with poor outcome and attenuated response to chemotherapy. In both genetically engineered mouse models and murine xenograft models of human MIBC, we demonstrate that tumors expressing ARF failed to respond to treatment with the platinum-based chemotherapy agent cisplatin. Resistance was mediated in part by the integrin-binding protein ITGB3BP (CENPR) and reflected ARF-dependent impairment of protein translation, which was exaggerated by drug treatment. Overall, our results highlight a context-dependent role for ARF in modulating the drug response of bladder cancer. Cancer Res; 77(4); 1035-46. ©2017 AACR.


Assuntos
Proteína Supressora de Tumor p14ARF/fisiologia , Neoplasias da Bexiga Urinária/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Linhagem Celular Tumoral , Cisplatino/uso terapêutico , Dano ao DNA , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Invasividade Neoplásica , Proteínas Nucleares/fisiologia , Proteína Supressora de Tumor p14ARF/análise , Proteína Supressora de Tumor p53/fisiologia , Neoplasias da Bexiga Urinária/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Oncotarget ; 7(30): 47609-47619, 2016 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-27356744

RESUMO

ARF couples with TP53 in a canonical signaling pathway to activate cellular senescence for tumor suppressive function under oncogenic insults. However, the mechanisms on aberrant elevation of ARF in cancers are still poorly understood. We previously showed that ARF (p14ARF in human and p19Arf in mouse) elevation correlates with PTEN loss and stabilizes SLUG to reduce cell adhesion in prostate cancer (PCa). Here we report that ARF is essential for MMP7 expression, E-Cadherin decrease and the anchorage loss to the extracellular matrix (ECM) in PCa in vitro and in vivo. We found that Mmp7 is aberrantly elevated in cytosol and nucleus of malignant prostate tumors of Pten/Trp53 mutant mice. Interestingly, p19Arf deficiency strikingly decreases Mmp7 levels but increases E-Cadherin in Pten/Trp53/p19Arf mice. ARF knockdown markedly reduces MMP7 in human PCa cells. Conversely, tetracycline-inducible expression of ARF increases MMP7 with a decrease of E-Cadherin in PCa cells. Importantly, MMP7 physically binds ARF to show the co-localization in nucleus. Co-expression of MMP7 and ARF promotes cell migration, and MMP7 knockdown decreases wound healing in PCa cells. Furthermore, MMP7 elevation correlates with ARF expression in advanced human PCa. Our findings reveal for the first time that the crosstalk between ARF and MMP7 in nucleus contributes to ECM network in tumor microenvironments in vivo, implicating a novel therapeutic target for advanced PCa treatment.


Assuntos
Núcleo Celular/metabolismo , Metaloproteinase 7 da Matriz/fisiologia , Neoplasias da Próstata/patologia , Microambiente Tumoral , Proteína Supressora de Tumor p14ARF/fisiologia , Caderinas/análise , Movimento Celular , Progressão da Doença , Matriz Extracelular/metabolismo , Humanos , Masculino
6.
Oncogene ; 35(13): 1692-702, 2016 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-26119940

RESUMO

Sex-determining region Y box 6 (SOX6) has been described as a tumor-suppressor gene in several cancers. Our previous work has suggested that SOX6 upregulated p21(Waf1/Cip1)(p21) expression in a p53-dependent manner; however, the underlying mechanism has remained elusive. In this study, we confirmed that SOX6 can suppress cell proliferation in vitro and in vivo by stabilizing p53 protein and subsequently upregulating p21. Co-immunoprecipitation and immunocytofluorescence assays demonstrated that SOX6 can promote formation of the p14ARF-HDM2-p53 ternary complex by promoting translocation of p14ARF (p14 alternate reading frame tumor suppressor) to the nucleoplasm, thereby inhibiting HDM2-mediated p53 nuclear export and degradation. Chromatin immunoprecipitation combined with PCR assay proved that SOX6 can bind to a potential binding site in the regulatory region of the c-Myc gene. Furthermore, we confirmed that SOX6 can downregulate the expression of c-Myc, as well as its direct target gene nucleophosmin 1 (NPM1), and that the SOX6-induced downregulation of NPM1 is linked to translocation of p14ARF to the nucleoplasm. Finally, we showed that the highly conserved high-mobility group (HMG) domain of SOX6 is required for SOX6-mediated p53 stabilization and tumor inhibitory activity. Collectively, these results reveal a new mechanism of SOX6-mediated tumor suppression involving p21 upregulation via the p14ARF-HDM2-p53 axis in an HMG domain-dependent manner.


Assuntos
Transformação Celular Neoplásica/genética , Proteínas Proto-Oncogênicas c-mdm2/fisiologia , Fatores de Transcrição SOXD/fisiologia , Proteína Supressora de Tumor p14ARF/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Proteína Supressora de Tumor p53/farmacocinética , Animais , Proliferação de Células/genética , Feminino , Genes Supressores de Tumor/fisiologia , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Nucleofosmina , Estrutura Terciária de Proteína/genética , Fatores de Transcrição SOXD/química , Fatores de Transcrição SOXD/genética , Transdução de Sinais/fisiologia , Células Tumorais Cultivadas
7.
J Cell Physiol ; 231(2): 336-44, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25752701

RESUMO

Weakening the Spindle Assembly Checkpoint by reduced expression of its components induces chromosome instability and aneuploidy that are hallmarks of cancer cells. The tumor suppressor p14(ARF) is overexpressed in response to oncogenic stimuli to stabilize p53 halting cell progression. Previously, we found that lack or reduced expression of p14(ARF) is involved in the maintenance of aneuploid cells in primary human cells, suggesting that it could be part of a pathway controlling their proliferation. To investigate this aspect further, p14(ARF) was ectopically expressed in HCT116 cells after depletion of the Spindle Assembly Checkpoint MAD2 protein that was used as a trigger for aneuploidy. p14(ARF) Re-expression reduced the number of aneuploid cells in MAD2 post-transcriptionally silenced cells. Also aberrant mitoses, frequently displayed in MAD2-depleted cells, were decreased when p14(ARF) was expressed at the same time. In addition, p14(ARF) ectopic expression in MAD2-depleted cells induced apoptosis associated with increased p53 protein levels. Conversely, p14(ARF) ectopic expression did not induce apoptosis in HCT116 p53KO cells. Collectively, our results suggest that the tumor suppressor p14(ARF) may have an important role in counteracting proliferation of aneuploid cells by activating p53-dependent apoptosis.


Assuntos
Aneuploidia , Apoptose/fisiologia , Proteína Supressora de Tumor p14ARF/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Apoptose/genética , Proliferação de Células/genética , Proliferação de Células/fisiologia , Técnicas de Inativação de Genes , Células HCT116 , Humanos , Pontos de Checagem da Fase M do Ciclo Celular/genética , Pontos de Checagem da Fase M do Ciclo Celular/fisiologia , Proteínas Mad2/genética , Proteínas Mad2/fisiologia , Mitose/genética , Mitose/fisiologia , Interferência de RNA , Proteína Supressora de Tumor p14ARF/genética , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
8.
J Mol Cell Biol ; 7(2): 154-67, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25691462

RESUMO

Ribosomes are among the most fundamental molecular machines in all cells, as they are required for protein synthesis. Most structural rRNA components are generated in the nucleolus and assembled into pre-ribosomal particles. Here we show Apak, a previously identified p53 inhibitor, as a novel ribosomal stress response protein. In unstressed cells, Apak is bound to the deSUMOylase SENP1 in the nucleoplasm and targeted for proteasomal degradation by MDM2 ubiquitin ligase. Upon ribosomal stress, SENP1 dissociates from Apak and the tumor suppressor protein ARF couples Ubc9 with Apak to promote Apak SUMOylation on zinc fingers. This results in Apak protein stabilization and translocation to the nucleolus, where Apak inhibits the pre-rRNA synthesis. These findings provide a molecular mechanism whereby ARF coordinates Apak to regulate ribosome biogenesis upon cellular stress.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Nucléolo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Precursores de RNA/biossíntese , RNA Ribossômico/biossíntese , Sumoilação , Proteína Supressora de Tumor p14ARF/fisiologia , Transporte Ativo do Núcleo Celular , Inibidor p16 de Quinase Dependente de Ciclina , Regulação da Expressão Gênica , Células HeLa , Humanos , Estabilidade Proteica , Precursores de RNA/genética , RNA Ribossômico/genética , Enzimas de Conjugação de Ubiquitina/metabolismo
9.
Gut ; 64(7): 1040-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25080447

RESUMO

OBJECTIVE: Infection with Helicobacter pylori is the strongest known risk factor for adenocarcinoma of the stomach. Tumorigenic transformation of gastric epithelium induced by H. pylori is a highly complex process driven by an active interplay between bacterial virulence and host factors, many aspects of which remain obscure. In this work, we investigated the degradation of p53 tumour suppressor induced by H. pylori. DESIGN: Expression of p53 protein in gastric biopsies was assessed by immunohistochemistry. Gastric cells were co-cultured with H. pylori strains isolated from high-gastric risk and low-gastric risk areas and assessed for expression of p53, p14ARF and cytotoxin-associated gene A (CagA) by immunoblotting. siRNA was used to inhibit activities of ARF-BP1 and Human Double Minute 2 (HDM2) proteins. RESULTS: Our analysis demonstrated that H. pylori strains expressing high levels of CagA virulence factor and associated with a higher gastric cancer risk more strongly suppress p53 compared with low-risk strains in vivo and in vitro. We found that degradation of p53 induced by bacterial CagA protein is mediated by host HDM2 and ARF-BP1 E3 ubiquitin ligases, while the p14ARF protein counteracts H. pylori-induced signalling. CONCLUSIONS: Our results provide novel evidence that tumorigenicity associated with H. pylori infection is linked to inhibition of p53 protein by CagA. We propose a model in which CagA-induced degradation of p53 protein is determined by a relative level of p14ARF. In cells in which p14ARF levels were decreased due to hypermethylation or deletion of the p14ARF gene, H. pylori efficiently degraded p53, whereas p53 is protected in cells expressing high levels of p14ARF.


Assuntos
Antígenos de Bactérias/fisiologia , Proteínas de Bactérias/fisiologia , Neoplasias Gástricas/microbiologia , Proteína Supressora de Tumor p14ARF/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Antígenos de Bactérias/classificação , Proteínas de Bactérias/classificação , Linhagem Celular Tumoral , Epitélio/metabolismo , Mucosa Gástrica/microbiologia , Humanos , Imuno-Histoquímica , Neoplasias Gástricas/fisiopatologia
10.
Oncogene ; 34(13): 1688-97, 2015 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-24769896

RESUMO

Upregulated expression of nucleolar GTPase nucleostemin (NS) has been associated with increased cellular proliferation potential and tumor malignancy during cancer development. Recent reports attribute the growth regulatory effects of NS protein to its role in facilitating ribosome production. However, the oncogenic potential of NS remains unclear, as imbalanced levels of NS have been reported to exert growth inhibitory effect by modulating p53 tumor-suppressor activity. It also remains in questions if aberrant NS levels might have a p53-independent role in regulation of cell proliferation and growth. In this study, we performed affinity purification and mass spectrometry analysis to explore protein-protein interactions influencing NS growth regulatory properties independently of p53 tumor suppressor. We identified the alternative reading frame (ARF) protein as a key protein associating with NS and further verified the interaction through in vitro and in vivo assays. We demonstrated that NS is able to regulate cell cycle progression by regulating the stability of the ARF tumor suppressor. Furthermore, overexpression of NS suppressed ARF polyubiquitination by its E3 ligase Ubiquitin Ligase for ARF and elongated its half-life, whereas knockdown of NS led to the decrease of ARF levels. Also, we found that NS can enhance NPM stabilization of ARF. Thus, we propose that in the absence of p53, ARF can be stabilized by NS and nucleophosmin to serve as an alternative tumor-suppressor surveillance, preventing potential cellular transformation resulting from the growth-inducing effects of NS overexpression.


Assuntos
Proteínas de Transporte/antagonistas & inibidores , Proteínas de Ligação ao GTP/fisiologia , Proteínas Nucleares/fisiologia , Proteína Supressora de Tumor p14ARF/fisiologia , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Linhagem Celular Tumoral , Pontos de Checagem da Fase G1 do Ciclo Celular , Proteínas de Ligação ao GTP/química , Humanos , Proteínas Nucleares/química , Estabilidade Proteica , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-mdm2/fisiologia , Proteína Supressora de Tumor p14ARF/química , Proteína Supressora de Tumor p53/fisiologia
11.
Endocrinology ; 155(11): 4329-40, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25137025

RESUMO

Acromegaly is caused by somatotroph cell adenomas (somatotropinomas [ACROs]), which secrete GH. Human and rodent somatotroph cells express the RET receptor. In rodents, when normal somatotrophs are deprived of the RET ligand, GDNF (Glial Cell Derived Neurotrophic Factor), RET is processed intracellularly to induce overexpression of Pit1 [Transcription factor (gene : POUF1) essential for transcription of Pituitary hormones GH, PRL and TSHb], which in turn leads to p19Arf/p53-dependent apoptosis. Our purpose was to ascertain whether human ACROs maintain the RET/Pit1/p14ARF/p53/apoptosis pathway, relative to nonfunctioning pituitary adenomas (NFPAs). Apoptosis in the absence and presence of GDNF was studied in primary cultures of 8 ACROs and 3 NFPAs. Parallel protein extracts were analyzed for expression of RET, Pit1, p19Arf, p53, and phospho-Akt. When GDNF deprived, ACRO cells, but not NFPAs, presented marked level of apoptosis that was prevented in the presence of GDNF. Apoptosis was accompanied by RET processing, Pit1 accumulation, and p14ARF and p53 induction. GDNF prevented all these effects via activation of phospho-AKT. Overexpression of human Pit1 (hPit1) directly induced p19Arf/p53 and apoptosis in a pituitary cell line. Using in silico studies, 2 CCAAT/enhancer binding protein alpha (cEBPα) consensus-binding sites were found to be 100% conserved in mouse, rat, and hPit1 promoters. Deletion of 1 cEBPα site prevented the RET-induced increase in hPit1 promoter expression. TaqMan qRT-PCR (real time RT-PCR) for RET, Pit1, Arf, TP53, GDNF, steroidogenic factor 1, and GH was performed in RNA from whole ACRO and NFPA tumors. ACRO but not NFPA adenomas express RET and Pit1. GDNF expression in the tumors was positively correlated with RET and negatively correlated with p53. In conclusion, ACROs maintain an active RET/Pit1/p14Arf/p53/apoptosis pathway that is inhibited by GDNF. Disruption of GDNF's survival function might constitute a new therapeutic route in acromegaly.


Assuntos
Adenoma/patologia , Apoptose/efeitos dos fármacos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Adenoma Hipofisário Secretor de Hormônio do Crescimento/patologia , Neoplasias Hipofisárias/patologia , Adenoma/genética , Animais , Apoptose/genética , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Adenoma Hipofisário Secretor de Hormônio do Crescimento/genética , Humanos , Neoplasias Hipofisárias/genética , Proteínas Proto-Oncogênicas c-ret/fisiologia , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fator de Transcrição Pit-1/fisiologia , Células Tumorais Cultivadas , Proteína Supressora de Tumor p14ARF/fisiologia , Proteína Supressora de Tumor p53/fisiologia
13.
Mol Cell ; 51(1): 46-56, 2013 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-23747016

RESUMO

ARF suppresses aberrant cell growth upon c-Myc overexpression by activating p53 responses. Nevertheless, the precise mechanism by which ARF specifically restrains the oncogenic potential of c-Myc without affecting its normal physiological function is not well understood. Here, we show that low levels of c-Myc expression stimulate cell proliferation, whereas high levels inhibit by activating the ARF/p53 response. Although the mRNA levels of ARF are induced in both scenarios, the accumulation of ARF protein occurs only when ULF-mediated degradation of ARF is inhibited by c-Myc overexpression. Moreover, the levels of ARF are reduced through ULF-mediated ubiquitination upon DNA damage. Blocking ARF degradation by c-Myc overexpression dramatically stimulates the apoptotic responses. Our study reveals that ARF stability control is crucial for differentiating normal (low) versus oncogenic (high) levels of c-Myc expression and suggests that differential effects on ULF- mediated ARF ubiquitination by c-Myc levels act as a barrier in oncogene-induced stress responses.


Assuntos
Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteína Supressora de Tumor p14ARF/fisiologia , Animais , Apoptose , Proteínas de Transporte/metabolismo , Proteínas de Transporte/fisiologia , Linhagem Celular , Proliferação de Células , Dano ao DNA , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Camundongos , Proteínas Proto-Oncogênicas c-myc/genética , RNA Mensageiro/metabolismo , Proteína Supressora de Tumor p14ARF/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/fisiologia
14.
Cancer Res ; 73(9): 2829-39, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23423975

RESUMO

Cellular senescence, a state of cell-cycle arrest accompanied by dramatic morphologic and metabolic changes, is a central means by which cells respond to physiologic stress and oncogene activity. Senescence is thought to play important roles in aging and in tumor suppression, yet the dynamics by which senescent cells are formed, their effects on tissue function and their eventual fate are poorly understood. To study cellular senescence within an adult tissue, we developed transgenic mice inducibly expressing p14(ARF) (human ortholog of murine p19(ARF)), a central activator of senescence. Induction of p14(ARF) in the epidermis rapidly led to widespread apoptosis and cell-cycle arrest, a stage that was transient, and was followed by p53-dependent cellular senescence. The endogenous Cdkn2a products p19(ARF) and p16(Ink4a) were activated by the transgenic p14(ARF) through p53, revealing a senescence-promoting feed-forward loop. Commitment of cells to senescence required continued p14(ARF) expression, indicating that entry into this state depends on a persistent signal. However, once formed, senescent cells were retained in the epidermis, often for weeks after transgene silencing, indicating an absence of an efficient rapidly acting mechanism for their removal. Stem cells in the hair follicle bulge were largely protected from apoptosis upon p14(ARF) induction, but irreversibly lost their ability to proliferate and initiate follicle growth. Interestingly, induction of epidermal hyperplasia prevented the appearance of senescent cells upon p14(ARF) induction. Our findings provide basic insights into the dynamics of cellular senescence, a central tumor- suppressive mechanism, and reveal the potential for prolonged retention of senescent cells within tissues.


Assuntos
Apoptose , Epiderme/metabolismo , Proteína Supressora de Tumor p14ARF/metabolismo , Animais , Ciclo Celular , Senescência Celular , Inativação Gênica , Folículo Piloso/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Modelos Biológicos , RNA/metabolismo , Transgenes , Proteína Supressora de Tumor p14ARF/fisiologia , Proteína Supressora de Tumor p53/metabolismo , beta-Galactosidase/metabolismo
15.
Oncogene ; 32(35): 4120-9, 2013 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-23045280

RESUMO

Our recent study showed critical roles of Dmp1 as a sensor of oncogenic Ras, HER2/neu signaling and activation of the Arf-p53 pathway. To elucidate the role of human DMP1 (hDMP1) in breast cancer, one hundred and ten pairs of human breast cancer specimen were studied for the alterations of the hDMP1-ARF-Hdm2-p53 pathway with follow up of clinical outcomes. Loss of heterozygosity (LOH) of the hDMP1 locus was found in 42% of human breast carcinomas, while that of INK4a/ARF and p53 were found in 20 and 34%, respectively. Hdm2 amplification was found in 13% of the same sample, which was found independently of LOH for hDMP1. Conversely, LOH for hDMP1 was found in mutually exclusive fashion with that of INK4a/ARF and p53, and was associated with low Ki67 index and diploid karyotype. Consistently, LOH for hDMP1 was associated with luminal A category and longer relapse-free survival, while that of p53 was associated with non-luminal A and shorter survival. Thus, loss of hDMP1 could define a new disease category associated with prognosis of breast cancer patients. Human breast epithelial cells/cancer cells with wild-type p53 were sensitive to growth inhibition by activated Dmp1:ER while those that delete p14(ARF) or p53, and/or Hdm2 amplification showed partial or nearly complete resistance, indicating that p53 is a critical target for hDMP1 to exhibit its biological activity.


Assuntos
Neoplasias da Mama/mortalidade , Proteínas Proto-Oncogênicas c-mdm2/fisiologia , Fatores de Transcrição/fisiologia , Proteína Supressora de Tumor p14ARF/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Feminino , Humanos , Perda de Heterozigosidade , Invasividade Neoplásica , Prognóstico , Transdução de Sinais
16.
PLoS One ; 7(7): e42210, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22860085

RESUMO

CDK5 regulatory subunit associated protein 3 (CDK5RAP3) is a novel activator of PAK4 and processes important pro-metastatic function in hepatocarcinogenesis. However, it remains unclear if there are other mechanisms by which CDK5RAP3 promotes HCC metastasis. Here, we showed that in CDK5RAP3 stable knockdown SMMC-7721 HCC cells, p14(ARF) tumor suppressor was upregulated at protein and mRNA levels, and ectopic expression of CDK5RAP3 was found to repress the transcription of p14(ARF). Using chromatin immunoprecipitation assay, we demonstrated that CDK5RAP3 bound to p14(ARF) promoter in vivo. Furthermore, knockdown of p14(ARF) in CDK5RAP3 stable knockdown HCC cells reversed the suppression of HCC cell invasiveness mediated by knockdown of CDK5RAP3. Taken together, our findings provide the new evidence that overexpression of CDK5RAP3 promotes HCC metastasis via downregulation of p14(ARF).


Assuntos
Carcinoma Hepatocelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Neoplasias Hepáticas/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Proteína Supressora de Tumor p14ARF/fisiologia , Carcinoma Hepatocelular/patologia , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Hepáticas/patologia , Microscopia Confocal , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Supressora de Tumor p14ARF/genética , Proteínas Supressoras de Tumor
17.
PLoS One ; 7(7): e42246, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22860097

RESUMO

As part of a cell's inherent protection against carcinogenesis, p14ARF is upregulated in response to hyperproliferative signalling to induce cell cycle arrest. This property makes p14ARF a leading candidate for cancer therapy. This study explores the consequences of reactivating p14ARF in breast cancer and the potential of targeting p14ARF in breast cancer treatment. Our results show that activation of the p14ARF-p53-p21-Rb pathway in the estrogen sensitive MCF-7 breast cancer cells induces many hallmarks of senescence including a large flat cell morphology, multinucleation, senescence-associated-ß-gal staining, and rapid G1 and G2/M phase cell cycle arrest. P14ARF also induces the expression of the proto-oncogene cyclin D1, which is most often associated with a transition from G1-S phase and is highly expressed in breast cancers with poor clinical prognosis. In this study, siRNA knockdown of cyclin D1, p21 and p53 show p21 plays a pivotal role in the maintenance of high cyclin D1 expression, cell cycle and growth arrest post-p14ARF induction. High p53 and p14ARF expression and low p21/cyclin D1 did not cause cell-cycle arrest. Knockdown of cyclin D1 stops proliferation but does not reverse senescence-associated cell growth. Furthermore, cyclin D1 accumulation in the nucleus post-p14ARF activation correlated with a rapid loss of nucleolar Ki-67 protein and inhibition of DNA synthesis. Latent effects of the p14ARF-induced cellular processes resulting from high nuclear cyclin D1 accumulation included a redistribution of Ki-67 into the nucleoli, aberrant nuclear growth (multinucleation), and cell proliferation. Lastly, downregulation of cyclin D1 through inhibition of ER abrogated latent recurrence. The mediation of these latent effects by continuous expression of p14ARF further suggests a novel mechanism whereby dysregulation of cyclin D1 could have a double-edged effect. Our results suggest that p14ARF induced-senescence is related to late-onset breast cancer in estrogen responsive breast cancers and/or the recurrence of more aggressive breast cancer post-therapy.


Assuntos
Neoplasias da Mama/metabolismo , Núcleo Celular/metabolismo , Ciclina D1/genética , Processamento Pós-Transcricional do RNA/fisiologia , Proteína Supressora de Tumor p14ARF/fisiologia , Neoplasias da Mama/patologia , Ciclo Celular , Feminino , Técnicas de Silenciamento de Genes , Humanos , Células MCF-7 , Prognóstico , Proto-Oncogene Mas , RNA Interferente Pequeno
19.
J Clin Invest ; 122(4): 1283-95, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22378045

RESUMO

Malignant gliomas are the most common and the most lethal primary brain tumors in adults. Among malignant gliomas, 60%-80% show loss of P14ARF tumor suppressor activity due to somatic alterations of the INK4A/ARF genetic locus. The tumor suppressor activity of P14ARF is in part a result of its ability to prevent the degradation of P53 by binding to and sequestering HDM2. However, the subsequent finding of P14ARF loss in conjunction with TP53 gene loss in some tumors suggests the protein may have other P53-independent tumor suppressor functions. Here, we report what we believe to be a novel tumor suppressor function for P14ARF as an inhibitor of tumor-induced angiogenesis. We found that P14ARF mediates antiangiogenic effects by upregulating expression of tissue inhibitor of metalloproteinase-3 (TIMP3) in a P53-independent fashion. Mechanistically, this regulation occurred at the gene transcription level and was controlled by HDM2-SP1 interplay, where P14ARF relieved a dominant negative interaction of HDM2 with SP1. P14ARF-induced expression of TIMP3 inhibited endothelial cell migration and vessel formation in response to angiogenic stimuli produced by cancer cells. The discovery of this angiogenesis regulatory pathway may provide new insights into P53-independent P14ARF tumor-suppressive mechanisms that have implications for the development of novel therapies directed at tumors and other diseases characterized by vascular pathology.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Regulação Neoplásica da Expressão Gênica , Glioblastoma/irrigação sanguínea , Proteínas de Neoplasias/fisiologia , Neovascularização Patológica/fisiopatologia , Inibidor Tecidual de Metaloproteinase-3/biossíntese , Proteína Supressora de Tumor p14ARF/fisiologia , Animais , Neoplasias Encefálicas/metabolismo , Linhagem Celular Transformada , Quimiotaxia/efeitos dos fármacos , Neovascularização da Córnea/fisiopatologia , Meios de Cultivo Condicionados/farmacologia , Células Endoteliais/patologia , Fibroblastos/efeitos dos fármacos , Genes p53 , Glioblastoma/metabolismo , Humanos , Camundongos , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Neovascularização Patológica/genética , Proteínas Proto-Oncogênicas c-mdm2/fisiologia , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Proteínas Recombinantes de Fusão/fisiologia , Fator de Transcrição Sp1/fisiologia , Inibidor Tecidual de Metaloproteinase-3/antagonistas & inibidores , Inibidor Tecidual de Metaloproteinase-3/genética , Inibidor Tecidual de Metaloproteinase-3/fisiologia , Transcrição Gênica , Células Tumorais Cultivadas/metabolismo , Regulação para Cima
20.
Curr Mol Med ; 12(6): 704-15, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22292438

RESUMO

Oxidative stress as a result of either exogenous stimuli or cellular metabolism affects several cellular processes such as proliferation, apoptosis, cell death and senescence. Consequently, it is implicated in the pathogenesis of various human diseases like cancer, diabetes mellitus, atherosclerosis, neurodegenerative diseases and aging. Oxidative stress is implicated in carcinogenesis either by directly provoking DNA damage or through the regulation of intracellular signaling cascades. In both cases the cellular response to oxidative stress is determined by the cellular context. ARF, the alternative protein product of the CDKN2A locus has been recently recognized as a novel sensor of oxidative stress, in a ß-catenin and Hsp70-mediated manner. Since, improved understanding of cellular responses to oxidative stress may facilitate the design of novel antineoplastic regimens, we herein review the mechanisms by which oxidative stress promotes carcinogenesis, focusing on the role of ARF as a sensor of oxidative stress.


Assuntos
Estresse Oxidativo , Proteína Supressora de Tumor p14ARF/genética , Animais , Transformação Celular Neoplásica/metabolismo , Dano ao DNA , Humanos , Mutagênese , Neoplasias/etiologia , Neoplasias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p14ARF/metabolismo , Proteína Supressora de Tumor p14ARF/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...