Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
1.
Clin Transl Oncol ; 24(2): 266-275, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34318428

RESUMO

PURPOSE: Increasing evidences suggest dysfunctions of microRNAs (miRNAs) are playing important part in tumors. Therefore, the role of miR-802 in osteosarcoma (OS) was exploited. The object was to evaluate the effect of miR-802 and verify its influence on p27 Kip1 (p27) in OS. METHODS: RT-qPCR experiment was used to detect miR-802 and p27 expression in OS tissues and cells. We explored the function of miR-802 through Transwell assays. The phosphoinositide 3-kinase (PI3K)/AKT serine/threonine kinase pathway and epithelial-mesenchymal transition (EMT) was detected by Western blot assays. Luciferase assay was used to testify the target of miR-802. RESULTS: MiR-802 expression was elevated in OS, which was related to poor clinical outcome in OS patients. MiR-802 overexpression promoted OS migration, invasion and EMT. Further, p27 is a direct target of miR-802. P27 elevation counteracted the promotion effect of OS on EMT, migration and invasion induced by miR-802. In addition, miR-802 overexpression inactivated PI3K/AKT pathway via targeting p27 in OS. CONCLUSION: MiR-802 promoted the progress of EMT, migration and invasion in OS via targeting p27. This newly identified miR-802/p27/PI3K/AKT axis may represent potential targets for OS.


Assuntos
Neoplasias Ósseas/etiologia , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , MicroRNAs/fisiologia , Osteossarcoma/etiologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Adolescente , Neoplasias Ósseas/patologia , Progressão da Doença , Feminino , Humanos , Masculino , Osteossarcoma/patologia , Adulto Jovem
2.
Sci Rep ; 9(1): 18693, 2019 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-31822694

RESUMO

Cell cycle stimulation is a major transforming mechanism of Myc oncoprotein. This is achieved through at least three concomitant mechanisms: upregulation of cyclins and Cdks, downregulation of the Cdk inhibitors p15 and p21 and the degradation of p27. The Myc-p27 antagonism has been shown to be relevant in human cancer. To be degraded, p27 must be phosphorylated at Thr-187 to be recognized by Skp2, a component of the ubiquitination complex. We previously described that Myc induces Skp2 expression. Here we show that not only Cdk2 but Cdk1 phosphorylates p27 at the Thr-187. Moreover, Myc induced p27 degradation in murine fibroblasts through Cdk1 activation, which was achieved by Myc-dependent cyclin A and B induction. In the absence of Cdk2, p27 phosphorylation at Thr-187 was mainly carried out by cyclin A2-Cdk1 and cyclin B1-Cdk1. We also show that Cdk1 inhibition was enough for the synthetic lethal interaction with Myc. This result is relevant because Cdk1 is the only Cdk strictly required for cell cycle and the reported synthetic lethal interaction between Cdk1 and Myc.


Assuntos
Proteína Quinase CDC2/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Animais , Proteína Quinase CDC2/fisiologia , Ciclo Celular , Pontos de Checagem do Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Divisão Celular , Linhagem Celular , Quinase 2 Dependente de Ciclina , Quinase 4 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p15/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Feminino , Células HEK293 , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Proteínas Proto-Oncogênicas c-myc/fisiologia , Transdução de Sinais
3.
PLoS Genet ; 15(3): e1008002, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30893315

RESUMO

Mammary epithelial progenitors are the normal cell-of-origin of breast cancer. We previously defined a population of p27+ quiescent hormone-responsive progenitor cells in the normal human breast whose frequency associates with breast cancer risk. Here, we describe that deletion of the Cdkn1b gene encoding the p27 cyclin-dependent kinase inhibitor in the estrogen-induced mammary tumor-susceptible ACI rat strain leads to a decrease in the relative frequencies of Cd49b+ mammary luminal epithelial progenitors and pregnancy-related differentiation. We show by comprehensive gene expression profiling of purified progenitor and differentiated mammary epithelial cell populations that p27 deletion has the most pronounced effects on luminal progenitors. Cdkn1b-/- females have decreased fertility, but rats that are able to get pregnant had normal litter size and were able to nurse their pups implying that loss of p27 in ACI rats does not completely abrogate ovarian function and lactation. Reciprocal mammary gland transplantation experiments indicate that the p27-loss-induced changes in mammary epithelial cells are not only caused by alterations in their intrinsic properties, but are likely due to altered hormonal signaling triggered by the perturbed systemic endocrine environment observed in Cdkn1b-/- females. We also observed a decrease in the frequency of mammary epithelial cells positive for progesterone receptor (Pr) and FoxA1, known direct transcriptional targets of the estrogen receptor (Erα), and an increase in phospho-Stat5 positive cells commonly induced by prolactin (Prl). Characterization of genome-wide Pr chromatin binding revealed distinct binding patterns in mammary epithelial cells of Cdkn1b+/+ and Cdkn1b-/- females and enrichment in genes with known roles in Notch, ErbB, leptin, and Erα signaling and regulation of G1-S transition. Our data support a role for p27 in regulating the pool size of hormone-responsive luminal progenitors that could impact breast cancer risk.


Assuntos
Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Animais , Animais Geneticamente Modificados/genética , Neoplasias da Mama/genética , Diferenciação Celular , Proliferação de Células/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Células Endócrinas/fisiologia , Células Epiteliais , Receptor alfa de Estrogênio , Estrogênios , Feminino , Predisposição Genética para Doença/genética , Humanos , Integrina alfa1 , Glândulas Mamárias Animais , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Gravidez , Progesterona , Ratos , Ratos Endogâmicos ACI , Ratos Sprague-Dawley , Receptores de Estrogênio , Receptores de Progesterona , Fatores de Risco , Transdução de Sinais , Células-Tronco
4.
DNA Repair (Amst) ; 69: 63-72, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30075372

RESUMO

The cell cycle is controlled by precise mechanisms to prevent malignancies such as cancer, and the cell needs these tight and advanced controls. Cyclin dependent kinase inhibitor p27 (also known as KIP1) is a factor that inhibits the progression of the cell cycle by using specific molecular mechanisms. The inhibitory effect of p27 on the cell cycle is mediated by CDKs inhibition. Other important functions of p27 include cell proliferation, cell differentiation and apoptosis. Post- translational modification of p27 by phosphorylation and ubiquitination respectively regulates interaction between p27 and cyclin/CDK complex and degradation of p27. In this review, we focus on the multiple function of p27 in cell cycle regulation, apoptosis, epigenetic modifications and post- translational modification, and briefly discuss the mechanisms and factors that have important roles in p27 functions.


Assuntos
Ciclo Celular , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Animais , Apoptose , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Epigênese Genética , Regulação da Expressão Gênica , Humanos , Processamento de Proteína Pós-Traducional
6.
Tumour Biol ; 39(4): 1010428317694574, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28381184

RESUMO

Glioblastoma multiforme is the most common primary brain tumor and is highly lethal. This study aims to figure out signatures for predicting the survival time of patients with glioblastoma multiforme. Clinical information, messenger RNA expression, microRNA expression, and single-nucleotide polymorphism array data of patients with glioblastoma multiforme were retrieved from The Cancer Genome Atlas. Patients were separated into two groups by using 1 year as a cutoff, and a logistic regression model was used to figure out any variables that can predict whether the patient was able to live longer than 1 year. Furthermore, Cox's model was used to find out features that were correlated with the survival time. Finally, a Cox model integrated the significant clinical variables, messenger RNA expression, microRNA expression, and single-nucleotide polymorphism was built. Although the classification method failed, signatures of clinical features, messenger RNA expression levels, and microRNA expression levels were figured out by using Cox's model. However, no single-nucleotide polymorphisms related to prognosis were found. The selected clinical features were age at initial diagnosis, Karnofsky score, and race, all of which had been suggested to correlate with survival time. Both of the two significant microRNAs, microRNA-221 and microRNA-222, were targeted to p27Kip1 protein, which implied the important role of p27Kip1 on the prognosis of glioblastoma multiforme patients. Our results suggested that survival modeling was more suitable than classification to figure out prognostic biomarkers for patients with glioblastoma multiforme. An integrated model containing clinical features, messenger RNA levels, and microRNA expression levels was built, which has the potential to be used in clinics and thus to improve the survival status of glioblastoma multiforme patients.


Assuntos
Neoplasias Encefálicas/mortalidade , Glioblastoma/mortalidade , Neoplasias Encefálicas/genética , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Feminino , Glioblastoma/genética , Humanos , Modelos Logísticos , Masculino , MicroRNAs/análise , Polimorfismo de Nucleotídeo Único , Prognóstico , Modelos de Riscos Proporcionais
7.
Clin Res Hepatol Gastroenterol ; 41(4): 445-458, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28286054

RESUMO

OBJECTIVE: To investigate the role of chromosome region maintenance-1 (CRM1) in Crohn's disease (CD) and its potential pathological mechanisms. METHODS: The expression and distribution of CRM1 in mucosal biopsies from patients with active CD and normal controls were detected by immunohistochemistry (IHC). We established a murine model of acute colitis induced by 2,4,6-trinitrobenzene sulfonic acid (TNBS). Western blot was performed to investigate the expression levels of CRM1, apoptotic markers (active caspase-3 and cleaved PARP), p27kip1 and p-p27ser10. IHC was performed to evaluate the distribution of CRM1, and double immunofluorescence (IF) was performed to evaluate the co-localization of CRM1 and active capase-3. Cells of the human intestinal epithelial cell line HT-29 were incubated with tumor necrosis factor-α (TNF-α) to establish an apoptotic in vitro model. Western blot was performed to determine the expression levels of CRM1, active caspase-3, cleaved PARP and p-p27ser10. Cytoplasmic and nuclear extracts were assessed to examine the translocation of CRM1. The interaction between CRM1 and p27kip1 was assessed by co-immunoprecipitation (co-IP) assays. Furthermore, we used small interfering RNA (siRNA) to knock down the protein expression of CRM1 in HT-29 cells and then measured the expression of active caspase-3, cleaved PARP and p-p27ser10. Flow cytometry was used to determine the effect of CRM1 on intestinal epithelial cell (IEC) apoptosis. RESULTS: We observed up-regulation of CRM1 accompanied by elevated levels of IEC apoptotic markers (active caspase-3 and cleaved PARP) and p-p27ser10 in IECs of patients with active CD and in TNBS-induced colitis model cells. However, the expression of p27kip1 was negatively correlated with the expression patterns of CRM1, p-p27ser10 and apoptotic biochemical markers. Co-localization of CRM1 and active caspase-3 in IECs of the TNBS group further indicated the possible involvement of CRM1 in IEC apoptosis. By employing TNF-α-treated HT-29 cells as an in vitro IEC apoptosis model, we found that the expression levels of CRM1 and p-p27ser10 were in accordance with active caspase-3 and cleaved PARP. In addition, immunoprecipitation confirmed the physical interaction between CRM1 and p27kip1. siRNA knockdown of CRM1 significantly inhibited the phosphorylation of p27kip1 and the expression of active caspase-3 and cleaved PARP. In addition, flow cytometry analysis also showed that silencing CRM1 by siRNA inhibited TNF-α-induced cellular apoptosis in HT-29 cells. CONCLUSIONS: Up-regulated CRM1 may facilitate IEC apoptosis possibly through p27kip1 in CD, indicating an important role of CRM1 in the pathophysiology of CD.


Assuntos
Apoptose , Doença de Crohn/patologia , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Células Epiteliais , Carioferinas/fisiologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Animais , Humanos , Mucosa Intestinal/citologia , Camundongos , Proteína Exportina 1
8.
Nucleic Acids Res ; 45(9): 5086-5099, 2017 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-28158851

RESUMO

The cyclin-dependent kinase inhibitor p27Kip1 (p27) also behaves as a transcriptional repressor. Data showing that the p300/CBP-associated factor (PCAF) acetylates p27 inducing its degradation suggested that PCAF and p27 could collaborate in the regulation of transcription. However, this possibility remained to be explored. We analyzed here the transcriptional programs regulated by PCAF and p27 in the colon cancer cell line HCT116 by chromatin immunoprecipitation sequencing (ChIP-seq). We identified 269 protein-encoding genes that contain both p27 and PCAF binding sites being the majority of these sites different for PCAF and p27. PCAF or p27 knock down revealed that both regulate the expression of these genes, PCAF as an activator and p27 as a repressor. The double knock down of PCAF and p27 strongly reduced their expression indicating that the activating role of PCAF overrides the repressive effect of p27. We also observed that the transcription factor Pax5 interacts with both p27 and PCAF and that the knock down of Pax5 induces the expression of p27/PCAF target genes indicating that it also participates in the transcriptional regulation mediated by p27/PCAF. In summary, we report here a previously unknown mechanism of transcriptional regulation mediated by p27, Pax5 and PCAF.


Assuntos
Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Regulação da Expressão Gênica , Fator de Transcrição PAX5/fisiologia , Fatores de Transcrição de p300-CBP/fisiologia , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Células Cultivadas , Cromatina/metabolismo , Células HCT116 , Humanos , Células MCF-7 , Camundongos , Ligação Proteica , Proteínas/genética , Análise Serial de Tecidos , Transcrição Gênica
9.
Nat Commun ; 7: 13719, 2016 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-27941799

RESUMO

Activation of NOTCH signalling is associated with advanced prostate cancer and treatment resistance in prostate cancer patients. However, the mechanism that drives NOTCH activation in prostate cancer remains still elusive. Moreover, preclinical evidence of the therapeutic efficacy of NOTCH inhibitors in prostate cancer is lacking. Here, we provide evidence that PTEN loss in prostate tumours upregulates the expression of ADAM17, thereby activating NOTCH signalling. Using prostate conditional inactivation of both Pten and Notch1 along with preclinical trials carried out in Pten-null prostate conditional mouse models, we demonstrate that Pten-deficient prostate tumours are addicted to the NOTCH signalling. Importantly, we find that pharmacological inhibition of γ-secretase promotes growth arrest in both Pten-null and Pten/Trp53-null prostate tumours by triggering cellular senescence. Altogether, our findings describe a novel pro-tumorigenic network that links PTEN loss to ADAM17 and NOTCH signalling, thus providing the rational for the use of γ-secretase inhibitors in advanced prostate cancer patients.


Assuntos
PTEN Fosfo-Hidrolase/genética , Neoplasias da Próstata/tratamento farmacológico , Receptores Notch/antagonistas & inibidores , Proteína ADAM17/genética , Proteína ADAM17/metabolismo , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Senescência Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Humanos , Masculino , Camundongos , PTEN Fosfo-Hidrolase/metabolismo , Neoplasias da Próstata/patologia , Receptores Notch/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tetra-Hidronaftalenos/uso terapêutico , Regulação para Cima , Valina/análogos & derivados , Valina/uso terapêutico
10.
Biomed Pharmacother ; 84: 1967-1971, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27842905

RESUMO

OBJECTIVE: The aim of this study is to explore the influence of miR-148a on cell proliferation and cell cycle of multiple myeloma (MM) cell line RPMI8226 and the related molecular mechanism. METHODS: The expression of miR-148a and CDKN1B in MM cells and primary cells of normal bone marrow were determined by RT-PCR and western blotting. The cell proliferation and cell cycle of miR-148a knockdown MM cells and normal MM cells were determined by flow cytometry. The protein expression of p-NPAT, p-Rb and p-CDC6 was determined in normal and miR-148a knockdown MM cells. Luciferase reported assay was used to explore the relationship between miR-148a and CDKN1B. RESULTS: The level of miR-148a in MM cells was much higher than that in primary cells from healthy bone marrow samples, while the expression of CDKN1B was lower in MM cells. After knockdown of miR-148a, cell cycle mainly distributed at G0/G1 and the proliferation capacity of MM cells decreased. Knockdown of miR-148a significantly reduced protein expression of p-NPAT, p-Rb and p-CDC6. Luciferase reported assay showed that miR-148a could directly target CDKN1B at 3'-UTR. CONCLUSIONS: High level of miR-148a inhibits CDK activity and promotes the proliferation of MM cells at least partly by downregulating CDKN1B.


Assuntos
Inibidor de Quinase Dependente de Ciclina p27/fisiologia , MicroRNAs/biossíntese , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Inibidor de Quinase Dependente de Ciclina p27/antagonistas & inibidores , Técnicas de Silenciamento de Genes , Humanos
11.
Oncotarget ; 7(34): 54102-54119, 2016 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-27472461

RESUMO

Glioblastoma multiforme (GBM) is an aggressive cancer with current therapies only marginally impacting on patient survival. Glioma stem cells (GSCs), a subpopulation of highly tumorigenic cells, are considered major contributors to glioma progression and play seminal roles in therapy resistance, immune evasion and increased invasion. Despite clinical relevance, effective/selective therapeutic targeting strategies for GSCs do not exist, potentially due to the lack of a definitive understanding of key regulators of GSCs. Consequently, there is a pressing need to identify therapeutic targets and novel options to effectively target this therapy-resistant cell population. The precise roles of GSCs in governing GBM development, progression and prognosis are under intense scrutiny, but key upstream regulatory genes remain speculative. MDA-9/Syntenin (SDCBP), a scaffold protein, regulates tumor pathogenesis in multiple cancers. Highly aggressive cancers like GBM express elevated levels of MDA-9 and contain increased populations of GSCs. We now uncover a unique function of MDA-9 as a facilitator and determinant of glioma stemness and survival. Mechanistically, MDA-9 regulates multiple stemness genes (Nanog, Oct4 and Sox2) through activation of STAT3. MDA-9 controls survival of GSCs by activating the NOTCH1 pathway through phospho-Src and DLL1. Once activated, cleaved NOTCH1 regulates C-Myc expression through RBPJK, thereby facilitating GSC growth and proliferation. Knockdown of MDA-9 affects the NOTCH1/C-Myc and p-STAT3/Nanog pathways causing a loss of stemness and initiation of apoptosis in GSCs. Our data uncover a previously unidentified relationship between MDA-9 and GSCs, reinforcing relevance of this gene as a potential therapeutic target in GBM.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , Células-Tronco Neoplásicas/fisiologia , Sinteninas/fisiologia , Animais , Astrócitos/fisiologia , Neoplasias Encefálicas/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Feminino , Glioma/tratamento farmacológico , Humanos , Camundongos , Células-Tronco Neoplásicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/fisiologia , Receptor Notch1/fisiologia , Fator de Transcrição STAT3/fisiologia , Sinteninas/antagonistas & inibidores , Sinteninas/genética
12.
Cell Death Differ ; 23(3): 430-41, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26292757

RESUMO

Cell-cycle inhibitors of the Ink4 and Cip/Kip families are involved in cellular senescence and tumor suppression. These inhibitors are individually dispensable for the cell cycle and inactivation of specific family members results in increased proliferation and enhanced susceptibility to tumor development. We have now analyzed the consequences of eliminating a substantial part of the cell-cycle inhibitory activity in the cell by generating a mouse model, which combines the absence of both p21(Cip1) and p27(Kip1) proteins with the endogenous expression of a Cdk4 R24C mutant insensitive to Ink4 inhibitors. Pairwise combination of Cdk4 R24C, p21-null and p27-null alleles results in frequent hyperplasias and tumors, mainly in cells of endocrine origin such as pituitary cells and in mesenchymal tissues. Interestingly, complete abrogation of p21(Cip1) and p27(Kip1) in Cdk4 R24C mutant mice results in a different phenotype characterized by perinatal death accompanied by general hypoplasia in most tissues. This phenotype correlates with increased replicative stress in developing tissues such as the nervous system and subsequent apoptotic cell death. Partial inhibition of Cdk4/6 rescues replicative stress signaling as well as p53 induction in the absence of cell-cycle inhibitors. We conclude that one of the major physiological activities of cell-cycle inhibitors is to prevent replicative stress during development.


Assuntos
Proteínas Inibidoras de Quinase Dependente de Ciclina/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Replicação do DNA , Animais , Autorrenovação Celular , Quinase 4 Dependente de Ciclina/fisiologia , Genes Letais , Hemangiossarcoma/genética , Camundongos , Camundongos Knockout , Células-Tronco Neurais/fisiologia , Neoplasias Hipofisárias/genética , Estresse Fisiológico
13.
Cell Cycle ; 14(23): 3734-47, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26654769

RESUMO

Seven-in-absentia homolog (SIAH) proteins are evolutionary conserved RING type E3 ubiquitin ligases responsible for the degradation of key molecules regulating DNA damage response, hypoxic adaptation, apoptosis, angiogenesis, and cell proliferation. Many studies suggest a tumorigenic role for SIAH2. In breast cancer patients SIAH2 expression levels correlate with cancer aggressiveness and overall patient survival. In addition, SIAH inhibition reduced metastasis in melanoma. The role of SIAH1 in breast cancer is still ambiguous; both tumorigenic and tumor suppressive functions have been reported. Other studies categorized SIAH ligases as either pro- or antimigratory, while the significance for metastasis is largely unknown. Here, we re-evaluated the effects of SIAH1 and SIAH2 depletion in breast cancer cell lines, focusing on migration and invasion. We successfully knocked down SIAH1 and SIAH2 in several breast cancer cell lines. In luminal type MCF7 cells, this led to stabilization of the SIAH substrate Prolyl Hydroxylase Domain protein 3 (PHD3) and reduced Hypoxia-Inducible Factor 1α (HIF1α) protein levels. Both the knockdown of SIAH1 or SIAH2 led to increased apoptosis and reduced proliferation, with comparable effects. These results point to a tumor promoting role for SIAH1 in breast cancer similar to SIAH2. In addition, depletion of SIAH1 or SIAH2 also led to decreased cell migration and invasion in breast cancer cells. SIAH knockdown also controlled microtubule dynamics by markedly decreasing the protein levels of stathmin, most likely via p27(Kip1). Collectively, these results suggest that both SIAH ligases promote a migratory cancer cell phenotype and could contribute to metastasis in breast cancer.


Assuntos
Neoplasias da Mama/genética , Movimento Celular/genética , Invasividade Neoplásica/genética , Proteínas Nucleares/fisiologia , Ubiquitina-Proteína Ligases/fisiologia , Apoptose/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Técnicas de Silenciamento de Genes , Humanos , Células MCF-7 , Microtúbulos/metabolismo , Metástase Neoplásica/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Interferência de RNA , Estatmina/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
14.
Proc Natl Acad Sci U S A ; 112(45): 13916-21, 2015 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-26512117

RESUMO

The cyclin-dependent kinase (CDK) inhibitor p27(kip1) is a critical regulator of the G1/S-phase transition of the cell cycle and also regulates microtubule (MT) stability. This latter function is exerted by modulating the activity of stathmin, an MT-destabilizing protein, and by direct binding to MTs. We recently demonstrated that increased proliferation in p27(kip1)-null mice is reverted by concomitant deletion of stathmin in p27(kip1)/stathmin double-KO mice, suggesting that a CDK-independent function of p27(kip1) contributes to the control of cell proliferation. Whether the regulation of MT stability by p27(kip1) impinges on signaling pathway activation and contributes to the decision to enter the cell cycle is largely unknown. Here, we report that faster cell cycle entry of p27(kip1)-null cells was impaired by the concomitant deletion of stathmin. Using gene expression profiling coupled with bioinformatic analyses, we show that p27(kip1) and stathmin conjunctly control activation of the MAPK pathway. From a molecular point of view, we observed that p27(kip1), by controlling MT stability, impinges on H-Ras trafficking and ubiquitination levels, eventually restraining its full activation. Our study identifies a regulatory axis controlling the G1/S-phase transition, relying on the regulation of MT stability by p27(kip1) and finely controlling the spatiotemporal activation of the Ras-MAPK signaling pathway.


Assuntos
Ciclo Celular , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Microtúbulos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Animais , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Ativação Enzimática , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Estatmina/metabolismo
15.
Cell Cycle ; 14(13): 2109-20, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26030190

RESUMO

UNLABELLED: Cancer stem-like cells (CSCs) are a rare subpopulation of cancer cells capable of propagating the disease and causing cancer recurrence. In this study, we found that the cellular localization of PKB/Akt kinase affects the maintenance of CSCs. When Akt tagged with nuclear localization signal (Akt-NLS) was overexpressed in SKBR3 and MDA-MB468 cells, these cells showed a 10-15% increase in the number of cells with CSCs enhanced ALDH activity and demonstrated a CD44(+High)/CD24(-Low) phenotype. This effect was completely reversed in the presence of Akt-specific inhibitor, triciribine. Furthermore, cells overexpressing Akt or Akt-NLS were less likely to be in G0/G1 phase of the cell cycle by inactivating p21(Waf1/Cip1) and exhibited increased clonogenicity and proliferation as assayed by colony-forming assay (mammosphere formation). Thus, our data emphasize the importance the intracellular localization of Akt has on stemness in human breast cancer cells. It also indicates a new robust way for improving the enrichment and culture of CSCs for experimental purposes. Hence, it allows for the development of simpler protocols to study stemness, clonogenic potency, and screening of new chemotherapeutic agents that preferentially target cancer stem cells. SUMMARY: The presented data, (i) shows new, stemness-promoting role of nuclear Akt/PKB kinase, (ii) it underlines the effects of nuclear Akt on cell cycle regulation, and finally (iii) it suggests new ways to study cancer stem-like cells.


Assuntos
Neoplasias da Mama/metabolismo , Núcleo Celular/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Células-Tronco Neoplásicas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/química , Feminino , Humanos , Proteínas Proto-Oncogênicas c-akt/análise
16.
J Neurochem ; 134(5): 879-91, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26086369

RESUMO

Zinc plays an important role in the development and maintenance of central neural system. Zinc deficiency has been known to alter normal brain function, whose molecular mechanism remains largely elusive. In the present study, we established a zinc deficiency-exposed rat model, and, using western blot and immunohistochemical analyses, found that the expression of FoxO3a and p27(kip1) was remarkably up-regulated in the rat brain hippocampus. Immunofluorescence assay showed that FOXO3a and p27(kip1) were significantly co-localized with nestin, the marker of neural stem cells (NSCs). Furthermore, we identified that the proportion of proliferating NSCs was markedly decreased in zinc-deficient rat hippocampaus. Using C17.2 neural stem cells, it was revealed that exposure to zinc chelator N,N,N',N'-tetrakis-(2-pyridylmethy) ethylenediamine induced the expression of FoxO3a and p27(kip1) , which coincided with reduced NSC proliferation. Furthermore, depletion of FoxO3a inhibited p27(kip1) expression and restored the growth of NSCs. On the basis of these data, we concluded that FoxO3a/p27(kip1) signaling might play a significant role in zinc deficiency-induced growth impairment of NSCs and consequent neurological disorders. We describe here that zinc deficiency induces the proliferative impairment of hippocampal neural stem cells partially through the activation of FOXO3a-p27 axis in rats. Neural progenitor cells exhibited significantly up-regulated expression of FOXO3a and p27 after zinc deficiency in vivo and in vitro. Depletion of FOXO3a ameliorates zinc deficiency-induced expression of p27 and growth impairment of neural stem cells. We provide novel insight into the mechanisms underlying zinc deficiency-induced neurological deficits.


Assuntos
Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Fatores de Transcrição Forkhead/fisiologia , Hipocampo/patologia , Células-Tronco Neurais/patologia , Zinco/deficiência , Animais , Ciclo Celular , Divisão Celular , Quelantes/farmacologia , Inibidor de Quinase Dependente de Ciclina p27/biossíntese , Inibidor de Quinase Dependente de Ciclina p27/genética , Etilenodiaminas/farmacologia , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/antagonistas & inibidores , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Técnicas de Silenciamento de Genes , Hipocampo/metabolismo , Masculino , Nestina/análise , Células-Tronco Neurais/metabolismo , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Regulação para Cima , Zinco/fisiologia
18.
Mol Cell Endocrinol ; 412: 205-15, 2015 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-25960164

RESUMO

Our group has previously shown that corticotropin releasing factor (CRF) inhibits proliferation of human endocrine-related cancer cell lines via the activation of CRF type-1 receptors (CRF-R1). Tumors originating from the nervous system also express CRF receptors but their role on neoplastic cell proliferation was poorly investigated. Here we investigated the effect of CRF receptor stimulation on nervous system-derived cancer cells, using the SK-N-SH (N) human neuroblastoma cell line as an experimental model. We found that SK-N-SH (N) cells express functionally active CRF-R1, whose activation by CRF and the cognate peptide urocortin (UCN) is associated to reduced cell proliferation and motility, as well as neuronal-like differentiation. UCN did not interfere with cell viability and cell-cycle arrest. Those effects seem to be mediated by a mechanism involving the activation of cAMP/PKA/CREB pathway and the subsequent downstream increase in p27(Kip1) and underphosphorylated retinoblastoma protein levels, as well as reduced c-Myc mRNA accumulation.


Assuntos
Diferenciação Celular , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Movimento Celular , Hormônio Liberador da Corticotropina/fisiologia , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Humanos , Neuroblastoma , Proteínas Proto-Oncogênicas c-myc/genética , Proteína do Retinoblastoma/metabolismo , Urocortinas/fisiologia
19.
Oncogene ; 34(4): 496-505, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-24469036

RESUMO

Many oncogenic viruses activate nuclear factor-κB (NF-κB) as a part of their replicative cycles. We have shown recently that persistent and potentially oncogenic activation of NF-κB by the human T-lymphotropic virus 1 (HTLV-1) oncoprotein Tax immediately triggers a host senescence response mediated by cyclin-dependent kinase inhibitors: p21(CIP1/WAF1) (p21) and p27(Kip1) (p27) Here we demonstrate that RelA/NF-κB activation by Kaposi sarcoma herpesvirus (KSHV) latency protein vFLIP also leads to p21/p27 upregulation and G1 cell cycle arrest. Remarkably, KSHV vCyclin, another latency protein coexpressed with vFLIP from a bicistronic latency-specific mRNA, was found to prevent the senescence and G1 arrest induced by HTLV-1 Tax and vFLIP, respectively. This is because of the known ability of vCyclin/cyclin-dependent kinase 6 complex to resist p21 and p27 inhibition and cause p27 degradation. In KSHV-transformed BCBL-1 cells, sustained vFLIP expression with small hairpin RNAs-mediated vCyclin depletion resulted in G1 arrest. The functional interdependence of vFLIP and vCyclin explains why they are cotranslated from the same viral mRNA. Importantly, deregulation of the G1 cyclin-dependent kinase can facilitate chronic I-κB kinases/NF-κB activation.


Assuntos
Pontos de Checagem da Fase G1 do Ciclo Celular , NF-kappa B/metabolismo , Proteínas Virais/fisiologia , Quinase 6 Dependente de Ciclina/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Produtos do Gene tax , Células HeLa , Humanos , Quinase I-kappa B/fisiologia
20.
In Vitro Cell Dev Biol Anim ; 50(8): 778-85, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24764126

RESUMO

Cell transplantation therapy with oligodendrocyte precursor cells (OPCs) is a promising and effective treatment for diseases involving demyelination in the central nervous system (CNS). In previous studies, we succeeded in producing O4(+) oligodendrocytes (OLs) from mouse- and human-induced pluripotent stem cells (iPSCs) in vitro; however, the efficiency of differentiation into OLs was lower for iPSCs than that for embryonic stem cells (ESCs). To clarify the cause of this difference, we compared the expression of proteins that contribute to OL differentiation in mouse iPSC-derived cells and in mouse ESC-derived cells. The results showed that the expression levels of cyclin dependent kinase inhibitor P27/Kip1, mitogen-activated protein kinase (MAPK) JNK3, and transcription factor Mash1 were lower in iPSC-derived cells. In contrast, the expression levels of MAPK P38α, P38γ, and thyroid hormone receptor ß1 were higher in iPSC-derived cells. We attempted to compensate for the expression changes in P27/Kip1 protein and Mash1 protein in iPSC-derived cells through retrovirus vector-mediated gene expression. Although the overexpression of Mash1 had no effect, the overexpression of P27/Kip1 increased the differentiation efficiency of iPSC-derived cells into O4(+) OLs.


Assuntos
Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Oligodendroglia/fisiologia , Animais , Western Blotting , Diferenciação Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p27/biossíntese , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Expressão Gênica/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Camundongos , Oligodendroglia/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA