Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 573: 93-99, 2021 10 08.
Article in English | MEDLINE | ID: mdl-34403810

ABSTRACT

ATF4 is a crucial transcription factor in the integrated stress response, a major adaptive signaling pathway activated by tumor microenvironment and therapeutic stresses. BRAF inhibitors, such as vemurafenib, induce ATF4 in BRAF-mutated melanoma cells, but the mechanisms of ATF4 induction are not fully elucidated. Here, we show that ATF4 expression can be upregulated by eukaryotic initiation factor 4B (eIF4B) in BRAF-mutated A375 cells. Indeed, eIF4B knockout (KO) prevented ATF4 induction and activation of the uORF-mediated ATF4 translation mechanism during vemurafenib treatment, which were effectively recovered by the rescue of eIF4B. Transcriptome analysis revealed that eIF4B KO selectively influenced ATF4-target gene expression among the overall gene expression changed by vemurafenib. Interestingly, eIF4B supported cellular proliferation under asparagine-limited conditions, possibly through the eIF4B-ATF4 pathway. Our findings indicate that eIF4B can regulate ATF4 expression, thereby contributing to cellular stress adaptation, which could be targeted as a therapeutic approach against malignancies, including melanoma.


Subject(s)
Activating Transcription Factor 4/genetics , Asparagine/metabolism , Eukaryotic Initiation Factors/metabolism , Melanoma/genetics , Proto-Oncogene Proteins B-raf/genetics , Activating Transcription Factor 4/metabolism , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Eukaryotic Initiation Factors/deficiency , Humans , Melanoma/drug therapy , Melanoma/pathology , Proto-Oncogene Proteins B-raf/metabolism , Tumor Cells, Cultured , Vemurafenib/pharmacology
2.
RNA ; 27(9): 991-1003, 2021 09.
Article in English | MEDLINE | ID: mdl-34108230

ABSTRACT

Mammalian RNA interference (RNAi) is often linked to the regulation of gene expression in the cytoplasm. Synthetic RNAs, however, can also act through the RNAi pathway to regulate transcription and splicing. While nuclear regulation by synthetic RNAs can be robust, a critical unanswered question is whether endogenous functions for nuclear RNAi exist in mammalian cells. Using enhanced crosslinking immunoprecipitation (eCLIP) in combination with RNA sequencing (RNA-seq) and multiple AGO knockout cell lines, we mapped AGO2 protein binding sites within nuclear RNA. The strongest AGO2 binding sites were mapped to micro RNAs (miRNAs). The most abundant miRNAs were distributed similarly between the cytoplasm and nucleus, providing no evidence for mechanisms that facilitate localization of miRNAs in one compartment versus the other. Beyond miRNAs, most statistically significant AGO2 binding was within introns. Splicing changes were confirmed by RT-PCR and recapitulated by synthetic miRNA mimics complementary to the sites of AGO2 binding. These data support the hypothesis that miRNAs can control gene splicing. While nuclear RNAi proteins have the potential to be natural regulatory mechanisms, careful study will be necessary to identify critical RNA drivers of normal physiology and disease.


Subject(s)
Alternative Splicing , Argonaute Proteins/genetics , Eukaryotic Initiation Factors/genetics , MicroRNAs/genetics , RNA, Nuclear/genetics , Argonaute Proteins/deficiency , Base Pairing , Base Sequence , Binding Sites , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cytoplasm/genetics , Cytoplasm/metabolism , Eukaryotic Initiation Factors/deficiency , Exons , HCT116 Cells , Humans , Immunoprecipitation , Introns , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Oligoribonucleotides/genetics , Oligoribonucleotides/metabolism , Protein Binding , RNA, Nuclear/metabolism , Sequence Analysis, RNA
3.
Proc Natl Acad Sci U S A ; 116(36): 18060-18067, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31427534

ABSTRACT

Translational control plays a key role in regulation of neuronal activity and behavior. Deletion of the translational repressor 4E-BP2 in mice alters excitatory and inhibitory synaptic functions, engendering autistic-like behaviors. The contribution of 4E-BP2-dependent translational control in excitatory and inhibitory neurons and astrocytic cells to these behaviors remains unknown. To investigate this, we generated cell-type-specific conditional 4E-BP2 knockout mice and tested them for the salient features of autism, including repetitive stereotyped behaviors (self-grooming and marble burying), sociability (3-chamber social and direct social interaction tests), and communication (ultrasonic vocalizations in pups). We found that deletion of 4E-BP2 in GABAergic inhibitory neurons, defined by Gad2, resulted in impairments in social interaction and vocal communication. In contrast, deletion of 4E-BP2 in forebrain glutamatergic excitatory neurons, defined by Camk2a, or in astrocytes, defined by Gfap, failed to cause autistic-like behavioral abnormalities. Taken together, we provide evidence for an inhibitory-cell-specific role of 4E-BP2 in engendering autism-related behaviors.


Subject(s)
Autistic Disorder/metabolism , Behavior, Animal , Eukaryotic Initiation Factors/deficiency , GABAergic Neurons/metabolism , Interneurons/metabolism , Protein Biosynthesis , Animals , Astrocytes/metabolism , Astrocytes/pathology , Autistic Disorder/genetics , Autistic Disorder/pathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , GABAergic Neurons/pathology , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Interneurons/pathology , Mice , Mice, Knockout
4.
Cell Death Dis ; 10(2): 57, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30670698

ABSTRACT

Physiological stress conditions attenuate global mRNA translation via modifications of key eukaryotic initiation factors. However, non-canonical translation initiation mechanisms allow cap-independent translation of certain mRNAs. We have previously demonstrated that eIF5B promotes cap-independent translation of the mRNA encoding the antiapoptotic factor, XIAP, during cellular stress. Here, we show that depletion of eIF5B sensitizes glioblastoma multiforme cells to TRAIL-induced apoptosis by a pathway involving caspases-8, -9, and -7, with no significant effect on cell cycle progression. eIF5B promotes evasion of apoptosis by promoting the translation of several IRES-containing mRNAs, encoding the antiapoptotic proteins XIAP, Bcl-xL, cIAP1, and c-FLIPS. We also show that eIF5B promotes translation of nuclear factor erythroid 2-related factor 2 and suggest that reactive oxygen species contribute to increased apoptosis under conditions of eIF5B depletion. Finally, eIF5B depletion leads to decreased activation of the canonical NF-κB pathway. Taken together, our data suggest that eIF5B represents a regulatory node, allowing cancer cells to evade apoptosis by promoting the translation of pro-survival proteins from IRES-containing mRNAs.


Subject(s)
Eukaryotic Initiation Factors/genetics , Glioblastoma/genetics , Glioblastoma/pathology , Apoptosis/drug effects , Apoptosis/physiology , Cell Line, Tumor , Cell Survival/physiology , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Drug Resistance, Neoplasm , Eukaryotic Initiation Factors/deficiency , Eukaryotic Initiation Factors/metabolism , Glioblastoma/drug therapy , Glioblastoma/metabolism , HEK293 Cells , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Inhibitor of Apoptosis Proteins/genetics , NF-E2-Related Factor 2/biosynthesis , NF-E2-Related Factor 2/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Recombinant Proteins/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Transfection , Up-Regulation , X-Linked Inhibitor of Apoptosis Protein/biosynthesis , X-Linked Inhibitor of Apoptosis Protein/genetics , bcl-X Protein/biosynthesis , bcl-X Protein/genetics
5.
Biochem Biophys Res Commun ; 503(3): 2033-2039, 2018 09 10.
Article in English | MEDLINE | ID: mdl-30078681

ABSTRACT

4-Amino-2-Trifluoromethyl-Phenyl Retinate (ATPR), an all-trans retinoic acid (ATRA) derivative, possesses the ability to relief several carcinoma. Here, we explored the potential molecular mechanism of eukaryotic translation initiation factor 6 (eIF6) in ATPR-induced leukemia cell differentiation. Our research showed that ATPR could inhibit cell proliferation and promote cell differentiation in several leukemia cell lines. Besides, ATPR remarkably reduced the expression of eIF6 in vitro. Interestingly, the reduction of eIF6 contributed to restraining proliferation of K562 cells by inhibiting CyclinD1, C-myc and blocking cell cycle, as well as promoting differentiation of K562 cells by increasing the expression of C/EBPε, cell surface antigen CD11b and inducing renal-shrinkage of nuclear. Furthermore, the over-expression of eIF6 restrained the effects of ATPR on cell proliferation and maturation in K562 cells. In Addition, Notch1/CBF-1 signal activated by Chrysin could increase expression of eIF6 and restrain the differentiation in ATPR-induced K562 cells. Taken together, all above results indicated that ATPR induced differentiation of leukemia cells by decreasing eIF6 through Notch1/CBF-1 signal, which might exert an innovative treatment for leukemia.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Differentiation/drug effects , Eukaryotic Initiation Factors/deficiency , Leukemia/metabolism , Leukemia/pathology , Retinoids/pharmacology , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Humans , K562 Cells , Leukemia/genetics , Retinoids/chemistry , Structure-Activity Relationship , THP-1 Cells , Tumor Cells, Cultured
6.
Cell Rep ; 21(6): 1507-1520, 2017 Nov 07.
Article in English | MEDLINE | ID: mdl-29117557

ABSTRACT

Regular endurance training improves muscle oxidative capacity and reduces the risk of age-related disorders. Understanding the molecular networks underlying this phenomenon is crucial. Here, by exploiting the power of computational modeling, we show that endurance training induces profound changes in gene regulatory networks linking signaling and selective control of translation to energy metabolism and tissue remodeling. We discovered that knockdown of the mTOR-independent factor Eif6, which we predicted to be a key regulator of this process, affects mitochondrial respiration efficiency, ROS production, and exercise performance. Our work demonstrates the validity of a data-driven approach to understanding muscle homeostasis.


Subject(s)
Eukaryotic Initiation Factors/metabolism , Exercise , Muscle, Skeletal/metabolism , Acetylation , Animals , Calorimetry , Chromatography, High Pressure Liquid , Down-Regulation , Energy Metabolism/physiology , Eukaryotic Initiation Factors/deficiency , Eukaryotic Initiation Factors/genetics , Gene Regulatory Networks , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Oligonucleotide Array Sequence Analysis , Oxygen/metabolism , Physical Conditioning, Animal , Proteome/analysis , Reactive Oxygen Species/metabolism , Ribosomes/metabolism , Tandem Mass Spectrometry , Transcription, Genetic , Up-Regulation
7.
J Neurosci ; 36(11): 3170-83, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26985028

ABSTRACT

The removal of apoptotic cell corpses is important for maintaining homeostasis. Previously, defects in apoptotic cell clearance have been linked to neurodegeneration. However, the mechanisms underlying this are still poorly understood. In this study, we report that the absence of the phagocytic receptor Draper in glia leads to a pronounced accumulation of apoptotic neurons in the brain of Drosophila melanogaster. These dead cells persist in the brain throughout the lifespan of the organism and are associated with age-dependent neurodegeneration. Our data indicate that corpses persist because of defective phagosome maturation, rather than recognition defects. TORC1 activation, or inhibition of Atg1, in glia is sufficient to rescue corpse accumulation as well as neurodegeneration. These results suggest that phagocytosis of apoptotic neurons by glia during development is essential for brain homeostasis in adult flies. Furthermore, it suggests that TORC1 regulates Draper-mediated phagosome maturation. SIGNIFICANCE STATEMENT: Previously, defects in dead cell clearance were linked to neurodegeneration, but the exact mechanisms are not well understood. In this study, we report that the absence of an engulfment receptor leads to a pronounced accumulation of dead neurons in the brain of the fruit fly Drosophila melanogaster. These dead cells persist in the brain throughout the lifespan of the organism and are associated with age-dependent neurodegeneration. Our data indicate that corpses persist because of defective degradation of cells rather than recognition of dead cells.


Subject(s)
Apoptosis/physiology , Drosophila Proteins/metabolism , Nerve Degeneration/genetics , Neuroglia/pathology , Phagocytosis/physiology , Transcription Factors/metabolism , Age Factors , Animals , Animals, Genetically Modified , Brain/pathology , Caenorhabditis elegans Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Embryo, Nonmammalian , Eukaryotic Initiation Factors/deficiency , Eukaryotic Initiation Factors/genetics , Gene Expression Regulation/genetics , Larva , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Degeneration/pathology , Neuroglia/ultrastructure , Neurons/metabolism , Neurons/pathology , Neurons/ultrastructure , RNA Interference/physiology , Transcription Factors/genetics
8.
PLoS One ; 9(6): e99582, 2014.
Article in English | MEDLINE | ID: mdl-24945486

ABSTRACT

Sepsis decreases skeletal muscle protein synthesis in part by impairing mTOR activity and the subsequent phosphorylation of 4E-BP1 and S6K1 thereby controlling translation initiation; however, the relative importance of changes in these two downstream substrates is unknown. The role of 4E-BP1 (and -BP2) in regulating muscle protein synthesis was assessed in wild-type (WT) and 4E-BP1/BP2 double knockout (DKO) male mice under basal conditions and in response to sepsis. At 12 months of age, body weight, lean body mass and energy expenditure did not differ between WT and DKO mice. Moreover, in vivo rates of protein synthesis in gastrocnemius, heart and liver did not differ between DKO and WT mice. Sepsis decreased skeletal muscle protein synthesis and S6K1 phosphorylation in WT and DKO male mice to a similar extent. Sepsis only decreased 4E-BP1 phosphorylation in WT mice as no 4E-BP1/BP2 protein was detected in muscle from DKO mice. Sepsis decreased the binding of eIF4G to eIF4E in WT mice; however, eIF4E•eIF4G binding was not altered in DKO mice under either basal or septic conditions. A comparable sepsis-induced increase in eIF4B phosphorylation was seen in both WT and DKO mice. eEF2 phosphorylation was similarly increased in muscle from WT septic mice and both control and septic DKO mice, compared to WT control values. The sepsis-induced increase in muscle MuRF1 and atrogin-1 (markers of proteolysis) as well as TNFα and IL-6 (inflammatory cytokines) mRNA was greater in DKO than WT mice. The sepsis-induced decrease in myocardial and hepatic protein synthesis did not differ between WT and DKO mice. These data suggest overall basal protein balance and synthesis is maintained in muscle of mice lacking both 4E-BP1/BP2 and that sepsis-induced changes in mTOR signaling may be mediated by a down-stream mechanism independent of 4E-BP1 phosphorylation and eIF4E•eIF4G binding.


Subject(s)
Carrier Proteins/genetics , Eukaryotic Initiation Factors/genetics , Gene Expression Regulation, Neoplastic , Muscle, Skeletal/metabolism , Phosphoproteins/genetics , Sepsis/genetics , Adaptor Proteins, Signal Transducing , Animals , Cell Cycle Proteins , Elongation Factor 2 Kinase/genetics , Elongation Factor 2 Kinase/metabolism , Eukaryotic Initiation Factors/deficiency , Female , Gene Deletion , Interleukin-6/genetics , Interleukin-6/metabolism , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/pathology , Myocardium/metabolism , Myocardium/pathology , Phosphoproteins/deficiency , Phosphorylation , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Sepsis/metabolism , Sepsis/pathology , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tripartite Motif Proteins , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
9.
Reproduction ; 148(1): 87-98, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24700326

ABSTRACT

Oocyte quality is a well-established determinant of embryonic fate. However, the molecular participants and biological markers that affect and may predict adequate embryonic development are largely elusive. Our aim was to identify the components of the oocyte molecular machinery that part take in the production of a healthy embryo. For this purpose, we used an animal model, generated by us previously, the oocytes of which do not express Cx43 (Cx43(del/del)). In these mice, oogenesis appears normal, fertilisation does occur, early embryonic development is successful but implantation fails. We used magnetic resonance imaging analysis combined with histological examination to characterise the embryonic developmental incompetence. Reciprocal embryo transfer confirmed that the blastocyst evolved from the Cx43(del/del) oocyte is responsible for the implantation disorder. In order to unveil the genes, the impaired expression of which brings about the development of defective embryos, we carried out a genomic screening of both the oocytes and the resulting blastocysts. This microarray analysis revealed a low expression of Egr1, Rpl21 and Eif4a1 in Cx43(del/del) oocytes and downregulation of Rpl15 and Eif4g2 in the resulting blastocysts. We propose that global deficiencies in genes related to the expression of ribosomal proteins and translation initiation factors in apparently normal oocytes bring about accumulation of defects, which significantly compromise their developmental capacity. The blastocysts resulting from such oocytes, which grow within a confined space until implantation, may be unable to generate enough biological mass to allow their expansion. This information could be implicated to diagnosis and treatment of infertility, particularly to IVF.


Subject(s)
Blastocyst/metabolism , Embryo Implantation, Delayed/genetics , Gene Expression Regulation, Developmental , Oocytes/metabolism , Protein Biosynthesis/genetics , Animals , Connexin 43/deficiency , Connexin 43/genetics , Embryo Transfer , Eukaryotic Initiation Factors/deficiency , Eukaryotic Initiation Factors/genetics , Female , Genotype , Magnetic Resonance Imaging , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Pregnancy , Ribosomal Proteins/deficiency , Ribosomal Proteins/genetics
10.
Nature ; 493(7432): 371-7, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23172145

ABSTRACT

Hyperconnectivity of neuronal circuits due to increased synaptic protein synthesis is thought to cause autism spectrum disorders (ASDs). The mammalian target of rapamycin (mTOR) is strongly implicated in ASDs by means of upstream signalling; however, downstream regulatory mechanisms are ill-defined. Here we show that knockout of the eukaryotic translation initiation factor 4E-binding protein 2 (4E-BP2)-an eIF4E repressor downstream of mTOR-or eIF4E overexpression leads to increased translation of neuroligins, which are postsynaptic proteins that are causally linked to ASDs. Mice that have the gene encoding 4E-BP2 (Eif4ebp2) knocked out exhibit an increased ratio of excitatory to inhibitory synaptic inputs and autistic-like behaviours (that is, social interaction deficits, altered communication and repetitive/stereotyped behaviours). Pharmacological inhibition of eIF4E activity or normalization of neuroligin 1, but not neuroligin 2, protein levels restores the normal excitation/inhibition ratio and rectifies the social behaviour deficits. Thus, translational control by eIF4E regulates the synthesis of neuroligins, maintaining the excitation-to-inhibition balance, and its dysregulation engenders ASD-like phenotypes.


Subject(s)
Autistic Disorder/genetics , Autistic Disorder/physiopathology , Eukaryotic Initiation Factor-4E/metabolism , Protein Biosynthesis , Animals , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Eukaryotic Initiation Factor-4E/antagonists & inhibitors , Eukaryotic Initiation Factors/deficiency , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Male , Mice , Mice, Knockout , Phenotype , Synapses/metabolism
11.
Genes Dev ; 26(7): 693-704, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22474261

ABSTRACT

Argonaute proteins (Ago1-4) are essential components of the microRNA-induced silencing complex and play important roles in both microRNA biogenesis and function. Although Ago2 is the only one with the slicer activity, it is not clear whether the slicer activity is a universally critical determinant for Ago2's function in mammals. Furthermore, functional specificities associated with different Argonautes remain elusive. Here we report that microRNAs are randomly sorted to individual Argonautes in mammals, independent of the slicer activity. When both Ago1 and Ago2, but not either Ago1 or Ago2 alone, are ablated in the skin, the global expression of microRNAs is significantly compromised and it causes severe defects in skin morphogenesis. Surprisingly, Ago3 is able to load microRNAs efficiently in the absence of Ago1 and Ago2, despite a significant loss of global microRNA expression. Quantitative analyses reveal that Ago2 interacts with a majority of microRNAs (60%) in the skin, compared with Ago1 (30%) and Ago3 (<10%). This distribution is highly correlated with the abundance of each Argonaute, as quantified by shotgun proteomics. The quantitative correlation between Argonautes and their associated microRNAs is conserved in human cells. Finally, we measure the absolute expression of Argonaute proteins and determine that their copy number is ~1.4 × 10(5) to 1.7 × 10(5) molecules per cell. Together, our results reveal a quantitative picture for microRNA activity in mammals.


Subject(s)
Argonaute Proteins/metabolism , Eukaryotic Initiation Factors/metabolism , Gene Expression Regulation, Developmental , Animals , Argonaute Proteins/deficiency , Argonaute Proteins/genetics , Cell Proliferation , Eukaryotic Initiation Factors/deficiency , Eukaryotic Initiation Factors/genetics , Melanoma/genetics , Melanoma/metabolism , Mice , Mice, Knockout , MicroRNAs/metabolism , Skin/cytology , Skin/metabolism
12.
Am J Pathol ; 180(3): 1121-1135, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22234171

ABSTRACT

Protein synthesis is a tightly regulated, energy-consuming process. The control of mRNA translation into protein is fundamentally important for the fine-tuning of gene expression; additionally, precise translational control plays a critical role in many cellular processes, including development, cellular growth, proliferation, differentiation, synaptic plasticity, memory, and learning. Eukaryotic translation initiation factor 4h (Eif4h) encodes a protein involved in the process of protein synthesis, at the level of initiation phase. Its human homolog, WBSCR1, maps on 7q11.23, inside the 1.6 Mb region that is commonly deleted in patients affected by the Williams-Beuren syndrome, which is a complex neurodevelopmental disorder characterized by cardiovascular defects, cerebral dysplasias and a peculiar cognitive-behavioral profile. In this study, we generated knockout mice deficient in Eif4h. These mice displayed growth retardation with a significant reduction of body weight that began from the first week of postnatal development. Neuroanatomical profiling results generated by magnetic resonance imaging analysis revealed a smaller brain volume in null mice compared with controls as well as altered brain morphology, where anterior and posterior brain regions were differentially affected. The inactivation of Eif4h also led to a reduction in both the number and complexity of neurons. Behavioral studies revealed severe impairments of fear-related associative learning and memory formation. These alterations suggest that Eif4h might contribute to certain deficits associated with Williams-Beuren syndrome.


Subject(s)
Eukaryotic Initiation Factors/deficiency , Eukaryotic Initiation Factors/genetics , Growth Disorders/genetics , Learning Disabilities/genetics , Memory Disorders/genetics , Williams Syndrome/genetics , Animals , Brain/growth & development , Brain/metabolism , Conditioning, Psychological/physiology , Eukaryotic Initiation Factors/metabolism , Exploratory Behavior/physiology , Fear , Female , Gene Deletion , Heterozygote , Male , Mice , Mice, Knockout , Muscle Fatigue/genetics , Mutagenesis, Insertional , Organ Size , Psychomotor Performance/physiology , RNA, Messenger/metabolism , Williams Syndrome/psychology
13.
RNA ; 17(10): 1858-69, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21878547

ABSTRACT

Small RNAs loaded into Argonaute proteins direct silencing of complementary target mRNAs. It has been proposed that multiple, imperfectly complementary small interfering RNAs or microRNAs, when bound to the 3' untranslated region of a target mRNA, function cooperatively to silence target expression. We report that, in cultured human HeLa cells and mouse embryonic fibroblasts, Argonaute1 (Ago1), Ago3, and Ago4 act cooperatively to silence both perfectly and partially complementary target RNAs bearing multiple small RNA-binding sites. Our data suggest that for Ago1, Ago3, and Ago4, multiple, adjacent small RNA-binding sites facilitate cooperative interactions that stabilize Argonaute binding. In contrast, small RNAs bound to Ago2 and pairing perfectly to an mRNA target act independently to silence expression. Noncooperative silencing by Ago2 does not require the endoribonuclease activity of the protein: A mutant Ago2 that cannot cleave its mRNA target also silences noncooperatively. We propose that Ago2 binds its targets by a mechanism fundamentally distinct from that used by the three other mammalian Argonaute proteins.


Subject(s)
Eukaryotic Initiation Factors/metabolism , RNA Interference , RNA, Small Interfering/genetics , Animals , Base Sequence , Cells, Cultured , Eukaryotic Initiation Factors/deficiency , Humans , Mice , Protein Binding
14.
Methods Mol Biol ; 725: 295-313, 2011.
Article in English | MEDLINE | ID: mdl-21528461

ABSTRACT

Argonautes (Agos) are core effectors of RNA silencing. In several nonmammalian organisms, multiple Agos are known to exhibit specialized functions for distinct RNA silencing pathway. Mammals have four closely related Agos. To examine the functions of mammalian Agos in the microRNA silencing pathway, we generated mouse embryonic stem (ES) cells that are nullizygous for all Agos. This chapter describes a variety of techniques including BAC recombineering, gene targeting, and inducible Cre-loxP recombination, used to generate inducible Ago knock-out ES cells. The Ago-deficient ES cells provide an important tool for the study of mammalian RNA silencing.


Subject(s)
Embryonic Stem Cells/metabolism , Eukaryotic Initiation Factors/deficiency , Molecular Biology/methods , Promoter Regions, Genetic/drug effects , Animals , Cell Line , Chromosomes, Artificial, Bacterial/genetics , Chromosomes, Artificial, Bacterial/metabolism , Estrogen Antagonists/pharmacology , Eukaryotic Initiation Factors/genetics , Gene Order , Gene Targeting , Genetic Vectors/genetics , Genetic Vectors/metabolism , Integrases/metabolism , Mice , Promoter Regions, Genetic/genetics , Recombination, Genetic/genetics , Research Design , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology
15.
Mol Cell ; 37(6): 797-808, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20347422

ABSTRACT

The eIF4E-binding proteins (4E-BPs) repress translation initiation by preventing eIF4F complex formation. Of the three mammalian 4E-BPs, only 4E-BP2 is enriched in the mammalian brain and plays an important role in synaptic plasticity and learning and memory formation. Here we describe asparagine deamidation as a brain-specific posttranslational modification of 4E-BP2. Deamidation is the spontaneous conversion of asparagines to aspartates. Two deamidation sites were mapped to an asparagine-rich sequence unique to 4E-BP2. Deamidated 4E-BP2 exhibits increased binding to the mammalian target of rapamycin (mTOR)-binding protein raptor, which effects its reduced association with eIF4E. 4E-BP2 deamidation occurs during postnatal development, concomitant with the attenuation of the activity of the PI3K-Akt-mTOR signaling pathway. Expression of deamidated 4E-BP2 in 4E-BP2(-/-) neurons yielded mEPSCs exhibiting increased charge transfer with slower rise and decay kinetics relative to the wild-type form. 4E-BP2 deamidation may represent a compensatory mechanism for the developmental reduction of PI3K-Akt-mTOR signaling.


Subject(s)
Brain/metabolism , Eukaryotic Initiation Factors/metabolism , Protein Processing, Post-Translational , Synaptic Transmission , Amino Acid Sequence , Animals , Animals, Newborn , Cells, Cultured , Eukaryotic Initiation Factors/chemistry , Eukaryotic Initiation Factors/deficiency , Eukaryotic Initiation Factors/genetics , Humans , Kinetics , Mice , Mice, Knockout , Molecular Sequence Data , Organ Specificity , Phosphorylation , Protein Transport , Sequence Alignment , Sequence Homology, Amino Acid
16.
Chem Biol ; 16(12): 1240-9, 2009 Dec 24.
Article in English | MEDLINE | ID: mdl-20064434

ABSTRACT

The signal transduction pathway wherein mTOR regulates cellular growth and proliferation is an active target for drug discovery. The search for new mTOR inhibitors has recently yielded a handful of promising compounds that hold therapeutic potential. This search has been limited by the lack of a high-throughput assay to monitor the phosphorylation of a direct rapamycin-sensitive mTOR substrate in cells. Here we describe a novel cell-based chemical genetic screen useful for efficiently monitoring mTOR signaling to 4E-BPs in response to stimuli. The screen is based on the nuclear accumulation of eIF4E, which occurs in a 4E-BP-dependent manner specifically upon inhibition of mTOR signaling. Using this assay in a small-scale screen, we have identified several compounds not previously known to inhibit mTOR signaling, demonstrating that this method can be adapted to larger screens.


Subject(s)
Drug Discovery/methods , Eukaryotic Initiation Factor-4E/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins , Cell Line , Cell Nucleus/metabolism , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factors/deficiency , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Mice , Mice, Knockout , Morpholines/pharmacology , Phosphoproteins/deficiency , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
17.
Nature ; 452(7185): 323-8, 2008 Mar 20.
Article in English | MEDLINE | ID: mdl-18272964

ABSTRACT

Transcriptional activation of cytokines, such as type-I interferons (interferon (IFN)-alpha and IFN-beta), constitutes the first line of antiviral defence. Here we show that translational control is critical for induction of type-I IFN production. In mouse embryonic fibroblasts lacking the translational repressors 4E-BP1 and 4E-BP2, the threshold for eliciting type-I IFN production is lowered. Consequently, replication of encephalomyocarditis virus, vesicular stomatitis virus, influenza virus and Sindbis virus is markedly suppressed. Furthermore, mice with both 4E- and 4E-BP2 genes (also known as Eif4ebp1 and Eif4ebp2, respectively) knocked out are resistant to vesicular stomatitis virus infection, and this correlates with an enhanced type-I IFN production in plasmacytoid dendritic cells and the expression of IFN-regulated genes in the lungs. The enhanced type-I IFN response in 4E-BP1-/- 4E-BP2-/- double knockout mouse embryonic fibroblasts is caused by upregulation of interferon regulatory factor 7 (Irf7) messenger RNA translation. These findings highlight the role of 4E-BPs as negative regulators of type-I IFN production, via translational repression of Irf7 mRNA.


Subject(s)
Immunity, Innate/immunology , Interferon Regulatory Factor-7/biosynthesis , Protein Biosynthesis , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins , Cells, Cultured , Dendritic Cells/immunology , Embryo, Mammalian/cytology , Eukaryotic Initiation Factors/deficiency , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Fibroblasts/virology , Gene Deletion , Immunity, Innate/genetics , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , Interferon Type I/biosynthesis , Interferon Type I/immunology , Mice , Mice, Knockout , Phosphoproteins/deficiency , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vesicular stomatitis Indiana virus/physiology , Virus Physiological Phenomena , Virus Replication
18.
J Clin Invest ; 117(2): 387-96, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17273556

ABSTRACT

The most common pathology associated with obesity is insulin resistance, which results in the onset of type 2 diabetes mellitus. Several studies have implicated the mammalian target of rapamycin (mTOR) signaling pathway in obesity. Eukaryotic translation initiation factor 4E-binding (eIF4E-binding) proteins (4E-BPs), which repress translation by binding to eIF4E, are downstream effectors of mTOR. We report that the combined disruption of 4E-BP1 and 4E-BP2 in mice increased their sensitivity to diet-induced obesity. Increased adiposity was explained at least in part by accelerated adipogenesis driven by increased expression of CCAAT/enhancer-binding protein delta (C/EBPdelta), C/EBPalpha, and PPARgamma coupled with reduced energy expenditure, reduced lipolysis, and greater fatty acid reesterification in the adipose tissue of 4E-BP1 and 4E-BP2 double KO mice. Increased insulin resistance in 4E-BP1 and 4E-BP2 double KO mice was associated with increased ribosomal protein S6 kinase (S6K) activity and impairment of Akt signaling in muscle, liver, and adipose tissue. These data clearly demonstrate the role of 4E-BPs as a metabolic brake in the development of obesity and reinforce the idea that deregulated mTOR signaling is associated with the development of the metabolic syndrome.


Subject(s)
Eukaryotic Initiation Factors/deficiency , Insulin Resistance/physiology , Obesity/etiology , Phosphoproteins/deficiency , Adaptor Proteins, Signal Transducing , Adipogenesis , Animals , Base Sequence , Carrier Proteins/genetics , Cell Cycle Proteins , DNA Primers/genetics , Diet/adverse effects , Eukaryotic Initiation Factors/genetics , Humans , Insulin Resistance/genetics , Lipid Metabolism , Liver/metabolism , Liver/pathology , Metabolic Syndrome/etiology , Mice , Mice, Congenic , Mice, Inbred BALB C , Mice, Knockout , Mice, Mutant Strains , Obesity/genetics , Obesity/physiopathology , Phosphoproteins/genetics , Protein Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases
19.
Neurobiol Learn Mem ; 87(2): 248-56, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17029989

ABSTRACT

The requirement for de novo protein synthesis during multiple forms of learning, memory and behavior is well-established; however, we are only beginning to uncover the regulatory mechanisms that govern this process. In order to determine how translation initiation is regulated during neuroplasticity we engineered mutant C57Bl/6J mice that lack the translation repressor eukaryotic initiation factor 4E-binding protein 2 (4E-BP2) and have previously demonstrated that 4E-BP2 plays a critical role in hippocampus-dependent synaptic plasticity and memory. Herein, we examined the 4E-BP2 knockout mice in a battery of paradigms to address motor activity and motor skill learning, anxiety and social dominance behaviors, working memory and conditioned taste aversion. We found that the 4E-BP2 knockout mice demonstrated altered activity in the rotating rod test, light/dark exploration test, spontaneous alternation T-maze and conditioned taste aversion test. The information gained from these studies builds a solid foundation for future studies on the specific role of 4E-BP2 in various types of behavior, and for a broader, more detailed examination of the mechanisms of translational control in the brain.


Subject(s)
Behavior, Animal/physiology , Eukaryotic Initiation Factors/deficiency , Animals , Avoidance Learning/physiology , Exploratory Behavior/physiology , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Psychomotor Performance/physiology , Reaction Time/genetics , Rotarod Performance Test/methods , Taste , Time Factors
20.
J Neurosci ; 25(42): 9581-90, 2005 Oct 19.
Article in English | MEDLINE | ID: mdl-16237163

ABSTRACT

Long-lasting synaptic plasticity and memory requires mRNA translation, yet little is known as to how this process is regulated. To explore the role that the translation repressor 4E-BP2 plays in hippocampal long-term potentiation (LTP) and learning and memory, we examined 4E-BP2 knock-out mice. Interestingly, genetic elimination of 4E-BP2 converted early-phase LTP to late-phase LTP (L-LTP) in the Schaffer collateral pathway, likely as a result of increased eIF4F complex formation and translation initiation. A critical limit for activity-induced translation was revealed in the 4E-BP2 knock-out mice because L-LTP elicited by traditional stimulation paradigms was obstructed. Moreover, the 4E-BP2 knock-out mice also exhibited impaired spatial learning and memory and conditioned fear-associative memory deficits. These results suggest a crucial role for proper regulation of the eIF4F complex by 4E-BP2 during LTP and learning and memory in the mouse hippocampus.


Subject(s)
Eukaryotic Initiation Factor-4F/physiology , Eukaryotic Initiation Factors/physiology , Hippocampus/physiology , Memory/physiology , Neuronal Plasticity/physiology , Protein Biosynthesis/physiology , Repressor Proteins/physiology , Animals , Eukaryotic Initiation Factor-4F/deficiency , Eukaryotic Initiation Factors/deficiency , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Repressor Proteins/genetics , Synapses/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...