Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biochem Pharmacol ; 190: 114616, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34022189

RESUMEN

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.


Asunto(s)
Antraciclinas/farmacología , Roturas del ADN de Doble Cadena/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Factor 3 de Iniciación Eucariótica/biosíntesis , Animales , Antraciclinas/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Reparación del ADN/fisiología , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/fisiología , Factor 3 de Iniciación Eucariótica/genética , Femenino , Estudios de Seguimiento , Células HeLa , Humanos , Células MCF-7 , Ratones , Células 3T3 NIH
2.
Science ; 370(6518): 853-856, 2020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-33184215

RESUMEN

Shutoff of global protein synthesis is a conserved response to cellular stresses. This general phenomenon is accompanied by the induction of distinct gene programs tailored to each stress. Although the mechanisms driving repression of general protein synthesis are well characterized, how cells reprogram the translation machinery for selective gene expression remains poorly understood. Here, we found that the noncanonical 5' cap-binding protein eIF3d was activated in response to metabolic stress in human cells. Activation required reduced CK2-mediated phosphorylation near the eIF3d cap-binding pocket. eIF3d controls a gene program enriched in factors important for glucose homeostasis, including members of the mammalian target of rapamycin (mTOR) pathway. eIF3d-directed translation adaptation was essential for cell survival during chronic glucose deprivation. Thus, this mechanism of translation reprogramming regulates the cellular response to metabolic stress.


Asunto(s)
Factor 3 de Iniciación Eucariótica/biosíntesis , Glucosa/deficiencia , Biosíntesis de Proteínas , Estrés Fisiológico , Adaptación Fisiológica , Supervivencia Celular , Factor 3 de Iniciación Eucariótica/genética , Células HEK293 , Humanos , Fosforilación , Serina-Treonina Quinasas TOR/metabolismo
3.
Metab Eng ; 59: 98-105, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32061967

RESUMEN

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.


Asunto(s)
Factor 3 de Iniciación Eucariótica , Regulación de la Expresión Génica , Proteínas Proto-Oncogénicas c-myc , Animales , Células CHO , Cricetulus , Factor 3 de Iniciación Eucariótica/biosíntesis , Factor 3 de Iniciación Eucariótica/genética , Células HEK293 , Humanos , Ratones , Células 3T3 NIH , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética
4.
Nucleic Acids Res ; 48(7): 3816-3831, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-31996915

RESUMEN

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.


Asunto(s)
Factor 3 de Iniciación Eucariótica/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Ováricas/genética , Biosíntesis de Proteínas , Proteínas de Unión al ARN/metabolismo , Adenosina/análogos & derivados , Animales , Carcinogénesis , Línea Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Factor 3 de Iniciación Eucariótica/biosíntesis , Femenino , Humanos , Ratones Desnudos , Invasividad Neoplásica , Metástasis de la Neoplasia , Oncogenes , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , ARN Mensajero/química , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/fisiología
5.
Cell Death Dis ; 10(9): 623, 2019 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-31423012

RESUMEN

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.


Asunto(s)
Factor 3 de Iniciación Eucariótica/biosíntesis , Neoplasias Gástricas/metabolismo , Animales , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Línea Celular Tumoral , Proliferación Celular/fisiología , Progresión de la Enfermedad , Regulación hacia Abajo , Factor 3 de Iniciación Eucariótica/genética , Factor 3 de Iniciación Eucariótica/metabolismo , Femenino , Gastritis/genética , Gastritis/metabolismo , Gastritis/patología , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/patología , Helicobacter pylori/genética , Helicobacter pylori/metabolismo , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Metástasis de la Neoplasia , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Transducción de Señal/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/patología , Transfección
6.
J Biol Chem ; 294(7): 2267-2278, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30573685

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptor alfa de Estrógeno/metabolismo , Factor 3 de Iniciación Eucariótica/biosíntesis , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/metabolismo , Transducción de Señal , Regiones no Traducidas 3' , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proliferación Celular , Supervivencia Celular , Receptor alfa de Estrógeno/genética , Factor 3 de Iniciación Eucariótica/genética , Femenino , Humanos , Células MCF-7 , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas de Neoplasias/genética
7.
Tissue Cell ; 53: 23-29, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30060823

RESUMEN

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.


Asunto(s)
Factor 3 de Iniciación Eucariótica/biosíntesis , Fase G1 , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/biosíntesis , Fase de Descanso del Ciclo Celular , Neoplasias Gástricas/metabolismo , Regulación hacia Arriba , Línea Celular Tumoral , Factor 3 de Iniciación Eucariótica/genética , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Proteínas de Neoplasias/genética , Neoplasias Gástricas/genética
8.
Oncotarget ; 7(14): 18541-57, 2016 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-26988917

RESUMEN

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.


Asunto(s)
Clusterina/metabolismo , Factor 3 de Iniciación Eucariótica/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Línea Celular Tumoral , Factor 3 de Iniciación Eucariótica/genética , Factor 3 de Iniciación Eucariótica/metabolismo , Factor 3 de Iniciación Eucariótica/farmacología , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Transducción de Señal , Transfección
9.
Mol Cancer Res ; 13(10): 1421-30, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26056130

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/metabolismo , Factor 3 de Iniciación Eucariótica/biosíntesis , Neoplasias Pulmonares/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/fisiología , Transición Epitelial-Mesenquimal , Factor 3 de Iniciación Eucariótica/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Transducción de Señal
10.
Oncol Rep ; 33(6): 2954-62, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25823503

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas/genética , Proliferación Celular/genética , Factor 3 de Iniciación Eucariótica/genética , Glioma/genética , Adolescente , Adulto , Anciano , Apoptosis/genética , Neoplasias Encefálicas/patología , Carcinogénesis , Supervivencia sin Enfermedad , Factor 3 de Iniciación Eucariótica/antagonistas & inhibidores , Factor 3 de Iniciación Eucariótica/biosíntesis , Femenino , Regulación Neoplásica de la Expresión Génica , Glioma/patología , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , ARN Mensajero/biosíntesis
11.
Eur J Hum Genet ; 23(11): 1573-80, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25669430

RESUMEN

Type 1 narcolepsy, an autoimmune disease affecting hypocretin (orexin) neurons, is strongly associated with HLA-DQB1*06:02. Among polymorphisms associated with the disease is single-nucleotide polymorphism rs2305795 (c.*638G>A) located within the P2RY11 gene. P2RY11 is in a region of synteny conserved in mammals and zebrafish containing PPAN, EIF3G and DNMT1 (DNA methyltransferase 1). As mutations in DNMT1 cause a rare dominant form of narcolepsy in association with deafness, cerebellar ataxia and dementia, we questioned whether the association with P2RY11 in sporadic narcolepsy could be secondary to linkage disequilibrium with DNMT1. Based on genome-wide association data from two cohorts of European and Chinese ancestry, we found that the narcolepsy association signal drops sharply between P2RY11/EIF3G and DNMT1, suggesting that the association with narcolepsy does not extend into the DNMT1 gene region. Interestingly, using transethnic mapping, we identified a novel single-nucleotide polymorphism rs3826784 (c.596-260A>G) in the EIF3G gene also associated with narcolepsy. The disease-associated allele increases EIF3G mRNA expression. EIF3G is located in the narcolepsy risk locus and EIF3G expression correlates with PPAN and P2RY11 expression. This suggests shared regulatory mechanisms that might be affected by the polymorphism and are of relevance to narcolepsy.


Asunto(s)
Factor 3 de Iniciación Eucariótica/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Narcolepsia/genética , Alelos , Animales , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , Etnicidad/genética , Factor 3 de Iniciación Eucariótica/biosíntesis , Femenino , Regulación de la Expresión Génica , Cadenas beta de HLA-DQ/genética , Humanos , Masculino , Mutación , Narcolepsia/patología , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Polimorfismo de Nucleótido Simple , Receptores Purinérgicos P2/biosíntesis , Receptores Purinérgicos P2/genética
12.
FASEB J ; 28(6): 2696-704, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24558198

RESUMEN

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.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Factor 3 de Iniciación Eucariótica/genética , Neoplasias Pulmonares/metabolismo , MicroARNs/fisiología , Regiones no Traducidas 3'/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo , Factor 3 de Iniciación Eucariótica/biosíntesis , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/biosíntesis , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba
13.
Methods Enzymol ; 536: 133-47, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24423273

RESUMEN

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.


Asunto(s)
Saccharomyces cerevisiae/metabolismo , Tampones (Química) , Fraccionamiento Celular , Factor 3 de Iniciación Eucariótica/biosíntesis , Factor 3 de Iniciación Eucariótica/genética , Factor 3 de Iniciación Eucariótica/aislamiento & purificación , Expresión Génica , Regiones Promotoras Genéticas , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Saccharomyces cerevisiae/crecimiento & desarrollo , Proteínas de Saccharomyces cerevisiae/biosíntesis , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/aislamiento & purificación
14.
Gene ; 532(2): 177-85, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24084365

RESUMEN

The gene encoding eIF3g (TaeIF3g), one of the 11 subunits of eukaryotic translation initiation factor 3 (eIF3), was cloned from wheat for carrying out its functional analysis. Transgenic expression of TaeIF3g enhanced the tolerance of TaeIF3g-overexpressing parental yeast cells and Arabidopsis plants under different abiotic stress conditions. Compared to untransformed plants, TaeIF3g-overexpressing Arabidopsis thaliana plants exhibited significantly higher survival rate, soluble proteins and photosynthetic efficiency, and enhanced protection against photooxidative stress under drought conditions. This study provides first evidence that TaeIF3g imparts stress tolerance and could be a potential candidate gene for developing crop plants tolerant to abiotic stress.


Asunto(s)
Arabidopsis/fisiología , Factor 3 de Iniciación Eucariótica/genética , Proteínas de Plantas/genética , Saccharomyces cerevisiae/fisiología , Estrés Fisiológico , Triticum/fisiología , Ácido Abscísico/farmacología , Ácido Abscísico/fisiología , Arabidopsis/genética , Productos Agrícolas/genética , Factor 3 de Iniciación Eucariótica/biosíntesis , Expresión Génica , Germinación , Datos de Secuencia Molecular , Presión Osmótica , Reguladores del Crecimiento de las Plantas/farmacología , Reguladores del Crecimiento de las Plantas/fisiología , Proteínas de Plantas/biosíntesis , Raíces de Plantas/genética , Raíces de Plantas/fisiología , Brotes de la Planta/fisiología , Saccharomyces cerevisiae/genética , Ácido Salicílico/farmacología , Tolerancia a la Sal/genética , Triticum/genética
15.
J Biol Chem ; 288(42): 30087-30093, 2013 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-24003236

RESUMEN

Mammalian eIF3 is composed of 13 subunits and is the largest eukaryotic initiation factor. eIF3 plays a key role in protein biosynthesis. However, it is not fully understood how different subunits contribute to the structural integrity and function of the eIF3 complex. Whether eIF3 is essential for embryonic development and homeostasis is also not known. Here, we show that eIF3m null embryos are lethal at the peri-implantation stage. Compound heterozygotes (eIF3m(flox)(/-)) or FABP4-Cre-mediated conditional knock-out mice are lethal at mid-gestation stages. Although the heterozygotes are viable, they show markedly reduced organ size and diminished body weight. Acute ablation of eIF3m in adult mouse liver leads to rapidly decreased body weight and death within 2 weeks; these effects are correlated with a severe decline of protein biogenesis in the liver. Protein analyses reveal that eIF3m deficiency significantly impairs the integrity of the eIF3 complex due to down-regulation of multiple other subunits. Two of the subunits, eIF3f and eIF3h, are stabilized by eIF3m through subcomplex formation. Therefore, eIF3m is required for the structural integrity and translation initiation function of eIF3. Furthermore, not only is eIF3m an essential gene, but its expression level is also important for mouse embryonic development and the control of organ size.


Asunto(s)
Implantación del Embrión/fisiología , Factor 3 de Iniciación Eucariótica/biosíntesis , Regulación del Desarrollo de la Expresión Génica/fisiología , Homeostasis/fisiología , Complejos Multiproteicos/metabolismo , Biosíntesis de Proteínas/fisiología , Animales , Factor 3 de Iniciación Eucariótica/genética , Células HEK293 , Heterocigoto , Humanos , Hígado , Ratones , Ratones Noqueados , Complejos Multiproteicos/genética , Tamaño de los Órganos/fisiología
16.
Hepatology ; 58(1): 239-50, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23460382

RESUMEN

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.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Factor 3 de Iniciación Eucariótica/biosíntesis , Factor 3 de Iniciación Eucariótica/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Proteína Fosfatasa 2/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores
17.
Protein Expr Purif ; 87(1): 5-10, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23063735

RESUMEN

Many biologically important factors are composed of multiple subunits. To study the structure and function of the protein complexes and the role of each subunit, a rapid and efficient method to prepare recombinant protein complexes is needed. In this work, we established an in vitro reconstitution system of eukaryotic translation initiation factor (eIF) 3, a protein complex consisting of 11 distinct subunits. A HeLa cell-derived in vitro coupled transcription/translation system was programmed with multiple plasmids encoding the 11 eIF3 subunits in total. After incubation for several hours, the eIF3 complex was purified through tag-dependent affinity chromatography. When eIF3l, one of the nonessential subunits of eIF3, was not expressed, the eIF3 complex that was devoid of eIF3l was still obtained. Both the 11 subunits complex and the eIF3l-less complex were as active as native eIF3 as observed by a reconstituted translation initiation assay system. In conclusion, the cell-free co-expression system should be a feasible and rapid system to reconstitute protein complexes.


Asunto(s)
Factor 3 de Iniciación Eucariótica/biosíntesis , Biosíntesis de Proteínas , Subunidades de Proteína/biosíntesis , Sistema Libre de Células , Cromatografía de Afinidad , Factor 3 de Iniciación Eucariótica/genética , Factor 3 de Iniciación Eucariótica/aislamiento & purificación , Células HeLa , Humanos , Subunidades de Proteína/genética , Subunidades de Proteína/aislamiento & purificación , Eliminación de Secuencia , Transcripción Genética
18.
Am J Physiol Cell Physiol ; 303(1): C102-11, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22555848

RESUMEN

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.


Asunto(s)
Apoptosis , Gránulos Citoplasmáticos/metabolismo , Proteínas de Choque Térmico/biosíntesis , Mucosa Intestinal/metabolismo , Poliaminas/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/biosíntesis , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Apoptosis/efectos de los fármacos , Arsenitos/farmacología , Línea Celular , Cicloheximida/farmacología , Gránulos Citoplasmáticos/ultraestructura , Eflornitina/farmacología , Células Epiteliales/metabolismo , Factor 3 de Iniciación Eucariótica/biosíntesis , Ornitina Descarboxilasa/biosíntesis , Ornitina Descarboxilasa/genética , Inhibidores de la Ornitina Descarboxilasa , Estrés Oxidativo , Proteínas de Unión a Poli(A)/biosíntesis , Proteínas de Unión a Poli(A)/genética , Interferencia de ARN , ARN Interferente Pequeño , Proteínas de Unión al ARN/biosíntesis , Proteínas de Unión al ARN/metabolismo , Ratas , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
19.
Braz J Med Biol Res ; 43(10): 920-30, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20922269

RESUMEN

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.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Factor 3 de Iniciación Eucariótica/biosíntesis , Regulación Neoplásica de la Expresión Génica/genética , Biosíntesis de Proteínas/genética , Proliferación Celular , Transformación Celular Neoplásica/genética , Factor 3 de Iniciación Eucariótica/genética , Humanos , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética
20.
Anticancer Res ; 30(4): 1047-55, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20530408

RESUMEN

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.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Factor 3 de Iniciación Eucariótica/biosíntesis , Diferenciación Celular/fisiología , Neoplasias Colorrectales/patología , Humanos , Immunoblotting , Inmunohistoquímica , Microscopía Confocal , Microscopía Electrónica , Persona de Mediana Edad , Estadificación de Neoplasias
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...