Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Biochem Pharmacol ; 190: 114616, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34022189

RESUMO

BACKGROUND: Anthracycline are inhibitors of topoisomerase II leading to DNA double strand breaks, and it is widely used for treatment of breast cancer. eIF3a is the largest subunit of eukaryotic translation initiation factor 3 (eIF3) and highly expressed in breast cancer. In this study, we investigated the role of eIF3a in DSB DNA repair and the response of breast cancer patients to anthracycline-based chemotherapy. METHODS: MTT assay was used to detect anthracycline sensitivity in cell lines. Real-time reverse transcriptase PCR, western blotting and immunofluorescence were performed to assess changes in gene expression levels. Cometassay and end-joining activity assay were conducted to explore the effect of eIF3a in NHEJ repair. Luciferase reporter assay was performed to detect LIG4 5'UTR activity. Immunohistochemistry was used to detect eIF3a, LIG4 and DNA-PKcs expression levels in breast cancer tissues. RESULTS: The results showed that eIF3a increased cellular response to anthracyclines by regulating DSB repair activity via influencing the expression of LIG4 and DNA-PKcs at translational level. Breast cancer patients with high level of eIF3a or low level of LIG4 or low level of DNA-PKcs had better anthracycline-based chemotherapy prognosis compared. Moreover, Combined expressions of eIF3a, LIG4 and DNA-PKcs could be better to predict PFS in breast cancer patients with anthracycline-based chemotherapy. CONCLUSION: Our findings suggest that eIF3a effects anthracycline-based chemotherapy response by regulating DSB DNA repair.


Assuntos
Antraciclinas/farmacologia , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Fator de Iniciação 3 em Eucariotos/biossíntese , Animais , Antraciclinas/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Reparo do DNA/fisiologia , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos/fisiologia , Fator de Iniciação 3 em Eucariotos/genética , Feminino , Seguimentos , Células HeLa , Humanos , Células MCF-7 , Camundongos , Células NIH 3T3
2.
Metab Eng ; 59: 98-105, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32061967

RESUMO

There is a desire to engineer mammalian host cell lines to improve cell growth/biomass accumulation and recombinant biopharmaceutical protein production in industrially relevant cell lines such as the CHOK1 and HEK293 cell lines. The over-expression of individual subunits of the eukaryotic translation factor eIF3 in mammalian cells has previously been shown to result in oncogenic properties being imparted on cells, including increased cell proliferation and growth and enhanced global protein synthesis rates. Here we report on the engineering of CHOK1 and HEK cells to over-express the eIF3i and eIF3c subunits of the eIF3 complex and the resultant impact on cell growth and a reporter of exogenous recombinant protein production. Transient over-expression of eIF3i in HEK293 and CHOK1 cells resulted in a modest increase in total eIF3i amounts (maximum 40% increase above control) and an approximate 10% increase in global protein synthesis rates in CHOK1 cells. Stable over-expression of eIF3i in CHOK1 cells was not achievable, most likely due to the already high levels of eIF3i in CHO cells compared to HEK293 cells, but was achieved in HEK293 cells. HEK293 cells engineered to over-express eIF3i had faster growth that was associated with increased c-Myc expression, achieved higher cell biomass and gave enhanced yields of a reporter of recombinant protein production. Whilst CHOK1 cells could not be engineered to over-express eIF3i directly, they could be engineered to over-express eIF3c, which resulted in a subsequent increase in eIF3i amounts and c-Myc expression. The CHOK1 eIF3c engineered cells grew to higher cell numbers and had enhanced cap- and IRES-dependent recombinant protein synthesis. Collectively these data show that engineering of subunits of the eIF3 complex can enhance cell growth and recombinant protein synthesis in mammalian cells in a cell specific manner that has implications for the engineering or selection of fast growing or high producing cells for production of recombinant proteins.


Assuntos
Fator de Iniciação 3 em Eucariotos , Regulação da Expressão Gênica , Proteínas Proto-Oncogênicas c-myc , Animais , Células CHO , Cricetulus , Fator de Iniciação 3 em Eucariotos/biossíntese , Fator de Iniciação 3 em Eucariotos/genética , Células HEK293 , Humanos , Camundongos , Células NIH 3T3 , Proteínas Proto-Oncogênicas c-myc/biossíntese , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética
3.
Nucleic Acids Res ; 48(7): 3816-3831, 2020 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-31996915

RESUMO

N 6-Methyladenosine (m6A) is the most abundant RNA modification in mammal mRNAs and increasing evidence suggests the key roles of m6A in human tumorigenesis. However, whether m6A, especially its 'reader' YTHDF1, targets a gene involving in protein translation and thus affects overall protein production in cancer cells is largely unexplored. Here, using multi-omics analysis for ovarian cancer, we identified a novel mechanism involving EIF3C, a subunit of the protein translation initiation factor EIF3, as the direct target of the YTHDF1. YTHDF1 augments the translation of EIF3C in an m6A-dependent manner by binding to m6A-modified EIF3C mRNA and concomitantly promotes the overall translational output, thereby facilitating tumorigenesis and metastasis of ovarian cancer. YTHDF1 is frequently amplified in ovarian cancer and up-regulation of YTHDF1 is associated with the adverse prognosis of ovarian cancer patients. Furthermore, the protein but not the RNA abundance of EIF3C is increased in ovarian cancer and positively correlates with the protein expression of YTHDF1 in ovarian cancer patients, suggesting modification of EIF3C mRNA is more relevant to its role in cancer. Collectively, we identify the novel YTHDF1-EIF3C axis critical for ovarian cancer progression which can serve as a target to develop therapeutics for cancer treatment.


Assuntos
Fator de Iniciação 3 em Eucariotos/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Ovarianas/genética , Biossíntese de Proteínas , Proteínas de Ligação a RNA/metabolismo , Adenosina/análogos & derivados , Animais , Carcinogênese , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Fator de Iniciação 3 em Eucariotos/biossíntese , Feminino , Humanos , Camundongos Nus , Invasividade Neoplásica , Metástase Neoplásica , Oncogenes , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , RNA Mensageiro/química , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/fisiologia
4.
Cell Death Dis ; 10(9): 623, 2019 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-31423012

RESUMO

Eukaryotic translation initiation factor 3 (eIF3) plays an important role in the regulation of mRNA translation, cell growth and cancer development. eIF3b is the main scaffolding subunit in the eIF3 complex and has been demonstrated to contribute to the development of several cancers. First, our study found that the downregulation of eIF3b could inhibit the proliferation and metastasis of gastric cancer cells by regulating the expression of cancer-related genes. In addition, the expression of eIF3b correlated with the stage and progression of gastric cancer and was shown to be upregulated in human chronic gastritis and in gastric cancer tissues compared with the expression of eIF3b in normal gastric tissues. Moreover, Helicobacter pylori (H. pylori) infection could upregulate the expression of eIF3b in gastric cancer cells, suggesting that eIF3b might be involved in the carcinogenic process of H. pylori. The above findings identified the oncogenic role of eIF3b in gastric cancer development, and this may contribute to the exploration and discovery of novel therapeutic targets for gastric cancer treatment.


Assuntos
Fator de Iniciação 3 em Eucariotos/biossíntese , Neoplasias Gástricas/metabolismo , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Progressão da Doença , Regulação para Baixo , Fator de Iniciação 3 em Eucariotos/genética , Fator de Iniciação 3 em Eucariotos/metabolismo , Feminino , Gastrite/genética , Gastrite/metabolismo , Gastrite/patologia , Infecções por Helicobacter/genética , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/patologia , Helicobacter pylori/genética , Helicobacter pylori/metabolismo , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Metástase Neoplásica , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Transdução de Sinais/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/patologia , Transfecção
5.
J Biol Chem ; 294(7): 2267-2278, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30573685

RESUMO

Approximately two thirds of all breast cancer cases are estrogen receptor (ER)-positive. The treatment of this breast cancer subtype with endocrine therapies is effective in the adjuvant and recurrent settings. However, their effectiveness is compromised by the emergence of intrinsic or acquired resistance. Thus, identification of new molecular targets can significantly contribute to the development of novel therapeutic strategies. In recent years, many studies have implicated aberrant levels of translation initiation factors in cancer etiology and provided evidence that identifies these factors as promising therapeutic targets. Accordingly, we observed reduced levels of the eIF3 subunit eIF3f in ER-positive breast cancer cells compared with ER-negative cells, and determined that low eIF3f levels are required for proper proliferation and survival of ER-positive MCF7 cells. The expression of eIF3f is tightly controlled by ERα at the transcriptional (genomic pathway) and translational (nongenomic pathway) level. Specifically, estrogen-bound ERα represses transcription of the EIF3F gene, while promoting eIF3f mRNA translation. To regulate translation, estrogen activates the mTORC1 pathway, which enhances the binding of eIF3 to the eIF4F complex and, consequently, the assembly of the 48S preinitiation complexes and protein synthesis. We observed preferential translation of mRNAs with highly structured 5'-UTRs that usually encode factors involved in cell proliferation and survival (e.g. cyclin D1 and survivin). Our results underscore the importance of estrogen-ERα-mediated control of eIF3f expression for the proliferation and survival of ER-positive breast cancer cells. These findings may provide rationale for the development of new therapies to treat ER-positive breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Receptor alfa de Estrogênio/metabolismo , Fator de Iniciação 3 em Eucariotos/biossíntese , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/metabolismo , Transdução de Sinais , Regiões 3' não Traduzidas , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proliferação de Células , Sobrevivência Celular , Receptor alfa de Estrogênio/genética , Fator de Iniciação 3 em Eucariotos/genética , Feminino , Humanos , Células MCF-7 , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas de Neoplasias/genética
6.
Tissue Cell ; 53: 23-29, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30060823

RESUMO

Eukaryotic translation initiation factor 3 subunit H (EIF3H) is required for the progression of several types of cancer. However, little is known about the function of EIF3H in gastric carcinoma. To address this issue, in the present study, we investigated EIF3H genetic alterations in and expression of EIF3H in gastric cancer tissue samples using cBioPortal and Oncomine databases. Endogenous EIF3H expression was knocked down in MGC80-3 and AGS gastric cancer cell lines by lentivirus-mediated RNA interference. We confirmed the knockdown efficiency by quantitative real-time PCR and western blotting and evaluated the effects of EIF3H silencing on cell proliferation of gastric cancer with the cell viability and colony formation assays and by flow cytometry. The OncoPrint of EIF3H generated using cBioPortal indicated that EIF3H genetic alterations (mutation, deletion and amplification) were present in two gastric cancer sample sets. The Oncomine analysis revealed that EIF3H mRNA level was upregulated in gastric cancer tissues. EIF3H knockdown inhibited cell proliferation and colony formation in gastric cancer lines and led to cell cycle arrest at the G0/G1 phase, while inducing apoptosis via up- and downregulation of pro- and anti-apoptotic factors, respectively. These results indicate that EIF3H can serve as a novel therapeutic target for the clinical treatment of gastric cancer.


Assuntos
Fator de Iniciação 3 em Eucariotos/biossíntese , Fase G1 , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/biossíntese , Fase de Repouso do Ciclo Celular , Neoplasias Gástricas/metabolismo , Regulação para Cima , Linhagem Celular Tumoral , Fator de Iniciação 3 em Eucariotos/genética , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Proteínas de Neoplasias/genética , Neoplasias Gástricas/genética
7.
Oncotarget ; 7(14): 18541-57, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-26988917

RESUMO

Clusterin is a secretory heterodimeric glycoprotein and the overexpression of secretory clusterin (sCLU) promotes cancer cell proliferation and reduces chemosensitivity. Therefore, sCLU might be an effective target for anticancer therapy. In the current study, we identified eIF3f as a novel CLU-interacting protein and demonstrated its novel function as a CLU inhibitor. The overexpression of eIF3f retarded cancer cell growth significantly and induced apoptosis. In addition, eIF3f interacted with the α-chain (1-227) of sCLU. This interaction blocked modification of psCLU, thereby decreasing the expression and secretion of α/ß CLU. Consequently, the overexpression of eIF3f suppressed Akt and ERK signaling and subsequently depleted CLU expression. In addition, eIF3F stabilized p53, which increased the expression of p21 and Bax. Interestingly, the expression of Bax was increased without the activation of p53. eIF3f injected into a xenograft model of human cervical cancer in nude mice markedly inhibited tumor growth. The identification of this novel function of eIF3f as a sCLU inhibitor might open novel avenues for developing improved strategies for CLU-targeted anti-cancer therapies.


Assuntos
Clusterina/metabolismo , Fator de Iniciação 3 em Eucariotos/biossíntese , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Linhagem Celular Tumoral , Fator de Iniciação 3 em Eucariotos/genética , Fator de Iniciação 3 em Eucariotos/metabolismo , Fator de Iniciação 3 em Eucariotos/farmacologia , Técnicas de Silenciamento de Genes , Células HEK293 , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Transdução de Sinais , Transfecção
8.
Mol Cancer Res ; 13(10): 1421-30, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26056130

RESUMO

UNLABELLED: The eIF3e protein is a component of the multisubunit eIF3 complex, which is essential for cap-dependent translation initiation. Decreased eIF3e expression is often observed in breast and lung cancer and has been shown to induce epithelial-to-mesenchymal transition (EMT) in breast epithelial cells by an unknown mechanism. Here, we study the effect of decreased eIF3e expression in lung epithelial cells by creating stable clones of lung epithelial cells (A549) that express an eIF3e-targeting shRNA. Our data indicate that decreased eIF3e expression in lung epithelial cells leads to EMT, as it does in breast epithelial cells. Importantly, we show that decreased eIF3e expression in both lung and breast epithelial cells leads to the overproduction of the TGFß cytokine and that inhibition of TGFß signaling can reverse eIF3e-regulated EMT in lung epithelial cells. In addition, we discovered that several mRNAs that encode important EMT regulators are translated by a cap-independent mechanism when eIF3e levels are reduced. These findings indicate that EMT mediated by a decrease in eIF3e expression may be a general phenomenon in epithelial cells and that it requires activation and maintenance of the TGFß signaling pathway. IMPLICATIONS: These results indicate that inhibition of TGFß signaling could be an efficient way to prevent metastasis in patients with NSCLC that display reduced eIF3e expression.


Assuntos
Neoplasias da Mama/metabolismo , Fator de Iniciação 3 em Eucariotos/biossíntese , Neoplasias Pulmonares/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Transição Epitelial-Mesenquimal , Fator de Iniciação 3 em Eucariotos/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Transdução de Sinais
9.
Oncol Rep ; 33(6): 2954-62, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25823503

RESUMO

Eukaryotic initiation factor 3, subunit c (eIF3c), an oncogene overexpressed in human cancers, plays an important role in cell tumorigenesis and proliferation. However, studies assessing its function in gliomas are scarce. The present study evaluated for the first time, the role of eIF3c in gliomas. Immunohistochemistry was carried out to assess eIF3c expression in 95 human glioma samples and normal brain tissues. Then, the eIF3c mRNA levels were detected in tumor and normal brain specimens by quantitative RT-PCR. In addition, eIF3c mRNA levels were assessed in four glioma cell lines (U87, U251, A172 and U373) by semi-quantitative RT-PCR. The RNA interference (RNAi) technology was employed to knock down the eIF3c gene in the U251 cells. Western blot analysis, BrdU assay and flow cytometry were used to measure eIF3c protein levels, cell proliferation, cell apoptosis and cell cycle, respectively. The eIF3c protein was overexpressed in the human glioma specimens. In agreement, the eIF3c mRNA expression levels were significantly higher in the human glioma tissues compared with the normal brain samples (P<0.0001). In addition, eIF3c mRNA was detected in all the glioma cell lines. Silencing the eIF3c gene in the U251 cells by RNAi significantly suppressed cell proliferation (P<0.01) and increased apoptosis (P<0.01). Finally, a stark decrease was observed in the G1 phase cell number (P<0.01), while the S and G2 phase cells were significantly increased (P<0.01) after eIF3c knockdown. These findings suggest that eIF3c is overexpressed in human gliomas and essential for their proliferation and survival. Therefore, inhibiting eIF3c expression may constitute an effective therapy for human glioma.


Assuntos
Neoplasias Encefálicas/genética , Proliferação de Células/genética , Fator de Iniciação 3 em Eucariotos/genética , Glioma/genética , Adolescente , Adulto , Idoso , Apoptose/genética , Neoplasias Encefálicas/patologia , Carcinogênese , Intervalo Livre de Doença , Fator de Iniciação 3 em Eucariotos/antagonistas & inibidores , Fator de Iniciação 3 em Eucariotos/biossíntese , Feminino , Regulação Neoplásica da Expressão Gênica , Glioma/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , RNA Mensageiro/biossíntese
10.
FASEB J ; 28(6): 2696-704, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24558198

RESUMO

TGFßR1 plays an important role in TGF-ß signaling transduction and serves as a tumor suppressor. Our previous studies show that reduced expression of TGFßR1 is common in non-small cell lung cancer (NSCLC) and TGFßR1 variants confer risk of NSCLC. However, the epigenetic mechanisms underlying the role of TGFßR1 in NSCLC carcinogenesis are still elusive. We investigated the function and regulation of TGF-ß signaling-based miRNAs in NSCLC. Computational algorithms predicted that the 3'-untranslated region (3'-UTR) of TGFßR1 is a target of miR-142-3p. Here a luciferase reporter assay confirmed that miR-142-3p can directly bind to 3'-UTR of TGFßR1. Overexpression of miR-142-3p in NSCLC A549 cells suppressed expression of TGFßR1 mRNA and protein, while knockdown of endogenous miR-142-3p led to increased expression of TGFßR1. On TGF-ß1 stimulation, stable overexpression of miR-142-3p attenuated phosphorylation of SMAD3, an indispensable downstream effector in canonical TGF-ß/Smad signaling, via repression of TGFßR1 in A549 cells. Furthermore, miR-142-3p-mediated down-regulation of TGFßR1 weakened TGF-ß-induced growth inhibition effect, and this effect was reversed by stable knockdown of endogenous miR-142-3p in A549 cells. In NSCLC tissues, miR-142-3p expression was increased and inversely correlated with TGFßR1 expression. These data demonstrate that miR-142-3p influences the proliferation of NSCLC cells through repression of TGFßR1.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Fator de Iniciação 3 em Eucariotos/genética , Neoplasias Pulmonares/metabolismo , MicroRNAs/fisiologia , Regiões 3' não Traduzidas/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo , Fator de Iniciação 3 em Eucariotos/biossíntese , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/biossíntese , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta1/metabolismo , Regulação para Cima
11.
Methods Enzymol ; 536: 133-47, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24423273

RESUMO

Yeast is an excellent system for the expression of recombinant eukaryotic proteins. Both endogenous and heterologous proteins can be overexpressed in yeast (Phan et al., 2001; Ton and Rao, 2004). Because yeast is easy to manipulate genetically, a strain can be optimized for the expression of a specific protein. Many eukaryotic proteins contain posttranslational modifications that can be performed in yeast but not in bacterial expression systems. In comparison with mammalian cell culture expression systems, growing yeast is both faster and less expensive, and large-scale cultures can be performed using fermentation. While several different yeast expression systems exist, this chapter focuses on the budding yeast Saccharomyces cerevisiae and will briefly describe some options to consider when selecting vectors and tags to be used for protein expression. Throughout this chapter, the expression and purification of yeast eIF3 is shown as an example alongside a general scheme outline.


Assuntos
Saccharomyces cerevisiae/metabolismo , Soluções Tampão , Fracionamento Celular , Fator de Iniciação 3 em Eucariotos/biossíntese , Fator de Iniciação 3 em Eucariotos/genética , Fator de Iniciação 3 em Eucariotos/isolamento & purificação , Expressão Gênica , Regiões Promotoras Genéticas , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Saccharomyces cerevisiae/crescimento & desenvolvimento , Proteínas de Saccharomyces cerevisiae/biossíntese , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/isolamento & purificação
12.
Hepatology ; 58(1): 239-50, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23460382

RESUMO

UNLABELLED: Eukaryotic translation initiation factor 3 subunit I (eIF3I) with transforming capability is often overexpressed in human hepatocellular carcinoma (HCC) but its oncogenic mechanisms remain unknown. We demonstrate that eIF3I is overexpressed in various cancers along with activated Akt1 phosphorylation and kinase activity in an eIF3I dose-dependent manner. A novel eIF3I and Akt1 protein interaction was identified in HCC cell lines and tissues and was required for eIF3I-mediated activation of Akt1 signaling. Expression of either antisense eIF3I or dominant negative Akt1 mutant suppressed eIF3I-mediated Akt1 oncogenic signaling and various other tumorigenic effects. Oncogenic domain mapping of the eIF3I and Akt1 interaction suggested that the C-terminal eIF3I interacted with the Akt1 kinase domain and conferred the majority of oncogenic functions. In addition, eIF3I interaction with Akt1 prevented PP2A dephosphorylation of Akt1 and resulted in constitutively active Akt1 oncogenic signaling. Importantly, concordant expression of endogenous eIF3I and phospho-Akt1 was detected in HCC cell lines and tissues. Treatment of eIF3I overexpressing HCC cells with the Akt1 specific inhibitor API-2 suppressed eIF3I-mediated tumorigenesis in vitro and in vivo. CONCLUSION: We describe a constitutive Akt1 oncogenic mechanism resulting from interaction of overexpressed eIF3I with Akt1 that prevents PP2A-mediated dephosphorylation. Overexpression of eIF3I in HCC is oncogenic and is a surrogate marker and therapeutic target for treatment with Akt1 inhibitors.


Assuntos
Carcinoma Hepatocelular/metabolismo , Fator de Iniciação 3 em Eucariotos/biossíntese , Fator de Iniciação 3 em Eucariotos/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Proteína Fosfatase 2/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores
13.
Am J Physiol Cell Physiol ; 303(1): C102-11, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22555848

RESUMO

Polyamines regulate multiple signaling pathways and are implicated in many aspects of cellular functions, but the exact molecular processes governed by polyamines remain largely unknown. In response to environmental stress, repression of translation is associated with the assembly of stress granules (SGs) that contain a fraction of arrested mRNAs and are thought to function as mRNA storage. Here we show that polyamines modulate the assembly of SGs in normal intestinal epithelial cells (IECs) and that induced SGs following polyamine depletion are implicated in the protection of IECs against apoptosis. Increasing the levels of cellular polyamines by ectopic overexpression of the ornithine decarboxylase gene decreased cytoplasmic levels of SG-signature constituent proteins eukaryotic initiation factor 3b and T-cell intracellular antigen-1 (TIA-1)-related protein and repressed the assembly of SGs induced by exposure to arsenite-induced oxidative stress. In contrast, depletion of cellular polyamines by inhibiting ornithine decarboxylase with α-difluoromethylornithine increased cytoplasmic eukaryotic initiation factor 3b and TIA-1 related protein abundance and enhanced arsenite-induced SG assembly. Polyamine-deficient cells also exhibited an increase in resistance to tumor necrosis factor-α/cycloheximide-induced apoptosis, which was prevented by inhibiting SG formation with silencing SG resident proteins Sort1 and TIA-1. These results indicate that the elevation of cellular polyamines represses the assembly of SGs in normal IECs and that increased SGs in polyamine-deficient cells are crucial for increased resistance to apoptosis.


Assuntos
Apoptose , Grânulos Citoplasmáticos/metabolismo , Proteínas de Choque Térmico/biossíntese , Mucosa Intestinal/metabolismo , Poliaminas/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/biossíntese , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Apoptose/efeitos dos fármacos , Arsenitos/farmacologia , Linhagem Celular , Cicloeximida/farmacologia , Grânulos Citoplasmáticos/ultraestrutura , Eflornitina/farmacologia , Células Epiteliais/metabolismo , Fator de Iniciação 3 em Eucariotos/biossíntese , Ornitina Descarboxilase/biossíntese , Ornitina Descarboxilase/genética , Inibidores da Ornitina Descarboxilase , Estresse Oxidativo , Proteínas de Ligação a Poli(A)/biossíntese , Proteínas de Ligação a Poli(A)/genética , Interferência de RNA , RNA Interferente Pequeno , Proteínas de Ligação a RNA/biossíntese , Proteínas de Ligação a RNA/metabolismo , Ratos , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
14.
Braz J Med Biol Res ; 43(10): 920-30, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20922269

RESUMO

Maintenance of cell homeostasis and regulation of cell proliferation depend importantly on regulating the process of protein synthesis. Many disease states arise when disregulation of protein synthesis occurs. This review focuses on mechanisms of translational control and how disregulation results in cell malignancy. Most translational controls occur during the initiation phase of protein synthesis, with the initiation factors being the major target of regulation through their phosphorylation. In particular, the recruitment of mRNAs through the m7G-cap structure and the binding of the initiator methionyl-tRNA(i) are frequent targets. However, translation, especially of specific mRNAs, may also be regulated by sequestration into processing bodies or stress granules, by trans-acting proteins or by microRNAs. When the process of protein synthesis is hyper-activated, weak mRNAs are translated relatively more efficiently, leading to an imbalance of cellular proteins that promotes cell proliferation and malignant transformation. This occurs, for example, when the cap-binding protein, eIF4E, is overexpressed, or when the methionyl-tRNA(i)-binding factor, eIF2, is too active. In addition, enhanced activity of eIF3 contributes to oncogenesis. The importance of the translation initiation factors as regulators of protein synthesis and cell proliferation makes them potential therapeutic targets for the treatment of cancer.


Assuntos
Transformação Celular Neoplásica/metabolismo , Fator de Iniciação 3 em Eucariotos/biossíntese , Regulação Neoplásica da Expressão Gênica/genética , Biossíntese de Proteínas/genética , Proliferação de Células , Transformação Celular Neoplásica/genética , Fator de Iniciação 3 em Eucariotos/genética , Humanos , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética
15.
Anticancer Res ; 30(4): 1047-55, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20530408

RESUMO

BACKGROUND: P150, a 150 kDa protein, was isolated from virally and oncogene-transformed mouse cell lines, partially purified and cloned. P150 is part of the large subunit of the eukaryotic translation initiation factor 3 with sequence homology to centrosomin A. A significant correlation between p150 expression and malignancy in breast, cervical and esophageal cancer have recently been demonstrated. MATERIALS AND METHODS: Here, 110 colorectal carcinomas of different grades and stages, including lymph node and liver metastases were compared to adjacent normal mucosa by immunohistochemistry of P150. Western blot analysis of selected cases confirmed the expression levels determined by immunohistochemistry. Additionally, immuno-electron and laser scanning microscopy (LSM) was performed. RESULTS: All investigated carcinomas revealed high levels of p150 protein compared to normal adjacent mucosa. The staining intensity was slightly heterogeneous, and positivity was correlated to the tumor grade with statistically significant differences of p150 expression between normal and neoplastic mucosa (p<0.0001, Kruskal-Wallis test). Western blots confirmed higher expression levels of p150 in the tumor. Immunogold labelling and LSM investigation showed high expression levels of p150 on the rough endoplasmic reticulum and polyribosomes, indicating that p150 is translationally active in these tumors. CONCLUSION: Thus, we propose that p150 plays an important role in development and growth of colorectal carcinomas. Furthermore, p150 expression might provide us with reliable information on the biological behaviour of tumors and the clinical course of the disease.


Assuntos
Neoplasias Colorretais/metabolismo , Fator de Iniciação 3 em Eucariotos/biossíntese , Diferenciação Celular/fisiologia , Neoplasias Colorretais/patologia , Humanos , Immunoblotting , Imuno-Histoquímica , Microscopia Confocal , Microscopia Eletrônica , Pessoa de Meia-Idade , Estadiamento de Neoplasias
16.
Proteomics ; 8(17): 3450-68, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18752215

RESUMO

Benzo[a]pyrene (B[a]P) is an ubiquitous environmental carcinogen produced during incomplete combustion of organic substances. Anti-benzo[a]pyrene-7,8-dihydrodiol-9,10-epoxide (BPDE), is the most carcinogenic form of the ultimate metabolites of B[a]P. The goal of this study was to investigate the responses of human amniotic epithelial FL cells to BPDE at different time intervals after exposure and to find potential biomarkers involved in these responses. Cells were treated with 0.05 microM BPDE for 2 h and incubated for another 3, 12, and 24 h to obtain protein extracts which were resolved by 2-DE and visualized by silver staining. Sixty-four spots were up-regulated while 66 were down-regulated following BPDE exposure. These altered spots were excised from the gels and analyzed by MALDI-TOF-MS. The analysis led to the identification of 84 proteins affected by BPDE. These proteins were involved in regulation of transcription, cell cycle, apoptosis, transport, signal transduction, metabolism,and so forth. Among them, subunits of eukaryotic translation initiation factor 3 (EIF3) including EIF3S2, EIF3S3, EIF3S12, and EIF5A, component proteins of ubiquitin-proteasome system (ubiquitin carboxyl-terminal esterase L3, proteasome beta 4 subunit, and proteasome beta 3 subunit) and 14-3-3 proteins (14-3-3 zeta and epsilon) have not been previously associated with a response to BPDE exposure. All these results aid our understanding of the mechanism of BPDE induced cell defensive responses and hazardous effects as well as providing the possibility of the establishment of potential biomarkers.


Assuntos
Proteínas 14-3-3/biossíntese , 7,8-Di-Hidro-7,8-Di-Hidroxibenzo(a)pireno 9,10-óxido/toxicidade , Carcinógenos/toxicidade , Células Epiteliais/efeitos dos fármacos , Fator de Iniciação 3 em Eucariotos/biossíntese , Complexo de Endopeptidases do Proteassoma/biossíntese , Ubiquitina Tiolesterase/biossíntese , Âmnio/citologia , Linhagem Celular , Proteínas do Citoesqueleto/biossíntese , Regulação para Baixo , Eletroforese em Gel Bidimensional , Ribonucleoproteínas Nucleares Heterogêneas Grupo K , Histona-Lisina N-Metiltransferase/biossíntese , Humanos , Ribonucleoproteínas/biossíntese , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Regulação para Cima
17.
J Biol Chem ; 283(35): 24047-60, 2008 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-18544531

RESUMO

Dysregulation of protein synthesis has been implicated in oncogenesis through a mechanism whereby "weak" mRNAs encoding proteins involved in cell proliferation are strongly translated when the protein synthesis apparatus is activated. Previous work has determined that many cancer cells contain high levels of eIF3h, a protein subunit of translation initiation factor eIF3, and overexpression of eIF3h malignantly transforms immortal NIH-3T3 cells. This is a general feature of eIF3h, as high levels also affect translation, proliferation, and a number of malignant phenotypes of CHO-K1 and HeLa cells and, most significantly, of a primary prostate cell line. Furthermore, overexpressed eIF3h inhibits Myc-dependent induction of apoptosis of primary prostate cells. eIF3h appears to function through translation, as the initial appearance of overexpressed eIF3h in rapidly induced NIH-3T3 cells correlates tightly with the stimulation of protein synthesis and the generation of malignant phenotypes. This oncogenic potential of eIF3h is enhanced by phosphorylation at Ser(183). Finally, reduction of eIF3h levels in breast and prostate cancer cell lines by short interfering RNA methods reduces their rates of proliferation and anchorage-independent growth in soft agar. The results provide compelling evidence that high eIF3h levels directly stimulate protein synthesis, resulting in the establishment and maintenance of the malignant state in cells.


Assuntos
Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Fator de Iniciação 3 em Eucariotos/biossíntese , Biossíntese de Proteínas , Subunidades Proteicas/biossíntese , Proteínas Proto-Oncogênicas c-myc/metabolismo , Animais , Apoptose/genética , Células CHO , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Cricetinae , Cricetulus , Fator de Iniciação 3 em Eucariotos/antagonistas & inibidores , Fator de Iniciação 3 em Eucariotos/genética , Células HeLa , Humanos , Camundongos , Células NIH 3T3 , Fosforilação , Biossíntese de Proteínas/genética , Subunidades Proteicas/genética , Proteínas Proto-Oncogênicas c-myc/genética , RNA Interferente Pequeno/genética
18.
Differentiation ; 75(7): 652-61, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17381544

RESUMO

Eukaryotic initiation factor 3a (eIF3a) has been suggested to play a regulatory role in mRNA translation. Decreased eIF3a expression has been observed in differentiated cells while higher levels have been observed in cancer cells. However, whether eIF3a plays any role in differentiation and development is currently unknown. Here, we investigated eIF3a expression during mouse development and its role in differentiation of colon epithelial cells. We found that eIF3a expression was higher in fetal tissues compared with postnatal ones. Its expression in intestine, stomach, and lung abruptly stopped on the 18th day in gestation but persisted in liver, kidney, and heart throughout the postnatal stage at decreased levels. Similarly, eIF3a expression in colon cancer cell lines, HT-29 and Caco-2, drastically decreased prior to differentiation. Enforced eIF3a expression inhibited while knocking it down using small interference RNA promoted Caco-2 differentiation. Thus, eIF3a may play some roles in development and differentiation and that the decreased eIF3a expression may be a pre-requisite of intestinal epithelial cell differentiation.


Assuntos
Diferenciação Celular/fisiologia , Fator de Iniciação 3 em Eucariotos/fisiologia , Mucosa Intestinal/citologia , Mucosa Intestinal/embriologia , Animais , Células CACO-2 , Células Clonais , Colo/citologia , Colo/embriologia , Colo/metabolismo , Fator de Iniciação 3 em Eucariotos/antagonistas & inibidores , Fator de Iniciação 3 em Eucariotos/biossíntese , Fator de Iniciação 3 em Eucariotos/imunologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células HT29 , Humanos , Soros Imunes/biossíntese , Soros Imunes/fisiologia , Mucosa Intestinal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Subunidades Proteicas/fisiologia , Coelhos
19.
Toxicol Lett ; 169(1): 82-90, 2007 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-17275222

RESUMO

In this study, we have investigated effects of benzo(a)pyrene (BP) on growth and functional differentiation of mouse bone marrow (BM)-derived dendritic cells (DC). 1 microM BP dramatically inhibited growth of BM cultured in the presence of granulocyte-macrophage colony stimulating factor (GM-CSF) and interleukin-4 (IL-4). Although little alterations in surface expression of CD11c, major histocompatibility complex (MHC II), and CD86 molecules characteristic of mature DC were induced by BP, production of cytokines including IL-12, IL-10, and TNF-alpha, and allogeneic T cell stimulating ability were severely impaired. Some of the effects of BP were dependent on arylhydrocarbon receptor (AhR), because alpha-naphthoflavone, an AhR antagonist, suppressed the effects of BP on IL-12 production and T cell stimulating ability, but not on DC proliferation. Expression of RelB, a transcription factor necessary for DC differentiation and function, and eIF3 p170, a subunit of eukaryotic translation initiation factor (eIF)3, was reduced upon BP treatment.


Assuntos
Benzo(a)pireno/toxicidade , Células da Medula Óssea/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Fator de Iniciação 3 em Eucariotos/biossíntese , Imunossupressores , Fator de Transcrição RelB/biossíntese , Animais , Antígeno B7-2/biossíntese , Antígeno CD11c/biossíntese , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Citocinas/biossíntese , Regulação para Baixo/efeitos dos fármacos , Fator de Iniciação 3 em Eucariotos/genética , Citometria de Fluxo , Genes MHC da Classe II/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Indicadores e Reagentes , Interleucina-4/biossíntese , Teste de Cultura Mista de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Dibenzodioxinas Policloradas/toxicidade , RNA/biossíntese , RNA/isolamento & purificação , Receptores de Hidrocarboneto Arílico/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Teratogênicos/toxicidade , Fator de Transcrição RelB/genética
20.
J Biol Chem ; 282(8): 5790-800, 2007 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-17170115

RESUMO

Transcriptional and post-transcriptional regulatory mechanisms are commonly accepted paradigms of tumorigenesis. The view is emerging that deregulation of translation contributes importantly to cancer development, a role not generally appreciated before. Eukaryotic initiation factor eIF3 contains at least thirteen non-identical subunits, named from eIF3a to eIF3m, and plays an essential role in the rate-limiting initiation phase of translation. Increased mRNA and protein levels of the eIF3a, -3b, -3c, -3h, and -3i subunits have been detected in a wide variety of human tumors and are frequently identified as prognostic biomarkers for poor clinical outcome. However, it remains to be established whether up-regulation of eIF3 subunits is a consequence or a cause of the malignant phenotypes. Here we report that ectopic expression of eIF3a, -3b, -3c, -3h, or -3i in stably transfected NIH3T3 cells leads to a number of oncogenic properties: decreased doubling times, increased clonogenicity and viability, facilitated S-phase entry, attenuation of apoptosis, formation of transformed foci, and anchorage-independent growth. Only overexpression of the transforming subunits results in a stimulation of initiation and global protein synthesis rates and enhanced translation of poorly translated mRNAs that encode growth-regulating proteins, including cyclinD1, c-Myc, fibroblast growth factor-2, and ornithine decarboxylase, which may be responsible for oncogenic malignancy in the transformed cell lines. Based on these results, we hypothesize that eIF3 contributes to hyperactivation of the translation initiation machinery and thereby may play an important role in neoplasia. Cancer cells appear to require an aberrantly activated translational state to survive, suggesting that the initiation factors may be promising therapeutic targets for treating cancer.


Assuntos
Transformação Celular Neoplásica/metabolismo , Fator de Iniciação 3 em Eucariotos/biossíntese , Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Iniciação Traducional da Cadeia Peptídica , Fase S , Animais , Apoptose/genética , Linhagem Celular Transformada , Sobrevivência Celular/genética , Transformação Celular Neoplásica/genética , Fator de Iniciação 3 em Eucariotos/genética , Fibroblastos/patologia , Expressão Gênica , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Camundongos , Células NIH 3T3 , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Subunidades Proteicas/biossíntese , Subunidades Proteicas/genética , Fase S/genética , Regulação para Cima/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA