Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Biochemistry (Mosc) ; 88(11): 1786-1799, 2023 Nov.
Article in English | MEDLINE | ID: mdl-38105199

ABSTRACT

In response to stress stimuli, eukaryotic cells typically suppress protein synthesis. This leads to the release of mRNAs from polysomes, their condensation with RNA-binding proteins, and the formation of non-membrane-bound cytoplasmic compartments called stress granules (SGs). SGs contain 40S but generally lack 60S ribosomal subunits. It is known that cycloheximide, emetine, and anisomycin, the ribosome inhibitors that block the progression of 80S ribosomes along mRNA and stabilize polysomes, prevent SG assembly. Conversely, puromycin, which induces premature termination, releases mRNA from polysomes and stimulates the formation of SGs. The same effect is caused by some translation initiation inhibitors, which lead to polysome disassembly and the accumulation of mRNAs in the form of stalled 48S preinitiation complexes. Based on these and other data, it is believed that the trigger for SG formation is the presence of mRNA with extended ribosome-free segments, which tend to form condensates in the cell. In this study, we evaluated the ability of various small-molecule translation inhibitors to block or stimulate the assembly of SGs under conditions of severe oxidative stress induced by sodium arsenite. Contrary to expectations, we found that ribosome-targeting elongation inhibitors of a specific type, which arrest solitary 80S ribosomes at the beginning of the mRNA coding regions but do not interfere with all subsequent ribosomes in completing translation and leaving the transcripts (such as harringtonine, lactimidomycin, or T-2 toxin), completely prevent the formation of arsenite-induced SGs. These observations suggest that the presence of even a single 80S ribosome on mRNA is sufficient to prevent its recruitment into SGs, and the presence of extended ribosome-free regions of mRNA is not sufficient for SG formation. We propose that mRNA entry into SGs may be mediated by specific contacts between RNA-binding proteins and those regions on 40S subunits that remain inaccessible when ribosomes are associated.


Subject(s)
Protein Biosynthesis , Stress Granules , RNA, Messenger/metabolism , Cytoplasmic Granules , Ribosomes/metabolism , Protein Synthesis Inhibitors/pharmacology , RNA-Binding Proteins/metabolism
2.
Int J Mol Sci ; 24(24)2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38138978

ABSTRACT

Upstream open reading frames (uORFs) are a frequent feature of eukaryotic mRNAs. Upstream ORFs govern main ORF translation in a variety of ways, but, in a nutshell, they either filter out scanning ribosomes or allow downstream translation initiation via leaky scanning or reinitiation. Previous reports concurred that eIF4G2, a long-known but insufficiently studied eIF4G1 homologue, can rescue the downstream translation, but disagreed on whether it is leaky scanning or reinitiation that eIF4G2 promotes. Here, we investigated a unique human mRNA that encodes two highly conserved proteins (POLGARF with unknown function and POLG, the catalytic subunit of the mitochondrial DNA polymerase) in overlapping reading frames downstream of a regulatory uORF. We show that the uORF renders the translation of both POLGARF and POLG mRNAs reliant on eIF4G2. Mechanistically, eIF4G2 enhances both leaky scanning and reinitiation, and it appears that ribosomes can acquire eIF4G2 during the early steps of reinitiation. This emphasizes the role of eIF4G2 as a multifunctional scanning guardian that replaces eIF4G1 to facilitate ribosome movement but not ribosome attachment to an mRNA.


Subject(s)
Peptide Chain Initiation, Translational , Ribosomes , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , 5' Untranslated Regions , Ribosomes/metabolism , Reading Frames , Open Reading Frames , Protein Biosynthesis , DNA Polymerase gamma/genetics , DNA Polymerase gamma/metabolism
3.
RNA ; 29(3): 282-299, 2023 03.
Article in English | MEDLINE | ID: mdl-36517212

ABSTRACT

The eukaryotic initiation factor 4G2 (eIF4G2, DAP5, Nat1, p97) was discovered in 1997. Over the past two decades, dozens of papers have presented contradictory data on eIF4G2 function. Since its identification, eIF4G2 has been assumed to participate in noncanonical translation initiation mechanisms, but recent results indicate that it can be involved in scanning as well. In particular, eIF4G2 provides leaky scanning through some upstream open reading frames (uORFs), which are typical for long 5' UTRs of mRNAs from higher eukaryotes. It is likely the protein can also help the ribosome overcome other impediments during scanning of the 5' UTRs of animal mRNAs. This may explain the need for eIF4G2 in higher eukaryotes, as many mRNAs that encode regulatory proteins have rather long and highly structured 5' UTRs. Additionally, they often bind to various proteins, which also hamper the movement of scanning ribosomes. This review discusses the suggested mechanisms of eIF4G2 action, denotes obscure or inconsistent results, and proposes ways to uncover other fundamental mechanisms in which this important protein factor may be involved in higher eukaryotes.


Subject(s)
Eukaryotic Initiation Factor-4G , Peptide Chain Initiation, Translational , Protein Biosynthesis , Animals , 5' Untranslated Regions/genetics , Eukaryota/genetics , Eukaryotic Initiation Factor-4G/genetics , Eukaryotic Initiation Factor-4G/metabolism , Proteins/metabolism , Ribosomes/genetics , Ribosomes/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
4.
Nucleic Acids Res ; 50(2): 1111-1127, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35018467

ABSTRACT

eIF4G2 (DAP5 or Nat1) is a homologue of the canonical translation initiation factor eIF4G1 in higher eukaryotes but its function remains poorly understood. Unlike eIF4G1, eIF4G2 does not interact with the cap-binding protein eIF4E and is believed to drive translation under stress when eIF4E activity is impaired. Here, we show that eIF4G2 operates under normal conditions as well and promotes scanning downstream of the eIF4G1-mediated 40S recruitment and cap-proximal scanning. Specifically, eIF4G2 facilitates leaky scanning for a subset of mRNAs. Apparently, eIF4G2 replaces eIF4G1 during scanning of 5' UTR and the necessity for eIF4G2 only arises when eIF4G1 dissociates from the scanning complex. In particular, this event can occur when the leaky scanning complexes interfere with initiating or elongating 80S ribosomes within a translated uORF. This mechanism is therefore crucial for higher eukaryotes which are known to have long 5' UTRs with highly frequent uORFs. We suggest that uORFs are not the only obstacle on the way of scanning complexes towards the main start codon, because certain eIF4G2 mRNA targets lack uORF(s). Thus, higher eukaryotes possess two distinct scanning complexes: the principal one that binds mRNA and initiates scanning, and the accessory one that rescues scanning when the former fails.


Subject(s)
Eukaryotic Initiation Factor-4G/metabolism , RNA, Messenger/metabolism , Ribosomes/metabolism , Humans , Open Reading Frames , Protein Biosynthesis
5.
Biochemistry (Mosc) ; 86(9): 1060-1094, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34565312

ABSTRACT

Viruses exploit the translation machinery of an infected cell to synthesize their proteins. Therefore, viral mRNAs have to compete for ribosomes and translation factors with cellular mRNAs. To succeed, eukaryotic viruses adopt multiple strategies. One is to circumvent the need for m7G-cap through alternative instruments for ribosome recruitment. These include internal ribosome entry sites (IRESs), which make translation independent of the free 5' end, or cap-independent translational enhancers (CITEs), which promote initiation at the uncapped 5' end, even if located in 3' untranslated regions (3' UTRs). Even if a virus uses the canonical cap-dependent ribosome recruitment, it can still perturb conventional ribosomal scanning and start codon selection. The pressure for genome compression often gives rise to internal and overlapping open reading frames. Their translation is initiated through specific mechanisms, such as leaky scanning, 43S sliding, shunting, or coupled termination-reinitiation. Deviations from the canonical initiation reduce the dependence of viral mRNAs on translation initiation factors, thereby providing resistance to antiviral mechanisms and cellular stress responses. Moreover, viruses can gain advantage in a competition for the translational machinery by inactivating individual translational factors and/or replacing them with viral counterparts. Certain viruses even create specialized intracellular "translation factories", which spatially isolate the sites of their protein synthesis from cellular antiviral systems, and increase availability of translational components. However, these virus-specific mechanisms may become the Achilles' heel of a viral life cycle. Thus, better understanding of the unconventional mechanisms of viral mRNA translation initiation provides valuable insight for developing new approaches to antiviral therapy.


Subject(s)
Eukaryotic Cells/virology , Peptide Chain Initiation, Translational/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Animals , Eukaryotic Cells/physiology , Humans , Internal Ribosome Entry Sites/physiology , RNA, Circular/genetics , Viral Proteins/physiology
6.
Int J Mol Sci ; 21(5)2020 Feb 29.
Article in English | MEDLINE | ID: mdl-32121426

ABSTRACT

The closed-loop model of eukaryotic translation states that mRNA is circularized by a chain of the cap-eIF4E-eIF4G-poly(A)-binding protein (PABP)-poly(A) interactions that brings 5' and 3' ends together. This circularization is thought to promote the engagement of terminating ribosomes to a new round of translation at the same mRNA molecule, thus enhancing protein synthesis. Despite the general acceptance and the elegance of the hypothesis, it has never been proved experimentally. Using continuous in situ monitoring of luciferase synthesis in a mammalian in vitro system, we show here that the rate of translation initiation at capped and polyadenylated reporter mRNAs increases after the time required for the first ribosomes to complete mRNA translation. Such acceleration strictly requires the presence of a poly(A)-tail and is abrogated by the addition of poly(A) RNA fragments or m7GpppG cap analog to the translation reaction. The optimal functional interaction of mRNA termini requires 5' untranslated region (UTR) and 3' UTR of moderate lengths and provides stronger acceleration, thus a longer poly(A)-tail. Besides, we revealed that the inhibitory effect of the dominant negative R362Q mutant of initiation factor eIF4A diminishes in the course of translation reaction, suggesting a relaxed requirement for ATP. Taken together, our results imply that, upon the functional looping of an mRNA, the recycled ribosomes can be recruited to the start codon of the same mRNA molecule in an eIF4A-independent fashion. This non-canonical closed-loop assisted reinitiation (CLAR) mode provides efficient translation of the functionally circularized mRNAs.


Subject(s)
Peptide Chain Initiation, Translational/genetics , Poly A/genetics , Protein Biosynthesis/genetics , RNA, Messenger/chemistry , 3' Untranslated Regions/genetics , Animals , Cell-Free System , Cyclization , Eukaryotic Initiation Factor-4E/chemistry , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4G/chemistry , Eukaryotic Initiation Factor-4G/genetics , Mice , Poly A/chemistry , RNA Caps/chemistry , RNA Caps/genetics
7.
RNA ; 25(7): 757-767, 2019 07.
Article in English | MEDLINE | ID: mdl-31010886

ABSTRACT

Poly(rC)-binding protein 2 (PCBP2, hnRNP E2) is one of the most abundant RNA-binding proteins in mammalian cells. In humans, it exists in seven isoforms, which are assumed to play similar roles in cells. The protein is shown to bind 3'-untranslated regions (3'-UTRs) of many mRNAs and regulate their translation and/or stability, but nothing is known about the functional consequences of PCBP2 binding to 5'-UTRs. Here we show that the PCBP2 isoform f interacts with the 5'-UTRs of mRNAs encoding eIF4G2 (a translation initiation factor with a yet unknown mechanism of action, also known as DAP5) and Cyclin I, and inhibits their translation in vitro and in cultured cells, while the PCBP2 isoform e only affects Cyclin I translation. Furthermore, eIF4G2 participates in a cap-dependent translation of the PCBP2 mRNA. Thus, PCBP2 and eIF4G2 seem to regulate one another's expression via a novel type of feedback loop formed by the translation initiation factor and the RNA-binding protein.


Subject(s)
5' Untranslated Regions/genetics , Eukaryotic Initiation Factor-4G/genetics , Protein Biosynthesis , RNA, Messenger/genetics , RNA-Binding Proteins/metabolism , Cells, Cultured , Eukaryotic Initiation Factor-4G/metabolism , Humans , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics
8.
Data Brief ; 23: 103701, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30815525

ABSTRACT

TMA20 (MCT-1), TMA22 (DENR) and TMA64 (eIF2D) are eukaryotic translation factors involved in ribosome recycling and re-initiation. They operate with P-site bound tRNA in post-termination or (re-)initiation translation complexes, thus participating in the removal of 40S ribosomal subunit from mRNA stop codons after termination and controlling translation re-initiation on mRNAs with upstream open reading frames (uORFs), as well as de novo initiation on some specific mRNAs. Here we report ribosomal profiling data of S.cerevisiae strains with individual deletions of TMA20, TMA64 or both TMA20 and TMA64 genes. We provide RNA-Seq and Ribo-Seq data from yeast strains grown in the rich YPD or minimal SD medium. We illustrate our data by plotting differential distribution of ribosomal-bound mRNA fragments throughout uORFs in 5'-untranslated region (5' UTR) of GCN4 mRNA and on mRNA transcripts encoded in MAT locus in the mutant and wild-type strains, thus providing a basis for investigation of the role of these factors in the stress response, mating and sporulation. We also document a shift of transcription start site of the APC4 gene which occurs when the neighboring TMA64 gene is replaced by the standard G418-resistance cassette used for the creation of the Yeast Deletion Library. This shift results in dramatic deregulation of the APC4 gene expression, as revealed by our Ribo-Seq data, which can be probably used to explain strong genetic interactions of TMA64 with genes involved in the cell cycle and mitotic checkpoints. Raw RNA-Seq and Ribo-Seq data as well as all gene counts are available in NCBI Gene Expression Omnibus (GEO) repository under GEO accession GSE122039 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE122039).

9.
Biochimie ; 157: 92-101, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30419262

ABSTRACT

Short upstream open reading frames (uORFs) are the most prevalent cis-acting regulatory elements in the mammalian transcriptome which can orchestrate mRNA translation. Apart from being "passive roadblocks" that decrease expression of the main coding regions, particular uORFs can serve as specific sensors for changing conditions, thus regulating translation in response to cell stress. Here we report a novel uORF-based regulatory mechanism that is employed under conditions of hyperosmotic stress by at least two human mRNAs, coding for translation reinitiation/recycling factor eIF2D and E3 ubiquitin ligase MDM2. This novel mode of translational control selectively downregulates their expression and requires as few as one uORF. Using a set of reporter mRNAs and fleeting mRNA transfection (FLERT) technique, we provide evidence that the phenomenon does not rely on delayed reinitiation, altered AUG recognition, ribosome stalling, mRNA destabilization or other known mechanisms. Instead, it is based on events taking place at uORF stop codon or immediately downstream. Functional aspects and implications of the novel regulatory mechanism to cell physiology are discussed.


Subject(s)
Codon, Initiator/metabolism , Eukaryotic Initiation Factor-2/biosynthesis , Open Reading Frames , Osmotic Pressure , Protein Biosynthesis , Proto-Oncogene Proteins c-mdm2/biosynthesis , Codon, Initiator/genetics , Eukaryotic Initiation Factor-2/genetics , HEK293 Cells , Humans , Proto-Oncogene Proteins c-mdm2/genetics , RNA Stability
10.
Trends Biochem Sci ; 43(11): 882-895, 2018 11.
Article in English | MEDLINE | ID: mdl-29789219

ABSTRACT

Eukaryotic translation initiation relies on the m7G cap present at the 5' end of all mRNAs. Some viral mRNAs employ alternative mechanisms of initiation based on internal ribosome entry. The 'IRES ideology' was adopted by researchers to explain the differential translation of cellular mRNAs when the cap recognition is suppressed. However, some cellular IRESs have already been challenged and others are awaiting their validation. As an alternative cap-independent mechanism, we propose adopting the concept of cap-independent translation enhancers (CITEs) for mammalian mRNAs. Unlike IRESs, CITEs can be located both within 5' and 3' UTRs and bind mRNA-recruiting translational components. The respective 5' UTRs are then inspected by the scanning machinery essentially in the same way as under cap-dependent translation.


Subject(s)
5' Untranslated Regions , Peptide Chain Initiation, Translational , RNA Caps/metabolism , RNA, Messenger/metabolism , Ribosomes/metabolism , Animals , Humans , RNA Caps/genetics , RNA, Messenger/genetics
11.
Gene ; 651: 174-182, 2018 Apr 20.
Article in English | MEDLINE | ID: mdl-29414693

ABSTRACT

Eukaryotic cells evolved highly complex and accurate protein synthesis machinery that is finely tuned by various signaling pathways. Dysregulation of translation is a hallmark of many diseases, including cancer, and thus pharmacological approaches to modulate translation become very promising. While there has been much progress in our understanding of mammalian mRNA-specific translation control, surprisingly, relatively little is known about whether and how the protein components of the translation machinery shape translation of their own mRNAs. Here we analyze mammalian mRNAs encoding components of the translation initiation machinery for potential regulatory features such as 5'TOP motifs, TISU motifs, poor start codon nucleotide context and upstream open reading frames.


Subject(s)
Eukaryotic Initiation Factors/genetics , Gene Expression Regulation , RNA, Messenger/metabolism , 5' Untranslated Regions , Animals , Humans , Mammals , Protein Biosynthesis , RNA 5' Terminal Oligopyrimidine Sequence
12.
Cell Mol Life Sci ; 74(8): 1431-1455, 2017 04.
Article in English | MEDLINE | ID: mdl-27853833

ABSTRACT

The idea of internal initiation is frequently exploited to explain the peculiar translation properties or unusual features of some eukaryotic mRNAs. In this review, we summarize the methods and arguments most commonly used to address cases of translation governed by internal ribosome entry sites (IRESs). Frequent mistakes are revealed. We explain why "cap-independent" does not readily mean "IRES-dependent" and why the presence of a long and highly structured 5' untranslated region (5'UTR) or translation under stress conditions cannot be regarded as an argument for appealing to internal initiation. We carefully describe the known pitfalls and limitations of the bicistronic assay and artefacts of some commercially available in vitro translation systems. We explain why plasmid DNA transfection should not be used in IRES studies and which control experiments are unavoidable if someone decides to use it anyway. Finally, we propose a workflow for the validation of IRES activity, including fast and simple experiments based on a single genetic construct with a sequence of interest.


Subject(s)
Internal Ribosome Entry Sites , 5' Untranslated Regions , Animals , Eukaryotic Initiation Factors/metabolism , Humans , Peptide Chain Initiation, Translational , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomes/genetics , Ribosomes/metabolism , Transfection
13.
Sci Rep ; 6: 37905, 2016 11 28.
Article in English | MEDLINE | ID: mdl-27892500

ABSTRACT

mRNAs lacking 5' untranslated regions (leaderless mRNAs) are molecular relics of an ancient translation initiation pathway. Nevertheless, they still represent a significant portion of transcriptome in some taxons, including a number of eukaryotic species. In bacteria and archaea, the leaderless mRNAs can bind non-dissociated 70 S ribosomes and initiate translation without protein initiation factors involved. Here we use the Fleeting mRNA Transfection technique (FLERT) to show that translation of a leaderless reporter mRNA is resistant to conditions when eIF2 and eIF4F, two key eukaryotic translation initiation factors, are inactivated in mammalian cells. We report an unconventional translation initiation pathway utilized by the leaderless mRNA in vitro, in addition to the previously described 80S-, eIF2-, or eIF2D-mediated modes. This mechanism is a bacterial-like eIF5B/IF2-assisted initiation that has only been reported for hepatitis C virus-like internal ribosome entry sites (IRESs). Therefore, the leaderless mRNA is able to take any of four different translation initiation pathways in eukaryotes.


Subject(s)
Eukaryotic Cells/physiology , Peptide Chain Initiation, Translational/physiology , RNA, Messenger/metabolism , Cell-Free System , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , HEK293 Cells , Hepatitis C/genetics , Humans , Internal Ribosome Entry Sites , Multiprotein Complexes , Protein Biosynthesis , RNA, Messenger/genetics , Saccharomyces cerevisiae/genetics , Transfection/methods
14.
Nucleic Acids Res ; 44(4): 1882-93, 2016 Feb 29.
Article in English | MEDLINE | ID: mdl-26717981

ABSTRACT

During eukaryotic translation initiation, 43S ribosomal complex scans mRNA leader unless an AUG codon in an appropriate context is found. Establishing the stable codon-anticodon base-pairing traps the ribosome on the initiator codon and triggers structural rearrangements, which lead to Pi release from the eIF2-bound GTP. It is generally accepted that AUG recognition by the scanning 43S complex sets the final point in the process of start codon selection, while latter stages do not contribute to this process. Here we use translation reconstitution approach and kinetic toe-printing assay to show that after the 48S complex is formed on an AUG codon, in case GTP hydrolysis is impaired, the ribosomal subunit is capable to resume scanning and slides downstream to the next AUG. In contrast to leaky scanning, this sliding is not limited to AUGs in poor nucleotide contexts and occurs after a relatively long pause at the recognized AUG. Thus, recognition of an AUG per se does not inevitably lead to this codon being selected for initiation of protein synthesis. Instead, it is eIF5-induced GTP hydrolysis and Pi release that irreversibly trap the 48S complex, and this complex is further stabilized by eIF5B and 60S joining.


Subject(s)
Eukaryotic Initiation Factor-2/genetics , Peptide Initiation Factors/genetics , Protein Biosynthesis , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Ribosomes/genetics , Anticodon/genetics , Codon/genetics , Escherichia coli , Eukaryotic Initiation Factor-2/metabolism , Guanosine Triphosphate/metabolism , HeLa Cells , Humans , Hydrolysis , Kinetics , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Peptide Initiation Factors/metabolism , RNA-Binding Proteins/metabolism , Eukaryotic Translation Initiation Factor 5A
15.
Biochimie ; 121: 228-37, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26700150

ABSTRACT

Unspliced human immunodeficiency virus-1 (HIV-1) mRNA is capped and therefore can be translated via conventional scanning mechanism. In addition, its 5' untranslated region (5'UTR) is thought to function as an internal ribosome entry site (IRES) during G2/M-phase of cell cycle or when cap-dependent translation is inhibited. Recently, customary methods of internal initiation demonstrating have been challenged, and consequently existence of certain IRESs of cellular origin has been put under question. Since a precise knowledge of translation initiation mechanism used by HIV may be important for cure development, presence of the IRES in HIV-1 mRNA demands a careful reexamination using contemporary stringent criteria. The key point of our strategy is to compare translation efficiency of bicistronic mRNA bearing HIV-1 unspliced mRNA 5' UTR in the intercistronic position to that of the corresponding capped monocistronic mRNA. This approach allows determination of internal initiation contribution into the overall level of particular mRNA translation. We found that both in cell-free systems and in cultured cells monocistronic mRNA with HIV-1 unspliced mRNA 5'UTR is translated significantly better than bicistronic one. Importantly, it is also true for G2/M-phase stalled cells or for cells under conditions of inhibited cap-dependent translation. Thus, in our hands contribution of internal ribosome entry into the overall level of translation driven by HIV-1 unspliced mRNA 5'UTR is negligible, and 5'-dependent scanning is a primary mechanism of its translation initiation.


Subject(s)
HIV-1/genetics , HIV-1/metabolism , Internal Ribosome Entry Sites/genetics , 5' Untranslated Regions/genetics , Cell Line , Humans , Jurkat Cells/metabolism , RNA, Messenger/genetics
16.
Gene ; 578(1): 1-6, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26680098

ABSTRACT

Protein synthesis in eukaryotes is subject to stringent control. The misregulation of translation of certain mRNAs is often a hallmark of many diseases, including malignancies and autoimmune disorders. To understand why and how it happens, it is important to investigate the translational control of specific mRNAs. In this case, one could use reporter mRNAs in order to identify cis-acting elements responsible for regulation. Here we overview plasmid DNA (pDNA) and mRNA transfections, their pitfalls and limitations, as well as some emerging applications for mRNA transfection.


Subject(s)
Mammals/metabolism , Plasmids/genetics , Protein Biosynthesis , RNA, Messenger/genetics , Animals , Genes, Reporter , Humans , Mammals/genetics , Regulatory Sequences, Nucleic Acid , Transfection
17.
Elife ; 4: e03971, 2015 Jan 26.
Article in English | MEDLINE | ID: mdl-25621764

ABSTRACT

Eukaryotic cells rapidly reduce protein synthesis in response to various stress conditions. This can be achieved by the phosphorylation-mediated inactivation of a key translation initiation factor, eukaryotic initiation factor 2 (eIF2). However, the persistent translation of certain mRNAs is required for deployment of an adequate stress response. We carried out ribosome profiling of cultured human cells under conditions of severe stress induced with sodium arsenite. Although this led to a 5.4-fold general translational repression, the protein coding open reading frames (ORFs) of certain individual mRNAs exhibited resistance to the inhibition. Nearly all resistant transcripts possess at least one efficiently translated upstream open reading frame (uORF) that represses translation of the main coding ORF under normal conditions. Site-specific mutagenesis of two identified stress resistant mRNAs (PPP1R15B and IFRD1) demonstrated that a single uORF is sufficient for eIF2-mediated translation control in both cases. Phylogenetic analysis suggests that at least two regulatory uORFs (namely, in SLC35A4 and MIEF1) encode functional protein products.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Gene Expression Regulation/genetics , Open Reading Frames/genetics , Protein Biosynthesis/genetics , Repressor Proteins/metabolism , Arsenites/pharmacology , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Mutagenesis, Site-Directed , Oxidative Stress/drug effects , Oxidative Stress/genetics , Protein Biosynthesis/drug effects , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomes/drug effects , Ribosomes/metabolism , Sodium Compounds/pharmacology
18.
Crit Rev Biochem Mol Biol ; 49(2): 164-77, 2014.
Article in English | MEDLINE | ID: mdl-24520918

ABSTRACT

The conventional paradigm of translation initiation in eukaryotes states that the cap-binding protein complex eIF4F (consisting of eIF4E, eIF4G and eIF4A) plays a central role in the recruitment of capped mRNAs to ribosomes. However, a growing body of evidence indicates that this paradigm should be revised. This review summarizes the data which have been mostly accumulated in a post-genomic era owing to revolutionary techniques of transcriptome-wide analysis. Unexpectedly, these techniques have uncovered remarkable diversity in the recruitment of cellular mRNAs to eukaryotic ribosomes. These data enable a preliminary classification of mRNAs into several groups based on their requirement for particular components of eIF4F. They challenge the widely accepted concept which relates eIF4E-dependence to the extent of secondary structure in the 5' untranslated regions of mRNAs. Moreover, some mRNA species presumably recruit ribosomes to their 5' ends without the involvement of either the 5' m(7)G-cap or eIF4F but instead utilize eIF4G or eIF4G-like auxiliary factors. The long-standing concept of internal ribosome entry site (IRES)-elements in cellular mRNAs is also discussed.


Subject(s)
RNA, Messenger/metabolism , Ribosomes/metabolism , Transcriptome , Animals , Eukaryotic Initiation Factors/metabolism , Humans , Protein Biosynthesis , RNA, Messenger/chemistry , RNA, Messenger/genetics , Ribosomes/chemistry , Ribosomes/genetics
19.
Nucleic Acids Res ; 41(3): 1807-16, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23268449

ABSTRACT

Resistance of translation of some eukaryotic messenger RNAs (mRNAs) to inactivation of the cap-binding factor eIF4E under unfavorable conditions is well documented. To date, it is the mechanism of internal ribosome entry that is predominantly thought to underlay this stress tolerance. However, many cellular mRNAs that had been considered to contain internal ribosome entry sites (IRESs) failed to pass stringent control tests for internal initiation, thus raising the question of how they are translated under stress conditions. Here, we show that inserting an eIF4G-binding element from a virus IRES into 5'-UTRs of strongly cap-dependent mRNAs dramatically reduces their requirement for the 5'-terminal m(7)G-cap, though such cap-independent translation remains dependent on a vacant 5'-terminus of these mRNAs. Importantly, direct binding of eIF4G to the 5'-UTR of mRNA makes its translation resistant to eIF4F inactivation both in vitro and in vivo. These data may substantiate a new paradigm of translational control under stress to complement IRES-driven mechanism of translation.


Subject(s)
5' Untranslated Regions , Peptide Chain Initiation, Translational , Eukaryotic Initiation Factor-4F/metabolism , Eukaryotic Initiation Factor-4G/metabolism , HEK293 Cells , Humans , RNA Caps/physiology , Ribosomes/metabolism
20.
FEBS Lett ; 586(23): 4139-43, 2012 Nov 30.
Article in English | MEDLINE | ID: mdl-23085065

ABSTRACT

We have previously shown that translation driven by the 5' UTR of Apaf-1 mRNA is relatively efficient in the absence of m7G-cap, but no IRES is involved. Nevertheless, it may be speculated that a "silent" IRES is activated under apoptosis conditions. Here, we show that translation of the mRNA with the Apaf-1 5' UTR is relatively resistant to apoptosis induced by etoposide when eIF4E is sequestered by 4E-BP and eIF4G is partially cleaved. However, translation under these conditions remains governed by 5' end-dependent scanning. We hypothesize that the observed phenomenon is based on the intrinsic low cap-dependence of the Apaf-1 5' UTR.


Subject(s)
5' Untranslated Regions/genetics , Apoptotic Protease-Activating Factor 1/genetics , Apoptotic Protease-Activating Factor 1/metabolism , RNA, Messenger/genetics , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Cell Line , Humans
SELECTION OF CITATIONS
SEARCH DETAIL