Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 187
Filter
Add more filters

Publication year range
1.
Annu Rev Biochem ; 90: 321-348, 2021 06 20.
Article in English | MEDLINE | ID: mdl-33770447

ABSTRACT

Influenza virus RNA-dependent RNA polymerase (FluPol) transcribes the viral RNA genome in the infected cell nucleus. In the 1970s, researchers showed that viral transcription depends on host RNA polymerase II (RNAP II) activity and subsequently that FluPol snatches capped oligomers from nascent RNAP II transcripts to prime its own transcription. Exactly how this occurs remains elusive. Here, we review recent advances in the mechanistic understanding of FluPol transcription and early events in RNAP II transcription that are relevant to cap-snatching. We describe the known direct interactions between FluPol and the RNAP II C-terminal domain and summarize the transcription-related host factors that have been found to interact with FluPol. We also discuss open questions regarding how FluPol may be targeted to actively transcribing RNAP II and the exact context and timing of cap-snatching, which is presumed to occur after cap completion but before the cap is sequestered by the nuclear cap-binding complex.


Subject(s)
Host-Pathogen Interactions/physiology , Orthomyxoviridae/enzymology , RNA-Dependent RNA Polymerase/metabolism , Transcription, Genetic , Viral Proteins/metabolism , Humans , Orthomyxoviridae/pathogenicity , RNA Cap-Binding Proteins/genetics , RNA Cap-Binding Proteins/metabolism , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , RNA-Dependent RNA Polymerase/genetics , Viral Proteins/genetics
2.
Genes Dev ; 34(17-18): 1113-1127, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32873578

ABSTRACT

The largely nuclear cap-binding complex (CBC) binds to the 5' caps of RNA polymerase II (RNAPII)-synthesized transcripts and serves as a dynamic interaction platform for a myriad of RNA processing factors that regulate gene expression. While influence of the CBC can extend into the cytoplasm, here we review the roles of the CBC in the nucleus, with a focus on protein-coding genes. We discuss differences between CBC function in yeast and mammals, covering the steps of transcription initiation, release of RNAPII from pausing, transcription elongation, cotranscriptional pre-mRNA splicing, transcription termination, and consequences of spurious transcription. We describe parameters known to control the binding of generic or gene-specific cofactors that regulate CBC activities depending on the process(es) targeted, illustrating how the CBC is an ever-changing choreographer of gene expression.


Subject(s)
Gene Expression Regulation , RNA Cap-Binding Proteins/metabolism , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional/genetics , Animals , Cell Nucleus/metabolism , Humans , Open Reading Frames/genetics , Saccharomyces cerevisiae
3.
Nat Rev Mol Cell Biol ; 16(3): 190-202, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25650800

ABSTRACT

Transcription termination occurs when the polymerase is released after a transcription event, thus delimitating transcription units; however, the functional importance of termination extends beyond the mere definition of gene borders. By determining the cellular fate of the generated transcripts, transcription termination pathways shape the transcriptome. Recent reports have underscored the crucial role of these pathways in limiting the extent of pervasive transcription, which has attracted interest in post-initiation events in gene expression control. Transcription termination pathways involved in the production of non-coding RNAs - such as the Nrd1-Nab3-Sen1 (NNS) pathway in yeast and the cap-binding complex (CBC)-ARS2 pathway in humans - are key determinants of transcription quality control. Understanding the mechanisms leading to the timely and efficient dismantling of elongation complexes remains a major unmet challenge, but new insights into the molecular basis of termination at mRNA-coding and non-coding RNA gene targets have been gained in eukaryotes.


Subject(s)
Gene Expression Regulation , RNA Polymerase II/genetics , Transcription Termination, Genetic , Transcriptome , Animals , DNA Helicases/genetics , DNA Helicases/metabolism , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA Cap-Binding Proteins/genetics , RNA Cap-Binding Proteins/metabolism , RNA Helicases/genetics , RNA Helicases/metabolism , RNA Polymerase II/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction
4.
Mol Cell ; 73(2): 314-324.e4, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30527663

ABSTRACT

Macroautophagy/autophagy is a key catabolic recycling pathway that requires fine-tuned regulation to prevent pathologies and preserve homeostasis. Here, we report a new post-transcriptional pathway regulating autophagy involving the Pat1-Lsm (Lsm1 to Lsm7) mRNA-binding complex. Under nitrogen-starvation conditions, Pat1-Lsm binds a specific subset of autophagy-related (ATG) transcripts and prevents their 3' to 5' degradation by the exosome complex, leading to ATG mRNA stabilization and accumulation. This process is regulated through Pat1 dephosphorylation, is necessary for the efficient expression of specific Atg proteins, and is required for robust autophagy induction during nitrogen starvation. To the best of our knowledge, this work presents the first example of ATG transcript regulation via 3' binding factors and exosomal degradation.


Subject(s)
Autophagy-Related Proteins/metabolism , Autophagy , Nitrogen/deficiency , RNA Cap-Binding Proteins/metabolism , RNA Stability , RNA, Fungal/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , 3' Untranslated Regions , Autophagy-Related Proteins/genetics , Binding Sites , Gene Expression Regulation, Fungal , Humans , Jurkat Cells , Multiprotein Complexes , Phosphorylation , Protein Binding , RNA Cap-Binding Proteins/genetics , RNA, Fungal/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction
5.
FASEB J ; 38(1): e23367, 2024 01.
Article in English | MEDLINE | ID: mdl-38095329

ABSTRACT

Leishmania encode six paralogs of the cap-binding protein eIF4E and five eIF4G candidates, forming unique complexes. Two cap-binding proteins, LeishIF4E1 and LeishIF4E2, do not bind any identified LeishIF4Gs, thus their roles are intriguing. Here, we combine structural prediction, proteomic analysis, and interaction assays to shed light on LeishIF4E2 function. A nonconserved C-terminal extension was identified through structure prediction and sequence alignment. m7 GTP-binding assays involving both recombinant and transgenic LeishIF4E2 with and without the C-terminal extension revealed that this extension functions as a regulatory gate, modulating the cap-binding activity of LeishIF4E2. The interactomes of the two LeishIF4E2 versions were investigated, highlighting the role of the C-terminal extension in binding to SLBP2. SLBP2 is known to interact with a stem-loop structure in the 3' UTRs of histone mRNAs. Consistent with the predicted inhibitory effect of SLBP2 on histone expression in Xenopus laevis, a hemizygous deletion mutant of LeishIF4E2, exhibited an upregulation of several histones. We therefore propose that LeishIF4E2 is involved in histone expression, possibly through its interaction between SLBP2 and LeishIF4E2, thus affecting cell cycle progression. In addition, cell synchronization showed that LeishIF4E2 expression decreased during the S-phase, when histones are known to be synthesized. Previous studies in T. brucei also highlighted an association between TbEIF4E2 and SLBP2, and further reported on an interaction between TbIF4E2 and S-phase-abundant mRNAs. Our results show that overexpression of LeishIF4E2 correlates with upregulation of cell cycle and chromosome maintenance proteins. Along with its effect on histone expression, we propose that LeishIF4E2 is involved in cell cycle progression.


Subject(s)
Leishmania , RNA Cap-Binding Proteins/metabolism , Histones/metabolism , Proteomics , RNA, Messenger/metabolism , Cell Cycle , Protein Binding
6.
Genes Dev ; 31(11): 1147-1161, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28698298

ABSTRACT

The eIF4E homologous protein (4EHP) is thought to repress translation by competing with eIF4E for binding to the 5' cap structure of specific mRNAs to which it is recruited through interactions with various proteins, including the GRB10-interacting GYF (glycine-tyrosine-phenylalanine domain) proteins 1 and 2 (GIGYF1/2). Despite its similarity to eIF4E, 4EHP does not interact with eIF4G and therefore fails to initiate translation. In contrast to eIF4G, GIGYF1/2 bind selectively to 4EHP but not eIF4E. Here, we present crystal structures of the 4EHP-binding regions of GIGYF1 and GIGYF2 in complex with 4EHP, which reveal the molecular basis for the selectivity of the GIGYF1/2 proteins for 4EHP. Complementation assays in a GIGYF1/2-null cell line using structure-based mutants indicate that 4EHP requires interactions with GIGYF1/2 to down-regulate target mRNA expression. Our studies provide structural insights into the assembly of 4EHP-GIGYF1/2 repressor complexes and reveal that rather than merely facilitating 4EHP recruitment to transcripts, GIGYF1/2 proteins are required for repressive activity.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Gene Expression Regulation/genetics , RNA Cap-Binding Proteins/metabolism , RNA, Messenger/genetics , Carrier Proteins/genetics , Cell Line , Crystallization , Eukaryotic Initiation Factor-4E , HEK293 Cells , Humans , Models, Molecular , Mutation , Protein Binding/genetics , Protein Stability , Protein Structure, Quaternary , RNA Cap-Binding Proteins/chemistry
7.
Cell ; 138(2): 224-6, 2009 Jul 23.
Article in English | MEDLINE | ID: mdl-19632172

ABSTRACT

In this issue, Sabin et al. (2009) and Gruber et al. (2009) reveal the protein Ars2 as a versatile regulator of RNA silencing. They show that Ars2 stimulates microRNA processing, contributes to antiviral resistance in flies, and is important for cell proliferation in mammals.


Subject(s)
Nuclear Proteins/metabolism , RNA Cap-Binding Proteins/metabolism , Animals , Cell Survival , Humans , Nuclear Proteins/immunology , RNA Interference
8.
Pharmacol Res ; 198: 107008, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37995895

ABSTRACT

LSM1 is part of the cytoplasmic protein complex Lsm1-7-Pat1 and is likely involved in pre-mRNA degradation by aiding U4/U6 snRNP formation. More research is needed to uncover LSM1's potential in breast cancer (BRCA) clinical pathology, the tumor immune microenvironment, and precision oncology. We discovered LSM1 as a diagnostic marker for advanced BRCA with poor survival, using a multi-omics approach. We studied LSM1 expression across BRCA regions and its link to immune cells through various methods, including spatial transcriptomics and single-cell RNA-sequencing. We also examined how silencing LSM1 affects mitochondrial function and energy metabolism in the tumor environment. These findings were confirmed using 54 BRCA patient biopsies and tissue microarrays. Immunofluorescence and bioinformatics assessed LSM1's connection to clinicopathological features and prognosis. This study uncovers gene patterns linked to breast cancer, with LSM1 linked to macrophage energy processes. Silencing LSM1 in breast cancer cells disrupts mitochondria and energy metabolism. Spatial analysis aligns with previous results, showing LSM1's connection to macrophages. Biopsies confirm LSM1 elevation in advanced breast cancer with increased macrophage presence. To summarize, LSM1 changes may drive BRCA progression, making it a potential diagnostic and prognostic marker. It also influences energy metabolism and the tumor's immune environment during metastasis, showing promise for precision medicine and drug screening in BRCA.


Subject(s)
Breast Neoplasms , Saccharomyces cerevisiae Proteins , Humans , Female , RNA-Binding Proteins/genetics , RNA Cap-Binding Proteins/genetics , RNA Cap-Binding Proteins/metabolism , Saccharomyces cerevisiae/genetics , RNA, Messenger/metabolism , Breast Neoplasms/genetics , Tumor-Associated Macrophages/metabolism , Precision Medicine , Tumor Microenvironment , Proto-Oncogene Proteins/metabolism
9.
Genes Dev ; 28(4): 357-71, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24532714

ABSTRACT

The mammalian target of rapamycin (mTOR) promotes cell growth and proliferation by promoting mRNA translation and increasing the protein synthetic capacity of the cell. Although mTOR globally promotes translation by regulating the mRNA 5' cap-binding protein eIF4E (eukaryotic initiation factor 4E), it also preferentially regulates the translation of certain classes of mRNA via unclear mechanisms. To help fill this gap in knowledge, we performed a quantitative proteomic screen to identify proteins that associate with the mRNA 5' cap in an mTOR-dependent manner. Using this approach, we identified many potential regulatory factors, including the putative RNA-binding protein LARP1 (La-related protein 1). Our results indicate that LARP1 associates with actively translating ribosomes via PABP and that LARP1 stimulates the translation of mRNAs containing a 5' terminal oligopyrimidine (TOP) motif, encoding for components of the translational machinery. We found that LARP1 associates with the mTOR complex 1 (mTORC1) and is required for global protein synthesis as well as cell growth and proliferation. Together, these data reveal important molecular mechanisms involved in TOP mRNA translation and implicate LARP1 as an important regulator of cell growth and proliferation.


Subject(s)
Autoantigens/metabolism , Gene Expression Regulation , Proteomics , Pyrimidines/metabolism , RNA, Messenger/genetics , Ribonucleoproteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autoantigens/genetics , Cell Line , Cell Line, Tumor , Cells, Cultured , HEK293 Cells , Humans , Mice , RNA Cap-Binding Proteins/metabolism , Ribonucleoproteins/genetics , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , SS-B Antigen
10.
Genes Dev ; 28(8): 835-40, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24736843

ABSTRACT

40S ribosomes are loaded onto capped mRNAs via the multisubunit translation initiation factors eIF3 and eIF4F. While eIF4E is the eIF4F cap recognition component, the eIF4G subunit associates with 40S-bound eIF3. How this intricate process is coordinated remains poorly understood. Here, we identify an eIF3 subunit that regulates eIF4F modification and show that eIF3e is required for inducible eIF4E phosphorylation. Significantly, recruitment of the eIF4E kinase Mnk1 (MAPK signal-integrating kinase 1) to eIF4F depended on eIF3e, and eIF3e was sufficient to promote Mnk1-binding to eIF4G. This establishes a mechanism by which 40S ribosome loading imparts a phosphorylation mark on the cap-binding eIF4F complex that regulates selective mRNA translation and is synchronized by a specific eIF3 subunit.


Subject(s)
Eukaryotic Initiation Factor-3/metabolism , Protein Subunits/metabolism , RNA Cap-Binding Proteins/metabolism , Ribosome Subunits, Small, Eukaryotic/metabolism , Chromatography , Eukaryotic Initiation Factor-3/genetics , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Peptide Chain Initiation, Translational , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Transport , RNA Cap-Binding Proteins/chemistry , Signal Transduction
11.
J Biol Chem ; 295(51): 17781-17801, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33454014

ABSTRACT

Knockout mouse models have been extensively used to study the antiviral activity of IFIT (interferon-induced protein with tetratricopeptide repeats). Human IFIT1 binds to cap0 (m7GpppN) RNA, which lacks methylation on the first and second cap-proximal nucleotides (cap1, m7GpppNm, and cap2, m7GpppNmNm, respectively). These modifications are signatures of "self" in higher eukaryotes, whereas unmodified cap0-RNA is recognized as foreign and, therefore, potentially harmful to the host cell. IFIT1 inhibits translation at the initiation stage by competing with the cap-binding initiation factor complex, eIF4F, restricting infection by certain viruses that possess "nonself" cap0-mRNAs. However, in mice and other rodents, the IFIT1 orthologue has been lost, and the closely related Ifit1b has been duplicated twice, yielding three paralogues: Ifit1, Ifit1b, and Ifit1c. Although murine Ifit1 is similar to human IFIT1 in its cap0-RNA-binding selectivity, the roles of Ifit1b and Ifit1c are unknown. Here, we found that Ifit1b preferentially binds to cap1-RNA, whereas binding is much weaker to cap0- and cap2-RNA. In murine cells, we show that Ifit1b can modulate host translation and restrict WT mouse coronavirus infection. We found that Ifit1c acts as a stimulatory cofactor for both Ifit1 and Ifit1b, promoting their translation inhibition. In this way, Ifit1c acts in an analogous fashion to human IFIT3, which is a cofactor to human IFIT1. This work clarifies similarities and differences between the human and murine IFIT families to facilitate better design and interpretation of mouse models of human infection and sheds light on the evolutionary plasticity of the IFIT family.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Coronavirus/growth & development , Coronavirus/genetics , Protein Biosynthesis , RNA Cap-Binding Proteins/metabolism , RNA Caps/metabolism , RNA-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Coronavirus/metabolism , Disease Models, Animal , HEK293 Cells , Humans , Mice , Mice, Knockout , Models, Molecular , Mutation , Protein Binding , RAW 264.7 Cells , RNA-Binding Proteins/genetics
12.
PLoS Pathog ; 15(12): e1008155, 2019 12.
Article in English | MEDLINE | ID: mdl-31856218

ABSTRACT

Cellular response to environmental challenges requires immediate and precise regulation of transcriptional programs. During viral infections, this includes the expression of antiviral genes that are essential to combat the pathogen. Transcribed mRNAs are bound and escorted to the cytoplasm by the cap-binding complex (CBC). We recently identified a protein complex consisting of NCBP1 and NCBP3 that, under physiological conditions, has redundant function to the canonical CBC, consisting of NCBP1 and NCBP2. Here, we provide evidence that NCBP3 is essential to mount a precise and appropriate antiviral response. Ncbp3-deficient cells allow higher virus growth and elicit a reduced antiviral response, a defect happening on post-transcriptional level. Ncbp3-deficient mice suffered from severe lung pathology and increased morbidity after influenza A virus challenge. While NCBP3 appeared to be particularly important during viral infections, it may be more broadly involved to ensure proper protein expression.


Subject(s)
Orthomyxoviridae Infections/immunology , RNA Cap-Binding Proteins/immunology , RNA Cap-Binding Proteins/metabolism , Animals , Influenza A virus/immunology , Mice , Mice, Knockout , Orthomyxoviridae Infections/metabolism , Protein Biosynthesis/physiology
13.
PLoS Genet ; 14(7): e1007563, 2018 07.
Article in English | MEDLINE | ID: mdl-30059503

ABSTRACT

RNA-binding proteins (RBPs) establish the cellular fate of a transcript, but an understanding of these processes has been limited by a lack of identified specific interactions between RNA and protein molecules. Using MS2 RNA tagging, we have purified proteins associated with individual mRNA species induced by osmotic stress, STL1 and GPD1. We found members of the Lsm1-7/Pat1 RBP complex to preferentially bind these mRNAs, relative to the non-stress induced mRNAs, HYP2 and ASH1. To assess the functional importance, we mutated components of the Lsm1-7/Pat1 RBP complex and analyzed the impact on expression of osmostress gene products. We observed a defect in global translation inhibition under osmotic stress in pat1 and lsm1 mutants, which correlated with an abnormally high association of both non-stress and stress-induced mRNAs to translationally active polysomes. Additionally, for stress-induced proteins normally triggered only by moderate or high osmostress, in the mutants the protein levels rose high already at weak hyperosmosis. Analysis of ribosome passage on mRNAs through co-translational decay from the 5' end (5P-Seq) showed increased ribosome accumulation in lsm1 and pat1 mutants upstream of the start codon. This effect was particularly strong for mRNAs induced under osmostress. Thus, our results indicate that, in addition to its role in degradation, the Lsm1-7/Pat1 complex acts as a selective translational repressor, having stronger effect over the translation initiation of heavily expressed mRNAs. Binding of the Lsm1-7/Pat1p complex to osmostress-induced mRNAs mitigates their translation, suppressing it in conditions of weak or no stress, and avoiding a hyperresponse when triggered.


Subject(s)
Osmotic Pressure/physiology , RNA Cap-Binding Proteins/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/physiology , Glycerol-3-Phosphate Dehydrogenase (NAD+)/genetics , Glycerol-3-Phosphate Dehydrogenase (NAD+)/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Protein Binding/physiology , Protein Biosynthesis/physiology , RNA Cap-Binding Proteins/genetics , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics
14.
Int J Mol Sci ; 22(23)2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34884522

ABSTRACT

Leishmania parasites are digenetic protists that shuffle between sand fly vectors and mammalian hosts, transforming from flagellated extracellular promastigotes that reside within the intestinal tract of female sand flies to the obligatory intracellular and non-motile amastigotes within mammalian macrophages. Stage differentiation is regulated mainly by post-transcriptional mechanisms, including translation regulation. Leishmania parasites encode six different cap-binding proteins, LeishIF4E1-6, that show poor conservation with their counterparts from higher eukaryotes and among themselves. In view of the changing host milieu encountered throughout their life cycle, we propose that each LeishIF4E has a unique role, although these functions may be difficult to determine. Here we characterize LeishIF4E-6, a unique eIF4E ortholog that does not readily associate with m7GTP cap in either of the tested life forms of the parasite. We discuss the potential effect of substituting two essential tryptophan residues in the cap-binding pocket, expected to be involved in the cap-binding activity, as judged from structural studies in the mammalian eIF4E. LeishIF4E-6 binds to LeishIF4G-5, one of the five eIF4G candidates in Leishmania. However, despite this binding, LeishIF4E-6 does not appear to function as a translation factor. Its episomal overexpression causes a general reduction in the global activity of protein synthesis, which was not observed in the hemizygous deletion mutant generated by CRISPR-Cas9. This genetic profile suggests that LeishIF4E-6 has a repressive role. The interactome of LeishIF4E-6 highlights proteins involved in RNA metabolism such as the P-body marker DHH1, PUF1 and an mRNA-decapping enzyme that is homologous to the TbALPH1.


Subject(s)
Eukaryotic Initiation Factor-4F/metabolism , Leishmania/metabolism , Protozoan Proteins/metabolism , RNA Cap Analogs/genetics , RNA Cap-Binding Proteins/metabolism , Amino Acid Sequence , Eukaryotic Initiation Factor-4F/chemistry , Eukaryotic Initiation Factor-4F/genetics , Leishmania/genetics , Leishmania/growth & development , Protein Biosynthesis , Protein Conformation , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , RNA Cap Analogs/metabolism , RNA Cap-Binding Proteins/genetics , Sequence Homology
15.
Biochem Biophys Res Commun ; 526(4): 1143-1149, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32327257

ABSTRACT

In this study, we examined the impact of roscovitine, a cyclin-dependent kinase inhibitor (CDKI) that has entered phase I and II clinical trials, on influenza A viruses (IAVs) and its antiviral mechanism. The results illustrated that roscovitine inhibited multiple subtypes of influenza strains dose-dependently, including A/WSN/1933(H1N1), A/Aichi/2/68 (H3N2) and A/FM1/47 (H1N1) with IC50 value of 3.35 ± 0.39, 7.01 ± 1.84 and 5.99 ± 1.89 µM, respectively. Moreover, roscovitine suppressed the gene transcription and genome replication steps in the viral life cycle. Further mechanistic studies indicated that roscovitine reduced viral polymerase activity and bound specifically to the viral PB2cap protein by fluorescence polarization assay (FP) and surface plasmon resonance (SPR). Therefore, we believed roscovitine, as a PB2cap inhibitor, was a prospective antiviral agent to be developed as therapeutic treatment against influenza A virus infection.


Subject(s)
Antiviral Agents/pharmacology , Influenza A virus/physiology , Protein Kinase Inhibitors/pharmacology , RNA Cap-Binding Proteins/metabolism , RNA-Dependent RNA Polymerase/metabolism , Roscovitine/pharmacology , Viral Proteins/metabolism , Virus Replication/drug effects , Animals , DNA-Directed RNA Polymerases/metabolism , Dogs , Genome, Viral , Humans , Influenza A virus/drug effects , Influenza A virus/genetics , Madin Darby Canine Kidney Cells , Protein Kinase Inhibitors/chemistry , Roscovitine/chemistry , Transcription, Genetic/drug effects , Virus Internalization/drug effects , Virus Replication/genetics
16.
Biochem Biophys Res Commun ; 533(3): 391-396, 2020 12 10.
Article in English | MEDLINE | ID: mdl-32962861

ABSTRACT

The interferon-induced proteins with tetratricopeptide repeats (IFITs) are a family of RNA-binding proteins that are very highly expressed during antiviral response of immune system. IFIT proteins recognize and tightly bind foreign RNA particles. These are primarily viral RNAs ended with triphosphate at the 5' or lacking methylation of the first cap-proximal nucleotide but also in vitro transcribed RNA synthesized in the laboratory. Recognition of RNA by IFIT proteins leads to the formation of stable RNA/IFIT complexes and translational shut off of non-self transcripts. Here, we present a fluorescent-based assay to study the interaction between RNA molecules and IFIT family proteins. We have particularly focused on two representatives of this family: IFIT1 and IFIT5. We found a probe that competitively with RNA binds the positively charged tunnel in these IFIT proteins. The use of this probe for IFIT titration allowed us to evaluate the differences in binding affinities of mRNAs with different variants of 5' ends.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Anilino Naphthalenesulfonates/chemistry , Biological Assay , Fluorescent Dyes/chemistry , Neoplasm Proteins/chemistry , RNA Cap-Binding Proteins/chemistry , RNA Caps/chemistry , RNA-Binding Proteins/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Binding Sites , Binding, Competitive , Humans , Hydrogen Bonding , Kinetics , Molecular Docking Simulation , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Protein Binding , Protein Conformation , RNA Cap Analogs/chemistry , RNA Cap Analogs/metabolism , RNA Cap-Binding Proteins/genetics , RNA Cap-Binding Proteins/metabolism , RNA Caps/genetics , RNA Caps/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Spectrometry, Fluorescence , Static Electricity , Thermodynamics
17.
Mol Cell ; 46(6): 847-58, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22578813

ABSTRACT

Translational control of gene expression plays a key role in many biological processes. Consequently, the activity of the translation apparatus is under tight homeostatic control. eIF4E, the mRNA 5' cap-binding protein, facilitates cap-dependent translation and is a major target for translational control. eIF4E activity is controlled by a family of repressor proteins, termed 4E-binding proteins (4E-BPs). Here, we describe the surprising finding that despite the importance of eIF4E for translation, a drastic knockdown of eIF4E caused only minor reduction in translation. This conundrum can be explained by the finding that 4E-BP1 is degraded in eIF4E-knockdown cells. Hypophosphorylated 4E-BP1, which binds to eIF4E, is degraded, whereas hyperphosphorylated 4E-BP1 is refractory to degradation. We identified the KLHL25-CUL3 complex as the E3 ubiquitin ligase, which targets hypophosphorylated 4E-BP1. Thus, the activity of eIF4E is under homeostatic control via the regulation of the levels of its repressor protein 4E-BP1 through ubiquitination.


Subject(s)
Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , RNA Cap-Binding Proteins/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Cycle Proteins , HEK293 Cells , HeLa Cells , Homeostasis , Humans , Mice , Models, Biological , Molecular Sequence Data , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Biosynthesis , RNA Cap-Binding Proteins/metabolism , Transfection , Ubiquitin/metabolism
18.
Mol Cell ; 48(2): 207-18, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22959269

ABSTRACT

Shortening eukaryotic poly(A) tails represses mRNA translation and induces mRNA turnover. The major cytoplasmic deadenylase, the Ccr4-Not complex, is a conserved multisubunit assembly. Ccr4-Not is organized around Not1, a large scaffold protein that recruits two 3'-5' exoribonucleases, Caf1 and Ccr4. We report structural studies showing that the N-terminal arm of yeast Not1 has a HEAT-repeat structure with domains related to the MIF4G fold. A MIF4G domain positioned centrally within the Not1 protein recognizes Caf1, which in turn binds the LRR domain of Ccr4 and tethers the Ccr4 nuclease domain. The interactions that form the nuclease core of the Ccr4-Not complex are evolutionarily conserved. Their specific disruption affects cell growth and mRNA deadenylation and decay in vivo in yeast. Thus, the N-terminal arm of Not1 forms an extended platform reminiscent of scaffolding proteins like eIF4G and CBP80, and places the two nucleases in a pivotal position within the Ccr4-Not complex.


Subject(s)
Cell Cycle Proteins , Ribonucleases , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Transcription Factors , Binding Sites , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Crystallography, X-Ray , Eukaryotic Initiation Factor-4G/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Protein Binding , Protein Conformation , Protein Structure, Tertiary , RNA Cap-Binding Proteins/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonucleases/chemistry , Ribonucleases/metabolism , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism
19.
Nucleic Acids Res ; 46(11): 5792-5808, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29554310

ABSTRACT

Initially identified as a factor involved in tyrosine kinase receptor signaling, Grb10-interacting GYF protein 2 (GIGYF2) has later been shown to interact with the 5' cap-binding protein 4EHP as part of a translation repression complex, and to mediate post-transcriptional repression of tethered reporter mRNAs. A current model proposes that GIGYF2 is indirectly recruited to mRNAs by specific RNA-binding proteins (RBPs) leading to translation repression through its association with 4EHP. Accordingly, we recently observed that GIGYF2 also interacts with the miRNA-induced silencing complex and probably modulates its translation repression activity. Here we have further investigated how GIGYF2 represses mRNA function. In a tethering reporter assay, we identify three independent domains of GIGYF2 with repressive activity. In this assay, GIGYF2-mediated repression is independent of 4EHP but largely dependent on the CCR4/NOT complex that GIGYF2 recruits through multiple interfaces. Importantly, we show that GIGYF2 is an RBP and identify for the first time endogenous mRNA targets that recapitulate 4EHP-independent repression. Altogether, we propose that GIGYF2 has two distinct mechanisms of repression: one depends on 4EHP binding and mainly affects translation; the other is 4EHP-independent and involves the CCR4/NOT complex and its deadenylation activity.


Subject(s)
Carrier Proteins/metabolism , Protein Biosynthesis , RNA Stability , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Carrier Proteins/chemistry , Eukaryotic Initiation Factor-4E , HeLa Cells , Humans , Protein Domains , RNA Cap-Binding Proteins/metabolism , RNA-Binding Proteins/chemistry , Ribonucleases/metabolism
20.
Proc Natl Acad Sci U S A ; 114(21): 5425-5430, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28487484

ABSTRACT

MicroRNAs (miRNAs) play critical roles in a broad variety of biological processes by inhibiting translation initiation and by destabilizing target mRNAs. The CCR4-NOT complex effects miRNA-mediated silencing, at least in part through interactions with 4E-T (eIF4E transporter) protein, but the precise mechanism is unknown. Here we show that the cap-binding eIF4E-homologous protein 4EHP is an integral component of the miRNA-mediated silencing machinery. We demonstrate that the cap-binding activity of 4EHP contributes to the translational silencing by miRNAs through the CCR4-NOT complex. Our results show that 4EHP competes with eIF4E for binding to 4E-T, and this interaction increases the affinity of 4EHP for the cap. We propose a model wherein the 4E-T/4EHP interaction engenders a closed-loop mRNA conformation that blocks translational initiation of miRNA targets.


Subject(s)
MicroRNAs/metabolism , RNA Cap-Binding Proteins/metabolism , RNA Interference , RNA-Induced Silencing Complex/metabolism , Eukaryotic Initiation Factor-4E , HEK293 Cells , HeLa Cells , Humans , Nucleocytoplasmic Transport Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL