Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
2.
Mol Plant Pathol ; 21(3): 303-317, 2020 03.
Article in English | MEDLINE | ID: mdl-31860775

ABSTRACT

Soybean mosaic virus (SMV), a potyvirus, is the most prevalent and destructive viral pathogen in soybean-planting regions of China. Moreover, other potyviruses, including bean common mosaic virus (BCMV) and watermelon mosaic virus (WMV), also threaten soybean farming. The eukaryotic translation initiation factor 4E (eIF4E) plays a critical role in controlling resistance/susceptibility to potyviruses in plants. In the present study, much higher SMV-induced eIF4E1 expression levels were detected in a susceptible soybean cultivar when compared with a resistant cultivar, suggesting the involvement of eIF4E1 in the response to SMV by the susceptible cultivar. Yeast two-hybrid and bimolecular fluorescence complementation assays showed that soybean eIF4E1 interacted with SMV VPg in the nucleus and with SMV NIa-Pro/NIb in the cytoplasm, revealing the involvement of VPg, NIa-Pro, and NIb in SMV infection and multiplication. Furthermore, transgenic soybeans silenced for eIF4E were produced using an RNA interference approach. Through monitoring for viral symptoms and viral titers, robust and broad-spectrum resistance was confirmed against five SMV strains (SC3/7/15/18 and SMV-R), BCMV, and WMV in the transgenic plants. Our findings represent fresh insights for investigating the mechanism underlying eIF4E-mediated resistance in soybean and also suggest an effective alternative for breeding soybean with broad-spectrum viral resistance.


Subject(s)
Glycine max/genetics , Glycine max/virology , Potyvirus/immunology , RNA Interference/immunology , Disease Resistance/genetics , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/physiology , Plants, Genetically Modified/genetics , Plants, Genetically Modified/virology
3.
Med Sci Monit ; 25: 7936-7941, 2019 Oct 23.
Article in English | MEDLINE | ID: mdl-31641096

ABSTRACT

BACKGROUND Eukaryotic initiation factor 4E (eIF4E) has been reported to act as a prognostic biomarker in various cancers, but its actual effect on basal cell cancer (BCC) of the skin is rarely reported. Our research measured eIF4E levels and discussed its consequence in BCC of the skin. MATERIAL AND METHODS Semi-quantitative real-time polymerase chain reaction (RT-PCR) and western blotting analysis were used to detect relative expression level of eIF4E in specimens at both mRNA and protein levels. The relationship of eIF4E level with clinical profiles was analyzed via chi-square test. Additionally, prognostic value of eIF4E was analyzed via Kaplan-Meier and cox regression analysis. RESULTS We found that eIF4E was over-expressed in tumor tissues, in comparison to bordering cancer-free tissue samples. Besides, elevated eIF4E level exhibited a strong relation to metastasis, TNM stage, and differentiation. Kaplan-Meier analysis revealed cases harboring high eIF4E levels faced shortened overall survival compared to cases of low levels (log rank test, P=0.018). Moreover, eIF4E could act as an independent biomarker for the prognosis of BCC of the skin, according to Cox regression analysis. CONCLUSIONS The level of eIF4E was upregulated and significantly correlated with the development of BCC of the skin. Thus, it might be a promising prognostic biomarker and therapy target for BCC of the skin.


Subject(s)
Eukaryotic Initiation Factor-4E/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/mortality , Adult , Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Basal Cell/metabolism , Carcinoma, Basal Cell/mortality , China , Eukaryotic Initiation Factor-4E/physiology , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Recurrence, Local/metabolism , Neoplasms, Basal Cell , Prognosis , Skin/pathology
4.
Brain ; 142(10): 3158-3175, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31365052

ABSTRACT

Huntington's disease is a neurodegenerative disorder caused by a CAG repeat expansion in exon 1 of the huntingtin gene. Striatal projection neurons are mainly affected, leading to motor symptoms, but molecular mechanisms involved in their vulnerability are not fully characterized. Here, we show that eIF4E binding protein (4E-BP), a protein that inhibits translation, is inactivated in Huntington's disease striatum by increased phosphorylation. Accordingly, we detected aberrant de novo protein synthesis. Proteomic characterization indicates that translation specifically affects sets of proteins as we observed upregulation of ribosomal and oxidative phosphorylation proteins and downregulation of proteins related to neuronal structure and function. Interestingly, treatment with the translation inhibitor 4EGI-1 prevented R6/1 mice motor deficits, although corticostriatal long-term depression was not markedly changed in behaving animals. At the molecular level, injection of 4EGI-1 normalized protein synthesis and ribosomal content in R6/1 mouse striatum. In conclusion, our results indicate that dysregulation of protein synthesis is involved in mutant huntingtin-induced striatal neuron dysfunction.


Subject(s)
Eukaryotic Initiation Factor-4E/physiology , Huntington Disease/genetics , Protein Biosynthesis/physiology , Animals , Behavior, Animal , Corpus Striatum/metabolism , Disease Models, Animal , Eukaryotic Initiation Factor-4E/genetics , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Interneurons/metabolism , Male , Mice , Mice, Transgenic , Neostriatum/pathology , Nerve Degeneration/pathology , Neurons/metabolism , Nuclear Proteins/genetics , Phosphorylation , Proteomics
5.
Nucleic Acids Res ; 47(13): 7035-7048, 2019 07 26.
Article in English | MEDLINE | ID: mdl-31114929

ABSTRACT

The eIF4E-homologous protein (4EHP) is a translational repressor that competes with eIF4E for binding to the 5'-cap structure of specific mRNAs, to which it is recruited by protein factors such as the GRB10-interacting GYF (glycine-tyrosine-phenylalanine domain) proteins (GIGYF). Several experimental evidences suggest that GIGYF proteins are not merely facilitating 4EHP recruitment to transcripts but are actually required for the repressor activity of the complex. However, the underlying molecular mechanism is unknown. Here, we investigated the role of the uncharacterized Drosophila melanogaster (Dm) GIGYF protein in post-transcriptional mRNA regulation. We show that, when in complex with 4EHP, Dm GIGYF not only elicits translational repression but also promotes target mRNA decay via the recruitment of additional effector proteins. We identified the RNA helicase Me31B/DDX6, the decapping activator HPat and the CCR4-NOT deadenylase complex as binding partners of GIGYF proteins. Recruitment of Me31B and HPat via discrete binding motifs conserved among metazoan GIGYF proteins is required for downregulation of mRNA expression by the 4EHP-GIGYF complex. Our findings are consistent with a model in which GIGYF proteins additionally recruit decapping and deadenylation complexes to 4EHP-containing RNPs to induce translational repression and degradation of mRNA targets.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/genetics , Eukaryotic Initiation Factor-4E/physiology , Gene Expression Regulation , RNA Cap-Binding Proteins/physiology , RNA, Messenger/genetics , Repressor Proteins/physiology , Amino Acid Sequence , Animals , Conserved Sequence , DEAD-box RNA Helicases/physiology , Down-Regulation , Endopeptidases/physiology , Genes, Reporter , Multiprotein Complexes , Protein Biosynthesis , RNA Caps/genetics , RNA Caps/metabolism , RNA Stability/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/physiology , Ribonucleases/physiology , Sequence Alignment , Sequence Homology, Amino Acid
6.
Oncogene ; 38(13): 2241-2262, 2019 03.
Article in English | MEDLINE | ID: mdl-30478448

ABSTRACT

The poor outcomes in infant acute lymphoblastic leukemia (ALL) necessitate new treatments. Here we discover that EIF4E protein is elevated in most cases of infant ALL and test EIF4E targeting by the repurposed antiviral agent ribavirin, which has anticancer properties through EIF4E inhibition, as a potential treatment. We find that ribavirin treatment of actively dividing infant ALL cells on bone marrow stromal cells (BMSCs) at clinically achievable concentrations causes robust proliferation inhibition in proportion with EIF4E expression. Further, we find that ribavirin treatment of KMT2A-rearranged (KMT2A-R) infant ALL cells and the KMT2A-AFF1 cell line RS4:11 inhibits EIF4E, leading to decreases in oncogenic EIF4E-regulated cell growth and survival proteins. In ribavirin-sensitive KMT2A-R infant ALL cells and RS4:11 cells, EIF4E-regulated proteins with reduced levels of expression following ribavirin treatment include MYC, MCL1, NBN, BCL2 and BIRC5. Ribavirin-treated RS4:11 cells exhibit impaired EIF4E-dependent nuclear to cytoplasmic export and/or translation of the corresponding mRNAs, as well as reduced phosphorylation of the p-AKT1, p-EIF4EBP1, p-RPS6 and p-EIF4E signaling proteins. This leads to an S-phase cell cycle arrest in RS4:11 cells corresponding to the decreased proliferation. Ribavirin causes nuclear EIF4E to re-localize to the cytoplasm in KMT2A-AFF1 infant ALL and RS4:11 cells, providing further evidence for EIF4E inhibition. Ribavirin slows increases in peripheral blasts in KMT2A-R infant ALL xenograft-bearing mice. Ribavirin cooperates with chemotherapy, particularly L-asparaginase, in reducing live KMT2A-AFF1 infant ALL cells in BMSC co-cultures. This work establishes that EIF4E is broadly elevated across infant ALL and that clinically relevant ribavirin exposures have preclinical activity and effectively inhibit EIF4E in KMT2A-R cases, suggesting promise in EIF4E targeting using ribavirin as a means of treatment.


Subject(s)
Eukaryotic Initiation Factor-4E/genetics , Molecular Targeted Therapy/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Ribavirin/therapeutic use , Cell Line, Tumor , Child, Preschool , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Eukaryotic Initiation Factor-4E/physiology , Gene Expression Profiling , Gene Expression Regulation, Leukemic/drug effects , Humans , Indoles , Infant , Microarray Analysis , Multigene Family/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Biosynthesis/drug effects , Pyrroles/therapeutic use , Signal Transduction/drug effects
7.
Biochim Biophys Acta Mol Cell Res ; 1866(1): 83-89, 2019 01.
Article in English | MEDLINE | ID: mdl-30401537

ABSTRACT

Protein synthesis is one of the most complex and energy-consuming processes in eukaryotic cells and therefore is tightly regulated. One of the main mechanisms of translational control is post-translational modifications of the components of translational apparatus. Phosphorylation status of translation factors depends on the balanced action of kinases and phosphatases. While many kinase-dependent events are well defined, phosphatases that counteract phosphorylation are rarely determined. This mini-review focuses on the regulation of activity of translational initiation factors by serine/threonine phosphatases.


Subject(s)
Phosphoprotein Phosphatases/metabolism , Phosphoprotein Phosphatases/physiology , Protein Processing, Post-Translational/physiology , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-2/physiology , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4E/physiology , Eukaryotic Initiation Factors/metabolism , Eukaryotic Initiation Factors/physiology , Peptide Initiation Factors/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Biosynthesis/physiology , Protein Kinases , Protein Phosphatase 1/metabolism , Protein Phosphatase 1/physiology , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/physiology , Ribosomal Protein S6 Kinases/metabolism
8.
Chin J Integr Med ; 24(12): 920-924, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29752696

ABSTRACT

OBJECTIVE: To investigate the anti-leukemia effect of total saponins of Rubus parvifolius L. (TSRP) on K562 cell xenografts in nude mice and the mechanisms of action. METHODS: The K562 cell xenografts in nude mice were established, and then randomly divided into 5 groups, the control group, the cytosine arabinoside group(Ara-c) and 3 TSRP groups (20, 40 and 100 mg/kg). The tumor volume and mass of each group of nude mice were measured and the anti-tumor rates of TSRP were calculated subsequently. The apoptosis status of tumor cells was detected by hematoxylin-eosin (HE) and terminal dexynucleotidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL) staining analysis. Finally, the activities of apoptosis related signaling of signal transducer and activator of transcription 3 (STAT3), eukaryotic initiation factor 4E (eIF4E) and B-cell lymphoma-2 (bcl-2) were determined with immunohistochemistry tests. RESULTS: Subcutaneous injection of K562 cells induced tumor formation in nude mice, and the TSRP treated group showed a signifificant inhibitory effect on tumor formation. The nude mice treated with TSRP showed a signifificant decrease in tumor growth rate and tumor weight in comparison to the control group (all P<0.05). The HE staining and TUNEL assay showed that TSRP induced cell death by apoptosis. The immunohistochemical assay showed down-regulation of the bcl-2 gene in the TSRP treated cells. The phosphorylation levels of eIF4E and STAT3 were decreased obviously after the treatment of TSRP. CONCLUSION: TSRP had an excellent tumor-suppressing effect on K562 cells in the nude mice xenograft model, suggesting that TSPR can be developed as a promising anti-chronic myeloide leukemia drug.


Subject(s)
Eukaryotic Initiation Factor-4E/physiology , Leukemia/drug therapy , Rubus , STAT3 Transcription Factor/physiology , Saponins/pharmacology , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Humans , K562 Cells , Leukemia/pathology , Male , Mice , Rubus/chemistry , Xenograft Model Antitumor Assays
9.
Nat Struct Mol Biol ; 23(5): 387-94, 2016 05.
Article in English | MEDLINE | ID: mdl-27065194

ABSTRACT

The mRNAs stored in oocytes undergo general decay during the maternal-zygotic transition (MZT), and their stability is tightly interconnected with meiotic cell-cycle progression. However, the factors that trigger decay of maternal mRNA and couple this event to oocyte meiotic maturation remain elusive. Here, we identified B-cell translocation gene-4 (BTG4) as an MZT licensing factor in mice. BTG4 bridged CNOT7, a catalytic subunit of the CCR4-NOT deadenylase, to eIF4E, a key translation initiation factor, and facilitated decay of maternal mRNA. Btg4-null females produced morphologically normal oocytes but were infertile, owing to early developmental arrest. The intrinsic MAP kinase cascade in oocytes triggered translation of Btg4 mRNA stored in fully grown oocytes by targeting the 3' untranslated region, thereby coupling CCR4-NOT deadenylase-mediated decay of maternal mRNA with oocyte maturation and fertilization. This is a key step in oocyte cytoplasmic maturation that determines the developmental potential of mammalian embryos.


Subject(s)
Cell Cycle Proteins/physiology , Oocytes/physiology , 3' Untranslated Regions , Animals , Base Sequence , Cells, Cultured , Eukaryotic Initiation Factor-4E/physiology , Exoribonucleases , Female , Gene Expression , Gene Expression Regulation, Developmental , Gene Knockout Techniques , MAP Kinase Signaling System , Male , Meiosis , Mice, Inbred ICR , Mice, Knockout , Proteins/metabolism , RNA Stability , Repressor Proteins , Ribonucleases
10.
Dev Biol ; 410(1): 14-23, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26721418

ABSTRACT

Maternal expression of the translational regulator 4EHP (eIF4E-Homologous Protein) has an established role in generating protein gradients essential for specifying the Drosophila embryonic pattern. We generated a null mutation of 4EHP, which revealed for the first time that it is essential for viability and for completion of development. In fact, 4EHP null larvae, and larvae ubiquitously expressing RNAi targeting 4EHP, are developmentally delayed, fail to grow and eventually die. In addition, we found that expressing RNAi that targets 4EHP specifically in the prothoracic gland disrupted ecdysone biosynthesis, causing a block of the transition from the larval to pupal stages. This phenotype can be rescued by dietary administration of ecdysone. Consistent with this, 4EHP is highly expressed in the prothoracic gland and it is required for wild type expression levels of steroidogenic enzymes. Taken together, these results uncover a novel essential function for 4EHP in regulating ecdysone biosynthesis.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/embryology , Ecdysone/biosynthesis , Eukaryotic Initiation Factor-4E/physiology , Animals , Cell Size , Larva/growth & development , Pupa/growth & development , RNA Interference , Thorax/embryology , Thorax/metabolism
11.
Biochim Biophys Acta ; 1859(7): 896-905, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26708722

ABSTRACT

Nonsense-mediated mRNA decay (NMD) modulates the level of mRNA harboring a premature termination codon (PTC) in a translation-dependent manner. Inhibition of translation is known to impair NMD; however, few studies have investigated the correlation between enhanced translation and increased NMD. Here, we demonstrate that insulin signaling events increase translation, leading to an increase in NMD of eIF4E-bound transcripts. We provide evidence that (i) insulin-mediated enhancement of translation augments NMD and rapamycin abrogates this enhancement; (ii) an increase in AKT phosphorylation due to inhibition of PTEN facilitates NMD; (iii) insulin stimulation increases the binding of up-frameshift factor 1 (UPF1), most likely to eIF4E-bound PTC-containing transcripts; and (iv) insulin stimulation induces the colocalization of UPF1 and eIF4E in processing bodies. These results illustrate how extracellular signaling promotes the removal of eIF4E-bound NMD targets.


Subject(s)
Eukaryotic Initiation Factor-4E/physiology , Insulin/pharmacology , Nonsense Mediated mRNA Decay/drug effects , Animals , HeLa Cells , Humans , Protein Biosynthesis/drug effects , Protein Biosynthesis/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Sirolimus/pharmacology , Up-Regulation/drug effects , Up-Regulation/genetics
12.
Mol Med Rep ; 12(6): 7971-8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26498338

ABSTRACT

Eukaryotic translation initiation factor 4E (eIF4E) was shown to be upregulated in malignant human tumors. To assess the effect of downregulation of eIF4E on the proliferation and invasiveness of a human lung adenocarcinoma cell line, a short hairpin (sh)RNA targeting eIF4E was constructed and transfected into A549 human lung adenocarcinoma cells. The expression of eIF4E was determined by reverse transcription­quantitative polymerase chain reaction and western blotting. Cell viability was assessed using a Cell Counting kit­8, and apoptosis levels and cell cycle distribution were assessed by flow cytometry. Invasiveness was assessed using Transwell chambers. Transfection of the A549 cells with eIF4E targeting shRNA reduced the mRNA and protein expression levels of eIF4E by >70% 48 and 72 h following transfection, and eIF4E targeting shRNA­transfected cells were significantly less viable compared with the cells transfected with scrambled shRNA. The rate of apoptosis was also significantly increased, significantly more cells were in the G0/G1 phase and fewer were in the S phase, indicating cell cycle arrest. The fraction of transfected cells migrating across Transwell inserts were also reduced. In conclusion, inhibition of eIF4E suppressed cell growth and invasion, induced apoptosis and cell cycle arrest, suggesting that eIF4E may be a potential therapeutic target in lung adenocarcinoma.


Subject(s)
Adenocarcinoma/pathology , Eukaryotic Initiation Factor-4E/physiology , Lung Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma of Lung , Apoptosis/genetics , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Gene Knockdown Techniques , Humans , Lung Neoplasms/genetics , Neoplasm Invasiveness/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering , Transfection
13.
Neurobiol Dis ; 83: 67-74, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26306459

ABSTRACT

Autism spectrum disorder (ASD) is a group of heritable disorders with complex and unclear etiology. Classic ASD symptoms include social interaction and communication deficits as well as restricted, repetitive behaviors. In addition, ASD is often comorbid with intellectual disability. Fragile X syndrome (FXS) is the leading genetic cause of ASD, and is the most commonly inherited form of intellectual disability. Several mouse models of ASD and FXS exist, however the intellectual disability observed in ASD patients is not well modeled in mice. Using the Fmr1 knockout mouse and the eIF4E transgenic mouse, two previously characterized mouse models of fragile X syndrome and ASD, respectively, we generated the eIF4E/Fmr1 double mutant mouse. Our study shows that the eIF4E/Fmr1 double mutant mice display classic ASD behaviors, as well as cognitive dysfunction. Importantly, the learning impairments displayed by the double mutant mice spanned multiple cognitive tasks. Moreover, the eIF4E/Fmr1 double mutant mice display increased levels of basal protein synthesis. The results of our study suggest that the eIF4E/Fmr1 double mutant mouse may be a reliable model to study cognitive dysfunction in the context of ASD.


Subject(s)
Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/psychology , Cognition Disorders/genetics , Disease Models, Animal , Eukaryotic Initiation Factor-4E/physiology , Fragile X Mental Retardation Protein/physiology , Memory/physiology , Animals , Anxiety/genetics , Behavior, Animal/physiology , Conditioning, Classical/physiology , Eukaryotic Initiation Factor-4E/genetics , Fear/physiology , Fragile X Mental Retardation Protein/genetics , Hippocampus/metabolism , Interpersonal Relations , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutation
14.
Nat Neurosci ; 18(6): 855-62, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25915475

ABSTRACT

The circadian (∼24 h) clock is continuously entrained (reset) by ambient light so that endogenous rhythms are synchronized with daily changes in the environment. Light-induced gene expression is thought to be the molecular mechanism underlying clock entrainment. mRNA translation is a key step of gene expression, but the manner in which clock entrainment is controlled at the level of mRNA translation is not well understood. We found that a light- and circadian clock-regulated MAPK/MNK pathway led to phosphorylation of the cap-binding protein eIF4E in the mouse suprachiasmatic nucleus of the hypothalamus, the locus of the master circadian clock in mammals. Phosphorylation of eIF4E specifically promoted translation of Period 1 (Per1) and Period 2 (Per2) mRNAs and increased the abundance of basal and inducible PER proteins, which facilitated circadian clock resetting and precise timekeeping. Together, these results highlight a critical role for light-regulated translational control in the physiology of the circadian clock.


Subject(s)
Behavior, Animal/physiology , Circadian Rhythm/physiology , Eukaryotic Initiation Factor-4E/physiology , Animals , Behavior, Animal/radiation effects , Brain Chemistry/genetics , Brain Chemistry/physiology , Circadian Rhythm/radiation effects , Gene Expression Regulation/radiation effects , Light , MAP Kinase Signaling System/radiation effects , Mice , Mice, Inbred C57BL , Period Circadian Proteins/genetics , Period Circadian Proteins/physiology , Phosphorylation , Suprachiasmatic Nucleus/metabolism , Suprachiasmatic Nucleus/physiology
16.
J Invest Dermatol ; 135(5): 1368-1376, 2015 May.
Article in English | MEDLINE | ID: mdl-25615552

ABSTRACT

In eukaryotic cells, the rate-limiting component for cap-dependent mRNA translation is the translation initiation factor eIF4E. eIF4E is overexpressed in a variety of human malignancies, but whether it has a role in melanoma remains obscure. We hypothesized that eIF4E promotes melanoma cell proliferation and facilitates the development of acquired resistance to the BRAF inhibitor vemurafenib. We show that eIF4E is overexpressed in a panel of melanoma cell lines, compared with immortalized melanocytes. Knockdown of eIF4E significantly repressed the proliferation of a subset of melanoma cell lines. Moreover, in BRAF(V600E) melanoma cell lines, vemurafenib inhibits 4E-BP1 phosphorylation, thus promoting its binding to eIF4E. Cap-binding and polysome profiling analysis confirmed that vemurafenib stabilizes the eIF4E-4E-BP1 association and blocks mRNA translation, respectively. Conversely, in cells with acquired resistance to vemurafenib, there is an increased dependence on eIF4E for survival; 4E-BP1 is highly phosphorylated and thus eIF4E-4E-BP1 associations are impeded. Moreover, increasing eIF4E activity by silencing 4E-BP1/2 renders vemurafenib-responsive cells more resistant to BRAF inhibition. In conclusion, these data suggest that therapeutically targeting eIF4E may be a viable means of inhibiting melanoma cell proliferation and overcoming vemurafenib resistance.


Subject(s)
Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Eukaryotic Initiation Factor-4E/physiology , Indoles/pharmacology , Melanoma/pathology , Skin Neoplasms/pathology , Sulfonamides/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Eukaryotic Initiation Factor-4E/drug effects , Eukaryotic Initiation Factor-4E/genetics , Gene Knockdown Techniques , Gene Silencing , Humans , Phosphoproteins/metabolism , Phosphorylation , Proto-Oncogene Proteins B-raf , RNA, Small Interfering/pharmacology , Vemurafenib
17.
Oncogene ; 34(27): 3593-604, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25241901

ABSTRACT

High activation of the PI3K-AKT-mTOR pathway is characteristic for T-cell acute lymphoblastic leukemia (T-ALL). The activity of the master regulator of this pathway, PTEN, is often impaired in T-ALL. However, experimental evidence suggests that input from receptor tyrosine kinases (RTKs) is required for sustained mTOR activation, even in the absence of PTEN. We previously reported the expression of Neurotrophin receptor tyrosine kinases (TRKs) and their respective ligands in primary human leukemia samples. In the present study we aimed to dissect the downstream signaling cascades of TRK-induced T-ALL in a murine model and show that T-ALLs induced by deregulated receptor tyrosine kinase signaling acquire activating mutations in Notch1 and lose PTEN during clonal evolution. Some clones additionally lost one allele of the homeodomain transcription factor Cux1. All events independently led to a gradual hyperactivation of both mTORC1 and mTORC2 signaling. We dissected the role of the individual mTOR complexes by shRNA knockdown and found that the separate depletion of mTORC1 or mTORC2 reduced the growth of T-ALL blasts, but was not sufficient to induce apoptosis. In contrast, knockdown of the mTOR downstream effector eIF4E caused a striking cytotoxic effect, demonstrating a critical addiction to cap-dependent mRNA-translation. Although high mTORC2-AKT activation is commonly associated with drug-resistance, we demonstrate that T-ALL displaying a strong mTORC2-AKT activation were specifically susceptible to 4EGI-1, an inhibitor of the eIF4E-eIF4G interaction. To decipher the mechanism of 4EGI-1, we performed a genome-wide analysis of mRNAs that are translationally regulated by 4EGI-1 in T-ALL. 4EGI-1 effectively reduced the ribosomal occupancy of mRNAs that were strongly upregulated in T-ALL blasts compared with normal thymocytes including transcripts important for translation, mitochondria and cell cycle progression, such as cyclins and ribosomal proteins. These data suggest that disrupting the eIF4E-eIF4G interaction constitutes a promising therapy strategy in mTOR-deregulated T-cell leukemia.


Subject(s)
Eukaryotic Initiation Factor-4E/physiology , Leukemia, T-Cell/genetics , Leukemia, T-Cell/metabolism , Multiprotein Complexes/metabolism , Protein Biosynthesis , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Leukemic , Humans , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Biosynthesis/genetics , Signal Transduction , Tumor Cells, Cultured
18.
Cell Rep ; 9(5): 1742-1755, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25466251

ABSTRACT

Fragile X syndrome (FXS) is the leading genetic cause of autism. Mutations in Fmr1 (fragile X mental retardation 1 gene) engender exaggerated translation resulting in dendritic spine dysmorphogenesis, synaptic plasticity alterations, and behavioral deficits in mice, which are reminiscent of FXS phenotypes. Using postmortem brains from FXS patients and Fmr1 knockout mice (Fmr1(-/y)), we show that phosphorylation of the mRNA 5' cap binding protein, eukaryotic initiation factor 4E (eIF4E), is elevated concomitant with increased expression of matrix metalloproteinase 9 (MMP-9) protein. Genetic or pharmacological reduction of eIF4E phosphorylation rescued core behavioral deficits, synaptic plasticity alterations, and dendritic spine morphology defects via reducing exaggerated translation of Mmp9 mRNA in Fmr1(-/y) mice, whereas MMP-9 overexpression produced several FXS-like phenotypes. These results uncover a mechanism of regulation of synaptic function by translational control of Mmp-9 in FXS, which opens the possibility of new treatment avenues for the diverse neurological and psychiatric aspects of FXS.


Subject(s)
Benzofurans/pharmacology , Eukaryotic Initiation Factor-4E/physiology , Fragile X Syndrome/drug therapy , Matrix Metalloproteinase 9/genetics , Protein Biosynthesis/drug effects , Adenosine Triphosphatases/antagonists & inhibitors , Animals , Autistic Disorder/enzymology , Benzofurans/therapeutic use , Brain/enzymology , Cation Transport Proteins/antagonists & inhibitors , Cells, Cultured , Copper-Transporting ATPases , Dendritic Spines/pathology , Enzyme Induction/drug effects , Female , Fragile X Syndrome/enzymology , Fragile X Syndrome/genetics , Humans , Male , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Phosphorylation , Protein Processing, Post-Translational , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism
20.
J Neurosci ; 34(27): 9034-9, 2014 Jul 02.
Article in English | MEDLINE | ID: mdl-24990923

ABSTRACT

Memory retrieval, often termed reconsolidation, can render previously consolidated memories susceptible to manipulation that can lead to alterations in memory strength. Although it is known that reconsolidation requires mammalian target of rapamycin complex 1 (mTORC1)-dependent translation, the specific contributions of its downstream effectors in reconsolidation are unclear. Using auditory fear conditioning in mice, we investigated the role of eukaryotic translation initiation factor 4E (eIF4E)-eIF4G interactions and p70 S6 kinase polypeptide 1 (S6K1) in reconsolidation. We found that neither 4EGI-1 (2-[(4-(3,4-dichlorophenyl)-thiazol-2-ylhydrazono)-3-(2-nitrophenyl)]propionic acid), an inhibitor of eFI4E-eIF4G interactions, nor PF-4708671 [2-((4-(5-ethylpyrimidin-4-yl)piperazin-1-yl)methyl)-5-(trifluoromethyl)-1H-benzo[d]imidazole], an inhibitor of S6K1, alone blocked the reconsolidation of auditory fear memory. In contrast, using these drugs in concert to simultaneously block eIF4E-eIF4G interactions and S6K1 immediately after memory reactivation significantly attenuated fear memory reconsolidation. Moreover, the combination of 4EGI-1 and PF-4708671 further destabilized fear memory 10 d after memory reactivation, which was consistent with experiments using rapamycin, an mTORC1 inhibitor. Furthermore, inhibition of S6K1 immediately after retrieval resulted in memory destabilization 10 d after reactivation, whereas inhibition of eIF4E-eIF4G interactions did not. These results indicate that the reconsolidation of fear memory requires concomitant association of eIF4E to eIF4G as well as S6K1 activity and that the persistence of memory at longer intervals after memory reactivation also requires mTORC1-dependent processes that involve S6K1. These findings suggest a potential mechanism for how mTORC1-dependent translation is fine tuned to alter memory persistence.


Subject(s)
Avoidance Learning/physiology , Conditioning, Classical/physiology , Fear/physiology , Mental Recall/physiology , Multiprotein Complexes/physiology , TOR Serine-Threonine Kinases/physiology , Acoustic Stimulation , Animals , Avoidance Learning/drug effects , Conditioning, Classical/drug effects , Cues , Electroshock , Eukaryotic Initiation Factor-4E/antagonists & inhibitors , Eukaryotic Initiation Factor-4E/physiology , Eukaryotic Initiation Factor-4G/antagonists & inhibitors , Eukaryotic Initiation Factor-4G/physiology , Hydrazones , Imidazoles/pharmacology , Male , Mechanistic Target of Rapamycin Complex 1 , Memory, Long-Term/drug effects , Memory, Long-Term/physiology , Mental Recall/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitro Compounds/pharmacology , Piperazines/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/deficiency , Ribosomal Protein S6 Kinases, 90-kDa/physiology , Sirolimus/pharmacology , Thiazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL