Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Genes (Basel) ; 14(11)2023 Nov 14.
Article in English | MEDLINE | ID: mdl-38003019

ABSTRACT

Translation initiation in eukaryotes is regulated at several steps, one of which involves the availability of the cap binding protein to participate in cap-dependent protein synthesis. Binding of eIF4E to translational repressors (eIF4E-binding proteins [4E-BPs]) suppresses translation and is used by cells to link extra- and intracellular cues to protein synthetic rates. The best studied of these interactions involves repression of translation by 4E-BP1 upon inhibition of the PI3K/mTOR signaling pathway. Herein, we characterize a novel 4E-BP, C8ORF88, whose expression is predominantly restricted to early spermatids. C8ORF88:eIF4E interaction is dependent on the canonical eIF4E binding motif (4E-BM) present in other 4E-BPs. Whereas 4E-BP1:eIF4E interaction is dependent on the phosphorylation of 4E-BP1, these sites are not conserved in C8ORF88 indicating a different mode of regulation.


Subject(s)
Carrier Proteins , Eukaryotic Initiation Factor-4E , Carrier Proteins/metabolism , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Binding , Phosphorylation
2.
Proc Natl Acad Sci U S A ; 120(25): e2300008120, 2023 06 20.
Article in English | MEDLINE | ID: mdl-37307456

ABSTRACT

mRNA translation initiation plays a critical role in learning and memory. The eIF4F complex, composed of the cap-binding protein eIF4E, ATP-dependent RNA helicase eIF4A, and scaffolding protein eIF4G, is a pivotal factor in the mRNA translation initiation process. eIF4G1, the major paralogue of the three eIF4G family members, is indispensable for development, but its function in learning and memory is unknown. To study the role of eIF4G1 in cognition, we used an eIF4G1 haploinsufficient (eIF4G1-1D) mouse model. The axonal arborization of eIF4G1-1D primary hippocampal neurons was significantly disrupted, and the mice displayed impairment in hippocampus-dependent learning and memory. Translatome analysis showed that the translation of mRNAs encoding proteins of the mitochondrial oxidative phosphorylation (OXPHOS) system was decreased in the eIF4G1-1D brain, and OXPHOS was decreased in eIF4G1-silenced cells. Thus, eIF4G1-mediated mRNA translation is crucial for optimal cognitive function, which is dependent on OXPHOS and neuronal morphogenesis.


Subject(s)
Eukaryotic Initiation Factor-4G , Oxidative Phosphorylation , Animals , Mice , RNA, Messenger , Peptide Chain Initiation, Translational , Morphogenesis , DNA Helicases
3.
J Mol Biol ; 434(9): 167523, 2022 05 15.
Article in English | MEDLINE | ID: mdl-35248544

ABSTRACT

The CCR4-NOT deadenylase complex is a major post-transcriptional regulator of eukaryotic gene expression. CNOT7 and CNOT8 are both vertebrate homologs of the yeast CCR4-NOT catalytic subunit Caf1. They are highly similar and are sometimes considered redundant, but Cnot7 and Cnot8 knockout mice exhibit different phenotypes, implying distinct physiological functions. In this study, we reveal a non-reciprocal effect of CNOT7 on CNOT8, in which CNOT8 protein is increased in the depletion of CNOT7 without corresponding changes in mRNA levels whereas CNOT7 is not affected by the loss of CNOT8. Cnot8 mRNA may be bound by the CCR4-NOT complex, suggesting that CCR4-NOT might directly regulate CNOT8 expression. Cnot8 mRNA is relatively unstable, but Cnot7 knockdown did not stabilize Cnot8 mRNA, nor did it increase translation. CNOT8 protein was also less stable than CNOT7. CNOT7 showed greater affinity than CNOT8 for the CCR4-NOT scaffold protein CNOT1 and was able to block CNOT8 from binding to CNOT1. Depletion of CNOT7 increased CNOT8 incorporation into the CCR4-NOT complex and stabilized CNOT8. These data suggest that CNOT7 is the dominant paralog in CCR4-NOT and that CNOT7 and CNOT8 protein stability is regulated in distinct ways.


Subject(s)
Exoribonucleases , Repressor Proteins , Transcription Factors , Animals , Exoribonucleases/genetics , Exoribonucleases/metabolism , Mice , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Ribonucleases/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
4.
iScience ; 24(10): 103151, 2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34646989

ABSTRACT

Control of mRNA stability and degradation is essential for appropriate gene expression, and its dysregulation causes various disorders, including cancer, neurodegenerative diseases, diabetes, and obesity. The 5'-3' exoribonuclease XRN1 executes the last step of RNA decay, but its physiological impact is not well understood. To address this, forebrain-specific Xrn1 conditional knockout mice (Xrn1-cKO) were generated, as Xrn1 null mice were embryonic lethal. Xrn1-cKO mice exhibited obesity with leptin resistance, hyperglycemia, hyperphagia, and decreased energy expenditure. Obesity resulted from dysregulated communication between the central nervous system and peripheral tissues. Moreover, expression of mRNAs encoding proteins that regulate appetite and energy expenditure was dysregulated in the hypothalamus of Xrn1-cKO mice. Therefore, we propose that XRN1 function in the hypothalamus is critical for maintenance of metabolic homeostasis.

5.
Cell Mol Life Sci ; 78(19-20): 6709-6719, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34559254

ABSTRACT

Eukaryotic initiation factor (eIF) 4F plays a central role in the ribosome recruitment phase of cap-dependent translation. This heterotrimeric complex consists of a cap binding subunit (eIF4E), a DEAD-box RNA helicase (eIF4A), and a large bridging protein (eIF4G). In mammalian cells, there are two genes encoding eIF4A (eIF4A1 and eIF4A2) and eIF4G (eIF4G1 and eIF4G3) paralogs that can assemble into eIF4F complexes. To query the essential nature of the eIF4F subunits in normal development, we used CRISPR/Cas9 to generate mouse strains with targeted ablation of each gene encoding the different eIF4F subunits. We find that Eif4e, Eif4g1, and Eif4a1 are essential for viability in the mouse, whereas Eif4g3 and Eif4a2 are not. However, Eif4g3 and Eif4a2 do play essential roles in spermatogenesis. Crossing of these strains to the lymphoma-prone Eµ-Myc mouse model revealed that heterozygosity at the Eif4e or Eif4a1 loci significantly delayed tumor onset. Lastly, tumors derived from Eif4e∆38 fs/+/Eµ-Myc or Eif4a1∆5 fs/+/Eµ-Myc mice show increased sensitivity to the chemotherapeutic agent doxorubicin, in vivo. Our study reveals that eIF4A2 and eIF4G3 play non-essential roles in gene expression regulation during embryogenesis; whereas reductions in eIF4E or eIF4A1 levels are protective against tumor development in a murine Myc-driven lymphoma setting.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Eukaryotic Initiation Factor-4F/genetics , Animals , Female , Gene Expression Regulation/genetics , Heterozygote , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Subunits/genetics , Spermatogenesis/genetics
6.
Commun Biol ; 3(1): 476, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32859966

ABSTRACT

Pancreatic ß-cells are responsible for production and secretion of insulin in response to increasing blood glucose levels. Defects in ß-cell function lead to hyperglycemia and diabetes mellitus. Here, we show that CNOT3, a CCR4-NOT deadenylase complex subunit, is dysregulated in islets in diabetic db/db mice, and that it is essential for murine ß cell maturation and identity. Mice with ß cell-specific Cnot3 deletion (Cnot3ßKO) exhibit impaired glucose tolerance, decreased ß cell mass, and they gradually develop diabetes. Cnot3ßKO islets display decreased expression of key regulators of ß cell maturation and function. Moreover, they show an increase of progenitor cell markers, ß cell-disallowed genes, and genes relevant to altered ß cell function. Cnot3ßKO islets exhibit altered deadenylation and increased mRNA stability, partly accounting for the increased expression of those genes. Together, these data reveal that CNOT3-mediated mRNA deadenylation and decay constitute previously unsuspected post-transcriptional mechanisms essential for ß cell identity.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Insulin-Secreting Cells/pathology , Transcription Factors/metabolism , Animals , Cell Count , Cell Differentiation/genetics , Diabetes Mellitus, Experimental/genetics , Disease Models, Animal , Glucose/toxicity , Glucose Tolerance Test , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Lipids/toxicity , Male , Mice, Knockout , Models, Biological , Obesity/pathology , Phenotype , Proteome/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptome/genetics
7.
Life Sci Alliance ; 3(5)2020 05.
Article in English | MEDLINE | ID: mdl-32238456

ABSTRACT

The biological significance of deadenylation in global gene expression is not fully understood. Here, we show that the CCR4-NOT deadenylase complex maintains expression of mRNAs, such as those encoding transcription factors, cell cycle regulators, DNA damage response-related proteins, and metabolic enzymes, at appropriate levels in the liver. Liver-specific disruption of Cnot1, encoding a scaffold subunit of the CCR4-NOT complex, leads to increased levels of mRNAs for transcription factors, cell cycle regulators, and DNA damage response-related proteins because of reduced deadenylation and stabilization of these mRNAs. CNOT1 suppression also results in an increase of immature, unspliced mRNAs (pre-mRNAs) for apoptosis-related and inflammation-related genes and promotes RNA polymerase II loading on their promoter regions. In contrast, mRNAs encoding metabolic enzymes become less abundant, concomitant with decreased levels of these pre-mRNAs. Lethal hepatitis develops concomitantly with abnormal mRNA expression. Mechanistically, the CCR4-NOT complex targets and destabilizes mRNAs mainly through its association with Argonaute 2 (AGO2) and butyrate response factor 1 (BRF1) in the liver. Therefore, the CCR4-NOT complex contributes to liver homeostasis by modulating the liver transcriptome through mRNA deadenylation.


Subject(s)
Homeodomain Proteins/metabolism , Liver/metabolism , Receptors, CCR4/metabolism , Animals , Cytoplasm/metabolism , Female , Homeodomain Proteins/genetics , Homeostasis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Poly A/genetics , RNA Stability , RNA, Messenger/genetics , Receptors, CCR4/genetics , Ribonucleases/genetics , TATA-Binding Protein Associated Factors/metabolism , Transcription Factors/genetics
8.
RNA Biol ; 17(3): 403-416, 2020 03.
Article in English | MEDLINE | ID: mdl-31924127

ABSTRACT

Shortening of mRNA poly(A) tails (deadenylation) to trigger their decay is mediated mainly by the CCR4-NOT deadenylase complex. While four catalytic subunits (CNOT6, 6L 7, and 8) have been identified in the mammalian CCR4-NOT complex, their individual biological roles are not fully understood. In this study, we addressed the contribution of CNOT7/8 to viability of primary mouse embryonic fibroblasts (MEFs). We found that MEFs lacking CNOT7/8 expression [Cnot7/8-double knockout (dKO) MEFs] undergo cell death, whereas MEFs lacking CNOT6/6L expression (Cnot6/6l-dKO MEFs) remain viable. Co-immunoprecipitation analyses showed that CNOT6/6L are also absent from the CCR4-NOT complex in Cnot7/8-dKO MEFs. In contrast, either CNOT7 or CNOT8 still interacts with other subunits in the CCR4-NOT complex in Cnot6/6l-dKO MEFs. Exogenous expression of a CNOT7 mutant lacking catalytic activity in Cnot7/8-dKO MEFs cannot recover cell viability, even though CNOT6/6L exists to some extent in the CCR4-NOT complex, confirming that CNOT7/8 is essential for viability. Bulk poly(A) tail analysis revealed that mRNAs with longer poly(A) tails are more numerous in Cnot7/8-dKO MEFs than in Cnot6/6l-dKO MEFs. Consistent with elongated poly(A) tails, more mRNAs are upregulated and stabilized in Cnot7/8-dKO MEFs than in Cnot6/6l-dKO MEFs. Importantly, Cnot6/6l-dKO mice are viable and grow normally to adulthood. Taken together, the CNOT7/8 catalytic subunits are essential for deadenylation, which is necessary to maintain cell viability, whereas CNOT6/6L are not.


Subject(s)
Exoribonucleases/metabolism , RNA, Messenger/metabolism , Receptors, CCR4/metabolism , Repressor Proteins/metabolism , Animals , Cell Survival/genetics , Exoribonucleases/genetics , Female , Fibroblasts/cytology , Fibroblasts/physiology , Male , Mice, Knockout , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Poly A/genetics , Poly A/metabolism , Protein Subunits , RNA Stability , RNA, Messenger/genetics , Receptors, CCR4/genetics , Repressor Proteins/genetics
9.
PLoS Pathog ; 14(8): e1007264, 2018 08.
Article in English | MEDLINE | ID: mdl-30138450

ABSTRACT

Herpes Simplex Virus 1 (HSV1) is amongst the most clinically advanced oncolytic virus platforms. However, efficient and sustained viral replication within tumours is limiting. Rapamycin can stimulate HSV1 replication in cancer cells, but active-site dual mTORC1 and mTORC2 (mammalian target of rapamycin complex 1 and 2) inhibitors (asTORi) were shown to suppress the virus in normal cells. Surprisingly, using the infected cell protein 0 (ICP0)-deleted HSV1 (HSV1-dICP0), we found that asTORi markedly augment infection in cancer cells and a mouse mammary cancer xenograft. Mechanistically, asTORi repressed mRNA translation in normal cells, resulting in defective antiviral response but also inhibition of HSV1-dICP0 replication. asTORi also reduced antiviral response in cancer cells, however in contrast to normal cells, transformed cells and cells transduced to elevate the expression of eukaryotic initiation factor 4E (eIF4E) or to silence the repressors eIF4E binding proteins (4E-BPs), selectively maintained HSV1-dICP0 protein synthesis during asTORi treatment, ultimately supporting increased viral replication. Our data show that altered eIF4E/4E-BPs expression can act to promote HSV1-dICP0 infection under prolonged mTOR inhibition. Thus, pharmacoviral combination of asTORi and HSV1 can target cancer cells displaying dysregulated eIF4E/4E-BPs axis.


Subject(s)
Herpes Simplex/pathology , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/genetics , Immediate-Early Proteins/genetics , Neoplasms/virology , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Catalytic Domain/drug effects , Cell Cycle Proteins , Cells, Cultured , Chlorocebus aethiops , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Herpes Simplex/complications , Herpes Simplex/genetics , Humans , Immediate-Early Proteins/deficiency , Mice , Neoplasms/complications , Neoplasms/genetics , Neoplasms/pathology , Organisms, Genetically Modified , Phosphoproteins/genetics , Phosphoproteins/metabolism , Signal Transduction/genetics , TOR Serine-Threonine Kinases/chemistry , Ubiquitin-Protein Ligases/deficiency , Vero Cells
10.
Oncotarget ; 8(31): 50542-50556, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28881582

ABSTRACT

Metformin inhibits the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway, which is frequently upregulated in hepatocellular carcinoma (HCC). Metformin has also been shown to induce apoptosis in this cancer. Here, we investigate whether metformin-induced apoptosis in HCC is mediated by the downstream mTORC1 effectors eukaryotic initiation factor 4E and (eIF4E)-binding proteins (4E-BPs). Further, we ask whether changes in 4E-BPs activity during metformin treatment negatively regulate translation of the anti-apoptotic myeloid cell leukemia 1 (Mcl-1) mRNA. A genetic HCC mouse model was employed to assess the ability of metformin to reduce tumor formation, induce apoptosis, and control 4E-BP1 activation and Mcl-1 protein expression. In parallel, the HCC cell line Huh7 was transduced with scrambled shRNA (control) or shRNAs targeting 4E-BP1 and 4E-BP2 (4E-BP knock-down (KD)) to measure differences in mRNA translation, apoptosis, and Mcl-1 protein expression after metformin treatment. In addition, immunohistochemical staining of eIF4E and 4E-BP1 protein levels was addressed in a HCC patient tissue microarray. We found that metformin decreased HCC tumor burden, and tumor tissues showed elevated apoptosis with reduced Mcl-1 and phosphorylated 4E-BP1 protein levels. In control but not 4E-BP KD Huh7 cells, metformin induced apoptosis and repressed Mcl-1 mRNA translation and protein levels. Immunostaining of HCC patient tumor tissues revealed a varying ratio of eIF4E/4E-BP1 expression. Our results propose that metformin induces apoptosis in mouse and cellular models of HCC through activation of 4E-BPs, thus tumors with elevated expression of 4E-BPs may display improved clinical chemopreventive benefit of metformin.

11.
Proc Natl Acad Sci U S A ; 113(44): 12360-12367, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27791185

ABSTRACT

Translational control of gene expression plays a key role during the early phases of embryonic development. Here we describe a transcriptional regulator of mouse embryonic stem cells (mESCs), Yin-yang 2 (YY2), that is controlled by the translation inhibitors, Eukaryotic initiation factor 4E-binding proteins (4E-BPs). YY2 plays a critical role in regulating mESC functions through control of key pluripotency factors, including Octamer-binding protein 4 (Oct4) and Estrogen-related receptor-ß (Esrrb). Importantly, overexpression of YY2 directs the differentiation of mESCs into cardiovascular lineages. We show that the splicing regulator Polypyrimidine tract-binding protein 1 (PTBP1) promotes the retention of an intron in the 5'-UTR of Yy2 mRNA that confers sensitivity to 4E-BP-mediated translational suppression. Thus, we conclude that YY2 is a major regulator of mESC self-renewal and lineage commitment and document a multilayer regulatory mechanism that controls its expression.


Subject(s)
Alternative Splicing/physiology , Cell Differentiation , Cell Self Renewal/physiology , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Transcription Factors/metabolism , Animals , Blastocyst/metabolism , Carrier Proteins/metabolism , Cell Lineage , Cell Self Renewal/genetics , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Introns , Mice , Mice, Knockout , Models, Biological , Octamer Transcription Factor-3/metabolism , Phosphoproteins , Polypyrimidine Tract-Binding Protein/genetics , Protein Biosynthesis/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Estrogen/metabolism , Transcription Factors/genetics , Transcription, Genetic/physiology , YY1 Transcription Factor/metabolism
12.
Nat Commun ; 7: 12188, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27432119

ABSTRACT

Parkinson's disease gene leucine-rich repeat kinase 2 (LRRK2) has been implicated in a number of processes including the regulation of mitochondrial function, autophagy and endocytic dynamics; nevertheless, we know little about its potential role in the regulation of synaptic plasticity. Here we demonstrate that postsynaptic knockdown of the fly homologue of LRRK2 thwarts retrograde, homeostatic synaptic compensation at the larval neuromuscular junction. Conversely, postsynaptic overexpression of either the fly or human LRRK2 transgene induces a retrograde enhancement of presynaptic neurotransmitter release by increasing the size of the release ready pool of vesicles. We show that LRRK2 promotes cap-dependent translation and identify Furin 1 as its translational target, which is required for the synaptic function of LRRK2. As the regulation of synaptic homeostasis plays a fundamental role in ensuring normal and stable synaptic function, our findings suggest that aberrant function of LRRK2 may lead to destabilization of neural circuits.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Neuromuscular Junction/metabolism , Synapses/metabolism , Animals , Furin/metabolism , HEK293 Cells , Homeostasis , Humans , Larva/metabolism , Mutant Proteins/metabolism , Mutation/genetics , Neurotransmitter Agents/metabolism , Protein Biosynthesis , RNA Caps/metabolism , Signal Transduction , Synaptic Transmission , Synaptic Vesicles/metabolism
13.
Diabetes ; 65(8): 2235-48, 2016 08.
Article in English | MEDLINE | ID: mdl-27217487

ABSTRACT

The mammalian target of rapamycin complex 1 (mTORC1) regulates several biological processes, although the key downstream mechanisms responsible for these effects are poorly defined. Using mice with deletion of eukaryotic translation initiation factor 4E-binding protein 2 (4E-BP2), we determine that this downstream target is a major regulator of glucose homeostasis and ß-cell mass, proliferation, and survival by increasing insulin receptor substrate 2 (IRS2) levels and identify a novel feedback mechanism by which mTORC1 signaling increases IRS2 levels. In this feedback loop, we show that 4E-BP2 deletion induces translation of the adaptor protein SH2B1 and promotes the formation of a complex with IRS2 and Janus kinase 2, preventing IRS2 ubiquitination. The changes in IRS2 levels result in increases in cell cycle progression, cell survival, and ß-cell mass by increasing Akt signaling and reducing p27 levels. Importantly, 4E-BP2 deletion confers resistance to cytokine treatment in vitro. Our data identify SH2B1 as a major regulator of IRS2 stability, demonstrate a novel feedback mechanism linking mTORC1 signaling with IRS2, and identify 4E-BP2 as a major regulator of proliferation and survival of ß-cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Eukaryotic Initiation Factors/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin-Secreting Cells/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle/genetics , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Cell Survival/genetics , Cell Survival/physiology , Eukaryotic Initiation Factors/genetics , Insulin Receptor Substrate Proteins/genetics , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Protein Stability , Signal Transduction/genetics , Signal Transduction/physiology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
14.
Mol Cell ; 61(5): 760-773, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26942679

ABSTRACT

MicroRNAs predominantly decrease gene expression; however, specific mRNAs are translationally upregulated in quiescent (G0) mammalian cells and immature Xenopus laevis oocytes by an FXR1a-associated microRNA-protein complex (microRNP) that lacks the microRNP repressor, GW182. Their mechanism in these conditions of decreased mTOR signaling, and therefore reduced canonical (cap-and-poly(A)-tail-mediated) translation, remains undiscovered. Our data reveal that mTOR inhibition in human THP1 cells enables microRNA-mediated activation. Activation requires shortened/no poly(A)-tail targets; polyadenylated mRNAs are partially activated upon PAIP2 overexpression, which interferes with poly(A)-bound PABP, precluding PABP-enhanced microRNA-mediated inhibition and canonical translation. Consistently, inhibition of PARN deadenylase prevents activation. P97/DAP5, a homolog of canonical translation factor, eIF4G, which lacks PABP- and cap binding complex-interacting domains, is required for activation, and thereby for the oocyte immature state. P97 interacts with 3' UTR-binding FXR1a-associated microRNPs and with PARN, which binds mRNA 5' caps, forming a specialized complex to translate recruited mRNAs in these altered canonical translation conditions.


Subject(s)
Cellular Senescence , MicroRNAs/metabolism , Oocytes/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Ribonucleoproteins/metabolism , 3' Untranslated Regions , Animals , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Binding Sites , Cell Line , Eukaryotic Initiation Factor-4G/genetics , Eukaryotic Initiation Factor-4G/metabolism , Exoribonucleases/genetics , Exoribonucleases/metabolism , Gene Expression Profiling/methods , Humans , MicroRNAs/genetics , Proteomics/methods , RNA Caps/genetics , RNA Caps/metabolism , RNA Interference , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Ribonucleoproteins/genetics , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Transfection , Xenopus laevis
15.
J Biol Chem ; 290(26): 15996-6020, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-25940091

ABSTRACT

The mammalian target of rapamycin complex 1 (mTORC1) is a critical regulator of protein synthesis. The best studied targets of mTORC1 in translation are the eukaryotic initiation factor-binding protein 1 (4E-BP1) and ribosomal protein S6 kinase 1 (S6K1). In this study, we identify the La-related protein 1 (LARP1) as a key novel target of mTORC1 with a fundamental role in terminal oligopyrimidine (TOP) mRNA translation. Recent genome-wide studies indicate that TOP and TOP-like mRNAs compose a large portion of the mTORC1 translatome, but the mechanism by which mTORC1 controls TOP mRNA translation is incompletely understood. Here, we report that LARP1 functions as a key repressor of TOP mRNA translation downstream of mTORC1. Our data show the following: (i) LARP1 associates with mTORC1 via RAPTOR; (ii) LARP1 interacts with TOP mRNAs in an mTORC1-dependent manner; (iii) LARP1 binds the 5'TOP motif to repress TOP mRNA translation; and (iv) LARP1 competes with the eukaryotic initiation factor (eIF) 4G for TOP mRNA binding. Importantly, from a drug resistance standpoint, our data also show that reducing LARP1 protein levels by RNA interference attenuates the inhibitory effect of rapamycin, Torin1, and amino acid deprivation on TOP mRNA translation. Collectively, our findings demonstrate that LARP1 functions as an important repressor of TOP mRNA translation downstream of mTORC1.


Subject(s)
Autoantigens/metabolism , Down-Regulation , Membrane Glycoproteins/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , Ribonucleoproteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Autoantigens/genetics , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1 , Membrane Glycoproteins/genetics , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Protein Binding , RNA, Long Noncoding , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Regulatory-Associated Protein of mTOR , Ribonucleoproteins/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , SS-B Antigen
16.
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
17.
Neuron ; 79(4): 712-24, 2013 Aug 21.
Article in English | MEDLINE | ID: mdl-23972597

ABSTRACT

Protein synthesis is critical for circadian clock function, but little is known of how translational regulation controls the master pacemaker in mammals, the suprachiasmatic nucleus (SCN). Here we demonstrate that the pivotal translational repressor, the eukaryotic translational initiation factor 4E binding protein 1 (4E-BP1), is rhythmically regulated via the mechanistic target of rapamycin (mTOR) signaling in the SCN and preferentially represses vasoactive intestinal peptide (Vip) mRNA translation. Knockout (KO) of Eif4ebp1 (gene encoding 4E-BP1) leads to upregulation of VIP and higher amplitude of molecular rhythms in the SCN. Consequently, the 4E-BP1 null mice exhibit accelerated re-entrainment to a shifted light/dark cycle and are more resistant to the rhythm-disruptive effects of constant light. Conversely, in Mtor(+/-) mice VIP expression is decreased and susceptibility to the effects of constant light is increased. These results reveal a key role for mTOR/4E-BP1-mediated translational control in regulating entrainment and synchrony of the master clock.


Subject(s)
Carrier Proteins/physiology , Circadian Rhythm/physiology , Gene Expression Regulation/physiology , Phosphoproteins/physiology , Signal Transduction/physiology , Suprachiasmatic Nucleus/metabolism , TOR Serine-Threonine Kinases/physiology , Adaptor Proteins, Signal Transducing , Animals , Anthraquinones/pharmacology , Butadienes/pharmacology , Cell Cycle Proteins , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Eukaryotic Initiation Factors , Gene Expression Regulation/drug effects , Indoles/pharmacology , Light , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitriles/pharmacology , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Phosphoproteins/deficiency , Phosphorylation/drug effects , Phosphorylation/genetics , Purines/pharmacology , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Vasoactive Intestinal Peptide/genetics , Vasoactive Intestinal Peptide/metabolism
18.
PLoS Biol ; 11(5): e1001564, 2013.
Article in English | MEDLINE | ID: mdl-23700384

ABSTRACT

Translation control often operates via remodeling of messenger ribonucleoprotein particles. The poly(A) binding protein (PABP) simultaneously interacts with the 3' poly(A) tail of the mRNA and the eukaryotic translation initiation factor 4G (eIF4G) to stimulate translation. PABP also promotes miRNA-dependent deadenylation and translational repression of target mRNAs. We demonstrate that isoform 2 of the mouse heterogeneous nuclear protein Q (hnRNP-Q2/SYNCRIP) binds poly(A) by default when PABP binding is inhibited. In addition, hnRNP-Q2 competes with PABP for binding to poly(A) in vitro. Depleting hnRNP-Q2 from translation extracts stimulates cap-dependent and IRES-mediated translation that is dependent on the PABP/poly(A) complex. Adding recombinant hnRNP-Q2 to the extracts inhibited translation in a poly(A) tail-dependent manner. The displacement of PABP from the poly(A) tail by hnRNP-Q2 impaired the association of eIF4E with the 5' m(7)G cap structure of mRNA, resulting in the inhibition of 48S and 80S ribosome initiation complex formation. In mouse fibroblasts, silencing of hnRNP-Q2 stimulated translation. In addition, hnRNP-Q2 impeded let-7a miRNA-mediated deadenylation and repression of target mRNAs, which require PABP. Thus, by competing with PABP, hnRNP-Q2 plays important roles in the regulation of global translation and miRNA-mediated repression of specific mRNAs.


Subject(s)
Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , MicroRNAs/metabolism , Animals , Binding Sites , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Fibroblasts/metabolism , HeLa Cells , Humans , Mice , MicroRNAs/genetics
19.
Neuron ; 78(2): 298-311, 2013 Apr 24.
Article in English | MEDLINE | ID: mdl-23622065

ABSTRACT

Control of protein synthesis is critical for synaptic plasticity and memory formation. However, the molecular mechanisms linking neuronal activity to activation of mRNA translation are not fully understood. Here, we report that the translational repressor poly(A)-binding protein (PABP)-interacting protein 2A (PAIP2A), an inhibitor of PABP, is rapidly proteolyzed by calpains in stimulated neurons and following training for contextual memory. Paip2a knockout mice exhibit a lowered threshold for the induction of sustained long-term potentiation and an enhancement of long-term memory after weak training. Translation of CaMKIIα mRNA is enhanced in Paip2a⁻/⁻ slices upon tetanic stimulation and in the hippocampus of Paip2a⁻/⁻ mice following contextual fear learning. We demonstrate that activity-dependent degradation of PAIP2A relieves translational inhibition of memory-related genes through PABP reactivation and conclude that PAIP2A is a pivotal translational regulator of synaptic plasticity and memory.


Subject(s)
Long-Term Potentiation/genetics , Memory/physiology , Neurons/physiology , Synapses/physiology , Tumor Suppressor Proteins/metabolism , Adenosine Triphosphatases/pharmacology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calpain/pharmacology , Cells, Cultured , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Dactinomycin/pharmacology , Enzyme Inhibitors/pharmacology , Fear/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hippocampus/cytology , Long-Term Potentiation/drug effects , Male , Memory/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , N-Methylaspartate/pharmacology , Neurons/drug effects , Oligodeoxyribonucleotides/pharmacology , Poly(A)-Binding Proteins , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/metabolism , RNA-Binding Proteins , Reaction Time/drug effects , Reaction Time/genetics , Repressor Proteins , Tumor Suppressor Proteins/genetics
20.
Cancer Res ; 72(24): 6468-76, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23100465

ABSTRACT

Active-site mTOR inhibitors (asTORi) hold great promise for targeting dysregulated mTOR signaling in cancer. Because of the multifaceted nature of mTORC1 signaling, identification of reliable biomarkers for the sensitivity of tumors to asTORi is imperative for their clinical implementation. Here, we show that cancer cells acquire resistance to asTORi by downregulating eukaryotic translation initiation factor (eIF4E)-binding proteins (4E-BPs-EIF4EBP1, EIF4EBP2). Loss of 4E-BPs or overexpression of eIF4E renders neoplastic growth and translation of tumor-promoting mRNAs refractory to mTOR inhibition. Conversely, moderate depletion of eIF4E augments the anti-neoplastic effects of asTORi. The anti-proliferative effect of asTORi in vitro and in vivo is therefore significantly influenced by perturbations in eIF4E/4E-BP stoichiometry, whereby an increase in the eIF4E/4E-BP ratio dramatically limits the sensitivity of cancer cells to asTORi. We propose that the eIF4E/4E-BP ratio, rather than their individual protein levels or solely their phosphorylation status, should be considered as a paramount predictive marker for forecasting the clinical therapeutic response to mTOR inhibitors.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Eukaryotic Initiation Factor-4E/physiology , Molecular Targeted Therapy , Neoplasms/diagnosis , Neoplasms/drug therapy , Phosphoproteins/physiology , Protein Kinase Inhibitors/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antineoplastic Agents/therapeutic use , Biomarkers, Pharmacological/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/physiology , Cell Cycle Proteins , Cells, Cultured , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Hep G2 Cells , Humans , Mice , Mice, Knockout , NIH 3T3 Cells , Neoplasms/genetics , Neoplasms/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Prognosis , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...